Chronic rhinosinusitis (CRS) is a heterogeneous disease characterized by localized inflammation of the upper airways. CRS includes two main phenotypes, namely, CRS with nasal polyps and CRS without nasal polyps. The phenotype-based classification method cannot reflect the pathological mechanism. The endotype-based classification method has been paid more and more attention by researchers. It is mainly divided into type 2 and non-type 2 endotypes. The mechanism driving the pathogenesis of non-type 2 inflammation is currently unknown. In this review, the PubMed and Web of Science databases were searched to conduct a critical analysis of representative literature works on the pathogenesis of non-type 2 inflammation in CRS published in the past decade. This review summarizes the latest evidence that may lead to the pathogenesis of non-type 2 inflammation. It is the main method that analyzing the pathogenesis from the perspective of immunology. Genomics and proteomics technique provide new approaches to the study of the pathogenesis. Due to differences in race, environment, geography, and living habits, there are differences in the occurrence of non-type 2 inflammation, which increase the difficulty of understanding the pathogenesis of non-type 2 inflammation in CRS. Studies have confirmed that non-type 2 endotype is more common in Asian patients. The emergence of overlap and unclassified endotypes has promoted the study of heterogeneity in CRS. In addition, as the source of inflammatory cells and the initiation site of the inflammatory response, microvessels and microlymphatic vessels in the nasal mucosal subepithelial tissue participate in the inflammatory response and tissue remodeling. It is uncertain whether CRS patients affect the risk of infection with SARS-CoV-2. In addition, the pathophysiological mechanism of non-type 2 CRS combined with COVID-19 remains to be further studied, and it is worth considering how to select the befitting biologics for CRS patients with non-type 2 inflammation.

Chronic rhinosinusitis (CRS) is a complex inflammatory disease characterized by localized inflammation of the upper airway and sinuses lasting at least 12 weeks and leading to a significant decline in quality of life [1]. About 10% of the world’s population is afflicted by the disease, resulting in reduced quality of life and productivity [2]. According to the results of endoscopic and sinus computed tomography examinations, CRS is usually divided into two main phenotypes [3], namely, CRS with nasal polyps (CRSwNP) [4] and CRS without nasal polyps (CRSsNP) [5]. CRSsNP is usually attributed to the mechanical obstruction of sinus drainage channels caused by secondary inflammation and infection [6]. CRSwNP is considered to be a diffuse eosinophilic mucosal inflammatory response process [7]. In addition, other phenotypes include aspirin-exacerbated respiratory disease [8, 9], infectious CRS [10, 11], allergic fungal rhinosinusitis [12], cystic fibrosis [13, 14], etc. However, the traditional phenotype-based classification method severely underestimates the complex cellular and molecular biological mechanisms [15, 16].

Currently, the mechanisms driving different phenotypes in CRS are unknown [17]. The purpose of this review was to summarize the latest evidence on the pathogenesis of non-type 2 inflammation in CRS, extract the main points of representative literature works in the past 10 years, reorganize the main research directions on the pathogenesis of non-type 2 inflammation, and update the relevant information on the heterogeneity. The participation of microvascular or microlymphangiogenesis in inflammatory cell recruitment, tissue remodeling, and inflammatory response in non-type 2 CRS is emphasized. Their potential contributions to the pathogenesis of non-type 2 inflammation in CRS are speculated. Hopefully, the study of nasal mucosal epithelium and subepithelial tissue will form a complete closed loop. The novelty and contribution of this review are to sort out the research direction of the pathogenesis of non-type 2 inflammation in CRS in the past 10 years and guide the balance of research on the nasal epithelium and subepithelial tissue. The potential roles of immune mechanisms within microvessels and lymphatic vessels in the pathogenesis of non-type 2 inflammation in CRS are predicted. A detailed explanation of the abbreviations used in this article can be found in the online supplementary material at https://doi.org/10.1159/000532067.

Different groups have proposed the possible pathogenesis of CRS from multiple perspectives in the past 10 years. They elaborated on the possible pathogenesis of CRS from the perspectives of genetics [18‒20] and immunology [21, 22]. Most of existing studies focus on the nasal mucosal epithelium [23‒25] and ignore the influence of immune function in blood vessels and lymphatic vessels in subepithelial tissue.

Existing studies on the pathogenesis of non-type 2 inflammation in CRS need to be improved in at least the following aspects. First, the research directions are scattered. It is hoped that a clear structure can be sorted out by the review. Second, current researches focus on the epithelium of the nasal mucosa [23, 24, 26] and ignore the impact of the immune regulation mechanism of microvessels and microlymphatic vessels. Currently, studies on microvascular and microlymphangiogenesis have been matured in the fields of tumor, trauma, transplantation, and immunology, which undoubtedly provide new ideas for the exploration of the pathogenesis of non-type 2 inflammation in CRS.

Phenotype

Phenotypes are usually classified according to clinically observed features [6]. CRSwNP and CRSsNP appear different from each other endoscopically and have long been thought to be pathophysiologically distinct [27]. It has been reported that approximately one-third of CRS suffer from CRSwNP [28], whereas the occurrence of CRSsNP is higher than that of CRSwNP [29]. Stevens et al. [30] reported the correlation between the endotypes and the clinical phenotypes. CRS patients with mixed type 2 and type 3 endotypes often have clinical manifestations shared by both type 2 and type 3 inflammatory endotypes. It is speculated that the clinical manifestations are directly related to the endotypes of inflammation in CRS.

Endotype

In order to better understand the pathogenesis of CRS [31], researchers have introduced the concept of endotypes [32, 33]. Endotypes refer to pathophysiological mechanisms that can be identified by specific biomarkers [34, 35]. Endotypes are classified based on histological features such as trends in neutrophils, eosinophils, and cytokines. Biomarkers for endotype identification in CRS can be determined directly from biological tissues of patients [36]. Commonly used biological tissues include peripheral blood, turbinate tissue, and sinus mucosa tissue [37]. The sinus mucosa is the most commonly used tissue as the site of disease occurrence [38‒40].

Currently, CRS mainly includes two endotypes, namely, type 2 and non-type 2 endotypes. Type 2 endotype usually receives more attention from researchers due to refractory nature. Non-type 2 endotype often receives less attention [41]. The complicating matter is that a certain endotype may be associated with multiple clinical phenotypes, and endotypes may contribute to the heterogeneity of phenotypes in CRS. Inflammation in CRSsNP and CRSwNP has been reported to be highly heterogeneous [42]. Each phenotype can exhibit a type 2 inflammatory endotype and a non-type 2 inflammatory endotype [43].

The concept of endotype has been paid attention by researchers after it was proposed. Tomassen et al. [44] pioneered the definition of endotype in CRS and distinguished 10 endotypes by cluster analysis of biomarkers. Bachert et al. [21] proposed to differentiate endotypes in CRS based on the pattern of inflammatory cells, specifically T cells and innate lymphoid cells (ILCs), which control major cytokines and chemokines, leading to the accumulation of eosinophils and neutrophils. Therefore, the publication of the above work opened the prelude to the study of endotypes. Liao et al. [45] used cluster analysis to identify endotypes in CRS and divided them into 7 clusters. Cluster 1 is comparable to typical eosinophilic CRSwNP (eCRSwNP). Most of them are refractory CRS with severe clinical symptoms and high recurrence rate. Clusters 3 and 6 are characterized by severe or moderate neutrophil inflammation, respectively, as well as elevated interleukin 8 (IL-8) levels and a high proportion of refractory CRS.

The 2020 European Position Paper on Rhinosinusitis and Nasal Polyps (EPOS) guidelines recommend the evaluation and treatment of primary CRS inflammatory disease in the context of type 2 or non-type 2 endotype [1]. Studies have found that for most patients, the main difference is between type 2 and non-type 2 inflammation [46]. Two major endotypes can be found in the CRSwNP and CRSsNP phenotypes [47]. The non-type 2 classification includes all other unknown factors regarding inflammation and may change over time [43]. There are also literature works that divide non-type 2 inflammatory endotypes into type 1 and type 3 endotypes [48].

The research scope of the pathophysiology of CRS includes microbial flora [7, 49], environment [1], genetic factors [20], and immune responses [50]. Research on the pathophysiology of CRS has always been a hotspot in this field. Bachert et al. [21] reported four effective methods and pointed out the direction for revealing the pathology of CRS. The first is to analyze surgically collected patient tissues, such as sinus, nasal polyp (NP) tissue, and blood samples. The second is to establish animal models. The third is to establish a model that simulates the situation in the human body, such as explant culture of sinus tissue and NPs. The fourth is to observe the therapeutic response to biological products, such as anti-IgE and anti-cytokine therapy [51]. Lam et al. [52] reported six hypotheses on the pathogenesis of CRS, namely, the fungal hypothesis [24], the superantigen hypothesis, the biofilm hypothesis, the microbiome hypothesis [53], the eicosanoid hypothesis [54], and the immune barrier hypothesis [55]. By combing literature works in the past 10 years, it can be seen that the research on the pathophysiology of CRS is mainly based on the above six hypotheses. We will review the relevant research progress on the pathogenesis of non-type 2 inflammation in CRS in the past 10 years.

The Concept of Non-Type 2 Inflammation

Non-type 2 endotype in CRS can be subdivided into type 1 and type 3/type 17 endotypes according to inflammatory cytokines [56, 57], as shown in Figure 1 [6, 21, 42].

Fig. 1.

Inflammatory pathways in non-type 2 inflammation in CRS. Non-type 2 immune responses can be subdivided into type 1 and type 3/17 inflammation. Type 1 inflammatory mechanisms involve epithelial responses to environmental stimuli and induce Th1 cell and Th17 cell differentiation. Type 1 inflammation is characterized by preferential expression of the Th1 cytokine IFN-γ produced by Th1 cells, NK cells, and ILC1s. Th1 cells activate macrophages to produce IFN-γ. Type 3 inflammation is a response to extracellular bacteria and fungi that stimulate epithelial cells to produce osteopontin. Osteopontin stimulates DCs to induce Th17 cell differentiation. Th3 cytokines IL-17 and IL-22 are produced by Th17 cells and ILC3s, which lead to chronic inflammation. Neutrophil recruitment, activation, and proliferation occur in the tissue after type 1 and type 3 responses are activated. Vascular permeability increases, and tissue remodeling occurs in the tissue. Ang, angiopoietins; DCs, dendritic cells; IgG, immunoglobulin G; IFN-γ, interferon-γ; IL, interleukin; ILC1s, type 1 innate lymphoid cells; ILC2s, type 2 innate lymphoid cells; ILC3s, type 3 innate lymphoid cells; M1, type 1 macrophage; NKs, natural killer cells; Th1, T helper type 1; Th3, T helper type 3; Th17, T helper type 17; Treg, T regulatory; TNF, tumor necrosis factor; TSLP, thymic stromal lymphopoietin; VEGF, vascular endothelial growth factor.

Fig. 1.

