ArticlePDF Available

Prognostic significance and immune escape implication of tumor-infiltrating neutrophil plasticity in human head and neck squamous cell carcinoma

Authors:

Abstract

Tumor-infiltrating neutrophils play a crucial role in the progression of head and neck squamous cell carcinoma (HNSCC). Here, we aimed to statistically quantify the plasticity of HNSCC-infiltrating N2/N1 neutrophils and examine its impacts on survival and immune infiltration landscape. A retrospective study of 80 patients who underwent curative surgical resection for HNSCC between 2014 and 2017 was conducted in this study. HNSCC-infiltrating neutrophil phenotypes were classified using immunofluorescence staining, and the N2/N1 neutrophil plasticity was evaluated via the ratio of N2/N1 neutrophils. We then assessed the correlations between N2/N1 neutrophil plasticity, clinicopathological characteristics, and immune infiltration landscape using rigorous statistical methods. Infiltration variations of N1 and N2 neutrophils were observed between the tumor nest (TN) and tumor stroma (TS), with TN exhibiting higher N2 neutrophil infiltration and lower N1 neutrophil infiltration. High ratios of N2/N1 neutrophils were correlated with advanced TNM stage, large tumor size and invasion of adjacent tissue. High infiltration of N2 neutrophils was associated with decreased overall and relapse-free survival, which were opposite for N1 neutrophils. The independent prognostic role of N2/N1 neutrophil plasticity, particularly within the TN region, was confirmed by multivariate analyses. Moreover, the ratio of N2/N1 neutrophils within the TN region showed correlations with high CD8+ T cells infiltration and low FOXP3+ Tregs infiltration. We identify HNSCC-infiltrating N2/N1 neutrophil plasticity as a crucial prognostic indictor which potentially reflects the tumor microenvironment (TME) and immune escape landscape within HNSCC tissues. Further investigations and validations may provide novel therapeutic strategies for personalized immunomodulation in HNSCC patients.
Vol.:(0123456789)
Human Cell
https://doi.org/10.1007/s13577-024-01024-7
RESEARCH ARTICLE
Prognostic significance andimmune escape implication
oftumor‑infiltrating neutrophil plasticity inhuman head andneck
squamous cell carcinoma
XiaokeZhu1 · YuHeng1· DuoZhang1· DiTang1· JianZhou1· HanqingLin1· JingyuMa1· XupingDing2· LeiTao1·
LimingLu2
Received: 20 August 2023 / Accepted: 27 December 2023
© The Author(s) under exclusive licence to Japan Human Cell Society 2024
Abstract
Tumor-infiltrating neutrophils play a crucial role in the progression of head and neck squamous cell carcinoma (HNSCC).
Here, we aimed to statistically quantify the plasticity of HNSCC-infiltrating N2/N1 neutrophils and examine its impacts on
survival and immune infiltration landscape. A retrospective study of 80 patients who underwent curative surgical resection
for HNSCC between 2014 and 2017 was conducted in this study. HNSCC-infiltrating neutrophil phenotypes were classified
using immunofluorescence staining, and the N2/N1 neutrophil plasticity was evaluated via the ratio of N2/N1 neutrophils. We
then assessed the correlations between N2/N1 neutrophil plasticity, clinicopathological characteristics, and immune infiltra-
tion landscape using rigorous statistical methods. Infiltration variations of N1 and N2 neutrophils were observed between
the tumor nest (TN) and tumor stroma (TS), with TN exhibiting higher N2 neutrophil infiltration and lower N1 neutrophil
infiltration. High ratios of N2/N1 neutrophils were correlated with advanced TNM stage, large tumor size and invasion of
adjacent tissue. High infiltration of N2 neutrophils was associated with decreased overall and relapse-free survival, which
were opposite for N1 neutrophils. The independent prognostic role of N2/N1 neutrophil plasticity, particularly within the
TN region, was confirmed by multivariate analyses. Moreover, the ratio of N2/N1 neutrophils within the TN region showed
correlations with high CD8+ T cells infiltration and low FOXP3+ Tregs infiltration. We identify HNSCC-infiltrating N2/
N1 neutrophil plasticity as a crucial prognostic indictor which potentially reflects the tumor microenvironment (TME) and
immune escape landscape within HNSCC tissues. Further investigations and validations may provide novel therapeutic
strategies for personalized immunomodulation in HNSCC patients.
Keywords Head and neck squamous cell carcinoma· Tumor-infiltrating neutrophils· Neutrophil plasticity· Prognoses·
Immune infiltration landscape· Immunosuppressive microenvironment
Abbreviations
ANT Adjacent normal tissues
CI Confidence interval
H&E Haematoxylin & eosin
HGF Hepatocyte growth factor
HPV Human papillomavirus
HR Hazard ratio
IF Immunofluorescence
IHC Immunohistochemistry
MMP9 Matrix metalloproteinase 9
NETs Neutrophil extracellular traps
OS Overall survival
PGE2 Prostaglandin E2
RFS Relapse-free survival
TANs Tumor-associated neutrophils
TME Tumor microenvironment
Xiaoke Zhu and Yu Heng contributed equally to this work.
* Lei Tao
doctortaolei@163.com; lei.tao@fdeent.org
* Liming Lu
lulunew2003@163.com; youlanda2009@sjtu.edu.cn
1 Department ofOtolaryngology, Shanghai Key Clinical
Disciplines ofOtorhinolaryngology, Eye Ear Nose
andThroat Hospital, Fudan University, 83 Fenyang Road,
Shanghai200031, People’sRepublicofChina
2 Department ofImmunology andMicrobiology,
Shanghai Institute ofImmunology, Shanghai Jiao Tong
University School ofMedicine, Shanghai200025,
People’sRepublicofChina
X.Zhu et al.
TN Tumor nest
TS Tumor stroma
HNSCC Head and neck squamous cell carcinoma
TMA Tissue microarray
Introduction
Head and neck squamous cell carcinomas (HNSCC) repre-
sent the sixth most common malignancy worldwide, encom-
passing cancers of oral cavity, pharynx and larynx [1]. The
pathogenesis of HNSCC can be attributed to a range of etio-
logical factors, including alcohol abuse, tobacco consump-
tion, and infection with human papillomavirus (HPV) [2,
3]. Although significant advancements have been made in
the diagnosis and treatment of HNSCC, the 5-year overall
survival (OS) rate for locally advanced HNSCC remains
suboptimal at approximately 50%, with even poorer progno-
ses observed in HPV-negative HNSCC cases [4, 5]. Recent
studies have elucidated the complex tumor microenviron-
ment (TME), which comprises a heterogeneous infiltrating
immune cell and stromal cell compartment that interacts
with neighboring tumor cells [68]. These interactions
are crucial for the growth, progression, and metastasis of
HNSCC [6].
HNSCC is recognized for its immunogenic nature, and
several studies have reported favorable outcomes of immu-
notherapy using immune checkpoint inhibitors (e.g., anti-
PD1/PDL1 blockade) in the treatment of a particular sub-
set of HNSCC patients [9, 10]. Nevertheless, the response
rate to anti-PD1 immunotherapy remains relatively low
at approximately 20%, potentially due to the variation in
immune cell infiltration landscape among patients [5, 6].
This underscores the pressing need to identify specific
HNSCC-infiltrating cell subtypes correlated with prognoses
and develop multimode therapeutic strategies for remodel-
ling the HNSCC TME.
Accumulating evidences highlight the crucial role of
tumor-infiltrating macrophages as a key immune cell popu-
lation within the TME, exhibiting significant plasticity in
both M2-polarized pro-tumoral and M1-polarized antitu-
moral phenotypes [11, 12]. As another critical immune cell
population within the TME, tumor-associated neutrophils
(TANs) also substantially contribute greatly to the HNSCC
microenvironment, with both peripheral neutrophils and
TANs implicated in tumor progression [1315]. Despite
this, the precision role of TANs remains to be elucidated;
some studies have associated TANs with favorable outcomes
[16], while others have linked them to poor prognosis [7,
17]. TANs can exert antitumoral function by modulating
IL12 production in macrophages and orchestrating antitumor
immunity [16]. Conversely, TANs may also facilitate tumor
progression and metastasis through directly promoting
tumor cell proliferation, angiogenesis, immunosuppres-
sion, and extracellular matrix remodeling and extravasation
[1719]. Therefore, the neutrophil diversity and plasticity
of tumors may undertake the two-sided functions of TANs
within the TME.
In a manner akin to tumor-infiltrating macrophages,
Fridlender etal. [20] reported that TANs can be polarized
into two distinctive phenotypes: pro-tumoral N2 neutro-
phils and antitumoral N1 neutrophils. N2 neutrophils pro-
mote tumor cell proliferation and contribute to angiogen-
esis through the secretion of cytokines such as neutrophil
elastase, prostaglandin E2 (PGE2), and matrix metallopro-
teinase 9 (MMP9) [21]. They also induce immunosuppres-
sion by upregulating the expression of immune checkpoints
(e.g., PD-L1 and VISTA) [7, 19], and recruiting immuno-
suppressive cells into the TME (e.g., regulatory T cells) [22].
Although TANs have been shown to play a prognostic role
in tumors [7], the plasticity and implications of HNSCC-
infiltrating N2/N1 neutrophils remains uncertain.
In this study, we aimed to investigate the plasticity of
HNSCC-infiltrating N2/N1 neutrophils within the TME and
explore its correlation with the infiltration of other immune
cells. Moreover, we sought to determine the prognostic role
of HNSCC-infiltrating N2/N1 neutrophils. To our knowl-
edge, this is the first study to evaluate the prognostic impli-
cations of the plasticity of HNSCC-infiltrating N2/N1 neu-
trophils in HNSCC patients undergoing curative resection.
Material andmethods
Study population andtissue microarrays
In this study, we recruited a cohort of HNSCC patients at our
medical center who underwent curable surgical treatments
between June 2014 and September 2017. The inclusion and
exclusion criteria for candidates were stringent, requiring
a de novo pathohistological diagnosis of HNSCC without
prior treatment and the absence of distant metastasis. This
study adhered to the principles of the Declaration of Helsinki
and received approval from the Medical Research Council
of the Eye & ENT Hospital, Fudan University, Shanghai,
China (No. KJ2008-01). Furthermore, participation in the
study was consented by all patients.
To examine the plasticity of HNSCC-infiltrating N2/
N1 neutrophils within the TME, tissue microarray (TMA)
samples were collected from HNSCC patients, consisting
of 80 pairs of tissue specimens, including HNSCC tissues
and adjacent normal tissues (ANT). The HNSCC TMAs
were serially sliced, with each TMA section contain-
ing 4-μm-thick tissues. Our study collected the following
clinical and pathological data: age, sex, smoking and drink-
ing history, history of vocal leukoplakia, primary subsite,
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
pathological grade, tumor size, clinical stage, metastatic
lymph node status, and follow-up data.
