Intradermal vaccination of HPV-16 E6 synthetic peptides conjugated to an optimized Toll-like receptor 2 ligand shows safety and potent T cell immunogenicity in patients with HPV-16 positive (pre-)malignant lesions

Frank M Speetjens, Marij J P Welters, Marije Slingerland, Mariette I E van Poelgeest, Peggy J de Vos van Steenwijk, Inge Roozen, Sanne Boekestijn, Nikki M Loof, Gijs G Zom, A Rob P M Valentijn, Willem-Jan Krebber, Nico J Meeuwenoord, Catharina A H Janssen, Cornelis J M Melief, Gijs A van der Marel, Dmitri V Filippov, Sjoerd H van der Burg, Hans Gelderblom, Ferry Ossendorp, Frank M Speetjens, Marij J P Welters, Marije Slingerland, Mariette I E van Poelgeest, Peggy J de Vos van Steenwijk, Inge Roozen, Sanne Boekestijn, Nikki M Loof, Gijs G Zom, A Rob P M Valentijn, Willem-Jan Krebber, Nico J Meeuwenoord, Catharina A H Janssen, Cornelis J M Melief, Gijs A van der Marel, Dmitri V Filippov, Sjoerd H van der Burg, Hans Gelderblom, Ferry Ossendorp

Abstract

Background: Amplivant is a molecularly optimized Toll-like receptor 2 ligand that can be covalently conjugated to tumor peptide antigens. In preclinical models, amplivant-adjuvanted synthetic long peptides (SLPs) strongly enhanced antigen presentation by dendritic cells, T cell priming and induction of effective antitumor responses. The current study is a first-in-human trial to investigate safety and immunogenicity of amplivant conjugated to human papillomavirus (HPV) 16-SLP.

Methods: A dose escalation phase I vaccination trial was performed in 25 patients treated for HPV16 positive (pre-)malignant lesions. Amplivant was conjugated to two SLPs derived from the two most immunodominant regions of the HPV16 E6 oncoprotein. The vaccine, containing a mix of these two conjugates in watery solution without any other formulation, was injected intradermally three times with a 3-week interval in four dose groups (1, 5, 20 or 50 µg per conjugated peptide). Safety data were collected during the study. Peptide-specific T cell immune responses were determined in blood samples taken before, during and after vaccination using complementary immunological assays.

Results: Toxicity after three amplivant-conjugated HPV16-SLP vaccinations was limited to grade 1 or 2, observed as predominantly mild skin inflammation at the vaccination site and sometimes mild flu-like symptoms. Adverse events varied from none in the lowest dose group to mild/moderate vaccine-related inflammation in all patients and flu-like symptoms in three out of seven patients in the highest dose group, after at least one injection. In the lowest dose group, vaccine-induced T cell responses were observed in the blood of three out of six vaccinated persons. In the highest dose group, all patients displayed a strong HPV16-specific T cell response after vaccination. These HPV16-specific T cell responses lasted until the end of the trial.

Conclusions: Amplivant-conjugated SLPs can safely be used as an intradermal therapeutic vaccine to induce robust HPV16-specific T cell immunity in patients previously treated for HPV16 positive (pre-) malignancies. Increased vaccine dose was associated with a higher number of mild adverse events and with stronger systemic T cell immunity.

Trial registration numbers: NCT02821494 and 2014-000658-12.

Keywords: Adjuvants, Immunologic; Immunogenicity, Vaccine; Immunotherapy; Vaccination.

