ArticlePDF AvailableLiterature Review

Chemotherapy and immunotherapy for recurrent and metastatic head and neck cancer: a systematic review

Authors:
  • Azienda Ospedaliera Santi Paolo e Carlo - Polo Universitario

Abstract and Figures

Head and neck cancer (HNC) is a fatal malignancy with an overall long-term survival of about 50% for all stages. The diagnosis is not rarely delayed, and the majority of patients present with loco-regionally advanced disease. The rate of second primary tumors after a diagnosis of HNC is about 3–7% per year, the highest rate among solid tumors. Currently, a single-modality or a combination of surgery, radiotherapy and chemotherapy (CHT), is the standard treatment for stage III–IV HNC. For the recurrent/metastatic setting, in the last 40 years great efforts have been made in order to develop a more effective CHT regimen, from the use of methotrexate alone, to the combination of cisplatin (CDDP) and 5-fluorouracile (5FU) or paclitaxel. Recently, the introduction of cetuximab, an anti-EGFR monoclonal antibody, to the CDDP–5FU doublet (EXTREME regimen) has improved the overall response rate, the progression-free survival and the overall survival (OS) compared to CHT alone. Nowadays, the EXTREME regimen is the standard of care for the first-line treatment of recurrent/metastatic head and neck carcinoma (RMHNC). In the last years, new promising therapies for RMHNC such as immune checkpoint inhibitors (ICIs), which have demonstrated favorable results in second-line clinical trials, gained special interest. Nivolumab and pembrolizumab are the first two ICIs able to prolong OS in the second-, later-line and platinum-refractory setting, with tolerable toxicities. This review summarizes the current state of the art in RMHNC treatment options.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Medical Oncology (2018) 35:37
https://doi.org/10.1007/s12032-018-1096-5
REVIEW ARTICLE
Chemotherapy andimmunotherapy forrecurrent andmetastatic head
andneck cancer: asystematic review
AlessandroGuidi1 · CarlaCodecà1· DarisFerrari1
Received: 11 January 2018 / Accepted: 5 February 2018 / Published online: 13 February 2018
© Springer Science+Business Media, LLC, part of Springer Nature 2018
Abstract
Head and neck cancer (HNC) is a fatal malignancy with an overall long-term survival of about 50% for all stages. The
diagnosis is not rarely delayed, and the majority of patients present with loco-regionally advanced disease. The rate of
second primary tumors after a diagnosis of HNC is about 3–7% per year, the highest rate among solid tumors. Currently,
a single-modality or a combination of surgery, radiotherapy and chemotherapy (CHT), is the standard treatment for stage
III–IV HNC. For the recurrent/metastatic setting, in the last 40years great efforts have been made in order to develop a more
effective CHT regimen, from the use of methotrexate alone, to the combination of cisplatin (CDDP) and 5-fluorouracile
(5FU) or paclitaxel. Recently, the introduction of cetuximab, an anti-EGFR monoclonal antibody, to the CDDP–5FU doublet
(EXTREME regimen) has improved the overall response rate, the progression-free survival and the overall survival (OS)
compared to CHT alone. Nowadays, the EXTREME regimen is the standard of care for the first-line treatment of recurrent/
metastatic head and neck carcinoma (RMHNC). In the last years, new promising therapies for RMHNC such as immune
checkpoint inhibitors (ICIs), which have demonstrated favorable results in second-line clinical trials, gained special interest.
Nivolumab and pembrolizumab are the first two ICIs able to prolong OS in the second-, later-line and platinum-refractory
setting, with tolerable toxicities. This review summarizes the current state of the art in RMHNC treatment options.
Keywords Immunotherapy· Chemotherapy· Monoclonal antibodies· Recurrent/metastatic head and neck cancer
Introduction
Head and neck cancer (HNC) represents the sixth leading
cancer by incidence worldwide [1], and it is defined as a
heterogeneous variety of malignant tumors which includes
oral cavity, oropharynx, hypopharynx, nasopharynx, larynx,
nasal fossa, paranasal sinuses, thyroid, salivary glands and
vermilion surfaces. This kind of cancer constitutes about 3%
of all newly diagnosed malignant tumors in humans, and this
percentage is likely to increase in the future. All over the
world head and neck tumors account for more than 550,000
new cases and 380,000 deaths annually. (In Europe, this
statistics declines to 250,000 new cases and 63,500 deaths
annually in 2012.) [2] Nearly all the cases of head and neck
malignancies (95%) are represented by squamous cell car-
cinoma (HNSCC) arising in the oral cavity and pharynx.
Adenocarcinomas, adenoid cystic carcinomas and mucoepi-
dermoid varieties are more frequent in the salivary glands,
while in the thyroid gland the most frequent histological
subtypes include papillary, follicular giant cell, Hürthle cell
carcinomas and lymphomas.
Despite the recent advances in treatment, including chem-
otherapy (CHT), radiotherapy (RTX) and the most promis-
ing immunotherapy, the global, long-term survival rate is
still under 50%, and this rate decreases to 19% if patients
are diagnosed in very advanced stage [3]. This trend can
be attributed to various causes, first of all the persistence
of inescapable and strong risk factors such as smoking and
alcohol, the new outbreak of human papillomavirus (HPV)
infection, the diagnostic delay of this kind of cancers and the
frequent onset of multiple primary tumors. Indeed, the rate
of second primary tumor is near 3–7% per year. No other
malignancies have a rate as higher as this [4, 5].
* Alessandro Guidi
alessandro.guidi@live.com
Carla Codecà
carla.codeca@asst-santipaolocarlo.it
Daris Ferrari
daris.ferrari@asst-santipaolocarlo.it
1 Department ofMedical Oncology, San Paolo Hospital, Via
Antonio di Rudinì, 8, 20142Milan, Italy
Medical Oncology (2018) 35:37
1 3
37 Page 2 of 12
Risk factors andbiological behavior
ofHNSCCs
Risk factors
The major risk factors for HNSCC are cigarette smoking
and alcohol consumption; for this reason, European and
North America males are more frequently affected than
females with a ratio ranging from 2:1 to 4:1. The propor-
tion of incidence due to alcohol and tobacco use is near
72, 4% of which due to alcohol alone, 33% due to tobacco
alone and 35% due to the combined action of alcohol and
tobacco [6]. Another important risk factor associated with
oropharyngeal carcinoma is represented by HPV [7]. In
particular, HPV-16 genotype has been identified as a caus-
ative agent in almost 50% of oropharyngeal malignancies
in patients with no other classical risk factors, especially
when localized at the base of the tongue and in the tonsil-
lar region. HPVs-18, 31 and 33 are also responsible for
HNSCC, but they are less frequent than genotype 16 [7].
Nowadays, the demographic pattern of HNC is chang-
ing. In the last 30years, there was a steady decline in the
number of HNSCC caused by tobacco and alcohol, but on
the contrary, there was an increase in the number of HPV-
related cases, for which sexual behavior is the most accred-
ited risk factor. For this reason, at present, patients affected
by HNSCC are more likely to be younger adults aged
40–50years [8]. While tobacco- and alcohol-related oro-
pharyngeal tumors are more frequent in African-Americans,
malignancies caused by HPV are diagnosed more frequently
among Caucasians. Overall survival (OS) is higher for HPV-
related HNSCC (about 79–82% of patients is still alive at
5years) than for tobacco-related ones (46%).
Also in the recurrent and/or metastatic context, the OS
for HPV-positive tumors is better than their HPV-negative
counterparts, with a 2-year OS rate of about 55 versus
28%. This favorable trend is likely explained by the fact
that carcinomas associated with HPV are more responsive
to treatment than those HPV-unrelated.
Other known risk factors for HNC include familiarity,
poor oral hygiene, chronic irritation of mucous membranes
(e.g., irritation from dental prostheses), actinic radiation
and pipe smoke for lip cancer, malnutrition (vitamin A
deficiency), high consumption of salted meat or fish,
immunosuppression (HIV, GVHD, transplant) and, within
the Asian population, betel mastication [9].
Recurrent HNSCCs
“Field cancerization” or “condemned mucosa” is an
expression, coined in 1953 by Slaughter [10], that is
referred to the presence of transformed stem cells near
the area of the primary tumor and well explains the natu-
ral trend of head and neck tumors to recur. According to
this theory, multiple primary tumors are the expression of
multiple independent foci of altered-by-carcinogens stem
cells. According to the “field cancerization,” a patient who
has survived for 5years from the diagnosis has a prob-
ability of developing one other primary tumor of about
35% in the same time lapse [3]. Therefore, the diagnosis of
premalignant lesions, such as leukoplakia or erythroplakia,
should be as earlier as possible.
Metastatic process
To better understand the reasoning behind the use of tradi-
tional and innovative therapies, it is important to know how
the metastatic behavior is acquired by cancer cells. The most
frequent sites where HNSCC metastasizes are the lymph
nodes of the cervical region and the lungs. The involvement
of different anatomical sites is much less frequent.
The so-called permeation mode was a common thought
in the past which was used to explain how primary tumor
cells spread to other organs. Following the “permeation
mode” theory, the metastatic process of HNSCC is the result
of tumor cells migration into lymphatic vessels. Regional
lymph nodes act as filters in order to prevent distant metas-
tasis [11]. This theory was accepted only for a short period
because several studies in the 1960s demonstrated that
lymph nodes could be easily overcome by tumor cells even
when they were not saturated [12, 13]. Nowadays it is gen-
erally accepted that the metastatic process is an active and
complex process rather than a passive one as described in the
1960s [14]. The first step is tumor cell detachment followed
by degradation of the extracellular matrix (ECM). This
process is regulated by matrix metalloproteinases (MMPs)
produced by cancer cells and stromal cells (e.g., fibroblasts
and inflammatory cells), and it is called epithelial-to-mes-
enchymal transition (EMT). The following step is tumor cell
dissemination, which can be local, lymphatic or blood dis-
semination. The interactions between receptors of both can-
cer cells and target organs are most important in the last step
of metastatic invasion, and they are the reason why a tumor
preferentially metastasizes to certain organs rather than oth-
ers. With regard to HNSCC, cell-surface receptors such as
epidermal growth factor receptor (EGFR) and neurotrophin
receptor B (TrkB), as well as the presence of inflammatory
cytokines such as interleukin-1b, have all been implicated in
the induction of EMT [14]. Cytokines such as interleukin-8
(IL-8), platelet-derived growth factor (PDGF), hepatocyte
growth factor (HGF) and vascular endothelial growth factor
(VEGF) are expressed by HNSCC tumor cells secondary to
constitutive activation of mitogen-activated protein kinase,
nuclear factor-kappa B and signal transducer and activator
Medical Oncology (2018) 35:37
1 3
Page 3 of 12 37
of transcription 3 intracellular signaling cascades, as well as
by immune and other stromal cells within the tumor micro-
environment. VEGF family members, in particular, seem to
correlate with elevated lymphatic vessel density in tumor
specimens and the presence of cervical metastasis [14].
However, further studies to assess the metastatic phenotype
of HNSCC malignant cells are needed.
