Investigative Ophthalmology & Visual Science Cover Image for Volume 60, Issue 6
May 2019
Volume 60, Issue 6
Open Access
Immunology and Microbiology  |   May 2019
PD-L1/PD1 Expression, Composition of Tumor-Associated Immune Infiltrate, and HPV Status in Conjunctival Squamous Cell Carcinoma
Author Affiliations & Notes
  • Priyadharsini Nagarajan
    Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Christian El-Hadad
    Orbital Oncology and Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Stephen K. Gruschkus
    Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Jing Ning
    Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Courtney W. Hudgens
    Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Oded Sagiv
    Orbital Oncology and Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Neil Gross
    Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Michael T. Tetzlaff
    Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
    Department of Translational and Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Bita Esmaeli
    Orbital Oncology and Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
  • Correspondence: Bita Esmaeli, Orbital Oncology and Ophthalmic Plastic Surgery, Department of Plastic Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1488, Houston, TX 77030, USA; [email protected]
Investigative Ophthalmology & Visual Science May 2019, Vol.60, 2388-2398. doi:https://doi.org/10.1167/iovs.19-26894
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Priyadharsini Nagarajan, Christian El-Hadad, Stephen K. Gruschkus, Jing Ning, Courtney W. Hudgens, Oded Sagiv, Neil Gross, Michael T. Tetzlaff, Bita Esmaeli; PD-L1/PD1 Expression, Composition of Tumor-Associated Immune Infiltrate, and HPV Status in Conjunctival Squamous Cell Carcinoma. Invest. Ophthalmol. Vis. Sci. 2019;60(6):2388-2398. https://doi.org/10.1167/iovs.19-26894.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

Purpose: Conjunctival squamous cell carcinoma (SCC), a type of ocular surface neoplasia, is primarily treated by surgical resection and topical immuno- or chemotherapy. Metastatic disease may be treated with systemic chemo- or immunotherapy, albeit with variable response. The purpose of this study was to determine whether immune checkpoint blockade might be considered in the management of conjunctival SCC.

Methods: In this retrospective study, we evaluated tumor programmed death-ligand 1 (PD-L1) expression, high-risk human papillomavirus (HPV) status, and immunohistochemical expression of cluster of differentiation 3 (CD3), cluster of differentiation 8 (CD8), and programmed death 1 (PD1) in tumor-associated immune infiltrate in a series of 31 conjunctival SCCs.

Results: PD-L1 expression in ≥1% of tumor cells was noted in 14 conjunctival SCCs (47%) and was more prevalent in invasive than in situ SCC and among tumors with higher American Joint Committee on Cancer (AJCC) T category (≥T3 versus ≤T2). The density of CD3-positive T cells was higher in primary than recurrent tumors and higher in invasive than in situ tumors. Density of CD3-positive and CD8-positive T cells was higher in higher AJCC stage tumors. Density of CD8-positive T cells was higher in HPV-positive than HPV-negative tumors. PD-L1 expression correlated with a higher density of CD3-, CD8-, and PD1-positive cells in the tumor-associated immune infiltrate but not with HPV status.

Conclusions: Our findings demonstrate that PD-L1 is expressed in almost half of conjunctival SCCs. The density of tumor-associated immune cells correlated with invasive SCC, stage, and HPV status in conjunctival SCC. Our findings support further studies to establish the potential application of immune checkpoint blockade in the management of conjunctival SCC.

