Immune-based biomarker accurately predicts response to imiquimod immunotherapy in cervical high-grade squamous intraepithelial lesions

Background The complete response rate of cervical high-grade squamous intraepithelial lesion (cHSIL) patients to imiquimod immunotherapy is approximately 60%. Consequently, many patients are exposed to unnecessary adverse effects of imiquimod. On the other hand, conventional surgical large loop excision therapy is associated with increased risk of premature births in subsequent pregnancies. An in-depth analysis of the cHSIL immune microenvironment was performed in order to identify and develop a predictive biomarker for response to imiquimod, to maximize therapy efficacy and to avoid adverse effects in patients unlikely to respond. Methods Biopsies of 35 cHSIL patients, before and 10 weeks on imiquimod treatment, were analyzed by two multispectral seven-color immunofluorescence panels for T cell and myeloid cell composition in relation to treatment response. Based on these results a simplified immunohistochemical detection protocol was developed. Samples were scanned with the Vectra multispectral imaging system and cells were automatically identified using machine learning. Results The immune microenvironment of complete responders (CR) is characterized by a strong and coordinated infiltration by T helper cells (activated PD1+/type 1 Tbet+), M1-like macrophages (CD68+CD163-) and dendritic cells (CD11c+) prior to imiquimod. The lesions of non-responders (NRs) displayed a high infiltration by CD3+FOXP3+ regulatory T cells. At 10 weeks on imiquimod, a strong influx of intraepithelial and stromal CD4+ T cells was observed in CR but not NR patients. A steep decrease in macrophages occurred both in CR and NR patients, leveling the pre-existing differences in myeloid cell composition between the two groups. Based on the pre-existing immune composition differences, the sum of intraepithelial CD4 T cell, macrophage and dendritic cell counts was used to develop a quantitative simplified one color immunohistochemical biomarker, the CHSIL immune biomarker for imiquimod (CIBI), which can be automatically and unbiasedly quantified and has an excellent predictive capacity (receiver operating characteristic area under the curve 0.95, p<0.0001). Conclusion The capacity of cHSIL patients to respond to imiquimod is associated with a pre-existing coordinated local immune process, fostering an imiquimod-mediated increase in local T cell infiltration. The CIBI immunohistochemical biomarker has strong potential to select cHSIL patients with a high likelihood to experience a complete response to imiquimod immunotherapy.

[1]  L. Kooreman,et al.  Topical Imiquimod Treatment of High-grade Cervical Intraepithelial Neoplasia (TOPIC-3): A Nonrandomized Multicenter Study , 2022, Journal of immunotherapy.

[2]  C. Gerestein,et al.  Efficacy and Long-term Outcomes of Repeated Large Loop Excision of the Transformation Zone of the Cervix , 2022, Obstetrics and gynecology.

[3]  Marieke E. Ijsselsteijn,et al.  Tumor-specific T cells support chemokine-driven spatial organization of intratumoral immune microaggregates needed for long survival , 2022, Journal for ImmunoTherapy of Cancer.

[4]  K. Schmeler,et al.  Topical Imiquimod for the Treatment of High-Grade Squamous Intraepithelial Lesions of the Cervix , 2021, Obstetrics and gynecology.

[5]  Frits Koning,et al.  ImaCytE: Visual Exploration of Cellular Micro-Environments for Imaging Mass Cytometry Data , 2021, IEEE Transactions on Visualization and Computer Graphics.

[6]  Adam J. Rubin,et al.  Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma , 2020, Cell.

[7]  S. H. van der Burg,et al.  Lack of myeloid cell infiltration as an acquired resistance strategy to immunotherapy , 2020, Journal for ImmunoTherapy of Cancer.

[8]  S. H. van der Burg,et al.  A pre‐existing coordinated inflammatory microenvironment is associated with complete response of vulvar high‐grade squamous intraepithelial lesions to different forms of immunotherapy , 2020, International journal of cancer.

[9]  S. H. van der Burg,et al.  Pre-existing inflammatory immune microenvironment predicts the clinical response of vulvar high-grade squamous intraepithelial lesions to therapeutic HPV16 vaccination , 2020, Journal for ImmunoTherapy of Cancer.

[10]  Holger Moch,et al.  The single-cell pathology landscape of breast cancer , 2020, Nature.

[11]  S. H. van der Burg,et al.  Tbet-positive regulatory T cells accumulate in oropharyngeal cancers with ongoing tumor-specific type 1 T cell responses , 2019, Journal of Immunotherapy for Cancer.

[12]  T. Wouters,et al.  Systemic adverse events in imiquimod use for cervical intraepithelial neoplasia – A case series , 2019, Case reports in women's health.

[13]  S. H. van der Burg,et al.  The immune microenvironment in vulvar (pre)cancer: review of literature and implications for immunotherapy , 2018, Expert opinion on biological therapy.

[14]  S. H. van der Burg,et al.  The Anatomical Location Shapes the Immune Infiltrate in Tumors of Same Etiology and Affects Survival , 2018, Clinical Cancer Research.

[15]  N. Bercovici,et al.  Macrophages impede CD8 T cells from reaching tumor cells and limit the efficacy of anti–PD-1 treatment , 2018, Proceedings of the National Academy of Sciences.

[16]  S. H. van der Burg,et al.  Therapeutic Peptide Vaccine-Induced CD8 T Cells Strongly Modulate Intratumoral Macrophages Required for Tumor Regression , 2015, Cancer Immunology Research.

[17]  Xuezheng Sun,et al.  Worldwide incidence of cervical lesions: a systematic review , 2014, Epidemiology and Infection.

[18]  M. Candiani,et al.  Prognostic Significance of Immunohistochemical Phenotypes in Patients Treated for High-Grade Cervical Intraepithelial Neoplasia , 2013, BioMed research international.

[19]  Z. Trajanoski,et al.  Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. , 2013, Immunity.

[20]  S. Kōzuma,et al.  The Prevalence Of Cervical Regulatory T Cells in HPV‐Related Cervical Intraepithelial Neoplasia (CIN) Correlates Inversely with Spontaneous Regression of CIN , 2013, American journal of reproductive immunology.

[21]  J. Baak,et al.  Interaction of epithelial biomarkers, local immune response and condom use in cervical intraepithelial neoplasia 2-3 regression. , 2012, Gynecologic oncology.

[22]  G. Heinze,et al.  Treatment of Cervical Intraepithelial Neoplasia With Topical Imiquimod: A Randomized Controlled Trial , 2012, Obstetrics and gynecology.

[23]  A. Malpica,et al.  The impact of epithelial biomarkers, local immune response and human papillomavirus genotype in the regression of cervical intraepithelial neoplasia grades 2–3 , 2011, Journal of Clinical Pathology.

[24]  R. Clark,et al.  Human Papillomavirus 16-Associated Cervical Intraepithelial Neoplasia in Humans Excludes CD8 T Cells from Dysplastic Epithelium , 2010, The Journal of Immunology.

[25]  M. Stanley,et al.  Characterising the local immune responses in cervical intraepithelial neoplasia: a cross‐sectional and longitudinal analysis , 2008, BJOG : an international journal of obstetrics and gynaecology.

[26]  N. Aaronson,et al.  Treatment of vulvar intraepithelial neoplasia with topical imiquimod. , 2008, The New England journal of medicine.

[27]  K. Syrjänen,et al.  Macrophages, inflammation and risk of cervical intraepithelial neoplasia (CIN) progression--clinicopathological correlation. , 2007, Gynecologic oncology.