Immunohistochemistry andimmunofluorescence
Haematoxylin & eosin (H&E), immunohistochemistry (IHC)
staining and immunofluorescence (IF) staining were per-
formed on formalin-fixed and paraffin-embedded sections of
TMA, as previously describe [7, 11]. H&E staining was uti-
lized to evaluate routine histological features and delineate
the tumor stroma (TS) and tumor nest (TN) areas (Supple-
mentary Fig.1), which were confirmed by two board-certi-
fied pathologists. Following the manufacturer’s instructions,
IHC staining was performed using the specified antibod-
ies: anti-CD3 (Abcam, ab16669, 1:150), anti-CD4 (Abcam,
ab133616, 1:500), anti-CD8(Abcam, ab217344, 1:2000),
anti-FOXP3(Abcam, ab20034, 1:500). Three high-power
fields (HPF) of version at 400× magnification were used to
estimate the average number of cells per HPF.
For IF staining, the primary antibodies used were anti-
CD66b (BioLegend, 305102, clone G10F5, 1:100), anti-
CD206 (Abcam, ab252921, 1:1000), anti-iNOS (Abcam,
ab283655, 1:200) monoclonal antibodies, which were
incubated at 4 overnight. Subsequently, TMA sections
were incubated in the dark for 60min with Cy3 (Abcam,
ab97035, 1:200), Alexa Fluor 488 (Abcam, ab150077,
1:200), and Cy5 (Abcam, ab6564, 1:200). Finally, DAPI
(Solarbio, C0060) was used for a 20-min incubation. The
results were visualized using a fluorescence microscope
(Olympus). CD66b+CD206+iNOS neutrophils and a few
CD66b+CD206+iNOS+ neutrophils were defined as HNSCC-
infiltrating N2 neutrophils, while CD66b+CD206iNOS+
neutrophils and a few CD66b+CD206iNOS neutrophils
were defined as HNSCC-infiltrating N1 neutrophils (Sup-
plementary Fig.2) [23]. The average numbers of N2 and N1
neutrophils per HPF in the TN, TS, combined tumor area
and ANT, respectively, were counted across three separate
400× HPF.
Study outcomes
The primary outcome of this study was overall survival
(OS), calculated from the date of curative surgery to all-
cause mortality or the last follow-up. The secondary out-
come was relapse-free survival (RFS), defined as the interval
from surgery to any disease recurrence (local, regional, or
distant).
Statistical analysis
All statistical analyses were performed using GraphPad
version 7.0 and SPSS software version 24.0. Categorical
variables were presented as frequency (percentage), and
continuous variables were presented as median (range). The
optimal cutoff values of the immune cells were determined
using the X-tile software, with the cutoff value of N2/N1
plasticity set at 1. Chi-square (χ2) tests were employed to
evaluate the associations between HNSCC-infiltrating N2/
N1 neutrophils and clinicopathological characteristics, while
Spearman’s correlation analyses were performed to assess
the relationships between two continuous variables. Survival
was presented using the Kaplan–Meier curves and compared
using the log-rank tests. Univariate and multivariate analyses
were conducted to assess the independent prognostic role of
variables. Sensitivity analyses were performed by restricting
cases to patients diagnosed with laryngeal SCC and those
who were older than 65years. Statistical significance was
deemed when the p value < 0.05.
Results
Patient characteristics
All patients included in this study were male, with a signifi-
cant proportion having a history of smoking (n = 56, 70%)
and drinking (n = 43, 53.8%). The majority of patients had
tumors originating from the larynx (n = 61, 76.3%), while
the remainder had tumors from the hypopharynx (n = 19,
23.8%). A total of 62 (77.5%) patients presented with
advanced T stage (i.e., T3–4), while 41 (51.2%) had adverse
N stage (i.e., N1–2). The detailed demographic and clinico-
pathological features of the cases were presented in Table1.
HNSCC‑infiltrating N2/N1 neutrophil plasticity
intheTME ofHNSCC
According to the indication of H&E staining, the num-
ber of CD66b+CD206+iNOS N2 neutrophils and
CD66b+CD206iNOS+ N1 neutrophils in the TN, TS, com-
bined tumor area and ANT were calculated, respectively
(Fig.1A, Supplementary Fig.2) [23]. Our study observed
a higher median number of N1 neutrophils (8 [range, 1–31]
in HNSCC vs. 4 [range, 0–13] in ANT) and N2 neutrophils
(4 [range, 0–41] vs. 0 [range, 0–4]) in HNSCC tissue than in
the ANT, which were confirmed by both unpaired and paired
analyses (Supplementary Fig.3, Supplementary Table1).
The ratio of HNSCC-infiltrating N2/N1 neutrophils was also
higher in HNSCC tissue (0.5 [range, 0–4.2]) than that of
ANT (0, [range 0–2]).
The count of HNSCC-infiltrating N1 neutrophils was higher
than that of N2 neutrophils in the TS region (p < 0.0001) and
combine tumor area (p = 0.0023), while the number of two
neutrophil phenotypes were comparable in the TN (p = 0.8411)
as indicated by both unpaired and paired analyses (Fig.1B–D).
The number of N1 neutrophils was highest in the TS, while
X.Zhu et al.
the number of N2 neutrophils and the ratio of N2/N1 neu-
trophils were highest in the TN (Fig.1E–G). The number of
total CD66b+ neutrophils were comparable in the TN and TS
regions (p = 0.5421, Fig.1H).
Correlations betweentheplasticity ofN2/N1
neutrophils andclinicopathological features
The cutoff values of the HNSCC-infiltrating N1 and N2 neu-
trophils in the TN were determined as 3 and 2, respectively.
This resulted in the categorization of 35 cases into the TN high
HNSCC-infiltrating N1 subgroup and 43 patients into the TN
high HNSCC-infiltrating N2 subgroup. Notably, high infiltra-
tion of HNSCC-infiltrating N1 neutrophils in the TN was sig-
nificantly related to primary tumor subsite (p = 0.022), T stage
(p = 0.026), as well as history of diabetes (p = 0.037) but not
with other features (Supplementary Table2). Conversely, the
high infiltration of HNSCC-infiltrating N2 phenotype in TN
was significantly related only to smoking history (p = 0.016)
and N stage (p = 0.026, Supplementary Table3). In the TS
region, the determined cutoff values of the HNSCC-infiltrat-
ing N2 and N1 phenotypes were 1 and 5, respectively. This
resulted in 39 patients being classified as TS high HNSCC-
infiltrating N1 subgroup and 41 patients as TS high HNSCC-
infiltrating N2 subgroup. No significant correlations were
identified between high infiltration of TS N1 neutrophils
and other factors, while the high infiltration of TS N2 neu-
trophils displayed a notable correlation with smoking history
(p = 0.036, Supplementary Table2,3). In terms of combined
tumor area, the threshold values of the HNSCC-infiltrating
N1 and N2 phenotypes were 7 and 4, respectively. Correlation
analyses revealed that high infiltration of N2 neutrophils in
combined tumor area was significantly related solely to smok-
ing history (p = 0.032, Supplementary Table2,3).
Furthermore, the HNSCC-infiltrating N2/N1 plasticity
was assessed by the N2/N1 ratio to account for variations
between N2 and N1 neutrophils. Patients with an N2/N1
ratio exceeding 1 were classified into the high N2/N1 ratio
subgroup. Correlation analyses (Table2) revealed that a high
N2/N1 ratio within the TN was significantly associated with
smoking history (p = 0.009), T stage (p = 0.004), N stage
(p = 0.005), TNM stage (p = 0.011), tumor size (p = 0.033),
and invasion of adjacent tissue (p = 0.006). A high N2/N1
ratio within the TS was significantly related only to N stage
(p = 0.024), and a high ratio of N2/N1 phenotypes in the
combined tumor area was associated with smoking history
(p = 0.034), primary site (p = 0.031), and N stage (p = 0.006).
Relationship betweentheinfiltration ofN2/N1
neutrophils andother immune cells inHNSCC tissue
The TMA sections were additionally stained with CD3, CD4,
CD8 and FOXP4 to gain a more profound understanding
Table 1 Demographic and clinicopathological characteristics of the
patients with HNSCC
HNSCC head and neck squamous cell carcinoma, LNs lymph nodes,
Tre g regulatory T cell
Variables
Total, n 80
Age at diagnosis, median (range) 63 (46–80)
Smoking history, n (%)
No 24 (30%)
Yes 56 (70%)
Drinking history, n (%)
No 37 (46.2%)
Yes 43 (53.8%)
Diabetes, n (%)
No 69 (86.3%)
Yes 11 (13.2%)
Hypertension, n (%)
No 57 (71.3%)
Yes 23 (28.7%)
History of vocal leukoplakia, n (%)
No 77 (96.3%)
Yes 3 (3.7%)
Primary site, n (%)
Larynx 61 (76.3%)
Hypopharynx 19 (23.8%)
T stage, n (%)
T1–2 18 (22.5%)
T3–4 62 (77.5%)
N stage, n (%)
N0 39 (48.8%)
N1–2 41 (51.2%)
TNM stage, n (%)
I–II 11 (13.8%)
III 69 (86.2%)
Tumor size, n (%)
≤ 3.5cm 40 (50.0%)
> 3.5cm 40 (50.0%)
Differentiation, n (%)
Well or moderately 71 (88.8%)
Poorly 9 (11.3%)
Number of metastatic LNs, n (%)
0 39 (48.8%)
1 14 (17.4%)
> 1 27 (33.8%)
Invasion of adjacent tissue, n (%)
No 25 (31.3%)
Yes 55 (68.2%)
HNSCC-infiltrating CD3+ T cells, median (range) 100 (7–301)
HNSCC-infiltrating CD4+ T cells, median (range) 76 (5–223)
HNSCC-infiltrating CD8+ T cells, median (range) 18 (1–151)
HNSCC-infiltrating FOXP3+ Treg cells, median
(range)
8 (0–167)
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
N1 N2
0
5
10
15
20
25
Cell count / HPF
N1 N2
0
10
20
30
40
Cell count / HPF
ns
N1 N2
0
5
10
15
20
25
Cell count / HPF
N1 N2
0
10
20
30
40
Cell count / HPF
ns
Tumor
S
trom
a
Tu
m
o
r N
est
N1 N2
0
10
20
30
40
50
Cell count / HPF
N1 N2
0
10
20
30
40
50
Cell count / HPF
C
o
m
b
in
ed
T
u
m
o
r Ar
ea
TN TS ANT
0
5
10
15
20
25
Cell count / HPF
TN TS ANT
0
5
10
15
20
25
Cell count / HPF
TN TS ANT
0
10
20
30
40
Cell count / HPF
TN TS ANT
0
10
20
30
40
Cell count / HPF
TN TS ANT
0
10
20
30
40
50
Cell count / HPF
ns
TN TS ANT
0
10
20
30
40
50
Cell count / HPF
ns
TN TS ANT
0
2
4
6
8
10
N2/N1 Ratio
TN TS ANT
0
2
4
6
8
10
N2/N1 Ratio
N1
N
2
Total Neutrophils
N2
/
N1 Ratio
B
C
D
E
G
F
H
50 50 50
50 50 50
50 50 50
50 50 50
50 50 50
200 200 200 200 200 200
A
Fig. 1 Identification of the HNSCC-infiltrating N2/N1 neutro-
phils plasticity. Representative immunofluorescence staining (A) of
HNSCC-infiltrating N2 neutrophils (yellow arrow) and N1 neutro-
phils (white arrow); paired and unpaired comparisons of the counts of
HNSCC-infiltrating N2/N1 neutrophils in TN (B), TS (C), and com-
bined tumor area (D). Paired and unpaired comparisons of HNSCC-
infiltrating N1 neutrophil counts (E), N2 neutrophil counts (F), N2/
N1 ratios (G) and total neutrophil counts (H) within different regions
(TN vs. TS vs. ANT). ****p < 0.0001, ***p < 0.001, **p < 0.01, ns
p > 0.05. TN Tumor nest, TS tumor stroma, ANT adjacent normal tis-
sue, HPF high-power field
X.Zhu et al.