Ocular surface squamous neoplasia encompasses a spectrum of disease that includes conjunctival premalignant dysplasia, carcinoma in situ, and invasive conjunctival squamous cell carcinoma (SCC).1 The annual incidence of ocular surface squamous neoplasia ranges from 0.02 to 3.5 cases per 1 million population depending on the geographic location and its attendant degree of ultraviolet B light exposure.28 Incidence is higher close to the equator and decreases with increasing latitude.9 Additional predisposing factors include smoking; human papillomavirus (HPV) infection; immunosuppression, including human immunodeficiency virus (HIV)/AIDS, hematolymphoid malignancies, and transplant; chronic inflammatory states such as allergic conjunctivitis and cicatricial pemphigoid; male sex; and Fitzpatrick type I and II skin.7,1016 
Surgery and adjuvant cryotherapy have historically been considered standard for local management of conjunctival SCC; however, topical and/or intralesional chemotherapy and immunotherapy with interferon have recently gained popularity.1724 In the United States, the reported 1-year local recurrence rates after definitive therapy with any of the above-mentioned modalities range from 3% to 10%,25,26 although the rates are much higher among immunocompromised patients.27 The rate of regional lymph node metastasis is 0% to 16%,27 but systemic dissemination is rare.24,28,29 Patients with metastatic disease may require systemic therapy with cytotoxic agents and/or EGFR inhibitors and radiation therapy.24,30,31 Patients with locally advanced and/or recurrent conjunctival SCC with orbital invasion may require orbital exenteration, a radical and debilitating surgery. Invasive conjunctival SCC with orbital invasion is endemic in parts of Africa and leads to significant morbidity.12,32,33 Thus, new treatments are needed to effectively prevent recurrence and preserve ocular function. 
Conjunctival SCC shares several features with cutaneous SCC, including demographic characteristics (predominance in males and individuals with Fitzpatrick type I and II skin), etiopathogenic factors (high cumulative ultraviolet B light exposure, immunosuppression, and chronic inflammatory states), management strategies (surgical resection, cryo-, photodynamic, topical, systemic, and radiation therapies), and genomic alterations (gains in 3q22.3–3q28 and 5p and losses in 9p, 13q, 17p, and 18q).34 Moreover, the presence of primary cutaneous SCC or basal cell carcinoma is correlated with increased risk for developing conjunctival SCC.8 
Management of advanced cutaneous SCC has not been standardized; however, since the advent of immune checkpoint therapy, studies have evaluated the role of tumor-infiltrating lymphocytes35,36 and programmed death-ligand 1 (PD-L1) expression in tumor cells in cutaneous SCC37,38 and have established the efficacy of programmed death 1 (PD1) blockade in the management of advanced cutaneous SCC.39,40 To explore whether use of PD1/PD-L1-based immunotherapy might also be feasible in conjunctival SCC, we evaluated, in a series of 31 conjunctival SCCs, the expression of PD-L1 within tumor cells; the composition and density of tumor-associated immune infiltrates; relationships between PD-L1 expression, immune infiltrates, and clinicopathologic features, including HPV status; and relationships between various patient and disease features and prognosis. 
Methods
Selection of Cases and Collation of Clinicopathologic Data
This study was approved by the Institutional Review Board of The University of Texas MD Anderson Cancer Center and adhered to the tenets of the Declaration of Helsinki. We identified 42 patients treated by the ophthalmic plastic surgeon (BE), between July 2009 and April 2017, of which, we were able to procure tissue blocks with residual tumor from the archived surgical specimens for 31 patients. For each patient, we collected demographic features (age at initial diagnosis and at presentation to our institution, sex, and ethnicity), risk factors for conjunctival SCC (cutaneous SCC, hematolymphoid malignancy, HIV positivity, and transplant), primary tumor features (laterality, anatomic site, disease status at presentation [primary or recurrent], in situ versus invasive disease, and size/diameter of invasive carcinoma), extraconjunctival extension, American Joint Committee on Cancer (AJCC) stage (Supplementary Table S1), local recurrence (anatomic site and date), metastases (regional or distant and date), and vital status at last follow-up and cause of death. Types of surgery and adjuvant therapy were also recorded. 
Immunohistochemistry and In Situ Hybridization
Immunohistochemical studies were performed on 4-μm-thick tissue sections using a Leica Bond III autostainer, using the following antibodies: cluster of differentiation 3 (CD3) (catalog no. A0452; dilutions 1:100; Dako, Carpinteria, CA, USA), cluster of differentiation 8 (CD8) (catalog no. MS457s; dilutions 1:25; Life Sciences Technology, Waltham, MA, USA), PD1 (catalog no. ab137132; dilutions 1:250; Abcam, Burlingame, CA, USA), and PD-L1 (catalog no. 13684S; dilutions 1:100; Cell Signaling Technology, Danvers, MA, USA). RNA in situ hybridization (ISH) to identify high-risk HPV (types 16, 18, 31, 33, 35, 45, 52, and 58) was performed using the RNAscope 2.5 LS assay. 
Image Analysis
Slides immunostained for PD-L1 or subjected to HPV ISH were evaluated in a blinded manner by two pathologists (MTT and PN). PD-L1 expression was visually estimated as the percentage of tumor cells that displayed complete or partial membranous staining. Tumors with PD-L1 staining in ≥1% of tumor cells were considered PD-L1 positive, on the basis of previous studies of cutaneous SCC.37,38,41 Of the 31 cases evaluated, 1 did not contain tumor cells for analysis on the PD-L1-immunostained slide. HPV status was determined by visual evaluation for the presence of punctate nuclear signals within tumor nuclei at 100× magnification and was scored as positive or negative. 
Automated image analysis for quantification of lymphocytes expressing CD3, CD8, and PD1 was performed as described previously.42 Briefly, the immunostained slides were scanned at 200× magnification by using the Aperio Scanscope AT Turbo instrument (Leica Biosystems, Buffalo Grove, IL, USA). Using the Aperio ImageScope image analysis software, we used 3 to 5 squares, each measuring 0.25 mm2, to designate regions of interest (ROI) for analysis. First, a square was drawn in the region of tumor-stroma interface with the highest density of immune infiltrate, and then 2 to 4 additional squares (depending on the size of tumor available for evaluation) were drawn in peritumoral and intratumoral regions with progressively decreasing density of immune infiltrate. To ensure optimal image analysis of tumor-associated immune infiltrate, the ROIs were first selected on CD3-immunostained slides, and then the corresponding ROIs were designated on CD8- and PD1-immunostained slides. Positive cells were counted within each square, tabulated either as raw number or as the percentage of the nucleated cells in the designated regions, and reported as overall (average of all designated squares) and “hotspot” (highest-density square) per mm2 for each sample. 
Statistical Analysis
The clinical and histopathologic characteristics were summarized using descriptive statistics. Correlations between immunohistochemical, demographic, and clinicopathologic factors were assessed using the Wilcoxon rank sum, Fisher's exact, and Pearson's correlation tests. Recurrence-free survival (RFS) was defined as the time from surgery to disease recurrence or death from any cause, whichever occurred first. Metastasis-free survival (MFS) was defined as the time from surgery to diagnosis of metastasis or death from any cause, whichever occurred first. Disease-free survival (DFS) was defined as the time from surgery to death. Patients not experiencing the event of interest were censored at their date of last follow-up for each survival outcome, and the survival curves were estimated by the Kaplan-Meier method.43 Cox regression modeling using Firth's penalized likelihood approach was used to evaluate correlations between demographic, clinicopathologic, and immunohistochemical features and survival outcomes. All statistical tests used a significance level of 5% and were performed using SAS 9.3 for Windows (SAS Institute Inc., Cary, NC, USA). 
Results
Demographic and clinicopathologic characteristics of the cohort are summarized in Table 1. Twenty-eight of the 31 patients in our cohort (90%) were white; and the cohort included 16 men and 15 women, with a mean age at presentation of 61.3 years. Six patients had at least one risk factor for conjunctival SCC, and cutaneous SCC was the most common. The left eye was involved in 23 patients (74%), and 17 patients (54%) had contiguous involvement of multiple parts of the ocular surface epithelium. Seven patients (23%) presented with recurrent disease. Twenty-three patients (74%) had invasive SCC and eight (26%) had in situ tumor. There was a slight predominance of AJCC T category T3 or higher tumors (n = 17; 55%) (Supplementary Table S1). Orbital exenteration was necessary for local disease control in 9 patients (29%), and 18 patients (58%) underwent adjuvant therapy, most commonly topical chemotherapy (11 patients; 36%). Five patients (16%) experienced local recurrence after curative surgery, and two patients developed regional metastases. Of this group, one patient refused treatment and died with disease 13.3 months after diagnosis of regional metastases; the other patient was alive without disease at 31.9 months after diagnosis of regional metastases. Four patients (13%) died, and one of which was due to conjunctival SCC (described above). Twenty-eight patients (90%) were disease free at their last follow-up. 
Table 1
 
Demographic, Clinicopathologic, and Immunohistochemical Parameters in Patients With Conjunctival SCC and Correlation With Tumor HPV Status and Proportion of Tumor Cells Expressing PD-L1
Table 1
 
