Skip to main content
Erschienen in: Journal of Translational Medicine 1/2019

Open Access 01.12.2019 | Research

SUPREME-HN: a retrospective biomarker study assessing the prognostic value of PD-L1 expression in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck

verfasst von: Sara I. Pai, Ezra E. W. Cohen, Derrick Lin, George Fountzilas, Edward S. Kim, Holger Mehlhorn, Neus Baste, Daniel Clayburgh, Loren Lipworth, Carlo Resteghini, Nawar Shara, Takashi Fujii, Jun Zhang, Michael Stokes, Huifen Wang, Philip Twumasi-Ankrah, Sophie Wildsmith, Asud Khaliq, Giovanni Melillo, Norah Shire

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2019

Abstract

Background

Programmed cell death ligand-1 (PD-L1) expression on tumor cells (TCs) is associated with improved survival in patients with head and neck squamous cell carcinoma (HNSCC) treated with immunotherapy, although its role as a prognostic factor is controversial. This study investigates whether tumoral expression of PD-L1 is a prognostic marker in patients with recurrent and/or metastatic (R/M) HNSCC treated with standard chemotherapy.

Methods

This retrospective, multicenter, noninterventional study assessed PD-L1 expression on archival R/M HNSCC tissue samples using the VENTANA PD-L1 (SP263) Assay. PD-L1 high was defined as PD-L1 staining of ≥ 25% TC, with exploratory scoring at TC ≥ 10% and TC ≥ 50%. The primary objective of this study was to estimate the prognostic value of PD-L1 status in terms of overall survival (OS) in patients with R/M HNSCC.

Results

412 patients (median age, 62.0 years; 79.9% male; 88.2% Caucasian) were included from 19 sites in seven countries. 132 patients (32.0%) had TC ≥ 25% PD-L1 expression; 199 patients (48.3%) and 85 patients (20.6%) had TC ≥ 10% and ≥ 50%, respectively. OS did not differ significantly across PD-L1 expression (at TC ≥ 25% cutoff median OS: 8.2 months vs TC < 25%, 10.1 months, P = 0.55) or the ≥ 10% and ≥ 50% cutoffs (at TC ≥ 10%, median OS: 9.6 months vs TC < 10%, 9.4 months, P = 0.32, and at TC ≥ 50%, median OS 7.9 vs TC < 50%, 10.0 months, P = 0.39, respectively).

Conclusions

PD-L1 expression, assessed using the VENTANA PD-L1 (SP263) Assay, was not prognostic of OS in patients with R/M HNSCC treated with standard of care chemotherapies.
Trial registration ClinicalTrials.gov, NCT02543476. Registered September 4, 2015.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CI
confidence interval
CT
computed tomography
ECOG
Eastern Cooperative Oncology Group
FFPE
formalin-fixed, paraffin-embedded
HNSCC
head and neck squamous cell carcinoma
HPV
human papillomavirus
HR
hazard ratio
IC
immune cell
IHC
immunohistochemistry
IO
immuno-oncology
ORR
objective response rate
OS
overall survival
PD-1
programmed cell death-1
PD-L1
programmed cell death-ligand 1
PFS
progression-free survival
PS
performance status
RECIST
Response Evaluation Criteria In Solid Tumors
R/M HNSCC
recurrent and/or metastatic head and neck squamous cell carcinoma
TC
tumor cell

Background

Approximately 60% of patients with head and neck squamous cell carcinoma (HNSCC) are diagnosed with locally advanced disease, which has a 5-year overall survival (OS) rate of approximately 30% [1]. Most patients with HNSCC will eventually experience either local or distant recurrence [2], while approximately 10% of patients with HNSCC will initially present with metastatic disease [3]. Patients with recurrent and/or metastatic (R/M) HNSCC have historically had a poor prognosis [4]. Traditional standard of care for first-line therapy in patients with R/M HNSCC is platinum-based chemotherapy plus cetuximab and 5-fluorouracil [5, 6], yielding a median OS of approximately 10 months [7]. However, this is usually only appropriate for patients who have an acceptable Eastern Cooperative Oncology Group performance status (ECOG PS) and are able to tolerate platinum-based therapy. Patients with R/M HNSCC treated in the second-line setting have a poorer prognosis, with median OS of approximately 4–8 months [3, 8, 9]. Standard therapy in this setting includes single-agent therapies (e.g. methotrexate, docetaxel, or cetuximab) which yield objective response rates (ORRs) of 4–13% in the platinum-refractory setting [3, 8, 9]. More recently, phase III studies have demonstrated that immuno-oncology (IO) agents targeting programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) improve OS in both the first-line and second-line settings, with median OS of approximately 13–15 months and 7–8 months, respectively [1014].
PD-L1 is expressed on antigen-presenting cells and other immune cells (ICs) and is upregulated on HNSCC tumor cells (TCs) [15, 16]. The presence of PD-L1 can be readily detected by immunohistochemistry (IHC) staining [16]. Evidence is building that PD-L1 expression on TCs is associated with improved survival in patients with HNSCC treated with IO agents and yet the role of PD-L1 in outcomes irrespective of treatment (i.e. prognosis) is still unclear, with conflicting reports of PD-L1 as both a negative and positive prognostic factor [1723]. Therefore, the SUPREME-HN study was conducted to investigate the possible prognostic role PD-L1 expression on TCs has in patients with R/M HNSCC. Here, we describe patient characteristics, OS, and other clinical outcomes related to PD-L1 expression independent of treatment choice [20, 24].

Methods

Study design

SUPREME-HN was a retrospective, international, multicenter, noninterventional cohort study based on data derived from established medical records and analysis of archival tumor samples (ClinicalTrials.gov identifier: NCT02543476); for the purposes of this study and for patient selection, the index date was defined as the date of diagnosis of R/M disease not amenable to local therapy.

Patient population

Patients aged ≥ 18 years with histologically confirmed HNSCC of the oral cavity (tongue, gum, floor of mouth, or other/unspecified part of the mouth), oropharynx, larynx, or hypopharynx were eligible if they had R/M disease not amenable to local therapy with curative intent (surgery, radiation therapy, chemo-radiation). Patients with locally advanced disease amenable to curative local therapy were excluded as were patients who had received prior IO treatment with anti-cytotoxic T-lymphocyte-associated antigen 4, or anti-PD-1, anti-PD-L1, or anti-PD-L2 antibodies for HNSCC.

Procedures

Archival tumor samples (< 5 years old) were obtained anytime during the disease history from patients who were diagnosed between March 1, 2011 and June 30, 2015. Biopsies or resections from the primary site, lymph node, or distant metastatic sites were provided for analysis as formalin-fixed, paraffin-embedded (FFPE) blocks or sections < 60 days old.
For patients with more than one tissue sample, the most recent sample from the index date was used to determine PD-L1 expression. PD-L1 IHC staining of FFPE tissue samples was performed using the VENTANA PD-L1 (SP263) Assay on the automated Ventana BenchMark ULTRA® platform (Ventana Medical Systems Inc., Tucson, AZ, USA) [25]. PD-L1 expression was scored by pathologists trained by the manufacturer, at an approved central testing laboratory. PD-L1 expression was evaluated for a cutoff of ≥ 25% of TCs with membrane staining for PD-L1 at any intensity (TC ≥ 25%). Exploratory scoring was assessed at TC ≥ 10% and TC ≥ 50%. Patient characteristics were collected including ECOG PS at the index date, smoking habits, alcohol consumption, human papilloma virus (HPV) status, HIV status, and medical history. Tumor characteristics, treatment patterns, and outcome measures were recorded.

