eISSN: 1897-4309
ISSN: 1428-2526
Contemporary Oncology/Współczesna Onkologia
Current issue Archive Manuscripts accepted About the journal Supplements Addendum Special Issues Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
3/2022
vol. 26
 
Share:
Share:
Original paper

Correlation of PD-L1 expression with toxicities and response in oropharyngeal cancers treated with definitive chemoradiotherapy

Smriti Srivastava
1
,
Madhup Rastogi
1
,
Ajeet Kumar Gandhi
1
,
Rohini Khurana
1
,
Rahat Hadi
1
,
Shantanu Sapru
1
,
Anoop Srivastava
2
,
Avinav Bharati
2
,
Nuzhat Husain
3
,
Surendra Prasad Mishra
2
,
Kamal Sahni
1

1.
Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
2.
Medical Physics, Department of Radiation Oncology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
3.
Department of Pathology, Dr Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
Contemp Oncol (Pozn) 2022; 26: 180–186
Online publish date: 2022/09/07
Article file
- Correlation of PD-L1.pdf  [0.20 MB]
Get citation
 
PlumX metrics:
 

Introduction

Head and neck squamous cell carcinoma (HNSCC) accounts for 0.2 million cases every year in India. Of these, oropharyngeal squamous cell carcinoma (OPSCC) comprise 20,000 cases per year [1]. Concurrent chemoradiotherapy (CCRT) is the current standard of care for locally advanced OPSCC. Treatment intensification with CCRT has led to increased acute and late toxicities [24]. There are several predictive factors for development of radiation morbidities, such as biologically effective dose, radiation techniques, and use of concurrent chemotherapy [5]. However, these factors are not sufficient to explain the inter-individual variability among patients for development of radiation toxicities. Gene modifications in biological pathways like DNA repair, cell cycle, and apoptosis have been found to be predictive of acute radiation toxicities in HNSCC like XRCC, RAD51, GSTP, etc. [6, 7]. Furthermore, the immune system of the patient is instrumental in determining radiation repair, and immune escape mechanisms may determine the individual response to radiation therapy (RT). Identification of novel biomarkers modulating immune response, like cytotoxic T lymphocyte-associated antigen 4, lymphocyte-activation gene-3, T-cells immunoglobulin mucin protein-3, and programmed death receptor ligand 1 (PD-L1), may predict the radiation response and toxicities in HNSCC [8].

Programmed death receptor 1 (PD1) is a member of the extended family of T-cell regulators expressed on the surface of activated T-cells, B-cells, and macrophages, and its ligand PD-L1 is a cell surface glycoprotein expressed on T-cells, macrophages, cancer cells, and other tissues. Co-expression of PD-1 and PD-L1 inhibits proliferation of lymphocytes and T-cells mediated cytokine secretion. The PD-1/PD-L1 interaction protects healthy cells from excessive inflammatory response. However, in the tumour microenvironment (TME) this interaction causes inhibition of activated T-cell proliferation and promotes apoptosis of T-cells, resulting in enhanced tumour cell growth [9].

The critical role of PD-L1 in HNSCC carcinogenesis was demonstrated by Zheng et al. [10]. Recent studies suggest that the PD-1/PD-L1 axis is closely related to HPV-associated HNSCC, suggesting the relevance of PD-L1 expression on tumour cells (TCs) and T-cells in OPSCC [11, 12]. The association of PD-L1 expression with clinicopathological features and its prognostic significance remains controversial [1316] . While some studies have reported poor clinical outcomes [14, 17] in PD-L1 expressing HNSCC, others have found improved survival [8, 15]. Although some studies demonstrate a change in PD-L1 expression level by local fractionated RT [18, 19], the correlation of PD-L1 expression with radiation toxicities remains unknown. So, we conducted this prospective study to evaluate the correlation of PD-L1 expression in tumour and tumour infiltrating lymphocytes (TILs) with acute radiation toxicity and response in patients with OPSCC treated with definitive CCRT.