Pathology ofmetastatic HNSCCs
Like any other cancer, HNSCC is the result of cumulative
mutations that cause the activation of oncogenes and the
inactivation of tumor suppressor genes in a clonal cell popu-
lation. HNSCCs normally arise from premalignant lesions,
such as leukoplakia or erythroplakia. Leukoplakia is defined
by World Health Organization as a “predominantly white
lesion of the oral mucosa that cannot be characterized as any
other definable lesion.” About 50% of these lesions contain
a loss of heterozygosity (LOH) in the 3p and 9p21 chro-
mosome arms. This LOH causes the inactivation of p16,
a cyclin-dependent kinase inhibitor (CDKI), and the con-
sequent alteration of cell cycle. A subsequent LOH of the
chromosomal region p13 on chromosome 17 leads to the
loss of tumor suppressor gene p53, the so-called guardian of
the genome, and is associated with progression from hyper-
plasia/mild dysplasia to advanced dysplasia/CIS (carcinoma
insitu) [3]. About 60% of HNSCCs carry the genetic muta-
tion which leads to the suppression of p53; the remaining
cases are supposed to be deleted in other proteins involved
in the same p53 pathway, or to undergo p53-independent
malignant progression. Since 2011, when the first exome
sequencing of HNSCC was published, different altered
pathways of growth have been identified in the cells. Apart
from the herein mentioned deletion of p53, mutations in
phosphoinositide 3-kinase (PI3K)–PTEN–AKT pathway
have been discovered in 10–20% of HNSCCs, especially in
those infected by HPV, whereas the inactivation of NOTCH1
(which encodes for a homonymous cell-surface receptor)
was found in 10–19% of head and neck malignancies. A
smaller proportion of HNSCCs, about 8–9%, is carrier of
mutations in caspase 8 (CASP8), which lead to dysregula-
tion of apoptosis and to the immortal profile acquired by
malignant cells [15].
Genetic alterations and deletions on chromosome 4q, 6p,
8p, 13q, 14q and amplification on 11q13, which causes the
overexpression of cyclin D1, are also responsible for the
metastatic potential acquired by HNSCC cells [3].
EGFR
Since 1960s, when growth factors were first discovered [16],
a large number of studies have been conducted in order to
better understand their involvement in tumor cell growth and
their role as targets of new therapies. In particular, EGFR
(ErbB1 or HER1) has been identified in many human can-
cers, such as lung and breast cancers. Also in HNSCCs, both
EGFR and transforming growth factor alpha (TGF-α, one of
EGFR ligands) have been observed to be overexpressed. In
particular, EGFR is elevated in 38–47% of HNSCCs [17].
EGFR is a cell-surface tyrosine-kinase receptor, and it is a
member of the ErbB family receptors which also includes
HER2/neu (ErbB2), HER3 (ErbB3) and HER4 (ErbB4). The
activation of EGFR by ligands such as EGF or TGF-α leads
to DNA synthesis and cell proliferation through the MAPK,
Akt or IP3 pathways.
Elevated expression of EGFR in HNSCC was observed
in advanced-stage and poorly differentiated tumors, and the
highest levels of EGFR correlate with poor prognosis. It was
also observed that EGFR is upregulated even in the normal
tissue surrounding the tumor, further supporting the field
cancerization theory [18].
Chemotherapy (CHT)
The history of medical treatment for recurrent/metastatic
head and neck cancer (RMHNC) began in 1970 with tra-
ditional drugs, methotrexate (MTX) above all, that offered
limited results to the patients in terms of response rate
(RR) and survival [1921]. In subsequent years, different
drugs such as 5-fluorouracil (5FU), bleomycin, doxoru-
bicin, cyclophosphamide, hydroxyurea and carboplatin
demonstrated good activity with 15–45% RR in phase II
studies [2224], but unfortunately, no phase III trials were
performed to evaluate any advantage versus best support-
ive care (BSC). Then, cisplatin (CDDP) was studied in
this patient population and demonstrated high RR and an
improvement in OS compared to BSC and MTX [2529].
In order to ameliorate efficacy, the association of CDDP
and 5FU was tested in different trials because of its activ-
ity and favorable safety profile [30, 31]. The combination
therapy yielded higher rate of objective response, without
prolonging OS, compared to single-agent treatment. As a
consequence, CDDP plus 5FU became the main regimen
used worldwide, because response may be so important in
these often symptomatic patients, and moreover, toxicity
was manageable with appropriate supportive care. More
complex regimens combining CDDP, MTX, vincristine
and bleomycin were studied and compared with CDDP
alone or in combination with 5FU, with poorer survival
results and high rate of toxicity, so they warranted no
further studies [32]. With the introduction of taxanes,
demonstrating promising activity in terms of RR in HNC
[3337], investigators tried to improve the efficacy of
treatment with paclitaxel and CDDP in combination. A
phase III trial that compared CDDP plus 5FU and CDDP
Medical Oncology (2018) 35:37
1 3
37 Page 4 of 12
plus paclitaxel showed no difference in survival between
the two arms, with similar toxicity [38]. The same group
tried to get better results with high-dose paclitaxel and
CDDP, but the trial showed no survival advantage and
excessive toxicity in the experimental arm [39]. The effi-
cacy of paclitaxel was also evaluated in a phase II study
in combination with carboplatin and demonstrated a
disease control rate (DCR) of 51.8% and few G3–4 side
effects [40]. Recently, buparlisib, a pan-PI3K inhibitor,
was studied in a phase II trial with paclitaxel [41]. In this
trial, patients with RMHNC resistant to platinum-based
CHT were randomly assigned to receive second-line treat-
ment with paclitaxel plus buparlisib or placebo. Median
progression-free survival (PFS) was 4.6months in the
buparlisib group and 3.5months in the placebo group
(P=0.011, hazard ratio [HR] 0.65). The treatment with
buparlisib was associated with a manageable safety pro-
file with hyperglycemia being the most common adverse
event. This study suggests that buparlisib plus paclitaxel
could be an effective second-line treatment, but a phase III
trial is warranted to confirm these promising results. Nab-
paclitaxel (paclitaxel bound to nanoparticle albumin as a
delivery vehicle) is a new taxane with good clinical activ-
ity in many solid tumors. There are no data about its activ-
ity as monotherapy in RMHNC neither about efficacy in
patients that progressed during treatment with a different
taxane. Nonetheless, a retrospective analysis of patients
treated with nab-paclitaxel monotherapy, conducted in a
single institution, showed clinical benefit for patients with
taxane-resistant disease [42].
Cabazitaxel is a second-generation taxane that improves
survival in patients with metastatic castration-resistant
prostate cancer resistant to docetaxel [43]. Unfortunately,
a phase II trial showed that cabazitaxel has low activity in
RMHNC previously treated with platinum-based therapy,
and it has an unfavorable toxicity profile [44].
Pemetrexed is a drug with promising activity that was
evaluated in combination with CDDP in a phase III study.
This large trial randomized 795 patients with RMHNC to
CDDP plus pemetrexed versus CDDP plus placebo. The
results were negative as an advantage in OS, the primary
endpoint, was not reached. OS in the pemetrexed–CDDP
arm was 7.3months, while in the placebo–CDDP arm it
was 6.3months (HR 0.87; 95% confidence interval [CI],
0.75–1.02; P=0.082). Looking at subgroups, in patients
with performance status 0 or 1, pemetrexed–CDDP
led to longer OS and PFS than placebo–CDDP (8.4 vs.
6.7months; HR 0.83; P=0.026; 4.0 vs. 3.0months;
HR 0.84; P=0.044, respectively). Also among patients
with oropharyngeal cancers, pemetrexed–CDDP resulted
in longer OS and PFS than placebo–CDDP (9.9 vs.
6.1months; HR 0.59; P=0.002; 4.0 vs. 3.4months; HR,
0.73; P=0.047, respectively). Despite this interesting
result, the combination cannot be considered a standard
treatment [45].
Since the activity of doublet with classical agents was dis-
appointing, investigators tried to get better results in phase
II trials with more complex regimens including paclitaxel,
ifosfamide and platinum analogue, achieving high rate of
objective response and median OS of 9–11months, but tox-
icity has always been a critical issue [46, 47].
Monoclonal antibodies (mAbs)
Cetuximab is a recombinant IgG1 antibody directed against
the external domain of EGFR, which blocks ligand-medi-
ated activation of EGFR pathway and stimulates antibody-
dependent cellular cytotoxicity [48, 49]. The efficacy of
cetuximab in RMHNC was first evaluated as monotherapy
in patients refractory to platinum-based therapy [50]. In this
trial, DCR was 46% and median time to progression 70days;
treatment was well tolerated. On the base of these findings,
a phase III trial was conducted by the Eastern Cooperative
Oncology Group, evaluating the activity of CDDP plus
cetuximab or placebo [51]. The addition of cetuximab to
CDDP significantly improved RR (overall RR was 26% in
experimental arm vs. 10% in placebo arm) without prolong-
ing significantly OS and PFS. In order to optimize the effi-
cacy of treatment, cetuximab was added to a standard dou-
blet with CDDP and 5FU in the EXTREME study [52]. This
trial enrolled 442 patients with untreated RMHNC that were
randomized to CDDP or carboplatin and 5FU or the same
combination plus cetuximab at the standard dose of 400mg
per square meter for the first dose followed by 250mg per
square meter weekly. Patients received up to 6 cycles of
CHT. Cetuximab was maintained until progression or unac-
ceptable toxicity. The primary endpoint was OS, and for the
first time after many years the experimental arm improved
significantly OS with an absolute advantage of 2.6months
(7.4months in the CHT arm vs. 10.1 in the CHT plus cetuxi-
mab arm). The addition of cetuximab also prolonged PFS
and increased the RR. The incidence of grade 3–4 adverse
events was comparable between the two treatments, except
for higher rates of skin toxicity, hypomagnesemia and sepsis
in the cetuximab group. There were no cetuximab-related
deaths. The benefit of cetuximab was greater for patients
younger than 65years, those with Karnofsky performance
status≥80% and those who received CDDP. The results of
this study led to consider CDDP plus 5FU and cetuximab,
the so-called EXTREME regimen, the new standard of care
for patients with RMHNC and good performance status.
This regimen might result difficult to administer for many
patients because of the risk of adverse events related to high
dose of CDDP. As a consequence, some authors tried to find
an alternative combination with the same efficacy. Nakano
Medical Oncology (2018) 35:37
1 3
Page 5 of 12 37
and colleagues [53] conducted a retrospective analysis of
patients with RMHNC treated with a cetuximab-containing
regimen. The efficacy of weekly paclitaxel plus cetuximab
was compared to the EXTREME regimen. A total of 86
patients were included. Response rates were similar in the
two cohorts (45% in the paclitaxel–cetuximab arm vs. 51%
in the CDDP–5FU–cetuximab arm), while patients treated
with paclitaxel and cetuximab had a significant advantage
in PFS (6.0 vs. 5.0months, P=0.027). This schedule could
represent a useful alternative option for patients refractory
or ineligible to CDDP.
To avoid the risks of toxicity associated with 5FU and the
need of a central catheter for the 96-h infusions, Bossi etal.
[54] tried to substitute 5FU with paclitaxel and compared a
3-drug (CDDP–paclitaxel–cetuximab) with a two-drug regi-
men (CDDP and cetuximab) in the phase II non-inferiority
study B490. A total of 201 patients were randomized 1:1 to
each regimen. For the primary endpoint, PFS, the 2-drug
arm resulted non-inferior to the 3-drug one (6 vs. 7months,
HR 0.99). There were also no differences in OS between
the two groups of patients, with a median OS of 13months
for patients receiving CDDP–cetuximab and 11months for
those treated with CDDP, paclitaxel and cetuximab with an
HR of 0.77. The overall RR was 41.8% with the two drugs
versus 51.7% with the addition of paclitaxel. The results
in terms of all efficacy endpoints were similar or supe-
rior to those reported in the EXTREME study. Grade≥3
adverse events were 76% with CDDP and cetuximab, 73%
with CDDP, paclitaxel and cetuximab. No toxic deaths
were observed, and compared to EXTREME, the rate of
life-threatening toxicities (cardiac and septic) was reduced.
This study showed that 5FU can be substituted by pacli-
taxel, mostly in patients that need a tumor shrinkage, and
that the 2-drug regimen can also be an option with similar
results in terms of PFS and OS. These regimens may repre-
sent a potential less toxic backbone for new combinations
with emerging immunotherapies, such as the new checkpoint
inhibitors.
