You are on page 1of 18

Immunol Res (2013) 57:5269

DOI 10.1007/s12026-013-8462-3

IMMUNOLOGY & MICROBIOLOGY IN MIAMI

The immune system and head and


neck squamous cell carcinoma:
from carcinogenesis to new
therapeutic opportunities
Monika E. Freiser Paolo Serafini Donald T. Weed
Monika E. Freiser Paolo Serafini Donald T. Weed

Published online: 12 November 2013


Springer Science+Business Media New York 2013

Abstract Head and neck squamous cell carcinomas (HNSCCs) exhibit complex interactions with the host immune
system that may simultaneously explain resistance to various therapeutic modalities and that may also provide opportu-
nities for therapeutic intervention. Discoveries in immunologic research over the last decade have led to an increased
understanding of these interactions as well as the development of a multitude of investigational immunotherapies. Here, we
describe the interaction between HNSCC and the immune system, including a discussion of immune cells involved with
tumor carcinogenesis and the role of immune-modulating factors derived from tumors. We also describe the current
immunotherapeutic approaches being investigated for HNSCC, including a discussion of the successes and limitations.
With this review, we hope to present HNSCC as a model to guide future research in cancer immunology.

Keywords Head and neck squamous cell carcinoma  Immunosuppression  Immunotherapy  Tumor vaccines  Antibody
therapies  Human papilloma virus

Introduction fatality rate remains high, with about 56 % of patients


dying from their disease [2]. This speaks to the limitations
Head and neck squamous cell carcinomas (HNSCCs) rep- in treatment efficacy of the currently available standard of
resent about 95 % of tumors originating from the epithe- care treatment options employing combinations of surgery,
lium of the upper aerodigestive tract and globally account radiotherapy, and chemotherapy. These limitations high-
for the sixth most common malignancy [1]. Over 634,000 light a need for new therapeutic modalities and approaches
new cases were reported worldwide in 2008, and the case in HNSCC, a need that is being met by an expanding role
for immunotherapy as a fourth treatment option for this
P. Serafini (&)
disease. Much has been discovered about the interaction of
Department of Microbiology and Immunology, University of the immune system and HNSCC tumors, both in the
Miami Miller School of Medicine, RMSB 3075, 1600 NW 10th development of the disease and in the mechanisms of
Avenue, Miami, FL 33136, USA tumor resistance. Understanding this interaction is essential
e-mail: pserafini@med.miami.edu
to developing successful immunotherapeutic interventions.
D. T. Weed (&)
Division of Head and Neck Surgery, Department of
Otolaryngology, University of Miami Miller School of
Interaction between HNSCC and the immune system
Medicine, 1475 NW 12th Avenue, 3rd Flr, Suite 3550, Miami,
FL 33136, USA
e-mail: dweed@med.miami.edu The immune system is intimately involved in the process of
HNSCC carcinogenesis (Fig. 1a). Ideally, when an epi-
M. E. Freiser
thelial cell acquires a deleterious mutation that cannot be
University of Miami Miller School of Medicine, 1475 NW 12th
Avenue, 3rd Flr, Suite 3550, Miami, FL 33136, USA repaired, it should undergo apoptosis [3]. When this fails,
e-mail: mfreiser@med.miami.edu the immune system should recognize the cell as abnormal

123
Immunology & Microbiology in Miami (2013) 57:5269 53

Fig. 1 The interaction between immune system and HNSCC. a back to the tumor where they exert their tumoricidal action (4). To
Anatomical location of HNSCC. The micro- and macro-environment counteract the immune surveillance, neoplastic cells are edited to be
of HNSCC is characterized by different immunological components ignored, to directly inhibit T-cell activity, or to secrete tumor-derived
that include lymph nodes, tonsils, resident immune cells of the factors (TDF) that inhibit DC migration to the lymph nodes (5a) and
mucosa (i.e., Langerhans cells), and immune-stimulatory elements alter the myelopoiesis (5b) inducing the generation of MDSCs and
(i.e., oral bacteria). b Schematic diagram of the interactions between Tregs. These cells inhibit the immune surveillance (6) by inhibiting
HNSCC and the immune system. Langerhans cells and resident DC maturation and migration, by preventing T-cell expansion, by
myeloid DCs or DCs infiltrating the mucosa uptake the tumor directly inhibiting the effector function of T cells or by promoting
antigens (1) and migrate (2) to the draining lymph nodes to cross- T-cell anergy
present them to the effector T cells (3). Once activated, T cells move

and attack it accordingly; this concept of immune system Immunoediting selects for tumor cells
recognition and elimination of transformed cells is known
as immunosurveillance, first introduced in 1970 by Burnet Immunoediting is a concept first introduced by Dunn and
and Thomas [4, 5]. However, there is a failure of this Schreiber in 2002 [5, 15] that describes how tumor cells
paradigm with all clinically evident cancers, and growing develop in the setting of a healthy immune system. Cancer
evidence demonstrates that certain immune cells may in immunoediting is composed of three processes: elimina-
fact promote the development and survival of neoplastic tion, equilibrium, and escape. Immunosurveillance occurs
cells. during the elimination process, whereby the immune sys-
Perhaps the most basic evidence of a relationship tem recognizes transitioned cells and induces cell death
between HNSCC and the immune system is that immu- through various mechanisms. Most preclinical lesions are
nodeficiency increases HNSCC risk [6]. For example, likely cleared this way [15]. Of note, aerodigestive epi-
premalignant leukoplakia develops in 13 % of renal thelial cells encounter foreign particles and bacteria more
transplant patients as compared to 0.6 % of age- and sex- often than many tissues, and yet most patients do not suffer
matched individuals, and about 10 % will become frank from perpetual mucosal inflammation. This natural ten-
HNSCC [7, 8]. Increased incidence of HNSCC has been dency for tolerance may predispose to tolerance toward
observed in bone marrow transplant patients [911] and transitioned cells [16]. Those transitioned cells that are
HIV-positive patients [12]. While HNSCC is not an AIDS- capable of surviving initially enter into the equilibrium
defining illness, HIV-positive patients develop HNSCC phase, whereby Darwinian pressures continue to select for
earlier in life with more advanced disease and poorer the tumor cells that develop mutations allowing continued
prognosis [13]. As significant as immunodeficiency may be survival. The escape process occurs once a tumor cell has
in HNSCC risk elevation, most patients have normal the means to efficiently overcome the immune system.
immune systems when the cancer develops. In the immu- These three processes may co-exist in a collection of tumor
nocompetent patient, the process to blame is immunoedit- cells. By the time a tumor becomes clinically evident,
ing [14]. immunoediting has already selected for the cells that are

123
54 Immunology & Microbiology in Miami (2013) 57:5269

best suited to survive in an immunologically intact promoting factors have been identified in HNSCC,
environment. including Galectin-1 [23], FASL [24, 25], TRAIL, and
PDL-1 [26]. These factors expressed by the malignant cells
Mechanisms of immune system escape seem to induce T-cell apoptosis when binding to the cog-
nate receptor. A factor commonly present in human pap-
There are four main ways that selective pressures during illoma virus positive (HPV?) HNSCC cells is PDL-1,
the equilibrium process are thought to lead to HNSCC which binds with PD-1 on the T cells and promotes anergy,
escape in immunocompetent patients: tumor cell evasion of exhaustion, or apoptosis [27, 28]. Direct inhibition of local
detection, tumor resistance to attack, direct tumor cell T cells by tumor cells is not the only immunosuppressive
inhibition of local immune function, and indirect inhibition strategy present. Indeed, it is becoming clear that HNSCC
of immune function by way of recruitment of immuno- cells can promote an aberrant hematopoiesis that affects the
suppressive players [16]. While evidence exists supporting entire immune system.
all four, it is now clear that selection of malignant cells
with direct or indirect intrinsic immunosuppressive activity HNSCC cells indirectly inhibit the immune system by
is particularly common. recruiting and directing an army of pro-tumoral cells

HNSCC cells evade immune system detection It is now evident that HNSCC tumors secrete factors that
alter normal hematopoiesis and induce the production and
Head and neck squamous cell carcinoma cells may trick the recruitment of cells from the innate and adaptive immune
immune system into disregarding them as normal cells. system to effectively counter and silence their anti-tumoral
This can be achieved by decreasing the expression of counterparts. These cells are part of a network of cells
surface MHC class I molecules [17] or by impairing their modulated by HNSCC tumors. It is important to understand
antigen processing machinery [16]. HNSCC may also lose this network in order to best design immunotherapeutic
immune-dominant epitopes during immunoediting [6]. strategies.
However, immune system evasion is not thought to play a
large role as most HNSCC tumors have abnormal antigens
that are recognized by the immune system even though the Cellular network of immune modulation in HNSCC
immune system does not effectively react. Lists detailing
many of these tumor-associated antigens (TAAs) have been The HNSCC microenvironment usually contains a signifi-
published recently [16, 1821]. In many cases, the specific cant amount of non-cancerous stromal cells in addition to
CD8? T-cell targeting the TAA is actually present in the neoplastic cells; these include tumor-associated macro-
patient. Furthermore, patients TAA-specific CD8? T cells phages (TAMs), myeloid derived suppressor cells
that are expanded in vitro and exposed to autologous tumor (MDSCs), immature dendritic cells (iDCs), and various
cells can lyse the tumor cells when incubated with stimu- types of T cells (Fig. 1b). These immune cells are not
latory factors like IFN-c [6]. innocent bystanders; indeed, they play an important role in
disease outcome. Depending on the composition and acti-
HNSCC cells resist immune attack vation status of these cells and the surrounding signaling
factors, the net effect can be either favorable or detrimental
Head and neck squamous cell carcinoma tumors may to tumor progression and metastasis. Table 1 summarizes
express receptors typically found on immune cells that the involvement of specific stromal cells and the net
induce anti-apoptotic pathways [6]. One such receptor is immunosuppressive effect.
toll-like receptor 4 (TLR-4), which normally binds to
lipopolysaccharide, found on bacteria. TLR-4 typically Effector T cells
helps protect immune cells during natural killer (NK) cell
attack, and on tumor cells may allow them to resist NK- Cell-mediated immunity is considered to be the most
induced apoptosis [22]. important process for antitumor immunity; intact CD8?
CD4? T helper 1 (Th1) function is essential for effective
HNSCC cells express or secrete factors that directly inhibit immunosurveillance and tumor containment [4, 14]. As
the immune system mentioned above, tumor antigen-specific CD8? T cells
have been identified in patients, yet these cells are unable
As is the case with many cancers, most HNSCC tumors to fully perform their tumoricidal action because of a
express or secrete factors that directly inhibit the immune suppressive network orchestrated by the tumor [6]. HNSCC
system in the microenvironment. A number of apoptosis- patients have fewer lymphocytes in their blood [29] but a