Demographic, Clinicopathologic, and Immunohistochemical Parameters in Patients With Conjunctival SCC and Correlation With Tumor HPV Status and Proportion of Tumor Cells Expressing PD-L1
The majority of the conjunctival SCCs in our cohort were HPV negative (n = 23; 74%) (Table 1). Most of the HPV-negative primary tumors were AJCC T category ≤T2 (13/23, 57%), whereas most of the HPV-positive primary tumors were AJCC ≥T3 (7/8, 88%; P < 0.05) (Fig. 1A). The HPV-positive tumors exhibited a higher incidence of extraconjunctival extension than the HPV-negative tumors did (50% vs. 13%, P = 0.05) (Fig. 1B). 
Figure 1
 
Correlation between HPV status and (A) AJCC T category and (B) extraconjunctival extension status in conjunctival SCC.
Figure 1
 
Correlation between HPV status and (A) AJCC T category and (B) extraconjunctival extension status in conjunctival SCC.
Fourteen of 30 tumors (47%) were PD-L1 positive (staining in ≥1% of tumor cells); of which in 5 tumors, more than 10% of tumor cells expressed PD-L1 (Table 1). PD-L1 positivity was more prevalent among invasive SCCs (n = 13 of 22, 59%) than among in situ tumors (n = 1 of 8, 13%) (P = 0.04) (Figs. 2A–E) and more prevalent among tumors with higher (≥T3) AJCC T category (11 of 14, 79%, including the 2 tumors that gave rise to regional nodal metastasis) than among tumors with lower (≤T2) AJCC T category (5 of 16, 31%) (P = 0.01) (Fig. 2F). No correlation was noted between HPV status and tumor PD-L1 expression, irrespective of the percentage of tumor cells expressing PD-L1 (≥1% vs. 1%–10% and ≥10%) (Table 2; Fig. 3). 
Figure 2
 
PD-L1 expression in conjunctival SCC and correlation between PD-L1 expression and tumor type and AJCC T category of conjunctival SCC. (AD) Representative micrographs showing PD-L1 expression in (A, B) in situ and (C, D) invasive conjunctival SCCs with (A, C) <1% and (B, D) ≥1% of tumor cells staining positive for PD-L1 (magnification ×200). Inset: corresponding field in hematoxylin-eosin-stained section (magnification ×200). (E, F) Correlation between PD-L1 staining and (E) tumor type and (F) AJCC T category. PD-L1 status not available in one case because of inadequate tumor for analysis.
Figure 2
 
PD-L1 expression in conjunctival SCC and correlation between PD-L1 expression and tumor type and AJCC T category of conjunctival SCC. (AD) Representative micrographs showing PD-L1 expression in (A, B) in situ and (C, D) invasive conjunctival SCCs with (A, C) <1% and (B, D) ≥1% of tumor cells staining positive for PD-L1 (magnification ×200). Inset: corresponding field in hematoxylin-eosin-stained section (magnification ×200). (E, F) Correlation between PD-L1 staining and (E) tumor type and (F) AJCC T category. PD-L1 status not available in one case because of inadequate tumor for analysis.
Table 2
 
Correlation Between Tumor HPV Status and PD-L1 Expression
Table 2
 
Correlation Between Tumor HPV Status and PD-L1 Expression
Figure 3
 
Expression of immune markers in primary conjunctival squamous cell carcinoma, with respect to HPV status: negative (AL) versus positive (MX) and PD-L1 expression in <1% (AF, MR) vs. ≥1% (GL, SX) of tumor cells. High-risk HPV: A, G, M, S (400× magnification); B, H, N, T (100× magnification); PD-L1: C, I, O, U (100× magnification); CD3: D, J, P, V (100× magnification); CD8: E, K, Q, W (100× magnification); PD1: F, L, R, X (100× magnification).
Figure 3
 
Expression of immune markers in primary conjunctival squamous cell carcinoma, with respect to HPV status: negative (AL) versus positive (MX) and PD-L1 expression in <1% (AF, MR) vs. ≥1% (GL, SX) of tumor cells. High-risk HPV: A, G, M, S (400× magnification); B, H, N, T (100× magnification); PD-L1: C, I, O, U (100× magnification); CD3: D, J, P, V (100× magnification); CD8: E, K, Q, W (100× magnification); PD1: F, L, R, X (100× magnification).
When the spatial distribution of CD3-, CD8- and PD1-positive immune infiltrate in and around the tumors were evaluated, we did not identify any obvious differences with respect to the HPV status or the pattern/percentage of PD-L1 expression in the primary (Fig. 3) or metastatic (Supplementary Fig. S1) tumors. However, the following correlations were observed between primary tumor features and tumor-associated immune infiltrate (Table 3; Supplementary Table S2; Fig. 4): (1) patients with recurrent tumors had lower hotspot CD3 expression than patients presenting with primary tumors (P = 0.04); (2) patients with invasive SCC had higher overall CD3 expression than patients with in situ tumors (P = 0.03); (3) AJCC T category ≥T3 was correlated with higher CD3 expression (overall, P = 0.03; hotspot, P = 0.05) and overall CD8 expression (P = 0.03), compared to T category ≤T2; (4) HPV-positive tumors had higher overall CD8 expression than HPV-negative tumors (P = 0.04); and (5) PD-L1 expression in ≥1% of tumor cells correlated with higher CD3 (overall, P = 0.0001; hotspot, P = 0.0003), CD8 (overall, P = 0.0002; hotspot, P = 0.001), and PD1 (overall, P < 0.0001; hotspot, P = 0.0002) expression in tumor-associated immune infiltrate. 
Table 3
 
Correlation Between Clinicopathologic Characteristics, HPV and PD-L1 Status, and Immune Infiltrates
Table 3
 
Correlation Between Clinicopathologic Characteristics, HPV and PD-L1 Status, and Immune Infiltrates
Figure 4
 
Relationships between immune infiltrate status and proportion of tumor cells expressing PD-L1 in conjunctival SCC. Shown are distributions of percentages of (A) CD3-positive, (B) CD8-positive, and (C) PD1-positive cells in the tumor-associated immune infiltrate according to PD-L1 expression.
Figure 4
 