Material and methods

This was a single-institution prospective observational study conducted from Dec 2017 to March 2021. The enrolment period was from Jan 2018 to April 2019. The study protocol was approved by the Institutional Ethical Committee (IEC no. 29/17). It included patients with histopathologically proven squamous cell carcinoma of the oropharynx aged between 18 and 80 years, with stage II to IVA disease, Karnofsky performance status ≥ 70, and normal haematological, renal, and hepatic functions. Patients were required to sign an informed consent form before enrolment in the study. Pre-treatment workup included baseline haematological parameters, chest X-Ray PA view, contrast-enhanced computed tomography (CECT) of face and neck region, and direct laryngoscopy. P16 expression was tested and patients were staged according to American Joint Committee on Cancer recommendations (eighth edition 2018) [20].

Immuno-histochemistry for PD-L1 expression

Biopsy samples or slide/blocks were sent to the pathology department of the institute. The tissue samples were formalin fixed and paraffin embedded. Then 3–4 µm sections were cut and subsequently stained with haematoxylin and eosin to check for the presence of tumour. Immuno-histochemistry (IHC) was applied using antibodies to PD-L1 (clone SP263) in the automated IHC slide staining system, VENTANA BenchMark XT according to manufacturer’s instructions. Deparaffinization, retrieval, and staining were done onboard (Ventana Medical Systems, Inc., Tucson, USA). Programmed death receptor ligand 1 expression was recorded on TCs and TILs. Programmed death receptor ligand 1 expression was considered positive if >1% of cells showed moderate to intense membranous or cytoplasmic staining with grading as follows: 1–10% (1+); 11–50% (2+), and > 50% (3+). The absence of membranous expression of TCs was deemed to be PD-L1 negative. Programmed death receptor ligand 1 expression in TILs (PD-L1-TILs) was reported separately (Fig. 1).

Fig. 1

Immunohistochemical staining for programmed death receptor ligand 1 (PD-L1) in patients with squamous cell carcinoma oropharynx. PD-L1 expression scored 3+ (strong membrane staining in more than 50% of tumour cells [TCs]) (A), PD-L1 expression in tumour cell scored 1+ (weak to moderate expression in 10% TCs) (B), PD-L1 expression in 2% TCs and 10% in tumour infiltrating lymphocytes (C)

/f/fulltexts/WO/47517/WO-26-47517-g001_min.jpg

Treatment

Definitive CCRT with curative intent was given to all patients depending on their clinical stage and general condition. Contrast-enhanced computed tomography simulation was performed using appropriate thermoplastic cast immobilization, and images of 3 mm slice thickness were acquired. Data were transferred to a treatment planning system (XIO Version 5.0/MONACO version 5.11.02) using DICOM (Digital Imaging and Communications in Medicine) protocol 3.0. Delineation of target volumes and organ at risk volumes were performed as defined in International Commission on Radiation Units and Measurements report numbers 50 and 62 [21]. A target dose of 66 to 70 Gray (Gy) in 33 to 35 fractions in 2 phases over a period of 6 to 7 weeks, 5 days a week was planned using a shrinking field technique.

In the first phase, 44 Gy was delivered using a parallel opposed field to bilateral face and neck and a low anterior neck field. The second phase was delivered using an off-cord bilateral parallel opposed field with or without posterior neck electron boost to a dose of 22–26 Gy. Posterior neck was boosted with 6–12 MeV electrons using R-90 as prescription isodose line if clinically indicated. Set up verification was done with EPID (Electronic Portal Imaging Device) twice a week to ensure proper positioning during the entire course of RT. Treatment delivery was done using a linear accelerator (Infinity, Synergy and Compact; Elekta) having MLC with leaf width of 1 cm using 6 MV photons. Injection of cisplatin 40 mg/m2 weekly was given as concurrent chemotherapy. Patients were assessed at least once during RT, and toxicities were recorded by the physician weekly as per Radiation Therapy Oncology Group (RTOG) acute toxicity criteria RTOG [22].

