Você está na página 1de 25

Accepted Manuscript

Nasal polyposis pathophysiology: Endotype and phenotype open


issues

Giuseppe Brescia, Claudia Zanotti, Daniela Parrino, Umberto


Barion, Gino Marioni

PII: S0196-0709(18)30178-9
DOI: doi:10.1016/j.amjoto.2018.03.020
Reference: YAJOT 1996
To appear in:
Received date: 22 February 2018

Please cite this article as: Giuseppe Brescia, Claudia Zanotti, Daniela Parrino, Umberto
Barion, Gino Marioni , Nasal polyposis pathophysiology: Endotype and phenotype open
issues. The address for the corresponding author was captured as affiliation for all authors.
Please check if appropriate. Yajot(2017), doi:10.1016/j.amjoto.2018.03.020

This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. Please note that during the production process errors may
be discovered which could affect the content, and all legal disclaimers that apply to the
journal pertain.
ACCEPTED MANUSCRIPT
1

Nasal polyposis pathophysiology:

Endotype and phenotype open issues.

Giuseppe Brescia, MDa*;

Claudia Zanotti, MDa; Daniela Parrino, MDa; Umberto Barion, MDa;

Gino Marioni, MDa.

PT
a
Department of Neurosciences DNS, Otolaryngology Section, Padova University,

RI
Padova, Italy.

SC
Running title: Endotyping CRSwNP

Word count: 3051


NU
Number of figures:0

Number of tables:0
MA

A preliminary version of this review was presented at the 63rd Meeting of “Alta
D

Italia” Group of Otolaryngology, Padova (Italy), December 2, 2017.


E
PT

Corresponding author
*
Giuseppe Brescia, MD; Department of Neurosciences DNS, Otolaryngology
CE

Section, Padova University, Via Giustiniani 2, 35128 Padova, Italy, fax

+39 049 8213113, e-mail giuseppe.brescia@aopd.veneto.it


AC
ACCEPTED MANUSCRIPT
2

ABSTRACT

Purpose: Endotyping chronic rhinosinusitis with nasal polyps

(CRSwNP) poses a challenge for rhinologists nowadays. Phenotyping

CRSwNP proved inappropriate as an approach to their classification

because of their common clinical features. Endotyping, being

PT
based on the pathogenic mechanism, provides a precise picture more

RI
appropriate for use in clinical practice. Patients’ treatment and

SC
follow-up can thus be tailored to cope with the degree of

aggressiveness of a specific CRSwNP endotype.


NU
The aim of this study was to analyze the available

information about the main currently accepted endotypes of CRSwNP;


MA

furthermore, we reported and commented evidence regarding some

clinical conditions associated with nasal polyposis which could be


D

related with new endotypes.


E

Materials and Methods: Pubmed and Scopus electronic database were


PT

searched. The main available studies about CRSwNP endotyping


CE

published predominantly in the last 5 years were critically

analyzed.
AC

Results: The pathophysiological features of some asthma-related

CRSwNP (allergic fungal rhinosinusitis, aspirin-exacerbated

respiratory disease) are quite well understood, including them

among known endotypes of CRSwNP. On the other hand, because of

their known pathophysiological mechanisms, some well-known

diseases associated with aggressive forms of CRSwNP, such as


ACCEPTED MANUSCRIPT
3

eosinophilic granulomatosis with polyangiitis, primary ciliary

dyskinesia and cystic fibrosis, should be investigated as

potentially related with CRSwNP endotypes.

Conclusions: CRSwNP comprises several inflammatory endotypes

defined by different pathogenic mechanisms. These endotypes

correlate with the disease’s clinical manifestations and behavior.

PT
A thorough understanding of CRSwNP endotypes will enable targeted

RI
medical therapies and tailored follow-up protocols.

SC
Key words: Chronic rhinosinusitis; nasal polyps; endotypes;
NU
aspirin-exacerbated respiratory disease; allergic fungal

rhinosinusitis; eosinophilic granulomatosis with polyangiitis.


MA
E D
PT
CE
AC
ACCEPTED MANUSCRIPT
4

1. Introduction

Chronic rhinosinusitis (CRS) is commonly divided into two

phenotype-based groups on the presence (CRSwNP) or absence of

nasal polyps (CRSsNP), which are clinically diagnosed by nasal

endoscopy and/or computed tomography (CT) of the paranasal

sinuses. It is clear that this classification is over simple,

PT
since the same patients may or may not have nasal polyps at

RI
different times in their clinical history, even after appropriate

SC
medical or surgical treatment.