Relationships between immune infiltrate status and proportion of tumor cells expressing PD-L1 in conjunctival SCC. Shown are distributions of percentages of (A) CD3-positive, (B) CD8-positive, and (C) PD1-positive cells in the tumor-associated immune infiltrate according to PD-L1 expression.
Univariate Cox regression and Kaplan-Meier analyses did not reveal significant correlations between patient, tumor, or immune features and RFS, MFS, or DFS (Supplementary Table S2). However, the correlation between hotspot CD3 expression and DFS approached significance (P = 0.09) by univariate analysis. Also, MFS and DFS appeared to be slightly lower in patients with PD-L1-positive SCCs than PD-L1-negative tumors, although this was not statistically significant. 
Discussion
To the best of our knowledge, this is one of the first studies to survey the prevalence of PD-L1 expression in conjunctival SCC and to evaluate the prognostic significance of tumor cell expression of PD-L1 and the density and composition of the tumor-associated immune infiltrates in conjunctival SCC. We and others have previously evaluated the expression of these markers in the context of response to immune checkpoint therapy in other malignancies, such as cutaneous malignancies such as Merkel cell carcinoma42 and sebaceous carcinoma.44 Many studies evaluating the PD-L1/PD1 axis often use only PD-L1 and PD145,46 or PD-L1 alone.47 We chose to include CD3 and CD8 as well because CD3- and CD8-positive immune infiltrate density correlated with outcome in cutaneous malignancies such as Merkel cell carcinoma.42 Inclusion of CD4 or FoxP3 did not proffer additional advantage because there was no significant association with clinicopathologic parameters or outcome.44,48 Therefore, we chose to evaluate the expression of only CD3, CD8, PD-L1, and PD1 in this exploratory study. This small panel of immune markers was also advantageous because, in some cases, only limited amount of tumor was available for evaluation. 
In this study, we found that the tumor cells in almost half of conjunctival SCCs (47%) expressed PD-L1. The frequency of PD-L1 positivity in tumor cells was higher among invasive tumors and in tumors with higher AJCC T category (≥T3) and increased with increasing density of CD3-positive, CD8-positive, and PD1-positive cells in the tumor-associated immune infiltrate. Furthermore, we found that the density of CD3-positive cells in the inflammatory infiltrate was lower in patients presenting with recurrent tumors and the relative proportion of CD8-positive T lymphocytes was higher among HPV-positive tumors. PD-L1 expression did not correlate with outcome in our cohort, although there appeared to be a trend toward slightly lower MFS and DFS in patients with PD-L1-positive tumors. 
We did not observe an independent association between the presence of risk factors such as cutaneous SCC or immunosuppression (Table 1; Supplementary Table S2) and expression of immune markers in our cohort. However, when immunosuppression was considered separately, the majority of patients with immunosuppression had lower PD-L1 expression, as well as lower overall numbers of immune cells positive for CD3, CD8, and PD1 (Supplementary Table S3). This finding, although statistically insignificant, suggests that the composition and density of tumor-associated immune infiltrate are likely influenced by multiple elements, which include systemic factors such as the patient's general immune competence, HIV status, and age-related immune senescence, as well as local factors such as HPV infection, chronic inflammatory diseases and ultraviolet light-induced immune dysregulation.11,49,50 Therefore, studies with larger cohorts will be needed to evaluate if and how systemic and/or local immune dysregulation may affect the expression of immune markers and whether they may be modified to enhance antitumor effects against conjunctival SCC. 
Our demonstration of PD-L1 expression in almost half of conjunctival SCC tumor samples is similar to the reported frequencies of PD-L1 expression in primary oropharyngeal mucosal SCC of head and neck (39.2%–87%)48,5153 and cutaneous SCC (25%–65%).38,5456 Our finding that the frequency of PD-L1 positivity in neoplastic cells increased with increasing conjunctival SCC AJCC T category is also similar to trends previously noted in cutaneous SCC. In a study of 45 cases of primary cutaneous SCC, the frequency of PD-L1 expression was reported to be higher in tumors with high-risk features for metastasis (50%–70%) than in low-risk tumors (20%–26%).37,38 The reported frequency of PD-L1 expression in samples of metastases from patients with cutaneous SCC ranges from 39.5% to 100%.37,38,52 In a study of 83 patients with head and neck cutaneous SCC, Amoils et al.56 reported that the percentage of tumor cells positive for PD-L1 was higher in metastatic tumor samples than in primary tumor samples. 
The demonstration of PD-L1 expression in conjunctival SCC raises the possibility of applying immune checkpoint blockade with agents targeting the PD1/PD-L1 axis—particularly among patients with locally advanced conjunctival SCC or those with orbital invasion, who are currently treated with radical disfiguring and disabling surgery, and/or among patients with metastatic disease, who tend to die of disease as there are currently few effective treatments.24 Recent studies have demonstrated that inhibition of the PD1/PD-L1 axis is often effective in the management of advanced cutaneous SCC.39 Similar, but inconsistent trends have also been noted in SCCs of other mucosal and cutaneous origins, and the inhibition of PD1/PD-L1 axis has been successful in the management of these malignancies.39,5760 
Immunohistochemical expression of PD-L1 on tumor cells and/or tumor-infiltrating immune cells has been shown to correlate with favorable response to PD1/PD-L1 inhibitor therapy in several malignancies, including melanoma61 and nonsquamous non-small-cell lung carcinoma.62 However, in other cancers, such as renal cell carcinoma and pulmonary SCCs, response to inhibition of the PD1/PD-L1 axis did not appear to correlate with PD-L1 expression on neoplastic cells.6365 Thus, in the majority of the previously evaluated malignancies, PD-L1 expression on tumor cells may be predictive of response to PD1/PD-L1 inhibitor therapy, particularly when supplemented by characteristics of immune infiltrate and other previously established biomarkers specific to that cancer as well as other tumor-related factors, such as tumor mutational burden.66,67 However, PD-L1 expression status may not be independently predictive of response to PD1/PD-L1 inhibitor therapy.68 For example, in the CheckMate 141 trial, patients with advanced, recurrent, or metastatic mucosal SCC of oral, oropharyngeal, hypopharyngeal, or laryngeal origin (that had progressed within 6 months from the last dose of platinum-based chemotherapy) were randomized to receive nivolumab monotherapy or investigator's choice single-agent chemotherapeutic agent.69 Nivolumab therapy increased overall survival (OS) at 18, 24, and 30 months, irrespective of PD-L1 expression (<1% vs. ≥1%) and HPV status.70 