123
Immunology & Microbiology in Miami (2013) 57:5269 55

Table 1 Cellular network of immune modulation specimen correlates with better survival in HNSCC
Cell Involvement Net effect
patients [3133]. Nevertheless, defects are commonly
found in the effector cells of patients with HNSCC. Even
Cytotoxic T Recognize tumor CD8? T cells system after curative resection of HNSCC cancers, patients have
cells (CD8?) antigens but are impairment [36]
reduced T-cell counts for several years [34]. Circulating
suppressed
CD8? T cells in the blood of most HNSCC patients have
Helper T cells T helper 1 suppressed, T Humoral response
(CD4?) helper 2 favored; T favored, which is an increased ratio of pro-apoptotic protein BAX to anti-
helper 2 cells express ineffective against apoptotic factor Bcl-2 [25]. Intrinsic molecular defects are
IL-4, IL-10, IL-13 tumor; cytotoxic detectable in the T cells, including downregulation of the
activity inhibited [16] n-chain of the CD3 complex, a low responsiveness to IL-2
Natural killer NKG2D internalized, Failure to destroy tumor [35], and a reduced proliferative capability to mitogenic
cells inhibitory receptors cells [36, 38]
activated, reduced stimulation [36].
expression of
cytotoxic substances, Natural killer cells
decreased CD1d-
restricted NK T cells
Natural killer cells of the innate immune system typically
Dendritic cells Impaired maturation; Tolerogenic
immature DCs lack environment; partake in immunosurveillance and antitumor immunity
sufficient CD80 and insufficient T-cell [37]. While NK cells should be able to clear tumor cells
CD86 to co-stimulate; stimulation [48, 80] with decreased MHC class I expression and perform anti-
promote T regulatory body-dependent cellular cytotoxicity, successful HNSCC
cell proliferation
cells inhibit NK function [36]. NK cell cytotoxicity
Tumor- M2 phenotype; produce Impairs T helper
associated IL-10 TGF-b, VEGF; 1/CD8? pathway and
depends on the relative stimulation of activating versus
macrophages secrete remodeling instead promotes inhibitory receptors: tumor cells ensure that inhibitory
(TAMs) proteases production of Treg signals dominate [38]. In addition, through secretion of
cells; contributes to TGF-b1 and a soluble form of MHC-I-related chain, tumor
tumor stabilization
and angiogenesis [49]
cells cause reduced expression of the activating receptors
Myeloid Produce TGF-b, Severe cytotoxic T-cell
NKG2D and CD16 [36, 39]. Tumor cells resist NK attack
derived peroxynitrite; induce dysfunction [58] by the production of anti-apoptotic molecules [22].
suppressor local L-arginine HNSCC patients have severe reductions in their circulating
cells starvation; Secrete CD1d-restricted NK T (iNKT) cells [40], which are
(MDSCs) remodeling proteases
important for activating effector cells [36].
Regulatory T Secrete TGF-b, IL-10; T-cell anergy or death
cells (Tregs) induce local and APC cell
tryptophan starvation dysfunction [36] Dendritic cells
Cytokine Imbalanced toward T Impaired cell-mediated
profile helper 2/humoral immunity [80] Dendritic cells (DCs) have a critical function as antigen-
phenotype presenting cells [41] but in HNSCC this function is also
B cells Poor generation of high- Impairment of impaired. The environment in which HNSCC develops,
affinity tumor antigen phagocytic effector
aerodigestive tract mucosa, is rich in immature myeloid
antibodies or cells renders antibody
antibodies produced recognition useless DCs and Langerhans cells [42]. Upon encountering
do not inherently harm [80] inflammatory signals (IL-1, TNF-a, etc.) or microbial
tumor cells products (i.e., TLR ligands) under physiological condi-
Complement Tumor cells express No complement damage tions, DCs migrate to secondary lymphoid organs and
cascade anti-complement to tumor cells [80]; mature into antigen-presenting cells capable of cross
factors complement proteins
may promote stimulating, priming, and promoting the expansion of
mutagenesis [81] effector T cells. Increased presence of DCs in tumor
Mast cells Secrete proteolytic Tumor migration and specimens is associated with longer disease-free survival
enzymes invasion [52] and decreased recurrence [43, 44], and fewer DCs are
found in high-grade tumors [45] and lymph nodes with
metastatic disease [46]. To avoid immune system recog-
relatively high presence of effector memory T cells [30], nition, tumors frequently prevent DC maturation and
further suggesting T-cell recognition of tumor. Albeit instead attract abnormal, immature DCs (iDCs) that actu-
stunted, effector T cells may still achieve some anti- ally promote T-cell dysfunction [5]. Immature DCs express
tumoral effect: effector T-cell infiltration in a tumor low levels of the necessary co-stimulatory CD80 and CD86

123
56 Immunology & Microbiology in Miami (2013) 57:5269

molecules as well as MHC-II, which are necessary for residues of important signaling proteins in the IL-2
antigen presentation to CD4? T cells. These tolerogenic receptor pathway including JAK1, JAK3, STAT5, ERK,
DCs also induce production and proliferation of T regula- and AKT, effectively rendering T cells unresponsive to IL-2
tory cells, which are immunosuppressive immune cells [58]. In addition, MDSCs can produce high quantities of
discussed below. Defects in DC maturation are also visible peroxynitrite which induces apoptosis [63] or anergy [64]
systemically: HNSCC patients with advanced disease have in activated T cells. NOS and peroxynitrate metabolites
fourfold reduced levels of circulating myeloid-derived DCs have been found in HNSCC tumor beds [65, 66], and tumor
[47] as these cells accumulate in the lymph node sinuses infiltration with CD34? MDSCs [59, 6769] is a negative
and tumor site in an immature form [48]. In addition to prognostic factor. MDSCs, like TAMs, also secrete matrix
iDCs, HNSCC tumors also simultaneously attract other remodeling proteases and series proteases, contributing to
immature myeloid cells, including macrophages, granulo- tumor dissemination [5052].
cytes, and myeloid-derived suppressor cells, which all
contribute to a pro-tumoral environment. Regulatory T cells

Tumor-associated macrophages CD4?CD25?Foxp3? regulatory T cells (Tregs) are par-


ticularly important for the maintenance of peripheral tol-
Tumor-associated macrophages (TAMs) are macrophages erance and have been implicated in tumor-induced
known to support tumor progression via promotion of immunosuppression by many studies. Tregs inhibit T-cell
humoral immunity and hindrance of cell-mediated immu- activity via a number of mechanisms [70], including via the
nity [49]. TAMs are similar to M2 macrophages in that production of TGF-b and IL-10 or the hydrolysis of
they produce IL-10 and transforming growth factor b extracellular ATP to ADP or AMP by the ectoenzyme
(TGF-b) and secrete matrix remodeling proteases and CD39 [71]. Tregs also promote the generation of indole-
serine proteases that are associated with more advanced amine-2,3-dioxygenase (IDO) positive tolerogenic DCs by
tumor grade and metastasis [5052], higher microvessel expressing the cytotoxic T-lymphocyte antigen-4 (CTLA-4)
density in the tumor, and increased bioavailability of and engaging CD80 and CD86 on DCs [5, 70]. An increase
VEGF [36]. TAMs can also directly secrete VEGF [36, 53, in CD4?CD25highFOXP3? has been associated with a poor
54]. A paracrine loop has been suggested between TAMs prognosis in many cancers [72]. Controversy does exist,
and tumor cells: tumor cells attract TAMs by secreting however, as FOXP3? cells infiltrating the tumor have been
monocyte chemotactic protein-1 (MCP-1) and TGF-b1, proposed either as a positive [73, 74] or as a negative
and meanwhile, TAMs secrete VEGF, IL-8, TNF-a, and prognostic factor in HNSCC [75, 76]. This controversy was
IL-1, which stimulate tumors to secrete more VEGF and recently resolved by using computer-assisted image quan-
IL-8 [1, 36, 55]. TAMs may contribute directly to extra- tification and by taking into consideration the subcellular
capsular extension and lymph node metastasis as increased localization of FOXP3 [77]. Indeed, FOXP3? can be
TAM infiltration correlates with tumors with these features expressed also by non-regulatory, activated effector T cells
[20, 56]. TAMs accumulate in the areas of fibrin deposition [78]. While in regulatory T cells FOXP3 is found mainly in
in the tumor and peritumoral tissue, which suggests that the nucleus, in activated T cells FOXP3 seems to be
they aid in tumor matrix stabilization as well as angio- localized mostly in the cytoplasm [79]. By taking advan-
genesis [36, 57]. tage of these two conditions, a retrospective study in
patients with oral squamous cell carcinoma found that
Myeloid derived suppressor cells while FOXP3 in the cytoplasm of tumor infiltrating CD4?
cells correlated with favorable prognosis, its nuclear
MDSCs have emerged as a powerful mediator of the localization strongly predicted recurrence [77]. The ratio
immunosuppressive state. Recruited directly by tumors via between nuclear and cytoplasmic FOXP3 expression
tumor-derived factors, MDSCs are CD34?CD33? imma- within the tumor infiltrating CD4? T cells is proposed as a
ture myeloid cells that suppress CD8? T-cell proliferation powerful prognostic marker in HNSCC [77].
and promote tolerance through a variety of mechanisms,
including by producing TGF-b, removing L-arginine
(L-Arg) from the microenvironment, and nitrosylating Tumor-derived factors in HNSCC
important T-cell motifs [58, 59]. L-Arg is essential for
correct CD3 complex expression in T lymphocytes [60]. Head and neck squamous cell carcinoma cells effectively
MDSCs upregulate the enzyme L-arginase or the enzyme hijack the immune system and control distribution of
nitric oxide synthase (NOS), both of which partake in supportive resources such as nutrients both locally and
L-Arg metabolism [61, 62]. MDSCs S-nitrosylate cysteine distantly via release of tumor-derived factors (TDFs) [16].

123
Immunology & Microbiology in Miami (2013) 57:5269 57

These soluble factors are also able to condition distant sites Treg production [36]. Non-steroidal anti-inflammatory
for eventual metastasis, leading to a tumor-driven macro- drugs, which target COX-2, have been shown to exert
environment beyond the local microenvironment. Numer- chemoprotective effects on HNSCC cancer development
ous TDFs have been identified for HNSCC, including [95].
granulocytemacrophage colony-stimulating factor (GM-
CSF), prostaglandin E2 (PGE2), vascular endothelial Vascular endothelial growth factor
growth factor (VEGF), and various cytokines and chemo-
kines. A number of these factors are not completely pro- Released by most tumors, VEGF is well known for its role
tumor or anti-tumor, but rather play a dual role depending in tumor angiogenesis [9698]. VEGF is also intimately
on elements such as dose, concentration, and duration of involved with MDSCs, with evidence suggesting that
exposure. VEGF administration leads to inhibition of DC develop-
ment and an increased number of MDSCs [99]. Meanwhile,
Granulocyte macrophage-colony stimulating factor MDSCs secrete matrix metalloproteinase 9 (MMP-9),
which breaks down matrix proteins and allows for VEGF
Granulocyte macrophage-colony stimulating factor appears permeation through the extracellular matrix [100]. In
to assume both anti-tumoral and pro-tumoral roles HNSCC, VEGF-A expression correlated with microvessel
depending on variations in amount, systemic concentration, density, disease progression, a reduced number of mature
and duration of exposure. About 31 % of tested cancer lines DCs, and an increased number of immature DCs and
including HNSCC secrete GM-CSF [82, 83], and it is MDSCs [101]. Elevated VEGF levels is a poor prognostic
associated with a negative prognosis [83]. At higher doses, factor and correlates with tumor stage and lymph node
GM-CSF recruits MDSCs and promotes their differentiation status in HNSCC [102].
into effective pro-tumoral cells. Administration of tumor-
transduced GM-CSF, recombinant GM-CSF protein, or Cytokines
high doses of GM-CSF vaccines leads to emergence of
MDSC cells [82, 84, 85], whereas administration of GM- Cytokines are released not only by recruited immunosup-
CSF neutralizing antibody in tumor conditioned media pressive immune cells, but also by tumor cells that release
inhibits MDSC differentiation [86]. On the other hand, GM- them to orchestrate a pro-tumoral effect [16]. HNSCC was
CSF has been shown to elicit powerful immune responses shown to produce IL-13 and IL-4, which promote the T
when combined with c-irradiated tumor cell vaccines in the helper 2 response and an anti-inflammatory state [103,
clinical setting and as a result has been used as an immune 104]. Many HNSCC tumors have IL-4 and IL-13 receptors
adjuvant to augment antitumor immunity [87, 88]. GM-CSF (IL4Ra and IL13Ra), which are constitutively over-
is also given to decrease the incidence of mucositis during expressed, leading to excess arginase production and pro-
chemotherapy [89] as well as prevent potentially life- motion of neoplastic proliferation [105]. IL4Ra expression
threatening neutropenia. A bystander vaccine strategy was on MDSCs and monocytes is required for their suppressive
used in which the antigen dose and steric hindrance were phenotype [106, 107] and survival [107]. Most HNSCC
maintained constant to test the doseeffect of GM-CSF, and patients also have increased blood levels of IL-6 [108],
a threshold was identified above which significant immu- which is associated with a poor prognosis. IL-6 affects the
nosuppression mediated by MDSC recruitment was induced differentiation of myeloid lineages [109] by the activation
[85]. This may also explain why clinical trials demonstrated of STAT3 and may inhibit the maturation of DCs in par-
a negative effect at doses between 100 and 500 lg, but not ticular [110]. Introducing IL-6 antibodies leads to restora-
at repeated low doses of 4080 lg for 15 days [90]. tion of DC differentiation [111]. Indeed, bone marrow cells
treated with GM-CSF and either IL-6 or G-CSF were
Prostaglandins shown to differentiate into suppressive MDSCs [112]. IL-6
also contributes to systemic symptoms seen in some
Head and neck squamous cell carcinoma tumors frequently HNSCC patients including weight loss, night sweats, and
overexpress cyclooxygenase-2 (COX-2) [91, 92] as do fevers [113].
MDSCs, leading to the production of the eicosanoid pros-
taglandin E2 (PGE2), which correlates with arginase Chemokines
overexpression, STAT3 and STAT1 phosphorylation, IL-10
and MIP-2 production, and resulting immunosuppression Chemokines are important chemotactic molecules that can
[93]. PGE2 is able to activate the suppressive phenotype in not only bind to each other to form various signaling het-
MDSCs and facilitate the differentiation of MDSCs from erodimers [114, 115] but that can also bind to multiple
their hematopoietic precursors [94]. PGE2 can also induce receptors. These receptors, in turn, can be activated by