Panitumumab is a fully humanized anti-EGFR mAb used
for the treatment of metastatic colorectal cancer [55]. In the
SPECTRUM study, patients with RMHNC candidates to a
first-line treatment were randomly assigned to CHT with
CDDP and 5FU or the same treatment plus panitumumab
[56]. Panitumumab was administered on day 1 of each cycle
before cytotoxic drugs at the dosage of 9mg/kg. CHT con-
tinued until disease progression or for a maximum of six
cycles. Patients in the panitumumab group could receive
the mAb until disease progression or unacceptable toxicity.
Unfortunately the trial did not meet OS, its primary end-
point. Median OS was 11.1months in the panitumumab
arm and 9months in CHT only arm (HR 0.873, P=0.14).
Median OS was longer for patients with p16-negative tumors
in the panitumumab arm than in the control arm (11.7 vs.
8.6months, HR 0.73, P=0.015). Grade 3–4 adverse events
were more frequent with CHT plus panitumumab. Panitu-
mumab was then studied in the PARTNER trial, a phase
II study that evaluated the activity of a first-line treatment
with CDDP and docetaxel with or without panitumumab in
patients with RMHNC [57]. In order to limit toxicity, only
patients younger than 70years were enrolled and primary
prophylaxis with granulocyte colony-stimulating factors was
mandatory. PFS, the primary endpoint, was 6.9months in
the experimental arm versus 5.5months in the CHT alone
arm (HR 0.62, P=0.048). There was no difference in OS
between the two treatment groups (12.9months with CHT
plus panitumumab and 13.8months with CHT alone, HR
1.103), while grade 3–4 adverse events were more frequent
with CHT plus panitumumab than with CHT alone. Follow-
ing these data, panitumumab is not indicated in the treatment
of RMHNC.
Bevacizumab is a humanized mAb directed against
VEGF, approved for many solid tumors. VEGF hyper-
expression is correlated to poor prognosis in patients with
HNC. Preclinical and clinical activities of a combined treat-
ment with cetuximab and bevacizumab were evaluated in
human endothelial cells, head and neck and lung cancer
xenografts model systems and, afterward, in patients with
RMHNC [58]. This chemo-free treatment demonstrated
growth inhibition invitro and invivo and reduced tumor vas-
cularization. In the clinical phase II trial of the same report,
46 patients were enrolled. The RR was 16% and the disease
control rate (DCR) 73%; median PFS was 2.8months and
median OS 7.5months. The treatment was well tolerated
and grade 3–4 adverse events occurred in less than 10% of
patients.
Bevacizumab was also studied in combination with pem-
etrexed in patients with RMHNC in first-line setting in a
phase II trial [59]. The 2-drug regimen demonstrated prom-
ising clinical activity. Time to progression, the primary end-
point, was 5months, while OS was 11.3months and DCR
86%. Unfortunately, frequent serious bleeding events (15%),
including two deaths, grade 3–4 neutropenia and sepsis were
indeed a real concern for the safety of the regimen.
Tyrosine‑kinase inhibitors (TKIs)
Gefitinib and erlotinib are the two anti-EGFR TKIs approved
for the treatment of advanced non-small cell lung cancer
patients. These drugs target one of the most important path-
ways in lung and head and neck carcinogenesis and have
been tested in RMHNC.
In a phase II study, gefitinib obtained a RR of 10.6%
and a DCR of 53% at the dose of 500mg daily. Median
OS was 8.1months, and the drug was well tolerated [60].
A following trial evaluated the activity of gefitinib at the
Medical Oncology (2018) 35:37
1 3
37 Page 6 of 12
recommended dosage of 250mg daily [61]. The lower dos-
age, as used in lung cancer, had less activity than the dose
of 500mg daily: A partial RR of 1.4% and median OS of
5.5months were reported. Most interesting, a phase III
trial designed to compare gefitinib 250mg daily or 500mg
daily with the old MTX in second-line demonstrated that
gefitinib 250 and 500mg/day did not improve OS com-
pared to MTX, and the primary endpoint of the study was
not reached [62].
Gefitinib was also tested in addition to docetaxel [63].
The study was closed at an interim analysis for inefficacy.
The addition of gefitinib to docetaxel did not improve OS
in these patients.
Erlotinib was also evaluated in this setting and dem-
onstrated modest activity as monotherapy [64]. RMHNC
patients were treated with erlotinib 150mg/day. Objective
response rate was 4.3%. The median PFS was 9.6weeks,
and the median OS was 6months. Rash and diarrhea were
the main adverse events. Erlotinib was also combined with
bevacizumab [65] in a phase II study enrolling 48 patients,
treated with erlotinib 150mg/day and bevacizumab 15mg/
kg every 3weeks. Fifteen percentage of patients obtained
an objective response, and 31% had stable disease. Median
OS was 7.1months, and the treatment was well tolerated.
The trial suggests that blocking both EGFR and VEGF
might have activity in HNSCC, but no further results have
been published yet.
Afatinib is an irreversible pan-human epidermal recep-
tor (pan-HER) inhibitor that downregulates ErbB signal-
ing by covalently binding to the kinase domain of EGFR,
HER2 or HER4. In a phase II study, afatinib demonstrated
anti-tumor activity comparable to cetuximab. In this
trial, 124 pretreated patients were randomized to receive
afatinib 50mg/day or cetuximab 250mg per square meter
weekly until disease progression or intolerable toxicity
(stage I); then, a crossover to the other treatment (stage II)
was permitted. The first stage of the study demonstrated
similar tumor shrinkage, RR, DCR and PFS with the two
drugs. In stage II, DCR was higher with afatinib than with
cetuximab, suggesting that afatinib after cetuximab fail-
ure may be the optimal sequence. This is related to a lack
of cross-resistance between the two agents, but it is still
unclear why afatinib used after and not before cetuximab
yields a better DCR [66].
On the base of these results, the activity of afatinib was
further investigated in a phase III trial of second-line treat-
ment. It was associated with significantly higher DCR and
longer PFS compared to MTX. Moreover, patients treated
with afatinib had less pain and a significant delay in time
to clinical worsening. These favorable outcomes did not
translate into a prolonged OS, a secondary endpoint of
the trial [67].
Immunotherapy
Despite new advances in conventional therapies such as
surgery, RTX and CHT, survival of HNSCC is still poor.
Furthermore, these strategies, although conventional, can
result in serious complications, from pain to dysphagia and
malnutrition, risk of infection and psychological distress
[68, 69]. Patients suffering from advanced HNSCC are
faced with challenging treatments, so researchers have the
tremendous task to discover and develop new therapies
that combine high efficacy as well as low toxicity. Coming
from studies on the molecular biology and immunology of
cancer, immunotherapy has recently opened new interest-
ing perspectives for HNSCC treatment.
It is now well known that cancer cells develop the abil-
ity to evade anti-tumor immune attacks either by inhibiting
recognition of cancer-specific antigens by T cells, or by
causing dysfunction of CD8 cytotoxic T cells (CTL). The
possibility to revert these cellular alterations is the basis
of cancer immunotherapy, working under the regulation
of positive and negative co-signaling pathways, the so-
called immunologic checkpoints. The checkpoints of pro-
grammed cell death 1 (PD-1) and programmed cell death
ligand 1 (PD-L1) have important roles in the formation
of “immune privilege” regions, viral persistence, tumor
development and immune evasion [7072]. Following
“in vitro” and preclinical tests, various immune check-
point inhibitors (ICIs), such as anti-PD-1 mAbs and anti-
CTLA-4 mAbs, have demonstrated the potential to control
cancer by immune activation [7380]. Immune checkpoint
blockade is able to reactivate dysfunctional or exhausted T
cells by restoring tumor-specific immunity, a high-quality
program aimed at eliminating cancer cells (Fig.1).
The results of two recently published trials, the open-
label, multicenter, phase Ib trial (KEYNOTE-012) and
the randomized, open-label, phase III trial (CheckMate
141) [8183], demonstrated the clinical activity of two
antagonists of the PD-1/PD-L1 axis (pembrolizumab and
nivolumab) and led to their approval by the US Food and
Drug Administration (FDA) as first molecular-targeted
therapeutic drugs for HNSCC.
The PD‑1/PD‑L1 axis
PD-1 is a surface transmembrane glycoprotein consisting
of 268 amino acids, which belongs to the CD28/CTLA-4/
ICOS co-stimulatory receptor family. Its structure consists
of an extracellular region, a hydrophobic transmembrane
region and an intracellular region. Two independent tyros-
ine residues are located in the intracellular region, at the
tail end [8486]. The corresponding PD-1 ligands (PD-L1
Medical Oncology (2018) 35:37
1 3
Page 7 of 12 37
and PD-L2) belong to the B7 superfamily, which includes
the important B7–1 (cluster of differentiation [CD]80) [71,
87]. Similar to PD-1, the structures of PD-L1 and PD-L2
mainly comprise an extracellular region, a hydrophobic
transmembrane region and an intracellular region with a
short cytoplasmic tail of unknown function [84, 87]. The
affinity of the PD-L1 extracellular region with PD-1 is
lower than that of the PD-L2 extracellular region, but it
can also bind with the B7-A (CD80) extracellular region
[71, 84, 87].
PD-1 is expressed on activated T cells after induction
by a T cell antigen receptor and cytokine receptor [84],
and it is also expressed at low levels on double-negative
(CD4–CD8–) T cells in the thymus, activated natural
killer T cells, B cells and monocytes [71, 84]. PD-L1 is
expressed constitutively at low levels on antigen-presenting
cells (APCs), but this scanty expression can be enhanced
by inflammatory cytokines such as type I and type II
interferons, tumor necrosis factor α and VEGF [84, 88].
Increased expression of PD-1/PD-L1 in the microenviron-
ment of HNSCC is independent of HPV status [89].
Different mechanisms are involved in the upregulation
of PD-L1 expression by tumor cells. One possibility is the
activation of the EGFR and the PI3K–Akt or Janus kinase
2/signal transducer and activator of transcription 1 signal-
ing pathways [84, 90, 91]. Other mechanisms involved are
the amplification of genes coding PD-L1 (9p24.1) [84, 90]
and the induction of Epstein–Barr virus (EBV). Moreo-
ver, in tumor microenvironment, the stimulatory effects of
inflammatory factors can also induce PD-L1 expression,
interferon-γ being the most important stimulating factor
[72, 84, 88, 9193].
T cells are activated by a dual control system involving a
specific binding between a T cell receptor and a major his-
tocompatibility complex class and the interaction between
co-stimulatory molecules and the corresponding receptor
Fig. 1 a. Tumor cells can express PDL-1, which is normally
expressed on antigen-presenting cells (APCs) of the immune system
in order to inhibit an exaggerate immune response. b. New antibody
drugs have been developed to prevent the inactivation of immune
system against tumors. Nivolumab and pembrolizumab are the main
humanized antibodies tailored to block PD-1. Durvalumab is an
experimental IgG1 mAb against PDL-1
Medical Oncology (2018) 35:37
1 3
37 Page 8 of 12
on the T cell surface. To prevent T cell overstimulation, T
cell activity is finely regulated at checkpoints [71, 85, 86,
88]. After invasion by tumor cells, these signals are used to
inhibit T cell activation so as to evade attack by the immune
system. At present, a variety of inhibitors of immune check-
points have been studied, mainly mAbs already tested in the
clinical context. The anti-tumor effect is realized by multiple
actions comprising the inhibition of the activity of immune
checkpoints, the blockade of immunosuppression in the
tumor microenvironment and the reactivation of the immune
response of T cells to the tumor [71, 72, 8184, 86, 88, 94].