Our finding that PD-L1 expression correlated with increased density of CD3-positive, CD8-positive, and PD1-positive cells in the tumor-associated immune infiltrate is similar to previously reported findings in melanoma71 and head and neck cutaneous SCC.52 Although it is unusual that HPV-negative tumors predominated in our cohort, the relative proportion of CD8-positive T lymphocytes was higher among HPV-positive conjunctival SCC. This may be secondary to T-cell activation in response to viral antigens. There was no correlation between HPV status and PD-L1 expression in our cohort. Similar results have been noted in oropharyngeal SCC, where majority of the tumors expressed PD-L1, irrespective of HPV status.72 Therefore, it remains to be seen if PD-L1 expression may correlate with response to immunotherapy in conjunctival SCC. 
Although we did not find any correlation between PD-L1 expression and tumor recurrence, the density of CD3-positive T cells was significantly lower in patients presenting with recurrent tumors in our cohort. Whether this reflects effects of altered microenvironment and/or evasion of immune surveillance is unclear. Kamiya et al.73 found that a higher intensity of PD-L1 staining, but not the proportion of tumor cells with PD-L1 staining, correlated with nodal metastasis in cutaneous SCC. In contrast, Garcia-Pedrero et al.74 found that PD-L1 expression in more than 25% of tumor cells correlated with lymph node metastasis in head and neck cutaneous SCC. 
Several studies in other types of cutaneous and mucosal SCC have documented conflicting correlations between PD-L1 expression and clinical outcome. In head and neck cutaneous SCC, PD-L1 expression was reported to correlate with longer DFS.52 In oral mucosal SCC, one study suggested that PD-L1 expression may be an indicator of poor outcome,75 but a recent meta-analysis did not support this notion in oral SCC.76 In an analysis of 133 cases of oropharyngeal SCC, PD-L1 expression in tumor cells did not correlate with OS, irrespective of HPV status.72 Similarly, there was no clear association between PD-L1 positivity in tumor cells and OS in HPV-positive oropharyngeal carcinomas, whereas higher density of CD8-positive tumor-infiltrating lymphocytes correlated with improved OS.7779 On the other hand, PD-L1 expression correlated with increased risk for regional lymph node metastasis among head and neck mucosal SCCs, including those of oropharyngeal origin.80 
There are important limitations to the current study. There might be a bias for the referral of patients with high-risk and/or recurrent conjunctival SCC to our institution. Of these, patients were selected on the basis of availability of tumor samples for further analysis. Patients lacking adequate tumor samples and patients treated exclusively with topical or intralesional chemotherapy (who had no surgical specimen available for analysis) were excluded. In view of this selection bias, it is not surprising that PD-L1 expression did not predict clinical outcomes in our cohort. Some of the specimens included in our study were subjected to intraoperative frozen section evaluation; it is unclear if and how this might have affected the detection of PD-L1 expression. The percentage of CD3-positive, CD8-positive, and PD1-positive cells compared to all nucleated cells in the ROIs may have been underestimated in large tumors. Additionally, the size of our study cohort was small (n = 31) and there was a predominance of high-risk HPV-negative tumors in the cohort. Also, the length of follow-up (less than 2 years in some patients) may not be sufficient for long-term outcome analyses. Therefore, additional studies will be needed to corroborate our findings. Nevertheless, this initial survey of conjunctival SCC reveals that PD-L1 is expressed in almost half of tumors and correlates with increased density of CD3-positive, CD8-positive, and PD1-positive immune infiltrate, justifying the possible initiation of clinical trials to determine the efficacy of PD1/PD-L1 blockade in patients with locally advanced, recurrent, or metastatic conjunctival SCC. 
Acknowledgments
Supported by the National Cancer Institute under award number P30CA016672, which supports the MD Anderson Cancer Center Clinical Trials Support Resource. 
Disclosure: P. Nagarajan, None; C. El-Hadad, None; S.K. Gruschkus, None; J. Ning, None; C.W. Hudgens, None; O. Sagiv, None; N. Gross, None; M.T. Tetzlaff, None; B. Esmaeli, None 
References
Lee GA, Hirst LW. Ocular surface squamous neoplasia. Surv Ophthalmol. 1995; 39: 429–450.
Alves LF, Fernandes BF, Burnier JV, Zoroquiain P, Eskenazi DT, Burnier MNJr. Incidence of epithelial lesions of the conjunctiva in a review of 12,102 specimens in Canada (Quebec). Arq Bras Oftalmol. 2011; 74: 21–23.
Gichuhi S, Sagoo MS, Weiss HA, Burton MJ. Epidemiology of ocular surface squamous neoplasia in Africa. Trop Med Int Health. 2013; 18: 1424–1443.
Sun EC, Fears TR, Goedert JJ. Epidemiology of squamous cell conjunctival cancer. Cancer Epidemiol Biomarkers Prev. 1997; 6: 73–77.
Yang J, Foster CS. Squamous cell carcinoma of the conjunctiva. Int Ophthalmol Clin. 1997; 37: 73–85.
Emmanuel B, Ruder E, Lin SW, Abnet C, Hollenbeck A, Mbulaiteye S. Incidence of squamous-cell carcinoma of the conjunctiva and other eye cancers in the NIH-AARP Diet and Health Study. Ecancermedicalscience. 2012; 6: 254.
Kiire CA, Stewart RMK, Srinivasan S, Heimann H, Kaye SB, Dhillon B. A prospective study of the incidence, associations and outcomes of ocular surface squamous neoplasia in the United Kingdom. Eye (Lond). 2018; 33: 283–294.
McClellan AJ, McClellan AL, Pezon CF, Karp CL, Feuer W, Galor A. Epidemiology of ocular surface squamous neoplasia in a veterans affairs population. Cornea. 2013; 32: 1354–1358.
Newton R, Ferlay J, Reeves G, Beral V, Parkin DM. Effect of ambient solar ultraviolet radiation on incidence of squamous-cell carcinoma of the eye. Lancet. 1996; 347: 1450–1451.