123
58 Immunology & Microbiology in Miami (2013) 57:5269

different chemokines, and this allows for chemokine prevention strategies. Indeed, with the distribution of the
redundancy [116], robustness, integration [117], and syn- HPV vaccine in children and young adults, evidence of a
ergy [118]. Consequently, individual chemokines have decrease in HNSCC in the vaccinated cohorts is emerging
been shown both to promote tumor immunity, such as by [132].
recruiting T cells to the tumor site, and to promote tumor
survival, such as by recruiting MDSCs and TAMs [119].
CCL5 can complex with CXCL4 to influence monocyte Immunotherapy for HNSCC
arrest; the same CCL5 can increase the antitumor effect of
adoptively transplanted T cells, but only if intratumoral For HNSCC patients, a number of immunotherapeutic
CD11c? cells are depleted [120]. CCL21 induces Th1 strategies are being explored, and many treatments are
polarization and boosts DNA vaccine effects, and yet the currently being tested in clinical trials. There are two main
same chemokine is secreted by many human tumors [121] categories of immunotherapeutic strategies: antigen non-
and the expression of its receptor CCR7 correlates with specific and antigen-specific. Antigen non-specific thera-
metastasis in HNSCC [122]. There are many more exam- pies are designed to broadly enhance the immune response,
ples of the contradictory roles of chemokines, but research either by stimulating anti-tumoral mediators or by revers-
is revealing more about the mechanisms behind each ing the immunosuppression put in place by the tumor.
beneficial or deleterious role. For example, CCL2 is known Antigen-specific therapies are designed to cause a focused
to attract both cytotoxic T cells [123], MDSCs, and TAMs immune response toward the tumor cells directly. Both
[124127], yet T cells remain trapped in the stroma sur- therapy types have yielded promising results, yet in most
rounding cancer cells, whereas TAMs and MDSC can cases there are limitations that need to be addressed.
freely travel within [128]. This occurs because reactive Antigen non-specific therapies are frequently limited by
nitrogen species produced by TAMs and neoplastic cells systemic toxicity, and antigen-specific therapies are often
induce nitration/nitrosylation of CCL2 that, once nitrosy- limited by the powerful immunosuppressive environment
lated, can no longer recruit cytotoxic T cells, but that can put forth by the tumor. Both therapy types may exhibit
still recruit myeloid cells to the tumor [129]. Understanding variable efficacy depending on the specific mutations
chemokine interactions may be imperative for designing present in a given tumor. Combination immunotherapies
immunotherapies capable of reaching the tumor and are being explored in order to overcome these limitations.
exerting the desired tumoricidal or immunostimulatory
effect.
Antigen non-specific therapies

HPV1 HNSCC and the immune system Immune stimulation: cytokine treatment

Whether a HNSCC tumor is HPV?or HPV- may have As detailed above, tumors utilize cytokines to orchestrate
important implications for its interaction with the immune immunosuppression and create a pro-tumoral milieu.
system. Notably, patients with HPV? tumors have a better Cytokine treatments attempt to shift the balance away from
survival than those with HPV- disease: three-year survival the pro-tumoral, T helper-2 cytokine predominance and
is 84 versus 57 %, and the median overall survival is toward anti-tumoral T helper-1/cytotoxic predominance
fourfold higher at 131 versus 20 months [130]. What [80].
accounts for this difference? The answer may be that the
immune system is better able to assume a cytotoxic role. In Interleukin-2
a preclinical study on immune-deficient mice, no differ-
ence in tumor growth was observed between transplanted Interleukin-2 (IL-2) is one of the earliest cytokines
HPV? and HPV- tumors, while in immune-competent employed to treat HNSCC. Recombinant IL-2 given locally
mice, a significant delay in tumor progression was by peritumoral, [133] intranodal, or intraarterial infusion
observed and 2030 % of them cleared the tumor com- [134] to HNSCC patients leads to increases in intratumoral
pletely [131]. Tumor rejection was possible only when both NK cells and activity of tumor infiltrating lymphocytes
HPV-specific CD4? and CD8? T cells were present. (TILs). These observation might be clinically significant
Indeed, HPV-infected cells are positive for the non-self, since the injection of IL-2 preoperatively around the cer-
virus-associated E7 and E6 antigens that can evoke a vical lymph node chains prior to oral cavity or oropharynx
powerful, generic, antiviral response. The differences squamous cell carcinoma resection and radiotherapy led to
between HPV? and HPV- tumors may become more significant increases in disease-free and overall survival
important as we develop immunotherapies and immune [135]. These local treatments are generally well tolerated,

123
Immunology & Microbiology in Miami (2013) 57:5269 59

whereas systemic IL-2 is not. Systemic toxicities are sig- IL-6, or other tumor-derived factors, while others target
nificant and include hypotension, capillary leak syndrome, specific immunosuppressive populations such as MDSCs
and oliguria [134] with only partial response rates near and T regulatory cells.
18 % [136]. Following the principle of local delivery, the
fusion drug ALT-801, composed of IL-2 and a T-cell Tyrosine kinase inhibitors
receptor specific for the p53/HLA-A*0201 complex, has
been designed to target IL-2 to p53 positive tumors [133]. Tyrosine kinases are commonly overexpressed and acti-
This treatment leads to higher efficacy and lower toxicity. vated in many tumors including HNSCC [142]. Tumor-
derived factors promote also the aberrant activation of
Interleukin-12 these kinases in the hematopoietic system, leading to the
accumulation of, for example, MDSCs. Suntinib is a mol-
Interleukin-12 (IL-12) has been used with a similar goal of ecule that inhibits multiple tyrosine kinases (VEGFR-1,
immunostimulation in HNSCC patients. Intratumoral IL-12 VEGFR-2, VEGFR-3, PDGFR, c-kit, ret, STAT3, etc.),
injection led to increased B-cell tumor and lymph node and, as expected, shows potent effects against MDSCs
infiltration, B-cell activation, and a highly significant IgG [143]. It has been shown to reverse MDSC accumulation
subclass switch measured in the plasma, indicating a switch and induce tumor cell apoptosis in renal cell carcinoma.
to Th1 phenotype [93]. IL-12 increased NK cells in the [144]. Clinical trials using suntinib in HNSCC patients
tumor as well as led to a 128-fold increase in IFN-c mRNA demonstrated significant toxicities including lymphopenia,
in lymph nodes [137, 138]. IL-12 is currently being neutropenia, and thrombocytopenia [145] and poor thera-
investigated in clinical trials [93] for efficacy. peutic efficacy [146, 147]. A number of other selective
kinase inhibitors are currently in development [142]. Some
Interferon-gamma tyrosine kinase inhibitor drugs are monoclonal antibodies
and will be discussed below.
Systemic IFN-c therapy has been attempted in HNSCC
patients with 3 of 8 responding with minimal toxicity in 1a,25-Dihydroxyvitamin D3
one study and 4 of 9 responding in a second study [139].
However, another trial failed to show any objective Vitamin D has been shown to induce the maturation of
response [140]. MDSCs into immune-stimulatory DCs [148, 149] as well
as inhibit tumor production of VEGF and hypoxia factor 1a
Interferon-alpha (HIF-1a) [150], which are involved with the induction of
MDSCs by the tumor. Although it is not clear whether
IFN-a has been used in combination with cisplatin and 5- vitamin D acts on MDSCs directly or indirectly, it has been
fluorouracil in the treatment of advanced esophageal tested in HNSCC. Patients treated with vitamin D preop-
squamous cell carcinoma with an overall response rate of eratively had reduced amounts of MDSCs in the tumor
55 % but was associated with significant toxicity. When and increased amounts of mature DCs and activated T cells
combined with IL-12, IFN-a produced significant tumor (CD4? and CD8?) [151]. More importantly, the time
regression in 2 of 11 advanced HNSCC patients [141]. A to recurrence was doubled in the patients who received
recent phase II trial investigated IFN-a combined with vitamin D. Nevertheless, more studies are needed to fur-
isotretinoin and vitamin E in locally advanced HNSCC, ther characterize the impact of vitamin D on HNSCC
showing 5-year progression-free survival rate and overall survival.
survival rate of 80 and 81.3 %, respectively [93].
Cox-2 inhibitors
Reversal of immunosuppression
COX-2 inhibitors reverse the PGE2 overproduction by
Reversal of tumor-induced immunosuppression is the sec- tumors and thus provide an opportunity to block MDSC
ond class of antigen non-specific strategies employed in differentiation and activation [152]. Celecoxib, a specific
HNSCC. These strategies are designed to (1) reestablish a COX-2 inhibitor, when used in combination with radio-
micro- and macro-environment favorable for immunosur- therapy and Erlotinib, an EGFR receptor blocker, led to
veillance; (2) deplete the suppressive populations that are 37 % progression-free survival and 60 % locoregional
recruited by the tumor; and (3) block the molecular control at 1 year [153]. When combined with the EGFR
mechanism of negative regulators of the adequate immune receptor blocker gefitinib, 22 % of unresectable or meta-
response. Several of these therapies aim to inhibit aberrant static HNSCC demonstrated a partial response [154].
intracellular pathways involving VEGF, PGE2, GM-CSF, While tolerated well during the study periods, celecoxib is

123
60 Immunology & Microbiology in Miami (2013) 57:5269

associated with dose-dependent cardiovascular morbidity disease during treatment according to radiographic review,
so its use in long-term treatment may be limited [155]. and overall survival was 69 % at 36 months [162].
Additional antigen non-specific therapies are currently
Phosphodiesterase inhibitors being tested in other cancers; a recently published review
discusses anticipated HNSCC clinical trials based on these
Phosphodiesterase-5 (PDE5) can be targeted to reverse additional therapies [36].
MDSC suppression. Its inhibition, by controlling the
intracellular concentration of cGMP, is sufficient to inhibit
arginase 1, NOS2, and IL4a on MDSCs, thus reducing Antigen-specific therapies
their suppressive ability [156]. Moreover, it has been
shown that administration of the PDE5 inhibitors sildenafil Antigen-specific therapies are designed to enhance tumor-
or tadalafil was sufficient to significantly reduce tumor specific responses generated by the natural immune system
progression in murine models of breast and colon cancer and include antibody therapies, adoptive cell transfer, and
[156], while in a lymphoma model was able to restrain tumor vaccines.
MDSC-mediated expansion of tumor-specific Tregs [157].
More importantly, sildenafil was able to restore T-cell Antibody therapies
proliferation in anti-CD3/anti-CD28-stimulated peripheral
blood mononuclear cells (PBMCs) from HNSCC patients A number of monoclonal antibodies have been developed
[157]. Two clinical trials (NCT00843635, NCT 00894413) against a variety of tumor cell surface molecules. When the
have recently closed to accrual utilizing daily tadalafil antibody binds to the target molecule, it may inhibit the
administration prior to standard of care surgery or chemo- tumor cell by directly blocking the signaling pathway ini-
radiation therapy in patients with HNSCC. While results of tiated by the molecule, and/or by initiating cell-mediated
these trials are not yet published, interim analysis of cytotoxicity resulting in tumor cell death. In cell culture,
NCT00843635 suggests that PDE5 blockade lowers MDSC tumor cell apoptosis only occurs after antibody adminis-
and Treg concentrations in the blood and tumor tissue and tration when lymphocytes are present [163, 164]. The
expands the pool of tumor-specific CD8? cells (unpub- precise mechanism of tumor lysis is not well understood:
lished data, updated September 2013). complement-dependent cytotoxicity can be seen with some
antibody therapies, but this mechanism of cell death should
occur rapidly, as should NK cell-mediated cell death, and
Alkylating agents
yet tumor shrinkage is noted over weeks, not hours; this is
more consistent with a T-lymphocyte-mediated lytic effect
Tregs are particularly sensitive to alkylating agents, and
[164]. Fc gamma R polymorphisms on antibodies should
thus, alkylating agents may play an immunotherapeutic
variably influence any NK antibody-dependent cellular
role. Indeed, cyclophosphamide or ifosphamide at low
cytotoxicity, yet this is not well elucidated [164, 165]. The
doses can decrease Treg concentration in the blood [158,
IgG subtype of the antibody appears to influence the effi-
159]. Interestingly, when cyclophosphamide was combined
cacy of the therapy; IgG1 and IgG3 subclasses are more
with cisplatin and 13-cis retinoic acid, a response rate of
efficient than IgG2 and IgG4 at mediating lysis of target
72 % for metastatic HNSCC was observed [160].
cells [164, 166, 167]. Lysis of normal cells is also a risk as
normal cells also contain many of the surface antigen
IRX-2 molecules, yet antibody therapy does not typically result in
severe systemic toxicity [164].
Combination therapies may be more effective than single The most widely studied antibodies for HNSCC are
agents. A novel drug named IRX-2 was developed that those targeting the epidermal growth factor receptor
contains a mixture of the antitumoral cytokines IFN-c, IL-2, (EGFR), which is typically overexpressed on the majority
IL-6, IL-8, IL-1b, G-CSF, GM-CSF, and TNF-alpha, as of HNSCC cells [16]. Cetuximab, an immunotherapy cur-
well as agents that reverse immunosuppression, namely rently FDA approved for HNSCC, targets EGFR, which is
indomethacin and cyclophosphamide [161]. Zinc is also a receptor tyrosine kinase. A phase III trial in which ce-
included as it is a necessary mineral for the development tuximab was added to radiotherapy demonstrated an
of cellular immunity. Interestingly, zinc deficiency is increase in locoregional control from 14.9 to 24.4 months
observed in 50 % of patients with HNSCC and correlates (p = 0.005) with limited toxicity [168]. Overall 5-year
with higher cancer stage [162]. Recently tested in a phase survival was also improved from 36.4 to 45.6 %, primarily
II clinical trial, IRX-2 was given to Stage II-IVa HNSCC in patients that experienced at least a grade 2 acneiform
patients for 21 days preoperatively; 83 % had stable rash during treatment [169]. Efficacy of cetuximab and