Post-treatment visits were monthly for 3 months and then 2-monthly for the next 6 months and every 3 months thereafter. Three months after completion of treatment, or earlier if clinically indicated, response assessment was done using clinical examination, direct laryngoscopy, and CECT of the face and neck with or without biopsy or fine needle aspiration cytology of suspicious or persistent lesions. In accordance with World Health Organization (WHO) response assessment criteria, the response was classified into complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD) [23]. A schema of the study is presented in Figure 2.

Fig. 2

Depicting the study schema

/f/fulltexts/WO/47517/WO-26-47517-g002_min.jpg

Statistical analysis

Statistical analysis was done with statistical package for social sciences (SPSS, version 23.0, IBM). Mean and standard deviation were estimates of quantitative data. Programmed death receptor ligand 1 expression was correlated with the clinic-pathological parameters and response to treatment using Student’s t-test or χ2 test. WHO response was correlated with PD-L1 expression using χ2 test or Fisher exact test. Overall survival (OS) was calculated from date of registration until the date of death from any cause. Progression-free survival (PFS) was evaluated from the date of initiation of treatment until the date of disease progression (locoregional, distant, or both) or death. Survival rates were estimated using Kaplan-Meier method, and the log-rank test was used to compare survival outcomes. All reported p-values were 2-sided, and p ≤ 0.05 was considered significant.

Results

In total, 56 patients were enrolled, of whom 51 successfully completed treatment and were included in the study cohort. Three patients defaulted before initiation of treatment (due to personal reasons), and 2 patients expired during treatment (due to pneumonia and cardiac arrest respectively), and these 5 patients were excluded from the study.

Ten (19.6%) patients were positive for p16 expression. Twenty (39.2%) patients had PD-L1 expression in TCs (PD-L1-tumour +ve). Eight (15.7%) patients had 1+ PD-L1 expression, while 6 (11.8%) patients had 2+ and 3+ expression each. The median age was 55 years (range, 26–75 years). Clinicopathological correlation of PD-L1 expression on TCs with patient characteristics is summarized in Table 1. Eighteen (35.3%) patients had PD-L1 positivity in TILs (PD-L1-TILs +ve) and 11 (21.6%) had PD-L1 expression in both TCs and TILs. PD-L1 expression in TCs was significantly associated with PD-L1 expression in TILs (p = 0.034). Higher nodal burden was observed in PD-L1-TIL –ve than PD-L1-TILs +ve (65% vs. 44%, p = 0.03).

Table 1

Comparison of baseline patient characteristics stratified by the programmed death receptor ligand 1 expression in tumour cells

VariableAll patients (N = 51)PD-L1 positive (n = 20)PD-L1 negative (n = 31)p-value
Gender: male/female, n45 : 616 : 429 : 20.19
Mean age (range), y55 (26–75)56.5 (26–75)54 (38–71)0.25
Karnofsky performance status, n (%)1.00
≤ 8040 (78.5)16 (80)24 (77.4)
> 8011 (21.6)4 ( 20)7 (22.6)
Subsite, n (%)
Soft palate10 (19.6)2 (10)8 (25.8)
Tonsil and tonsillolingual sulcus7 (13.7)2 (10)5 (16.1)
Base of tongue29 (56.7)13 (65)16 (51.5)
Vallecula2 (3.9)2 (10)0 (0)
Posterior pharyngeal wall3 (5.9)1 (5)2 (6.5)
Tumour category, n (%)*
T1 + T210 (19.6)4 (20)6 (19.4)1.00
T3 + T441 (80.4)16 (80)25 (80.6)
Nodal category, n (%)
N0 + N118 (35)8 (40)10 (32)0.76
N2 + N333 (65)12 (60)21 (68)
Differentiation, n (%)
Well differentiated + moderately differentiated42 (82.5)15 (75)27 (87)0.28
Poorly differentiated9 (17.6)5 (25)4 (13)
Smoking history, n (%)#
Yes42 (82.6)14 (70)28 (90.3)0.12
No9 (17.4)6 (30)3 (9.7)
Tobacco chewing history, n (%)
Yes39 (76.5)14 (70)25 (80.6)0.50
No12 (23.5)6 (30)6 (19.4)
P16 expression, n (%)
Positive10 (19.6)5 (25)5 (16)0.49
Negative41 (80.4)15 (75)26 (84)