Study endpoints

The study primary endpoint was OS as defined from the date of diagnosis of R/M HNSCC (index date) to time of death due to any causes. OS was reported separately in predefined subgroups based on baseline characteristics (e.g. HPV status, anatomical site of tumor). Secondary endpoints included descriptive analyses of demographics and clinical characteristics distribution with PD-L1 as well as investigator-assessed ORR, duration of response, and progression-free survival (PFS). ORR (complete response + partial response) was based on Response Evaluation Criteria In Solid Tumors (RECIST) v1.1. PFS was assessed from the start of first-line therapy for R/M disease to progression on or after therapy, or death due to any cause (whichever came first), and from the start of second-line therapy to first documented disease progression or death due to any cause (whichever came first).

Statistical analyses

The sample size to support the primary endpoint was not known a priori and was driven by the number of patients at selected sites with available tissue samples. Based on assumptions of a PD-L1 high prevalence of 25% (TC ≥ 25%), a median OS of 10 months, uniform accrual over 52 months with 10 months’ follow-up from the last patient entering, and exponentially distributed survival times, it was determined post hoc that the study statistics could be powered to the 80% level (two-sided alpha 0.05) to detect a hazard ratio (HR) of 0.7 for PD-L1 high versus low/negative patients for a total of 396 patients and 278 deaths.
Time-to-event endpoints were described using the Kaplan–Meier method. Two-sided 95% confidence intervals (CIs) were provided for the main statistical estimators. OS and PFS were compared between patients with PD-L1 high and low/negative expression for the different cutoffs using a log-rank test at a 5% level of significance. Prognostic value of PD-L1 expression in terms of OS was investigated using a multivariable Cox proportional hazards model where covariates were selected by biological and clinical significance and included age, race, smoking status, alcohol use, metastatic disease, platinum-based therapy, and anatomical site as baseline covariates. Due to the retrospective design of the study, some data were unavailable for collection.

Results

Baseline characteristics

Nineteen sites in seven countries screened 513 patients with R/M HNSCC tumors not amenable to local therapy (e.g. surgery or radiation) or at stage IVC between March 1, 2011 and June 30, 2015. The majority of patients (n = 213; 51.7%) were from the United States, with the remainder from Greece (n = 57; 13.8%), Spain (n = 49; 11.9%), Germany (n = 35; 8.5%), Italy (n = 33; 8.0%), Japan (n = 15; 3.6%), and South Korea (n =10; 2.4%). Of the 513 patients, 412 met all eligibility criteria and comprised the full analysis set; PD-L1 expression was unknown in 16 (3.9%). The 16 patients with unknown PD-L1 expression were not included in prevalence assessments or outcome assessments unless otherwise stated. Most patients (n =400; 97.1%) provided one tissue sample, with 12 patients providing two samples for a total of 424 tissue samples. For patients who provided two samples, PD-L1 expression was determined independently on each sample, and the sample obtained closest to the index date was used to assess PD-L1 expression. Tumor samples were obtained from the primary site in 162/424 cases (38.2%), from recurrent disease in 179/424 cases (42.2%), and from distant sites in 83/424 cases (19.6%).
The median age of patients at or closest to the index date was 62.0 years (range 28.0–93.0; n = 411) (Table 1). There were 132 patients (32.0%) who were found to have TC ≥ 25% PD-L1 expression (Table 1) [2628]. Furthermore, 199 patients (48.3%) and 85 patients (20.6%) had TC ≥ 10% and ≥ 50%, respectively. Among 130 patients with HPV data, 37 were HPV-positive (28.5%). Of the HPV-positive patients, 8 (21.6%) had TC ≥ 25% PD-L1 expression, 17 (45.9%) had TC ≥ 10% PD-L1 expression, and 5 patients (13.5%) had TC ≥ 50% PD-L1 expression.
Table 1
Prevalence of PD-L1 expression based on baseline characteristics and HNSCC tumor characteristics
Characteristic, %
Na
PD-L1 TC ≥ 25% (n = 132)
PD-L1 TC < 25% (n = 264)
Median age, years (range)
 
62.0 (38.0–87.0)
62.0 (28.0–93.0)
  < 60
167
32.9
67.1
  ≥ 60
228
33.8
66.2
Sex
 Male
317
30.9
69.1
 Female
79
43.0
57.0
Race
Caucasian
339
32.7
67.3
 Black or African American
20
30.0
70.0
 Asian
22
50.0
50.0
Region
 United States
205
29.3
70.7
 Asia
22
50.0
50.0
 Europe
169
36.1
63.9
ECOG PS
 0
73
50.7
49.3
 1
87
32.2
67.8
  ≥ 2
41
26.8
73.2
Tobacco use
 Current
97
26.8
73.2
 Former
199
32.2
67.8
 Never
78
42.3
57.7
Alcohol consumption
 Current
148
26.4
73.6
 Former
123
32.5
67.5
HPV status
 Positive
37
21.6
78.4
 Negative
93
25.8
74.2
Timing of tissue sample extraction
 Pre-1st chemotherapy, %
202
30.2
69.8
 Post-1st chemotherapy, %
36
25.0
75.0
Type of tumor sample
 Surgical resection
186
34.9
65.1
 Surgical biopsy
199
32.2
67.8
 Punch biopsy
8
12.5
87.5
Location of tumor sample
 Primary tumor
153
34.0
66.0
 From recurrent disease
175
32.6
67.4
 From metastatic disease
80
33.8
66.3
Primary tumor site
 
132
261
 Oral cavity
108
43.5
56.5
 Oropharynx
61
34.4
65.6
 Hypopharynx
21
9.5
90.5
 Larynx
99
30.3
69.7
 Overlapping lesion
22
22.7
77.3
Stage at index dateb
 Stage 0–III
17
29.4
70.6
 Stage IVA
62
37.1
62.9
 Stage IVB
21
23.8
76.2
 Stage IVC
230
31.3
68.7
Time from diagnosis to index
 Median, months (range)
 
11.4 (0.0–475.9)
14.7 (0.0–349.8)
Sites of new metastases post index date
 Local lymph node
89
31.5
68.5
 Lung
77
27.3
72.7
 Bone
29
37.9
62.1
 Distant lymph node
23
34.8
65.2
 Liver
23
30.4
69.6
 Skin/soft tissue
21
42.9
57.1
 Head and neck
11
27.3
72.7
 Pleura
9
44.4
55.6
aPatients with PD-L1 result N = 396
bIndex date is defined as date of diagnosis of R/M HNSCC not amenable to local therapy
ECOG PS Eastern Cooperative Oncology Group performance status, HNSCC head and neck squamous cell carcinoma, HPV human papilloma virus, mo months, PD-L1 programmed cell death-ligand 1, R/M recurrent and/or metastatic, TC tumor cell
At TC ≥ 25%, the PD-L1 prevalence was higher among females (43.0% vs 30.9% for males), Asians (50.0% vs 32.7% and 30.0% for Caucasians and Black/African Americans, respectively), ECOG PS 0 (50.7% vs 32.2% and 26.8% for 1 and ≥ 2, respectively), and never smokers (42.3% vs 26.8% and 32.2% for current and former smokers, respectively) (Table 1). PD-L1 prevalence decreased with increasing ECOG PS values and was highest in never smokers (compared with current and former smokers) and former alcohol users (vs current).