123
Immunology & Microbiology in Miami (2013) 57:5269 61

other EGFR antibody therapies is limited by HNSCC opsonization and cytokine washout, the cells were rein-
resistance through upregulation of other receptor tyrosine troduced into the patients. One patient showed stable dis-
kinases such as HER2, HER3, and MET, altered EGFR ease for 6 months and another went into complete
function after nuclear translocation, and altered expression remission during the follow-up period of 27 months. A
of downstream effectors of EGFR signaling [142]. Com- recent study made use of bimodal ex vivo expansion to
bination therapy may be necessary to increase efficacy. A expand harvested tumor infiltrating lymphocytes using
number of additional antibody therapies for HNSCC are anti-CD3 antibody, feeder cells, and high-dose IL-2. Just
also being studied, including ones that target HER2/neu, 17 days were required to create up to 3,500-fold expansion
VEGF, CD45, CTLA-4, PDL-1, or carcinoembryonic of the T cells, most of which were T-effector memory cells
antigen (CEA) [93]. Various monoclonal antibody thera- [31]. This method may be used in the future for clinical
pies for HNSCC have been recently reviewed [36, 164, trials in HNSCC patients. Adoptive cell transfer techniques
170, 171]. have also been studied for NK T cells, and one study
Overall, antibody-based immunotherapy for HNSCC demonstrated tumor regression in 5 out of 10 locally
has the greatest clinical efficacy and the most widespread recurrent, operable HNSCC patients [176].
use in clinical practice today as compared to other immu- As with other immunotherapies, adoptive cell transfer
notherapies. However, further investigation is needed in has its limitations that still need to be overcome. Not only
order to increase antibody effectiveness, perhaps by com- is the technique cumbersome and technically challenging,
bining with agents that reverse local immunosuppression. but also many tumor-specific lymphocytes that pre-exist in
Conjugated antibodies, in which the antibody is conjugated the patient are anergic to in vitro re-stimulation [177]. A
with a radioactive substance, drug, or toxin, are an alter- new strategy involving gene modification of T lympho-
native strategy for antibody therapy designed to take cytes may be a solution [135, 178]; PBMCs from patients
advantage of antibody specificity as a means of targeted are retrovirally transduced with a T-cell receptor (TCR)
delivery of a non-immunotherapeutic agent. One conju- specific for the tumor, and additional genes that protect the
gated antibody tested in a HNSCC phase I clinical trial was lymphocytes from the tumor suppressive mechanisms can
186
Re-labeled humanized bivatuzumab, which demon- also be added [179]. In a study with melanoma patients,
strated that the drug was well tolerated and led to delivery PBMCs were transfected with high-affinity T-cell receptors
of a median of 12.4 Gy to the tumor cells [172]. Studies specific for p53HLA-A2 complex and were shown to be
evaluating the use of other labeled or loaded antibodies to able to recognize different human tumor cells lines
treat various cancer cell types are ongoing [173]. expressing p53 [180]. As many HNSCC tumors express
p53 [181, 182], this may become a potential therapy for
Adoptive cell transfer HNSCC patients.

Adoptive cell transfer involves harvesting and priming a


patients own tumor antigen-specific effector cells before Anti-tumor vaccines
re-introducing them to the patient. In one such trial, 17
patients with HNSCC were injected in the thigh with Dendritic cell-based vaccines
irradiated autologous tumor cells admixed with GM-CSF
followed by 3 additional injections of GM-CSF at the site Dendritic cell counts are generally reduced in HNSCC
[174]. The following week, the inguinal lymph nodes patients, but patient monocytes can be used to generate
draining the injection site were resected, and the lympho- autologous DCs in vitro, or allogeneic DC lines can be
cytes harvested from these nodes were activated with manipulated. These DCs loaded with TAAs such as p53
staphylococcal enterotoxin A and expanded in IL-2 [183], tumor RNA [184, 185], or tumor DNA [36] are
in vitro. The CD3-enriched cells were then infused back being used as vaccines in patients. These TAA-specific
into the patients peripherally. Three patients had disease DCs are able to produce IFN-c in vitro [184, 186]. Stage
stabilization where progression had been evident prior to I-IVa HNSCC patients with no active disease were vacci-
the treatment. Another trial involved 4 patients in which nated with autologous DC pulsed with p53 and a tetanus-
harvested peripheral blood mononuclear cells (PBMCs) derived helper peptide. A phase I clinical trial [187]
were collected by leukophoresis and then incubated ex vivo demonstrated the safety of this vaccine and an increase in
with catumaxomab, a monoclonal antibody that binds p53-specific T cells in 11 of 16 patients. Frequencies and
CD3? T cells with one arm and epithelial cell adhesion absolute numbers of Tregs were significantly decreased
molecule (EpCAM) with the other [175]. Catumaxomab- after vaccination. More importantly, disease-free survival
loaded PBMCs were shown to release IFN gamma and at 24 months appeared to be favorable as compared to
gramzyme B when coincubated with EpCAM? cells. After historical unvaccinated HNSCC patients.

123
62 Immunology & Microbiology in Miami (2013) 57:5269

Peptide-based vaccines developed locoregional recurrence, and disease-specific


survival was 82.4 % at median follow-up of 29 months.
Peptide-based vaccines, also known as Trojan vaccines, Additional clinical trials with OncoVEX are ongoing.
utilize a penetrin peptide sequence derived from HIV-TAT,
which allows the entire peptide to translocate through the
cell membrane and penetrate directly into the endoplasmic Therapeutic HPV vaccines in HNSCC
reticulum and the Golgi apparatus [188]. Once there, they
can form peptideHLA complexes without the need of Therapeutic HPV vaccines are being developed that target
proteasome processing and TAP transportation. This ther- E6 and E7, which are constitutively expressed in HPV-
apeutic modality has been attempted in a pilot study with infected cells [193]. These vaccines include the use of
HNSCC patients. MAGE 3 and HPV-16 peptides fused to HPV-E6 peptides, live attenuated listeria monocytogenes
the penetrin peptide were used in 4 consecutive immuni- bacteria carrying E7 fusion protein, Trojan peptides, vac-
zations with GM-CSF and Montanide ISA 51 as adjuvant. cinia based vaccines, naked DNA vaccines, and DC-based
Most patients demonstrated systemic immune response vaccines [194]. The vaccine utilizing listeria is interesting
against the Trojan and HLA-II-restricted peptides [189]. in that it takes advantage of the capacity of listeria to infect
Analysis of the tumor specimen from one patient after antigen-presenting cells and promote DC antigen cross-
treatment revealed significant infiltration of MAGE-spe- presentation; subsequently, effector T cells are activated
cific CD4? and CD8? T cells as well as large areas of while the innate immune system is simultaneously stimu-
apoptotic tumor cells. Phase II clinical trials using MAGE-A lated by the bacteria [195]. Preclinical data demonstrate
and HPV-16 antigens are currently ongoing [36]. that this technique induces the regression of established
tumors by reducing the suppressive activity of Tregs and
Whole tumor vaccines MDSCs at the tumor site, by promoting the chemotaxis and
maturation of DCs, and by generating memory effector T
Whole tumor vaccines attempt to target the whole reper- cells [196]. In a phase I trial of refractory cervical cancer
toire of tumor antigens so as to minimize the chance that patients receiving the listeria-based vaccine, 1-year sur-
further immunoediting and subsequent tumor resistance. vival increased to 53 % from the historical 5 %, with a
One technique involves re-injecting autologous irradiated significant reduction in tumor size in 33 % [197]. Various
tumor cells into patients along with GM-CSF and then clinical trials with HPV vaccines are ongoing [198].
harvesting the sentinel node lymphocytes that are theoret-
ically sensitized to multiple TAAs for subsequent adoptive
cell transfer [174], as already described above. Another Conclusion
strategy involves using oncolytic viruses with genes
encoding immune-stimulatory cytokines such as GM-CSF Recent advances in our understanding of the complex
[190]. The rationale for use of these viruses is that they interactions between HNSCC and the immune system have
selectively replicate in a tumor leading to lytic tumor cell led to a number of innovative immunotherapeutic strate-
death and also lead to release of tumor antigens along with gies. While most of these strategies are still investigational,
stimulating cytokines, inducing a tumor-specific immune their existence represents both a hope for improved
response that protects the host from local and distant HNSCC survival in the near future and an opportunity to
recurrence. A phase I trial of OncoVEXGM-CSF, which shape the future of immunologic cancer research. It is
consists of a herpes simplex virus expressing GM-CSF, increasingly apparent that efforts to generate an effective
was conducted on stage III/IV HNSCC patients. Pre- anti-tumor immune response must be coupled to strategies
liminary data demonstrated that 6 of 8 patients had a that work to abrogate the strong immunosuppressive net-
complete pathologic response with only mild toxicity work orchestrated by the tumor. As more is discovered
[191]. Another study was conducted in which the virus was about the role of immune system in carcinogenesis, the
injected intratumorally in stage III/IVA/IVB HNSCC opportunity for earlier intervention may also become pos-
patients in conjunction with neoadjuvant chemoradiother- sible. HNSCC remains a debilitating and deadly disease,
apy [192]. All patients underwent neck dissection 6 yet through the enthusiastic study of its interaction with the
10 weeks later. 82.3 % of the patients showed tumor immune system, the promise of immunotherapy is stronger
response by Response Evaluation Criteria in Solid Tumors, than ever.
and pathologic complete remission was confirmed in 93 %
Acknowledgments This work was supported by the Flight Atten-
of the patients at neck dissection. HSV was detected in
dant Medical Research Institute (FAMRI)s young investigator
injected and adjacent uninjected tumors at levels higher award, by the Bankhead Coley Cancer Research Program Grant
than the input dose, indicating viral replication. No patient 2BF0650904.