Inflammatory pathways in non-type 2 inflammation in CRS. Non-type 2 immune responses can be subdivided into type 1 and type 3/17 inflammation. Type 1 inflammatory mechanisms involve epithelial responses to environmental stimuli and induce Th1 cell and Th17 cell differentiation. Type 1 inflammation is characterized by preferential expression of the Th1 cytokine IFN-γ produced by Th1 cells, NK cells, and ILC1s. Th1 cells activate macrophages to produce IFN-γ. Type 3 inflammation is a response to extracellular bacteria and fungi that stimulate epithelial cells to produce osteopontin. Osteopontin stimulates DCs to induce Th17 cell differentiation. Th3 cytokines IL-17 and IL-22 are produced by Th17 cells and ILC3s, which lead to chronic inflammation. Neutrophil recruitment, activation, and proliferation occur in the tissue after type 1 and type 3 responses are activated. Vascular permeability increases, and tissue remodeling occurs in the tissue. Ang, angiopoietins; DCs, dendritic cells; IgG, immunoglobulin G; IFN-γ, interferon-γ; IL, interleukin; ILC1s, type 1 innate lymphoid cells; ILC2s, type 2 innate lymphoid cells; ILC3s, type 3 innate lymphoid cells; M1, type 1 macrophage; NKs, natural killer cells; Th1, T helper type 1; Th3, T helper type 3; Th17, T helper type 17; Treg, T regulatory; TNF, tumor necrosis factor; TSLP, thymic stromal lymphopoietin; VEGF, vascular endothelial growth factor.

Close modal

Cells and Cytokines in Non-Type 2 Inflammation

Type 1 inflammation is commonly seen with microorganisms [49, 58] and viruses [11, 59]. It is characterized by preferential expression of interferon-γ (IFN-γ) produced by Th1 cells, natural killer cells (NKs), and type 1 innate lymphoid cells (ILC1s) [6]. Th1 cells activate macrophages to produce IFN-γ which is a hallmark marker of type 1 inflammation. IFN-γ induces apoptosis in nasal epithelial cells and stimulates neutrophil activity [60]. Type 1 inflammation may contribute to disease chronicity and is generally more common in the CRSsNP phenotype [61, 62].

IL-6, IL-8, and tumor necrosis factor (TNF) have been shown to stimulate the production of IFN-γ, which further promotes type 1 immune responses [43]. Bequignon et al. [63] found that wound closure of injured epithelium in CRSwNP patients was significantly faster under IL-6 stimulation. It is suggested that IL-6 can induce epithelial cell proliferation and promote NP formation. Therefore, it is speculated that IL-6 may be an important cytokine in the physiopathology of NPs.

Type 1 inflammation has been reported in Asian patients or second-generation Asian CRS patients in the USA [64]. Klingler et al. [47] found that type 1 CRSsNP is characterized by the accumulation of Th1 cells, CD8+ cytotoxic T cells, and NKs. Type 1 CRSsNP is associated with viral infection and NK cell-mediated cytotoxicity. Wang et al. [65] found that type 1 endotype is the main endotype of CRSsNP in Asia. It is unclear why Asian patients are more likely to have type 1 inflammation in CRSwNP compared with patients from Western countries. It has not been determined whether genetic factors play key roles [19, 66].

Type 3 inflammation is a response to bacteria and fungi [67] that triggers epithelial cells to produce osteopontin, which stimulates DCs to induce Th17 cell differentiation. Type 3 inflammation is characterized by the production of the T helper type 3(Th3) cytokines IL-17 and IL-22 by Th17 cells and ILC3 [68]. It results in neutrophil recruitment, activation, and proliferation after activation of the type 3 immune responses [69]. On one hand, neutrophils have the innate immune ability to phagocytize microorganisms and produce antibacterial products. On the other hand, the inflammatory factors released by them can aggravate tissue damage and inflammatory response [70]. Wang et al. [71] found that elastase secreted by neutrophils is the main activator of IL-36γ. IL-36γ promotes the expression of IL-17A in neutrophils, which in turn upregulates the expression of IL-36γ in nasal epithelial cells and leads to a vicious cycle of persistent neutrophil inflammation in CRS. Kim et al. [72] found that anti-IL-33 treatment in a mouse model of CRS reduced the thickness of edematous mucosa, subepithelial deposition of collagen, and neutrophil infiltration, but not eosinophil infiltration. It is speculated that IL-33 may become a key mediator of the pathogenesis of CRSwNP by recruiting neutrophils [70]. Klingler et al. [47] found that type 3 CRSsNP was characterized by the accumulation of Th17 cells, B cells, DCs, M1 macrophages (type 1 macrophages), and neutrophils. It is speculated that type 3 CRSsNP is related to bacterial infection. Li et al. [73] found that the Th17/Treg (T regulatory) ratio and IL-6 levels were significantly elevated in both eCRSwNP and non-eCRSwNP. It is speculated that IL-6 may regulate the function of Th17, Treg cells and the Th17/Treg ratio. It plays a role in the pathogenesis of CRSwNP. In order to facilitate sorting out the key information, the trend of changes in Th1, Th2, and Th3 cytokines in CRS is summarized in Table 1 [45, 71, 73‒77].

Table 1.

Changes in Th1, Th2, and Th3 cytokines in the population

CytokinesDefinition of caseEndotypePopulationFindingYearCountryReferences
IFN-γ non-eCRSwNP Type 1/type 3 Adult Increased 2015 Japan Baba et al. [74
IL-17 CRSwNP Type 3 Adult Increased 2021 China Yu et al. [75
non-eCRSwNP Type 1/type 3 Adult Increased 2015 Japan Baba et al. [74
IL-6 CRSwNP Type 3 Adult Increased 2022 China Li et al. [73
CRSwNP Type 2/type 3 Adult Increased 2020 Switzerland Steiner et al. [76
IL-8 CRSwNP Type 3 Adult Increased 2018 China, USA Liao et al. [45
IL-4 CRSwNP Type 2 Mice Increased 2019 France, USA Rouyar et al. [77
IL-5 CRSwNP Type 2 Mice Increased 2019 France, USA Rouyar et al. [77
CRSwNP Type 2/type 3 Adult Increased 2020 Switzerland Steiner et al. [76
IL-13 CRSwNP Type 2 Mice Increased 2019 France, USA Rouyar et al. [77
CytokinesDefinition of caseEndotypePopulationFindingYearCountryReferences
IFN-γ non-eCRSwNP Type 1/type 3 Adult Increased 2015 Japan Baba et al. [74
IL-17 CRSwNP Type 3 Adult Increased 2021 China Yu et al. [75
non-eCRSwNP Type 1/type 3 Adult Increased 2015 Japan Baba et al. [74
IL-6 CRSwNP Type 3 Adult Increased 2022 China Li et al. [73
CRSwNP Type 2/type 3 Adult Increased 2020 Switzerland Steiner et al. [76
IL-8 CRSwNP Type 3 Adult Increased 2018 China, USA Liao et al. [45
IL-4 CRSwNP Type 2 Mice Increased 2019 France, USA Rouyar et al. [77
IL-5 CRSwNP Type 2 Mice Increased 2019 France, USA Rouyar et al. [77
CRSwNP Type 2/type 3 Adult Increased 2020 Switzerland Steiner et al. [76
IL-13 CRSwNP Type 2 Mice Increased 2019 France, USA Rouyar et al. [77

Heterogeneity of Non-Type 2 Inflammation

The complicating matter is that the prevalence of non-type 2 inflammation varies with geographically, environmental, genetic factors and lifestyles [78‒80]. Both type 2 and non-type 2 (type 1 and/or type 3) endotypes are present in Asian patients [2, 81].

Heterogeneity of CRSsNP

Studies found that type 1 and type 3 endotypes have been demonstrated in parts of Asia, Europe, and the USA. The difference is that type 1, type 3, or their mixed endotypes are common in CRSsNP in Asia [78]. Wang et al. [78] demonstrated that type 1 and mixed type 1/3 endotypes were predominant in CRSsNP patients in Beijing, China. Type 1, type 2, or type 3 marker was not elevated in patients in Chengdu, China. This suggests that there are also significant geographical differences within a country and inflammatory endotypes are more controlled by environment than by genetic factors. In general, CRSsNP is classified as type 1 inflammation based on the elevation of IFN-γ. It is a characteristic marker of type 1 inflammation [42]. However, recent studies found that elevated IFN-γ in CRSsNP could not be confirmed [65]. It is speculated that the inflammation may be transferred from type 1 to type 2. Therefore, the inflammatory endotypes in CRSsNP are highly heterogeneous, and the occurrence of each endotype varies from region to region, which undoubtedly increases the difficulty of studying non-type 2 inflammatory endotype.

Heterogeneity of CRSwNP

Asian CRSwNP patients especially in China, Korea, and Japan are characterized by Th1 and Th17 inflammation. They may have mixed type 1 or type 3 inflammation characteristics [82]. Compared with the inflammation observed in NPs of American or European patients, the neutrophils were more common [31]. Similarly, neutrophil inflammation may also play a role in the pathogenesis of Western NPs [83, 84]. Wang et al. [65] found that type 1, type 2, and type 3 inflammation showed diversity in Europe, Asia, and Oceania. Endoypes in CRSwNP are highly heterogeneous. The occurrence of each endotype varies geographically [65]. Studies have found that there are not only type 2 immune responses in CRSwNP but also non-type 2 immune responses [36]. Jiang et al. [85] found that eosinophilic inflammation was significantly enhanced in CRSwNP patients in central China over time. It may be related to increased T helper type 2 (Th2) responses and IgE production. Hulse et al. [86] found that second-generation Asian patients born in the USA had significantly fewer eosinophils in NP tissues than African-American and Hispanic patients living in the Greater Chicago area. It is suggested that genetic differences may play key roles in the development of NPs [66]. How do the environment [80] and genetics [19] influence the development of non-type 2 inflammation worthy of further study [87].

In addition, the study found that age may be one of the factors affecting the heterogeneity of CRS [88]. The function of innate and acquired immunity changes with age, which can be confirmed by observing the distribution of age-related immune cells in peripheral blood. The deterioration of immune function with age may affect the pathophysiological process of CRS [88]. Ryu et al. [89] reported differences in age-related cytokines change in CRS subtypes and control group. In the CRSsNP and non-eCRSwNP groups, type 2 cytokines were found to increase with age, and type 3 cytokines were found to decrease with age. In the eCRSwNP group, type 2 media were found to remain at high levels, and type 3 media increase with age. It is speculated that the age-related distribution of inflammatory mediators is different in CRS endotypes. In addition, the study suggests that smoking may be one of the factors influencing the age-related increase of inflammatory mediators in eCRSwNP. Kim et al. [90] reported a significant correlation between age and tissue eosinophils. They found that the proportion of non-eCRSwNP was significantly higher in younger patients than in older patients (79.2% vs. 56.6%). An association was hypothesized between age and non-type 2 inflammation in patients with CRSwNP. Understanding age-related immune regulation in CRS will be necessary to establish precision medicine in the future.

Application of Genomics and Proteomics Technique to Non-Type 2 Inflammation

Recently, genomics and proteomics approaches have played increasing roles in identifying immune mechanism in non-type 2 inflammatory endotype [91]. Wang et al. [65] found that NPs from non-type 2 CRSwNP patients exhibited IFN-γ-induced genes (CXCL9, CXCL10, and CXCL11), IL-17A-induced genes (CXCL6), and neutrophil chemokines (IL-8, CXCL1, and CXCL6). It is speculated that the mixed type 1 and type 3 endotypes are the most common endotypes in non-type 2 CRSwNP in Beijing. It suggests that non-type 2 CRSwNP in Asia may predominantly exhibit a mixed type 1 and type 3 endotypes with neutropenia [65, 91]. Large-scale transcriptome studies are needed to further define immune mechanisms in type 1 and type 3 CRSwNP in the future.

