A pre-existing coordinated inflammatory microenvironment is associated with complete response of vulvar high-grade squamous intraepithelial lesions to different forms of immunotherapy

Ziena Abdulrahman, Noel F C C de Miranda, Bart W J Hellebrekers, Peggy J de Vos van Steenwijk, Edith M G van Esch, Sjoerd H van der Burg, Mariette I E van Poelgeest, Ziena Abdulrahman, Noel F C C de Miranda, Bart W J Hellebrekers, Peggy J de Vos van Steenwijk, Edith M G van Esch, Sjoerd H van der Burg, Mariette I E van Poelgeest

Abstract

Immunotherapy of vulvar high-grade squamous intraepithelial lesion (vHSIL) is investigated as an alternative for surgery, because of high comorbidity and risk of recurrence. Limited evidence exists on the role and composition of the immune microenvironment in current immunotherapeutic approaches for vHSIL. The vHSIL of 29 patients biopsied before treatment with imiquimod were analyzed by two multiplex seven-color immunofluorescence panels to investigate the pre-existing T-cell and myeloid cell composition in relation to treatment response. The samples were scanned with the Vectra multispectral imaging system. Cells were automatically phenotyped and counted with inForm advanced image analysis software. Cell counts and composition were compared to that of vHSIL patients before therapeutic vaccination (n = 29) and to healthy vulva (n = 27). Our data show that the immune microenvironment of complete responders (CR) to imiquimod resembled the coordinated infiltration with type 1 CD4+ and CD8+ T cells and CD14+ inflammatory myeloid cells also found in healthy vulva. However, more CD8+ T cells and FoxP3+ regulatory T cells were present in CR. The lesions of partial responders (PR) lacked such a coordinated response and displayed an impaired influx of CD14+ inflammatory myeloid cells. Importantly, complete responses after imiquimod or therapeutic vaccination showed the same dependency on a pre-existing coordinated type 1 T-cell and CD14+ myeloid cell infiltration. In conclusion, a good clinical outcome after two different forms of immunotherapy for vHSIL is associated with the presence of a primary inflammatory process resulting in the coordinated influx of several types of immune cells which is then amplified.

Keywords: TLR7; imiquimod; immune microenvironment; therapeutic vaccine; vulvar HSIL.

Conflict of interest statement

The authors declare no potential conflict of interest.

© 2020 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.

Figures

FIGURE 1
FIGURE 1
Multiplex immunofluorescence to detect T cells and myeloid cells in vHSIL. A, Composite image including all individual markers of the seven‐color T‐cell panel staining, consisting of the markers CD3, CD8, FoxP3, Tbet, Tim3, PD‐1 and DAPI. B, Composite image including all individual markers of the seven‐color myeloid cell panel staining, consisting of the markers CD33, CD68, CD163, PD‐L1, CD14, CD11c and DAPI
FIGURE 2
FIGURE 2
The pre‐imiquimod TME differs between the different response groups. A, The numbers of intraepithelial and stromal infiltrating T‐cell and myeloid cell subtypes are presented as cells/mm2 epithelium and cells/mm2 stroma in the pre‐imiquimod vHSIL biopsies of 29 patients. Each dot represents an individual sample, the horizontal bars indicate the median cell counts and the vertical bars are the 95% confidence intervals. B, T‐cell and C, myeloid‐cell infiltrate of the pre‐imiquimod vHSIL patients when grouped according to response (PR, CR), and compared to healthy vulvar tissue (n = 27), presented as median cells/mm2 epithelium and median cells/mm2 stroma. A threshold of a median cell count ≥10 cells/mm2 in at least one group (NR, PR, CR or healthy vulva) was applied to study the changes in biologically common phenotypes. CR, complete responders (n = 14); PR, partial responders (n = 12)
FIGURE 3
FIGURE 3
Correlation between the absolute numbers of tissue infiltrating T cells and myeloid cells in the pre‐imiquimod vHSIL biopsies of partial responders and complete responders. Nonparametric Spearman r correlation analysis (two‐tailed) was performed to analyze the co‐infiltration of the indicated different immune cell subtypes in the epithelium (E) and stroma (S), and is shown in a heatmap for vHSIL partial responders and complete responders to topical imiquimod treatment. Red indicates a strong positive correlation between the two cell phenotypes, blue a strong negative correlation, the green squares indicate the most striking differences between the partial and complete responders. A threshold of a median cell count ≥10 cells/mm2 in at least one group (NR, PR, CR or healthy vulva) was applied to study the changes in biologically common phenotypes. CR, complete responders (n = 14); PR, partial responders (n = 12)
FIGURE 4
FIGURE 4
Fractional differences between the pre‐imiquimod and pre‐vaccination immune microenvironment in vHSIL, divided based on clinical response to the respective therapy. A, Fractional composition of the T cells and myeloid cells in the pre‐imiquimod vHSIL cohort, grouped according to response (NR: n = 3, PR: n = 12, CR: n = 14), and compared to healthy vulva (n = 27). B, Fractional composition of the T cells and myeloid cells in the pretherapeutic vaccination vHSIL cohort, grouped according to response (NR: n = 12, PR: n = 10, CR: n = 7), and compared to healthy vulva (n = 27)
FIGURE 5
FIGURE 5
Summary of complete responder's vHSIL immune microenvironment in the context of the mode of action of imiquimod and therapeutic vaccination. The vHSIL immune microenvironment of complete responders to imiquimod and therapeutic HPV16 synthetic long peptide (SLP) vaccination comprises a coordinated infiltration with type 1 (Tbet+) CD4+ and CD8+ T cells as well as CD14+ inflammatory myeloid cells. Imiquimod stimulates CD14+ myeloid cells to produce pro‐inflammatory cytokines which activate and attract the type 1 T cells. 13 Whereas, therapeutic vaccination stimulates the type 1 T‐cell response, which in turn produces cytokines that also stimulate the influx of the lesion by CD14+ myeloid cells. 9 This figure was created using adapted images of Servier Medical Art, licensed under a creative commons attribution 3.0 unported license