123
Immunology & Microbiology in Miami (2013) 57:5269 63

Conflict of interest The authors declare that they have no conflict 18. Schuler PJ, Boeckers P, Engers R, Boelke E, Bas M, Greve J,
of interest. et al. EGFR-specific T cell frequencies correlate with EGFR
expression in head and neck squamous cell carcinoma. J Transl
Med. 2011;9:168. doi:10.1186/1479-5876-9-168.
19. Hoffmann TK, Donnenberg AD, Finkelstein SD, Donnenberg
VS, Friebe-Hoffmann U, Myers EN, et al. Frequencies of tet-
References ramer? T cells specific for the wild-type sequence p53(264
272) peptide in the circulation of patients with head and neck
1. Duray A, Demoulin S, Hubert P, Delvenne P, Saussez S. cancer. Cancer Res. 2002;62(12):35219.
Immune suppression in head and neck cancers: a review. Clin 20. Uppaluri R, Dunn GP, Lewis JS Jr. Focus on TILs: prognostic
Dev Immunol. 2010;2010:701657. doi:10.1155/2010/701657. significance of tumor infiltrating lymphocytes in head and neck
2. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, Parkin DM. cancers. Cancer Immun. 2008;8:16.
GLOBOCAN 2008 v2.0, Cancer Incidence and Mortality 21. Schutt C, Bumm K, Mirandola L, Bernardini G, DCunha N,
Worldwide: IARC CancerBase No. 10 [Internet]. Lyon, France: Tijani L, et al. Immunological treatment options for locore-
International Agency for Research on Cancer 2010. gionally advanced head and neck squamous cell carcinoma. Int
3. Hunter AM, LaCasse EC, Korneluk RG. The inhibitors of Rev Immunol. 2012;31(1):2242. doi:10.3109/08830185.2011.
apoptosis (IAPs) as cancer targets. Apoptosis. 2007;12(9):1543 637253.
68. doi:10.1007/s10495-007-0087-3. 22. Szczepanski MJ, Czystowska M, Szajnik M, Harasymczuk M,
4. Burnet FM. The concept of immunological surveillance. Prog Boyiadzis M, Kruk-Zagajewska A, et al. Triggering of Toll-like
Exp Tumor Res. 1970;13:127. receptor 4 expressed on human head and neck squamous cell
5. French JD. Revisiting immune-based therapies for aggressive carcinoma promotes tumor development and protects the tumor
follicular cell-derived thyroid cancers. Thyroid. 2013;23(5): from immune attack. Cancer Res. 2009;69(7):310513. doi:10.
52942. doi:10.1089/thy2012.0566. 1158/0008-5472.can-08-3838.
6. Badoual C, Sandoval F, Pere H, Hans S, Gey A, Merillon N, 23. Saussez S, Camby I, Toubeau G, Kiss R. Galectins as modula-
et al. Better understanding tumor-host interaction in head and tors of tumor progression in head and neck squamous cell car-
neck cancer to improve the design and development of immu- cinomas. Head Neck. 2007;29(9):87484. doi:10.1002/hed.
notherapeutic strategies. Head Neck. 2010;32(7):94658. doi:10. 20559.
1002/hed.21346. 24. Gastman BR, Atarshi Y, Reichert TE, Saito T, Balkir L, Rabi-
7. Harris JP, Penn I. Immunosuppression and the development of nowich H, et al. Fas ligand is expressed on human squamous cell
malignancies of the upper airway and related structures. carcinomas of the head and neck, and it promotes apoptosis of T
Laryngoscope. 1981;91(4):5208. lymphocytes. Cancer Res. 1999;59(20):535664.
8. King GN, Healy CM, Glover MT, Kwan JT, Williams DM, 25. Gastman BR, Yin XM, Johnson DE, Wieckowski E, Wang GQ,
Leigh IM, et al. Increased prevalence of dysplastic and malig- Watkins SC, et al. Tumor-induced apoptosis of T cells: ampli-
nant lip lesions in renal-transplant recipients. N Engl J Med. fication by a mitochondrial cascade. Cancer Res. 2000;60(24):
1995;332(16):10527. doi:10.1056/NEJM199504203321602. 68117.
9. Avital I, Moreira AL, Klimstra DS, Leversha M, Papadopoulos 26. Kassouf N, Thornhill MH. Oral cancer cell lines can use mul-
EB, Brennan M, et al. Donor-derived human bone marrow cells tiple ligands, including Fas-L, TRAIL and TNF-alpha, to induce
contribute to solid organ cancers developing after bone marrow apoptosis in Jurkat T cells: possible mechanisms for immune
transplantation. Stem Cells. 2007;25(11):29039. doi:10.1634/ escape by head and neck cancers. Oral Oncol. 2008;44(7):672
stemcells.2007-0409. 82. doi:10.1016/j.oraloncology.2007.08.013.
10. Baker KS, DeFor TE, Burns LJ, Ramsay NK, Neglia JP, Rob- 27. Lyford-Pike S, Peng S, Young GD, Taube JM, Westra WH,
ison LL. New malignancies after blood or marrow stem-cell Akpeng B, et al. Evidence for a role of the PD-1:PD-L1 pathway
transplantation in children and adults: incidence and risk factors. in immune resistance of HPV-associated head and neck squa-
J Clin Oncol. 2003;21(7):13528. mous cell carcinoma. Cancer Res. 2013;. doi:10.1158/0008-
11. Bhatia S, Louie AD, Bhatia R, ODonnell MR, Fung H, Kashyap 5472.CAN-12-2384.
A, et al. Solid cancers after bone marrow transplantation. J Clin 28. Zha Y, Blank C, Gajewski TF. Negative regulation of T-cell
Oncol. 2001;19(2):46471. function by PD-1. Crit Rev Immunol. 2004;24(4):22937.
12. Haigentz M Jr. Aerodigestive cancers in HIV infection. Curr 29. Kuss I, Hathaway B, Ferris RL, Gooding W, Whiteside TL.
Opin Oncol. 2005;17(5):4748. Decreased absolute counts of T lymphocyte subsets and their
13. Singh B, Balwally AN, Shaha AR, Rosenfeld RM, Har-El G, relation to disease in squamous cell carcinoma of the head and
Lucente FE. Upper aerodigestive tract squamous cell carcinoma. neck. Clin Cancer Res. 2004;10(11):375562. doi:10.1158/
The human immunodeficiency virus connection. Arch Otolar- 1078-0432.CCR-04-0054.
yngol Head Neck Surg. 1996;122(6):63943. 30. Turksma A, Bontkes H, van den Heuvel H, de Gruijl T, von
14. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Blomberg B, Braakhuis B, et al. Effector memory T-cell
integrating immunitys roles in cancer suppression and promo- frequencies in relation to tumour stage, location and HPV
tion. Science. 2011;331(6024):156570. doi:10.1126/science. status in HNSCC patients. Oral Dis. 2012;. doi:10.1111/odi.
1203486. 12037.
15. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer 31. Junker N, Kvistborg P, Kollgaard T, Straten P, Andersen MH,
immunoediting: from immunosurveillance to tumor escape. Nat Svane IM. Tumor associated antigen specific T-cell populations
Immunol. 2002;3(11):9918. doi:10.1038/ni1102-991. identified in ex vivo expanded TIL cultures. Cell Immunol.
16. Allen CT, Judd NP, Bui JD, Uppaluri R. The clinical implica- 2012;273(1):19. doi:10.1016/j.cellimm.2011.12.004.
tions of antitumor immunity in head and neck cancer. Laryn- 32. Ogino T, Shigyo H, Ishii H, Katayama A, Miyokawa N, Har-
goscope. 2012;122(1):14457. doi:10.1002/lary.21913. abuchi Y, et al. HLA class I antigen down-regulation in primary
17. Ferris RL, Whiteside TL, Ferrone S. Immune escape associated laryngeal squamous cell carcinoma lesions as a poor prognostic
with functional defects in antigen-processing machinery in head marker. Cancer Res. 2006;66(18):92819. doi:10.1158/0008-
and neck cancer. Clin Cancer Res. 2006;12(13):38905. 5472.CAN-06-0488.

123
64 Immunology & Microbiology in Miami (2013) 57:5269

33. Wolf GT, Hudson JL, Peterson KA, Miller HL, McClatchey KD. 51. Pahler JC, Tazzyman S, Erez N, Chen YY, Murdoch C, Nozawa
Lymphocyte subpopulations infiltrating squamous carcinomas H, et al. Plasticity in tumor-promoting inflammation: impair-
of the head and neck: correlations with extent of tumor and ment of macrophage recruitment evokes a compensatory neu-
prognosis. Otolaryngol Head Neck Surg. 1986;95(2):14252. trophil response. Neoplasia. 2008;10(4):32940.
34. Kuss I, Hathaway B, Ferris RL, Gooding W, Whiteside TL. 52. Xiang M, Gu Y, Zhao F, Lu H, Chen S, Yin L. Mast cell tryptase
Imbalance in absolute counts of T lymphocyte subsets in promotes breast cancer migration and invasion. Oncol Rep.
patients with head and neck cancer and its relation to disease. 2010;23(3):6159.
Adv Otorhinolaryngol. 2005;62:16172. 53. Neuchrist C. Vascular endothelial growth factor (VEGF) and
35. Farace F, Angevin E, Vanderplancke J, Escudier B, Triebel F. microvessel density in squamous cell carcinomas of the larynx:
The decreased expression of CD3 zeta chains in cancer patients an immunohistochemical study. Acta Otolaryngol. 1999;119(6):
is not reversed by IL-2 administration. Int J Cancer. 1994; 7328.
59(6):7525. 54. Salven P, Heikkila P, Anttonen A, Kajanti M, Joensuu H.
36. Varilla V, Atienza J, Dasanu CA. Immune alterations and Vascular endothelial growth factor in squamous cell head and
immunotherapy prospects in head and neck cancer. Expert Opin neck carcinoma: expression and prognostic significance. Mod
Biol Ther. 2013;. doi:10.1517/14712598.2013.810716. Pathol. 1997;10(11):1128.
37. Sinkovics JG, Horvath JC. Human natural killer cells: a com- 55. Liss C, Fekete MJ, Hasina R, Lam CD, Lingen MW. Paracrine
prehensive review. Int J Oncol. 2005;27(1):547. angiogenic loop between head-and-neck squamous-cell carci-
38. Pegram HJ, Andrews DM, Smyth MJ, Darcy PK, Kershaw MH. nomas and macrophages. Int J Cancer. 2001;93(6):7815.
Activating and inhibitory receptors of natural killer cells. 56. Marcus B, Arenberg D, Lee J, Kleer C, Chepeha DB, Schmal-
Immunol Cell Biol. 2011;89(2):21624. doi:10.1038/icb.2010. bach CE, et al. Prognostic factors in oral cavity and oropha-
78. ryngeal squamous cell carcinoma. Cancer. 2004;101(12):2779
39. Duffey DC, Chen Z, Dong G, Ondrey FG, Wolf JS, Brown K, 87. doi:10.1002/cncr.20701.
et al. Expression of a dominant-negative mutant inhibitor-kap- 57. Bardos H, Juhasz A, Repassy G, Adany R. Fibrin deposition in
paBalpha of nuclear factor-kappaB in human head and neck squamous cell carcinomas of the larynx and hypopharynx.
squamous cell carcinoma inhibits survival, proinflammatory Thromb Haemost. 1998;80:76772.
cytokine expression, and tumor growth in vivo. Cancer Res. 58. Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E,
1999;59(14):346874. Serafini P, et al. Myeloid suppressor lines inhibit T cell
40. Molling JW, Langius JA, Langendijk JA, Leemans CR, Bontkes responses by an NO-dependent mechanism. J Immunol.
HJ, van der Vliet HJ, et al. Low levels of circulating invariant 2002;168(2):68995.
natural killer T cells predict poor clinical outcome in patients 59. Pak AS, Wright MA, Matthews JP, Collins SL, Petruzzelli GJ,
with head and neck squamous cell carcinoma. J Clin Oncol. Young MR. Mechanisms of immune suppression in patients
2007;25(7):8628. doi:10.1200/jco.2006.08.5787. with head and neck cancer: presence of CD34(?) cells which
41. Liu K, Nussenzweig MC. Origin and development of dendritic suppress immune functions within cancers that secrete granu-
cells. Immunol Rev. 2010;234(1):4554. doi:10.1111/j.0105- locyte-macrophage colony-stimulating factor. Clin Cancer Res.
2896.2009.00879.x. 1995;1(1):95103.
42. Valladeau J, Saeland S. Cutaneous dendritic cells. Semin 60. Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Pi-
Immunol. 2005;17(4):27383. doi:10.1016/j.smim.2005.05.009. azuelo MB, et al. Arginase I production in the tumor microen-
43. Yilmaz T, Gedikoglu G, Celik A, Onerci M, Turan E. Prognostic vironment by mature myeloid cells inhibits T-cell receptor
significance of Langerhans cell infiltration in cancer of the lar- expression and antigen-specific T-cell responses. Cancer Res.
ynx. Otolaryngol Head Neck Surg. 2005;132(2):30916. doi:10. 2004;64(16):583949.
1016/j.otohns.2004.04.018. 61. Bogdan C. Nitric oxide and the immune response. Nat Immunol.
44. Gallo O, Libonati GA, Gallina E, Fini-Storchi O, Giannini A, 2001;2(10):90716.
Urso C, et al. Langerhans cells related to prognosis in patients 62. Wu G, Morris SM Jr. Arginine metabolism: nitric oxide and
with laryngeal carcinoma. Arch Otolaryngol Head Neck Surg. beyond. Biochem J. 1998;336(Pt 1):117.
1991;117(9):100710. 63. Bronte V, Serafini P, De Santo C, Marigo I, Tosello V, Mazzoni
45. Albuquerque RL Jr, Miguel MC, Costa AL, Souza LB. Corre- A, et al. IL-4-induced arginase 1 suppresses alloreactive T cells
lation of c-erbB-2 and S-100 expression with the malignancy in tumor- bearing mice. J Immunol. 2003;170(1):2708.
grading and anatomical site in oral squamous cell carcinoma. Int 64. Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L,
J Exp Pathol. 2003;84(6):25965. et al. Altered recognition of antigen is a mechanism of CD8(?)
46. Li X, Takahashi Y, Sakamoto K, Nakashima T. Expression of T cell tolerance in cancer. Nat Med. 2007;13(7):82835.
dendritic cell phenotypic antigens in cervical lymph nodes of 65. Bentz BG, Haines GK 3rd, Radosevich JA. Increased protein
patients with hypopharyngeal and laryngeal carcinoma. J Lar- nitrosylation in head and neck squamous cell carcinogenesis.
yngol Otol Suppl. 2009;31:510. Head Neck. 2000;22(1):6470.
47. Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, 66. Rosbe KW, Prazma J, Petrusz P, Mims W, Ball SS, Weissler
et al. Clinical significance of defective dendritic cell differen- MC. Immunohistochemical characterization of nitric oxide
tiation in cancer. Clin Cancer Res. 2000;6(5):175566. synthase activity in squamous cell carcinoma of the head and
48. Gabrilovich D. Mechanisms and functional significance of neck. Otolaryngol Head Neck Surg. 1995;113(5):5419.
tumour-induced dendritic-cell defects. Nat Rev Immunol. 67. Pandit R, Lathers DM, Beal NM, Garrity T, Young MR. CD34?
2004;4(12):94152. immune suppressive cells in the peripheral blood of patients
49. Sica A, Larghi P, Mancino A, Rubino L, Porta C, Totaro MG, with head and neck cancer. Ann Otol Rhinol Laryngol.
et al. Macrophage polarization in tumour progression. Semin 2000;109(8 Pt 1):74954.
Cancer Biol. 2008;18(5):34955. doi:10.1016/j.semcancer.2008. 68. Wiers K, Wright MA, Vellody K, Young MR. Failure of tumor-
03.004. reactive lymph node cells to kill tumor in the presence of
50. Coussens LM, Tinkle CL, Hanahan D, Werb Z. MMP-9 sup- immune-suppressive CD34? cells can be overcome with vita-
plied by bone marrow-derived cells contributes to skin carci- min D3 treatment to diminish CD34? cell levels. Clin Exp
nogenesis. Cell. 2000;103(3):48190. Metastasis. 1998;16(3):27582.