PD-L1 – the programmed death receptor ligand 1

* All staging was performed as per American Joint Committee on Cancer Staging (eighth edition, 2018)

# For smoking status yes indicates active smokers at present or patient who smoked > 100 cigarettes in their lifetime, and no indicates patients who never smoked or who are currently non-smokers and smoked <100 cigarettes in their lifetime

The median RT dose was 70 Gy (range, 66–70 Gy), with a median RT duration of 50 days (range, 48–57 days). All patients received cisplatin-based CCRT with a median cumulative dose of 320 mg (range, 250–350). Forty-four (86.3%) patients received ≥ 5 cycles of CCRT. Seven patients received ≤ 4 cycles; of these, 2 were PD-L1-tumour +ve. Two patients received 68 and 66 Gy over 34 and 33 fractions, respectively, due to grade 4 mucositis; both were PD-L1-tumour –ve. Prophylactic nasogastric tube (NGT) or percutaneous endoscopic gastrostomy tube feeding was not used as per institutional policy. Nasogastric tube feeding was required in 6 patients, 4 in PD-L1 –ve group and 2 in PD-L1 +ve group (1 each in tumor and TILs). The median duration of NGT feeding was 25 days (18 days for PD-L1 +ve and 22 days for PD-L1 –ve). Highest grade of radiation toxicity observed during CTRT were taken into consideration. Radiation toxicities in PD-L1 +ve vs. PD-L1 –ve patients are compared in Table 2. On multivariable modelling, PD-L1 status was not found to be an independent predictor for severe acute radiation toxicities (all p-value > 0.05).

Table 2

Correlation of the programmed death receptor ligReferences and 1 expression in tumour and tumour-infiltrating lymphocytes with acute radiation toxicity

VariableAll patients N = 51 (%)PD-L1-tumour +ve n = 20 (%)PD-L1-tumour –ve n = 31 (%)p-valuePD-L1-TILs +ve n = 18 (%)PD-L1-TILs –ve n = 33 (%)p-value
Oral mucositis
Grade ≤ 228 (55)15 (75)13 (42)0.02514 (78)14 (42)0.020
Grade ≥ 323 (45)5 (25)18 (58)4 (22)19 (58)
Dysphagia
Grade ≤ 229 (57)15 (75)14 (45.2)0.04615 (83)14 (42)0.007
Grade ≥ 322 (43)5 (25)17 (54.8)3 (17)19 (58)
Xerostomia
Grade 121 (41)14 (70)12 (39)0.0412 (67)14 (42)0.144
Grade 230 (59)6 (30)19 (61)6 (33)19 (58)
Laryngeal toxicity
Grade ≤ 225 (49)11 (55)14 (45)0.57211 (61)14 (42)0.24
Grade ≥ 326 (51)9 (45)17 (55)7 (39)19 (58)
Skin reactions
Grade 121 (41)10 (50)11 (35)0.38611 (61)10 (30)0.04
Grade 230 (59)10 (50)20 (65)7 (39)23 (70)
Haematological toxicity
Grade 140 (78)15 (75)25 (80)0.73215 (83)25 (76)0.72
Grade 211 (22)5 (25)6 (20)3 (17)8 (24)

[i] PD-L1 – the programmed death receptor ligand 1, TILs – tumour infiltrating lymphocytes