Gichuhi S, Macharia E, Kabiru J, et al. Risk factors for ocular surface squamous neoplasia in Kenya: a case-control study. Trop Med Int Health. 2016; 21: 1522–1530.
Gichuhi S, Ohnuma S, Sagoo MS, Burton MJ. Pathophysiology of ocular surface squamous neoplasia. Exp Eye Res. 2014; 129: 172–182.
Guech-Ongey M, Engels EA, Goedert JJ, Biggar RJ, Mbulaiteye SM. Elevated risk for squamous cell carcinoma of the conjunctiva among adults with AIDS in the United States. Int J Cancer. 2008; 122: 2590–2593.
Kamal S, Kaliki S, Mishra DK, Batra J, Naik MN. Ocular surface squamous neoplasia in 200 patients: a case-control study of immunosuppression resulting from human immunodeficiency virus versus immunocompetency. Ophthalmology. 2015; 122: 1688–1694.
Kao AA, Galor A, Karp CL, Abdelaziz A, Feuer WJ, Dubovy SR. Clinicopathologic correlation of ocular surface squamous neoplasms at Bascom Palmer Eye Institute: 2001 to 2010. Ophthalmology. 2012; 119: 1773–1776.
Kheir WJ, Tetzlaff MT, Pfeiffer ML, et al. Epithelial, non-melanocytic and melanocytic proliferations of the ocular surface. Semin Diagn Pathol. 2016; 33: 122–132.
Shields CL, Chien JL, Surakiatchanukul T, Sioufi K, Lally SE, Shields JA. Conjunctival tumors: review of clinical features, risks, biomarkers, and outcomes—the 2017 J. Donald M. Gass Lecture. Asia Pac J Ophthalmol (Phila). 2017; 6: 109–120.
Cicinelli MV, Marchese A, Bandello F, Modorati G. Clinical management of ocular surface squamous neoplasia: a review of the current evidence. Ophthalmol Ther. 2018; 7: 247–262.
Santoni A, Thariat J, Maschi C, et al. Management of invasive squamous cell carcinomas of the conjunctiva Treatment of invasive conjunctival carcinoma. Am J Ophthalmol. 2019; 200: 1–9.
Galor A, Karp CL, Chhabra S, Barnes S, Alfonso EC. Topical interferon alpha 2b eye-drops for treatment of ocular surface squamous neoplasia: a dose comparison study. Br J Ophthalmol. 2010; 94: 551–554.
Karp CL, Galor A, Chhabra S, Barnes SD, Alfonso EC. Subconjunctival/perilesional recombinant interferon alpha2b for ocular surface squamous neoplasia: a 10-year review. Ophthalmology. 2010; 117: 2241–2246.
Mercado CL, Pole C, Wong J, et al. Surgical versus medical treatment for ocular surface squamous neoplasia: a quality of life comparison. Ocul Surf. 2019; 17: 60–63.
Sepulveda R, Pe'er J, Midena E, Seregard S, Dua HS, Singh AD. Topical chemotherapy for ocular surface squamous neoplasia: current status. Br J Ophthalmol. 2010; 94: 532–535.
Siedlecki AN, Tapp S, Tosteson AN, et al. Surgery versus interferon alpha-2b treatment strategies for ocular surface squamous neoplasia: a literature-based decision analysis. Cornea. 2016; 35: 613–618.
Yin VT, Merritt HA, Sniegowski M, Esmaeli B. Eyelid and ocular surface carcinoma: diagnosis and management. Clin Dermatol. 2015; 33: 159–169.
Galor A, Karp CL, Oellers P, et al. Predictors of ocular surface squamous neoplasia recurrence after excisional surgery. Ophthalmology. 2012; 119: 1974–1981.
Nanji AA, Moon CS, Galor A, Sein J, Oellers P, Karp CL. Surgical versus medical treatment of ocular surface squamous neoplasia: a comparison of recurrences and complications. Ophthalmology. 2014; 121: 994–1000.
Chauhan S, Sen S, Sharma A, et al. American Joint Committee on Cancer Staging and clinicopathological high-risk predictors of ocular surface squamous neoplasia: a study from a tertiary eye center in India. Arch Pathol Lab Med. 2014; 138: 1488–1494.
Erie JC, Campbell RJ, Liesegang TJ. Conjunctival and corneal intraepithelial and invasive neoplasia. Ophthalmology. 1986; 93: 176–183.
Rajeh A, Barakat F, Khurma S, et al. Characteristics, management, and outcome of squamous carcinoma of the conjunctiva in a single tertiary cancer center in Jordan. Int J Ophthalmol. 2018; 11: 1132–1138.
Desai SJ, Pruzan NL, Geske MJ, Jeng BH, Bloomer MM, Vagefi MR. Local and regional spread of primary conjunctival squamous cell carcinoma. Eye Contact Lens. 2018; 44: S312–S315.
Nair AG, Kaliki S, Mishra DK, Reddy VA, Naik MN. Neoadjuvant chemotherapy for invasive squamous cell carcinoma of the conjunctiva: a case report. Indian J Ophthalmol. 2015; 63: 927–929.
Reynolds JW, Pfeiffer ML, Ozgur O, Esmaeli B. Prevalence and severity of ocular surface neoplasia in African nations and need for early interventions. J Ophthalmic Vis Res. 2016; 11: 415–421.
Tornesello ML, Duraturo ML, Waddell KM, et al. Evaluating the role of human papillomaviruses in conjunctival neoplasia. Br J Cancer. 2006; 94: 446–449.
Asnaghi L, Alkatan H, Mahale A, et al. Identification of multiple DNA copy number alterations including frequent 8p11.22 amplification in conjunctival squamous cell carcinoma. Invest Ophthalmol Vis Sci. 2014; 55: 8604–8613.
Bauer C, Abdul Pari AA, Umansky V, et al. T-lymphocyte profiles differ between keratoacanthomas and invasive squamous cell carcinomas of the human skin. Cancer Immunol Immunother. 2018; 67: 1147–1157.
Linedale R, Schmidt C, King BT, et al. Elevated frequencies of CD8 T cells expressing PD-1, CTLA-4 and Tim-3 within tumour from perineural squamous cell carcinoma patients. PLoS One. 2017; 12: e0175755.
Garcia-Diez I, Hernandez-Ruiz E, Andrades E, et al. PD-L1 Expression is increased in metastasizing squamous cell carcinomas and their metastases. Am J Dermatopathol. 2018; 40: 647–654.
Slater NA, Googe PB. PD-L1 expression in cutaneous squamous cell carcinoma correlates with risk of metastasis. J Cutan Pathol. 2016; 43: 663–670.
Migden MR, Rischin D, Schmults CD, et al. PD-1 blockade with cemiplimab in advanced cutaneous squamous-cell carcinoma. N Engl J Med. 2018; 379: 341–351.
Stevenson ML, Wang CQ, Abikhair M, et al. Expression of programmed cell death ligand in cutaneous squamous cell carcinoma and treatment of locally advanced disease with pembrolizumab. JAMA Dermatol. 2017; 153: 299–303.
Schaper K, Kother B, Hesse K, Satzger I, Gutzmer R. The pattern and clinicopathological correlates of programmed death-ligand 1 expression in cutaneous squamous cell carcinoma. Br J Dermatol. 2017; 176: 1354–1356.
Feldmeyer L, Hudgens CW, Ray-Lyons G, et al. Density, distribution, and composition of immune infiltrates correlate with survival in Merkel cell carcinoma. Clin Cancer Res. 2016; 22: 5553–5563.
Kaplan E, Meier P. Nonparametric estimator from incomplete observations. J Am Stat Assoc. 1958; 53: 457–481.
Kandl TJ, Sagiv O, Curry JL, et al. High expression of PD-1 and PD-L1 in ocular adnexal sebaceous carcinoma. Oncoimmunology. 2018; 7: e1475874.
Thangarajah F, Morgenstern B, Pahmeyer C, et al. Clinical impact of PD-L1 and PD-1 expression in squamous cell cancer of the vulva [published online ahead of print April 10, 2019]. J Cancer Res Clin Oncol. doi:10.1007/s00432-019-02915-1.