Conflict of interest statement

Competing interests: FO, CJMM, DVF and GAvdM are inventors of a patent application related to the work in this article entitled 'Adjuvant compound', with publication number WO 2013/051936 and filing date October 4, 2012. CJMM and WJK receive a salary from ISA Pharmaceuticals BV and are in possession of ISA stock appreciation rights and are inventors on patents that are licensed to or owned by ISA Pharmaceuticals BV, dealing with synthetic long peptide vaccines. SHB is named as an inventor on the patent for the use of synthetic long peptides as vaccine. SHB serves as a paid member of the strategy board of ISA Pharmaceuticals and received honoraria as a consultant for PCI Biotech, IO Biotech and DC prime.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
A schematic overview of the vaccination scheme. Individuals received the vaccine injections within a dose escalation of 1, 5, 20 or 50 µg/peptide – conjugate in patients with HPV16+ tumors or premalignant lesions at weeks 0, 3 and 6. Before, after two injections, three and 20 weeks after last injection, blood was drawn for immunomonitoring. Safety was assessed throughout the whole trial using the CTCAE. CTCAE, Common Terminology Criteria for Adverse Events; HPV, human papillomavirus.
Figure 2
Figure 2
Proliferative and cytokine response rate increases with dose of conjugated peptide vaccine. (A) An example (patient #25) of proliferative response indicated by stimulation index (SI) in the left graph as determined by LST of fresh PBMC. In the middle and right graph the IFNγ and IL-5 production, respectively, as measured by CBA in the LST of the same patient. Peptide pool E6.2 and E6.4 (and to a lesser extent E6.3) contain the conjugated peptides as present in the vaccine. The time points of tested PBMC samples were baseline, week 6 (postvaccination 2), week 9 (postvaccination 3) and week 26 (follow-up). The cut-off in the left graph indicates the counts per minutes in negative control (cells in medium only) plus 3 × SD SI above this cut-off are considered a positive response. In the middle and right graph, the cut-off indicates the threshold of the CBA. (B) A summary of the responses for all patients is presented in a heat map. (C) The percentage of positive proliferative (SI) responses (calculated as number of positive responses divided by the total tested peptide pools times 100) per time point and per vaccine dose level (cohort) is depicted in the upper graph. The numbers above the bars indicate the number of patients. A two-way analysis of variance test was performed to determine the differences between the cohorts. (D) The MoM of SI (LST) was determined per patients per vaccine dose level (cohort) and each dot represents a patient. The Mann-Whithney test was used to determine differences between the dose levels. (E) The cytokines produced on recognition of HPV16 peptide pools and measured by CBA are displayed in a heat map. No cytokine production was measurable on E6.1 or the E7 peptides stimulation and therefore not shown here. CBA, cytometric bead array; LST, lymphocyte stimulation test; MoM, median of maximal response; PBMC, peripheral blood mononuclear cell; SI, stimulation index.
Figure 3
Figure 3
Frequency of IFNγ producing T cells increased with dose of vaccine. (A) An example of the 4-day IFNγ ELISpot results showing patient #19 against the two peptides in the vaccine (pept A: HPV16 E671-95 and pept B: HPV16 E6127-158). The specific spot counts are given per 105 cells. The cut-off defines a spot count of 10 above which a response is positive. (B) An overview of the ELISpot responses to the two vaccine peptides for all patients. (C) The percentage of positive ELISpot responses (calculated as number of positive responses divided by the total tested peptide pools times 100) per time point and per vaccine dose level (cohort) is shown. The numbers above the bars indicate the number of patients. A two-way analysis of variance test was performed to determine the differences between the cohorts. (D) Patients are grouped according to their dose of vaccine and the median of max (MoM, see Methods), is depicted for each patient by a dot. Statistical significant differences are indicated (Mann-Whitney test).
Figure 4
Figure 4
Both CD4+ and CD8+ T cell responses are induced by the conjugated peptide vaccine. A selected (based on responses in the 4-day IFNγ ELISpot assay as well as availability of PBMCs) group of six patients were analyzed in the intracellular cytokine staining (ICS) after a 10-day prestimulation. The PBMCs were only tested for the two vaccine peptides (pept A: HPV16 E671-95 and pept B: HPV16 E6127-158) and SEB was taken along as a positive control. (A) The individual frequencies of the peptide responses (indicated cytokine) per patient as percentage of CD4+ (upper graphs) or CD8+ (lower graphs) T cells are given. (B) A heat map of the ICS responses for both CD4+ (upper) and CD8+ (lower) T cells is displayed. The dose of the vaccine and patient number (ID) is given. PBMCs, peripheral blood mononuclear cells; SEB, staphylococcal enterotoxin B.