On response evaluation after 3 months of RT completion, overall CR was achieved in 67% patients while 24% had PR and 4% had PD, none of the patients had SD. Complete response was 76% vs. 67% and PR + PD was 23% vs. 10% respectively in PD-L1-tumour +ve vs. PD-L1-tumour –ve group (p = 0.74). At a median follow-up of 25 months (range,1–39 months), the 2-year OS rate of entire cohort was 58.8% and the 2-year PFS rate for entire cohort was 54.9% (median PFS – 28 months). Two-year OS rate for PD-L1-tumour –ve vs. PD-L1-tumour +ve seen in other tumour types and group was 58.1% and 60% [hazard ratio (HR), 1.21; 95% CI: 0.51–2.83; p = 0.65]. Two-year PFS rate was 51.6% in PD-L1-tumour –ve group v 60% for PD-L1-tumour +ve group (HR, 1.19; 95% CI: 0.54–2.58; p = 0.659) (Fig. 3).

Fig. 3

Kaplan-Meier’s analysis of survival rates. Progression-free survival (A), overall survival of tumour programmed death receptor ligand 1 positive vs. survival probability (B)

/f/fulltexts/WO/47517/WO-26-47517-g003_min.jpg

Complete response rate was 59% vs. 77% and PR + PD was 41.4% vs. 22.4%, respectively, in the PD-L1-TILs +ve vs. the PD-L1-TILs –ve group (p = 0.20). The two-year OS rate for the PD-L1-TILs –ve group vs. the PD-L1-TILs +ve group was 60.6% and 55.6%, respectively (HR, 0.829; 95% CI: 0.36–1.89; p = 0.65). The two-year PFS rate was 54.5% in the PD-L1-TILs –ve group vs. 55.6% in the PD-L1-TILs +ve group (HR, 1.21; 95% CI: 0.568–2.59; p = 0.619).

Discussion

The PD-1/PD-L1 axis has emerged as a valuable biomarker in predicting response to anti PD-1/PD-L1 therapy. The PD-L1 positivity in HNSCC ranges from 18–80% reported in various studies (11–15, 22). This high variability is also seen in other tumour types and is due to technical difficulties in determining PD-L1 expression [24, 25]. In our study, positive PD-L1 expression in TCs and TILs was observed in 39.2% and 35.2%, respectively, which is in concordance with contemporary studies. Although HPV status has been shown to have positive correlation with PD-L1 expression [26], Kim et al. [16] could not find any association between them. In our study no significant association of PD-L1 expression with clinicopathological parameters and HPV status was observed. However, in our study PD-L1 expression in TILs was significantly associated with N stage and PD-L1 expression in tumour.

Our study, to the best of our knowledge, is the first prospective study to correlate PD-L1 expression with both acute radiation toxicity and treatment response in patients of OPSCC treated with cisplatin based CCRT. We noted higher grade ≥ 3 maximal toxicities in terms of oral mucositis, dysphagia, xerostomia, and laryngeal toxicity in the PD-L1-tumour –ve group. Similar findings were also observed in PD-L1-TILs because grade ≥ 3 oral mucositis; dysphagia, and grade ≥ 2 skin reactions were more common in the PD-L1-TILs –ve group (Table 2). The programmed death receptor ligand 1 negative patients also had delayed healing of radiation reactions in comparison to their positive counterparts one month after completion of RT.

The immune effect of fractionated RT elucidates our finding. Radiation causes generation of reactive oxygen species and nitric oxide in the TME and breaks the DNA double strand, leading to release of cytokines, activation of toll-like receptors signalling immune response pathway, and secretion of pro-inflammatory cytokines such as interleukin-1 (IL-1), IL-2, -4, and -6, and tumour necrosis factor, leading to activation of immune cells. These mediators increase vascular permeability causing more infiltration and recruitment of inflammatory cells. This leads to proliferation of regulatory and helper T-cells. The dead cells at the site of irradiation also release damage-associated molecular patterns like adenosine triphosphate, which activate local dendritic cells, increasing antigen presentation [2729]. As demonstrated by Dovedi et al. [18], the PD1/PD-L1 expression in immune and TCs is upregulated by RT, which leads to functional anergy of T-cells and hence modulation of acute inflammatory response. This could be a plausible hypothesis suggesting fewer acute radiation morbidities in patients with increased PD-L1 expression. These findings concur with our finding of decreased radiation toxicity in patients with PD-L1 expression. Myers et al. also reported enhanced healthy tissue damage by T-cell activation with the addition of PD-1 blockade to RT in mice treated with RT and PD-1 blockade vs. RT alone [30].

