Pre-existing inflammatory immune microenvironment predicts the clinical response of vulvar high-grade squamous intraepithelial lesions to therapeutic HPV16 vaccination

Ziena Abdulrahman, Noel de Miranda, Edith M G van Esch, Peggy J de Vos van Steenwijk, Hans W Nijman, Marij J P Welters, Mariette I E van Poelgeest, Sjoerd H van der Burg, Ziena Abdulrahman, Noel de Miranda, Edith M G van Esch, Peggy J de Vos van Steenwijk, Hans W Nijman, Marij J P Welters, Mariette I E van Poelgeest, Sjoerd H van der Burg

Abstract

Background: Vulvar high-grade squamous intraepithelial lesion (vHSIL) is predominantly induced by high-risk human papilloma virus type 16 (HPV16). In two independent trials, therapeutic vaccination against the HPV16 E6 and E7 oncoproteins resulted in objective partial and complete responses (PRs/CRs) in half of the patients with HPV16+ vHSIL at 12-month follow-up. Here, the prevaccination and postvaccination vHSIL immune microenvironment in relation to the vaccine-induced clinical response was investigated.

Methods: Two novel seven-color multiplex immunofluorescence panels to identify T cells (CD3, CD8, Foxp3, Tim3, Tbet, PD-1, DAPI) and myeloid cells (CD14, CD33, CD68, CD163, CD11c, PD-L1, DAPI) were designed and fully optimized for formalin-fixed paraffin-embedded tissue. 29 prevaccination and 24 postvaccination biopsies of patients with vHSIL, and 27 healthy vulva excisions, were stained, scanned with the Vectra multispectral imaging system, and automatically phenotyped and counted using inForm advanced image analysis software.

Results: Healthy vulvar tissue is strongly infiltrated by CD4 and CD8 T cells expressing Tbet and/or PD-1 and CD14+HLA-DR+ inflammatory myeloid cells. The presence of such a coordinated pre-existing proinflammatory microenvironment in HPV16+ vHSIL is associated with CR after vaccination. In partial responders, a disconnection between T cell and CD14+ myeloid cell infiltration was observed, whereas clinical non-responders displayed overall lower immune cell infiltration. Vaccination improved the coordination of local immunity, reflected by increased numbers of CD4+Tbet+ T cells and HLA-DR+CD14+ expressing myeloid cells in patients with a PR or CR, but not in patients with no response. CD8+ T cell infiltration was not increased after vaccination.

Conclusion: A prevaccination inflamed type 1 immune contexture is required for stronger vaccine-induced immune infiltration and is associated with better clinical response. Therapeutic vaccination did not overtly increase immune infiltration of cold lesions.

Keywords: immunotherapy; therapeutic vaccination; tumor microenvironment; vulvar HSIL.

Conflict of interest statement

Competing interests: SHvdB is being named as an inventor on the patent for the use of synthetic long peptides as vaccine for the treatment of HPV-induced diseases, which is exploited by ISA Pharmaceuticals. SHvdB serves as a paid member of the strategy board of ISA Pharmaceuticals. No other potential conflicts of interest relevant to this article were reported.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Multiplex immunofluorescence staining to detect T cells and myeloid cells. Two multiplex panels were developed to detect T cells and myeloid cells in vulvar tissue. (A) The expression of each of the seven markers for T cells showing CD3, CD8, FoxP3, Tbet, Tim3, PD-1 and DAPI as well as a figure showing all seven markers simultaneously. Arrows indicate examples of analyzed phenotypes. (B) The expression of each of the seven markers for myeloid cells showing CD33, CD68, CD163, PD-L1, CD14, CD11c and DAPI as well as a figure showing all seven markers simultaneously. Arrows indicate examples of analyzed phenotypes.
Figure 2
Figure 2
Healthy vulvar tissue is infiltrated by activated type 1 T cells and several types of myeloid cells while the vulvar high-grade squamous intraepithelial lesion (vHSIL) immune infiltrate is highly heterogenic among different patients. The numbers of intraepithelial and stroma-infiltrating T cell and myeloid cell subtypes, of which the median cell count exceeded the threshold of ≥10 cells/mm2, are presented as cells/mm2 for (A) human papillomavirus-negative healthy vulva (n=27) and (B) vHSIL (n=29) before vaccination. Each dot represents an individual sample, the horizontal bars indicate the median cell counts, and the vertical bars are the 95% CIs.
Figure 3
Figure 3
The tumor microenvironment of complete responders (CR, n=7) displays a similar immune infiltration pattern as found in healthy vulvar tissue. The numbers of intraepithelial and stroma-infiltrating T cell and myeloid cell subtypes, of which the median cell count exceeded the threshold of ≥10 cells/mm2, are presented as cells/mm2 in the (A) prevaccination (n=29) and (B) postvaccination (n=24) vulvar high-grade squamous intraepithelial lesion biopsies of vaccinated patients grouped according to their best clinical response during the 12 months of follow-up, and compared with healthy vulvar tissue (n=27). NR, non-responders (n=12); PR, partial responders (n=10). Statistical differences in the cell types between patient groups are depicted in online supplementary files 6 and 9. (C) Highlighted T cell and myeloid cell subtypes that significantly differed among different response groups, shown as fractional differences in all CD3+ T cells (left) or all CD14+ and CD68+ myeloid cells (right), prevaccination and postvaccination.
Figure 4
Figure 4
Correlation between the absolute numbers of tissue-infiltrating T cells and myeloid cells in the pretreatment and post-treatment vulvar high-grade squamous intraepithelial lesion (vHSIL) biopsies and in healthy vulvar tissue. Non-parametric Spearman r correlation analysis (two tailed) was performed to analyze the coinfiltration of the indicated different immune cell subtypes in the epithelium (E) and stroma (S) and is shown in a heatmap for healthy vulvar tissue (n=27), prevaccination complete responder patients with vHSIL (n=7) and partial responder patients with vHSIL (n=10) as well as postvaccination partial responder patients with vHSIL (n=9).
Figure 5
Figure 5
Immune profile of non-responders (NR), partial responders (PR) and complete responders (CR) bases on the relative infiltration of significantly different immune cell subtypes and human papillomavirus (HPV)-specific T cell reactivity. The strength of the vaccine-induced HPV16-specific T cell response present in peripheral blood mononuclear cells (PBMC) as measured by the production of interferon γ (IFNγ) in the supernatant of PBMC stimulated with six different E6 and E7 peptide pools for clinical NR, PR and CR as determined previously by cytokine bead array is depicted as (A) the mean±SEM of all patients to all six peptide pools tested, before and after two and four vaccinations; and (B) the median of max calculated by taking the median of the highest responses to each peptide pool after two or four vaccinations. Shown is the mean±SEM of all medians per group. (C) and (D) The cell counts of all immune subsets of which the median cell count exceeded the threshold of ≥10 cells/mm2 and the numbers significantly differed between at least two of the patient groups (NR, PR, CR) either before vaccination (C) or after vaccination (D) as well as the median of max of IFNγ production as determined by cytometric bead array are depicted after the data were z-transformed to enable comparison of relative differences.