HNSCC tumor characteristics

The most common sites from which tumor samples were collected were oral cavity (35.0%; n = 143), larynx (33.5%; n = 137), and oropharynx (22.2%; n = 91). Oral cavity tumors (43.5%) showed the highest prevalence of PD-L1 expression (TC ≥ 25%), while the hypopharynx tumors were most often associated with PD-L1 < 25% (90.5%) (Table 1).
The prevalence of PD-L1 expression TC ≥ 25% was similar irrespective of whether the sample was collected from the primary tumor (34.0%), or recurrent (32.6%) or metastatic (33.8%) sites. There was also no difference in prevalence regarding the type of tumor sample used (34.9% in surgical resection vs 32.2% for surgical biopsy) (Table 1).

Treatment history

Among the total cohort of 412 patients, 238 patients (57.8%) received first-line chemotherapy and 84 patients (20.4%) received additional second-line chemotherapy after the index date (Table 2). A limited number of patients received subsequent lines of chemotherapy (n = 42; 10.2%). First-line chemotherapy was administered to 52.3% of patients in the PD-L1 TC ≥ 25% group and 60.2% in the PD-L1 TC < 25% group. Approximately 30% of patients underwent palliative surgical interventions and another ~ 30% underwent radiotherapy. The most common first-line targeted therapy was cetuximab (49.6%), and chemotherapy treatments were cisplatin (44.7%), 5-fluorouracil (36.5%), carboplatin (31.6%), paclitaxel (25.2%), and docetaxel (16.2%) (Table 2). The rates of prior first-line treatment with cetuximab and platinum-based therapy were similar for patients in either PD-L1 cohort. The most common second-line targeted therapy was cetuximab (33.3%), and chemotherapy treatments included paclitaxel (27.8%), carboplatin (22.2%), docetaxel (20.0%), and 5-fluorouracil (11.1%), again with no differences between PD-L1 expression cohorts (Table 2).
Table 2
Treatment history
Treatment history, n (%)
PD-L1 TC ≥ 25%
(n = 132a)
PD-L1 TC < 25%
(n = 264a)
Total (N = 412)
Palliative surgical interventions
44 (33.3)
74 (28.0)
123 (29.9)
Radiotherapy
43 (32.6)
63 (23.9)
113 (27.4)
Chemoradiation therapy
1 (0.8)
0 (0.0)
1 (0.2)
Line of chemotherapy, n
132
264
412
 1st
69 (52.3)
159 (60.2)
238 (57.8)
 2nd
24 (18.2)
55 (20.8)
84 (20.4)
  ≥ 3rd
8 (6.1)
28 (10.6)
42 (10.2)
Type of first-line chemotherapy, n
77
177
266
 Cetuximab
38 (49.4)
91 (51.4)
132 (49.6)
 Cisplatin
41 (53.2)
71 (40.1)
119 (44.7)
 Carboplatin
15 (19.5)
64 (36.2)
84 (31.6)
 Paclitaxel
10 (13.0)
56 (31.6)
67 (25.2)
 Docetaxel
18 (23.4)
22 (12.4)
43 (16.2)
 5-Fluorouracil
35 (45.5)
56 (31.6)
97 (36.5)
Type of second-line chemotherapy
25
60
90
 Cetuximab
8 (32.0)
21 (35.0)
30 (33.3)
 Cisplatin
1 (4.0)
4 (6.7)
5 (5.6)
 Carboplatin
6 (24.0)
13 (21.7)
20 (22.2)
 Paclitaxel
5 (20.0)
20 (33.3)
25 (27.8)
 Docetaxel
7 (28.0)
10 (16.7)
18 (20.0)
 5-Fluorouracil
5 (20.0)
5 (8.3)
10 (11.1)
aPatients with PD-L1 result N = 396
PD-L1 programmed cell death-ligand 1, TC tumor cell

Treatment outcomes

A total of 290 (70.4%) patients died during the study period. Median OS from the index date of R/M disease was 9.6 months (95% CI 8.3–10.8). Among the patients with known PD-L1 expression, OS did not differ significantly for PD-L1 TC ≥ 25% versus TC < 25% (median 8.2 vs 10.1 months, P = 0.55; Fig. 1a). This was also true for PD-L1 expression cutoffs of TC ≥ 10% versus TC < 10% (median 9.6 vs 9.4 months, P = 0.32; Fig. 1b) and TC ≥ 50% versus TC < 50% (median 7.9 vs 10.0 months, P = 0.39; Fig. 1c). Among the 130 patients with available HPV status, median OS was 10 months (95% CI 5.1–16.9) in patients with HPV-positive status and 8.3 months (95% CI 5.8–12.5) in those with HPV-negative status. There was no association of HPV status with PD-L1 expression.
The estimated median OS was 8.0 months (95% CI 6.3–10.0) in patients with oral cavity primary tumor site (n =143), 10.4 months (95% CI 6.9–14.9) in oropharynx (n =91), 12.5 months (95% CI 8.9–14.8) in larynx (n =137), 12.2 months (95% CI 5.7–21.0) in hypopharynx (n =27), and 4.0 months (95% CI 3.3–14.7) in patients with overlapping regions (n =11). The OS for patients with oral cavity tumors was numerically lower in the PD-L1 TC ≥ 25% population than in the PD-L1 TC < 25% population (median 6.9 months vs 9.7 months; log-rank test; P = 0.15). Similarly, for oropharyngeal primary site patients, those in the PD-L1 TC ≥ 25% population had a median OS of 6.3 months versus 14.8 months for patients in the PD-L1 TC < 25% population (log-rank test; P = 0.03) (Fig. 1d). In contrast, numerically longer survival was seen in the PD-L1 TC ≥ 25% population than in the PD-L1 TC < 25% population with hypopharyngeal primary tumors (median 21 months vs 12.2 months; log-rank test; P = 0.35).
Median PFS from the start of first- and second-line chemotherapy was 4.6 months (95% CI 4.0–5.0) and 2.8 months (95% CI 1.9–4.4), respectively. The PFS from the start of first-line chemotherapy did not differ significantly among patients with TC ≥ 25% PD-L1 expression versus TC < 25% (median: 4.2 vs 4.8 months, P = 0.37) (Fig. 2a). This was similar when TC ≥ 10% PD-L1 expression versus TC < 10% and TC ≥ 50% PD-L1 expression versus TC < 50% cutoff values were applied (median 4.4 vs 4.9 months, P = 0.544 and median 4.8 vs 4.5 months, P = 0.557, respectively). However, median PFS from the start of second-line chemotherapy was significantly different between patients with TC ≥ 25% PD-L1 (n = 25) expression versus those with TC < 25% (n = 58) (4.1 months vs 2.2 months, P = 0.04). The difference was also significant for patients with TC ≥ 10% PD-L1 (n = 38) expression versus those with TC < 10% (n = 45) (4.1 vs 2.1 months, P = 0.04) and those patients with TC ≥ 50% PD-L1 (n = 13) expression versus those with TC < 50% (n = 70) (6.3 vs 2.4 months, P = 0.03). However, these results must be weighed against the small sample size and lack of adjustment for any confounding factors (Fig. 2b). Validation in a larger cohort of patients is required.
Among the 98 patients who had a tumor response, according to RECIST, after treatment with first-line chemotherapy, ORR was 43.9% (95% CI 33.9–54.3). Patients with PD-L1-high expressing tumors (TC ≥ 25%) had an ORR of 40.0% (95% CI 21.1–61.3, n = 25) and those with TC < 25% had an ORR of 44.3% (95% CI 32.4–56.7, n = 70) (Table 3). Among the 30 patients treated with second-line chemotherapy who had a tumor response evaluated, the ORR was 13.3% (95% CI 3.8–30.7). The ORR observed for the TC ≥ 25% cohort was 20.0% (2/10 patients; 95% CI 2.5–55.6) and those with TC < 25% had an ORR of 5.6% (1/18 patients; 95% CI 0.1–27.3) (Table 3).
Table 3
Response and survival by PD-L1 expression
Endpoint, na (%)
PD-L1 TC ≥ 25%
PD-L1 TC < 25%
PD-L1 TC ≥ 10%
PD-L1 TC < 10%
PD-L1 TC ≥ 50%
PD-L1 TC < 50%
From diagnosis date to death
 Median OS, months (range)
8.2 (6.3–10.6)
10.1 (8.3–12.2)
9.6 (7.5–12.5)
9.4 (7.9–11.4)
7.9 (5.5–12.4)
10.0 (8.5–11.8)
 log-rank P value, PD-L1 high vs PD-L1 low/negative
0.55
0.32
0.39
From first-line therapy
 Number evaluable
25
70
    