Table 2 Associations between HNSCC-infiltrating N2/N1 neutrophils ratio and clinicopathological features among patients with HNSCC
Supplementary Table2 and 3 show the associations between HNSCC-infiltrating N2 or N1 neutrophils and clinicopathological features
HNSCC head and neck squamous cell carcinoma, LNs lymph nodes, Treg regulatory T cell
Bold = P value meets definition for statistical significance
Variables Tumor nest Tumor stroma Combined tumor area
Low N2/N1 High N2/N1 p value Low N2/N1 High N2/N1 p value Low N2/N1 High N2/N1 p value
Age at diagnosis 0.388 0.266 0.716
< 65/ ≥ 65 28/25 17/10 39/27 6/8 35/26 10/9
Smoking history 0.009 0.158 0.034
No/yes 21/32 3/24 22/44 2/12 22/39 2/17
Drinking history 0.098 0.384 0.346
No/yes 28/25 9/18 32/34 5/9 30/31 7/12
Diabetes 0.625 0.429 0.768
No/yes 45/8 24/3 56/10 13/1 53/8 16/3
Hypertension 0.690 0.188 0.396
No/yes 37/16 20/7 45/21 12/2 42/19 15/4
History of vocal leukoplakia 0.988 0.416 0.691
No/yes 51/2 26/1 63/3 14/0 59/2 18/1
Primary site 0.151 0.822 0.031
Larynx/Hypopharynx 43/10 18/9 50/16 11/3 50/11 11/8
T stage 0.004 0.916 0.152
T1–2/T3–4 17/36 1/26 15/51 3/11 16/45 2/17
N stage 0.005 0.024 0.006
N0/N1–2 32/21 7/29 36/30 3/11 35/26 4/15
TNM stage 0.011 0.429 0.219
I–II/III 11/42 0.27 10/56 1/13 10/51 1/18
Tumor size (cm) 0.033 0.556 0.431
≤ 3.5/ > 3.5 31/22 9/18 34/32 6/8 32/29 8/11
Differentiation 0.471 0.692 0.909
Well or moderately/poorly 48/5 23/4 50/7 12/2 54/7 17/2
Number of metastatic LNs 0.492 0.169 0.387
0/1/ > 1 29/8/16 11/5/11 36/9/21 4/4/6 32/8/21 8/5/6
Invasion of adjacent tissue 0.006 0.812 0.096
No/yes 22/31 3/24 21/45 4/10 22/39 3/16
Tumor-associated CD66b+
neutrophils
0.699 0.208 0.079
Low/high 24/29 11/16 31/35 4/10 30/31 5/14
HNSCC-infiltrating CD3+ T
cells
0.237 0.239 0.793
Low/high 24/29 16/11 35/31 5/9 31/30 9/10
HNSCC-infiltrating CD4+ T
cells
0.582 0.014 0.361
Low/high 26/27 15/12 38/28 3/11 33/28 8/11
HNSCC-infiltrating CD8+ T
cells
0.001 0.941 0.005
Low/high 23/30 22/5 37/29 8/6 29/32 16/3
HNSCC-infiltrating FOXP3+
Tregs
0.030 0.222 0.120
Low/high 35/18 11/16 40/26 6/8 38/23 8/11
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
of immune cytolysis and suppression mechanisms within
HNSCC tissue. The median number in the TMA was 100
(range 7–301) for CD3+ cells, 76 (range 5–223) for CD4+
T cells, 18 (range 1–151) for CD8+ T cell, and 8 (range
0–167) for FOXP3+ Treg cells (Table1). The optimal cutoff
values were determined as 100 for CD3+ cells, 80 for CD4+
T cells, 20 for CD8+ T cells, and 15 for FOXP3+ Tregs.
Interestingly, a higher number of CD8+ T cell was corre-
lated with improved survival (OS: p < 0.0001, HR 0.115,
95% IC: 0.034–0.384; RFS: p = 0.0111, HR 0.338, 95% CI
0.140–0.817). In contrast, elevated infiltration of FOXP3+
Tregs was associated with worse survival (OS: p = 0.0019,
HR 3.502, 95% IC: 1.528–8.023; RFS: p = 0.0051, HR
3.538, 95% CI 1.530–8.182, Fig.2, Supplementary Table4).
However, no correlations were observed between survival
and CD4+ T cells or CD3+ T cells (Fig.2, Supplementary
Table4).
High infiltration of HNSCC-infiltrating N2 phenotype,
especially within TN, was associated with low CD8+ T
cells infiltration and high FOXP3+ Tregs infiltration (Sup-
plementary Fig.4), while no significant correlations were
identified for HNSCC-infiltrating N1 phenotype (Supple-
mentary Fig.5). Moreover, a high N2/N1 ratio in the TN
was significantly related to low CD8+ T cells infiltration
(p = 0.001) and high FOXP3+ Tregs infiltration (p = 0.030),
as evidenced by χ2 tests (Table2) and further confirmed
through unpaired t tests (Fig.3) and Spearman’s correla-
tion analyses (Fig.3). In addition, our study evaluated the
correlation between HNSCC-infiltrating N2/N1 neutrophil
plasticity and the ratios of CD8/Treg, CD8/CD3 and Treg/
CD3, which were all significantly associated with survival
(Supplementary Fig.6–8). These intriguing findings sug-
gest that a high N2/N1 ratio may be related to lower CD8/
CD3 ratio, lower CD8/Treg ratio and higher Treg/CD3
ratio, with more significant associations observed within
the TN (Supplementary Fig.6–8). These correlations may
potentially indicate that HNSCC-infiltrating N2/N1 neu-
trophil plasticity reflects the immune escape landscape of
the TME.
Absent Present
CD8
Treg
CD3
CD4
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low CD8
High CD8
P < 0.0001
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low Treg
High Treg
P = 0.0019
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low CD4
High CD4
P = 0.4902
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low CD3
High CD3
P = 0.1189
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low CD8
High CD8
P = 0.0111
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low Treg
High Treg
P = 0.0051
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low CD4
High CD4
P = 0.8383
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low CD3
High CD3
P = 0.5060
CD
8
reg
C
D
4
CD
3
BA
C
D
E
50 50
50 50
50 50
50 50
Fig. 2 Representative immunohistochemistry staining for CD8+
T cells, Tregs, CD4+ T cells and CD3+ T cells (A). Kaplan–Meier
curves for OS and RFS by the infiltration variation of CD8+ T cells
(B), Tregs (C), CD4+ T cells (D) and CD3+ T cells (E). RFS relapse-
free survival, OS overall survival
X.Zhu et al.
0246810
0
50
100
150
200
Ratio of N2/N1
Count of CD8+ T cells / HPF
Pearson r = -0.2827
P value = 0.0111*
0246
0
50
100
150
200
Ratio of N2/N1
Count of CD8+ T cells / HPF
Pearson r = 0.0234
P value = 0.8368
012345
0
50
100
150
200
Ratio of N2/N1
Count of CD8+ T cells / HPF
Pearson r = -0.1668
P value = 0.1391
0246810
0
50
100
150
200
Ratio of N2/N1
Count of FOXP3+ Treg cells / HPF
Pearson r = 0.2253
P value = 0.0445*
0246
0
50
100
150
200
Ratio of N2/N1
Count of FOXP3+ Treg cells / HPF
Pearson r = 0.2023
P value = 0.0719
012345
0
50
100
150
200
Ratio of N2/N1
Count of FOXP3+ Treg cells / HPF
Pearson r = 0.2187
P value = 0.0498
0246810
0
50
100
150
200
Ratio of N2/N1
Count of CD4+ T cells / HPF
Pearson r = 0.0452
P value = 06905
0246
0
100
200
300
Ratio of N2/N1
Count of CD4+ T cells / HPF
Pearson r = 0.2064
P value = 00662
012345
0
100
200
300
Ratio of N2/N1
Count of CD4+ T cells / HPF
Pearson r = 0.1276
P value = 02593
0246810
0
50
100
150
200
Ratio of N2/N1
Count of CD3+ T cells / HPF
Pearson r = -0.0322
P value = 07769
0246
0
100
200
300
Ratio of N2/N1
Count of CD3+ T cells / HPF
Pearson r = 0.1577
P value = 01624
012345
0
100
200
300
Ratio of N2/N1
Count of CD3+ T cells / HPF
Pearson r = 0.0650
P value = 05668
T
umor nes
t
T
umor stroma
Combined tumor area
0
50
100
150
200
0
50
100
150
200
ns
0
50
100
150
200
0
50
100
150
200
0
50
100
150
200 ns
0
50
100
150
200
0
50
100
150
200
250
ns
0
50
100
150
200
250
0
50
100
150
200
250
ns
0
100
200
300
400
ns
0
100
200
300
400
ns
0
100
200
300
400 ns
CD
8
T
re
g
C
D
4
CD
3
C
D8 Tre
g
C
D
4
C
D
3
C
D
8
Treg
C
D
4
CD
3
T
u
m
o
r n
est
T
umor stroma
L
ow N2
/
N1
H
i
g
h N2/N
1
Low N2/N1
High N2/N
1
Low N2/N
1
Hi
g
h N2/N
1
Combined tumor are
a
A
B
C
D
E
F
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
Independent prognostic role ofHNSCC‑infiltrating
N2/N1 neutrophils inHNSCC
The Kaplan–Meier plots (Fig.4) and univariate analyses
(Fig.5) revealed that a higher infiltration of HNSCC-
infiltrating N1 neutrophils within the TN was signifi-
cantly associated with improved OS (HR, 0.262; 95%
CI 0.105–0.651; p = 0.004) and RFS (HR, 0.335; 95%
CI 0.140–0.800; p = 0.014). Conversely, worse OS and
RFS were observed in patients with a higher infiltration
of HNSCC-infiltrating N2 neutrophils in TN (OS: HR,
2.454; 95% CI 1.071–5.623; p = 0.034; RFS: HR, 1.906;
95% CI 0.848–4.284; p = 0.119). When assessing sur-
vival based on N2/N1 ratio, significant distinctions were
noticed between high and low N2/N1 subgroups within the
TN (OS: HR, 10.470; 95% CI 4.157–26.369; p < 0.0001;
RFS: HR, 5.092; 95% CI 2.261–11.469; p < 0.0001)
and in the combined tumor area (OS: HR, 6.211; 95%
CI 2.846–13.554; p < 0.0001; RFS: HR, 4.541; 95% CI
2.080–9.914; p < 0.0001).