It should be noted that sometimes the endotype does not belong to one of the above-mentioned type 1, type 2, or type 3. Nearly 25% of CRS patients have mixed endotypes. It is difficult to divide them into certain types of endotypes [92, 93]. Since inflammatory pathways can be related in multiple ways, it is understandable that CRS has crossed endotypes. For example, eCRSwNP may also have significant levels of neutrophils and non-type 2 cytokines [71]. Kato et al. [6] found that all three endotypes were elevated in patients with CRSsNP or CRSwNP. It indicates the existence of endotype overlap. It can be classified from the perspective of distinguishing which endotype is dominant.

In addition, sometimes the test results do not show visible elevations of type 1, type 2, or type 3 endotype markers in patients with CRSsNP. It cannot be classified under the current endotype classification system [6, 30]. Possible factors leading to this phenomenon include the several aspects. First, the unrecognized endotype markers of CRS patients may be slightly elevated, but the level of significant difference cannot be reached. Second, the patients may have type 1, type 2, or type 3 inflammation, but the researchers did not sample it. Furthermore, CRS patients may have unrecognized endotypes in addition to type 1, type 2, and type 3 [6]. It is required to understand the immune mechanisms and pathogenesis of unrecognized CRS in the future.

Research on microvascular permeability and microlymphangiogenesis has been successful in the fields of allergy, infection, and oncology [94‒98], which will provide inspiration for the study of the pathogenesis of non-type 2 inflammation in CRS. Currently, most studies on the pathogenesis of non-type 2 endotype focus on nasal mucosal epithelium [23, 25] and ignore the impact of immune mechanisms in blood vessels and lymphatic vessels in the subepithelial tissue.

Immune Regulation Mechanism of Microvessels in Non-Type 2 CRS

Under normal physiological conditions, the main function of microvessels is to maintain blood flow, transport nutrients to tissues for material exchange [99], and transport inflammatory cells to effector organs or tissues. Increased vascular permeability is often observed under inflammatory conditions and is accompanied by increased expression of inflammatory genes [100]. Inflammatory cells and cytokines have effects on vascular permeability and angiogenesis. On the contrary, the absorption and permeability of blood vessel wall have influences on the transport and penetration of inflammatory cells during the inflammatory process. According to the published literature works, research on inflammation-driven microvascular dysfunction in CRS has focused on type 2 inflammation [101‒103]. It is typically characterized by the observation of a large number of eosinophilic infiltrates. In addition, some studies did not target specific endotypes in CRS [104‒106]. Possibly, no significant changes in inflammatory cytokines were observed.

Currently, inflammation-induced vascular dysfunction and its associated genes and signaling pathways have not been identified in non-type 2 endotype in CRS. Khurana et al. [105] found that vascular biomarkers and signaling pathways were significantly overexpressed in patients with CRS. It was speculated that vascular dysfunction occurred. These pathways are usually quiescent in adults. When stimulated by inflammation or injury, the migration of inflammatory leukocytes increases which releases more inflammatory cytokines. It leads to remodeling of vascular endothelial cells, initiating new angiogenesis, and regulating blood flow and inflammatory cells from the vascular space to infiltrate into the tissue. Vascular leakiness and dysfunction occur, which allow increased exchange of macromolecules. Another study by Khurana et al. [104] found a significant increase in the number of blood vessels in CRS patients compared to the control group. Luukkainen et al. [106] found that the average density of all vessels in the NP group was significantly lower than in the control group in the area of the highest vessel density. It was previously believed that the blood vessel density in polyps may be similar to that in normal tissues [107]. A larger sample size is needed to confirm in future research.

The public literature works on microvascular dysfunction of CRS driven by non-type 2 inflammation have not been retrieved. Similar studies are mainly published in the fields of asthma [95, 108], atherosclerosis [109], diabetes [110], diabetic retinopathy (DR) [111, 112], tumor [97, 98, 113], blood-brain barrier (BBB) [114, 115], hypertension [116‒118], and so on. Cytokines have been found to play a role in inflammation-driven vascular dysfunction [113]. Increased vascular risk is associated with elevated levels of non-type 2 inflammatory cytokines, such as INF-γ, TNF-α, IL-6, and IL-8 [119]. IFN-γ is thought to play a role in antiviral response and vascular dysfunction [119]. TNF-α is an important cytokine in injured blood vessels, which can regulate the expression of VEGF, fibroblast growth factor, adhesion molecules, etc. [119]. IL-6 has been found to be involved in the pathogenesis of specific vascular diseases [119]. IL-8 is produced by monocytes, lymphocytes, fibroblasts, endothelial cells, epithelial cells, etc. and is only released under inflammatory conditions [97]. Inflammation is mostly associated with pathological angiogenesis [100]. Angiogenesis can promote inflammation and the progression of vascular diseases [120]. Therefore, angiogenesis may be a therapeutic target for the treatment of various human diseases.

Angiogenesis is the main feature of airway remodeling in asthma research, which is mainly mediated by VEGF [108]. Akdis et al. [95] found that IL-17 plays a role in the progression of inflammation in patients with asthma. IL-17 is secreted by Th17 cells and is a proinflammatory cytokine. Th17 cells upregulate VEGF production in bronchial epithelial cells [108] and promote the development of angiogenesis. In addition, Kudo et al. [94] also found that IL-17 has a similar proinflammatory effect in airway hyperresponsiveness.

Vascular inflammation plays an important role in the occurrence and progression of atherosclerosis [109]. Xu et al. [96] found that endothelial cells are activated in response to injury, and they produce IL-8, chemokines, interferons, intercellular adhesion molecule-1, vascular adhesion molecule-1, and other inflammatory factors. Proinflammatory mediators such as TNF-α stimulate endothelial cells to secrete other proinflammatory cytokines, including IL-6. They regulate chronic vascular inflammatory responses [121].

Studies have found that vascular inflammation can promote atherosclerosis and vascular endothelial dysfunction in diabetes [110], resulting in a high incidence of cardiovascular disease in diabetic patients. Based on the elevated concentrations detected, inflammatory mediators such as IL-6 and vascular adhesion molecule-1 were implicated in this process [110]. Ma et al. [110] showed that exercise has a significant benefit in relieving vascular inflammation. Proper endurance exercise plays an anti-inflammatory role by inhibiting proinflammatory cytokines.

DR is one of the common complications of microangiopathy in diabetes [112]. Symptoms of retinal ischemia, osmosis, and angiogenesis are common in patients with the disease. Tsai et al. [111] found that the expression levels of proinflammatory factors IL-1β and INF-γ were significantly increased in DR patients, while the expression level of IL-6 was not affected. Both VEGF and VEGF-A are upregulated in patients with DR. It is speculated that proinflammatory factors and angiogenic factors are related to inflammation and angiogenesis [111]. They play key roles in the development of DR. One possible reason for the absence of IL-6 upregulation in DR patients is the inhibitory effect of IL-1β on IL-6 signaling.

Inflammation in tumor angiogenesis is mainly related to tumor-associated macrophage recruitment [113]. Various inflammatory mediators secreted by tumor-associated macrophage act on tumor endothelial cells [113]. Tumor endothelial cell upregulates the production of proinflammatory cytokines and chemokines, thereby regulating the activity of inflammatory cells, recruiting them to the site of inflammation, and promoting tumor angiogenesis. These inflammatory molecules include IL-1β, IL-6, IL-8, and TNF-α [98]. Matsushima et al. [97] found that IL-8 was involved in tumor-related angiogenesis. IL-8 interacts with CXCR2 receptors on endothelial cells to induce angiogenesis. Anti-IL-8 antibody combined with anti-VEGFR may synergistically inhibit tumor-related angiogenesis.

The BBB is an important regulator of material movement between blood and nerve tissue [113]. Recruitment of proinflammatory chemokines (CCL2, CCL5, and CXCL10) and proinflammatory mediators (IL-17 and IL-22) occurs during BBB injury in most neurological diseases [114]. Proinflammatory factors (IL-1β, IL-6, and TNF-α) promote the proliferation of endothelial cells in the BBB and enhance angiogenesis during neuroinflammation [115]. Increased VEGF-A stimulates BBB leakage, secretion of proinflammatory cytokines, and infiltration of leukocytes, which lead to neuroinflammation [115].

The link between endothelial dysfunction and inflammatory cytokine production has been demonstrated [116]. IL-17A plays an important role in endothelial dysfunction associated with hypertension [118]. IL-17A enhances the proinflammatory effects of other cytokines on endothelial cells, vascular smooth muscle cells, and macrophages. IL-17A stimulation increased the expression of proinflammatory genes in cultured mouse vascular smooth muscle cells compared to control mice [118]. Overexpression of IL-17A in mice leads to increased production of reactive oxygen species, which in turn triggers vascular dysfunction and perivascular fibrosis. Tanase et al. [116] showed that chronic inflammation leads to increased vascular permeability and thrombosis associated with persistent hypertension. The serum levels of IL-1β, IL-6, IL-8, IL-17, IL-23, TGF-β, TNF-α, and other proinflammatory cytokines in hypertensive patients increase the production of reactive oxygen species, leading to vascular endothelial dysfunction. In addition, studies have found that elevated serum IL-6 levels can lead to increased production of hepatitis markers such as C-reactive protein (CRP) and increase vascular permeability, apoptosis, and thrombosis [116, 117].

The findings suggest that controlling angiogenesis and VEGF-A holds promise for improving quality of life and longevity in patients with heart disease [122]. Braile et al. [122] reported the expression of VEGF-A in cardiomyocytes (CM) and the role of VEGF-A in cardiovascular diseases. VEGF can cause cell-cell contact disruption and increased permeability. CM is the source of inflammatory cytokines and angiogenic factors, and it secretes TNF-α, IFN-γ, IL-6, ANGPT1, ANGPT2, VEGF-A, etc. The results suggest that the lack of VEGF-A expression in CM may affect myocardial angiogenesis and lead to impaired cardiac function.

Inflammation-driven microvascular dysfunction plays an important role in the progression of COVID-19 [123‒125]. Chen et al. [123] showed that after the SARS-CoV-2 virus entered human cells by binding with ACE2, many critically ill patients developed cytokine storm [126] or cytokine release syndrome [127]. Serum levels of inflammatory factors such as IL-6, IL-1, IFN-γ and endothelial activating markers such as vasculopathy factor were elevated, leading to endodermatitis and microvascular thrombosis [124, 125].

Angiogenesis and inflammation are features of almost all chronic liver diseases [113]. Vascular capillaries and abnormal organization interfere with the interaction between endothelial and cells, thus activating the immune response and leading to inflammation and chronic liver disease [128]. Activated liver endothelial cells have proinflammatory effects and secrete a variety of cytokines and chemokines such as TNF-α, IL-1, IL-6, and CCL2 [129], which promote the development of inflammation. Inflammatory cells recruited in the damaged liver microenvironment secrete angiogenic factors to promote liver angiogenesis, aggravate inflammation through metabolism, and form a cycle between liver angiogenesis and inflammation [129].

In conclusion, studies on microvascular dysfunction triggered by inflammation have focused on vascular endothelial cells, while the role of the pericytes remains to be investigated. Pericytes provide functional support for endothelium, which play important roles in the functional maintenance of the blood-retinal barrier and BBB [130, 131]. Previously, pericyte loss in both the blood-retinal barrier and BBB has been reported [132]. Pericyte loss leads to increased vascular permeability and tissue damage in diabetes mellitus [133, 134]. The role of pericytes in microvascular dysfunction triggered by non-type 2 inflammation in CRS has not been reported and further studies are needed.