 ORRb, n (%)
 Overall response rate
10 (40.0)
31 (44.3)
    
 Complete response
0 (0.0)
1 (1.4)
    
 Partial response
10 (40.0)
30 (42.9)
    
 Duration of response, n
8
24
    
 Median, weeks (range)
10.6 (0.1–28.7)
15.3 (1.7–52.3)
    
 PFS, n
73
169
110
132
41
201
Median, months (range)
4.2 (2.6–4.8)
4.8 (3.9–5.8)
4.4 (3.3–4.9)
4.9 (3.9–6.0)
4.8 (3.2–6.1)
4.5 (3.9–5.0)
log-rank P value, PD-L1 high vs PD-L1 low/negative
0.37
0.54
0.56
From second-line therapy
 Number evaluable
10
18
    
 ORRb, n (%)
 Overall response rate
2 (20.0)
1 (5.6)
    
 Complete response
0 (0.0)
0 (0.0)
    
 Partial response
2 (20.0)
1 (5.6)
    
 Duration of response, n
2
1
    
 Median, weeks (range)
10.6 (5.9–15.4)
1.3 (1.3–1.3)
    
 PFS, n
25
58
38
45
13
70
 Median, months (range)
4.1 (2.8–7.1)
2.2 (1.6–4.0)
4.1 (2.2–6.5)
2.1 (1.6–3.6)
6.3 (1.2–13.8)
2.4 (1.6–3.8)
 log-rank P value, PD-L1 high vs PD-L1 low/negative
0.04
0.04
0.03
aPatients with PD-L1 result N = 396
bORR measured by RESIST
ORR objective response rate, OS overall survival, PD-L1 programmed cell death-ligand 1, PFS progression-free survival, RECIST Response Evaluation Criteria In Solid Tumors

Multivariable risk factor analyses

PD-L1 expression TC ≥ 25%, was not identified as a significant predictor of risk of death, with an HR of 1.04 (95% CI 0.79–1.37; P = 0.79), nor were cutoffs TC ≥ 10% and TC ≥ 50% (HR 0.86; 95% CI 0.67–1.11; P = 0.25 and HR 1.14; 95% CI 0.83–1.56; P = 0.42, respectively) (Fig. 3; Table 4). Metastatic disease at the time of index date was associated with increased risk of death, whereas age ≥ 60 years, platinum-based therapy, and anatomic subsite of larynx were associated with a lower risk of death regardless of the PD-L1 cutoff used (Fig. 3; Table 4).
Table 4
Multivariable analysis of risk factors for analyses examining PFS or OS for all-comers
Category
PFS from start of first-line therapy (n = 253)
PFS from start of second-line therapy (n = 88)
OS from index date (n = 370)
HR
95% CI
P value
HR
95% CI
P value
HR
95% CI
P value
PD-L1 expression high vs lowa
1.09
(0.78–1.52)
0.63
0.58
(0.30–1.13)
0.11
1.04
(0.79–1.37)
0.790
Age < 60 vs ≥ 60 years
0.89
(0.67–1.18)
0.40
0.82
(0.45–1.47)
0.50
0.71
(0.55–0.92)
0.008
Race Caucasian vs other
0.59
(0.38–0.9)
0.02
0.61
(0.30–1.23)
0.16
0.92
(0.62–1.37)
0.689
Nonsmoker vs current/former smoker
0.81
(0.55–1.21)
0.31
0.97
(0.47–2.01)
0.93
1.20
(0.85–1.69)
0.292
Heavy alcohol use, no vs yes
0.97
(0.57–1.66)
0.90
1.11
(0.39–3.14)
0.85
1.33
(0.80–2.19)
0.268
Metastatic disease, no vs yes
1.27
(0.96–1.68)
0.10
0.59
(0.34–1.03)
0.06
1.42
(1.10–1.84)
0.008
Platinum-based therapy, no vs yes
1.10
(0.68–1.79)
0.70
3.084
(0.64–14.81)
0.16
0.70
(0.52–0.94)
0.018
Anatomical site vs oral cavity
 Oropharynx
0.89
(0.59–1.35)
0.58
0.88
(0.39–1.99)
0.75
0.74
(0.52–1.05)
0.089
 Hypopharynx
0.65
(0.36–1.17)
0.15
0.72
(0.30–1.75)
0.47
0.62
(0.36–1.05)
0.076
 Larynx
0.74
(0.51–1.06)
0.10
0.88
(0.44–1.76)
0.72
0.63
(0.46–0.86)
0.003
 Overlapping lesion
1.05
(0.47–2.33)
0.91
0.82
(0.16–4.10)
0.81
1.25
(0.65–2.38)
0.503
Statistically significant P values are in italics
aPatients with PD-L1 result N = 396
CI confidence interval, HR hazard ratio, OS overall survival, PD-L1 programmed cell death ligand-1, PFS progression-free survival