A different, more comprehensive approach is therefore needed


NU
to classify CRS more accurately. There is evidence to suggest that

some forms of nasal polyposis, associated with certain conditions


MA

(e.g. allergy, asthma, aspirin sensitivity), have particular

characteristics: local aggressiveness, a weak response to drugs,


D

and high recurrence rates after surgical treatment. Such forms are
E

quite homogeneous from the clinical viewpoint, and have been


PT

called "phenotypes", but this classification also reveals some


CE

weaknesses in clinical practice [1, 2]. In 2008, Anderson [3]

observed that intrinsic bronchial asthma (which could be


AC

considered as an "unicum" in terms of its clinical profile)

differed between young adult and elderly patients in terms of

severity and response to therapy. Surmising that the two forms of

asthma had a different pathogenesis, Anderson introduced the

concept of "endotype". Endotyping is based on the fact that some

diseases have the same pathogenic mechanism even though their


ACCEPTED MANUSCRIPT
5

etiology and final clinical features may differ.

The endotyping approach prompted the idea that some forms of

disease sharing the same mechanism of action could be treated with

targeted therapies to interrupt their progression. Investigations

revealed that some elements were capable of triggering a common

inflammatory mechanism [4, 5]. For example, bacteria, fungi,

PT
viruses, biofilms, and proteins are all able to activate T-helper

RI
(TH) cells, which shift from a quiescent native state (TH naive),

SC
through an intermediate T-helper Ø (THØ) stage, to reach a final T-

helper specialization stage as type 1, 2 and 17 (TH1, TH2, TH17).


NU
These activated lymphocytes produce a spectrum of substances

called interleukins (ILs). They can regulate the activity of other


MA

blood cells in the immune system, especially eosinophils and

basophils, giving rise to an inflammatory microenvironment. The


D

cells attracted to a site of inflammation are capable of producing


E

other cytokines that can enrich the microenvironment and


PT

facilitate the inflammatory process. Among these cytokines, at the


CE

endothelial level there are chemokines (eotaxins 1, 2 and 3) that

are able to activate and recruit eosinophils, basophils, and TH


AC

lymphocytes at the inflammatory site. Adhesins (intercellular

adhesion molecule-1, and vascular cell adhesion molecule-1) make

cells adhere to vessels and tissues, and cause cellular

transmigration through the endothelium. Then there are the metal

proteinases 2 and 9 which locally degrade the extracellular matrix

by allowing inflammatory cells to pass through and spread into the


ACCEPTED MANUSCRIPT
6

nasal polyp tissue. The final stage leads to the synthesis of DNA

transcription factors, such as G-A-T-A binding core sequence

transcription factors, or signal transducers and transcription

activators. These proteins produced by inflammatory cells migrate

into the cell nucleus, binding to specific DNA sites, and blocking

or triggering the action of RNA polymerase and RNA synthesis,

PT
which results in an altered cell turnover. For example, action on

RI
eosinophils has the effect of inhibiting apoptosis, leading to an

SC
exponential increase in eosinophils in nasal polyps.

It is now believed that T helper cells are compartmentalized:


NU
interleukins 4, 5 and 13 are produced, and eosinophils are

recruited in the TH2 environment, while interleukins 17 and 22 are


MA

mainly produced, and neutrophils are recruited in the TH1 and TH17

environments. It has also been suggested, however, that multiple


D

cell lines can coexist at the same site of inflammation.


E

We are still unable to classify all CRS endotypes. Some


PT

endotypes have been understood and associated with a clearly-


CE

defined form of CRS thanks to a close correspondence between

endotype and phenotype. Different forms of CRS sometimes seem to


AC

share the same pathogenesis (i.e. there may be several phenotypes

with the same endotype). Finally, as Anderson [3] suggested for

asthma, if the same form of CRS shows different clinical

characteristics (e.g. a phenotype with different degrees of

aggressiveness), this may be due to various pathogenic mechanisms

(different endotypes with the same phenotype). These complexities


ACCEPTED MANUSCRIPT
7

can only be clarified by means of a significant improvement in our

understanding of the pathogenic mechanisms involved.