Clinical application inHNC
PD‑1‑targeted drugs
The main PD-1-targeted drugs used to treat HNSCC are
pembrolizumab and nivolumab, both of which are human-
ized PD-1-inhibiting IgG4 mAbs with high specificity for
receptors [8183]. They have been used for the treatment
of patients with recurrent/metastatic HNSCC with disease
progression on or after platinum-containing CHT (Table1).
Pembrolizumab
Seiwert etal. [81] experienced pembrolizumab in a popu-
lation of 60 patients with recurrent/metastatic HNSCC
(KEYNOTE-O12). PD-L1 expression of≥1% was present
in tumor cells of all patients. The majority of cases (62%)
were HPV−, whereas 38% were HPV+. Pembrolizumab
(10mg/kg body weight, intravenous [iv]) was adminis-
tered biweekly for 2years. Median duration of follow-up
was 14 (range 4–14) months. The ORR was 18% (25% for
HPV+ and 14% for HPV−). While the median PFS was
only 2months, interestingly the median OS was 13months.
Treatment-related adverse events were reported in 63% of
the cases (17% grade 3–4), mainly fatigue and skin toxicity,
but there were no treatment-related deaths. Chow etal. [82]
used pembrolizumab in 132 adult patients with recurrent/
metastatic HNSCC irrespective of PD-L1 or HPV status.
Fifty-seven percent of the patients previously received two
or more lines of CHT. Most of them (71%) were HPV−.
Pembrolizumab (200mg, iv) was administered once every
3weeks for 2years. The median duration of follow-up was 9
(range 3–11) months. The ORR was 18%, and the response
rate of HPV+was 32%, while that of HPV−was 14%. A
statistically significant increase in ORR was observed for
PD-L1-positive versus PD-L1-negative patients (22 vs. 4%;
P=0.021). The median PFS was 2months, and the median
OS was 8months. Treatment-related adverse events were
reported in 62% of the patients, 9%≥grade 3. There were
no treatment-related deaths. Bauml etal. [95] used pem-
brolizumab in platinum- and cetuximab-refractory HNC.
Pembrolizumab (200mg, iv) was administered once every
3weeks for 2years in 50 patients. The median duration of
follow-up was 6.8months. ORR was 16% (95% CI 11–23%),
with a median duration of response of 8months (range 2+
to 12+months); 75% of the patients were still in response at
the time of analysis. RR was similar in all HPV and PD-L1
subgroups. Median PFS was 2.1months, and median OS
was 8months. Sixty-four percent of the patients experienced
a treatment-related adverse event, and 15% experienced a
grade≥3 event. Seven patients (4%) discontinued treatment,
and one treatment-related death was registered. That trial
showed that pembrolizumab demonstrated clinically mean-
ingful anti-tumor activity and an acceptable safety profile in
recurrent/metastatic HNSCC previously treated with plati-
num and cetuximab.
Nivolumab
Nivolumab recently demonstrated to be active in recurrent/
metastatic HNSCC and gained approval by the FDA after
the pivotal phase III trial by Ferris etal. [83]. In that trial,
361 patients with recurrent/metastatic HNSCC were rand-
omized in a 2:1 ratio to nivolumab (3mg/kg, iv) once every
2weeks, or standard therapy (either methotrexate, docetaxel,
or cetuximab of the investigator’s choice) until disease pro-
gression. The median duration of follow-up was 5.1months.
The ORR and partial response rate (PRR) were 13.3 and
10.8% for nivolumab, and 5.8 and 5.0% for standard therapy,
Table 1 Anti-PD-1 drugs currently tested for R/M HNSCC
Authors Drug Year Phase No. of patients mOS (months) mPFS (months)
Seiwert etal. [81] Pembrolizumab 2016 Ib 60 13 2
Chow etal. [82] Pembrolizumab 2016 132 8 2
Bauml etal. [95] Pembrolizumab 2017 50 8 2.1
Ferris etal. [83] Nivolumab versus standard therapy 2016 III 361 7.5—Nivolumab 2—Nivolumab
5.1—Standard therapy 2.3—Standard therapy
Segal etal. [97] Durvalumab (MEDI4736) 2015 I/II 62 Not evaluated Not evaluated
Fury etal. [98] Durvalumab (MEDI4736) 2014 I 50 Not evaluated Not evaluated
Medical Oncology (2018) 35:37
1 3
Page 9 of 12 37
respectively. The median PFS was 2months for nivolumab
and 2.3months for standard therapy. The median OS for
nivolumab was 7.5months and 36% of patients reached
1-year PFS, while for standard therapy it was 5.1months
and 16.6% patients reached 1-year PFS. Treatment-related
adverse events (fatigue, nausea and skin rash) were recorded
in 58.9% of the nivolumab patients compared to 77.5% in the
standard therapy group. Grade>3 treatment-related adverse
events were experienced by 13.1% of the nivolumab patients
and by 35.1% of the standard therapy patients. In conclusion,
nivolumab prolonged survival, as compared with standard
therapy, among patients with platinum-refractory HNSCC,
and was associated with fewer toxic effects of grade 3 or
4 than standard therapy. Moreover, nivolumab delayed the
time to deterioration of patient-reported quality-of-life out-
comes [96] among patients with treatment-refractory cancer
that negatively impacts on quality of life and leads to death.
PD‑L1‑targeted drugs
At present, the main PD-L1-targeted drug for HNSCC treat-
ment is durvalumab (MEDI4736), a human IgG1 mAb that
blocks PD-L1 binding to its receptors [71, 97, 98]. It has
been used for recurrent/metastatic HNSCC in two small tri-
als [97, 98] where it was administered (10mg/kg, iv) once
every 2weeks for 12months. At present, the data have been
published only in abstract form, so it is premature to con-
clude about a possible role of durvalumab in the clinical
practice.
Conclusion
The treatment of RMHNC is a hard task as the disease is
burdened by cumbersome morbidity and high mortality.
Recent trials have demonstrated how useful the advent of
immunotherapy in this dreary landscape could be, high-
lighting superior response and survival rates compared to
standard CHT. Furthermore, a well-defined prerogative of
immunotherapy seems to be a reduced rate of adverse events,
making it a safe and powerful new anticancer treatment.
Pembrolizumab has the highest ORR in HPV+HNSCC
patients and probably has the greatest potential among
PD-1-/PD-L1-targeting drugs. In the effort to find an effec-
tive and less toxic treatment, a promising approach will be
the evaluation of the combination of pembrolizumab and
radiotherapy in the loco-regionally advanced disease, with
the aim of decreasing local and distant relapse in the HPV
subgroup. For the next future, we need more clinical data
on the efficacy of the single agents, but it will be of utmost
interest the unexplored option to combine two or more drugs
together (immunotherapy or CHT), as well as the possibility
to substitute one drug in the standard EXTREME protocol
(5-FU for example) with a PD-1-/PD-L1-targeting drugs.
Finally, it is well known how cetuximab, a mAb directed
to EGFR, can be effective in HNC. As it has been demon-
strated, the blockade of PD-1/PD-L1 signaling can improve
anticancer effects of anti-EGFR agents [99], so the combined
application of PD-1-/PD-L1-targeted drugs and cetuximab
merits further research in order to discover favorable and
long-lasting clinical effects.
Compliance with ethical standards
Conflict of interest All authors declare that they have no conflict of
interest.
References
1. Kamangar F, Dores GM, Anderson WF. Patterns of cancer inci-
dence, mortality, and prevalence across five continents: defining
priorities to reduce cancer disparities in different geographic
regions of the world. J Clin Oncol. 2006;24(14):2137–50.
2. Global Burden of Disease Cancer Collaboration, Fitzmaurice C,
Allen C, etal. Global, Regional, and National Cancer Incidence,
Mortality, Years of Life Lost, Years Lived With Disability, and
Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to
2015: A Systematic Analysis for the Global Burden of Disease
Study. JAMA Oncol. 2016.
3. Kumar V, Abbas AK, Fausto N, Aster JC. Robbins and Cotran
pathologic basis of disease. 8th ed. Philadelphia: Saunders Else-
vier; 2010.
4. Haddad RI, Shin DM. Recent advances in head and neck cancer.
N Engl J Med. 2008;359:1143.
5. Day GL, Blot WJ. Second primary tumors in patients with oral
cancer. Cancer. 1992;70:14.
6. Van Dijk BAC, Gatta G, Capocaccia R, Pierannunzio D, Strojan P,
Licitra L. The RARECARE Working Group. Rare cancers of the
head and neck area in Europe. Eur J Cancer. 2012;48(6):783–96.
7. Gillison ML, etal. Evidence for a causal association between
human papillomavirus and a subset of head and neck cancers. J
Natl Cancer Inst. 2000;92:709–20.
8. Young D, Xiao CC, Murphy B, Moore M, Fakhry C, Day TA.
Increase in head and neck cancer in younger patients due to human
papillomavirus (HPV). Oral Oncol. 2015;51:727–30.
9. Yen TT, Lin WD, Wang CP, etal. The association of smoking,
alcoholic consumption, betel quid chewing and oral cavity cancer:
a cohort study. Eur Arch Otorhinolaryngol. 2008;265:1403–7.
10. Slaughter DP, etal. “Field cancerization” in oral stratified squa-
mous epithelium. Cancer. 1953;6:962.
11. Karatzanis AD, Koudounarakis E, Papadakis I, Velegrakis G.
Molecular pathways of lymphangiogenesis and lymph node
metastasis in head and neck cancer. Eur Arch Otorhinolaryngol.
2012;269:731–7.
12. Madden RE, Gyur L. Translymphnodal passage of tumor cells.
Oncology. 1968;22:281–9.
13. Fisher B, Fisher ER. Transmigration of lymph nodes by tumor
cells. Science. 1966;152:1397–8.
14. Allen CT, Law JH, Dunn GP, Uppaluri R. Emerging
insights into head and neck cancer metastasis. Head Neck.
2013;35(11):1669–78.
15. Stransky N, etal. The mutational landscape of head and neck
squamous cell carcinoma. Science. 2011;333(6046):1157–60.
Medical Oncology (2018) 35:37
1 3
37 Page 10 of 12
16. Cohen S. Origins of growth factors: NGF and EGF. Ann N Y
Acad Sci. 2004;1038:98–102.
17. Kalyankrishna S, Grandis JR. Epidermal growth fac-
tor receptor biology in head and neck cancer. J Clin Oncol.
2006;24(17):2666–72.
18. Grandis JR, Tweardy DJ. Elevated levels of transforming growth
factors alpha and epidermal growth factor receptor messenger
RNA are early markers in carcinogenesis in head and neck can-
cer. Cancer Res. 1993;53:3579–84.
19. Taylor SG IV, McGuire WP, Hauck WW, Showel JL, Lad TE.
A randomized comparison of high dose methotrexate versus
standard-dose weekly therapy in head and neck squamous can-
cer. J Clin Oncol. 1984;2(9):1006–11.
20. Woods RL, Fox RM, Tattersall MH. Methotrexate treatment of
squamous-cell head and neck cancers: dose-response evaluation.
Br Med J (Clin Res Ed). 1981;282(6264):600–2.
21. Pitman SW, Miller D, Weichselbaum R. Initial adjuvant therapy
in advanced squamous-cell carcinoma of the head and neck
employing weekly high dose methotrexate with leucovorin res-
cue. Laryngoscope. 1978;88(4):632–8.
22. Carter SK, Slavik M. Current investigational drugs of interest
in the chemotherapy program of the National Cancer Institute.
Natl Cancer Inst Monogr. 1977;45:101–21.
23. Wittes RE. Chemotherapy of head and neck cancer. Otolaryngol
Clin North Am. 1980;13:515–20.
24. Eisemberger M, Hornedo J, Silva H, Donehower R, Spaulding
M, Van Echo D. Carboplatin (NSC-241.240): an active platinum
analogue for the treatment of squamous-cell carcinoma of the
head and neck. J Clin Oncol. 1986;4:1506–9.