Naruse T, Yanamoto S, Okuyama K, et al. Immunohistochemical study of PD-1/PD-L1 axis expression in oral tongue squamous cell carcinomas: effect of neoadjuvant chemotherapy on local recurrence [published online ahead of print February 14, 2019]. Pathol Oncol Res. doi:10.1007/s12253-019-00606-3.
Miyazawa T, Marushima H, Saji H, et al. PD-L1 expression in non-small-cell lung cancer including various adenocarcinoma subtypes. Ann Thorac Cardiovasc Surg. 2019; 25: 1–9.
Cho YA, Yoon HJ, Lee JI, Hong SP, Hong SD. Relationship between the expressions of PD-L1 and tumor-infiltrating lymphocytes in oral squamous cell carcinoma. Oral Oncol. 2011; 47: 1148–1153.
Yamazaki S, Odanaka M, Nishioka A, et al. Ultraviolet B-induced maturation of CD11b-type langerin(-) dendritic cells controls the expansion of Foxp3(+) regulatory T cells in the skin. J Immunol. 2018; 200: 119–129.
Hart PH, Norval M. Ultraviolet radiation-induced immunosuppression and its relevance for skin carcinogenesis. Photochem Photobiol Sci. 2018; 17: 1872–1884.
Malm IJ, Bruno TC, Fu J, et al. Expression profile and in vitro blockade of programmed death-1 in human papillomavirus-negative head and neck squamous cell carcinoma. Head Neck. 2015; 37: 1088–1095.
Roper E, Lum T, Palme CE, et al. PD-L1 expression predicts longer disease free survival in high risk head and neck cutaneous squamous cell carcinoma. Pathology. 2017; 49: 499–505.
Ukpo OC, Thorstad WL, Lewis JS,Jr. B7-H1 expression model for immune evasion in human papillomavirus-related oropharyngeal squamous cell carcinoma. Head Neck Pathol. 2013; 7: 113–121.
Varki V, Ioffe OB, Bentzen SM, et al. PD-L1, B7-H3, and PD-1 expression in immunocompetent vs. immunosuppressed patients with cutaneous squamous cell carcinoma. Cancer Immunol Immunother. 2018; 67: 805–814.
Gambichler T, Gnielka M, Ruddel I, Stockfleth E, Stucker M, Schmitz L. Expression of PD-L1 in keratoacanthoma and different stages of progression in cutaneous squamous cell carcinoma. Cancer Immunol Immunother. 2017; 66: 1199–1204.
Amoils M, Kim J, Lee C, et al. PD-L1 Expression and tumor-infiltrating lymphocytes in high-risk and metastatic cutaneous squamous cell carcinoma. Otolaryngol Head Neck Surg. 2019; 160: 93–99.
Zandberg DP, Strome SE. The role of the PD-L1:PD-1 pathway in squamous cell carcinoma of the head and neck. Oral Oncol. 2014; 50: 627–632.
Guo W, Zhang F, Shao F, et al. PD-L1 expression on tumor cells associated with favorable prognosis in surgically resected esophageal squamous cell carcinoma. Hum Pathol. 2019; 84: 291–298.
Shen Z, Gu L, Mao D, Chen M, Jin R. Clinicopathological and prognostic significance of PD-L1 expression in colorectal cancer: a systematic review and meta-analysis. World J Surg Oncol. 2019; 17: 4.
Noh BJ, Kim JH, Eom DW. Prognostic significance of categorizing gastric carcinoma by PD-L1 expression and tumor infiltrating lymphocytes. Ann Clin Lab Sci. 2018; 48: 695–706.
Madore J, Vilain RE, Menzies AM, et al. PD-L1 expression in melanoma shows marked heterogeneity within and between patients: implications for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res. 2015; 28: 245–253.
Remon J, Chaput N, Planchard D. Predictive biomarkers for programmed death-1/programmed death ligand immune checkpoint inhibitors in nonsmall cell lung cancer. Curr Opin Oncol. 2016; 28: 122–129.
Gandini S, Massi D, Mandala M. PD-L1 expression in cancer patients receiving anti PD-1/PD-L1 antibodies: a systematic review and meta-analysis. Crit Rev Oncol Hematol. 2016; 100: 88–98.
Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014; 515: 563–567.
Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res. 2014; 20: 5064–5074.
Festino L, Botti G, Lorigan P, et al. Cancer treatment with anti-PD-1/PD-L1 agents: is PD-L1 expression a biomarker for patient selection? Drugs. 2016; 76: 925–945.
Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015; 348: 124–128.
Shen X, Zhao B. Efficacy of PD-1 or PD-L1 inhibitors and PD-L1 expression status in cancer: meta-analysis. BMJ. 2018; 362: k3529.
Ferris RL, Blumenschein GJr, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016; 375: 1856–1867.
Ferris RL, Blumenschein GJr, Fayette J, et al. Nivolumab vs investigator's choice in recurrent or metastatic squamous cell carcinoma of the head and neck: 2-year long-term survival update of CheckMate 141 with analyses by tumor PD-L1 expression. Oral Oncol. 2018; 81: 45–51.
Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med. 2012; 4: 127ra137.
Kim HS, Lee JY, Lim SH, et al. Association between PD-L1 and HPV status and the prognostic value of PD-L1 in oropharyngeal squamous cell carcinoma. Cancer Res Treat. 2016; 48: 527–536.
Kamiya S, Kato J, Kamiya T, et al. Association between PD-L1 expression and lymph node metastasis in cutaneous squamous cell carcinoma [published online ahead of print November 8, 2018]. Asia Pac J Clin Oncol. doi:10.1111/ajco.13102.
Garcia-Pedrero JM, Martinez-Camblor P, Diaz-Coto S, et al. Tumor programmed cell death ligand 1 expression correlates with nodal metastasis in patients with cutaneous squamous cell carcinoma of the head and neck. J Am Acad Dermatol 2017; 77: 527–533.
de Vicente JC, Rodriguez-Santamarta T, Rodrigo JP, Blanco-Lorenzo V, Allonca E, Garcia-Pedrero JM. PD-L1 expression in tumor cells is an independent unfavorable prognostic factor in oral squamous cell carcinoma. Cancer Epidemiol Biomarkers Prev. 2019; 28: 546–554.
Troiano G, Caponio VCA, Zhurakivska K, et al. High PD-L1 expression in the tumour cells did not correlate with poor prognosis of patients suffering for oral squamous cells carcinoma: a meta-analysis of the literature. Cell Prolif. 2019; 52: e12537.
Solomon B, Young RJ, Bressel M, et al. Prognostic significance of PD-L1(+) and CD8(+) immune cells in HPV(+) oropharyngeal squamous cell carcinoma. Cancer Immunol Res. 2018; 6: 295–304.
Oguejiofor K, Galletta-Williams H, Dovedi SJ, Roberts DL, Stern PL, West CM. Distinct patterns of infiltrating CD8+ T cells in HPV+ and CD68 macrophages in HPV- oropharyngeal squamous cell carcinomas are associated with better clinical outcome but PD-L1 expression is not prognostic. Oncotarget. 2017; 8: 14416–14427.
De Meulenaere A, Vermassen T, Aspeslagh S, et al. Tumor PD-L1 status and CD8(+) tumor-infiltrating T cells: markers of improved prognosis in oropharyngeal cancer. Oncotarget 2017; 8: 80443–80452.
Schneider S, Kadletz L, Wiebringhaus R, et al. PD-1 and PD-L1 expression in HNSCC primary cancer and related lymph node metastasis - impact on clinical outcome. Histopathology. 2018; 73: 573–584.
Figure 1
 