Discussion

In this study, we investigated if PD-L1 expression was associated with survival in patients treated with standard chemotherapy.
In the entire population of this study, PD-L1 was not prognostic for survival in patients with HNSCC who received standard chemotherapy regimens. This finding was consistent with observations in randomized controlled trials of similar patients with R/M HNSCC [10, 26, 29]. In CheckMate 141, for patients treated with investigator’s choice the median OS in PD-L1 TC ≥ 1% was slightly lower than in PD-L1 TC < 1% [4.6 months (95% CI 3.8–5.8) vs 5.8 months (95% CI 4.0–9.8)] [30]. In KEYNOTE-040 the survival of patients treated with investigator's choice of standard of care (methotrexate, docetaxel, or cetuximab) did not increase with increasing PD-L1 expression [12]. Similar results have also been observed in an evaluation of commercially obtained patient samples with stage I–IV HNSCC, in which PD-L1 expression was not prognostic for OS based on a TC ≥ 25% cutoff [31].
Currently accepted prognostic markers in HNSCC include HPV status in patients with oropharyngeal carcinoma and smoking status [32]. Other researchers have identified prognostic factors including age, race, ECOG PS, prior treatments [33], C-reactive protein, leukocyte levels, and time from diagnosis to relapse [34]. In a multivariable analysis of the SUPREME-HN study we found age, platinum therapy, primary tumor location, and metastatic disease to be associated with survival. It is not surprising that metastatic disease is associated with poorer survival, this variable has been incorporated in prognostic models of survival in advanced cancers [35]. Similarly, patients healthy enough to tolerate a platinum-based therapy might be expected to survive longer. The observation here of improved survival in older patients (≥ 60 years) compared with younger patients is somewhat counterintuitive; it is generally considered that older adults have comparable survival outcomes but with increased toxicity [36]. However, a non-significantly higher survival in patients > 65 years versus < 65 years has also been shown in patients treated with investigator's choice in a retrospective analysis of CheckMate 141 [37]. In both the SUPREME-HN and the CheckMate 141 studies, investigator's choice of standard of care was used. It is possible that elderly patients were treated with taxanes, rather than cisplatin and cetuximab, due to the higher toxicities associated with the latter therapies. Later publications have indicated that docetaxel improves OS over cisplatin [38]. One could speculate that investigators selected therapies for older patients based on the toxicity profile, which were later demonstrated to be more efficacious. Urba identified race (Caucasian vs other) as prognostic for OS and PFS. In the SUPREME-HN study an association was observed that was only significant for PFS from first-line therapy; possibly because there was a smaller non-Caucasian population in this study. In a univariate analysis, Urba identified primary tumor location as negatively prognostic for survival (oral cavity vs “other”, HR 1.37, 95% CI 1.15–1.63, P = 0.01) and associated with reduced PFS [33]. In the multivariable analysis of the SUPREME-HN study, patients with primary tumor locations of oropharynx and hypopharynx had improved OS compared with patients with oral cavity carcinoma and survival was significantly longer in patients with tumors in laryngeal versus oral cavity sites (HR 0.63, 95% CI 0.46–0.86, P = 0.003). Currently smoking and HPV status are considered to be major independent prognostic factors in patients with oropharyngeal cancer [32] and recent HNSCC randomized clinical trial studies have been stratified using PD-L1 and HPV, smoking status, and performance status [39]. The SUPREME-HN study shows meaningful survival differences by primary tumor location, raising the question whether site of tumor origin should also be considered in study design and patient treatment.
The PD-L1 prevalence at TC ≥ 25% was consistent across biopsy locations: 32.1% (primary tumor), 31.8% (recurrent site), and 32.5% (metastatic site). These data suggest that any tumor lesion can be used for PD-L1 testing for HNSCC, although in this study the primary and metastatic lesions were not from the same patient. Additionally, PD-L1 expression seems to be stable across the primary versus metastatic setting, only the punch biopsy gave lower PD-L1 expression.
The prevalence of PD-L1 varied according to a number of other factors; gender (higher in females), race, region, ECOG PS 0, oral cavity cancers, and never smokers. High PD-L1 prevalence has previously been significantly associated with females, never smokers, and oral cavity in other studies of second-line patients with HNSCC [23]. The PD-L1 TC ≥ 25% prevalence varied substantially depending on the primary tumor location; from 43.5% in oral cavity to 9.5% in hypopharyngeal (see Table 1). The median OS for patients with oral cavity carcinoma was lower in PD-L1 TC ≥ 25% than PD-L1 TC < 25% patients; poor prognosis in PD-L1 TC ≥ 25% oral cavity patients has been observed by others [18]. Likewise, for oropharyngeal primary site patients, median OS in patients with PD-L1 TC ≥ 25% was less than that seen for patients with PD-L1 TC < 25% (log-rank test; P = 0.03; Fig. 1d). Conversely, longer survival was seen in PD-L1 TC ≥ 25% than PD-L1 TC < 25% patients with hypopharyngeal primary tumors (21 months vs 12.2 months). These data indicate that for patients with tumors of oral cavity and oropharyngeal origin, PD-L1 expression is linked to shorter survival, whereas those with PD-L1 high hypopharyngeal primary tumors live longer.
Therefore, although PD-L1 was not prognostic in the entire SUPREME-HN cohort, our data indicate PD-L1 can be both positively and negatively prognostic depending on the primary tumor location. This finding may help to explain historical conflicting views of the prognostic value of PD-L1; for example, the finding that PD-L1 expression was positively prognostic in laryngeal squamous cell carcinoma [22] but conversely associated with poor prognosis in oral squamous cell carcinoma [17].
Study limitations pertain mainly to the retrospective study design, and hence, the reliance of available information in medical charts. Quantitative analyses of risk factors were limited due to missing information on performance status, HPV status, and small sample size. This study used an assay validated for PD-L1 expression on TCs and did not investigate the prognostic value of IC PD-L1 expression. PD-L1 expression in other cellular compartments of the tumor microenvironment may be indicative of survival. The variety of scoring methods used for determining PD-L1 positivity (TCs and/or ICs) may also contribute to the apparent contradictory publications regarding its prognostic value.
Tumor stage and grade at initial diagnosis were not available for all patients since they may have received initial care in a hospital other than the investigating site. Furthermore, the definition of R/M status may have led to the exclusion of patients who received local therapies for palliative purposes, as the treatment intent was not always mentioned in the patient’s medical records. Additionally, evaluations of tumor response and progression were not evaluated via blinded, independent committee review as would be the case in clinical trials, which can lead to some variability in results. PD-L1 expression was assessed using available tissue that was not necessarily obtained at the time of initial diagnosis or at the same stage of disease for all patients. Findings from additional exploratory analyses suggest that PD-L1 expression was lower in tissue samples obtained after a patient’s prior exposure to chemotherapy than prior to initiation of chemotherapy, irrespective of tissue origin (primary tumor, recurrent site, or metastatic site). A similar finding was observed for the subset of samples from the primary tumor obtained after exposure to radiotherapy.
Since starting the SUPREME-HN study a number of immunotherapies have been approved for use in R/M HNSCC. The approvals of PD-L1 assays as companion diagnostics demonstrates the predictive nature and the value of this biomarker. As the use of immunotherapy increases the opportunity diminishes to perform a prospective study in patients treated with non-immune based treatments and thus SUPREME-HN represents a unique historical record of the prognostic value of PD-L1.

Conclusion

There have been conflicting results reported regarding the prognostic value of PD-L1 expression on TCs. Early reports did not demonstrate any association between PD-L1 expression and OS, whereas other studies have suggested that PD-L1 expression may be associated with improved survival [1723]. In the SUPREME-HN study, PD-L1 expression using TCs at cutoff values of 10%, 25%, and 50% was not prognostic for survival in patients with HNSCC treated with standard therapies; however, PD-L1 expression may be positively or negatively prognostic when anatomic subsites within the head and neck are considered.
In evaluating the correlation of PD-L1 and survival, previous studies did not always account for confounding factors. Based on our analysis these factors, specifically HPV status, primary tumor location, and demographic factors, may be highly relevant to OS in patients with R/M HNSCC.