The multivariate analyses (Fig.5, Supplementary
Table5–7) demonstrated that after adjusted for prognostic
risk factors (i.e., tumor size, T stage, N stage, TNM stage,
HNSCC-infiltrating CD8+ T cells and HNSCC-infiltrating
FOXP3+ Treg cells, Supplementary Table4), low infiltra-
tion of HNSCC-infiltrating N1 neutrophils within the TN
(adjusted HRos, 0.355; 95% CI 0.132–0.950; p = 0.039),
high infiltration of HNSCC-infiltrating N2 neutrophils
within the TN (adjusted HRos, 3.069; 95% CI 1.144–8.233;
p = 0.026), high N2/N1 neutrophils ratio in the TN
(adjusted HRos, 4.458; 95% CI 1.384–14.366; p 0.012)
and in combined tumor area (adjusted HRos, 3.373; 95%
CI 1.269–8.965; p = 0.015) were all verified as independ-
ent prognostic factors for worse OS. Regarding RFS, low
infiltration of HNSCC-infiltrating N1 neutrophils within
the TN (adjusted HRRFS, 0.342; 95% CI 0.132–0.885;
p = 0.027), high N2/N1 neutrophils ratio within the TN
(adjusted HRos, 3.088; 95% CI 1.171–8.140; p = 0.023) and
the combined tumor area (adjusted HRos, 3.350; 95% CI
1.194–9.400; p = 0.022) were considered as independent
prognostic factors for worse RFS (Fig.5, Supplementary
Table5–7), after adjusted for corresponding RFS-related
risk factors (Supplementary Table4). The results of sensi-
tivity analysis were in alignment with our primary findings
(Supplementary Table8, 9).
Discussion
This study first evaluated the plasticity of HNSCC-infiltrat-
ing N2/N1 neutrophils within the TME and their prognos-
tic implications. Our findings support the notion that the
plasticity of HNSCC-infiltrating neutrophils, particularly
the ratio of N2/N1 neutrophils, serves as an independent
prognostic indicator for HNSCC and is related to adverse
clinical and pathological features. Furthermore, the plastic-
ity HNSCC-infiltrating N2/N1 neutrophils, especially within
TN, may potentially reflect the immune escape and immu-
nosuppressive state of TME.
Accumulating evidences suggests that neutrophils play
a pivotal role in modulating tumor onset and progression
[17]. They exhibit antitumoral functions, such as function-
ing as antigen-presenting cells [24], directly killing tumor
cells [25, 26], and mediating antibody-mediated trogoptosis
[27]. Neutrophils are also implicated in promoting tumor
progression by secreting MMP9, BV8 [17], hepatocyte
growth factor (HGF) [28], or by undergoing the process of
NETosis [29]. Consequently, these evidences raise the pos-
sibility of phenotypic plasticity and functional versatility
that dictate their response to TME cues [30, 31]. Various
markers, including CD66b, MPO, CD15 and HE staining,
have been employed to identify HNSCC-infiltrating neu-
trophils. CD66b is indispensable and crucial for neutrophil
transmigration and adhesion and is consistently expressed
in all stages of neutrophil differentiation [32]. Therefore,
our study utilized CD66b staining to identify neutrophils. In
addition, compelling evidences from previous studies have
identified distinct neutrophil phenotypes in cancer patients
[33]. However, markers for N1 and N2 neutrophil pheno-
types are not yet to be determined. Thus, it remains neces-
sary to identify appropriate molecular markers for N1 and
N2 neutrophils, respectively. Recent studies [23, 34] have
reported the effective use of CD206 and iNOS in identi-
fying neutrophil phenotypes, with their effects analogous
to their distinctive properties in macrophages [11]. In our
study, the expressions of CD206 and iNOS were assessed in
tissue-infiltrating CD66b+ neutrophils and were proven to be
suitable for identifying neutrophils with distinct prognostic
functions, indicating the diversity and plasticity of HNSCC-
infiltrating N2/N1 neutrophils within the TME.
Previous researches have demonstrated that tumor cells
could secrete numerous cytokines that facilitate the recruit-
ment of neutrophils, including members of the C-X-C
chemokine family, growth factor, IL-6, and GM-CSF [35,
36]. Furthermore, neutrophils exhibit high plasticity and
have remarkable adaptive abilities in response to differ-
ent TME cues. Fridlender etal. reported that transforming
growth factor-beta (TGF-β) could drive the polarization of
neutrophils toward a protumor phenotype (N2), whereas
Fig. 3 Comparisons of HNSCC-infiltrating CD8+ T cells, Tregs,
CD4+ T cells and CD3+ T cells between low and high ratio of N2/N1
neutrophils in TN (A), TS (B) and combined tumor area (C). Correla-
tions between HNSCC-infiltrating CD8+ T cells, Tregs, CD4+ T cells
and CD3+ T cells and the ratio of N2/N1 neutrophils in TN (D), TS
(E) and combined tumor area (F). TN tumor nest, TS tumor stroma,
HPF high-power field
X.Zhu et al.
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N1
High N1
P = 0.0017
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N1
High N1
P = 0.8830
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N1
High N1
P = 0.1841
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N2
High N2
P = 0.0283
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N2
High N2
P = 0.0548
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N2
High N2
P = 0.0708
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N2/N1
High N2/N1
P < 0.0001
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N2/N1
High N2/N1
P = 0.1114
0102030405060
0
20
40
60
80
100
Months
Overall survival rate, %
Low N2/N1
High N2/N1
P < 0.0001
Tu
m
o
r n
es
t
T
umor stroma
A
B
C
C
ombined tumor are
a
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N1
High N1
P = 0.0126
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N1
High N1
P = 0.8615
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N1
High N1
P = 0.2990
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N2
High N2
P = 0.1033
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N2
High N2
P = 0.3534
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N2
High N2
P = 0.3735
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N2/N1
High N2/N1
P < 0.0001
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N2/N1
High N2/N1
P = 0.2772
0102030405060
0
20
40
60
80
100
Months
Cumulative Incidence of relapse, %
Low N2/N1
High N2/N1
P < 0.0001
T
umor nes
t
T
umor stroma
C
ombined tumor are
a
D
E
F
Fig. 4 Kaplan–Meier plots for OS in HNSCC patients by the HNSCC-infiltrating N2/N1 neutrophil plasticity within TN (A), TS (B) and combined tumor area (C). Kaplan–Meier plots for OS
in patients with HNSCC by the plasticity of HNSCC-infiltrating N2/N1 neutrophils within TN (D), TS (E) and combined tumor area (F). RFS relapse-free survival, OS overall survival, TN
tumor nest, TS tumor stroma
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
0510 15 20
HROS(95% CI)
T
umor nes
t
Tumor-infiltrating N1
Low
Hi
g
h
Tumor-infiltratin
g
N2
Low
High
N2/N1 ratio
Low
H
ig
h
Co
m
b
in
ed
tu
m
o
r
a
r
ea
Tumor-infiltratin
g
N1
Low
H
i
g
h
Tumor-infiltratin
g
N2
Low
Hi
g
h
N2/N1 r
at
i
o
Low
H
i
gh
0.262(0.105-0.651)
2.454(1.071-5.623)
10.470(4.157-26.369)
0.945(0.443-2.017)
2.154(0.965-4.804)
1.989(0.838-4.718)
0.600(0.280-1.286)
2.027(0.926-4.436)
6.211(2.846-13.554)
0.004
0.034
< 0.0001
0.883
0.061
0.119
0.189
0.077
< 0.0001
0510 15 20
Adjusted HROS(95% CI)
0.355(0.132-0.950)
3.069(1.144-8.233)
4.458(1.384-14.366)
0.874(0.351-2.173)
2.485(0.725-8.513)
3.010(1.072-8.450)
0.572 0.216-1.515
1.61 0.540-4.794
3.373 1.269-8.965)
0.039
0.026
0.012
0.772
0.148
0.036
0.261
0.393
0.015
HR
OS
(
95
%
C
I)
Adj
uste
d
HR
OS
(95
%
C
I)
Tumor strom
a
Tumor-infiltratin
g
N1
Low
H
i
g
h
Tumor-in
f
iltrating N2
L
ow
Hi
g
h
N2/N1 ratio
Low
Hi
g
h
0.945
(
0.443-2.017
)
2
.154
(
0.965-4.804
)
1.989(0.838-4.718
)
0
.
883
0
.
061
0
.11
9
0
.874
(
0.351-2.173
)
2
.485(0.725-8.513)
3.010
(
1.072-8.450
)
0
.772
0
.14
8
0
.