The need for identifying individual phenotypes and endotypes

extends to the approaches that currently considered the mainstay

of treatment of CRS [6]. In particular, the endotypes

classificative approach has to focus on defining CRS pathogenic

PT
mechanisms rather than describing the inflammatory pattern of CRS

RI
even in an advanced biological way. The aim of this study was to

SC
analyze the available information about the main currently

accepted endotypes of CRSwNP [6, 7]; furthermore, we reported and


NU
commented evidence regarding some clinical conditions associated

with nasal polyposis which could be related with new endotypes.


MA
D

2. CRSwNP endotypes
E

In 2013, a preliminary attempt to classify CRSwNP endotypes was


PT

performed by Akdis et al [1]. Different CRSwNP endotypes can be


CE

characterized by differences in responsiveness to different

treatments, including topical intranasal corticosteroids and


AC

biological agents, such as anti–IL5 and anti-IgE antibody, and can

be based on different biomarkers that are linked to underlying

mechanisms. In 2016, Dennis et al. [2] proposed four distinct, but

partially overlapping, classification modalities for identifying

endotypes within the CRSwNP phenotype: 1) type 2 cytokine based

approach, 2) eosinophil-mediated approach, 3) immunoglobulin IgE-


ACCEPTED MANUSCRIPT
8

based approach, and 4) cysteinyl leukotriene (CysLT)-based

approach. In the same year, however, a multicentric investigation

by Tomassen et al. [8] proposed a classification for clusters

based on tissue analysis of IL-5, IFN-γ, IL-17A, TNF-α, IL-22, IL-

1β, IL-6, IL-8, cationic eosinophilic protein, myeloperoxidase,

TGF-β1, IgE, Staphylococcus aureus IgE specific for enterotoxin

PT
and albumin in CRS patients. The clustering of 173 CRS cases had

RI
resulted in 10 groups, of which 4 clusters with low or

SC
undetectable IL-5, eosinophilic cationic protein, IgE and albumin

concentrations and 6 clusters with high concentrations of such


NU
markers. The aim was to identify inflammatory endotypes of CRS

through biomarkers’ clusters analysis. Nevertheless the current


MA

difficulties in classifying CRSwNP into endotypes (one phenotype

could include several molecular mechanisms and one endotype could


D

include several phenotypes), very recently, Kim et al. [5]


E

concluded that endotypes depended on the epithelial barrier


PT

function, epithelial cytokines, and T cell subsets.


CE

2.1 CRSwNP and inflammatory cells


AC

As mentioned previously, the first step in the inflammatory

process in patients with CRSwNP involves nasal mucosa colonization

by immune system cells. The subsequent recruitment of other immune

system cells and the formation of the inflammatory

microenvironment give rise to a chronic disorder. Some cells

involved in this process are neutrophils and eosinophils [4, 5]


ACCEPTED MANUSCRIPT
9

through a process that involves basophils, type 2 innate lymphoid

cells, and type 2 helper lymphocytes as promoters [9, 10].

CRSwNP shows a great cytological variety. A first difference has

to do with geographical/racial distributions. In Western

countries, cases of eosinophilic CRSwNP are prevalent, sustained

by TH2 inflammation, whereas in Asia, eosinophilic and non-

PT
eosinophilic CRSwNP histotypes are equally represented [4]. In

RI
Western countries, tissue eosinophilia staging in cases of CRSwNP

SC
has now become sufficiently standardized: after staining with

hematoxylin & eosin, three microscopic high-power fields (HPF)


NU
(400x magnification) are examined. The number of eosinophils is

counted for each field, and the average for the three fields is
MA

calculated. Values ≥10 eosinophils/HPF characterize eosinophilic

CRSwNP, and values <10/HPF non-eosinophilic CRSwNP. In the Asian


D

rhinological literature, the cut-off is less clearly defined, and


E

can vary from 5 to more than 100 eosinophils/HPF. This variability


PT

may depend also on the greater heterogeneity of the T-helper cell


CE

lines; TH1, TH2 and TH17 environments coexist in the same patient,

and thus give rise to different cell lines [4].


AC

The eosinophilic histotype has been extensively studied and

found associated with the prognosis for CRSwNP [11-15]. Generally

speaking, eosinophilic CRSwNP are associated with higher

recurrence rates and shorter disease-free intervals after

treatment than non-eosinophilic or neutrophilic forms. Another

histotype, the basophilic one, has recently become the object of


ACCEPTED MANUSCRIPT
10

preliminary investigations [16] showing that basophils have a

common pathway with eosinophils, and are represented at the

inflammatory site, correlating strongly with eosinophilia and

prognosis.