Acknowledgements

The authors would like to thank the patients and their caregivers for their participation in this study. The authors also thank Armida Lefranc Torres for his role as research coordinator at Massachusetts Eye and Ear Infirmary/Massachusetts General Hospital. Medical writing support, which was in accordance with Good Publication Practice (GPP3) guidelines, was provided by Jubilee Stewart, PhD, and Edwin Thrower, PhD, of Parexel (Hackensack, NJ) and was funded by AstraZeneca.
The results of this study have been presented at the ASCO 2017 Congress, June 3–6, Chicago, IL, USA (interim analysis data) and at the ESMO 2017 Congress, September 8–12, Madrid, Spain (final study data).
This trial was performed in accordance with ethical principles consistent with the Declaration of Helsinki, International Conference on Harmonisation (ICH) guidelines, International Society for Pharmacoepidemiology (ISPE) (2007) Guidelines for Good Pharmacoepidemiology Practices (GPP) and applicable legislation. The Investigator(s) have performed this trial in accordance with the regulations and guidelines governing medical practice and ethics in the countries of the trial and in accordance with currently acceptable techniques and know-how. The final protocol of this trial, including the final version of the subject or next of kin/legal representative ICF, was approved or given a favorable opinion in writing by the Ethics Committee/Institutional Review Board (IRB)/Independent Ethics Committee (IEC). The Ethics Committee/IRB/IEC also approved any amendments to the protocol and all communication to patient or next of kin, according to local regulations.
Not applicable.