Immune Regulation Mechanism of Microlymphatic Vessels in Non-Type 2 CRS

Lymphatic vessels are part of the immune organs and are participants in inflammation. They are responsible for transporting and reabsorbing inflammatory cells [135]. They are closely related to inflammatory response. Quantification of blood and lymphatic vessel density has previously been used as a prognostic indicator in cancer tumors, especially melanoma and head and neck cancer [136]. Recently, researchers have begun to pay attention to the relationship between CRS and lymphatic dysregulation [106]. Luukkainen et al. [106] found that the relative density of lymphatic vessels in CRSwNP was significantly lower than that in the control group, both in the area of the highest vascular density and throughout the tissue. It is speculated that low relative density of lymphatic vessels may be related to NPs. Chauhan et al. [135] showed that IL-17 promoted the growth of lymphatic vessels by inducing increased expression of VEGF-D in lymphatic angiogenesis.

As CRS is a heterogeneous disease, the sample size may have contributed to sampling bias. The existing research on blood vessel and lymphatic vessel dysfunction in CRS has great limitations at present. Further studies with a larger sample size are needed to clarify the influence of lymphatic vessel dysfunction on the pathogenesis of non-type 2 endotype in CRS.

CRS and COVID-19 are the most common causes of olfactory dysfunction [46]. However, there is little scientific information on the link between the two. Marin et al. [46] reported the impact of the COVID-19 pandemic on olfactory function in patients with CRS. They found that type 2 eosinophilic inflammation may decrease the expression of ACE2 in sinus epithelial cells. They speculate that eosinophilic inflammation in CRS may play a protective role against olfactory dysfunction caused by SARS-CoV-2 infection. In addition, they found that increased ACE2 expression was associated with the expression of type 1 inflammatory cytokine IFN-γ in non-eCRSwNP patients. They suggest that non-eCRSwNP may increase susceptibility to SARS-CoV-2 infection. Wang et al. [10] found that the expression of ACE2 was significantly increased in the nasal tissues of non-eCRSwNP patients. It was positively correlated with the expression of IFN-γ but negatively correlated with the expression of tissue-infiltrating eosinophils, IL-5 and IL-13. They found that TMPRSS2 expression was reduced in the nasal tissue of CRSwNP patients and was independent of the inflammatory endotypes of CRSwNP. Therefore, it is speculated that IFN-γ-regulated ACE2 expression is increased in nasal tissues of non-eCRSwNP patients and is positively correlated with type 1 inflammation. Xu et al. [92] found that SARS-CoV-2 utilizes ACE2 as a receptor for entry into cells and infection. In non-eCRSwNP tissues, ACE2 expression was further upregulated by IFN-γ. It is speculated that type 1 inflammation may increase susceptibility to COVID-19. However, there is currently no evidence that people with non-type 2 CRS are at higher risk of contracting severe COVID-19.

Marin et al. [137] reported that type 2 inflammation in tissues of CRSwNP patients may be related to the downregulation of ACE2/TMPRSS2 in the olfactory neuroepithelium. It may reduce the risk of further damage to the olfactory neuroepithelium and the entry of SARS-CoV-2 into the brain. Lee et al. [138] found that a prominent type 2 immune response caused by NPs may contribute to reducing SARS-CoV-2 infectiousness and COVID-19 severity compared to CRSsNP. Similar findings were reported by Jian et al. [139], who found that type 2 inflammation in the airway may have a protective effect against COVID-19 infection or its severity. It is pointed out that the expression and regulation of ACE2 receptor and TMPRSS2 protease are the key points of study.

Forster-Ruhrmann et al. [140] reported a reduction of disease in patients with severe CRSwNP treated with dupilumab during COVID-19 infection. It is speculated that continuous treatment with dupilumab may reduce local inflammation, improve nasal breathing, and ultimately control the symptoms of CRSwNP. Klimek et al. [141] reported several biologics targeting related cytokines or effector cells for the treatment of CRS during the COVID-19 pandemic. Dupilumab was shown to reduce IgE production, Th2 differentiation, and recruitment of eosinophils. Mepolizumab and reslizumab can induce apoptosis and reduce recruitment of eosinophils. Benralizumab may deplete tissue of eosinophils. Omalizumab can reduce IgE receptor expression and reduce mast cell mediator release.

At present, it is uncertain whether patients with non-type 2 CRS and targeted biologics for patients with non-type 2 CRS affect the risk of infection with SARS-CoV-2. In addition, the protective hypothesis of CRS against COVID-19 and the pathophysiological mechanism of non-type 2 CRS combined with COVID-19 remain to be further studied.

The guidelines recommend saline irrigation and intranasal corticosteroids as standard treatment and oral corticosteroids and antibiotics as adjuvant treatment [1]. When CRS patients do not respond to drug therapy, surgery is recommended [142]. However, the strategy does not take into account underlying pathophysiological mechanisms and does not respond optimally to the heterogeneity of CRS. Recently, precision medicine has received more and more attention [61, 143].

Corticosteroids

Glucocorticoids (GCs) have anti-inflammatory, immunosuppressive, and anti-allergic effects [1]. Oral and topical GCs are considered to be effective treatments for CRS and are widely used in most patients [61, 144]. It has been reported that the effective rate of GC therapy for CRS is 50%–80% [145]. Corticosteroids can effectively inhibit type 2 inflammation in CRS, including eosinophils, ILC2s, and TH2 cells. However, some patients with CRS may not respond to GCs [145]. Studies by Milara et al. [146] showed that approximately 30% of CRS patients responded poorly to GCs. Yan et al. [147], Lou et al. [143], and Wu et al. [148] showed that the effect of GC treatment depended on the concentration of eosinophils or neutrophils. Although systemic corticosteroids may relieve inflammatory symptoms, long-term use may produce adverse effects [1]. The benefits of GCs in the treatment of non-type 2 CRS need to be supported by more clinical data.

Antibiotics

At present, microbial resistance to antibiotics is continuously increasing [145, 149]. Although oral antibiotics can alleviate acute respiratory symptoms, eliminating resistant bacteria remains a challenge [150]. There is conflicting evidence regarding the benefits of antibiotics as a form of treatment for CRS [151, 152], and recommendations for antibiotic use in CRS patients are controversial. Currently, the latest guidelines do not recommend the routine use of topical and intravenous antibiotics for the treatment of CRS [1, 151]. Actual evidence for the treatment of CRS with oral antibiotics remains scarce [145]. Carlton et al. [150] showed that topical antibiotic therapy is useful in the short term in CRS patients with refractory Staphylococcus aureus (S. aureus) infection. Although guidelines recommend against topical antibiotics, they are still used in clinical practice [150]. Miyake et al. [153] showed that local treatment has the potential to deliver higher drug concentrations to the disease site, reducing systemic side effects and antibiotic resistance. Miyake et al. [153] reviewed recent advances in topical drug therapy and compared the potential and shortcomings of topical drug therapy for CRS. Although the efficacy of local treatment varies, different local treatment strategies continue to emerge. It is speculated that local antibiotic therapy is a promising area.

Corticosteroids have a limited role in non-type 2 CRS, and treatment options often favor antibiotics and surgery [70]. In general, patients with CRSsNP are often treated with antibiotics [152]. The study found that patients with low tissue eosinophils benefited the most from antibiotic treatment, suggesting that patients with non-type 2 disease are most likely to benefit from the drugs [151, 152]. As the role of microbial infection in CRS patients is gradually explained, the rationality of antibiotic use will be reexamined [145]. Further validation of antimicrobial and anti-inflammatory effects of antibiotics on appropriate CRS patients is needed in the future to determine which patients are suitable for antibiotic treatment [145].

Surgery

Sinus surgery is another effective way to treat CRS when medications fail to control symptoms [152]. Endoscopic sinus surgery for the treatment of CRS can achieve the purpose of relieving the obstruction of the nasal passage and removing inflammatory tissue [152]. Patel et al. [152] proposed three criteria for determining the rationality of surgical treatment. The first is a Lund-McKay computed tomography score of 1 or above. The second is adequate medication before surgery, including a course of antibiotics in patients with CRSsNP or a short course of oral corticosteroids in patients with CRSwNP. Third, the SNOT-22 score can reach 20 or above after drug treatment.

One of the advantages of surgery is that the diseased tissue of the patient can be collected [70] to obtain the most direct information about the disease, which can be used to predict subsequent treatment measures and treatment outcomes. The disadvantage is that the condition may be repeated after surgery. It has been reported that approximately 40% of patients with CRS develop recurrent disease 3∼5 years after surgery [70, 154]. In addition, patients who choose surgery also bear risks such as accidental injury and anesthesia [155]. Seys et al. [156] showed that multiple nasal endoscopic surgeries can reduce patients’ sense of smell. Because CRS is a heterogeneous disease, the goals and effects of surgery may vary. Sometimes, surgery and other treatment strategies can be combined. For example, in patients with type 2 inflammation at highest risk of recurrence, sinusitis incision and debridement of inflammatory tissue may be combined with high-dose topical corticosteroid irrigation [152]. Studies have found that surgery may affect the classification of endotypes [151]. Type 2 inflammatory mediators are reduced after surgery, and more than 50% of patients have endotype conversion [157].

Biologic Therapies

Biotherapy is an important adjunct to the treatment of CRS [158] and is usually used in patients with refractory diseases and drug or surgical failures [151, 159]. Currently, the biologics used generally target patients with type 2 CRSwNP [158, 160]. The United States Food and Drug Administration (FDA) has approved dupilumab, omalizumab, and mepolizumab for the treatment of CRSwNP [41]. Biologic drugs mainly target downstream cytokines of type 2 inflammation, mainly targeting IgE, IL-4, IL-5, and IL-13 [152, 161]. The recruitment and survival of inflammatory cells are regulated by blocking the signaling of inflammatory mediators [160]. In phase III clinical trials, these biologics improved NP scores in patients with CRSwNP. They have positive effects on both NP size and outcome measures [41, 151].

Research on the mechanism and treatment of non-type 2 immune response lags far behind that of type 2 immune responses [162]. Currently, no biologics targeting non-type 2 factors have been used in CRS [163‒165]. The development of biologic therapies for non-type 2 inflammation seems to be more difficult than for type 2 inflammation [61]. Research on non-type 2 inflammation needs to be strengthened [61]. Similarly, type 2 asthma is well characterized, while there is a lack of adequate biomarkers and targeted treatment options for non-type 2 asthma [166, 167]. Targeting TH17 cell signaling may be an effective therapeutic option for patients with corticosteroid-insensitive asthma, including targeting TH17 cell-associated cytokines, cytokine receptors, and signaling pathways [168]. Brodalumab is a human, anti-IL-17RA antibody which may block IL-17A, IL-17F, and other IL-17 subtypes. Busse et al. [169] reported a randomized double-blind study of brodalumab targeting strategies blocking IL-17 receptor signaling in asthma treatment, which found no significant improvement in treatment. IL-6 is an important cytokine that induces TH17 cell differentiation [168]. Chen et al. [170] reported that blocking IL-6 signaling reduced TH2/TH17 cell response and improved symptoms in corticosteroid-insensitive asthmatic mice. Chu et al. [171] reported that anti-IL-6 antibodies reduced neutrophil and eosinophilic cytokines and chemokines in mice and reduced airway inflammation. Because TH17 cells are not homogenous [168], the possible consequences of blocking the TH17 cell pathway must be carefully evaluated to determine the risks. McDowell et al. [172] showed that IL-17 and IL-13 play mutually regulating roles in asthma, suggesting that targeting these pathways simultaneously may be more powerful for treatment. Choy et al. [173] showed that the expression of TH2 and TH17 induced gene signaling is mutually exclusive and reversely regulated in patients with asthma. Patients with a TH17 signature also had elevated levels of the type 2 biomarker. Therefore, they speculate that patients with type 2 asthma may shift between TH2 and Th17 molecular markers over time. Targeting both type 2 and type 3 cytokines in combination may be better than targeting one or the other alone.