The correlation of CCRT and PD-L1 expression has not been extensively studied. In a retrospective study of 92 patients by Fukushima et al. [31] better outcomes were observed in patients with high PD-L1 expression. A caveat to their study was the inclusion of patients who received induction chemotherapy and various CCRT regimens, unlike our study which has a homogenous treatment cohort. We observed better CR in PD-L1-tumour +ve patients, but this was statistically not significant (76% vs. 67%, p = 0.74). The impact of PD-L1 expression on clinical outcomes of patients has been studied in various solid tumours, but the correlation with prognosis remains to be deciphered. Two Meta-analysis done in HNSCC by Li et al. and Yang et al. could not establish a correlation of PD-L1 expression with OS and DFS; however, in the former, PD-L1 +ve patients showed improved PFS, while poor survival in the Asian population was observed in the latter [32, 33]. Our results could not show significant association of PD-L1 expression on tumour or TILs with OS and PFS in OPSCC patients. Larger cohort-based studies with longer follow-up are needed to establish an association.

The programmed death receptor ligand 1 mediates the inter-relationship between TCs and TILs. However, the PD-L1 upregulation could be an adaptive response resulting from a vigorous immune microenvironment which evinces anti-cancer effects [3436]. Multiple cell types like malignant, immune, and stromal cells and immune checkpoints impose more intricacy to TME (9,31,32). Most studies show that the pathogenesis of OPSCC (especially HPV positive) has an immunosuppressive origin and is related to the PD-1/PD-L1 pathway, but it is not yet established whether this pathway plays a greater role in HPV-positive cancers compared to HPV-negative ones [11, 16]. Therefore, PD-L1 expression alone may not be considered a strong prognostic factor in OPSCC.

A major limitation to our study was the small sample size and a shorter follow-up. Due to lack of specific data on the study outcomes and owing to limited resources, a sample size was also not specifically calculated for our study and hence the exact power of our study is unknown, and this is a limitation of our study. Owing to the small sample size, only qualitative expression of PD-L1 could be taken into consideration, so the difference in toxicity and outcome due to intensity of PD-L1 expression could not be established. The number of HPV positives was also very low, perhaps because of the regional population of patients included in our study.

Despite these shortcomings, we believe that the scarcity of data regarding PD-L1 expression in OPSCC and its effect on radiation toxicities in patients makes our study unique. The majority of the studies [18, 29] focused on effect of RT on changes in PD-L1 expression and the role of PD-L1 blockade therapy [37] in the outcome, but none of these discussed the outcome of RT in terms of toxicity and treatment response in patients with positive PD-L1 expression. Prospective use of PD-L1 expression as a predictor of radiation toxicities may guide early institution of supportive therapy and early consideration of NGT insertion or percutaneous endoscopic gastrostomy tube placement in patients having negative PD-L1 expression. This could reduce toxicity-related treatment interruption and decrease morbidity in patients, which will further lead to better treatment outcomes. This could serve as a potential biomarker for predicting acute radiation toxicities in this cohort of patients. The combination of immune check-point inhibitors with radiotherapy in HNSCC is an intensive area of ongoing research [38], and the findings from our study may guide future clinical trials in this setting.