Competing interests

SIP has served as a consultant or in an advisory role for AbbVie, AstraZeneca/MedImmune, Cue, EMD Serono, Merck, Newlink Genetics, Oncolys, Replimune, and Sensei; has received research funding for Abbvie, AstraZeneca/MedImmune, Cue, Merck, Tesaro; and received compensation for travel, accommodations and/or expenses from AbbVie, AstraZeneca/MedImmune, EMD Serono, Newlink Genetics, Oncolys, and Sensei. EEWC has been a consultant or held an advisory role for AstraZeneca, Bristol-Myers Squibb, EMD Serono, Human Longevity, Inc, Merck, Pfizer. GF has received honoraria from AstraZeneca; and served as a consutant or in an advisory role for Boehringer Ingelheim, Celgene, and Lilly. ESK has received honoraria from AstraZeneca, Boehringer Ingelheim, Celgene, and Lilly; has served as a consutant or in an advisory role for AstraZeneca, Boehringer Ingelheim, Celgene, and Lilly; and received compensation for travel, accommodations and or expenses from AstraZeneca, Boehringer Ingelheim, Celgene, and Lilly. NB has received honoraria from Merck Serono, MSD, AstraZeneca, and BMS; has served as a consutant or in an advisory role for BMS, Merck Serono, and Nanobiotix. DC has received research funding from AbbVie. MS is an employee of Evidera and provided epidemiological support to AZ in the development of this manuscript. HW, PT-A, SW, AK, GM, and NS are employees of AstraZeneca and may hold stock or other ownership in AstraZeneca. The other authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Monnerat C, Faivre S, Temam S, Bourhis J, Raymond E. End points for new agents in induction chemotherapy for locally advanced head and neck cancers. Ann Oncol. 2002;13(7):995–1006.CrossRef Monnerat C, Faivre S, Temam S, Bourhis J, Raymond E. End points for new agents in induction chemotherapy for locally advanced head and neck cancers. Ann Oncol. 2002;13(7):995–1006.CrossRef
2.
Zurück zum Zitat Vermorken JB, Specenier P. Optimal treatment for recurrent/metastatic head and neck cancer. Ann Oncol. 2010;21(Suppl 7):vii252–61.PubMed Vermorken JB, Specenier P. Optimal treatment for recurrent/metastatic head and neck cancer. Ann Oncol. 2010;21(Suppl 7):vii252–61.PubMed
3.
Zurück zum Zitat Zenda S, Onozawa Y, Boku N, Iida Y, Ebihara M, Onitsuka T. Single-agent docetaxel in patients with platinum-refractory metastatic or recurrent squamous cell carcinoma of the head and neck (SCCHN). Jpn J Clin Oncol. 2007;37(7):477–81.CrossRef Zenda S, Onozawa Y, Boku N, Iida Y, Ebihara M, Onitsuka T. Single-agent docetaxel in patients with platinum-refractory metastatic or recurrent squamous cell carcinoma of the head and neck (SCCHN). Jpn J Clin Oncol. 2007;37(7):477–81.CrossRef
4.
Zurück zum Zitat Leon X, Hitt R, Constenla M, Rocca A, Stupp R, Kovacs AF, et al. A retrospective analysis of the outcome of patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck refractory to a platinum-based chemotherapy. Clin Oncol (R Coll Radiol). 2005;17(6):418–24.CrossRef Leon X, Hitt R, Constenla M, Rocca A, Stupp R, Kovacs AF, et al. A retrospective analysis of the outcome of patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck refractory to a platinum-based chemotherapy. Clin Oncol (R Coll Radiol). 2005;17(6):418–24.CrossRef
5.
Zurück zum Zitat Gregoire V, Lefebvre JL, Licitra L, Felip E. Squamous cell carcinoma of the head and neck EHNS-ESMO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v184–6.CrossRef Gregoire V, Lefebvre JL, Licitra L, Felip E. Squamous cell carcinoma of the head and neck EHNS-ESMO-ESTRO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v184–6.CrossRef
7.
Zurück zum Zitat Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359(11):1116–27.CrossRef Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecki A, Rottey S, et al. Platinum-based chemotherapy plus cetuximab in head and neck cancer. N Engl J Med. 2008;359(11):1116–27.CrossRef
8.
Zurück zum Zitat Stewart JS, Cohen EE, Licitra L, Van Herpen CM, Khorprasert C, Soulieres D, et al. Phase III study of gefitinib compared with intravenous methotrexate for recurrent squamous cell carcinoma of the head and neck [corrected]. J Clin Oncol. 2009;27(11):1864–71.CrossRef Stewart JS, Cohen EE, Licitra L, Van Herpen CM, Khorprasert C, Soulieres D, et al. Phase III study of gefitinib compared with intravenous methotrexate for recurrent squamous cell carcinoma of the head and neck [corrected]. J Clin Oncol. 2009;27(11):1864–71.CrossRef
9.
Zurück zum Zitat Vermorken JB, Trigo J, Hitt R, Koralewski P, Diaz-Rubio E, Rolland F, et al. Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy. J Clin Oncol. 2007;25(16):2171–7.CrossRef Vermorken JB, Trigo J, Hitt R, Koralewski P, Diaz-Rubio E, Rolland F, et al. Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy. J Clin Oncol. 2007;25(16):2171–7.CrossRef
10.
Zurück zum Zitat Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, et al. Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study. J Clin Oncol. 2017;35(14):1542–9.CrossRef Bauml J, Seiwert TY, Pfister DG, Worden F, Liu SV, Gilbert J, et al. Pembrolizumab for platinum- and cetuximab-refractory head and neck cancer: results from a single-arm, phase II study. J Clin Oncol. 2017;35(14):1542–9.CrossRef
11.
Zurück zum Zitat Chow LQ, Haddad R, Gupta S, Mahipal A, Mehra R, Tahara M, et al. Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J Clin Oncol. 2016;34(32):3838–45.CrossRef Chow LQ, Haddad R, Gupta S, Mahipal A, Mehra R, Tahara M, et al. Antitumor activity of pembrolizumab in biomarker-unselected patients with recurrent and/or metastatic head and neck squamous cell carcinoma: results from the phase Ib KEYNOTE-012 expansion cohort. J Clin Oncol. 2016;34(32):3838–45.CrossRef
12.
Zurück zum Zitat Cohen EEW, Soulières D, Le Tourneau C, Dinis J, Licitra L, Ahn M-J, et al. Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study. Lancet. 2019;393(10167):156–67.CrossRef Cohen EEW, Soulières D, Le Tourneau C, Dinis J, Licitra L, Ahn M-J, et al. Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): a randomised, open-label, phase 3 study. Lancet. 2019;393(10167):156–67.CrossRef
13.
Zurück zum Zitat Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856–67.CrossRef Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856–67.CrossRef
15.
Zurück zum Zitat Kim JW, Eder JP. Prospects for targeting PD-1 and PD-L1 in various tumor types. Oncology. 2014;28(Suppl 3):15–28.PubMed Kim JW, Eder JP. Prospects for targeting PD-1 and PD-L1 in various tumor types. Oncology. 2014;28(Suppl 3):15–28.PubMed
16.
Zurück zum Zitat Feldman R, Gatalica Z, Knezetic J, Reddy S, Nathan CA, Javadi N, et al. Molecular profiling of head and neck squamous cell carcinoma. Head Neck. 2016;38(Suppl 1):E1625–38.CrossRef Feldman R, Gatalica Z, Knezetic J, Reddy S, Nathan CA, Javadi N, et al. Molecular profiling of head and neck squamous cell carcinoma. Head Neck. 2016;38(Suppl 1):E1625–38.CrossRef
17.
Zurück zum Zitat Lin YM, Sung WM, Hsieh MJ, Tsai SC, Lai HW, Yang SM, et al. High PD-L1 expression correlates with metastasis and poor prognosis in oral squamous cell carcinoma. PLoS ONE. 2015;10(11):e0142656.CrossRef Lin YM, Sung WM, Hsieh MJ, Tsai SC, Lai HW, Yang SM, et al. High PD-L1 expression correlates with metastasis and poor prognosis in oral squamous cell carcinoma. PLoS ONE. 2015;10(11):e0142656.CrossRef
18.
Zurück zum Zitat Ock CY, Kim S, Keam B, Kim M, Kim TM, Kim JH, et al. PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget. 2016;7(13):15901–14.CrossRef Ock CY, Kim S, Keam B, Kim M, Kim TM, Kim JH, et al. PD-L1 expression is associated with epithelial-mesenchymal transition in head and neck squamous cell carcinoma. Oncotarget. 2016;7(13):15901–14.CrossRef
19.
Zurück zum Zitat Oliveira-Costa JP, de Carvalho AF, da Silveira GG, Amaya P, Wu Y, Park KJ, et al. Gene expression patterns through oral squamous cell carcinoma development: PD-L1 expression in primary tumor and circulating tumor cells. Oncotarget. 2015;6(25):20902–20.CrossRef Oliveira-Costa JP, de Carvalho AF, da Silveira GG, Amaya P, Wu Y, Park KJ, et al. Gene expression patterns through oral squamous cell carcinoma development: PD-L1 expression in primary tumor and circulating tumor cells. Oncotarget. 2015;6(25):20902–20.CrossRef
20.
Zurück zum Zitat Pai S, Cohen E, Lin D, Fountzilas G, Kim ES, Mehlhorn H. A retrospective cohort study of PD-L1 expression in recurrent and/or metastatic squamous cell carcinoma of the head and neck (SUPREME-HN). J Clin Oncol. 2017;35(15 Suppl):6040.CrossRef Pai S, Cohen E, Lin D, Fountzilas G, Kim ES, Mehlhorn H. A retrospective cohort study of PD-L1 expression in recurrent and/or metastatic squamous cell carcinoma of the head and neck (SUPREME-HN). J Clin Oncol. 2017;35(15 Suppl):6040.CrossRef
21.
Zurück zum Zitat Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–56.CrossRef Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–56.CrossRef
22.
Zurück zum Zitat Vassilakopoulou M, Avgeris M, Velcheti V, Kotoula V, Rampias T, Chatzopoulos K, et al. Evaluation of PD-L1 expression and associated tumor-infiltrating lymphocytes in laryngeal squamous cell carcinoma. Clin Cancer Res. 2016;22(3):704–13.CrossRef Vassilakopoulou M, Avgeris M, Velcheti V, Kotoula V, Rampias T, Chatzopoulos K, et al. Evaluation of PD-L1 expression and associated tumor-infiltrating lymphocytes in laryngeal squamous cell carcinoma. Clin Cancer Res. 2016;22(3):704–13.CrossRef