References

    1. Galon J, Angell HK, Bedognetti D, et al. . The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures. Immunity 2013;39:11–26. 10.1016/j.immuni.2013.07.008
    1. Fridman WH, Pagès F, Sautès-Fridman C, et al. . The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012;12:298–306. 10.1038/nrc3245
    1. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized immunotherapy for human cancer. Science 2015;348:62–8. 10.1126/science.aaa4967
    1. Fesnak AD, June CH, Levine BL. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat Rev Cancer 2016;16:566–81. 10.1038/nrc.2016.97
    1. Sharma P, Allison JP. The future of immune checkpoint therapy. Science 2015;348:56–61. 10.1126/science.aaa8172
    1. Whiteside TL, Demaria S, Rodriguez-Ruiz ME, et al. . Emerging opportunities and challenges in cancer immunotherapy. Clin Cancer Res 2016;22:1845–55. 10.1158/1078-0432.CCR-16-0049
    1. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res 2016;22:1865–74. 10.1158/1078-0432.CCR-15-1507
    1. Tumeh PC, Harview CL, Yearley JH, et al. . Pd-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–71. 10.1038/nature13954
    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
    1. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197–218. 10.1038/s41573-018-0007-y
    1. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 2017;541:321–30. 10.1038/nature21349
    1. McLaughlin-Drubin ME, Munger K. Viruses associated with human cancer. Biochim Biophys Acta 2008;1782:127–50. 10.1016/j.bbadis.2007.12.005
    1. Welters MJP, Kenter GG, Piersma SJ, et al. . Induction of tumor-specific CD4+ and CD8+ T-cell immunity in cervical cancer patients by a human papillomavirus type 16 E6 and E7 long peptides vaccine. Clin Cancer Res 2008;14:178–87. 10.1158/1078-0432.CCR-07-1880
    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. Bagarazzi ML, Yan J, Morrow MP, et al. . Immunotherapy against HPV16/18 generates potent Th1 and cytotoxic cellular immune responses. Sci Transl Med 2012;4:155ra138–38. 10.1126/scitranslmed.3004414
    1. Kim TJ, Jin H-T, Hur S-Y, et al. . Clearance of persistent HPV infection and cervical lesion by therapeutic DNA vaccine in CIN3 patients. Nat Commun 2014;5:5317 10.1038/ncomms6317
    1. de Vos van Steenwijk PJ, Ramwadhdoebe TH, Goedemans R, et al. . Tumor-Infiltrating CD14-positive myeloid cells and CD8-positive T-cells prolong survival in patients with cervical carcinoma. Int J Cancer 2013;133:n/a–94. 10.1002/ijc.28309
    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. . Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med 2009;361:1838–47. 10.1056/NEJMoa0810097
    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. 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. Trimble CL, Morrow MP, Kraynyak KA, et al. . Safety, efficacy, and immunogenicity of VGX-3100, a therapeutic synthetic DNA vaccine targeting human papillomavirus 16 and 18 E6 and E7 proteins for cervical intraepithelial neoplasia 2/3: a randomised, double-blind, placebo-controlled phase 2B trial. Lancet 2015;386:2078–88. 10.1016/S0140-6736(15)00239-1
    1. Massarelli E, William W, Johnson F, 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 2018.
    1. Melief CJM, Welters MJP, Vergote I, et al. . Strong vaccine responses during chemotherapy are associated with prolonged cancer survival. Nature 2019.
    1. Morrow MP, Kraynyak KA, Sylvester AJ, et al. . Clinical and immunologic biomarkers for histologic regression of high-grade cervical dysplasia and clearance of HPV16 and HPV18 after immunotherapy. Clin Cancer Res 2018;24:276–94. 10.1158/1078-0432.CCR-17-2335