Brescia et al. [17] recently focused on the role of

eosinophilia and basophilia in CRSwNP patients’ blood. Tissue and

PT
blood eosinophilia were closely related in eosinophilic CRSwNP

RI
(including cases associated with asthma and allergy), and in

SC
aspirin-exacerbated respiratory disease. Blood basophilia also

correlated closely with blood eosinophilia in this form of CRSwNP.


NU
A role for blood sampling of these cells to predict the course of

CRSwNP was consequently hypothesized. Basophil and eosinophil


MA

levels in the blood were also found to vary significantly between

different endotypes and phenotypes. Some authors [16, 18-20] have


D

suggested considering blood and/or tissue basophil and eosinophil


E

levels for the purpose of CRSwNP endotyping.


PT
CE

2.2 Aspirin-exacerbated respiratory disease (AERD)

AERD has a known pathogenesis and is associated with severe asthma


AC

and recurrent nasal polyposis; it can be counted among CRSwNP

endotypes [5, 7].

AERD usually occurs in the third and fourth decades of life,

most frequently in males (M/F 2.3/1). It is characterized by a

reaction to aspirin or other non-steroidal anti-inflammatory drugs

(NSAIDs) that inhibit cyclooxygenase-1 (COX-1). The clinical


ACCEPTED MANUSCRIPT
11

features of AERD include asthma, eosinophilic nasal polyps, and

blood eosinophilia (often without atopia, but with intolerance to

alcoholic beverages) [2, 21].

Eosinophilic type CRSwNP accounts for 8.7% to 9.7% of all

CRSwNP patients [2, 21-23]. Asthma develops after the onset of

CRSwNP in 14.89% of all patients with severe asthma [22]. Not all

PT
AERD patients develop asthma, however (though they may suffer from

RI
abdominal and dermatological disorders as well as CRSwNP), which

SC
is why the term AERD is preferable to "aspirin-intolerant asthma".

From a pathophysiological standpoint, AERD has been


NU
associated with enzymatic defects in eicosanoid metabolism,

including a functional reduction of COX-1 enzymes with a decrease


MA

in prostaglandin levels, and an increase in 5-lipoxygenase

activity and cysteinyl-leukotriene (CysLT) levels, especially


D

leukotriene C4 [2]. High CysLT levels are related to the downstream


E

activation of effector cells, such as eosinophils, basophils and


PT

mast cells, which stimulate the inflammatory response within the


CE

sinonasal and respiratory mucosa. Other cytokines comprising the

inflammatory milieu of AERD include IL‐4, IL‐33, and interferon‐γ,


AC

which goes to show the mechanistic overlap between AERD and CRSwNP

in aspirin‐tolerant patients [2, 24].

AERD treatment begins with the avoidance of aspirin and

NSAIDs. Steroid treatments are administered systemically and

topically (inhalations). Anti-leukotriene therapy is indicated and

has recently been implemented using monoclonal antibodies capable


ACCEPTED MANUSCRIPT
12

of blocking IL-4, IL-5 and IL-13 [21]. Endoscopic sinus surgery

(ESS) for AERD offers symptomatic benefits and enables a more

limited use of systemic steroid therapy. ESS plus steroids

significantly improves patients’ asthma symptoms by reducing their

systemic inflammatory state [18]. If medical and surgical

treatments for AERD yield poor results, then aspirin

PT
desensitization should be considered [21].

RI
SC
2.3 Allergic fungal rhinosinusitis (AFRS)

Considering the classification of Dennis et al. [2], and in


NU
particular their IgE-based approach for endotyping CRSwNP

hypothesis, local IgE may be a stronger pathophysiological driver


MA

than systemic IgE; AFRS should also be counted among emerging

endotypes [5].
D

AFRS accounts for 5-10% of all cases of CRSwNP in


E

immunocompetent patients. It is caused mainly by Aspergillus,


PT

Bipolaris, Curvularia and Alternaria species colonizing the


CE

paranasal sinuses, and impairing mucociliary clearance [25]. In

1994, Bent and Kuhn [26] diagnosed AFRS in patients with type I
AC

hypersensitivity, nasal polyps, and eosinophilic mucin found at

the time of surgery and on CT scan. Asthma, positive fungal

staining or Charcot-Leyden crystals in surgical specimens, and

serum eosinophilia could also be found. These criteria are still

valid today, but it is important to emphasize that the presence of

fungi in the nasal sinus and serum IgE cannot always be confirmed.
ACCEPTED MANUSCRIPT
13

IgE are only found locally in some cases. Asthma reportedly

develops in 50% of AFRS patients, and becomes severe in 5-10% of

asthmatics. The Alternaria species has been associated with life-

threatening acute asthma attacks and asthma-related deaths [25].