25. Wittes RE, Cvitkovic E, Shah J, Gerold FP, Strong EW.
CIS-Dichlorodiammineplatinum(II) in the treatment of epi-
dermoid carcinoma of head and neck. Cancer Treat Rep.
1977;61(3):359–66.
26. Wittes R, Heller K, Randolph V, Howard J, Vallejo A, Farr H,
etal. Cis-Dichlorodiammineplatinum(II)-based chemotherapy as
initial treatment of advanced head and neck cancer. Cancer Treat
Rep. 1979;63(9–10):1533–8.
27. Grose WE, Lehane DE, Dixon DO, Fletcher WS, Stuckey
WJ. Comparison of methotrexate and cisplatin for patients
with advanced squamous-cell carcinoma of the head and neck
region: a Southwest Oncology Group Study. Cancer Treat Rep.
1985;69(6):577–81.
28. Morton RP, Rugman F, Dorman EB, Stoney PJ, Wilson JA, Cor-
mick MC, etal. Cisplatinum and bleomycin for advanced or recur-
rent squamous cell carcinoma of the head and neck: a randomized
factorial phase III controlled trial. Cancer Chemother Pharmacol.
1985;15(3):283–9.
29. Liverpool Head and Neck Oncology Group. A phase III rand-
omized trial of cisplatinum, methotrexate, cisplatinum+metho-
trexate and cisplatin+5-FU in end stage squamous carcinoma of
head and neck. Br J Cancer. 1990;62(1):171.
30. Forastiere AA, Metch B, Schuller DE, Ensley JF, Hutchins LF,
Triozzi P, etal. Randomized comparison of cisplatin plus fluoro-
uracil and carboplatin plus 5fluorouracil versus methotrexate in
advanced squamous-cell carcinoma of the head and neck: a South-
west Oncology Group Study. J Clin Oncol. 1992;10(8):1245–51.
31. Jacobs C, Lyman G, Velez-Garcia E, Sridhar KS, Knight W,
Hochster H, etal. A phase III randomized study comparing cis-
platin and fluorouracil as single agents and in combination for
advanced squamous cell carcinoma of the head and neck. J Clin
Oncol. 1992;10(2):257–63.
32. Clacel M, Vermorken JB, Cognetti F, Cappelaere P, de Mulder
PH, Schornagel JH, etal. Randomized comparison of cisplatin,
methotrexate, bleomycin and vincristine (CABO) versus cisplatin
and 5-fluorouracil (CF) versus cisplatin (C) in recurrent or meta-
static squamous-cell carcinoma of the head and neck. A phase III
study of the EORTC Head and Neck Cancer Cooperative Group.
Ann Oncol. 1994;5(6):521–6.
33. Catimel G, Verweij J, Mattijssen V, Hanauske A, Piccart M, Wan-
ders J, etal. Docetaxel (Taxotere). An active drug for the treat-
ment of patients with advanced squamous-cell carcinoma of the
head and neck—EORTC Early Clinical Trials Group. Ann Oncol.
1994;5:233–7.
34. Al Dreyfuss, Clark JR, Norris CM, Rossi RM, Lucarini JW, Busse
PM, etal. Docetaxel: an active drug for squamous-cell carcinoma
of the head and neck. J Clin Oncol. 1996;14:1672–8.
35. Schrijvers D, Vermorken JB. Taxanes in the treatment of head and
neck cancer. Curr Opin Oncol. 2005;17:218–24.
36. Forastiere AA, Shank D, Neuberg D, Taylor SG 4th, DeConti RG,
Adams G. Final report of a phase II evaluation of paclitaxel in
squamous-cell carcinoma of the head and neck: an Eastern Coop-
erative Oncology Group trial (PA390). Cancer. 1998;82:2270–4.
37. Smith RE, Thornoton DE, Allen J. A phase II trial of paclitaxel
in squamous-cell carcinoma of the head and neck with correlative
laboratory studies. Semin Oncol. 1995;22:41–6.
38. Gibson MK, Li Y, Murphy B, Hussain MH, De Conti RC, Ensley
J, etal. Randomized phase III evaluation of cisplatin plus fluoro-
uracil versus cisplatin plus paclitaxel in advanced head and neck
cancer (E1395): an intergroup trial of the Eastern Cooperative
Oncology Group. J Clin Oncol. 2005;23(15):3562–7.
39. Forastiere AA, Leong T, Rowinsky E, Murphy BA, Vlock DR,
DeConti RC, etal. Phase III comparison of high-dose pacli-
taxel+cisplatin+granulocyte colony-stimulating factor ver-
sus low-dose paclitaxel+cisplatin in advanced head and neck
cancer: Eastern Oncology Group Study E1393. J Clin Oncol.
2001;19(4):1088–95.
40. Ferrari D, Fiore J, Codecà C, Di Maria G, Bozzoni S, Bordin V,
etal. A phase II study of carboplatin and paclitaxel for recur-
rent or metastatic head and neck cancer. Anticancer Drugs.
2009;20(3):185–9.
41. Soulieres D, Faivre S, Mesia R, Remenar E, Li SH, Karpenko
A, etal. Buparlisib and paclitaxel in patients with platinum-pre-
treated recurrent or metastatic squamous-cell carcinoma of the
head and neck (BERIL-1): a randomized, double-blind, placebo-
controlled phase II trial. Lancet Oncol. 2017;18(3):323–35.
42. Lay J, Wildes TM, Daly K, Oppelt P, Adkins D. Clinical benefit
of nanoparticle albumin-bound-paclitaxel in recurrent/metastatic
head and neck squamous-cell carcinoma resistant to cremophor-
based paclitaxel or docetaxel. Med Oncol. 2017;34:28.
43. de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP,
Kocak I, etal. Prednisone plus cabazitaxel or mitoxantrone for
metastatic castration-resistant prostate cancer progressing after
docetaxel treatment: a randomized open label trial. Lancet.
2010;376(9747):1147–54.
44. Machielis JP, Van Maanen A, Vandenbulcke JM, Filleul B, Seront
E, Henry S, etal. Randomized phase II study of Cabazitaxel ver-
sus Methotrexate in patients with recurrent and/or metastatic
squamous-cell carcinoma of the head and neck previously treated
with platinum-based therapy. Oncologist. 2016;21(12):1416–7.
45. Urba S, van Herpen CM, Sahoo TP, Shin DM, Licitra L, Mezei
K, etal. Pemetrexed in combination with cisplatin versus cisplatin
monotherapy in patients with recurrent or metastatic head and
neck cancer: final results of a randomized, double-blind, placebo-
controlled, phase III study. Cancer. 2012;118(19):4694–705.
46. Shin DM, Khuri FR, Glisson BS, Ginsberg L, Papadimitrakopou-
lou VM, Clayman G, etal. Phase II study of paclitaxel, ifosfamide,
and carboplatin in patients with recurrent or metastatic head and
neck squamous cell carcinoma. Cancer. 2001;91(7):1316–23.
47. Shin DM, Gibson BS, Khuri FR, Ginsberg L, Papadimitrakopou-
lou V, Lee JJ, etal. Phase II study of paclitaxel, ifosfamide and
cisplatin in patients with recurrent head and neck squamous-cell
carcinoma. J Clin Oncol. 1998;16:1325–30.
Medical Oncology (2018) 35:37
1 3
Page 11 of 12 37
48. Li S, Schmitz KR, Jeffrey PD, Wiltzius PD, Kussie P, Ferguson
KM. Structural basis for the inhibition of the epidermal growth
factor receptor by cetuximab. Cancer Cell. 2005;7:301–11.
49. Taylor RJ, Chan SL, Wood A. FcγRIII polymorphism and cetuxi-
mab induced cytoxicity in squamous-cell carcinoma of the head
and neck. Cancer Immunol Immunother. 2009;58:997–1006.
50. Vermorken JB, Trigo J, Hitt R, Koralewski P, Diaz-Rubio E, Rol-
land F, etal. Open-label, uncontrolled, multicenter phase II study
to evaluate the efficacy and toxicity of cetuximab as single agent
in patients with recurrent and/or metastatic squamous-cell car-
cinoma of the head and neck who failed to respond to platinum-
based therapy. J Clin Oncol. 2007;25(16):2171–7.
51. Burtness B, Goldwasser MA, Flood W, Mattar B, Forastiere AA.
Eastern Cooperative Oncology Group. Phase III randomized trial
of cisplatin plus placebo compared with cisplatin plus cetuximab
in metastatic/recurrent head and neck cancer: an Eastern Coopera-
tive Oncology Group study. J Clin Oncol. 2005;23(34):8646–54.
52. Vermorken JB, Mesia R, Rivera F, Remenar E, Kawecky A, Rottey
S, etal. Platinum-based chemotherapy plus cetuximab in head and
neck canecer. N Engl J Med. 2008;359:1116–27.
53. Nakano K, Marshall S, Taira S, etal. A comparison of weekly
paclitaxel and cetuximab with the EXTREME regimen in the
treatment of recurrent/metastatic squamous cell head and neck
carcinoma. Oral Oncol. 2017;73:21–6.
54. Bossi P, Miceli R, Locati LD, etal. A randomized, phase 2 study
of cetuximab plus cisplatin with or without paclitaxel for the
first-line treatment of patients with recurrent and/or metastatic
squamous cell carcinoma of the head and neck. Ann Oncol.
2017;28(11):2820–6.
55. Peeters M, Cohn A, Kohne CH, Douillard JY. Panitumumab
in combination with cytotoxic chemotherapy for the treatment
of metastatic colorectal carcinoma. Clin Colorectal Cancer.
2012;11:14–23.
56. Vermorken JB, Stohlmacher-Williams J, Davidenko I, Licitra L,
Winquist E, Villanueva C, etal. Cisplatin and fluorouracil with
or without panitumumab in patients with recurrent or meta-
static squamous-cell carcinoma of the head and neck (SPEC-
TRUM): an open-label phase III randomized trial. Lancet Oncol.
2013;14(8):697–710.
57. Wirth LJ, Dakhil S, Kornek G, Axelrod R, Adkins D, Pant S, etal.
PARTNER: an open-label, phase II study of docetaxel/cisplatin
with or without Panitumumab as first-line treatment for recurrent
or metastatic squamous cell carcinoma of the head and neck. Oral
Oncol. 2016;61:31–40.
58. Argiris A, Kotsakis AP, Hoang T, Worden FP, Savvides P, Gib-
son MK, etal. Cetuximab and bevacizumab: preclinical data and
phase II trial in recurrent or metastatic squamous cell carcinoma
of the head and neck. Ann Oncol. 2013;24(1):220–5.
59. Argiris A, Karamouzis MV, Gooding WE, Branstetter BF, Zhong
S, Raez LE, etal. Phase II trial of pemetrexed and bevacizumab
in patients with recurrent or metastatic head and neck cancer. J
Clin Oncol. 2001;29(9):1140–5.
60. Cohen EE, Rosen F, Stadle WM, Recant W, Stenson K, Huo
D, etal. Phase II trial of ZD1839 in recurrent or metastatic
squamous-cell carcinoma of the head and neck. J Clin Oncol.
2003;21(10):1980–7.
61. Cohen EE, Kane MA, Brockstein BE, Mehrotra B, Huo D, Mauer
AM, etal. Phase II trial of gefitinib 250mg daily in patients with
recurrent and/or metastatic squamous-cell carcinoma of the head
and neck. Clin Cancer Res. 2005;11(23):8418–24.
62. Stewart JS, Cohen EE, Licitra L, Van Herpen CM, Khorprasert C,
Soulieres D, etal. Phase III study of gefitinib compared with intra-
venous methotrexate for recurrent squamous-cell carcinoma of the
head and neck [corrected]. J Clin Oncol. 2009;27(11):1864–71.