    1. van Esch EMG, van Poelgeest MIE, Kouwenberg S, et al. . Expression of coinhibitory receptors on T cells in the microenvironment of usual vulvar intraepithelial neoplasia is related to proinflammatory effector T cells and an increased recurrence-free survival. Int J Cancer 2015;136:E95–106. 10.1002/ijc.29174
    1. van Esch EMG, van Poelgeest MIE, Trimbos JBMZ, et al. . Intraepithelial macrophage infiltration is related to a high number of regulatory T cells and promotes a progressive course of HPV-induced vulvar neoplasia. Int J Cancer 2015;136:E85–94. 10.1002/ijc.29173
    1. Ijsselsteijn ME, Brouwer TP, Abdulrahman Z, et al. . Cancer immunophenotyping by seven-colour multispectral imaging without tyramide signal amplification. J Pathol Clin Res 2019;5:3–11. 10.1002/cjp2.113
    1. Daayana S, Elkord E, Winters U, et al. . Phase II trial of imiquimod and HPV therapeutic vaccination in patients with vulval intraepithelial neoplasia. Br J Cancer 2010;102:1129–36. 10.1038/sj.bjc.6605611
    1. Mauldin IS, Wages NA, Stowman AM, et al. . Topical treatment of melanoma metastases with imiquimod, plus administration of a cancer vaccine, promotes immune signatures in the metastases. Cancer Immunol Immunother 2016;65:1201–12. 10.1007/s00262-016-1880-z
    1. Barnetson RSC, Satchell A, Zhuang L, et al. . Imiquimod induced regression of clinically diagnosed superficial basal cell carcinoma is associated with early infiltration by CD4 T cells and dendritic cells. Clin Exp Dermatol 2004;29:639–43. 10.1111/j.1365-2230.2004.01614.x
    1. van der Sluis TC, Sluijter M, van Duikeren S, et al. . Therapeutic peptide vaccine-induced CD8 T cells strongly modulate intratumoral macrophages required for tumor regression. Cancer Immunol Res 2015;3:1042–51. 10.1158/2326-6066.CIR-15-0052
    1. Aulmann S, Schleibaum J, Penzel R, et al. . Gains of chromosome region 3q26 in intraepithelial neoplasia and invasive squamous cell carcinoma of the vulva are frequent and independent of HPV status. J Clin Pathol 2008;61:1034–7. 10.1136/jcp.2008.056275
    1. Bryndorf T, Kirchhoff M, Larsen J, et al. . The most common chromosome aberration detected by high-resolution comparative genomic hybridization in vulvar intraepithelial neoplasia is not seen in vulvar squamous cell carcinoma. Cytogenet Genome Res 2004;106:43–8. 10.1159/000078559
    1. Rosenthal AN, Ryan A, Hopster D, et al. . High frequency of loss of heterozygosity in vulval intraepithelial neoplasia (VIN) is associated with invasive vulval squamous cell carcinoma (VSCC). Int J Cancer 2001;94:896–900. 10.1002/ijc.1549
    1. Davoli T, Uno H, Wooten EC, et al. . Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 2017;355:eaaf8399 10.1126/science.aaf8399
    1. Davidson EJ, Boswell CM, Sehr P, et al. . Immunological and clinical responses in women with vulval intraepithelial neoplasia vaccinated with a vaccinia virus encoding human papillomavirus 16/18 oncoproteins. Cancer Res 2003;63:6032–41.
    1. Melief SM, Visconti VV, Visser M, et al. . Long-Term survival and clinical benefit from adoptive T-cell transfer in stage IV melanoma patients is determined by a Four-parameter tumor immune signature. Cancer Immunol Res 2017;5:170–9. 10.1158/2326-6066.CIR-16-0288
    1. Gajewski TF. The next hurdle in cancer immunotherapy: overcoming the Non-T-Cell-Inflamed tumor microenvironment. Semin Oncol 2015;42:663–71. 10.1053/j.seminoncol.2015.05.011
    1. Baldwin PJ, van der Burg SH, Boswell CM, et al. . Vaccinia-Expressed human papillomavirus 16 and 18 E6 and E7 as a therapeutic vaccination for vulval and vaginal intraepithelial neoplasia. Clin Cancer Res 2003;9:5205–13.
    1. Signorelli D, Giannatempo P, Grazia G, et al. . Patients selection for immunotherapy in solid tumors: overcome the naïve vision of a single biomarker. Biomed Res Int 2019;9056417.

Source: PubMed

3
Iratkozz fel