Conclusions

Patients with positive PD-L1 expression in TCs and TILs treated with definitive CCRT have significantly fewer acute radiation toxicities compared to their negative counterparts. The programmed death receptor ligand 1 expression alone could not be considered as a prognostic factor for survival in OPSCC. Our study provides encouraging data for the use of PD-L1 expression as a potential biomarker for acute radiation toxicities, but it requires further evaluation in a larger cohort to find its prognostic implication on clinical outcome in terms of tumour response and survival. This may help in the recognition of the cohort of patients who may need early and additional support during treatment.

Notes

[5] Conflicts of interest The authors declare no conflict of interest.

References

1 

Available from: https://gco.iarc.fr/ (accessed: July 20, 2021).

2 

Lacas B, Bourhis J, Overgard J, et al. Role of radiotherapy fractionation in head and neck cancers (MARCH): an updated meta-analysis. Lancet Oncol 2017; 18: 1221-1237.

3 

Adelstein DJ, Li Y, Adams GL, et al. An intergroup phase III comparison of standard radiation therapy and two schedules of concurrent chemoradiotherapy in patients with unresectable squamous cell head and neck cancer. J Clin Oncol 2003; 21: 92-98.

4 

Meyer F, Fortin A, Wang CS, et al. Predictors of severe acute and late toxicities in patients with localized head-and-neck cancer treated with radiation therapy. Int J Radiat Oncol Biol Phys 2012; 82: 1454-1462.

5 

Marcu L, Van Doorn T, Olver I. Cisplatin and radiotherapy in the treatment of locally advanced head and neck cancer: a review of their cooperation. Acta Oncol 2003; 42: 315-325.

6 

Normando AGC, Rocha CL, de Toledo IP, et al. Biomarkers in the assessment of oral mucositis in head and neck cancer patients: a systematic review and meta-analysis. Support Care Cancer 2017; 25: 2969-2988.

7 

Nanda SS, Gandhi AK, Rastogi M, et al. Evaluation of XRCC1 gene polymorphism as a biomarker in head and neck cancer patients undergoing chemoradiation therapy. Int J Radiat Oncol Biol Phys 2018; 101: 593-601.

8 

Kim HR, Ha SJ, Hong MH, et al. PD-L1 expression on immune cells, but not on tumor cells, is a favorable prognostic factor for head and neck cancer patients. Sci Rep 2016; 6: 36956.

9 

Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol 2008; 26: 677-704.

10 

Zheng A, Li F, Chen F, et al. PD-L1 promotes head and neck squamous cell carcinoma cell growth through mTOR signaling. Oncol Rep 2019; 41: 2833-2843.

11 

Lyford-Pike S, Peng S, Young GD, et al. Evidence for a role of the PD-1:PD-L1 pathway in immune resistance of HPV-associated head and neck squamous cell carcinoma. Cancer Res 2013; 73: 1733-1741.

12 

Badoual C, Hans S, Merillon N, et al. PD-1-expressing tumor-infiltrating T-cell are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res 2013; 73: 128-138.

13 

Satgunaseelan L, Gupta R, Madore J, et al. Programmed cell death-ligand 1 expression in oral squamous cell carcinoma is associated with an inflammatory phenotype. Pathology 2016; 48: 574-580.

14 

Müller T, Braun M, Dietrich D, et al. PD-L1: a novel prognostic biomarker in head and neck squamous cell carcinoma. Oncotarget 2017; 8: 52889-52900.

15 

Hong AM, Vilain RE, Romanes S, et al. PD-L1 expression in tonsillar cancer is associated with human papillomavirus positivity and improved survival: implications for anti-PD1 clinical trials. Oncotarget 2016; 7: 77010-77020.

16 

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.

17 

Wu P, Wu D, Li L, et al. PD-L1 and survival in solid tumors: a meta-analysis. PLoS One 2015; 10: e0131403.

18 

Dovedi SJ, Illidge TM. The antitumor immune response generated by fractionated radiation therapy may be limited by tumor cell adaptive resistance and can be circumvented by PD-L1 blockade. Oncoimmunology 2015; 4: e1016709.