In 2004, Collins et al. [27] concluded that AFRS was the

result of a local, not systemic, hypersensitivity reaction type I.

PT
Their conclusion was based on having found specific IgE in the

RI
mucin of AFRS patients. Their investigation underscored the role

SC
of an IgE-mediated hypersensitivity reaction, explaining why not

all AFRS patients had signs of systemic allergy. High levels of


NU
local and systemic IgE have been found in all forms of CRSwNP,

with the exception of AERD, and local IgE can be even more
MA

powerful as a driver of disease pathophysiology than systemic IgE

[2]. The most accredited hypothesis is that local sinonasal IgE


D

production contributes to the development of nasal polyps,


E

particularly in association with asthma [2]. Given the high


PT

mucosal IgE concentrations in nasal polyposis, using high-affinity


CE

IgE-blocking monoclonal antibodies may be appropriate as a

targeted treatment [2].


AC

More recently, other authors [25, 28] have suggested that

different immunological reactions against fungal antigens might

coexist, including: (i) type I hypersensitivity to fungal IgE;

(ii) type III hypersensitivity, with IgG forming a complex with

fungal antigens; and (iii) type IV hypersensitivity, with T-cell–

mediated eosinophilic inflammation. All of these inflammatory


ACCEPTED MANUSCRIPT
14

reactions interfere with mucus drainage from the nasal sinus,

subsequently causing fungal growth and the maintenance of

inflammation.

AFRS is usually treated surgically to remove the polyps and

eosinophilic mucin, clear the affected sinuses, and create an

access route for topical intranasal medication. If the sinuses are

PT
not cleared completely, the inflammatory process usually

RI
continues, and resistance to anti-inflammatory drugs develops.

SC
Patients should undergo nasal irrigations and take intranasal

steroids to reduce the inflammation and enable sinus drainage


NU
[25]. Immunotherapy can cure the allergic diseases, relieve

symptoms, and reduce the need for medication. Fungal extracts are
MA

not standardized, however, and immunotherapy is not recommended

for patients with asthma because it quite frequently prompts


D

adverse reactions [25]. Oral steroids are still recommended for


E

AFRS [29].
PT
CE

3. Potential new CRSwNP endotypes

Some diseases have very clear phenotypes associated with nasal


AC

polyps and are caused by well-known pathophysiological mechanisms.

Eosinophilic granulomatosis with polyangiitis (EGPA) -

formerly known as Churg-Strauss syndrome, primary ciliary

dyskinesia (PCD) [30] - also known as Kartagener syndrome, and

cystic fibrosis (CF) are considered phenotypes associated with CRS

that have a distinctive clinical profile and behavior.


ACCEPTED MANUSCRIPT
15

EGPA is a systemic vasculitis characterized by extravascular

granulomas, blood eosinophilia and eosinophilic tissue

infiltration. Clinically, it begins with allergic rhinitis, nasal

polyps, and asthma. With time, EGPA features blood and tissue

eosinophilia, chronic eosinophilic pneumonia, and/or eosinophilic

gastroenteritis. In advanced stages, a potentially life-

PT
threatening vasculitis involving several organs may be diagnosed.

RI
The diagnosis of EGPA is based on the presence of 4 or more

SC
American College of Rheumatology criteria: asthma, >10%

eosinophilia in blood, rhinosinusitis, pulmonary infiltrates


NU
(sometimes transient), histological evidence of vasculitis with

extravascular eosinophils, and mono- or poly-neuropathy [31]. The


MA

presence of CRS in more than 80% of patients with early EGPA

enables rhinologists to diagnose the disease at an early stage,


D

and prompt appropriate therapy can prevent it from progressing


E

further [32]. The known TH2 pathogenic mechanism seems to be


PT

involved, which triggers and maintains eosinophilic inflammation.


CE

In the setting of EGPA, eosinophilic polyposis is extremely

aggressive and resistant to common steroid treatments. Recurrences


AC

are frequent after endoscopic surgery, and immunosuppressants

should be added to steroids in some cases.