63. Argiris A, Ghebremichael M, Gilbert J, Lee JW, Sachidanandam
K, Kolesar JM, etal. Phase III randomized, placebo-controlled
trial of docetaxel with or without gefitinib in recurrent or meta-
static head and neck cancer: an Eastern Cooperative Oncology
Group trial. J Clin Oncol. 2013;31(11):1405–14.
64. Soulieres D, Senzer NN, Vokes EE, Hidalgo M, Agarwala SS,
Siu LL. Multicenter phase II study of erlotinib, an oral epider-
mal growth factor receptor tyrosine kinase inhibitor, in patients
with recurrent or metastatic squamous-cell cancer of the head
and neck. J Clin Oncol. 2004;22(1):77–85.
65. Cohen EE, Davis DW, Karrison TG, Seiwert TY, Wong SJ,
Nattam S, etal. Erlotinib and bevacizumab in patients with
recurrent or metastatic squamous-cell carcinoma of the head
and neck: a phase I–II study. Lancet Oncol. 2009;10(3):247–57.
66. Seiwert T, Fayette J, Cupissol D, DelCampo JM, Clement PM,
Hitt R, etal. A randomized, phase II study of afatinib versus
cetuximab in metastatic or recurrent squamous cell carcinoma
of the head and neck. Ann Oncol. 2014;25(9):1813–20.
67. Machiels JP, Haddad RI, Fayette J, Licitra LF, Tahara M, Ver-
morken JB, etal. Afatinib versus methotrexate as second-line
treatment in patients with recurrent or metastatic squamous-
cell carcinoma of the head and neck progressing on or after
platinum-based therapy (LUX Head and Neck 1): an open-label,
randomized phase III trial. Lancet Oncol. 2015;16(5):583–94.
68. Cohen EE, LaMonte SJ, Erb NL, etal. American cancer society
head and neck cancer survivorship care guideline. CA Cancer J
Clin. 2016;66(3):203–39.
69. Epstein JB, Thariat J, Bensadoun R-J, etal. Oral complications
of cancer and cancer therapy: from cancer treatment to survivor-
ship. CA Cancer J Clin. 2012;62(6):400–22.
70. Dorsey K, Agulnik M. Promising new molecular targeted thera-
pies in head and neck cancer. Drugs. 2013;73(4):315–25.
71. Pai SI, Zandberg DP, Strome SE. The role of antagonists of the
PD-1:PD-L1/PD-L2 axis in head and neck cancer treatment.
Oral Oncol. 2016;61:152–8.
72. Pai SI. Adaptive immune resistance in HPV-associated head
and neck squamous cell carcinoma. Oncoimmunology.
2013;2(5):e24065.
73. Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, etal.
Ipilimumab for patients with relapse after allogeneic transplan-
tation. N Engl J Med. 2016;375:143–53.
74. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA,
etal. Improved survival with ipilimumab in patients with meta-
static melanoma. N Engl J Med. 2010;363:711–23.
75. Larkin J, Hodi FS, Wolchok JD. Combined nivolumab and ipili-
mumab or monotherapy in untreated melanoma. N Engl J Med.
2015;373:1270–1.
76. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K,
etal. Nivolumab and ipilimumab versus ipilimumab in untreated
melanoma. N Engl J Med. 2015;372:2006–17.
77. Robert C, Ribas A, Wolchok JD, Hodi FS, Hamid O, etal.
Anti-programmed-death-receptor-1 treatment with pembroli-
zumab in ipilimumab-refractory advanced melanoma: a ran-
domised dose-comparison cohort of a phase 1 trial. Lancet.
2014;384:1109–17.
78. Pardoll DM. The blockade of immune checkpoints in cancer
immunotherapy. Nat Rev Cancer. 2012;12:252–64.
79. Weber JS, Gibney G, Sullivan RJ, Sosman JA, Slingluff CL Jr,
etal. Sequential administration of nivolumab and ipilimumab with
a planned switch in patients with advanced melanoma (Check-
Mate 064): an open-label, randomised, phase 2 trial. Lancet
Oncol. 2016;17:943–55.
80. Robert C, Soria JC, Eggermont AM. Drug of the year: pro-
grammed death-1 receptor/programmed death-1 ligand-1 receptor
monoclonal antibodies. Eur J Cancer. 2013;49:2968–71.
81. Seiwert TY, Burtness B, Mehra R, etal. Safety and clinical activ-
ity of pembrolizumab for treatment of recurrent or metastatic
squamous cell carcinoma of the head and neck (KEYNOTE-012):
Medical Oncology (2018) 35:37
1 3
37 Page 12 of 12
an open-label, multicentre, phase 1b trial. Lancet Oncol.
2016;17(7):956–65.
82. Chow LQ, Haddad R, Gupta S, etal. Antitumor activity of pem-
brolizumab 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.
83. Ferris RL, Blumenschein G, Fayette J, etal. Nivolumab for recur-
rent squamous-cell carcinoma of the head and neck. N Engl J
Med. 2016;375(19):1856–67.
84. Boussiotis VA. Molecular and biochemical aspects of the PD-1
checkpoint pathway. N Engl J Med. 2016;375(18):1767–78.
85. Zhang X, Schwartz JC, Guo X, etal. Structural and functional
analysis of the costimulatory receptor programmed death-1.
Immunity. 2004;20(3):337–47.
86. Cheng X, Veverka V, Radhakrishnan A, etal. Structure and inter-
actions of the human programmed cell death 1 receptor. J Biol
Chem. 2013;288(17):11771–85.
87. Yao Q, Fischer KP, Tyrrell DL, Gutfreund KS. The Pekin duck
programmed death-ligand 1: cDNA cloning, genomic structure,
molecular characterization and mRNA expression analysis. Int J
Immunogenet. 2015;42(2):111–20.
88. Lyford-Pike S, Peng S, Young GD, etal. Evidence for a role of
the PD-1:PD-L1 pathway in immune resistance of HPV-asso-
ciated head and neck squamous cell carcinoma. Cancer Res.
2013;73(6):1733–41.
89. Yu GT, Bu LL, Huang CF, etal. PD-1 blockade attenuates immu-
nosuppressive myeloid cells due to inhibition of CD47/SIRPα
axis in HPV negative head and neck squamous cell carcinoma.
Oncotarget. 2015;6(39):42067–80.
90. Green MR, Monti S, Rodig SJ, etal. Integrative analysis reveals
selective 9p24.1 amplification, increased PD-1 ligand expression,
and further induction via JAK2 in nodular sclerosing Hodgkin
lymphoma and primary mediastinal large B-cell lymphoma.
Blood. 2010;116(17):3268–77.
91. Akbay EA, Koyama S, Carretero J, etal. Activation of the PD-1
pathway contributes to immune escape in EGFR-driven lung
tumors. Cancer Discov. 2013;3(12):1355–63.
92. Concha-Benavente F, Srivastava RM, Trivedi S, etal. Identifica-
tion of the cell-intrinsic and -extrinsic pathways downstream of
EGFR and IFNγ that induce PD-L1 expression in head and neck
cancer. Cancer Res. 2016;76(5):1031–43.
93. Straub M, Drecoll E, Pfarr N, etal. CD274/PD-L1 gene ampli-
fication and PD-L1 protein expression are common events
in squamous cell carcinoma of the oral cavity. Oncotarget.
2016;7(11):12024–34.
94. Ibrahim R, Stewart R, Shalabi A. PD-L1 blockade for cancer treat-
ment: MEDI4736. Semin Oncol. 2015;42(3):474–83.
95. Bauml J. Preliminary results from KEYNOTE-055: Pembroli-
zumab after platinum and cetuximab failure in head and neck
squamous cell carcinoma (HNSCC). J Clin Oncol. 2016: ASCO
meeting abstracts:34(Suppl):abstr 6011.
96. Harrington KJ, etal. Nivolumab versus standard, single-agent
therapy of investigator’s choice in recurrent or metastatic squa-
mous cell carcinoma of the head and neck (CheckMate 141):
health-related quality-of-life results from a randomised, phase 3
trial. Lancet Oncol. 2017;18(8):1104–15.
97. Segal NH. Safety and efficacy of MEDI4736, an anti-PD-L1 anti-
body, in patients from a squamous cell carcinoma of the head
and neck (SCCHN) expansion cohort. J Clin Oncol. 2015:ASCO
meeting abstracts:33(Suppl):abstr 3011.
98. Fury M, Ou SI, Balmanoukian A, etal. Clinical activity and safety
of MEDI4736, an anti-PD-L1 antibody, in patients with head and
neck cancer. Ann Oncol. 2014;25(Suppl 4):iv341.
99. Jie HB, Srivastava RM, Argiris A, etal. Increased PD-1+and
TIM-3+TILs during cetuximab therapy inversely correlate with
response in head and neck cancer patients. Cancer Immunol Res.
2017;5(5):408–16.
... p16 and p53 are closely linked to HPV and its role in the progression and prognosis of HNSCC (45). Various factors, such as tobacco smoking, alcohol abuse and pollutants, can influence the transformation of normal cells into tumor cells (104). Significant therapeutic advancements have been made in the treatment of HNSCC. ...
... Radiotherapy targets specific areas where the tumor is located, using high-energy radiation to destroy cancer cells (165). Combining these treatments with immunotherapy, which activates the body's immune system to recognize and attack cancer cells, has a synergistic effect (104). Immunotherapy helps in enhancing the immune response, making it more effective in recognizing and eliminating cancer cells (166). ...
... It is also essential to evaluate the adequacy of various therapies before combining different immunotherapeutic agents (192). Finally, immunotherapy has shown a high efficacy for aerodigestive malignancies and has paved the way for an optimal methodology aiming at a novel therapeutic approach (104). However, despite the success, further efforts are needed to improve the clinical efficacy in patients with challenging HNSCC. ...
Article
Full-text available
Immunotherapy has become one of the most promising approaches in tumor therapy, and there are numerous associated clinical trials in China. As an immunosuppressive tumor, head and neck squamous cell carcinoma (HNSCC) carries a high mutation burden, making immune checkpoint inhibitors promising candidates in this field due to their unique mechanism of action. The present review outlines a comprehensive multidisciplinary cancer treatment approach and elaborates on how combining immunochemotherapy and immunoradiotherapy guidelines could enhance clinical efficacy in patients with HNSCC. Furthermore, the present review explores the immunology of HNSCC, current immunotherapeutic strategies to enhance antitumor activity, ongoing clinical trials and the future direction of the current immune landscape in HNSCC. Advanced‑stage HNSCC presents with a poor prognosis, low survival rates and minimal improvement in patient survival trends over time. Understanding the potential of immunotherapy and ways to combine it with surgery, chemotherapy and radiotherapy confers good prospects for the management of human papillomavirus (HPV)‑positive HNSCC, as well as other HPV‑positive malignancies. Understanding the immune system and its effect on HNSCC progression and metastasis will help to uncover novel biomarkers for the selection of patients and to enhance the efficacy of treatments. Further research on why current immune checkpoint inhibitors and targeted drugs are only effective for some patients in the clinic is needed; therefore, further research is required to improve the overall survival of affected patients.
... Cancers 2024, 16, 1354 2 of 23 types of HNC havd been distinguished. Here, we focus on the most common histological type of head and neck cancer, namely, squamous cell carcinoma (HNSCC), which accounts for over 90% of HNCs [3,4]. ...
... Platinum-based chemotherapy, paclitaxel (Taxol), docetaxel, and 5-fluorouracil (5-FU) are administered to patients with stages III/IV HNSCC as systemic treatments [6,8]. Cetuximab, a monoclonal antibody that targets the epidermal growth factor receptor (EGFR), applied together with cisplatin (cis-diamminedichloroplatinum(II); CDDP) and 5-FU, is used in recurrent/metastatic HNSCC treatment [3]. Recently, the PD-1 receptor protein-targeting drugs nivolumab and pembrolizumab have also been used in patients with recurrent/metastatic HNSCCs treated previously with platinum-containing CHT. ...