036
0510 15 20
HR
RFS
(95% CI)
T
u
m
o
r n
est
Tumor-infiltratin
g
N
1
Lo
w
Hi
gh
Tumor-infiltratin
g
N2
L
ow
Hig
h
N2/N1 rati
o
L
ow
Hig
h
Co
m
b
in
ed tu
m
o
r
a
r
ea
Tumor-in
f
iltratingN
1
L
ow
Hig
h
Tumor-in
f
iltrating N2
L
ow
Hi
gh
N2
/
N1 rati
o
Lo
w
Hi
gh
0.335(0.140-0.800)
1.906(0.848-4.284)
5.092(2.261-11.469)
0.863(0.399-1.870)
1.445(0.663-3.149)
1.659(0.663-4.148)
0.638(0.294-1.384)
1.372(0.634-2.970)
4.541(2.080-9.914)
0.014
0.119
< 0.0001
0.709
0.355
0.279
0.255
0.422
< 0.0001
0510 15 20
Adjusted HR
RFS
(95% CI)
0.342(0.132-0.885)
2.295(0.717-7.339)
3.088(1.171-8.140)
1.034(0.424-2.518)
0.873(0.286-2.664)
1.419(0.459-4.386)
0.736(0.287-1.885)
1.146(0.391-3.364)
3.350(1.194-9.400)
0.027
0.162
0.023
0.942
0.812
0.544
0.523
0.804
0.022
HR
R
F
S
(95
%C
I
)
Adj
uste
dHR
RF
S
(95
%C
I
)
T
umor stroma
Tumor-infiltrating N
1
L
o
w
Hi
g
h
Tumor-in
f
iltrating N2
Lo
w
Hi
g
h
N2/N1 r
at
i
o
Lo
w
Hig
h
0.863
(
0.399-1.870
)
1.445
(
0.663-3.149
)
1.659
(
0.663-4.148
)
0
.7
09
0
.
3
55
0
.27
9
1.034(0.424-2.518
)
0.873(0.286-2.664
)
1.419(0.459-4.386
)
0
.
9
4
2
0
.
8
1
2
0
.54
4
B
A
Fig. 5 Univariate and multivariate COX regression estimating the
prognostic value of HNSCC-infiltrating N2/N1 plasticity within dif-
ferent regions for OS (A) and RFS (B). The independent prognostic
value of HNSCC-infiltrating N2/N1 neutrophil plasticity for OS was
estimated through multivariate COX regression adjusted for primary
site, T stage, N stage, TNM stage, tumor size, HNSCC-infiltrating
CD8+ T cells and HNSCC-infiltrating FOXP3+ Tregs, which were
selected through univariate COX regression models (Supplementary
Table4); The independent prognostic value of HNSCC-infiltrating
N2/N1 neutrophil plasticity for RFS was estimated through multi-
variate COX regression adjusted for smoking history, primary site,
T stage, N stage, tumor size, HNSCC-infiltrating CD8+ T cells and
HNSCC-infiltrating FOXP3+ Tregs, which were selected through
univariate COX regression models (Supplementary Table4); Supple-
mentary Table5–7 showed the corresponding HRs for each variables
in multivariate COX regression models
X.Zhu et al.
TGF-β neutralization could induce the accumulation of neu-
trophils with an antitumor phenotype (N1) [20]. Previous
studies have also reported the abundance of TGF-β expres-
sion within HNSCC and the benefit of TGF-β blockade
in improving survival [37], which may correlate with our
finding that the infiltration of N2 neutrophils was higher in
HNSCC tissue than that in adjacent normal tissue. Notably,
we discovered that the infiltration pattern of N2/N1 neu-
trophils varied between the TN and TS, with a relatively
higher number of N2 neutrophils and a lower number of N1
neutrophils localized in the TN region compared to the TS.
Accordingly, the ratio of HNSCC-infiltrating N2/N1 neutro-
phils was also higher in the TN region than that in the TS.
Our study revealed that patients with a smoking history
tended to have a high ratio of HNSCC-infiltrating N2/N1
neutrophils in TN, consistent with previous studies that
smoking-induced chronic inflammation and nicotine expo-
sure could promote neutrophil polarization to the N2 phe-
notype [23, 38]. Furthermore, patients with higher N2/N1
ratios, particularly in TN, were found to be at an advanced
TNM stage, with larger tumor size and invasive growth into
adjacent tissue. The reason why increased N2 neutrophils
infiltrate advanced HNSCC requires additional discussion.
One possible explanation is that advanced HNSCC can
secrete more specific cytokines to homing N2 neutrophils,
like members of the C-X-C chemokine family and IL6 as
mentioned above [35, 36]. In addition, advanced HNSCC
may produce increased factors to drive the polarization of
neutrophils toward N2 phenotype [37]. Notably, prior studies
have reported that HNSCC-infiltrating N2 neutrophils could
promote tumor growth and invasion through the production
of neutrophil extracellular traps (NETs) and the secretion of
ROS, MMP9, HGF, myeloperoxidase, and oncostatin M [39,
40]. Thus, we hypothesis that a positive feedback loop may
exist between tumor progression and the accumulation of N2
neutrophils within tumor, which needs further investigation
in our future studies. That is, the development and growth
of HNSCC can attract the homing of N2 neutrophils or drive
the polarization of neutrophils toward N2 phenotype, and
in turn, the increased N2 neutrophils within HNSCC can
promote the progression of tumor via various pro-tumor
mechanisms. Further studies are warranted to elucidate this
association in an effort to find potential clues for future can-
cer therapy.
Increasing evidences have highlighted the significance of
cancer immune landscape in modulating tumor progression,
patient prognosis and immunotherapy effectiveness [41, 42].
CD8+ T cells have been shown to induce pyroptosis in tumor
cells through the release of TNF-α and IFN-γ [43], with
long-term survivors exhibiting higher densities of CD8+ T
cells in HNSCC tissue as evidenced by IHC and IF staining
[13]. these findings align with our observation that increased
CD8+ T cells infiltration correlates with improved survival
outcomes. Conversely, higher densities of FOXP3+ Tregs
have been linked to poorer survival outcomes due to the
expression of CTLA4 [44], which is also consistent with
our observations. We noted an increased number of FOXP3+
Tregs and a decreased number of CD8+ lymphocytes infil-
trating HNSCC tissue, accompanied by a high infiltration
of N2 phenotype or a high ratio of N2/N1 neutrophils in
TN. Moreover, we identified a negative relationship between
the HNSCC-infiltrating N2/N1 ratio and the CD8/CD3 and
CD8/Treg ratios, as well as a positive correlation between
the HNSCC-infiltrating N2/N1 ratio and the Treg/CD3 ratio.
These intriguing findings may be attributable the complex
interplay between immune cells within the HNSCC TME.
It is important to note that neutrophils primarily promote
tumor growth and immune escape through their inhibitory
effects on T lymphocytes [36]. HNSCC-infiltrating neu-
trophils have been shown to suppress the proliferation and
function of CD8+ T cells via PD-L1 and PD-1 binding [7]
or the release of immunosuppressive cytokines, such as argi-
nase 1 and ROS [45]. Meanwhile, Mishalian etal. reported
that HNSCC-infiltrating neutrophils secrete the chemokine
CCL17, which plays a crucial role in recruiting FOXP3+
Tregs [22]. Contrarily, a recent study demonstrated that neu-
trophil infiltration can enhance the prognostic significance
of CD8+ T cells infiltration in cancer, suggesting a potential
role in promoting antitumor immunity [46]. The intricate
interactions and crosstalk between HNSCC-infiltrating neu-
trophil phenotypes and T cells warrant further investigation
to elucidate novel therapeutic approaches for HNSCC.
In this study, we primarily assessed the prognostic
implications of HNSCC-infiltrating N2/N1 neutrophil
plasticity in HNSCC patients. Elevated N2 neutrophils
infiltration and a higher ratio of N2/N1 neutrophils in TN
significantly were correlated with poorer OS and RFS
in HNSCC patients, as indicated by the Kaplan–Meier
curves. Thereafter, univariate and multivariate COX
regression analyses identified the plasticity of HNSCC-
infiltrating N2/N1 neutrophils as an independent prognos-
tic predictor. One possible underlying mechanism involves
the interaction between neutrophils and tumor cells in TN,
which may induce the neutrophil polarization toward the
N2 phenotype and, in turn, directly facilitate tumor cell
proliferation [17]. In addition, neutrophils in TN may
release neutrophil extracellular traps, which could impede
T cell penetration and disrupt contact between immune
cytotoxic cells and tumor cells [47, 48]. Consequently,
incorporating N2/N1 neutrophil plasticity in TN into
the TNM staging system might provide a more accurate
prognostic classification for HNSCC patients. Moreover,
several studies [49, 50] have reported the role of HNSCC-
infiltrating neutrophils and NETs in mediating resistance
to immunotherapy. Therefore, further assessments of
the correlations between the HNSCC-infiltrating N2/N1
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
neutrophils plasticity, particularly in TN, and treatment
responses may yield valuable insights for selecting optimal
therapeutic strategies.
Despite the findings of our study, several limitations
warrant further discussion. First, the single-center and ret-
rospective nature of this study may constrain the general-
izability of our results, necessitating validation through
larger, multicenter, and prospective cohorts. Second, we
only included patients with laryngeal or hypopharyngeal
SCC, and future studies should investigate the prognos-
tic implications of HNSCC-infiltrating N2/N1 neutrophil
plasticity in other HNSCC subsites. Third, the underly-
ing mechanisms by which the HNSCC TME polarizes
HNSCC-infiltrating neutrophils towards N2 phenotype
remain unclear. Further studies should focus on elucidat-
ing the mechanisms of N2/N1 neutrophils plasticity, espe-
cially in TN, and their specific roles in HNSCC.
All in all, our study demonstrated the existence of
HNSCC-infiltrating N2/N1 neutrophils plasticity in HNSCC.
HNSCC-infiltrating N2/N1 neutrophil plasticity carries sub-
stantial implications regarding immune evasion and prog-
nostic outcomes. Moreover, HNSCC-infiltrating N2 neu-
trophils within the TN may serve a pivotal function in the
HNSCC TME. Further studies exploring neutrophils polari-
zation, especially within the TN region, and its associations
with immunopathological characteristics in HNSCC may
yield invaluable insights for therapeutic strategies.
Supplementary Information The online version contains supplemen-
tary material available at https:// doi. org/ 10. 1007/ s13577- 024- 01024-7.
Acknowledgements We thank all members of Professor Lu’s Labo-
ratory at Shanghai Institute of Immunology and Shanghai Jiaotong
University School of Medicine for their technical assistance.
Author contributions LLM, LT, and DZ designed the experiments.
XKZ and YH contributed to executing the described experiments. DT
and JZ was responsible for the bioinformatic analysis of the data. LLM,
LT, XKZ, and JYM analyzed the data. XKZ and XPD contributed to
the preparation of this manuscript, and all authors aided in editing the
final manuscript.