In 2015, Stevens et al. [33] said that almost all patients

with CF or PCD develop CRS, and it often proves more recalcitrant

to therapy in these patients than in cases of idiopathic CRS.

Patients with CF or PCD are predisposed to CRS because of a


ACCEPTED MANUSCRIPT
16

defective mucociliary clearance (MCC), which allows bacterial

colonization of the sinus. That is why CRSwNP in CF and PCD is

usually characterized by a neutrophilic histotype. The upper

airways play an important part in removing particulates and

pathogens from inspired air by means of MCC, a specialized

function unique to the airway epithelium. In CF, defects in the

PT
transmembrane conductance regulator anion channel result in an

RI
impaired salt and water secretion, and possibly also an enhanced

SC
absorption, causing mucus dehydration and impairing MCC [33]. CF

patients frequently develop severe recurrent sinonasal infections


NU
that may also seed or exacerbate lung infections. There are also

higher than normal levels of the epithelial anion transporter,


MA

pendrin, in nasal polyps than in uncinate tissue isolated from

healthy controls. An upregulated pendrin expression in the airway


D

has been linked to IL-4, IL-13, and IL-17A [34, 35]. Defects
E

involving the epithelial cell cilia can also affect MCC and
PT

contribute to chronic sinonasal inflammation. In PCD, for example,


CE

an abnormal ciliary function and/or structure result in an

impaired MCC and a higher incidence of upper respiratory


AC

infections. More commonly, though, ciliary dysfunctions are

acquired through exposure to environmental or microbial toxins,

and/or as a secondary consequence of disease via exposure to

inflammatory stimuli. In both CF and PCD, the pathogenic picture

seems to involve the known TH1 mechanism that triggers and

maintains neutrophilic inflammation [35].


ACCEPTED MANUSCRIPT
17

4. Conclusion

Current CRS phenotyping is of limited value because common

clinical manifestations seem to overlap in different forms of CRS.

This heterogeneity of behavior also collides with a homogeneous

approach to therapy, based mainly on the use of steroids and

PT
endoscopic sinus surgery, with variable results in terms of

RI
recurrence rates. Hence the need to endotype CRS in order to

SC
classify the variants of this disease no longer from the clinical

standpoint, but according to the pathogenic mechanisms behind it.


NU
For some CRSwNP, the pathogenic mechanism seems to be fairly

clear, and they take on the role of endotypes [5, 7]. Because of
MA

their quite well-known pathophysiological mechanism, several

phenotypes, associated with some of the more aggressive forms of


D

CRSwNP, such as EGPA, CF and PCD, may however be considered as


E

promising potential CRSwNP endotypes to investigate.


PT

Another challenge for modern rhinologists lies in correlating


CE

histotypes (eosinophils, basophils, neutrophils) with cytokines in

the local inflammatory microenvironment, and tissue DNA


AC

modifications. A growing interest in inflammatory blood cells

(eosinophils, basophils) has recently emerged: these cells seem to

correlate with histotypes, comorbidities (asthma, allergy),

endotyping, and CRS recurrence rates. It remains to be seen

whether this systemic phenomenon is a consequence of the local

inflammatory state, or whether local inflammation is an expression


ACCEPTED MANUSCRIPT
18

of a systemic disease. The main goal of this field of

investigation is to identify specific, not generic mechanisms

behind nasal polyposis in order to tailor endotype-based therapies

and thereby achieve significantly lower disease recurrence rates.

PT
RI
SC
Funding

This study was partly supported by grant No. DOR 1658072/16


NU
(G. Marioni) from the University of Padova, Italy.
MA

Declarations of Interest

None
D

Informed consent
E

Not applicable.
PT
CE

Aknowledgements

The authors thank Frances Coburn for correcting the English


AC

version of this paper.


ACCEPTED MANUSCRIPT
19

References

[1] Akdis CA, Bachert C, Cingi C, Dykewicz MS, Hellings

PW, Naclerio RM, et al. Endotypes and phenotypes of

chronic rhinosinusitis: a PRACTALL document of the European

Academy of Allergy and Clinical Immunology and the American

PT
Academy of Allergy, Asthma & Immunology.J Allergy Clin

Immunol 2013;131:1479-90.