The CXC chemokine 2 receptor (CXCR2) is a key mediator of neutrophil migration. Targeting CXCR2 may inhibit the progression of non-type 2 inflammatory mechanisms [174]. Nair et al. [175] and O‘Byrne et al. [176] reported that CXCR2 antagonists targeted the CXCR2 on neutrophils and blocked its activation through the chemokine IL-8. Although neutrophils were reduced in sputum and blood in patients with asthma, no clinical improvement was seen. Because the authors did not elaborate on possible factors that may have contributed to the deterioration of efficacy during the trial, the assessment of biologics may have deviated from their actual effect.

In addition to driving type 2 inflammation after release from epithelial cells, TSLP also plays a role in non-type 2 immune response [177]. TSLP blocking has been shown to be a promising approach for treating both type 2-driven and non-type 2-driven inflammation in asthma [177]. In particular, for patients with non-type 2 inflammation, treatment options are limited. Although the pathogenesis of the asthma is unknown, TSLP blocking has been shown to be effective in such patients [177].

For severe asthma patients with low type 2 inflammation, treatment targeting type 2 cytokines has no significant clinical effect [178]. It suggests that there is a gap between the means and the need to treat low type 2 inflammation and mixed type 2 and non-type 2 inflammation. Maun et al. [178] found that mast cell trypsin levels were correlated with asthma severity and that elevated trypsin in the airways of patients with severe asthma was independent of type 2 biomarker levels. For this type 2 inflammatory independent subgroup, they developed anti-trypsin antibodies as a therapeutic agent for severe asthma. The results indicate the potential role of neutralization of trypsin activity in the treatment of type 2 independent asthma.

The use of biologics in non-type 2 inflammatory diseases may interfere with COVID-19 vaccination response [179]. Anti-cytokine antibodies (e.g., anti-IL-6) may block the antiviral cellular response. Therefore, Jutel et al. [179] recommend a minimum 7-day gap between biotargeted non-type 2 immune responses and COVID-19 vaccination to determine the possible side effects of each other. Because of the differences between the effects of different biologics, it is worth considering how to select the best biologics [151].

Existing studies on the pathological mechanism of non-type 2 endotype in CRS have at least the following limitations [180]. First of all, there is a time limit. Most of the lesions have already occurred when sampling. It is difficult to determine the susceptibility genetic and environmental factors in the early stage of CRS [80]. It is impossible to obtain new information about the disease mechanism from the source. Second, how to select the most ideal anatomical site as the research object has not been specified yet. Inflammatory cytokines have been reported to be unevenly distributed throughout the sinus tissue, and cytokine levels may vary depending on the anatomical site of origin [92]. In addition, the control group used, including inferior turbinate, hook, or cribriform tissue, was also non-standardized in tissue sampling which makes comparison between studies challenging between different groups [37]. Finally, there is a lack of widely accepted animal models of CRS. Most studies have focused on examining nasal secretions, nasal mucosa, NPs, sinus secretions, sinus mucosa, and peripheral blood from human patients with CRSwNP or CRSsNP. Therefore, new perspectives are needed to understand the pathophysiology of non-type 2 endotype in CRS. Genomics and proteomics studies, which can examine the entire genetic information in an unbiased and independent manner [180], are effective ways to determine the pathogenesis of non-type 2 endotype in the future. In addition, further studies on the pathogenesis of non-type 2 endotype in CRS are needed at least in the following aspects [46, 48], as shown in Table 2 [46, 65, 78, 79, 103, 106, 181, 182].

Table 2.

Further investigation of pathogenesis in non-type 2 inflammation in CRS

What is knownQuestions and gaps in understandingPersonal viewpoints
ILC2s play key roles in the production of type 2 cytokines in CRSwNP [181What are the roles of ILC1s and ILC3s in the pathogenesis of non-type 2 endotype in CRS? ILC1s produce type 1 cytokine IFN-γ. ILC3s produce type 3 cytokines IL-17 and IL-22. They all participate in the inflammatory process of non-type 2 immune responses 
The prevalence of non-type 2 inflammation varies with geographically, environmental, genetic factors, and lifestyle [65, 78, 79Which factor plays a decisive role in non-type 2 endotype in CRS? Geography, genetic factors, age, environment, and lifestyle all influence the incidence of non-type 2 CRS. Environmental and lifestyle influences may be even greater 
Increased vascular permeability and decreased lymphatic vessel density have been confirmed in CRS [103, 106What are the vascular dysfunction and associated genes and signaling pathways in non-type 2 inflammation in CRS patients? Non-type 2 cytokines such as INF-γ, TNF-α, IL-6, IL-8, and IL-17 are upregulated, promoting chronic vascular inflammatory responses and leading to microvascular dysfunction. In addition to causing vascular endothelial dysfunction, the morphology and number of pericytes may change 
The global epidemic of COVID-19 has an impact on the phenotype of CRS [46, 182Will the global pandemic of COVID-19 lead to the emergence of new endotypes in CRS? Or will it cause a transfer between existing endotypes? Endotype transitions in CRS may occur during COVID-19 treatment, but new endotypes are not expected 
What is knownQuestions and gaps in understandingPersonal viewpoints
ILC2s play key roles in the production of type 2 cytokines in CRSwNP [181What are the roles of ILC1s and ILC3s in the pathogenesis of non-type 2 endotype in CRS? ILC1s produce type 1 cytokine IFN-γ. ILC3s produce type 3 cytokines IL-17 and IL-22. They all participate in the inflammatory process of non-type 2 immune responses 
The prevalence of non-type 2 inflammation varies with geographically, environmental, genetic factors, and lifestyle [65, 78, 79Which factor plays a decisive role in non-type 2 endotype in CRS? Geography, genetic factors, age, environment, and lifestyle all influence the incidence of non-type 2 CRS. Environmental and lifestyle influences may be even greater 
Increased vascular permeability and decreased lymphatic vessel density have been confirmed in CRS [103, 106What are the vascular dysfunction and associated genes and signaling pathways in non-type 2 inflammation in CRS patients? Non-type 2 cytokines such as INF-γ, TNF-α, IL-6, IL-8, and IL-17 are upregulated, promoting chronic vascular inflammatory responses and leading to microvascular dysfunction. In addition to causing vascular endothelial dysfunction, the morphology and number of pericytes may change 
The global epidemic of COVID-19 has an impact on the phenotype of CRS [46, 182Will the global pandemic of COVID-19 lead to the emergence of new endotypes in CRS? Or will it cause a transfer between existing endotypes? Endotype transitions in CRS may occur during COVID-19 treatment, but new endotypes are not expected 

Endotypes in CRS are mainly divided into type 2 and non-type 2 endotype. The mechanisms driving the pathogenesis of non-type 2 endotype in CRS are currently unknown. The occurrence of non-type 2 endotype in CRS varies with race, geography, environment, and lifestyle. Type 1, type 2, and type 3 and unrecognized endotypes in CRS promoted the study of CRS heterogeneity. The immune mechanisms of CRS with overlap and unrecognized endotypes remain to be further studied. In addition, as emerging aspects of the inflammatory response mechanism in CRS, lymphatic vessel density, vascular permeability, angiogenesis and their related genes and signaling pathways have not yet been identified. The impact of immune mechanisms in blood vessels and lymphatic vessels in the subepithelial tissue on CRS still needs to be confirmed in larger sample size studies. Finally, the pathophysiological mechanism of non-type 2 CRS combined with COVID-19 remains to be further studied. It is worth considering how to select the befitting biologics for CRS patients with non-type 2 endotype.

The authors have no conflicts of interest to declare.

This research was supported in part by the National Nature Science Foundation of China (81870701).

Na Cui, Xuewei Zhu, Chen Zhao, Cuida Meng, Jichao Sha, and Dongdong Zhu have contributed in writing and reviewing the final version of the manuscript.