Funding This study was supported by of the Major Clinical Research
Project of Shanghai Shen-kang Hospital Clinical Development
Center under Grant (Grant numbers: SHDC2020CR6011); the Sci-
ence and Technology Innovation Project of Shanghai Shen-kang
Hospital Clinical Development Center under Grant (Grant numbers:
SHDC12015114); the National Natural Science Foundation of China
under Grant (Grant numbers: 81772878, 82003178); the Shanghai
Municipal Key Clinical Specialty under Grant (Grant numbers: shslc-
zdzk00801); the Science and Technology Committee of Shanghai
under Grant (Grant numbers: 20MC1920200); the Science and Tech-
nology Commission of Shanghai Municipality under Grant (Grant
numbers: 20Y11902200); the Shanghai Anti-Cancer Development
Foundation under Grant (Grant numbers: H6001-008); and the Train-
ing Program of the Excellent Doctors of Fudan University under Grant
(Grant numbers: QT00140).
Data availability Data will be made available on request.
Declarations
Conflict of interest The authors have no relevant financial or non-fi-
nancial interests to disclose.
Ethics approval This study adhered to the principles of the Declaration
of Helsinki and received approval from the Medical Research Council
of the Eye & ENT Hospital, Fudan University, Shanghai, China (No.
KJ2008-01).
Patient consent for publication Consent obtained directly from
patient(s).
References
1. Marur S, Forastiere AA. Head and neck squamous cell carci-
noma: update on epidemiology, diagnosis, and treatment. Mayo
Clin Proc. 2016;91(3):386–96. https:// doi. org/ 10. 1016/j. mayocp.
2015. 12. 017.
2. Chow LQM. Head and neck cancer. N Engl J Med.
2020;382(1):60–72. https:// doi. org/ 10. 1056/ NEJMr a1715 715.
3. Zhu X, Zhou J, Zhou L, Zhang M, Gao C, Tao L. Association
between postoperative radiotherapy for young-onset head and
neck cancer and long-term risk of second primary malignancy:
a population-based study. J Transl Med. 2022;20(1):405. https://
doi. org/ 10. 1186/ s12967- 022- 03544-y.
4. Choi JH, Lee BS, Jang JY, Lee YS, Kim HJ, Roh J, etal. Single-
cell transcriptome profiling of the stepwise progression of head
and neck cancer. Nat Commun. 2023;14(1):1055. https:// doi. org/
10. 1038/ s41467- 023- 36691-x.
5. Wang X, Muzaffar J, Kirtane K, Song F, Johnson M, Schell MJ,
etal. T cell repertoire in peripheral blood as a potential biomarker
for predicting response to concurrent cetuximab and nivolumab in
head and neck squamous cell carcinoma. J Immunother Cancer.
2022;10(6): e004512. https:// doi. org/ 10. 1136/ jitc- 2022- 004512.
6. Ruffin AT, Li H, Vujanovic L, Zandberg DP, Ferris RL, Bruno
TC. Improving head and neck cancer therapies by immunomod-
ulation of the tumour microenvironment. Nat Rev Cancer.
2023;23(3):173–88. https:// doi. org/ 10. 1038/ s41568- 022- 00531-9.
7. Tang D, Zhang D, Heng Y, Zhu XK, Lin HQ, Zhou J, etal. Tumor-
infiltrating PD-L1+ neutrophils induced by GM-CSF suppress T
cell function in laryngeal squamous cell carcinoma and predict
unfavorable prognosis. J Inflamm Res. 2022;15:1079–97. https://
doi. org/ 10. 2147/ JIR. S3477 77.
8. Huang Q, Hsueh CY, Shen YJ, Guo Y, Huang JM, Zhang YF,
etal. Small extracellular vesicle-packaged TGFβ1 promotes the
reprogramming of normal fibroblasts into cancer-associated fibro-
blasts by regulating fibronectin in head and neck squamous cell
carcinoma. Cancer Lett. 2021;517:1–13. https:// doi. org/ 10. 1016/j.
canlet. 2021. 05. 017.
9. Ferris RL, Licitra L. PD-1 immunotherapy for recurrent or meta-
static HNSCC. Lancet. 2019;394(10212):1882–4. https:// doi. org/
10. 1016/ S0140- 6736(19) 32539-5.
10. Burtness B, Harrington KJ, Greil R, Soulières D, Tahara M,
de Castro JG, etal. Pembrolizumab alone or with chemo-
therapy versus cetuximab with chemotherapy for recurrent
or metastatic squamous cell carcinoma of the head and neck
(KEYNOTE-048): a randomised, open-label, phase 3 study.
Lancet. 2019;394(10212):1915–28. https:// doi. org/ 10. 1016/
S0140- 6736(19) 32591-7.
11. Heng Y, Zhu X, Lin H, Jingyu M, Ding X, Tao L, etal. CD206+
tumor-associated macrophages interact with CD4+ tumor-
infiltrating lymphocytes and predict adverse patient outcome
X.Zhu et al.
in human laryngeal squamous cell carcinoma. J Transl Med.
2023;21(1):167. https:// doi. org/ 10. 1186/ s12967- 023- 03910-4.
12. Sica A, Schioppa T, Mantovani A, Allavena P. Tumour-associated
macrophages are a distinct M2 polarised population promoting
tumour progression: potential targets of anti-cancer therapy. Eur
J Cancer. 2006;42(6):717–27. https:// doi. org/ 10. 1016/j. ejca. 2006.
01. 003.
13. Zhang D, Tang D, Heng Y, Zhu XK, Zhou L, Tao L, etal. Prog-
nostic impact of tumor-infiltrating lymphocytes in laryngeal squa-
mous cell carcinoma patients. Laryngoscope. 2021;131(4):E1249–
55. https:// doi. org/ 10. 1002/ lary. 29196.
14. Valero C, Pardo L, López M, García J, Camacho M, Quer M,
etal. Pretreatment count of peripheral neutrophils, monocytes,
and lymphocytes as independent prognostic factor in patients with
head and neck cancer. Head Neck. 2017;39(2):219–26. https:// doi.
org/ 10. 1002/ hed. 24561.
15. Rosculet N, Zhou XC, Ha P, Tang M, Levine MA, Neuner G, etal.
Neutrophil-to-lymphocyte ratio: prognostic indicator for head and
neck squamous cell carcinoma. Head Neck. 2017;39(4):662–7.
https:// doi. org/ 10. 1002/ hed. 24658.
16. Ponzetta A, Carriero R, Carnevale S, Barbagallo M, Molgora M,
Perucchini C, etal. Neutrophils driving unconventional T cells
mediate resistance against murine sarcomas and selected human
tumors. Cell. 2019;178(2):346-360.e24. https:// doi. org/ 10. 1016/j.
cell. 2019. 05. 047.
17. Hedrick CC, Malanchi I. Neutrophils in cancer: heterogeneous
and multifaceted. Nat Rev Immunol. 2022;22(3):173–87. https://
doi. org/ 10. 1038/ s41577- 021- 00571-6.
18. Antonio N, Bønnelykke-Behrndtz ML, Ward LC, Collin J, Chris-
tensen IJ, Steiniche T, etal. The wound inflammatory response
exacerbates growth of pre-neoplastic cells and progression to
cancer. EMBO J. 2015;34(17):2219–36. https:// doi. org/ 10. 15252/
embj. 20149 0147.
19. Xu W, Dong J, Zheng Y, Zhou J, Yuan Y, Ta HM, etal. Immune-
checkpoint protein VISTA regulates antitumor immunity by con-
trolling myeloid cell-mediated inflammation and immunosuppres-
sion. Cancer Immunol Res. 2019;7(9):1497–510. https:// doi. org/
10. 1158/ 2326- 6066. CIR- 18- 0489.
20. Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, etal.
Polarization of tumor-associated neutrophil phenotype by TGF-
beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16(3):183–94.
https:// doi. org/ 10. 1016/j. ccr. 2009. 06. 017.
21. Spiegel A, Brooks MW, Houshyar S, Reinhardt F, Ardolino M,
Fessler E, etal. Neutrophils suppress intraluminal NK cell-medi-
ated tumor cell clearance and enhance extravasation of dissemi-
nated carcinoma cells. Cancer Discov. 2016;6(6):630–49. https://
doi. org/ 10. 1158/ 2159- 8290. CD- 15- 1157.
22. Mishalian I, Bayuh R, Eruslanov E, Michaeli J, Levy L, Zolotarov
L, etal. Neutrophils recruit regulatory T-cells into tumors via
secretion of CCL17—a new mechanism of impaired antitumor
immunity. Int J Cancer. 2014;135(5):1178–86. https:// doi. org/ 10.
1002/ ijc. 28770.
23. Tyagi A, Sharma S, Wu K, Wu SY, Xing F, Liu Y, etal. Nico-
tine promotes breast cancer metastasis by stimulating N2 neutro-
phils and generating pre-metastatic niche in lung. Nat Commun.
2021;12(1):474. https:// doi. org/ 10. 1038/ s41467- 020- 20733-9.
24. Singhal S, Bhojnagarwala PS, O’Brien S, Moon EK, Garfall AL,
Rao AS, Quatromoni JG, etal. Origin and role of a subset of
tumor-associated neutrophils with antigen-presenting cell features
in early-stage human lung cancer. Cancer Cell. 2016;30(1):120–
35. https:// doi. org/ 10. 1016/j. ccell. 2016. 06. 001.
25. Gershkovitz M, Caspi Y, Fainsod-Levi T, Katz B, Michaeli J, Kha-
waled S, etal. TRPM2 mediates neutrophil killing of disseminated
tumor cells. Cancer Res. 2018;78(10):2680–90. https:// doi. org/ 10.
1158/ 0008- 5472. CAN- 17- 3614.
26. Massara M, Bonavita O, Savino B, Caronni N, Mollica Poeta V,
Sironi M, etal. ACKR2 in hematopoietic precursors as a check-
point of neutrophil release and anti-metastatic activity. Nat Com-
mun. 2018;9(1):676. https:// doi. org/ 10. 1038/ s41467- 018- 03080-8.
27. Matlung HL, Babes L, Zhao XW, van Houdt M, Treffers LW, van
Rees DJ, etal. Neutrophils kill antibody-opsonized cancer cells
by trogoptosis. Cell Rep. 2018;23(13):3946-3959.e6. https:// doi.
org/ 10. 1016/j. celrep. 2018. 05. 082.
28. He M, Peng A, Huang XZ, Shi DC, Wang JC, Zhao Q, etal.
Peritumoral stromal neutrophils are essential for c-Met-elicited
metastasis in human hepatocellular carcinoma. Oncoimmunology.
2016;5(10): e1219828. https:// doi. org/ 10. 1080/ 21624 02X. 2016.
12198 28.
29. Demers M, Wagner DD. NETosis: a new factor in tumor progres-
sion and cancer-associated thrombosis. Semin Thromb Hemost.
2014;40(3):277–83. https:// doi. org/ 10. 1055/s- 0034- 13707 65.