References

    1. van der Burg SH, Melief CJM. Therapeutic vaccination against human papilloma virus induced malignancies. Curr Opin Immunol 2011;23:252–7. 10.1016/j.coi.2010.12.010
    1. Kenter GG, Welters MJP, Valentijn ARPM, et al. . Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009;361:1838–47. 10.1056/NEJMoa0810097
    1. Melief CJM, Welters MJP, Vergote I, et al. . Strong vaccine responses during chemotherapy are associated with prolonged cancer survival. Sci Transl Med 2020;12. doi:10.1126/scitranslmed.aaz8235. [Epub ahead of print: 18 03 2020].
    1. van Poelgeest MIE, Welters MJP, Vermeij R, et al. . Vaccination against oncoproteins of HPV16 for noninvasive Vulvar/Vaginal lesions: lesion clearance is related to the strength of the T-cell response. Clin Cancer Res 2016;22:2342–50. 10.1158/1078-0432.CCR-15-2594
    1. Sahin U, Derhovanessian E, Miller M, et al. . Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017;547:222–6. 10.1038/nature23003
    1. Hu Z, Leet DE, Allesøe RL, et al. . Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat Med 2021;27:515–25. 10.1038/s41591-020-01206-4
    1. Saxena M, van der Burg SH, Melief CJM, et al. . Therapeutic cancer vaccines. Nat Rev Cancer 2021;21:360–78. 10.1038/s41568-021-00346-0
    1. Zom GGP, Khan S, Filippov DV, et al. . TLR ligand-peptide conjugate vaccines: toward clinical application. Adv Immunol 2012;114:177–201. 10.1016/B978-0-12-396548-6.00007-X
    1. Willems MMJHP, Zom GG, Khan S, et al. . N-tetradecylcarbamyl lipopeptides as novel agonists for Toll-like receptor 2. J Med Chem 2014;57:6873–8. 10.1021/jm500722p
    1. Zom GG, Willems MMJHP, Khan S, et al. . Novel TLR2-binding adjuvant induces enhanced T cell responses and tumor eradication. J Immunother Cancer 2018;6:146. 10.1186/s40425-018-0455-2
    1. Zom GG, Khan S, Britten CM, et al. . Efficient induction of antitumor immunity by synthetic toll-like receptor ligand-peptide conjugates. Cancer Immunol Res 2014;2:756–64. 10.1158/2326-6066.CIR-13-0223
    1. Khan S, Bijker MS, Weterings JJ, et al. . Distinct uptake mechanisms but similar intracellular processing of two different toll-like receptor ligand-peptide conjugates in dendritic cells. J Biol Chem 2007;282:21145–59. 10.1074/jbc.M701705200
    1. de Vos van Steenwijk PJ, Heusinkveld M, Ramwadhdoebe TH, et al. . An unexpectedly large polyclonal repertoire of HPV-specific T cells is poised for action in patients with cervical cancer. Cancer Res 2010;70:2707–17. 10.1158/0008-5472.CAN-09-4299
    1. de Vos van Steenwijk PJ, van Poelgeest MIE, Ramwadhdoebe TH, et al. . The long-term immune response after HPV16 peptide vaccination in women with low-grade pre-malignant disorders of the uterine cervix: a placebo-controlled phase II study. Cancer Immunol Immunother 2014;63:147–60. 10.1007/s00262-013-1499-2
    1. Kenter GG, Welters MJP, Valentijn ARPM, et al. . Phase I immunotherapeutic trial with long peptides spanning the E6 and E7 sequences of high-risk human papillomavirus 16 in end-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res 2008;14:169–77. 10.1158/1078-0432.CCR-07-1881