23.
Zurück zum Zitat Wildsmith S, Scott M, Midha A, Barker C, Whiteley J, Ratcliffe M, et al. PD-L1 expression in patients screened for phase 2 head and neck squamous cell carcinoma clinical studies (HAWK and CONDOR). Presented at: American Association for Cancer Research (AACR); April 14–18, 2018; Chicago, US. Poster 5530. Wildsmith S, Scott M, Midha A, Barker C, Whiteley J, Ratcliffe M, et al. PD-L1 expression in patients screened for phase 2 head and neck squamous cell carcinoma clinical studies (HAWK and CONDOR). Presented at: American Association for Cancer Research (AACR); April 14–18, 2018; Chicago, US. Poster 5530.
24.
Zurück zum Zitat Pai S, Cohen EE, Lin D, Fountzilas G, Kim ES, Mehlhorn H, et al. RetroSpective cohort stUdy of PD-L1 expression in REcurrent and/or MEtastatic squamous cell carcinoma of the head and neck (SUPREME-HN). Ann Oncol. 2017;28(Suppl 5):v372–94. Pai S, Cohen EE, Lin D, Fountzilas G, Kim ES, Mehlhorn H, et al. RetroSpective cohort stUdy of PD-L1 expression in REcurrent and/or MEtastatic squamous cell carcinoma of the head and neck (SUPREME-HN). Ann Oncol. 2017;28(Suppl 5):v372–94.
25.
Zurück zum Zitat Rebelatto MC, Midha A, Mistry A, Sabalos C, Schechter N, Li X, et al. Development of a programmed cell death ligand-1 immunohistochemical assay validated for analysis of non-small cell lung cancer and head and neck squamous cell carcinoma. Diagn Pathol. 2016;11(1):95.CrossRef Rebelatto MC, Midha A, Mistry A, Sabalos C, Schechter N, Li X, et al. Development of a programmed cell death ligand-1 immunohistochemical assay validated for analysis of non-small cell lung cancer and head and neck squamous cell carcinoma. Diagn Pathol. 2016;11(1):95.CrossRef
26.
Zurück zum Zitat Licitra LF, Haddad R, Even C, Tahara M, Dvorkin M, Ciuleanu T-E, et al. EAGLE: a phase 3, randomized, open-label study of durvalumab (D) with or without tremelimumab (T) in patients with recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC). Presented at: American Society of Clinical Oncology (ASCO); May 31–June 4, 2019; Chicago, IL. Poster 6012. Licitra LF, Haddad R, Even C, Tahara M, Dvorkin M, Ciuleanu T-E, et al. EAGLE: a phase 3, randomized, open-label study of durvalumab (D) with or without tremelimumab (T) in patients with recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC). Presented at: American Society of Clinical Oncology (ASCO); May 31–June 4, 2019; Chicago, IL. Poster 6012.
27.
Zurück zum Zitat Siu LL, Even C, Mesia R, Remenar E, Daste A, Delord JP, et al. Safety and efficacy of durvalumab with or without tremelimumab in patients with PD-L1-low/negative recurrent or metastatic HNSCC: the phase 2 CONDOR randomized clinical trial. JAMA Oncol. 2019;5(2):195–203.CrossRef Siu LL, Even C, Mesia R, Remenar E, Daste A, Delord JP, et al. Safety and efficacy of durvalumab with or without tremelimumab in patients with PD-L1-low/negative recurrent or metastatic HNSCC: the phase 2 CONDOR randomized clinical trial. JAMA Oncol. 2019;5(2):195–203.CrossRef
28.
Zurück zum Zitat Zandberg DP, Algazi A, Jimeno A, Good JS, Fayette J, Bouganim N, et al. Durvalumab for recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC): preliminary results from a single-arm, phase 2 study (HAWK). Ann Oncol. 2017;28(Suppl 5):372–94 (Abstract 1042O). Zandberg DP, Algazi A, Jimeno A, Good JS, Fayette J, Bouganim N, et al. Durvalumab for recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC): preliminary results from a single-arm, phase 2 study (HAWK). Ann Oncol. 2017;28(Suppl 5):372–94 (Abstract 1042O).
29.
Zurück zum Zitat Rischin D. Protocol-specified final analysis of the phase 3 KEYNOTE-048 trial of pembrolizumab (pembro) as first-line therapy for recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). Presented at: American Society of Cancer Oncology (ASCO); May 31–June 4, 2019; Chicago, IL. Abstract 6000. https://doi.org/10.1093/annonc/mdx374 Rischin D. Protocol-specified final analysis of the phase 3 KEYNOTE-048 trial of pembrolizumab (pembro) as first-line therapy for recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). Presented at: American Society of Cancer Oncology (ASCO); May 31–June 4, 2019; Chicago, IL. Abstract 6000. https://​doi.​org/​10.​1093/​annonc/​mdx374
30.
Zurück zum Zitat Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, 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.CrossRef Ferris RL, Blumenschein G Jr, Fayette J, Guigay J, Colevas AD, Licitra L, 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.CrossRef
32.
Zurück zum Zitat Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24–35.CrossRef Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-Tan PF, et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010;363(1):24–35.CrossRef
33.
Zurück zum Zitat Urba S, Gatz J, Shen W, Hossain A, Winfree K, Koustenis A, et al. Quality of life scores as prognostic factors of overall survival in advanced head and neck cancer: analysis of a phase III randomized trial of pemetrexed plus cisplatin versus cisplatin monotherapy. Oral Oncol. 2012;48(8):723–9.CrossRef Urba S, Gatz J, Shen W, Hossain A, Winfree K, Koustenis A, et al. Quality of life scores as prognostic factors of overall survival in advanced head and neck cancer: analysis of a phase III randomized trial of pemetrexed plus cisplatin versus cisplatin monotherapy. Oral Oncol. 2012;48(8):723–9.CrossRef
34.
Zurück zum Zitat Magnes T, Melchardt T, Weiss L, Mittermair C, Neureiter D, Klieser E, et al. Prognostic score in patients with recurrent or metastatic carcinoma of the head and neck treated with cetuximab and chemotherapy. PLoS ONE. 2017;12(7):e0180995.CrossRef Magnes T, Melchardt T, Weiss L, Mittermair C, Neureiter D, Klieser E, et al. Prognostic score in patients with recurrent or metastatic carcinoma of the head and neck treated with cetuximab and chemotherapy. PLoS ONE. 2017;12(7):e0180995.CrossRef
35.
Zurück zum Zitat Arkenau HT, Barriuso J, Olmos D, Ang JE, de Bono J, Judson I, et al. Prospective validation of a prognostic score to improve patient selection for oncology phase I trials. J Clin Oncol. 2009;27(16):2692–6.CrossRef Arkenau HT, Barriuso J, Olmos D, Ang JE, de Bono J, Judson I, et al. Prospective validation of a prognostic score to improve patient selection for oncology phase I trials. J Clin Oncol. 2009;27(16):2692–6.CrossRef
36.
Zurück zum Zitat VanderWalde NA, Fleming M, Weiss J, Chera BS. Treatment of older patients with head and neck cancer: a review. Oncologist. 2013;18(5):568–78.CrossRef VanderWalde NA, Fleming M, Weiss J, Chera BS. Treatment of older patients with head and neck cancer: a review. Oncologist. 2013;18(5):568–78.CrossRef
37.
Zurück zum Zitat Saba NF, Blumenschein G Jr, Guigay J, Licitra L, Fayette J, Harrington KJ, et al. Nivolumab versus investigator’s choice in patients with recurrent or metastatic squamous cell carcinoma of the head and neck: efficacy and safety in CheckMate 141 by age. Oral Oncol. 2019;96:7–14.CrossRef Saba NF, Blumenschein G Jr, Guigay J, Licitra L, Fayette J, Harrington KJ, et al. Nivolumab versus investigator’s choice in patients with recurrent or metastatic squamous cell carcinoma of the head and neck: efficacy and safety in CheckMate 141 by age. Oral Oncol. 2019;96:7–14.CrossRef
38.
Zurück zum Zitat Harari PM, Harris J, Kies MS, Myers JN, Jordan RC, Gillison ML, et al. Postoperative chemoradiotherapy and cetuximab for high-risk squamous cell carcinoma of the head and neck: radiation Therapy Oncology Group RTOG-0234. J Clin Oncol. 2014;32(23):2486–95.CrossRef Harari PM, Harris J, Kies MS, Myers JN, Jordan RC, Gillison ML, et al. Postoperative chemoradiotherapy and cetuximab for high-risk squamous cell carcinoma of the head and neck: radiation Therapy Oncology Group RTOG-0234. J Clin Oncol. 2014;32(23):2486–95.CrossRef
39.
Zurück zum Zitat Soulières D, Cohen EEW, Le Tourneau C, Dinis J, Licitra L, Ahn MJ, et al. Updated survival results of the KEYNOTE-040 study of pembrolizumab vs standard-of-care chemotherapy for recurrent or metastatic head and neck squamous cell carcinoma. Presented at: American Association for Cancer Research; April 14–18, 2018; Chicago, IL. Abstract CT115. Soulières D, Cohen EEW, Le Tourneau C, Dinis J, Licitra L, Ahn MJ, et al. Updated survival results of the KEYNOTE-040 study of pembrolizumab vs standard-of-care chemotherapy for recurrent or metastatic head and neck squamous cell carcinoma. Presented at: American Association for Cancer Research; April 14–18, 2018; Chicago, IL. Abstract CT115.
Metadaten
Titel
SUPREME-HN: a retrospective biomarker study assessing the prognostic value of PD-L1 expression in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck
verfasst von
Sara I. Pai
Ezra E. W. Cohen
Derrick Lin
George Fountzilas
Edward S. Kim
Holger Mehlhorn
Neus Baste
Daniel Clayburgh
Loren Lipworth
Carlo Resteghini
Nawar Shara
Takashi Fujii
Jun Zhang
Michael Stokes
Huifen Wang
Philip Twumasi-Ankrah
Sophie Wildsmith
Asud Khaliq
Giovanni Melillo
Norah Shire
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2019
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-019-02182-1

Weitere Artikel der Ausgabe 1/2019

Journal of Translational Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.