References

    1. Hurt CN, Jones S, Madden TA, et al. Recurrence of vulval intraepithelial neoplasia following treatment with cidofovir or imiquimod: results from a multicentre, randomised, phase II trial (RT3VIN). BJOG. 2018;125:1171‐1177.
    1. van Seters M, van Beurden M, ten Kate FJ, et al. Treatment of vulvar intraepithelial neoplasia with topical imiquimod. N Engl J Med. 2008;358:1465‐1473.
    1. van Poelgeest MI, Welters MJ, 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;22:2342‐2350.
    1. Stanley MA. Imiquimod and the imidazoquinolones: mechanism of action and therapeutic potential. Clin Exp Dermatol. 2002;27:571‐577.
    1. Schon MP, Schon M. Imiquimod: mode of action. Br J Dermatol. 2007;157(Suppl 2):8‐13.
    1. Schon M, Bong AB, Drewniok C, et al. Tumor‐selective induction of apoptosis and the small‐molecule immune response modifier imiquimod. J Natl Cancer Inst. 2003;95:1138‐1149.
    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;102:1129‐1136.
    1. Winters U, Daayana S, Lear JT, et al. Clinical and immunologic results of a phase II trial of sequential imiquimod and photodynamic therapy for vulval intraepithelial neoplasia. Clin Cancer Res. 2008;14:5292‐5299.
    1. Abdulrahman Z, de Miranda N, van Esch EMG, et al. Pre‐existing inflammatory immune microenvironment predicts the clinical response of vulvar high‐grade squamous intraepithelial lesions to therapeutic HPV16 vaccination. J Immunother Cancer. 2020;8:e000563.
    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.
    1. Santegoets S, de Groot AF, Dijkgraaf EM, et al. The blood mMDSC to DC ratio is a sensitive and easy to assess independent predictive factor for epithelial ovarian cancer survival. Onco Targets Ther. 2018;7:e1465166.
    1. Fridman WH, Zitvogel L, Sautes‐Fridman C, Kroemer G. The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol. 2017;14:717‐734.
    1. Gibson SJ, Imbertson LM, Wagner TL, et al. Cellular requirements for cytokine production in response to the immunomodulators imiquimod and S‐27609. J Interferon Cytokine Res. 1995;15:537‐545.
    1. van Poelgeest MI, van Seters M, van Beurden M, et al. Detection of human papillomavirus (HPV) 16‐specific CD4+ T‐cell immunity in patients with persistent HPV16‐induced vulvar intraepithelial neoplasia in relation to clinical impact of imiquimod treatment. Clin Cancer Res. 2005;11:5273‐5280.
    1. Welters MJ, 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 USA. 2010;107:11895‐11899.
    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;8:334ra52.
    1. Melief CJM, Welters MJ, Vergote I, et al. Strong vaccine responses during chemotherapy are associated with prolonged cancer survival. Sci Transl Med. 2020;12:eaaz8235.
    1. Wang E, Worschech A, Marincola FM. The immunologic constant of rejection. Trends Immunol. 2008;29:256‐262.
    1. Wang E, Monaco A, Monsurró V, et al. Antitumor vaccines, immunotherapy and the immunological constant of rejection. IDrugs. 2009;12:297.
    1. Karim R, Tummers B, Meyers C, et al. Human papillomavirus (HPV) upregulates the cellular deubiquitinase UCHL1 to suppress the keratinocyte's innate immune response. PLoS Pathog. 2013;9:e1003384.
    1. Tummers B, Goedemans R, Pelascini LP, et al. The interferon‐related developmental regulator 1 is used by human papillomavirus to suppress NFkappaB activation. Nat Commun. 2015;6:6537.
    1. Ma W, Melief CJ, van der Burg SH. Control of immune escaped human papilloma virus is regained after therapeutic vaccination. Curr Opin Virol. 2017;23:16‐22.
    1. Aulmann S, Schleibaum J, Penzel R, Schirmacher P, Gebauer G, Sinn HP. 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;61:1034‐1037.
    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;106:43‐48.
    1. Rosenthal AN, Ryan A, Hopster D, Surentheran T, Jacobs IJ. 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.
    1. Jones SEF, Hibbitts S, Hurt CN, et al. Human papillomavirus DNA methylation predicts response to treatment using Cidofovir and Imiquimod in Vulval intraepithelial Neoplasia 3. Clin Cancer Res. 2017;23:5460‐5468.

Source: PubMed

3
Subscribe