123
Immunology & Microbiology in Miami (2013) 57:5269 65

69. Young MR, Cigal M. Tumor skewing of CD34? cell differen- producing vaccines impair the immune response through the
tiation from a dendritic cell pathway into endothelial cells. recruitment of myeloid suppressor cells. Cancer Res.
Cancer Immunol Immunother. 2006;55(5):55868. 2004;64(17):633743. doi:10.1158/0008-5472.CAN-04-0757.
70. Zou W. Regulatory T cells, tumour immunity and immuno- 86. Young MR, Wright MA, Young ME. Antibodies to colony-
therapy. Nat Rev Immunol. 2006;6(4):295307. doi:10.1038/ stimulating factors block Lewis lung carcinoma cell stimulation
nri1806. of immune-suppressive bone marrow cells. Cancer Immuno-
71. Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of lImmunother. 1991;33:14652.
treg-mediated T cell suppression. Front Immunol. 2012;3:51. 87. Dranoff G. GM-CSF-based cancer vaccines. Immunol Rev.
doi:10.3389/fimmu.2012.00051. 2002;188:14754.
72. Curiel TJ. Tregs and rethinking cancer immunotherapy. J Clin 88. Dranoff G. GM-CSF-secreting melanoma vaccines. Oncogene.
Invest. 2007;117(5):116774. doi:10.1172/JCI31202. 2003;22(20):318892.
73. Pretscher D, Distel LV, Grabenbauer GG, Wittlinger M, Bu- 89. Biron P, Sebban C, Gourmet R, Chvetzoff G, Philip I, Blay JY.
ettner M, Niedobitek G. Distribution of immune cells in head Research controversies in management of oral mucositis. Sup-
and neck cancer: CD8? T-cells and CD20? B-cells in meta- port Care Cancer. 2000;8(1):6871.
static lymph nodes are associated with favourable outcome in 90. Parmiani G, Castelli C, Pilla L, Santinami M, Colombo MP,
patients with oro- and hypopharyngeal carcinoma. BMC Cancer. Rivoltini L. Opposite immune functions of GM-CSF adminis-
2009;9:292. doi:10.1186/1471-2407-9-292. tered as vaccine adjuvant in cancer patients. Ann Oncol.
74. Badoual C, Hans S, Rodriguez J, Peyrard S, Klein C. Agueznay 2007;18(2):22632.
Nel H et al. Prognostic value of tumor-infiltrating CD4? T-cell 91. Itoh S, Matsui K, Furuta I, Takano Y. Immunohistochemical
subpopulations in head and neck cancers. Clin Cancer Res. study on overexpression of cyclooxygenase-2 in squamous cell
2006;12(2):46572. doi:10.1158/1078-0432.CCR-05-1886. carcinoma of the oral cavity: its importance as a prognostic
75. Zhang YL, Li J, Mo HY, Qiu F, Zheng LM, Qian CN, et al. predictor. Oral Oncol. 2003;39(8):82935.
Different subsets of tumor infiltrating lymphocytes correlate 92. Nathan CO, Leskov IL, Lin M, Abreo FW, Shi R, Hartman GH,
with NPC progression in different ways. Mol Cancer. 2010;9:4. et al. COX-2 expression in dysplasia of the head and neck:
doi:10.1186/1476-4598-9-4. correlation with elF4E. Cancer. 2001;92(7):188895.
76. Distel LV, Fickenscher R, Dietel K, Hung A, Iro H, Zenk J, et al. 93. Rapidis AD, Wolf GT. Immunotherapy of head and neck cancer:
Tumour infiltrating lymphocytes in squamous cell carcinoma of current and future considerations. J Oncol. 2009;2009:346345.
the oro- and hypopharynx: prognostic impact may depend on doi:10.1155/2009/346345.
type of treatment and stage of disease. Oral Oncol. 94. Serafini P. Editorial: PGE2-producing MDSC: a role in tumor
2009;45(10):e16774. doi:10.1016/j.oraloncology.2009.05.640. progression? J Leukoc Biol. 2010;88(5):8279. doi:10.1189/jlb.
77. Weed DT, Walker G, De La Fuente AC, Nazarian R, Vella JL, 0510303.
Gomez-Fernandez CR, et al. FOXP3 subcellular localization 95. Hoshikawa H, Goto R, Mori T, Mitani T, Mori N. Expression of
predicts recurrence in oral squamous cell carcinoma. PLoS prostaglandin E2 receptors in oral squamous cell carcinomas
ONE. 2013;8(8):e71908. doi:10.1371/journal.pone.0071908. and growth inhibitory effects of an EP3 selective antagonist,
78. Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes ONO-AE3-240. Int J Oncol. 2009;34(3):84752.
RE. Transient expression of FOXP3 in human activated non- 96. Jain RK. Normalization of tumor vasculature: an emerging con-
regulatory CD4? T cells. Eur J Immunol. 2007;37(1):12938. cept in antiangiogenic therapy. Science. 2005;307(5706):5862.
doi:10.1002/eji.200636435. 97. Liang X, Yang D, Hu J, Hao X, Gao J, Mao Z. Hypoxia
79. Magg T, Mannert J, Ellwart JW, Schmid I, Albert MH. Sub- inducible factor-alpha expression correlates with vascular
cellular localization of FOXP3 in human regulatory and non- endothelial growth factor-C expression and lymphangiogenesis/
regulatory T cells. Eur J Immunol. 2012;42(6):162738. doi:10. angiogenesis in oral squamous cell carcinoma. Anticancer Res.
1002/eji.201141838. 2008;28(3A):165966.
80. Agada FO, Alhamarneh O, Stafford ND, Greenman J. Immu- 98. Shang ZJ, Li ZB, Li JR. VEGF is up-regulated by hypoxic
notherapy in head and neck cancer: current practice and future stimulation and related to tumour angiogenesis and severity of
possibilities. J Laryngol Otol. 2009;123(1):1928. doi:10.1017/ disease in oral squamous cell carcinoma: in vitro and in vivo
S0022215108003356. studies. Int J Oral Maxillofac Surg. 2006;35(6):5338. doi:10.
81. Rutkowski MJ, Sughrue ME, Kane AJ, Mills SA, Parsa AT. 1016/j.ijom.2005.09.006.
Cancer and the complement cascade. Mol Cancer Res. 99. Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S,
2010;8(11):145365. doi:10.1158/1541-7786.mcr-10-0225. et al. Vascular endothelial growth factor inhibits the develop-
82. Bronte V, Chappell DB, Apolloni E, Cabrelle A, Wang M, Hwu ment of dendritic cells and dramatically affects the differentia-
P, et al. Unopposed production of granulocyte-macrophage tion of multiple hematopoietic lineages in vivo. Blood.
colony-stimulating factor by tumors inhibits CD8? T cell 1998;92(11):415066.
responses by dysregulating antigen-presenting cell maturation. J 100. Yang L, DeBusk LM, Fukuda K, Fingleton B, Green-Jarvis B,
Immunol. 1999;162(10):572837. Shyr Y, et al. Expansion of myeloid immune suppressor
83. Young MR, Wright MA, Lozano Y, Prechel MM, Benefield J, Gr?CD11b? cells in tumor-bearing host directly promotes
Leonetti JP, et al. Increased recurrence and metastasis in patients tumor angiogenesis. Cancer Cell. 2004;6(4):40921.
whose primary head and neck squamous cell carcinomas 101. Strauss L, Volland D, Kunkel M, Reichert TE. Dual role of
secreted granulocyte-macrophage colony-stimulating factor and VEGF family members in the pathogenesis of head and neck
contained CD34? natural suppressor cells. Int J Cancer. cancer (HNSCC): possible link between angiogenesis and
1997;74(1):6974. immune tolerance. Med Sci Monit. 2005;11(8):BR28092.
84. Adeegbe D, Serafini P, Bronte V, Zoso A, Ricordi C, Inverardi 102. Karatzanis AD, Koudounarakis E, Papadakis I, Velegrakis G.
L. In vivo induction of myeloid suppressor cells and CD4Foxp3 Molecular pathways of lymphangiogenesis and lymph node
T regulatory cells prolongs skin allograft survival in mice. Cell metastasis in head and neck cancer. Eur Arch Otorhinolaryngol.
Transplant. 2011;. doi:10.3727/096368910X540621. 2012;269(3):7317. doi:10.1007/s00405-011-1809-2.
85. Serafini P, Carbley R, Noonan KA, Tan G, Bronte V, Borrello I. 103. Pries R, Thiel A, Brocks C, Wollenberg B. Secretion of tumor-
High-dose granulocyte-macrophage colony-stimulating factor- promoting and immune suppressive cytokines by cell lines of