Article
Full-text available
Simple Summary Head and neck squamous cell carcinoma is one of the most common cancers that arises in the upper aerodigestive tract. Patients suffering from this cancer have a high mortality risk, mainly due to local recurrence, resistance to chemo- and radiotherapy, and metastasis. The more aggressive behavior of this tumor is associated with epithelial–mesenchymal transition, a process described in both physiological, primarily during embryonic development, and pathological situations, including the progression of other types of tumors. Epithelial-to-mesenchymal transition is governed by various transcription factors that regulate target gene expression and play a role in the resistance to contemporary head and neck cancer therapies. This review presents the current knowledge of the main transcription factors involved in mesenchymal conversion and discusses their role in head and neck squamous cell carcinoma treatment. The main protein markers associated with this cancer type are also presented. Abstract Head and neck cancers (HNCs) are heterogeneous and aggressive tumors of the upper aerodigestive tract. Although various histological types exist, the most common is squamous cell carcinoma (HNSCC). The incidence of HNSCC is increasing, making it an important public health concern. Tumor resistance to contemporary treatments, namely, chemo- and radiotherapy, and the recurrence of the primary tumor after its surgical removal cause huge problems for patients. Despite recent improvements in these treatments, the 5-year survival rate is still relatively low. HNSCCs may develop local lymph node metastases and, in the most advanced cases, also distant metastases. A key process associated with tumor progression and metastasis is epithelial–mesenchymal transition (EMT), when poorly motile epithelial tumor cells acquire motile mesenchymal characteristics. These transition cells can invade different adjacent tissues and finally form metastases. EMT is governed by various transcription factors, including the best-characterized TWIST1 and TWIST2, SNAIL, SLUG, ZEB1, and ZEB2. Here, we highlight the current knowledge of the process of EMT in HNSCC and present the main protein markers associated with it. This review focuses on the transcription factors related to EMT and emphasizes their role in the resistance of HNSCC to current chemo- and radiotherapies. Understanding the role of EMT and the precise molecular mechanisms involved in this process may help with the development of novel anti-cancer therapies for this type of tumor.
... Various tumors, including laryngeal cancer, are increasingly being treated with immunotherapy (30). It has become common to combine immunotherapy and surgery with radiotherapy and chemotherapy to treat laryngeal cancer (31)(32)(33). A study carried out by us found that the expression of GALNT2 signi cantly increased the expression of immune checkpoint genes, including PD-1, PD-L1, PD-L2 and CTLA-4, indicating that patients with high expression of GALNT2 might be more suitable for immunotherapy. ...
Preprint
Full-text available
GALNT2 is associated with the occurrence and development of many tumors, but its role in laryngeal carcinoma has not been studied. This study used TCGA and GEO databases to analyze GALNT2 expression in laryngeal cancer and adjacent tissues. Western blot, immunohistochemistry, and RT-qPCR were used to validate results. Survival analysis assessed the impact of GALNT2 expression on patient prognosis. KEGG and GSEA enrichment analyses identified biological pathways associated with GALNT2. CIBERSORT algorithm examined the effect of GALNT2 on tumor immune cell infiltration. ENCORI database built a ceRNA network. TIMER2.0 database studied GALNT2 expression in various tumors. The results find that GALNT2 is highly expressed in laryngeal cancer patients and has prognostic implications. Enrichment analysis showed that GALNT2 is involved in various carcinogenic pathways. GALNT2 expression levels were significantly correlated with immune cell and immune checkpoint content in the tumor microenvironment. The HAGLR/miR-338-3p/GALNT2 regulatory axis may affect laryngeal cancer development. GALNT2 expression levels also differed significantly among other tumor types, as shown in our pan-cancer analysis. GALNT2 is a significant factor in the prognosis and immune cell infiltration of laryngeal cancer patients, indicating its potential as a useful prognostic marker for laryngeal cancer.
... Head and neck cancer squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, reporting more than 500,000 new cases and 300,000 deaths annually [1]. Despite multi-modality treatment with surgery, radiation, and/or chemotherapy, the recurrence and metastatic rates for HNSCC remain high, and patients suffering relapse remain at extremely high risk of dying from the disease [2,3]. Often, palliative chemotherapy is the only option; however, response rates to conventional chemotherapies remain poor, with combination chemotherapy response rates at approximately ~25% [4] and approximately 35% when chemotherapy is combined with cetuximab [5]. ...
Article
Full-text available
Recurrent or metastatic head and neck squamous cell carcinoma (RMHNSCC) is associated with a poor prognosis and short survival duration. There is an urgent need to identify personalized predictors of drug response to guide the selection of the most effective therapy for each individual recurrence. We tested the feasibility of patient-derived xenografts (PDX) for guiding their RMHNSCC salvage treatment. Fresh tumor samples from eligible, consented patients were implanted into mice. Established tumors were expanded in mouse PDX cohorts to identify responses to candidate salvage drug treatments in parallel testing. Patients alive and suitable for chemotherapy were treated based on responses determined by PDX testing. Nine patient tumors were successfully engrafted in mice with an average time of 89.2±41.7 days. Four patients' PDX models underwent parallel drug testing. Two patients received PDX-guided therapy. In one of these patients, single agents of cetuximab and paclitaxel demonstrated the best responses in the PDX model, and this patient exhibited sequential partial responses to each drug, including a 17-month clinical response to cetuximab. The main limitation of PDX testing for RMHNSCC was the time delay in obtaining testing results. Despite this, parallel PDX testing may be feasible for a subset of patients and appears to correlate with clinical benefit.
... Head and neck squamous cell carcinoma is an aggressive malignancy with high morbidity and represents the sixth most common cancer worldwide [1,2]. Radio-, chemo-and immuno-therapies are used in the management of head and neck squamous cell carcinoma [3][4][5][6][7][8]. However, there are several hurdles with these treatments, including failure in eradication of solid tumors, severe adverse effects and inefficient targeting capabilities, which lead to failure of treatments and stemness in metastasis. ...
Article
Full-text available
Background: Boron neutron capture therapy (BNCT) is a promising cancer treatment that eliminates tumor cells by triggering high-energy radiation within cancer cells. Aim: In vivo evaluation of poly(vinyl alcohol)/boric acid crosslinked nanoparticles (PVA/BA NPs) for BNCT. Materials & methods: PVA/BA NPs were synthesized and intravenously injected into tumor-bearing mice for BNCT. Results: The in vitro boron uptake of PVA/BA NPs in tumor cells was 70-fold higher than the required boron uptake for successful BNCT. In an in vivo study, PVA/BA NPs showed a 44.29% reduction in tumor size compared with clinically used boronophenylalanine for oral cancer in a murine model. Conclusion: PVA/BA NPs exhibited effective therapeutic results for oral cancer treatments in BNCT.
... The 5-year survival rates of squamous cell carcinoma (SCC) decrease from 93% in the case of stage I detection to 40% for stage IV. To date, there is no diagnostic method that can identify oral and oropharyngeal SCC in the early stage of the disease [1][2][3][4]. Moreover, acknowledging the increasing incidence of HNC in the last years, as well as the correlation between tobacco smoking and oral and oropharyngeal cancer (OOC) alongside intense tobacco consumption nowadays, a screening method is mandatory. ...
Article
Full-text available
Raman spectroscopy recently proved a tremendous capacity to identify disease-specific markers in various (bio)samples being a non-invasive, rapid, and reliable method for cancer detection. In this study, we first aimed to record vibrational spectra of salivary exosomes isolated from oral and oropharyngeal squamous cell carcinoma patients and healthy controls using surface enhancement Raman spectroscopy (SERS). Then, we assessed this method’s capacity to discriminate between malignant and non-malignant samples by means of principal component–linear discriminant analysis (PC-LDA) and we used area under the receiver operating characteristics with illustration as the area under the curve to measure the power of salivary exosomes SERS spectra analysis to identify cancer presence. The vibrational spectra were collected on a solid plasmonic substrate developed in our group, synthesized using tangential flow filtered and concentrated silver nanoparticles, capable of generating very reproducible spectra for a whole range of bioanalytes. SERS examination identified interesting variations in the vibrational bands assigned to thiocyanate, proteins, and nucleic acids between the saliva of cancer and control groups. Chemometric analysis indicated discrimination sensitivity between the two groups up to 79.3%. The sensitivity is influenced by the spectral interval used for the multivariate analysis, being lower (75.9%) when the full-range spectra were used.
Article
Full-text available
The use of conventional chemotherapy in conjunction with targeted and immunotherapy drugs has emerged as an option to limit the severity of side effects in patients diagnosed with head and neck cancer (HNC), particularly oropharyngeal cancer (OPC). OPC prevalence has increased exponentially in the past 30 years due to the prevalence of human papillomavirus (HPV) infection. This study reports a comprehensive review of clinical trials registered in public databases and reported in the literature (PubMed/Medline, Scopus, and ISI web of science databases). Of the 55 clinical trials identified, the majority (83.3%) were conducted after 2015, of which 77.7% were performed in the United States alone. Eight drugs have been approved by the FDA for HNC, including both generic and commercial forms: bleomycin sulfate, cetuximab (Erbitux), docetaxel (Taxotere), hydroxyurea (Hydrea), pembrolizumab (Keytruda), loqtorzi (Toripalimab-tpzi), methotrexate sodium (Trexall), and nivolumab (Opdivo). The most common drugs to treat HPV-associated OPC under these clinical trials and implemented as well for HPV-negative HNC include cisplatin, nivolumab, cetuximab, paclitaxel, pembrolizumab, 5-fluorouracil, and docetaxel. Few studies have highlighted the necessity for new drugs specifically tailored to patients with HPV-associated OPC, where molecular mechanisms and clinical prognosis are distinct from HPV-negative tumors. In this context, we identified most mutated genes found in HPV-associated OPC that can represent potential targets for drug development. These include TP53, PIK3CA, PTEN, NOTCH1, RB1, FAT1, FBXW7, HRAS, KRAS, and CDKN2A.