RI
[2] Dennis SK, Lam K, Luong A. A Review of Classification

SC
Schemes for Chronic Rhinosinusitis with Nasal Polyposis

Endotypes. Laryngoscope Investig Otolaryngol 2016;1:130–4.


NU
[3] Anderson GP. Endotyping asthma: new insights into key
MA

pathogenic mechanisms in a complex, heterogeneous disease.

Lancet 2008;372:1107-19.
D

[4] Wang X, Zhang N, Bo M, Holtappels G, Zheng M, Lou H, et


E

al. Diversity of TH cytokine profiles in patients with


PT

chronic rhinosinusitis: A multicenter study in Europe,

Asia, and Oceania. J Allergy Clin Immunol 2016;138:1344-53.


CE

[5] Kim DW and Cho SH. Emerging Endotypes of Chronic


AC

Rhinosinusitis and Its Application to Precision Medicine.

Allergy Asthma Immunol Res 2017;9:299-306.

[6] Gurrola J 2nd, Borish L. Chronic rhinosinusitis: Endotypes,

biomarkers, and treatment response. J Allergy Clin Immunol

2017;140:1499-1508.

[7] Stevens WW, Schleimer RP. Aspirin-Exacerbated Respiratory


ACCEPTED MANUSCRIPT
20

Disease as an Endotype of Chronic Rhinosinusitis. Immunol

Allergy Clin North Am 2017;36:669-680.

[8] Tomassen P, Vandeplas G, Van Zele T, Cardell LO, Arebro J,

Olze H, et al. Inflammatory endotypes of chronic

rhinosinusitis based on cluster analysis of biomarkers. J

Allergy Clin Immunol 2016;137:1449-1456.

PT
[9] Poposki JA, Klingler AI, Tan BK, Soroosh P, Banie H, Lewis

RI
G, et al. Group 2 innate lymphoid cells are elevated and

SC
activated in chronic rhinosinusitis with nasal polyps.

Immun Inflamm Dis 2017;5:233-243


NU
[10] Yamanishi Y, Karasuyama H. Basophil-derived IL-4 plays

versatile roles in immunity. Semin Immunopathol


MA

2016:38:615-22.

[11] Gitomer SA, Fountain CR, Kingdom TT, Getz AE, Sillau
D

SH, Katial RK, et al. Clinical Examination of Tissue


E

Eosinophilia in Patients with Chronic Rhinosinusitis and


PT

Nasal Polyposis. Otolaryngol Head Neck Surg 2016;155:173-8.


CE

[12] Tosun F, Arslan HH, Karslioglu Y, Deveci MS, Durmaz A.

Relationship between postoperative recurrence rate and


AC

eosinophil density of nasal polyps. Ann Otol Rhinol

Laryngol 2010;119:455–9.

[13] Brescia G, Marioni G, Franchella S, Ramacciotti

G, Velardita C, Giacomelli L, et al. Can a panel of

clinical, laboratory, and pathological variables pinpoint

patients with sinonasal polyposis at higher risk of


ACCEPTED MANUSCRIPT
21

recurrence after surgery? Am J Otolaryngol 2010;36:554-8.

[14] Brescia G, Marioni G, Franchella S, Ramacciotti

G, Giacomelli L, Marino F, et al. A prospective

investigation of predictive parameters for post-surgical

recurrences in sinonasal polyposis. Eur Arch

Otorhinolaryngol 2016;273:655-60.

PT
[15] Brescia G, Pedruzzi B, Barion U, Cinetto F, Giacomelli

RI
L, Martini A, et al. Are neutrophil-, eosinophil-, and

SC
basophil-to-lymphocyte ratios useful markers for

pinpointing patients at higher risk of recurrent sinonasal


NU
polyps? Am J Otolaryngol 2016;37:339-45.

[16] Mahdavinia M, Carter RG, Ocampo CJ, Stevens W, Kato A, Tan


MA

BK, et al. Basophils are elevated in nasal polyps of

patients with chronic rhinosinusitis without aspirin


D

sensitivity. J Allergy Clin Immunol 2014;133:1759-63.


E

[17] Brescia G, Barion U, Zanotti C, Giacomelli L, Martini


PT

A, Marioni G. The prognostic role of serum eosinophil and


CE

basophil levels in sinonasal polyposis. Int Forum Allergy

Rhinol 2017;7:261-67.
AC

[18] Brescia G, Barion U, Zanotti C, Parrino D, Marioni G. Pre-

and postoperative blood neutrophil- and eosinophil-to-

lymphocyte ratios in patients with sinonasal polyps. A

preliminary investigation. Allergy and Asthma Proceedings

2017;38:64-69.