30. Sagiv JY, Michaeli J, Assi S, Mishalian I, Kisos H, etal. Pheno-
typic diversity and plasticity in circulating neutrophil subpopula-
tions in cancer. Cell Rep. 2015;10(4):562–73. https:// doi. org/ 10.
1016/j. celrep. 2014. 12. 039.
31. Pylaeva E, Lang S, Jablonska J. The essential role of type I inter-
ferons in differentiation and activation of tumor-associated neutro-
phils. Front Immunol. 2016;7:629. https:// doi. org/ 10. 3389/ fimmu.
2016. 00629.
32. Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Manto-
vani A. Neutrophil diversity and plasticity in tumour progression
and therapy. Nat Rev Cancer. 2020;20(9):485–503. https:// doi. or g/
10. 1038/ s41568- 020- 0281-y.
33. Silvestre-Roig C, Fridlender ZG, Glogauer M, Scapini P.
Neutrophil diversity in health and disease. Trends Immunol.
2019;40(7):565–83. https:// doi. org/ 10. 1016/j. it. 2019. 04. 012.
34. Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend
or foe? Carcinogenesis. 2012;33(5):949–55. https:// doi. org/ 10.
1093/ carcin/ bgs123.
35. Wu Y, Zhao Q, Peng C, Sun L, Li XF, Kuang DM. Neutrophils
promote motility of cancer cells via a hyaluronan-mediated TLR4/
PI3K activation loop. J Pathol. 2011;225(3):438–47. https:// doi.
org/ 10. 1002/ path. 2947.
36. Geh D, Leslie J, Rumney R, Reeves HL, Bird TG, Mann DA.
Neutrophils as potential therapeutic targets in hepatocellular carci-
noma. Nat Rev Gastroenterol Hepatol. 2022;19(4):257–73. https://
doi. org/ 10. 1038/ s41575- 021- 00568-5.
37. Redman JM, Friedman J, Robbins Y, Sievers C, Yang X, Lassoued
W, etal. Enhanced neoepitope-specific immunity following neo-
adjuvant PD-L1 and TGF-β blockade in HPV-unrelated head and
neck cancer. J Clin Invest. 2022;132(18): e161400. https:// doi. org/
10. 1172/ JCI16 1400.
38. Tyagi A, Wu SY, Sharma S, Wu K, Zhao D, Deshpande R, etal.
Exosomal miR-4466 from nicotine-activated neutrophils pro-
motes tumor cell stemness and metabolism in lung cancer metas-
tasis. Oncogene. 2022;41(22):3079–92. https:// doi. org/ 10. 1038/
s41388- 022- 02322-w.
39. Li Q, Chen W, Li Q, Mao J, Chen X. A novel neutrophil extra-
cellular trap signature to predict prognosis and immunotherapy
response in head and neck squamous cell carcinoma. Front Immu-
nol. 2022;13:1019967. https:// doi. org/ 10. 3389/ fimmu. 2022. 10199
67.
40. Peng ZP, Jiang ZZ, Guo HF, Zhou MM, Huang YF, Ning WR,
etal. Glycolytic activation of monocytes regulates the accumula-
tion and function of neutrophils in human hepatocellular carci-
noma. J Hepatol. 2020;73(4):906–17. https:// doi. org/ 10. 1016/j.
jhep. 2020. 05. 004.
41. Gulati S, Crist M, Riaz MK, Takiar V, Lehn M, Monroe I, etal.
Durvalumab plus cetuximab in patients with recurrent or meta-
static head and neck squamous cell carcinoma: an open-label,
Prognostic significance andimmune escape implication oftumor‑infiltrating neutrophil…
non-randomized, phase-2 clinical trial. Clin Cancer Res. 2023.
https:// doi. org/ 10. 1158/ 1078- 0432. CCR- 22- 3886.
42. Damasio MPS, Nascimento CS, Andrade LM, de Oliveira VL,
Calzavara-Silva CE. The role of T-cells in head and neck squa-
mous cell carcinoma: From immunity to immunotherapy. Front
Oncol. 2022;12:1021609. https:// doi. org/ 10. 3389/ fonc. 2022.
10216 09.
43. Wang S, Wu ZZ, Zhu SW, Wan SC, Zhang MJ, Zhang BX, etal.
CTLA-4 blockade induces tumor pyroptosis via CD8+ T cells in
head and neck squamous cell carcinoma. Mol Ther. 2023;S1525–
0016(23):00121–31. https:// doi. org/ 10. 1016/j. ymthe. 2023. 02. 023.
44. Knitz MW, Bickett TE, Darragh LB, Oweida AJ, Bhatia S, Van
Court B, etal. Targeting resistance to radiation-immunotherapy
in cold HNSCCs by modulating the Treg-dendritic cell axis. J
Immunother Cancer. 2021;9(4): e001955. https:// doi. org/ 10. 1136/
jitc- 2020- 001955.
45. Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-
specific inhibition of CD8+ T cell response by immature myeloid
cells in cancer is mediated by reactive oxygen species. J Immunol.
2004;172(2):989–99. https:// doi. org/ 10. 4049/ jimmu nol. 172.2.
989.
46. Governa V, Trella E, Mele V, Tornillo L, Amicarella F, Cremo-
nesi E, etal. The interplay between neutrophils and CD8+ T
cells improves survival in human colorectal cancer. Clin Cancer
Res. 2017;23(14):3847–58. https:// doi. org/ 10. 1158/ 1078- 0432.
CCR- 16- 2047.
47. Pietrobon V, Marincola FM. Hypoxia and the phenomenon of
immune exclusion. J Transl Med. 2021;19(1):9. https:// doi. org/
10. 1186/ s12967- 020- 02667-4.
48. Teijeira Á, Garasa S, Gato M, Alfaro C, Migueliz I, Cirella A,
etal. CXCR1 and CXCR2 chemokine receptor agonists produced
by tumors induce neutrophil extracellular traps that interfere with
immune cytotoxicity. Immunity. 2020;52(5):856-871.e8. https://
doi. org/ 10. 1016/j. immuni. 2020. 03. 001.
49. Li K, Tandurella JA, Gai J, Zhu Q, Lim SJ, Thomas DL 2nd, etal.
Multi-omic analyses of changes in the tumor microenvironment
of pancreatic adenocarcinoma following neoadjuvant treatment
with anti-PD-1 therapy. Cancer Cell. 2022;40(11):1374-1391.e7.
https:// doi. org/ 10. 1016/j. ccell. 2022. 10. 001.
50. Canè S, Barouni RM, Fabbi M, Cuozzo J, Fracasso G, Adamo A,
etal. Neutralization of NET-associated human ARG1 enhances
cancer immunotherapy. Sci Transl Med. 2023;15(687): eabq6221.
https:// doi. org/ 10. 1126/ scitr anslm ed. abq62 21.
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds
exclusive rights to this article under a publishing agreement with the
author(s) or other rightsholder(s); author self-archiving of the accepted
manuscript version of this article is solely governed by the terms of
such publishing agreement and applicable law.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Background Tumor-associated macrophages (TAMs) are major component in the tumor microenvironment (TME) and play regulatory role in tumor progression. We aimed to investigate the infiltration and prognostic value of TAMs in laryngeal squamous cell carcinoma (LSCC) and to reveal the underlying mechanism of TAM subgroups in tumorigenesis. Methods Hematoxylin and eosin (HE) staining were performed to define the tumor nest and stroma of LSCC tissue microarrays. CD206 + /CD163 + and iNOS + TAM infiltrating profiles were obtained and analyzed through double-labeling immunofluorescence and immunohistochemical staining. The recurrence-free (RFS) and overall survival (OS) curves based on the infiltration of TAMs were plotted using the Kaplan-Meier method. Infiltration of macrophages, T lymphocytes and their corresponding subgroups were analyzed in fresh LSCC tissue samples by flow cytometry. Results We found that CD206 ⁺ rather than CD163 ⁺ M2-like TAMs were the most enriched population in the TME of human LSCC. CD206 ⁺ macrophages localized mostly in the tumor stroma (TS) rather than the tumor nest (TN) region. In contrast, relatively low infiltration of iNOS ⁺ M1-like TAMs were found in the TS and almost none in the TN region. High level of TS CD206 ⁺ TAM infiltration correlated with poor prognosis. Interestingly, we identified a HLA-DR high CD206 ⁺ macrophage subgroup that was significantly associated with the tumor-infiltrating CD4 ⁺ T lymphocytes and showed different surface costimulatory molecule expression than that of the HLA-DR low/ -CD206 ⁺ subgroup. Taken together, our results indicate that HLA-DR high -CD206 ⁺ is a highly activated subgroup of CD206 + TAMs that may interact with CD4 + T cells through MHC-II axis and promote tumorigenesis.
Article
Full-text available
Head and neck squamous cell carcinoma (HNSCC) undergoes stepwise progression from normal tissues to precancerous leukoplakia, primary HNSCC, and metastasized tumors. To delineate the heterogeneity of tumor cells and their interactions during the progression of HNSCC, we employ single-cell RNA-seq profiling for normal to metastasized tumors. We can identify the carcinoma in situ cells in leukoplakia lesions that are not detected by pathological examination. In addition, we identify the cell type subsets of the Galectin 7B (LGALS7B)-expressing malignant cells and CXCL8-expressing fibroblasts, demonstrating that their abundance in tumor tissue is associated with unfavorable prognostic outcomes. We also demonstrate the interdependent ligand-receptor interaction of COL1A1 and CD44 between fibroblasts and malignant cells, facilitating HNSCC progression. Furthermore, we report that the regulatory T cells in leukoplakia and HNSCC tissues express LAIR2, providing a favorable environment for tumor growth. Taken together, our results update the pathobiological insights into cell-cell interactions during the stepwise progression of HNSCCs.
Article
Full-text available
Successful pancreatic ductal adenocarcinoma (PDAC) immunotherapy necessitates optimization and maintenance of activated effector T cells (Teff). We prospectively collected and applied multi-omic analyses to paired pre- and post-treatment PDAC specimens collected in a platform neoadjuvant study of granulocyte-macrophage colony-stimulating factor-secreting allogeneic PDAC vaccine (GVAX) vaccine ± nivolumab (anti-programmed cell death protein 1 [PD-1]) to uncover sensitivity and resistance mechanisms. We show that GVAX-induced tertiary lymphoid aggregates become immune-regulatory sites in response to GVAX + nivolumab. Higher densities of tumor-associated neutrophils (TANs) following GVAX + nivolumab portend poorer overall survival (OS). Increased T cells expressing CD137 associated with cytotoxic Teff signatures and correlated with increased OS. Bulk and single-cell RNA sequencing found that nivolumab alters CD4+ T cell chemotaxis signaling in association with CD11b+ neutrophil degranulation, and CD8+ T cell expression of CD137 was required for optimal T cell activation. These findings provide insights into PD-1-regulated immune pathways in PDAC that should inform more effective therapeutic combinations that include TAN regulators and T cell activators.