1.
Fokkens WJ, Lund VJ, Hopkins C, Hellings PW, Kern R, Reitsma S, et al. European position paper on rhinosinusitis and nasal polyps 2020. Rhinology. 2020;58(Suppl S29):1–464.
2.
Sedaghat AR, Kuan EC, Scadding GK. Epidemiology of chronic rhinosinusitis: prevalence and risk factors. J Allergy Clin Immunol Pract. 2022;10(6):1395–403.
3.
Naclerio R, Mullol J, Stevens WW. A decade of clinical advances in chronic rhinosinusitis: 2012-2022. J Allergy Clin Immunol Pract. 2023;11(1):43–50.
4.
Kristjansson RP, Benonisdottir S, Davidsson OB, Oddsson A, Tragante V, Sigurdsson JK, et al. A loss-of-function variant in ALOX15 protects against nasal polyps and chronic rhinosinusitis. Nat Genet. 2019;51(2):267–76.
5.
Cho SH, Hamilos DL, Han DH, Laidlaw TM. Phenotypes of chronic rhinosinusitis. J Allergy Clin Immunol Pract. 2020;8(5):1505–11.
6.
Kato A, Peters AT, Stevens WW, Schleimer RP, Tan BK, Kern RC. Endotypes of chronic rhinosinusitis: relationships to disease phenotypes, pathogenesis, clinical findings, and treatment approaches. Allergy. 2022;77(3):812–26.
7.
Hamilos DL. Drivers of chronic rhinosinusitis: inflammation versus infection. J Allergy Clin Immunol. 2015;136(6):1454–9.
8.
Morales DR, Guthrie B, Lipworth BJ, Jackson C, Donnan PT, Santiago VH. NSAID-exacerbated respiratory disease: a meta-analysis evaluating prevalence, mean provocative dose of aspirin and increased asthma morbidity. Allergy. 2015;70(7):828–35.
9.
Divekar R, Patel N, Jin J, Hagan J, Rank M, Lal D, et al. Symptom-based clustering in chronic rhinosinusitis relates to history of aspirin sensitivity and postsurgical outcomes. J Allergy Clin Immunol Pract. 2015;3(6):934–40.e3.
10.
Wang M, Bu X, Fang G, Luan G, Huang Y, Akdis CA, et al. Distinct expression of SARS-CoV-2 receptor ACE2 correlates with endotypes of chronic rhinosinusitis with nasal polyps. Allergy. 2021;76(3):789–803.
11.
Wang H, Song J, Pan L, Deng YK, Yao Y, Wang ZC, et al. Regional differences in ACE2 expression in the sinonasal mucosa of adult Chinese patients with chronic rhinosinusitis. Allergy. 2021;76(5):1565–8.
12.
Hoyt AE, Borish L, Gurrola J, Payne S. Allergic fungal rhinosinusitis. J Allergy Clin Immunol Pract. 2016;4(4):599–604.
13.
Krajewska J, Zub K, Słowikowski A, Zatoński T. Chronic rhinosinusitis in cystic fibrosis: a review of therapeutic options. Eur Arch Otorhinolaryngol. 2022;279(1):1–24.
14.
Mainz JG, Koitschev A. Pathogenesis and management of nasal polyposis in cystic fibrosis. Curr Allergy Asthma Rep. 2012;12(2):163–74.
15.
Hellings PW, Fokkens WJ, Bachert C, Akdis CA, Bieber T, Agache I, et al. Positioning the principles of precision medicine in care pathways for allergic rhinitis and chronic rhinosinusitis: a EUFOREA-ARIA-EPOS-AIRWAYS ICP statement. Allergy. 2017;72(9):1297–305.
16.
Toro MDC, Antonio MA, Alves Dos Reis MG, de Assumpcao MS, Sakano E. Achieving the best method to classify Eosinophilic Chronic Rhinosinusitis: a systematic review. Rhinology. 2021;59(4):330–9.
17.
Steinke JW, Borish L. Chronic rhinosinusitis phenotypes. Ann Allergy Asthma Immunol. 2016;117(3):234–40.
18.
Mitts KB, Chang EH. Genetics of chronic rhinosinusitis. J Allergy Clin Immunol. 2020;145(3):777–9.
19.
Bohman A, Oscarsson M, Holmberg K, Johansson L, Millqvist E, Nasic S, et al. Heredity of nasal polyps. Rhin. 2015;53(1):25–8.
20.
Purnell PR, Addicks BL, Zalzal HG, Shapiro S, Wen S, Ramadan HH, et al. Single nucleotide polymorphisms in chemosensory pathway genes GNB3, TAS2R19, and TAS2R38 are associated with chronic rhinosinusitis. Int Arch Allergy Immunol. 2019;180(1):72–8.
21.
Bachert C, Akdis CA. Phenotypes and emerging endotypes of chronic rhinosinusitis. J Allergy Clin Immunol Pract. 2016;4(4):621–8.
22.
Karadeniz EB, Aliyeva C, Aydemir S, Karaaslan BG, Mustu U, Ada M, et al. Chronic rhinosinusitis in patients with primary immunodeficiency. Int Arch Allergy Immunol. 2023;184(3):302–10.
23.
Cornet ME, Kostamo K, Rinia AB, Zwinderman AH, van Egmond D, de Groot E, et al. Novel roles for nasal epithelium in the pathogenesis of chronic rhinosinusitis with nasal polyps. Rhinology. 2019;57(3):169–79.
24.
Ickrath P, Sprügel L, Beyersdorf N, Haug L, Scherzad A, Hagen R, et al. Aspergillus fumigatus-specific T cells in patients with chronic rhinosinusitis. Int Arch Allergy Immunol. 2023;184(5):502–12.
25.
Zhou M, Tan KS, Guan WJ, Jiang LJ, Deng J, Gao WX, et al. Proteomics profiling of epithelium-derived exosomes from nasal polyps revealed signaling functions affecting cellular proliferation. Respir Med. 2020;162:105871.
26.
Xia Y, Wang H, Yin J. The role of epithelial-mesenchymal transition in chronic rhinosinusitis. Int Arch Allergy Immunol. 2022;183(10):1029–39.
27.
Hopkins C. Chronic rhinosinusitis with nasal polyps. N Engl J Med. 2019;381(1):55–63.
28.
Ahn JC, Kim JW, Lee CH, Rhee CS. Prevalence and risk factors of chronic Rhinosinusitus, allergic rhinitis, and nasal septal deviation: results of the Korean national health and nutrition survey 2008-2012. JAMA Otolaryngol Head Neck Surg. 2016;142(2):162–7.
29.
Tan BK, Chandra RK, Pollak J, Kato A, Conley DB, Peters AT, et al. Incidence and associated premorbid diagnoses of patients with chronic rhinosinusitis. J Allergy Clin Immunol. 2013;131(5):1350–60.
30.
Stevens WW, Peters AT, Tan BK, Klingler AI, Poposki JA, Hulse KE, et al. Associations between inflammatory endotypes and clinical presentations in chronic rhinosinusitis. J Allergy Clin Immunol Pract. 2019;7(8):2812–20.e3.
31.
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 academy of allergy, asthma & immunology. J Allergy Clin Immunol. 2013;131(6):1479–90.
32.
Hamilos DL. Chronic rhinosinusitis endotyping: sharpening the focus on inflammation. J Allergy Clin Immunol. 2016;137(5):1457–9.
33.
Brescia G, Zanotti C, Parrino D, Barion U, Marioni G. Nasal polyposis pathophysiology: endotype and phenotype open issues. Am J Otolaryngol. 2018;39(4):441–4.
34.
Turner JH, Chandra RK, Li P, Bonnet K, Schlundt DG. Identification of clinically relevant chronic rhinosinusitis endotypes using cluster analysis of mucus cytokines. J Allergy Clin Immunol. 2018;141(5):1895–7.e7.
35.
Khan A, Vandeplas G, Huynh TMT, Joish VN, Mannent L, Tomassen P, et al. The Global Allergy and Asthma European Network (GALEN rhinosinusitis cohort: a large European cross-sectional study of chronic rhinosinusitis patients with and without nasal polyps. Rhin. 2019;57(1):32–42.
36.
Bachert C, Marple B, Hosemann W, Cavaliere C, Wen W, Zhang N. Endotypes of chronic rhinosinusitis with nasal polyps: pathology and possible therapeutic implications. J Allergy Clin Immunol Pract. 2020;8(5):1514–9.
37.
Huang Y, Zhang N, Xu Z, Zhang L, Bachert C. The development of the mucosal concept in chronic rhinosinusitis and its clinical implications. J Allergy Clin Immunol Pract. 2022;10(3):707–15.
38.
Pan L, Liao B, Guo CL, Liu JX, Wang H, Long XB, et al. Inflammatory features and predictors for postsurgical outcomes in patients with nasal polyps stratified by local and systemic eosinophilia. Int Forum Allergy Rhinol. 2021;11(5):846–56.
39.
Turner JH, Li P, Chandra RK. Mucus T helper 2 biomarkers predict chronic rhinosinusitis disease severity and prior surgical intervention. Int Forum Allergy Rhinol. 2018;8(10):1175–83.
40.
Ho J, Hamizan AW, Alvarado R, Rimmer J, Sewell WA, Harvey RJ. Systemic predictors of eosinophilic chronic rhinosinusitis. Am J Rhinol Allergy. 2018;32(4):252–7.
41.
Cardell LO, Stjärne P, Jonstam K, Bachert C. Endotypes of chronic rhinosinusitis: impact on management. J Allergy Clin Immunol. 2020;145(3):752–6.
42.
Kato A, Schleimer RP, Bleier BS. Mechanisms and pathogenesis of chronic rhinosinusitis. J Allergy Clin Immunol. 2022;149(5):1491–503.
43.
Grayson JW, Hopkins C, Mori E, Senior B, Harvey RJ. Contemporary classification of chronic rhinosinusitis beyond polyps vs No polyps: a review. JAMA Otolaryngol Head Neck Surg. 2020;146(9):831–8.
44.
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(5):1449–56.e4.
45.
Liao B, Liu JX, Li ZY, Zhen Z, Cao PP, Yao Y, et al. Multidimensional endotypes of chronic rhinosinusitis and their association with treatment outcomes. Allergy. 2018;73(7):1459–69.
46.
Marin C, Hummel T, Liu Z, Mullol J. Chronic rhinosinusitis and COVID-19. J Allergy Clin Immunol Pract. 2022;10(6):1423–32.
47.
Klingler AI, Stevens WW, Tan BK, Peters AT, Poposki JA, Grammer LC, et al. Mechanisms and biomarkers of inflammatory endotypes in chronic rhinosinusitis without nasal polyps. J Allergy Clin Immunol. 2021;147(4):1306–17.
48.
Czerwaty K, Piszczatowska K, Brzost J, Ludwig N, Szczepański MJ, Dżaman K. Immunological aspects of chronic rhinosinusitis. Diagnostics. 2022;12(10):2361.
49.
Choi EB, Hong SW, Kim DK, Jeon SG, Kim KR, Cho SH, et al. Decreased diversity of nasal microbiota and their secreted extracellular vesicles in patients with chronic rhinosinusitis based on a metagenomic analysis. Allergy. 2014;69(4):517–26.
50.
Tan BK, Peters AT, Schleimer RP, Hulse KE. Pathogenic and protective roles of B cells and antibodies in patients with chronic rhinosinusitis. J Allergy Clin Immunol. 2018;141(5):1553–60.
51.
Mortuaire G, Gengler I, Carpentier C, Szymanski C, Chenivesse C, Lefevre G. T helper 2 inflammatory markers are associated with recurrence in chronic rhinosinusitis with nasal polyps after endoscopic sinus surgery. Rhinology. 2020;58(5):444–50.
52.
Lam K, Schleimer R, Kern RC. The etiology and pathogenesis of chronic rhinosinusitis: a review of current hypotheses. Curr Allergy Asthma Rep. 2015;15(7):41.
53.
Calus L, Derycke L, Dullaers M, Van Zele T, De Ruyck N, Pérez-Novo C, et al. IL-21 is increased in nasal polyposis and after stimulation with Staphylococcus aureus enterotoxin B. Int Arch Allergy Immunol. 2017;174(3–4):161–9.
54.
Pezato R, Świerczyńska-Krępa M, Niżankowska-Mogilnicka E, Derycke L, Bachert C, Pérez-Novo CA. Role of imbalance of eicosanoid pathways and staphylococcal superantigens in chronic rhinosinusitis. Allergy. 2012;67(11):1347–56.
55.
Ono N, Kusunoki T, Miwa M, Hirotsu M, Shiozawa A, Ikeda K. Reduction in superoxide dismutase expression in the epithelial mucosa of eosinophilic chronic rhinosinusitis with nasal polyps. Int Arch Allergy Immunol. 2013;162(2):173–80.