[19] Brescia G, Barion U, Zanotti C, Cinetto F, Giacomelli


ACCEPTED MANUSCRIPT
22

L, Martini A. Blood eosinophil-to-basophil ratio in

patients with sinonasal polyps: Does it have a clinical

role? Ann Allergy Asthma Immunol2017;119:223-226.

[20] Marioni G, Zanotti C, Brescia G. Chronic rhinosinusitis

with nasal polyps in the elderly: assessing current

evidence. Allergy and Asthma Proceedings 2018;39:9-13.

PT
[21] Kennedy JL, Stoner AN, Borish L. Aspirin-exacerbated

RI
respiratory disease: Prevalence, diagnosis, treatment, and

SC
considerations for the future. Am J Rhinol Allergy

2016;30:407–13.
NU
[22] Rajan JP, Wineinger NE, Stevenson DD, White AA. Prevalence

of aspirin-exacerbated respiratory disease among asthmatic


MA

patients: A meta-analysis of the literature. J Allergy Clin

Immunol 2015;135:676–81.
D

[23] Orlandi RR, Kingdom TT, Hwang PH. International Consensus


E

Statement on Allergy and Rhinology: Rhinosinusitis


PT

Executive Summary. Int Forum Allergy Rhinol 2016;6:S3–21.


CE

[24] Steinke JW and Wilson JM. Aspirin-exacerbated respiratory

disease: pathophysiological insights and clinical advances.


AC

J Asthma Allergy 2016;9:37–43.

[25] Rick EM, Woolnough K, Pashley CH, Wardlaw AJ. Allergic

Fungal Airway Disease. J Investig Allergol Clin Immunol

2016;26:344-54.

[26] Bent JP 3rd, Kuhn FA. Diagnosis of allergic fungal

sinusitis. Otolaryngol Head Neck Surg 1994;111:580-8.


ACCEPTED MANUSCRIPT
23

[27] Collins M, Nair S, Smith W, Kette F, Gillis D, Wormald PJ.

Role of local immunoglobulin E production in the

pathophysiology of noninvasive fungal sinusitis.

Laryngoscope 2004;114:1242-6.

[28] Doellman MS, Dion GR, Weitzel EK, Reyes EG. Immunotherapy

in allergic fungal sinusitis: The controversy continues. A

PT
recent review of literature. Allergy Rhinol

RI
(Providence)2013;4:e32-5.

SC
[29] Patadia MO, Welch KC. Role of immunotherapy in allergic

fungal rhinosinusitis. Curr Opin Otolaryngol Head Neck Surg


NU
2015;23:21-8.

[30] Mygind N, Lund V. Intranasal corticosteroids for nasal


MA

polyposis: biological rationale, efficacy, and safety.

Treat Respir Med 2006;5:93-102.


D

[31] Jennette JC, Falk RJ, Bacon PA, Basu N, Cid MC, Ferrario F,
E

et al. 2012 revised international chapel hill consensus


PT

conference nomenclature of vasculitides. Arthritis &


CE

Rheumatisms 2013;65:1–11.

[32] Nakamaru Y, Takagi D, Suzuki M, Homma A, Morita S, Homma


AC

A, et al. Otologic and Rhinologic Manifestations of

Eosinophilic Granulomatosis with Polyangiitis. Audiol

Neurootol 2016;21:45-53.

[33] Stevens WW, Lee RJ, Schleimer RP, Cohen NA. Chronic

rhinosinusitis pathogenesis. J Allergy Clin Immunol

2015;136:1442-53.
ACCEPTED MANUSCRIPT
24

[34] Adams KM, Abraham V, Spielman D, Kolls JK, Rubenstein

RC, Conner GE, et al. IL-17A induces Pendrin expression and

chloride-bicarbonate exchange in human bronchial epithelial

cells. PLoSOne 2014;9:e103263.

[35] Nofziger C, Vezzoli V, Dossena S, Schönherr T, Studnicka

J, Nofziger J, et al. STAT6 links IL-4/IL-13 stimulation

PT
with pendrin expression in asthma and chronic obstructive

RI
pulmonary disease. Clin Pharmacol Ther 2011;90:399-405.

SC
NU
MA
E D
PT
CE
AC

Você também pode gostar