123
66 Immunology & Microbiology in Miami (2013) 57:5269

head and neck squamous cell carcinoma. In Vivo. 121. Shields JD, Fleury ME, Yong C, Tomei AA, Randolph GJ,
2006;20(1):458. Swartz MA. Autologous chemotaxis as a mechanism of tumor
104. Squarize CH, Castilho RM, Sriuranpong V, Pinto DS Jr, cell homing to lymphatics via interstitial flow and autocrine
Gutkind JS. Molecular cross-talk between the NFkappaB and CCR7 signaling. Cancer Cell. 2007;11(6):52638. doi:10.1016/
STAT3 signaling pathways in head and neck squamous cell j.ccr.2007.04.020.
carcinoma. Neoplasia. 2006;8(9):73346. doi:10.1593/neo. 122. Wang J, Xi L, Hunt JL, Gooding W, Whiteside TL, Chen Z,
06274. et al. Expression pattern of chemokine receptor 6 (CCR6) and
105. Kawakami K, Leland P, Puri RK. Structure, function, and tar- CCR7 in squamous cell carcinoma of the head and neck iden-
geting of interleukin 4 receptors on human head and neck cancer tifies a novel metastatic phenotype. Cancer Res. 2004;64(5):
cells. Cancer Res. 2000;60(11):29817. 18616.
106. Gallina G, Dolcetti L, Serafini P, Santo CD, Marigo I, Colombo 123. Brown CE, Vishwanath RP, Aguilar B, Starr R, Najbauer J,
MP, et al. Tumors induce a subset of inflammatory monocytes Aboody KS, et al. Tumor-derived chemokine MCP-1/CCL2 is
with immunosuppressive activity on CD8 T cells. J Clin Invest. sufficient for mediating tumor tropism of adoptively transferred
2006;116(10):277790. T cells. J Immunol. 2007;179(5):333241.
107. Roth F, De La Fuente AC, Vella JL, Zoso A, Inverardi L, 124. Bailey C, Negus R, Morris A, Ziprin P, Goldin R, Allavena P,
Serafini P. Aptamer-mediated blockade of IL4Ralpha triggers et al. Chemokine expression is associated with the accumulation
apoptosis of MDSCs and limits tumor progression. Cancer Res. of tumour associated macrophages (TAMs) and progression in
2012;72(6):137383. doi:10.1158/0008-5472.CAN-11-2772. human colorectal cancer. Clin Exp Metastasis. 2007;24(2):121
108. Riedel F, Zaiss I, Herzog D, Gotte K, Naim R, Hormann K. 30. doi:10.1007/s10585-007-9060-3.
Serum levels of interleukin-6 in patients with primary head and 125. Huang B, Lei Z, Zhao J, Gong W, Liu J, Chen Z, et al. CCL2/
neck squamous cell carcinoma. Anticancer Res. 2005;25(4): CCR2 pathway mediates recruitment of myeloid suppressor
27615. cells to cancers. Cancer Lett. 2007;252(1):8692. doi:10.1016/j.
109. Park SJ, Nakagawa T, Kitamura H, Atsumi T, Kamon H, Sawa canlet.2006.12.012.
S, et al. IL-6 regulates in vivo dendritic cell differentiation 126. Loberg RD, Day LL, Harwood J, Ying C, St John LN, Giles R,
through STAT3 activation. J Immunol. 2004;173(6):384454. et al. CCL2 is a potent regulator of prostate cancer cell migra-
110. Ratta M, Fagnoni F, Curti A, Vescovini R, Sansoni P, Oliviero tion and proliferation. Neoplasia. 2006;8(7):57886. doi:10.
B, et al. Dendritic cells are functionally defective in multiple 1593/neo.06280.
myeloma: the role of interleukin-6. Blood. 2002;100(1):2307. 127. Ueno T, Toi M, Saji H, Muta M, Bando H, Kuroi K, et al.
111. Hayashi T, Hideshima T, Akiyama M, Raje N, Richardson P, Significance of macrophage chemoattractant protein-1 in mac-
Chauhan D, et al. Ex vivo induction of multiple myeloma-spe- rophage recruitment, angiogenesis, and survival in human breast
cific cytotoxic T lymphocytes. Blood. 2003;102(4):143542. cancer. Clin Cancer Res. 2000;6(8):32829.
112. Marigo I, Bosio E, Solito S, Mesa C, Fernandez A, Dolcetti L, 128. Molon B, Viola A, Bronte V. Smoothing T cell roads to the
et al. Tumor-induced tolerance and immune suppression depend tumor: chemokine post-translational regulation. Oncoimmunol-
on the C/EBPbeta transcription factor. Immunity. 2010;32(6): ogy. 2012;1(3):3902. doi:10.4161/onci.19069.
790802. doi:10.1016/j.immuni.2010.05.010. 129. Molon B, Ugel S, Del Pozzo F, Soldani C, Zilio S, Avella D,
113. Hong DS, Angelo LS, Kurzrock R. Interleukin-6 and its receptor et al. Chemokine nitration prevents intratumoral infiltration of
in cancer: implications for translational therapeutics. Cancer. antigen-specific T cells. J Exp Med. 2011;208(10):194962.
2007;110(9):191128. doi:10.1002/cncr.22999. doi:10.1084/jem.20101956.
114. Azenshtein E, Luboshits G, Shina S, Neumark E, Shahbazian D, 130. Ang KK, Harris J, Wheeler R, Weber R, Rosenthal DI, Nguyen-
Weil M, et al. The CC chemokine RANTES in breast carcinoma Tan PF, et al. Human papillomavirus and survival of patients
progression: regulation of expression and potential mechanisms with oropharyngeal cancer. N Engl J Med. 2010;363(1):2435.
of promalignant activity. Cancer Res. 2002;62(4):1093102. doi:10.1056/NEJMoa0912217.
115. Sun Y, Cheng Z, Ma L, Pei G. Beta-arrestin2 is critically 131. Williams R, Lee DW, Elzey BD, Anderson ME, Hostager BS,
involved in CXCR4-mediated chemotaxis, and this is mediated Lee JH. Preclinical models of HPV? and HPV- HNSCC in
by its enhancement of p38 MAPK activation. J Biol Chem. mice: an immune clearance of HPV? HNSCC. Head Neck.
2002;277(51):492129. doi:10.1074/jbc.M207294200. 2009;31(7):9118. doi:10.1002/hed.21040.
116. Zlotnik A. Involvement of chemokine receptors in organ-spe- 132. Marty R, Roze S, Bresse X, Largeron N, Smith-Palmer J.
cific metastasis. Contrib Microbiol. 2006;13:1919. doi:10. Estimating the clinical benefits of vaccinating boys and girls
1159/000092973. against HPV-related diseases in Europe. BMC Cancer.
117. Nath A, Chattopadhya S, Chattopadhyay U, Sharma NK. Mac- 2013;13:10. doi:10.1186/1471-2407-13-10.
rophage inflammatory protein (MIP)1alpha and MIP1beta dif- 133. Fishman MN, Thompson JA, Pennock GK, Gonzalez R, Diez
ferentially regulate release of inflammatory cytokines and LM, Daud AI, et al. Phase I trial of ALT-801, an interleukin-2/
generation of tumoricidal monocytes in malignancy. Cancer T-cell receptor fusion protein targeting p53 (aa264272)/HLA-
Immunol Immunother. 2006;55(12):153441. doi:10.1007/ A*0201 complex, in patients with advanced malignancies. Clin
s00262-006-0149-3. Cancer Res. 2011;17(24):776575. doi:10.1158/1078-0432.
118. Vaday GG, Peehl DM, Kadam PA, Lawrence DM. Expression CCR-11-1817.
of CCL5 (RANTES) and CCR5 in prostate cancer. Prostate. 134. Rosenberg SA, Lotze MT, Yang JC, Aebersold PM, Linehan
2006;66(2):12434. doi:10.1002/pros.20306. WM, Seipp CA, et al. Experience with the use of high-dose
119. Viola A, Sarukhan A, Bronte V, Molon B. The pros and cons of interleukin-2 in the treatment of 652 cancer patients. Ann Surg.
chemokines in tumor immunology. Trends Immunol. 2012;. 1989;210(4):47484 (discussion 8485).
doi:10.1016/j.it.2012.05.007. 135. Rossig C, Brenner MK. Genetic modification of T lymphocytes
120. Nesbeth Y, Scarlett U, Cubillos-Ruiz J, Martinez D, Engle X, for adoptive immunotherapy. Mol Ther. 2004;10(1):518.
Turk MJ, et al. CCL5-mediated endogenous antitumor immunity doi:10.1016/j.ymthe.2004.04.014.
elicited by adoptively transferred lymphocytes and dendritic cell 136. Shirakura Y, Mizuno Y, Wang L, Imai N, Amaike C, Sato E,
depletion. Cancer Res. 2009;69(15):63318. doi:10.1158/0008- et al. T-cell receptor gene therapy targeting melanoma-associ-
5472.CAN-08-4329. ated antigen-A4 inhibits human tumor growth in non-obese

123
Immunology & Microbiology in Miami (2013) 57:5269 67

diabetic/SCID/gammacnull mice. Cancer Sci. 2012;103(1):17 factor pathway in human cancer cells. Mol Cancer Ther.
25. doi:10.1111/j.1349-7006.2011.02111.x. 2007;6(4):14339. doi:10.1158/1535-7163.MCT-06-0677.
137. van Herpen CM, van der Laak JA, de Vries IJ, van Krieken JH, 151. Walsh JE, Clark AM, Day TA, Gillespie MB, Young MR. Use
de Wilde PC, Balvers MG, et al. Intratumoral recombinant of alpha,25-dihydroxyvitamin D3 treatment to stimulate
human interleukin-12 administration in head and neck squamous immune infiltration into head and neck squamous cell carci-
cell carcinoma patients modifies locoregional lymph node noma. Hum Immunol. 2010;71(7):65965. doi:10.1016/j.
architecture and induces natural killer cell infiltration in the humimm.2010.04.008.
primary tumor. Clin Cancer Res. 2005;11(5):1899909. doi:10. 152. Ochoa AC, Zea AH, Hernandez C, Rodriguez PC. Arginase,
1158/1078-0432.ccr-04-1524. prostaglandins, and myeloid-derived suppressor cells in renal
138. Ochi T, Fujiwara H, Yasukawa M. Application of adoptive T- cell carcinoma. Clin Cancer Res. 2007;13(2 Pt 2):721s6s.
cell therapy using tumor antigen-specific T-cell receptor gene doi:10.1158/1078-0432.CCR-06-2197.
transfer for the treatment of human leukemia. J Biomed Bio- 153. Kao J, Genden EM, Chen CT, Rivera M, Tong CC, Mis-
technol. 2010;2010:521248. doi:10.1155/2010/521248. iukiewicz K, et al. Phase 1 trial of concurrent erlotinib, cele-
139. Richtsmeier WJ, Koch WM, McGuire WP, Poole ME, Chang coxib, and reirradiation for recurrent head and neck cancer.
EH. Phase I-II study of advanced head and neck squamous cell Cancer. 2011;117(14):317381. doi:10.1002/cncr.25786.
carcinoma patients treated with recombinant human interferon 154. Wirth LJ, Haddad RI, Lindeman NI, Zhao X, Lee JC, Joshi VA,
gamma. Arch Otolaryngol Head Neck Surg. 1990;116(11): et al. Phase I study of gefitinib plus celecoxib in recurrent or
12717. metastatic squamous cell carcinoma of the head and neck. J Clin
140. Mahjoubi R, Bachouchi M, Munck JN, Busson P, Gasmi J, Azli Oncol. 2005;23(28):697681. doi:10.1200/JCO.2005.02.4182.
N, et al. Phase II trial of recombinant interferon gamma in 155. Dajani EZ, Islam K. Cardiovascular and gastrointestinal toxicity
refractory undifferentiated carcinoma of the nasopharynx. Head of selective cyclo-oxygenase-2 inhibitors in man. J Physiol
Neck. 1993;15(2):1158. Pharmacol. 2008;59(Suppl 2):11733.
141. Urba SG, Forastiere AA, Wolf GT, Amrein PC. Intensive 156. Serafini P, Meckel K, Kelso M, Noonan K, Califano J, Koch W,
recombinant interleukin-2 and alpha-interferon therapy in et al. Phosphodiesterase-5 inhibition augments endogenous
patients with advanced head and neck squamous carcinoma. antitumor immunity by reducing myeloid-derived suppressor
Cancer. 1993;71(7):232631. cell function. J Exp Med. 2006;203(12):2691702.
142. Burtness B, Bauman JE, Galloway T. Novel targets in HPV- 157. Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived
negative head and neck cancer: overcoming resistance to EGFR suppressor cells promote cross-tolerance in B-cell lymphoma by
inhibition. Lancet Oncol. 2013;14(8):e3029. doi:10.1016/ expanding regulatory T cells. Cancer Res. 2008;68(13):5439
s1470-2045(13)70085-8. 49. doi:10.1158/0008-5472.CAN-07-6621.
143. Ozao-Choy J, Ma G, Kao J, Wang GX, Meseck M, Sung M, 158. Airoldi M, Cortesina G, Giordano C, Pedani F, Bumma C. If-
et al. The novel role of tyrosine kinase inhibitor in the reversal osfamide in the treatment of head and neck cancer. Oncology.
of immune suppression and modulation of tumor microenvi- 2003;65(Suppl 2):3743. doi:73356.
ronment for immune-based cancer therapies. Cancer Res. 159. Ghiringhelli F, Menard C, Puig PE, Ladoire S, Roux S, Martin F,
2009;69(6):251422. doi:10.1158/0008-5472.CAN-08-4709. et al. Metronomic cyclophosphamide regimen selectively depletes
144. Porta C, Paglino C, Imarisio I, Ganini C, Pedrazzoli P. Immu- CD4?CD25? regulatory T cells and restores T and NK effector
nological effects of multikinase inhibitors for kidney cancer: a functions in end stage cancer patients. Cancer Immunol Immun-
clue for integration with cellular therapies? J Cancer. other. 2007;56(5):6418. doi:10.1007/s00262-006-0225-8.
2011;2:3338. 160. Recchia F, Lalli A, Lombardo M, De Filippis S, Saggio G,
145. Choong NW, Kozloff M, Taber D, Hu HS, Wade J 3rd, Ivy P, Fabbri F, et al. Ifosfamide, cisplatin, and 13-Cis retinoic acid for
et al. Phase II study of sunitinib malate in head and neck patients with advanced or recurrent squamous cell carcinoma of
squamous cell carcinoma. Invest New Drugs. 2010;28(5):677 the head and neck: a phase III study. Cancer. 2001;92(4):814
83. doi:10.1007/s10637-009-9296-7. 21.
146. Fountzilas G, Fragkoulidi A, Kalogera-Fountzila A, Nikolaidou 161. Czystowska M, Han J, Szczepanski MJ, Szajnik M, Quadrini K,
M, Bobos M, Calderaro J, et al. A phase II study of sunitinib in Brandwein H, et al. IRX-2, a novel immunotherapeutic, protects
patients with recurrent and/or metastatic non-nasopharyngeal human T cells from tumor-induced cell death. Cell Death Differ.
head and neck cancer. Cancer Chemother Pharmacol. 2009;16(5):70818. doi:10.1038/cdd.2008.197.
2010;65(4):64960. doi:10.1007/s00280-009-1070-1. 162. Wolf GT, Fee WE Jr, Dolan RW, Moyer JS, Kaplan MJ, Spring
147. Machiels JP, Henry S, Zanetta S, Kaminsky MC, Michoux N, PM, et al. Novel neoadjuvant immunotherapy regimen safety
Rommel D, et al. Phase II study of sunitinib in recurrent or and survival in head and neck squamous cell cancer. Head Neck.
metastatic squamous cell carcinoma of the head and neck: 2011;33(12):166674. doi:10.1002/hed.21660.
GORTEC 2006-01. J Clin Oncol. 2010;28(1):218. doi:10.1200/ 163. Baselga J, Pfister D, Cooper MR, Cohen R, Burtness B, Bos M,
JCO.2009.23.8584. et al. Phase I studies of anti-epidermal growth factor receptor
148. Garrity T, Pandit R, Wright MA, Benefield J, Keni S, Young chimeric antibody C225 alone and in combination with cisplatin.
MR. Increased presence of CD34? cells in the peripheral blood J Clin Oncol. 2000;18(4):90414.
of head and neck cancer patients and their differentiation into 164. Ferris RL, Jaffee EM, Ferrone S. Tumor antigen-targeted,
dendritic cells. Int J Cancer. 1997;73(5):6639. monoclonal antibody-based immunotherapy: clinical response,
149. Young MR, Wright MA, Vellody K, Lathers DM. Skewed dif- cellular immunity, and immunoescape. J Clin Oncol.
ferentiation of bone marrow CD34? cells of tumor bearers from 2010;28(28):43909. doi:10.1200/JCO.2009.27.6360.
dendritic toward monocytic cells, and the redirection of differ- 165. Lopez-Albaitero A, Lee SC, Morgan S, Grandis JR, Gooding
entiation toward dendritic cells by 1alpha,25-dihydroxyvitamin WE, Ferrone S, et al. Role of polymorphic Fc gamma receptor
D3. Int J Immunopharmacol. 1999;21(10):67588. IIIa and EGFR expression level in cetuximab mediated, NK cell
150. Ben-Shoshan M, Amir S, Dang DT, Dang LH, Weisman Y, dependent in vitro cytotoxicity of head and neck squamous cell
Mabjeesh NJ. 1alpha,25-Dihydroxyvitamin D3 (Calcitriol) carcinoma cells. Cancer Immunol Immunother. 2009;58(11):
inhibits hypoxia-inducible factor-1/vascular endothelial growth 185364. doi:10.1007/s00262-009-0697-4.