56.
Shi LL, Song J, Xiong P, Cao PP, Liao B, Ma J, et al. Disease-specific T-helper cell polarizing function of lesional dendritic cells in different types of chronic rhinosinusitis with nasal polyps. Am J Respir Crit Care Med. 2014;190(6):628–38.
57.
Baba S, Kondo K, Toma-Hirano M, Kanaya K, Suzukawa K, Ushio M, et al. Local increase in IgE and class switch recombination to IgE in nasal polyps in chronic rhinosinusitis. Clin Exp Allergy. 2014;44(5):701–12.
58.
Hamilos DL. Host-microbial interactions in patients with chronic rhinosinusitis. J Allergy Clin Immunol. 2014;133(3):640–53.e4.
59.
Goggin RK, Bennett CA, Bialasiewicz S, Vediappan RS, Vreugde S, Wormald PJ, et al. The presence of virus significantly associates with chronic rhinosinusitis disease severity. Allergy. 2019;74(8):1569–72.
60.
Lee M, Kim DW, Khalmuratova R, Shin SH, Kim YM, Han DH, et al. The IFN-γ-p38, ERK kinase axis exacerbates neutrophilic chronic rhinosinusitis by inducing the epithelial-to-mesenchymal transition. Mucosal Immunol. 2019;12(3):601–11.
61.
Avdeeva K, Fokkens W. Precision medicine in chronic rhinosinusitis with nasal polyps. Curr Allergy Asthma Rep. 2018;18(4):25.
62.
Borish L, Hamilos DL. Endotype-phenotype correlation in chronic rhinosinusitis: is it time to think beyond polyposis?J Allergy Clin Immunol Pract. 2019;7(8):2821–2.
63.
Bequignon E, Mangin D, Bécaud J, Pasquier J, Angely C, Bottier M, et al. Pathogenesis of chronic rhinosinusitis with nasal polyps: role of IL-6 in airway epithelial cell dysfunction. J Transl Med. 2020;18(1):136.
64.
Stevens WW, Lee RJ, Schleimer RP, Cohen NA. Chronic rhinosinusitis pathogenesis. J Allergy Clin Immunol. 2015;136(6):1442–53.
65.
Wang X, Zhang N, Bo M, Holtappels G, Zheng M, Lou H, et al. Diversity of TH cytokine profiles in patients with chronic rhinosinusitis: a multicenter study in Europe, Asia, and Oceania. J Allergy Clin Immunol. 2016;138(5):1344–53.
66.
Mahdavinia M, Suh LA, Carter RG, Stevens WW, Norton JE, Kato A, et al. Increased noneosinophilic nasal polyps in chronic rhinosinusitis in US second-generation Asians suggest genetic regulation of eosinophilia. J Allergy Clin Immunol. 2015;135(2):576–9.
67.
Annunziato F, Romagnani C, Romagnani S. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol. 2015;135(3):626–35.
68.
Bachert C, Zhang N. Medical algorithm: diagnosis and treatment of chronic rhinosinusitis. Allergy. 2020;75(1):240–2.
69.
Wang M, Zhang N, Zheng M, Li Y, Meng LL, Ruan Y, et al. Cross-talk between TH2 and TH17 pathways in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2019;144(5):1254–64.
70.
Xu X, Ong YK, Wang Y. Novel findings in immunopathophysiology of chronic rhinosinusitis and their role in a model of precision medicine. Allergy. 2020;75(4):769–80.
71.
Wang H, Li ZY, Jiang WX, Liao B, Zhai GT, Wang N, et al. The activation and function of IL-36γ in neutrophilic inflammation in chronic rhinosinusitis. J Allergy Clin Immunol. 2018;141(5):1646–58.
72.
Kim DK, Jin HR, Eun KM, Mo JH, Cho SH, Oh S, et al. The role of interleukin-33 in chronic rhinosinusitis. Thorax. 2017;72(7):635–45.
73.
Li H, Wang Y, Wang J. Th17/Treg cells regulated by interleukin 6 in the pathogenesis of chronic rhinosinusitis with nasal polyps. Eur Arch Otohhinolaryngol. 2022;279(7):3493–501.
74.
Baba S, Kagoya R, Kondo K, Suzukawa M, Ohta K, Yamasoba T. T-cell phenotypes in chronic rhinosinusitis with nasal polyps in Japanese patients. Allergy Asthma Clin Immunol. 2015;11:33.
75.
Yu S, Cao C, Li Q, Wen X, Guo X, Bao Q, et al. Local IL-17 positive T cells are functionally associated with neutrophil infiltration and their development is regulated by mucosal microenvironment in nasal polyps. Inflamm Res. 2021;70(1):139–49.
76.
Steiner UC, Bischoff S, Valaperti A, Ikenberg K, Starzyk J, Bucher S, et al. Endotypes of chronic rhinosinusitis with nasal polyps with and without NSAID: intolerance. Rhinology. 2020;58(6):544–9.
77.
Rouyar A, Classe M, Gorski R, Bock MD, Le-Guern J, Roche S, et al. Type 2/Th2-driven inflammation impairs olfactory sensory neurogenesis in mouse chronic rhinosinusitis model. Allergy. 2019;74(3):549–59.
78.
Tan BK, Klingler AI, Poposki JA, Stevens WW, Peters AT, Suh LA, et al. Heterogeneous inflammatory patterns in chronic rhinosinusitis without nasal polyps in Chicago, Illinois. J Allergy Clin Immunol. 2017;139(2):699–703.e7.
79.
Stevens WW, Peters AT, Suh L, Norton JE, Kern RC, Conley DB, et al. A retrospective, cross-sectional study reveals that women with CRSwNP have more severe disease than men. Immun Inflamm Dis. 2015;3(1):14–22.
80.
Puxeddu I, Petrelli F, Cristofani-Mencacci L, Scarano M, Latorre M, De Rosa A, et al. Component-resolved diagnosis for endotyping patients with chronic rhinosinusitis with nasal polyps. Int Arch Allergy Immunol. 2022;183(7):770–7.
81.
Wen W, Liu W, Zhang L, Bai J, Fan Y, Xia W, et al. Increased neutrophilia in nasal polyps reduces the response to oral corticosteroid therapy. J Allergy Clin Immunol. 2012;129(6):1522–8.e5.
82.
Han X, Wu D, Sun Z, Sun H, Lv Q, Zhang L, et al. Type 1/type 2 inflammatory cytokines correlate with olfactory function in patients with chronic rhinosinusitis. Am J Otolaryngol. 2020;41(5):102587.
83.
Delemarre T, Holtappels G, De Ruyck N, Zhang N, Nauwynck H, Bachert C, et al. A substantial neutrophilic inflammation as regular part of severe type 2 chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2021;147(1):179–88.e2.
84.
Pothoven KL, Norton JE, Suh LA, Carter RG, Harris KE, Biyasheva A, et al. Neutrophils are a major source of the epithelial barrier disrupting cytokine oncostatin M in patients with mucosal airways disease. J Allergy Clin Immunol. 2017;139(6):1966–78.e9.
85.
Jiang WX, Cao PP, Li ZY, Zhai GT, Liao B, Lu X, et al. A retrospective study of changes of histopathology of nasal polyps in adult Chinese in central China. Rhinology. 2019;57(4):261–7.
86.
Hulse KE, Stevens WW, Tan BK, Schleimer RP. Pathogenesis of nasal polyposis. Clin Exp Allergy. 2015;45(2):328–46.
87.
Staudacher AG, Peters AT, Kato A, Stevens WW. Use of endotypes, phenotypes, and inflammatory markers to guide treatment decisions in chronic rhinosinusitis. Ann Allergy Asthma Immunol. 2020;124(4):318–25.
88.
Cho SH, Hong SJ, Han B, Lee SH, Suh L, Norton J, et al. Age-related differences in the pathogenesis of chronic rhinosinusitis. J Allergy Clin Immunol. 2012;129(3):858–60.e2.
89.
Ryu G, Dhong HJ, Park M, Hwang NY, Kim DK, Kim HY, et al. Age-associated changes in chronic rhinosinusitis endotypes. Clin Exp Allergy. 2020;50(5):585–96.
90.
Kim JW, Won TB, Woo H, Yang SK, Phannikul C, Ko SA, et al. Age related non-type 2 inflammation and its association with treatment outcome in patients with chronic rhinosinusitis with nasal polyp in Korea. Sci Rep. 2022;12(1):1671.
91.
Okada N, Nakayama T, Asaka D, Inoue N, Tsurumoto T, Takaishi S, et al. Distinct gene expression profiles and regulation networks of nasal polyps in eosinophilic and non-eosinophilic chronic rhinosinusitis. Int Forum Allergy Rhinol. 2018;8(5):592–604.
92.
Xu X, Reitsma S, Wang Y, Fokkens WJ. Highlights in the advances of chronic rhinosinusitis. Allergy. 2021;76(11):3349–58.
93.
Delemarre T, Holtappels G, De Ruyck N, Zhang N, Nauwynck H, Bachert C, et al. Type 2 inflammation in chronic rhinosinusitis without nasal polyps: another relevant endotype. J Allergy Clin Immunol. 2020;146(2):337–43.e6.
94.
Kudo M, Melton AC, Chen C, Engler MB, Huang KE, Ren X, et al. IL-17A produced by αβ T cells drives airway hyper-responsiveness in mice and enhances mouse and human airway smooth muscle contraction. Nat Med. 2012;18(4):547–54.
95.
Akdis M, Palomares O, van de Veen W, van Splunter M, Akdis CA. TH17 and TH22 cells: a confusion of antimicrobial response with tissue inflammation versus protection. J Allergy Clin Immunol. 2012;129(6):1438–49; quiz1450-1.
96.
Xu S, Ilyas I, Little PJ, Li H, Kamato D, Zheng X, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev. 2021;73(3):924–67.
97.
Matsushima K, Yang D, Oppenheim JJ. Interleukin-8: an evolving chemokine. Cytokine. 2022;153:155828.
98.
Kammerer T, Faihs V, Hulde N, Stangl M, Brettner F, Rehm M, et al. Hypoxic-inflammatory responses under acute hypoxia: in Vitro experiments and prospective observational expedition trial. Int J Mol Sci. 2020;21(3):1034.
99.
Monahan-Earley R, Dvorak AM, Aird WC. Evolutionary origins of the blood vascular system and endothelium. J Thromb Haemost. 2013;11(Suppl 1):46–66.
100.
Chen L, Deng H, Cui H, Fang J, Zuo Z, Deng J, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2017;9(6):7204–18.
101.
Hirshoren N, Neuman T, Gross M, Eliashar R. Angiogenesis in chronic rhinosinusitis with nasal polyps and in antrochoanal polyps. Inflamm Res. 2011;60(4):321–7.
102.
Karatzanis AD, Samara KD, Antoniou KM, Lymbouridou R, Chatzakis N, Spandidos DA, et al. Investigation of angiogenetic pathways in nasal polyposis. Mol Med Rep. 2012;5(5):1158–62.
103.
Khurana N, Yathavan B, Jedrzkiewicz J, Gill AS, Pulsipher A, Alt JA, et al. Vascular permeability in chronic rhinosinusitis enhances accumulation and retention of nanoscale pegylated liposomes. Nanomedicine. 2021;38:102453.
104.
Khurana N, Pulsipher A, Jedrzkiewicz J, Ashby S, Pollard CE, Ghandehari H, et al. Inflammation-driven vascular dysregulation in chronic rhinosinusitis. Int Forum Allergy Rhinol. 2021;11(6):976–83.
105.
Khurana N, Pulsipher A, Ghandehari H, Alt JA. Meta-analysis of global and high throughput public gene array data for robust vascular gene expression discovery in chronic rhinosinusitis: implications in controlled release. J Control Release. 2021;330:878–88.
106.
Luukkainen A, Seppälä M, Renkonen J, Renkonen R, Hagstrő M J, Huhtala H, et al. Low lymphatic vessel density associates with chronic rhinosinusitis with nasal polyps. Rhinology. 2017;55(2):181–91.
107.
Schleimer RP. Immunopathogenesis of chronic rhinosinusitis and nasal polyposis. Annu Rev Pathol. 2017;12:331–57.
108.
Meyer N, Akdis CA. Vascular endothelial growth factor as a key inducer of angiogenesis in the asthmatic airways. Curr Allergy Asthma Rep. 2013;13(1):1–9.
109.