19 

Lim SH, Hong M, Ahn S, et al. Changes in tumour expression of programmed death-ligand 1 after neoadjuvant concurrent chemoradiotherapy in patients with squamous oesophageal cancer. Eur J Cancer 2016; 52: 1-9.

20 

Amin MB, Edge SB. AJCC cancer staging system, 8th edition. Am Jt Commitee Cancer AJCC Cancer Staging Manual 8th Ed 2017, XVII, 1032.

21 

Newhauser W. International Commission on Radiation Units and Measurements Report 78: prescribing, recording and reporting proton-beam therapy. Radiat Prot Dosimetry 2009; 133: 60-62.

22 

Cox JD, Stetz JA, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European organization for research and treatment of cancer (EORTC). Int J Radiat Oncol Biol Phys 1995; 31: 1341-1346.

23 

Miller AB, Hoogstraten B, Staquet M, Winkler A. Reporting results of cancer treatment. Cancer 1981; 47: 207-214.

24 

Oliveira-Costa JP, de Carvalho AF, da Silveira GG, et al. Gene expression patterns through oral squamous cell carcinoma development: PD-L1 expression in primary tumor and circulating tumor cells. Oncotarget 2015; 6: 20902-20920.

25 

Lin YM, Sung WW, Hsieh MJ, et al. High PD-L1 expression correlates with metastasis and poor prognosis in oral squamous cell carcinoma. PLoS One 2015; 10: e0142656.

26 

Balermpas P, Rödel F, Krause M, et al. The PD-1/PD-L1 axis and human papilloma virus in patients with head and neck cancer after adjuvant chemoradiotherapy: a multicentre study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). Int J Cancer 2017; 141: 594-603.

27 

Maria OM, Eliopoulos N, Muanza T. Radiation-induced oral mucositis. Front Oncol 2017; 7: 89.

28 

Sprung CN, Forrester HB, Siva S, et al. Immunological markers that predict radiation toxicity. Cancer Lett 2015; 368: 191-197.

29 

Gong J, Le TQ, Massarelli E, et al. Radiation therapy and PD-1/PD-L1 blockade: the clinical development of an evolving anticancer combination. J Immunother Cancer 2018; 6: 46.

30 

Myers CJ, Lu B. Decreased survival after combining thoracic irradiation and an anti-PD-1 antibody correlated with increased T-cell infiltration into cardiac and lung tissues. Int J Radiat Oncol Biol Phys 2017; 99: 1129-1136.

31 

Fukushima Y, Someya M, Nakata K, et al. Influence of PD-L1 expression in immune cells on the response to radiation therapy in patients with oropharyngeal squamous cell carcinoma. Radiother Oncol 2018; 129: 409-414.

32 

Li J, Wang P, Xu Y. Prognostic value of programmed cell death ligand 1 expression in patients with head and neck cancer: a systematic review and meta-analysis. PLoS One 2017; 12: e0179536.

33 

Yang WF, Wong MCM, Thomson PJ, et al. The prognostic role of PD-L1 expression for survival in head and neck squamous cell carcinoma: a systematic review and meta-analysis. Oral Oncol 2018; 86: 81-90.

34 

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: 127ra37.

35 

De Ruiter EJ, Ooft ML, Devriese LA, et al. The prognostic role of tumor infiltrating T-lymphocytes in squamous cell carcinoma of the head and neck: a systematic review and meta-analysis. Oncoimmunology 2017; 6: e1356148.

36 

Puram SV, Tirosh I, Parikh AS, et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 2017; 171: 1611-1624.e24.

37 

Qiao XW, Jiang J, Pang X, et al. The evolving landscape of PD-1/PD-L1 pathway in head and neck cancer. Front Immunol 2020; 11: 1721.

38 

Biau J, Bourhis J. Combining immunotherapy and radiotherapy in head and neck squamous cell cancers: which perspectives? Curr Opin Oncol 2020; 32: 196-202.

Copyright: © 2022 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.