123
68 Immunology & Microbiology in Miami (2013) 57:5269

166. Kurai J, Chikumi H, Hashimoto K, Yamaguchi K, Yamasaki A, 181. Balz V, Scheckenbach K, Gotte K, Bockmuhl U, Petersen I, Bier
Sako T, et al. Antibody-dependent cellular cytotoxicity medi- H. Is the p53 inactivation frequency in squamous cell carcino-
ated by cetuximab against lung cancer cell lines. Clin Cancer mas of the head and neck underestimated? Analysis of p53
Res. 2007;13(5):155261. doi:10.1158/1078-0432.CCR-06- exons 2-11 and human papillomavirus 16/18 E6 transcripts in
1726. 123 unselected tumor specimens. Cancer Res. 2003;63(6):1188
167. Lopez-Albaitero A, Ferris RL. Immune activation by epidermal 91.
growth factor receptor specific monoclonal antibody therapy for 182. Boyle JO, Hakim J, Koch W, van der Riet P, Hruban RH, Roa RA,
head and neck cancer. Arch Otolaryngol Head Neck Surg. et al. The incidence of p53 mutations increases with progression of
2007;133(12):127781. doi:10.1001/archotol.133.12.1277. head and neck cancer. Cancer Res. 1993;53(19):447780.
168. Bonner JA, Harari PM, Giralt J, Azarnia N, Shin DM, Cohen 183. Turksma AW, Braakhuis BJ, Bloemena E, Meijer CJ, Leemans
RB, et al. Radiotherapy plus cetuximab for squamous-cell car- CR, Hooijberg E. Immunotherapy for head and neck cancer
cinoma of the head and neck. N Engl J Med. 2006;354(6):567 patients: shifting the balance. Immunotherapy. 2013;5(1):4961.
78. doi:10.1056/NEJMoa053422. doi:10.2217/imt.12.135.
169. Bonner JA, Harari PM, Giralt J, Cohen RB, Jones CU, Sur RK, 184. Bontkes HJ, Kramer D, Ruizendaal JJ, Meijer CJ, Hooijberg E.
et al. Radiotherapy plus cetuximab for locoregionally advanced Tumor associated antigen and interleukin-12 mRNA transfected
head and neck cancer: 5-year survival data from a phase 3 dendritic cells enhance effector function of natural killer cells
randomised trial, and relation between cetuximab-induced rash and antigen specific T-cells. Clin Immunol. 2008;127(3):375
and survival. Lancet Oncol. 2010;11(1):218. doi:10.1016/ 84. doi:10.1016/j.clim.2008.02.001.
S1470-2045(09)70311-0. 185. Gholamin M, Moaven O, Farshchian M, Mahmoudi M, Sankian
170. da Silva SD, Hier M, Mlynarek A, Kowalski LP, Alaoui-Jamali M, Memar B, et al. Induction of cytotoxic T lymphocytes
MA. Recurrent oral cancer: current and emerging therapeutic primed with tumor RNA-loaded dendritic cells in esophageal
approaches. Front Pharmacol. 2012;3:149. doi:10.3389/fphar. squamous cell carcinoma: preliminary step for DC vaccine
2012.00149. design. BMC Cancer. 2010;10:261. doi:10.1186/1471-2407-10-
171. Russell JS, Colevas AD. The use of epidermal growth factor 261.
receptor monoclonal antibodies in squamous cell carcinoma of 186. Chikamatsu K, Albers A, Stanson J, Kwok WW, Appella E,
the head and neck. Chemother Res Pract. 2012;2012:761518. Whiteside TL, et al. P53(110124)-specific human CD4? T-
doi:10.1155/2012/761518. helper cells enhance in vitro generation and antitumor function of
172. Borjesson PK, Postema EJ, Roos JC, Colnot DR, Marres HA, tumor-reactive CD8? T cells. Cancer Res. 2003;63(13):367581.
van Schie MH, et al. Phase I therapy study with (186)Re-labeled 187. Andrade P, Deleo A, Visus C, Butterfield L, Argiris A, Ferris
humanized monoclonal antibody BIWA 4 (bivatuzumab) in RL. Phase I adjuvant trial of multi-epitope p53 vaccine for
patients with head and neck squamous cell carcinoma. Clin patients with squamous cell carcinoma of the head and neck. J
Cancer Res. 2003;9(10 Pt 2):3961S72S. Clin Oncol. 2009;27:15s.
173. Goldenberg DM, Sharkey RM. Using antibodies to target cancer 188. Voskens CJ, Strome SE, Sewell DA. Synthetic peptide-based
therapeutics. Expert Opin Biol Ther. 2012;12(9):117390. cancer vaccines: lessons learned and hurdles to overcome. Curr
doi:10.1517/14712598.2012.693472. Mol Med. 2009;9(6):68393.
174. To WC, Wood BG, Krauss JC, Strome M, Esclamado RM, 189. Voskens CJ, Sewell D, Hertzano R, DeSanto J, Rollins S, Lee
Lavertu P, et al. Systemic adoptive T-cell immunotherapy in M, et al. inducTION of mage-A3 and HPV-16 immunity by
recurrent and metastatic carcinoma of the head and neck: a Trojan vaccines in patients with head and neck carcinoma. Head
phase 1 study. Arch Otolaryngol Head Neck Surg. 2000;126(10): Neck. 2012;34(12):173446. doi:10.1002/hed.22004.
122531. 190. Hu JC, Coffin RS, Davis CJ, Graham NJ, Groves N, Guest PJ,
175. Riechelmann H, Wiesneth M, Schauwecker P, Reinhardt P, et al. A phase I study of OncoVEXGM-CSF, a second-genera-
Gronau S, Schmitt A, et al. Adoptive therapy of head and neck tion oncolytic herpes simplex virus expressing granulocyte
squamous cell carcinoma with antibody coated immune cells: a macrophage colony-stimulating factor. Clin Cancer Res.
pilot clinical trial. Cancer Immunol Immunother. 2007;56(9): 2006;12(22):673747. doi:10.1158/1078-0432.CCR-06-0759.
1397406. doi:10.1007/s00262-007-0283-6. 191. Coffin R, Hingorani M, McNeish I, Sibtain A, Hamilton B, Love
176. Yamasaki K, Horiguchi S, Kurosaki M, Kunii N, Nagato K, C, et al. Phase I/II trial of OncoVEXGM-CSF combined with
Hanaoka H, et al. Induction of NKT cell-specific immune radical chemoradiation (CRT) in patients with newly diagnosed
responses in cancer tissues after NKT cell-targeted adoptive node-positive stage III/IV head and neck cancer (HNC). J Clin
immunotherapy. Clin Immunol. 2011;138(3):25565. doi:10. Oncol. 2007;25(18S):14095.
1016/j.clim.2010.11.014. 192. Harrington KJ, Hingorani M, Tanay MA, Hickey J, Bhide SA,
177. Dudley ME, Rosenberg SA. Adoptive-cell-transfer therapy for Clarke PM, et al. Phase I/II study of oncolytic HSV GM-CSF in
the treatment of patients with cancer. Nat Rev Cancer. combination with radiotherapy and cisplatin in untreated stage
2003;3(9):66675. doi:10.1038/nrc1167. III/IV squamous cell cancer of the head and neck. Clin Cancer
178. Govers C, Sebestyen Z, Coccoris M, Willemsen RA, Debets R. Res. 2010;16(15):400515. doi:10.1158/1078-0432.CCR-10-
T cell receptor gene therapy: strategies for optimizing transgenic 0196.
TCR pairing. Trends Mol Med. 2010;16(2):7787. doi:10.1016/ 193. Ma B, Yijie X, Hung C-F, Wu T-C. HPV and therapeutic vac-
j.molmed.2009.12.004. cines: where we stand in 2010. Curr Cancer Ther Rev.
179. Jorritsma A, Schotte R, Coccoris M, de Witte MA, Schumacher 2010;6:81103.
TN. Prospects and limitations of T cell receptor gene therapy. 194. Best SR, Niparko KJ, Pai SI. Biology of human papillomavirus
Curr Gene Ther. 2011;11(4):27687. infection and immune therapy for HPV-related head and neck
180. Cohen CJ, Zheng Z, Bray R, Zhao Y, Sherman LA, Rosenberg cancers. Otolaryngol Clin North Am. 2012;45(4):80722.
SA, et al. Recognition of fresh human tumor by human doi:10.1016/j.otc.2012.04.005.
peripheral blood lymphocytes transduced with a bicistronic 195. Wallecha A, French C, Petit R, Singh R, Amin A, Rothman J.
retroviral vector encoding a murine anti-p53 TCR. J Immunol. Lm-LLO-based immunotherapies and HPV-associated disease. J
2005;175(9):5799808. Oncol. 2012;2012:542851. doi:10.1155/2012/542851.

123
Immunology & Microbiology in Miami (2013) 57:5269 69

196. Gunn GR, Zubair A, Peters C, Pan ZK, Wu TC, Paterson Y. study of Lm-LLO-E7 in patients with advanced carcinoma of
Two Listeria monocytogenes vaccine vectors that express dif- the cervix. Vaccine. 2009;27(30):397583. doi:10.1016/j.
ferent molecular forms of human papilloma virus-16 (HPV-16) vaccine.2009.04.041.
E7 induce qualitatively different T cell immunity that correlates 198. Ma B, Maraj B, Tran NP, Knoff J, Chen A, Alvarez RD, et al.
with their ability to induce regression of established tumors Emerging human papillomavirus vaccines. Expert Opin Emerg
immortalized by HPV-16. J Immunol. 2001;167(11):64719. Drugs. 2012;17(4):46992. doi:10.1517/14728214.2012.744393.
197. Maciag PC, Radulovic S, Rothman J. The first clinical use of a
live-attenuated Listeria monocytogenes vaccine: a Phase I safety

123

You might also like