Article
Full-text available
Head and neck squamous cell carcinoma (HNSCC) encompass a group of complex entities of tumours affecting the aerodigestive upper tract. The main risk factors are strongly related to tobacco and alcohol consumption, but also HPV infection is often associated. Surgery, radiotherapy and/or chemotherapy are the standard treatments, though the 5-year overall survival is less than 50%. The advances in genomics, molecular medicine, immunology, and nanotechnology have shed a light on tumour biology which helps clinical researchers to obtain more efficacious and less toxic therapies. Head and neck tumours possess different immune escape mechanisms including diminishing the immune response through modulating immune checkpoints, in addition to the recruitment and differentiation of suppressive immune cells. The insights into the HNSCC biology and its strong interaction with the tumour microenvironment highlights the role of immunomodulating agents. Recently, the knowledge of the immunological features of these tumours has paved the way for the discovery of effective biomarkers that allow a better selection of patients with odds of improving overall survival through immunotherapy. Specially biomarkers regarding immune checkpoint inhibitors antibodies, such as anti-PD-1/PD-L1 and anti-CTLA-4 in combination with standard therapy or as monotherapy. New immunotherapies to treat head and neck cancer carcinomas, such as CAR T cells and nanoparticles have been the center of attention and in this review, we discuss the necessity of finding targets for the T cell in the cancer cells to generate CAR T cells, but also the relevance of evaluating specificity and safety of those therapies.
Article
Full-text available
Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignant cancers, and patients with HNSCC possess early metastases and poor prognosis. Systematic therapies (including chemotherapy, targeted therapy, and immunotherapy) are generally applied in the advanced/late stages of HNSCC, but primary and acquired resistance eventually occurs. At present, reliable biomarkers to predict the prognosis of HNSCC have not been completely identified. Recent studies have shown that neutrophil extracellular traps (NETs) are implicated in cancer progression, metastasis and cancer immune response, and NET-related gene signatures are associated with the prognosis of patients with several human cancers. To explore whether NET-related genes play crucial roles in HNSCC, we have performed systematic analysis and reported several findings in the current study. Firstly, we identified seven novel NET-related genes and developed a NET-score signature, which was highly associated with the clinicopathological and immune traits of the HNSCC patients. Then, we, for the first time, found that NIFK was significantly upregulated in HNSCC patient samples, and its levels were significantly linked to tumor malignancy and immune status. Moreover, functional experiments confirmed that NIFK was required for HNSCC cell proliferation and metastasis. Altogether, this study has identified a novel NET-score signature based on seven novel NET-related genes to predict the prognosis of HNSCC and NIFK has also explored a new method for personalized chemo-/immuno-therapy of HNSCC.
Article
Full-text available
Background Second primary malignancy (SPM) represents the leading long-term cause of death among patients with index head and neck squamous cell carcinoma (HNSCC). We aimed to quantify the association between postoperative radiotherapy (PORT) and the risk of SPM development for index HNSCC among adolescent and young patients, who are particularly vulnerable to radiation-associated impacts due to their increased tissue susceptibilities and longer life expectancies. Methods This study was conducted using the Surveillance, Epidemiology, and End Results (SEER) database to collect the data of 5 year survivors of index young-onset HNSCC from 1975 to 2011. The outcome of interest was SPM, a new, metachronous malignancy after the index HNSCC. Standardized incidence ratios (SIRs) and excess absolute risks (EARs) were used to quantify the PORT-associated risks externally, and relative risks (RRs) were estimated by the multivariate Poisson regression analysis to quantify the PORT-associated risks internally. Results Of the included 2771 5 year survivors with index young-onset HNSCCs, the receipt of PORT (37.6%) was associated with higher risk of SPMs (RR, 1.23; 95% CI 1.07 to 1.43). PORT-associated risks were elevated for the majority of sites, including head and neck (RR, 1.19; 95% CI 0.95 to 1.50) and lung (RR, 1.67; 95% CI 1.18 to 2.34). With regarding to the subsites of head and neck, RRs were above unity in oral cavity squamous cell carcinoma (SCC) (RR, 1.68; 95% CI 1.39 to 2.03) and laryngeal SCC (RR, 1.02; 95% CI 0.73 to 1.43). A relatively greater RR was observed for patients younger than 35 years (RR, 1.44, 95% CI 0.37 to 5.57) and those diagnosed with localized diseases (RR, 1.16, 95% CI 0.9 to 1.5). PORT-associated risks were increased remarkably after 15 years of follow-up (RR, 1.24; 95% CI 0.97 to 1.58). Conclusions An association was discovered between PORT treatment and increased long-term risk of SPM among patients with index young-onset HNSCC. The findings suggest long-term follow-up surveillance for these patients, particularly those with oral cavity SCC or laryngeal SCC.
Article
Myeloid cells can restrain antitumor immunity by metabolic pathways, such as the degradation of l-arginine, whose concentrations are regulated by the arginase 1 (ARG1) enzyme. Results from preclinical studies indicate the important role of arginine metabolism in pancreatic ductal adenocarcinoma (PDAC) progression, suggesting a potential for clinical application; however, divergent evolution in ARG1 expression and function in rodents and humans has restricted clinical translation. To overcome this dichotomy, here, we show that neutrophil extracellular traps (NETs), released by spontaneously activated neutrophils isolated from patients with PDAC, create a microdomain where cathepsin S (CTSS) cleaves human (h)ARG1 into different molecular forms endowed with enhanced enzymatic activity at physiological pH. NET-associated hARG1 suppresses T lymphocytes whose proliferation is restored by either adding a hARG1-specific monoclonal antibody (mAb) or preventing CTSS-mediated cleavage, whereas small-molecule inhibitors are not effective. We show that ARG1 blockade, combined with immune checkpoint inhibitors, can restore CD8+ T cell function in ex vivo PDAC tumors. Furthermore, anti-hARG1 mAbs increase the frequency of adoptively transferred tumor-specific CD8+ T cells in tumor and enhance the effectiveness of immune checkpoint therapy in humanized mice. Thus, this study shows that extracellular ARG1, released by activated myeloid cells, localizes in NETs, where it interacts with CTSS that in turn cleaves ARG1, producing major molecular forms endowed with different enzymatic activity at physiological pH. Once exocytosed, ARG1 activity can be targeted by mAbs, which bear potential for clinical application for the treatment of PDAC and require further exploration.
Article
Immune checkpoint blockade (ICB) treatment has demonstrated excellent medical effects in oncology, and it is one of the most sought-after immunotherapies for tumors. However, there are several issues with ICB therapy, including low response rates and a lack of effective efficacy predictors. Gasdermin-mediated pyroptosis is a typical inflammatory death mode. We discovered that increased expression of gasdermin protein was linked to a favorable tumor immune microenvironment and prognosis in head and neck squamous cell carcinoma (HNSCC). We used the mouse HNSCC cell lines 4MOSC1 (responsive to CTLA-4 blockade) and 4MOSC2 (resistant to CTLA-4 blockade) orthotopic models and demonstrated that CTLA-4 blockade treatment induced gasdermin-mediated pyroptosis of tumor cells, and gasdermin expression positively correlated to the effectiveness of CTLA-4 blockade treatment. We found that CTLA-4 blockade activated CD8+ T cells and increased the levels of IFN-γ and TNF-α cytokines in the tumor microenvironment. These cytokines synergistically activated the STAT1/IRF1 axis to trigger tumor cell pyroptosis and the release of large amounts of inflammatory substances and chemokines. Collectively, our findings revealed that CTLA-4 blockade triggered tumor cells pyroptosis via the release of IFN-γ and TNF-α from activated CD8+ T cells, providing a new perspective of ICB.
Article
Purpose: The efficacy of cetuximab is poor in metastatic head and neck squamous cell carcinoma (HNSCC). Cetuximab initiates natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity, with resultant recruitment of immune cells and suppression of anti-tumor immunity. We hypothesized that adding an immune checkpoint inhibitor (ICI) could overcome this and lead to an enhanced anti-tumor response. Patients and methods: A phase II study of cetuximab and durvalumab in metastatic HNSCC was conducted. Eligible patients had measurable disease. Patients who had received both cetuximab and an ICI were excluded. The primary endpoint was objective response rate (ORR) by RECIST 1.1 at six months. Results: As of April 2022, 35 patients enrolled, of whom 33 received at least 1 dose of durvalumab and were included in the response analysis. Eleven patients (33%) had received prior platinum-based chemotherapy, 10 an ICI (30%), and 1 patient (3%) cetuximab. ORR was 39% (13/33) with a median duration of response of 8.6 months (95% CI: 6.5, 16.8). Median progression-free and overall survivals were 5.8 months (95% CI: 3.7 to 14.1) and 9.6 months (95% CI: 4.8 to 16.3), respectively. There were sixteen grade 3 treatment-related adverse events (TRAEs) and one grade 4 TRAE, with no treatment-related deaths. Overall and progression-free survival did not correlate with PD-L1 status. NK cell cytotoxic activity was increased by cetuximab and further increased with the addition of durvalumab in responders. Conclusions: The combination of cetuximab and durvalumab demonstrated durable activity with a tolerable safety profile in metastatic HNSCC and warrants further investigation.
Article
Targeted immunotherapy has improved patient survival in head and neck squamous cell carcinoma (HNSCC), but less than 20% of patients produce a durable response to these treatments. Thus, new immunotherapies that consider all key players of the complex HNSCC tumour microenvironment (TME) are necessary to further enhance tumour-specific T cell responses in patients. HNSCC is an ideal tumour type in which to evaluate immune and non-immune cell differences because of two distinct TME aetiologies (human papillomavirus (HPV)-positive and HPV-negative disease), multiple anatomic sites for tumour growth, and clear distinctions between patients with locally advanced disease and those with recurrent and/or metastatic disease. Recent technological and scientific advancements have provided a more complete picture of all cellular constituents within this complex TME and have evaluated the interplay of both immune and non-immune cells within HNSCC. Here, we include a comprehensive analysis of the complete ecosystem of the HNSCC TME, performed utilizing data-rich resources such as The Cancer Genome Atlas, and cutting-edge techniques, such as single-cell RNA sequencing, high-dimensional flow cytometry and spatial multispectral imaging, to generate improved treatment strategies for this diverse disease. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and its incidence continues to rise, mostly owing to an increase in human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma. In this Review, the authors describe HPV-positive and HPV-negative HNSCC tumour microenvironments and discuss current and novel treatment modalities.