Correlation between HPV status and (A) AJCC T category and (B) extraconjunctival extension status in conjunctival SCC.
Figure 1
 
Correlation between HPV status and (A) AJCC T category and (B) extraconjunctival extension status in conjunctival SCC.
Figure 2
 
PD-L1 expression in conjunctival SCC and correlation between PD-L1 expression and tumor type and AJCC T category of conjunctival SCC. (AD) Representative micrographs showing PD-L1 expression in (A, B) in situ and (C, D) invasive conjunctival SCCs with (A, C) <1% and (B, D) ≥1% of tumor cells staining positive for PD-L1 (magnification ×200). Inset: corresponding field in hematoxylin-eosin-stained section (magnification ×200). (E, F) Correlation between PD-L1 staining and (E) tumor type and (F) AJCC T category. PD-L1 status not available in one case because of inadequate tumor for analysis.
Figure 2
 
PD-L1 expression in conjunctival SCC and correlation between PD-L1 expression and tumor type and AJCC T category of conjunctival SCC. (AD) Representative micrographs showing PD-L1 expression in (A, B) in situ and (C, D) invasive conjunctival SCCs with (A, C) <1% and (B, D) ≥1% of tumor cells staining positive for PD-L1 (magnification ×200). Inset: corresponding field in hematoxylin-eosin-stained section (magnification ×200). (E, F) Correlation between PD-L1 staining and (E) tumor type and (F) AJCC T category. PD-L1 status not available in one case because of inadequate tumor for analysis.
Figure 3
 
Expression of immune markers in primary conjunctival squamous cell carcinoma, with respect to HPV status: negative (AL) versus positive (MX) and PD-L1 expression in <1% (AF, MR) vs. ≥1% (GL, SX) of tumor cells. High-risk HPV: A, G, M, S (400× magnification); B, H, N, T (100× magnification); PD-L1: C, I, O, U (100× magnification); CD3: D, J, P, V (100× magnification); CD8: E, K, Q, W (100× magnification); PD1: F, L, R, X (100× magnification).
Figure 3
 
Expression of immune markers in primary conjunctival squamous cell carcinoma, with respect to HPV status: negative (AL) versus positive (MX) and PD-L1 expression in <1% (AF, MR) vs. ≥1% (GL, SX) of tumor cells. High-risk HPV: A, G, M, S (400× magnification); B, H, N, T (100× magnification); PD-L1: C, I, O, U (100× magnification); CD3: D, J, P, V (100× magnification); CD8: E, K, Q, W (100× magnification); PD1: F, L, R, X (100× magnification).
Figure 4
 
Relationships between immune infiltrate status and proportion of tumor cells expressing PD-L1 in conjunctival SCC. Shown are distributions of percentages of (A) CD3-positive, (B) CD8-positive, and (C) PD1-positive cells in the tumor-associated immune infiltrate according to PD-L1 expression.
Figure 4
 
Relationships between immune infiltrate status and proportion of tumor cells expressing PD-L1 in conjunctival SCC. Shown are distributions of percentages of (A) CD3-positive, (B) CD8-positive, and (C) PD1-positive cells in the tumor-associated immune infiltrate according to PD-L1 expression.
Table 1
 
Demographic, Clinicopathologic, and Immunohistochemical Parameters in Patients With Conjunctival SCC and Correlation With Tumor HPV Status and Proportion of Tumor Cells Expressing PD-L1
Table 1
 
Demographic, Clinicopathologic, and Immunohistochemical Parameters in Patients With Conjunctival SCC and Correlation With Tumor HPV Status and Proportion of Tumor Cells Expressing PD-L1
Table 2
 
Correlation Between Tumor HPV Status and PD-L1 Expression
Table 2
 
Correlation Between Tumor HPV Status and PD-L1 Expression
Table 3
 
Correlation Between Clinicopathologic Characteristics, HPV and PD-L1 Status, and Immune Infiltrates
Table 3
 
Correlation Between Clinicopathologic Characteristics, HPV and PD-L1 Status, and Immune Infiltrates
Supplement 1
Supplement 2
Supplement 3
Supplement 4
Supplement 5
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×