Article
Full-text available
Background Patients with platinum-refractory recurrent or metastatic squamous cell carcinoma of the head and neck have few treatment options and poor prognosis. Nivolumab significantly improved survival of this patient population when compared with standard single-agent therapy of investigator's choice in Checkmate 141; here we report the effect of nivolumab on patient-reported outcomes (PROs). Methods CheckMate 141 was a randomised, open-label, phase 3 trial in patients with recurrent or metastatic squamous cell carcinoma of the head and neck who progressed within 6 months after platinum-based chemotherapy. Patients were randomly assigned (2:1) to nivolumab 3 mg/kg every 2 weeks (n=240) or investigator's choice (n=121) of methotrexate (40–60 mg/m² of body surface area), docetaxel (30–40 mg/m²), or cetuximab (250 mg/m² after a loading dose of 400 mg/m²) until disease progression, intolerable toxicity, or withdrawal of consent. On Jan 26, 2016, the independent data monitoring committee reviewed the data at the planned interim analysis and declared overall survival superiority for nivolumab over investigator's choice therapy (primary endpoint; described previously). The protocol was amended to allow patients in the investigator's choice group to cross over to nivolumab. All patients not on active therapy are being followed for survival. As an exploratory endpoint, PROs were assessed at baseline, week 9, and every 6 weeks thereafter using the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire–Core 30 (QLQ-C30), the EORTC head and neck cancer-specific module (EORTC QLQ-H&N35), and the three-level European Quality of Life–5 Dimensions (EQ-5D) questionnaire. Differences within and between treatment groups in PROs were analysed by ANCOVA among patients with baseline and at least one other assessment. All randomised patients were included in the time to clinically meaningful deterioration analyses. Median time to clinically meaningful deterioration was analysed by Kaplan-Meier methods. CheckMate 141 was registered with ClinicalTrials.org, number NCT02105636. Findings Patients were enrolled between May 29, 2014, and July 31, 2015, and subsequently 361 patients were randomly assigned to receive nivolumab (n=240) or investigator's choice (n=121). Among them, 129 patients (93 in the nivolumab group and 36 in the investigator's choice group) completed any of the PRO questionnaires at baseline and at least one other assessment. Treatment with nivolumab resulted in adjusted mean changes from baseline to week 15 ranging from −2·1 to 5·4 across functional and symptom domains measured by the EORTC QLQ-C30, with no domains indicating clinically meaningful deterioration. By contrast, eight (53%) of the 15 domains in the investigator's choice group showed clinically meaningful deterioration (10 points or more) at week 15 (change from baseline range, −24·5 to 2·4). Similarly, on the EORTC QLQ-H&N35, clinically meaningful worsening at week 15 was seen in no domains in the nivolumab group and eight (44%) of 18 domains in the investigator's choice group. Patients in the nivolumab group had a clinically meaningful improvement (according to a difference of 7 points or greater) in adjusted mean change from baseline to week 15 on the EQ-5D visual analogue scale, in contrast to a clinically meaningful deterioration in the investigator's choice group (7·3 vs −7·8). Differences between groups were significant and clinically meaningful at weeks 9 and 15 in favour of nivolumab for role functioning, social functioning, fatigue, dyspnoea, and appetite loss on the EORTC QLQ-C30 and pain and sensory problems on the EORTC QLQ-H&N35. Median time to deterioration was significantly longer with nivolumab versus investigator's choice for 13 (37%) of 35 domains assessed across the three questionnaires. Interpretation In this exploratory analysis of CheckMate 141, nivolumab stabilised symptoms and functioning from baseline to weeks 9 and 15, whereas investigator's choice led to clinically meaningful deterioration. Nivolumab delayed time to deterioration of patient-reported quality-of-life outcomes compared with single-agent therapy of investigator's choice in patients with platinum-refractory recurrent or metastatic squamous cell carcinoma of the head and neck. In view of the major unmet need in this population and the importance of maintaining or improving quality of life for patients with recurrent or metastatic squamous cell carcinoma of the head and neck, these data support nivolumab as a new standard-of-care option in this setting. Funding Bristol-Myers Squibb.
Article
Background: B490 (EudraCT# 2011-002564-24) is a randomized, phase 2b, noninferiority study investigating the efficacy and safety of first-line cetuximab plus cisplatin with/without paclitaxel (CetCis versus CetCisPac) in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck (R/M SCCHN). Patients and methods: Eligible patients had confirmed R/M SCCHN (oral cavity/oropharynx/larynx/hypopharynx/paranasal sinus) and no prior therapy for R/M disease. Cetuximab was administered on day 1 (2-h infusion, 400 mg/m2), then weekly (1-h infusions, 250 mg/m2). Cisplatin was given as a 1-h infusion (CetCis arm: 100 mg/m2; CetCisPac arm: 75 mg/m2) on day 1 of each cycle for a maximum of six cycles. Paclitaxel was administered as a 3-h infusion (175 mg/m2) on day 1 of each cycle. After six cycles, maintenance cetuximab was administered until disease progression or unacceptable toxicity. The primary end point was progression-free survival (PFS). We assumed a noninferiority margin of 1.40 as compatible with efficacy. Results: A total of 201 patients were randomized 1 : 1 to each regimen; 191 were assessable. PFS with CetCis (median, 6 months) was noninferior to PFS with CetCisPac (median, 7 months) [HR for CetCis versus CetCisPac 0.99; 95% CI: 0.72-1.36, P = 0.906; margin of noninferiority (90% CI of 1.4) not reached]. Median overall survival was 13 versus 11 months (HR = 0.77; 95% CI: 0.53-1.11, P = 0.117). The overall response rates were 41.8% versus 51.7%, respectively (OR = 0.69; 95% CI: 0.38-1.20, P = 0.181). Grade ≥3 adverse event rates were 76% and 73% for CetCis versus CetCisPac, respectively, while grade 4 toxicities were lower in the two-drug versus three-drug arm (14% versus 33%, P = 0.015). No toxic death or sepsis were reported and cardiac events were negligible (1%). Conclusion: The two-drug CetCis regimen proved to be noninferior in PFS to a three-drug combination with CetCisPac. The median OS of both regimens is comparable with that observed in EXTREME, while the life-threatening toxicity rate appeared reduced. Clinical trial number: EudraCT# 2011-002564-24.
Article
Background The effectiveness of the combination chemotherapy of weekly paclitaxel and cetuximab has not yet been compared to that of the current standard regimen, EXTREME (combination of 5-fluorouracil, cisplatin and cetuximab). Methods We retrospectively reviewed the clinical records of R/M SCCHN patients who received cetuximab-containing chemotherapy as a first-line therapy; from these, patients receiving a weekly paclitaxel and cetuximab regimen (cohort A) and the EXTREME regimen (cohort B) were extracted. The responses, prognoses and adverse events of these two cohorts were evaluated. Results A total of 86 patients were included (cohort A, 49; cohort B, 36). Patients with histories of platinum-based chemotherapy were more frequently given the cohort A treatment. Though the response rates were similar in the two cohorts (45% in cohort A and 51% in cohort B; p = 0.83), the progression-free survival (PFS) was significantly more favorable in cohort A by the log-rank test (6.0 months vs 5.0 months; p = 0.027). In the Cox-regression hazard analyses, male gender (hazard ratio [HR] = 2.1, p = 0.010), older age (≥ 70 yo) (HR = 5.0, p = 0.018), PS 0 (HR = 2.2, p = 0.027), no history of platinum chemotherapy (HR = 3.2, p = 0.003) and the presence of a tracheostomy (HR = 2.3, p = 0.039) were favorable factors within cohort A. Conclusion In selected R/M SCCHN patients, the combination of weekly paclitaxel and cetuximab could be the better treatment option than the EXTREME regimen.
Article
Despite emerging appreciation for the important role of immune checkpoint receptors in regulating the effector functions of T cells, it is unknown whether their expression is involved in determining the clinical outcome in response to cetuximab therapy. We examined the expression patterns of immune checkpoint receptors (including PD-1, CTLA-4, and TIM-3) and cytolytic molecules (including granzyme B and perforin) of CD8+ tumorinfiltrating lymphocytes (TILs) and compared them to those of peripheral blood T lymphocytes (PBLs) in patients with head and neck cancer (HNSCC) during cetuximab therapy. The frequency of PD-1 and TIM-3 expression was significantly increased in CD8+ TILs compared to CD8+ PBLs (P = 0.008 and P = 0.02, respectively). This increased CD8+ TIL population co-expressed granzyme B/perforin and PD-1/TIM-3, which suggests a regulatory role for these immune checkpoint receptors in cetuximab- promoting cytolytic activities of CD8+ TIL. Indeed, the increased frequency of PD-1+ and TIM-3+ CD8+ TILs was inversely correlated with clinical outcome of cetuximab therapy. These findings support the use of PD-1 and TIM-3 as biomarkers to reflect immune status of CD8+ T cells in the tumor microenvironment during cetuximab therapy. Blockade of these immune checkpoint receptors might enhance cetuximab-based cancer immunotherapy to reverse CD8+ TIL dysfunction, thus potentially improving clinical outcomes of HNSCC patients.
Article
Our initial experience with weekly high dose methotrexate with leucovorin rescue (MTX-LCV), in advanced recurrent or metastatic squamous cell carcinoma of the head and neck with a 77% tumor response rate and high therapeutic index, prompted a trial of MTX-LCV as initial adjuvant therapy in high risk nonmetastatic patients. Results in 11 patients are presented and confirm the high response rate to MTX-LCV and the low incidence of myelotoxicity and mucositis, when concurrent urinary alkalinization is employed. Initial MTX-LCV administration has not compromised subsequent optimum aggressive combinations of surgery and radiation therapy. Cytoreduction with MTX-LCV may be safely used initially in combined therapy for high risk squamous cell carcinoma of the head and neck.
Article
PURPOSEWe conducted a phase II study designed to evaluate the activity, safety, and tolerability of docetaxel (Taxotere: Rhone-Poulenc Rorer Pharmaceuticals Inc, Collegeville, PA) in patients with advanced, incurable, or recurrent squamous cell carcinoma of the head and neck (SCCHN) who had not received prior palliative chemotherapy.PATIENTS AND METHODS Thirty-one patients with measurable, locoregional, or metastatic SCCHN were treated with docetaxel, administered at a dose of 100 mg/m2 as a 1-hour intravenous (i.v.) infusion once every 21 days on an outpatient basis. All patients were premedicated with dexamethasone, diphenhydramine, and cimetidine. Prophylactic administration of growth factors or antiemetics was not permitted.RESULTSThirty-one patients were treated. Twenty-nine patients were assessable for response and 30 for toxicity. Four of 31 patients (13%) achieved complete response (CR), nine (29%) achieved partial response had stable disease (SD) and seven (23%) experienced progression of disease ...
Article
PURPOSEThe Southwest Oncology Group (SWOG) conducted a randomized comparison of cisplatin plus fluorouracil (5-FU) and carboplatin plus 5-FU versus single-agent methotrexate (MTX) in patients with recurrent and metastatic squamous-cell carcinoma (SCC) of the head and neck. The primary objective was to compare separately the response rates of each combination regimen to standard weekly MTX.PATIENTS AND METHODS Two hundred seventy-seven patients diagnosed with SCC of the head and neck were randomized to one of three treatments: (1) cisplatin 100 mg/m2 intravenously (IV) on day 1 and 5-FU 1,000 mg/m2 per day for a 96-hour continuous infusion repeated every 21 days; (2) carboplatin 300 mg/m2 IV on day 1 and 5-FU 1,000 mg/m2 per day for a 96-hour continuous infusion repeated every 28 days; and (3) MTX 40 mg/m2 IV given weekly.RESULTSAll three treatment regimens were well tolerated. However, both hematologic and nonhematologic toxicities were significantly greater with cisplatin plus 5-FU compared with MTX (P = ...
Article
Background Docetaxel (Taxotere®) is a new cytotoxic agent acting as a promotor of tubulin polymerisation with broad spectrum antitumor activity in preclinical testing. Phase I clinical trials have shown promising activity of docetaxel in patients with breast, ovarian and lung carcinomas. The objective of this open multicentre phase II study was to determine the efficacy and tolerability of this agent in patients with head and neck cancer. Patients and methods Patients with proven advanced and/ or recurrent squamous cell carcinoma of the head and neck without prior chemotherapy for advanced disease were eligible for this trial. Docetaxel was given at a dose of 100 mg/ m² as a 1 hour infusion every 3 weeks. Dose reductions were performed according to hematological and non-hematologi-cal toxicities. No pre-medication was given to prevent hyper-sensitivity reactions. Results Fourty-three patients entered this trial: 39 patients were evaluable for toxicity and 37 patients were evalu-able for response. Sixty-five percent of the patients had loco-regional disease, 28% had metastatic disease, and 7% had both. Twenty-five percent of the patients had previously received neo-adjuvant cisplatin-based chemotherapy. A total of 166 docetaxel courses were administered. The most frequent side-effects associated with docetaxel were alopecia (90% of the patients), asthenia (69% of the patients) and short lasting neutropenia (grade 3–4 neutropenia in 61% of the courses). Fifty-four percent of the patients experienced skin toxicity, 23% experienced hypersensitivity reaction, and 31% developed peripheral edema. Ten partial and 2 complete responses were observed, yielding a response rate of 32% (95% confidence interval 17%–47%). Conclusion Docetaxel is an active drug in patients with advanced squamous cell carcinoma of the head and neck.