    1. van Poelgeest MIE, Welters MJP, van Esch EMG, et al. . HPV16 synthetic long peptide (HPV16-SLP) vaccination therapy of patients with advanced or recurrent HPV16-induced gynecological carcinoma, a phase II trial. J Transl Med 2013;11:88. 10.1186/1479-5876-11-88
    1. Zom GG, Welters MJP, Loof NM, et al. . TLR2 ligand-synthetic long peptide conjugates effectively stimulate tumor-draining lymph node T cells of cervical cancer patients. Oncotarget 2016;7:67087–100. 10.18632/oncotarget.11512
    1. Common terminology criteria for adverse events (CTCAE). Available:
    1. Welters MJ, van der Sluis TC, van Meir H, et al. . Vaccination during myeloid cell depletion by cancer chemotherapy fosters robust T cell responses. Sci Transl Med 2016;8:ra52. 10.1126/scitranslmed.aad8307
    1. Welters MJP, Kenter GG, de Vos van Steenwijk PJ, et al. . Success or failure of vaccination for HPV16-positive vulvar lesions correlates with kinetics and phenotype of induced T-cell responses. Proc Natl Acad Sci U S A 2010;107:11895–9. 10.1073/pnas.1006500107
    1. Welters MJP, de Jong A, van den Eeden SJF, et al. . Frequent display of human papillomavirus type 16 E6-specific memory T-helper cells in the healthy population as witness of previous viral encounter. Cancer Res 2003;63:636–41.
    1. Ott PA, Hu Z, Keskin DB, et al. . An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017;547:217–21. 10.1038/nature22991
    1. Deres K, Schild H, Wiesmüller KH, et al. . In vivo priming of virus-specific cytotoxic T lymphocytes with synthetic lipopeptide vaccine. Nature 1989;342:561–4. 10.1038/342561a0
    1. Gential GPP, Hogervorst TP, Tondini E, et al. . Peptides conjugated to 2-alkoxy-8-oxo-adenine as potential synthetic vaccines triggering TLR7. Bioorg Med Chem Lett 2019;29:1340–4. 10.1016/j.bmcl.2019.03.048
    1. Reintjens NRM, Tondini E, de Jong AR, et al. . Self-Adjuvanting cancer vaccines from Conjugation-Ready lipid A analogues and synthetic long peptides. J Med Chem 2020;63:11691–706. 10.1021/acs.jmedchem.0c00851
    1. Karbach J, Neumann A, Atmaca A, et al. . Efficient in vivo priming by vaccination with recombinant NY-ESO-1 protein and CpG in antigen naive prostate cancer patients. Clin Cancer Res 2011;17:861–70. 10.1158/1078-0432.CCR-10-1811
    1. Ohno S, Takano F, Ohta Y, et al. . Frequency of myeloid dendritic cells can predict the efficacy of Wilms' tumor 1 peptide vaccination. Anticancer Res 2011;31:2447–52.
    1. Stebbing J, Dalgleish A, Gifford-Moore A, et al. . An intra-patient placebo-controlled phase I trial to evaluate the safety and tolerability of intradermal IMM-101 in melanoma. Ann Oncol 2012;23:1314–9. 10.1093/annonc/mdr363
    1. Rittig SM, Haentschel M, Weimer KJ, et al. . Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients. Mol Ther 2011;19:990–9. 10.1038/mt.2010.289
    1. Sampson JH, Heimberger AB, Archer GE, et al. . Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J Clin Oncol 2010;28:4722–9. 10.1200/JCO.2010.28.6963
    1. van den Hende M, van Poelgeest MIE, van der Hulst JM, et al. . Skin reactions to human papillomavirus (HPV) 16 specific antigens intradermally injected in healthy subjects and patients with cervical neoplasia. Int J Cancer 2008;123:146–52. 10.1002/ijc.23502

Source: PubMed

Подписаться