Haybar H, Shokuhian M, Bagheri M, Davari N, Saki N. Involvement of circulating inflammatory factors in prognosis and risk of cardiovascular disease. J Mol Cell Cardiol. 2019;132:110–9.
110.
Ma Y, Liu H, Wang Y, Xuan J, Gao X, Ding H, et al. Roles of physical exercise-induced MiR-126 in cardiovascular health of type 2 diabetes. Diabetol Metab Syndr. 2022;14(1):169.
111.
Tsai T, Kuehn S, Tsiampalis N, Vu MK, Kakkassery V, Stute G, et al. Anti-inflammatory cytokine and angiogenic factors levels in vitreous samples of diabetic retinopathy patients. PLoS One. 2018;13(3):e0194603.
112.
Antonetti DA, Klein R, Gardner TW. Diabetic retinopathy. N Engl J Med. 2012;366(13):1227–39.
113.
Jeong JH, Ojha U, Lee YM. Pathological angiogenesis and inflammation in tissues. Arch Pharm Res. 2021;44(1):1–15.
114.
Daneman R, Prat A. The blood-brain barrier. Cold Spring Harb Perspect Biol. 2015;7(1):a020412.
115.
Chen CC, Chen YC, Hsiao HY, Chang C, Chern Y. Neurovascular abnormalities in brain disorders: highlights with angiogenesis and magnetic resonance imaging studies. J Biomed Sci. 2013;20(1):47.
116.
Tanase DM, Gosav EM, Radu S, Ouatu A, Rezus C, Ciocoiu M, et al. Arterial hypertension and interleukins: potential therapeutic target or future diagnostic marker?Int J Hypertens. 2019;2019:1–17.
117.
Teixeira BC, Lopes AL, Macedo RCO, Correa CS, Ramis TR, Ribeiro JL, et al. Inflammatory markers, endothelial function and cardiovascular risk. J Vasc Bras. 2014;13(2):108–15.
118.
Rodrigues-Diez RR, Tejera-Muñoz A, Orejudo M, Marquez-Exposito L, Santos-Sanchez L, Rayego-Mateos S, et al. Interleukin-17A: potential mediator and therapeutic target in hypertension. Nefrologia. 2021;41(3):244–57.
119.
Sprague AH, Khalil RA. Inflammatory cytokines in vascular dysfunction and vascular disease. Biochem Pharmacol. 2009;78(6):539–52.
120.
Li Y, Zhu H, Wei X, Li H, Yu Z, Zhang H, et al. LPS induces HUVEC angiogenesis in vitro through miR-146a-mediated TGF-β1 inhibition. Am J Transl Res. 2017;9(2):591–600.
121.
Libby P. Interleukin-1 beta as a target for atherosclerosis therapy: biological basis of CANTOS and beyond. J Am Coll Cardiol. 2017;70(18):2278–89.
122.
Braile M, Marcella S, Cristinziano L, Galdiero MR, Modestino L, Ferrara AL, et al. VEGF-A in cardiomyocytes and heart diseases. Int J Mol Sci. 2020;21(15):5294.
123.
Chen LYC, Hoiland RL, Stukas S, Wellington CL, Sekhon MS. Confronting the controversy: interleukin-6 and the COVID-19 cytokine storm syndrome. Eur Respir J. 2020;56(4):2003006.
124.
Herold T, Jurinovic V, Arnreich C, Lipworth BJ, Hellmuth JC, von Bergwelt-Baildon M, et al. Elevated levels of IL-6 and CRP predict the need for mechanical ventilation in COVID-19. J Allergy Clin Immunol. 2020;146(1):128–36.e4.
125.
Lucas C, Wong P, Klein J, Castro TBR, Silva J, Sundaram M, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature. 2020;584(7821):463–9.
126.
Fajgenbaum DC, June CH. Cytokine storm. N Engl J Med. 2020;383(23):2255–73.
127.
Kang S, Tanaka T, Inoue H, Ono C, Hashimoto S, Kioi Y, et al. IL-6 trans-signaling induces plasminogen activator inhibitor-1 from vascular endothelial cells in cytokine release syndrome. Proc Natl Acad Sci U S A. 2020;117(36):22351–6.
128.
Sanz-Cameno P, Trapero-Marugán M, Chaparro M, Jones EA, Moreno-Otero R. Angiogenesis: from chronic liver inflammation to hepatocellular carcinoma. J Oncol. 2010;2010:1–7.
129.
Lafoz E, Ruart M, Anton A, Oncins A, Hernández-Gea V. The endothelium as a driver of liver fibrosis and regeneration. Cells. 2020;9(4):929.
130.
Salameh TS, Shah GN, Price TO, Hayden MR, Banks WA. Blood-brain barrier disruption and neurovascular unit dysfunction in diabetic mice: protection with the mitochondrial carbonic anhydrase inhibitor topiramate. J Pharmacol Exp Ther. 2016;359(3):452–9.
131.
Hill J, Rom S, Ramirez SH, Persidsky Y. Emerging roles of pericytes in the regulation of the neurovascular unit in health and disease. J Neuroimmune Pharmacol. 2014;9(5):591–605.
132.
Tien T, Muto T, Barrette K, Challyandra L, Roy S. Downregulation of Connexin 43 promotes vascular cell loss and excess permeability associated with the development of vascular lesions in the diabetic retina. Mol Vis. 2014;20:732–41.
133.
Rom S, Zuluaga-Ramirez V, Gajghate S, Seliga A, Winfield M, Heldt NA, et al. Hyperglycemia-driven neuroinflammation compromises BBB leading to memory loss in both diabetes mellitus (DM) type 1 and type 2 mouse models. Mol Neurobiol. 2019;56(3):1883–96.
134.
Shah GN, Morofuji Y, Banks WA, Price TO. High glucose-induced mitochondrial respiration and reactive oxygen species in mouse cerebral pericytes is reversed by pharmacological inhibition of mitochondrial carbonic anhydrases: implications for cerebral microvascular disease in diabetes. Biochem Biophys Res Commun. 2013;440(2):354–8.
135.
Chauhan SK, Jin Y, Goyal S, Lee HS, Fuchsluger TA, Lee HK, et al. A novel pro-lymphangiogenic function for Th17/IL-17. Blood. 2011;118(17):4630–4.
136.
Tatti O, Gucciardo E, Pekkonen P, Holopainen T, Louhimo R, Repo P, et al. MMP16 mediates a proteolytic switch to promote cell-cell adhesion, collagen alignment, and lymphatic invasion in melanoma. Cancer Res. 2015;75(10):2083–94.
137.
Marin C, Tubita V, Langdon C, Fuentes M, Rojas-Lechuga MJ, Valero A, et al. ACE2 downregulation in olfactory mucosa: eosinophilic rhinosinusitis as COVID-19 protective factor?Allergy. 2021;76(9):2904–7.
138.
Lee SW, Kim SY, Moon SY, Yang JM, Ha EK, Jee HM, et al. Estimating COVID-19 infection and severity risks in patients with chronic rhinosinusitis: a Korean nationwide cohort study. J Allergy Clin Immunol Pract. 2021;9(6):2262–71.e2.
139.
Jian L, Yi W, Zhang N, Wen W, Krysko O, Song WJ, et al. Perspective: COVID-19, implications of nasal diseases and consequences for their management. J Allergy Clin Immunol. 2020;146(1):67–9.
140.
Förster-Ruhrmann U, Szczepek AJ, Bachert C, Olze H. COVID-19 in a patient with severe chronic rhinosinusitis with nasal polyps during therapy with dupilumab. J Allergy Clin Immunol. 2020;146(1):218–20.e2.
141.
Klimek L, Jutel M, Bousquet J, Agache I, Akdis CA, Hox V, et al. Management of patients with chronic rhinosinusitis during the COVID-19 pandemic-An EAACI position paper. Allergy. 2021;76(3):677–88.
142.
Bachert C, Pawankar R, Zhang L, Bunnag C, Fokkens WJ, Hamilos DL, et al. ICON: chronic rhinosinusitis. World Allergy Organ J. 2014;7(1):25.
143.
Lou H, Wang C, Zhang L. Endotype-driven precision medicine in chronic rhinosinusitis. Expert Rev Clin Immunol. 2019;15(11):1171–83.
144.
Wang C, Lou H, Wang X, Wang Y, Fan E, Li Y, et al. Effect of budesonide transnasal nebulization in patients with eosinophilic chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2015;135(4):922–9.e6.
145.
Gurrola J, 2nd, Borish L. Chronic rhinosinusitis: endotypes, biomarkers, and treatment response. J Allergy Clin Immunol. 2017;140(6):1499–508.
146.
Milara J, Morell A, Ballester B, Armengot M, Morcillo E, Cortijo J. MUC4 impairs the anti-inflammatory effects of corticosteroids in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2017;139(3):855–62.e13.
147.
Yan B, Lou H, Wang Y, Li Y, Meng Y, Qi S, et al. Epithelium-derived cystatin SN enhances eosinophil activation and infiltration through IL-5 in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2019;144(2):455–69.
148.
Wu D, Yan B, Wang Y, Zhang L, Wang C. Charcot-Leyden crystal concentration in nasal secretions predicts clinical response to glucocorticoids in patients with chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2019;144(1):345–8.e8.
149.
Andersson DI, Hughes D. Microbiological effects of sublethal levels of antibiotics. Nat Rev Microbiol. 2014;12(7):465–78.
150.
Carlton DA, Beahm DD, Chiu AG. Topical antibiotic therapy in chronic rhinosinusitis: an update. Int Forum Allergy Rhinol. 2019;9(S1):S27–31.
151.
Chapurin N, Wu J, Labby AB, Chandra RK, Chowdhury NI, Turner JH. Current insight into treatment of chronic rhinosinusitis: phenotypes, endotypes, and implications for targeted therapeutics. J Allergy Clin Immunol. 2022;150(1):22–32.
152.
Patel GB, Kern RC, Bernstein JA, Hae-Sim P, Peters AT. Current and future treatments of rhinitis and sinusitis. J Allergy Clin Immunol Pract. 2020;8(5):1522–31.
153.
Miyake MM, Bleier BS. Future topical medications in chronic rhinosinusitis. Int Forum Allergy Rhinol. 2019;9(S1):S32–46.
154.
van der Veen J, Seys SF, Timmermans M, Levie P, Jorissen M, Fokkens WJ, et al. Real-life study showing uncontrolled rhinosinusitis after sinus surgery in a tertiary referral centre. Allergy. 2017;72(2):282–90.
155.
Alanin MC, Laidlaw T, Society TS, Hopkins C. The Burden of Non-steroidal anti-inflammatory exacerbated respiratory disease from the patient's perspective - a qualitative analysis of posts from the Samter's Society. Rhinology. 2020;58(4):333–40.
156.
Seys SF, De Bont S, Fokkens WJ, Bachert C, Alobid I, Bernal-Sprekelsen M, et al. Real-life assessment of chronic rhinosinusitis patients using mobile technology: the mySinusitisCoach project by EUFOREA. Allergy. 2020;75(11):2867–78.
157.
Yancey KL, Li P, Huang LC, Sheng Q, Chandra RK, Chowdhury NI, et al. Longitudinal stability of chronic rhinosinusitis endotypes. Clin Exp Allergy. 2019;49(12):1637–40.
158.
Iqbal IZ, Kao SS, Ooi EH. The role of biologics in chronic rhinosinusitis: a systematic review. Int Forum Allergy Rhinol. 2020;10(2):165–74.
159.
Pauwels B, Jonstam K, Bachert C. Emerging biologics for the treatment of chronic rhinosinusitis. Expert Rev Clin Immunol. 2015;11(3):349–61.
160.
Bachert C, Gevaert P, Hellings P. Biotherapeutics in chronic rhinosinusitis with and without nasal polyps. J Allergy Clin Immunol Pract. 2017;5(6):1512–6.
161.
Manka LA, Wechsler ME. New biologics for allergic diseases. Expert Rev Clin Immunol. 2018;14(4):285–96.
162.
Jeong JS, Kim SR, Cho SH, Lee YC. Endoplasmic reticulum stress and allergic diseases. Curr Allergy Asthma Rep. 2017;17(12):82.
163.
Wang X, Sima Y, Zhao Y, Zhang N, Zheng M, Du K, et al. Endotypes of chronic rhinosinusitis based on inflammatory and remodeling factors. J Allergy Clin Immunol. 2023;151(2):458–68.
164.
Bachert C, Han JK, Desrosiers M, Hellings PW, Amin N, Lee SE, et al. Efficacy and safety of dupilumab in patients with severe chronic rhinosinusitis with nasal polyps (LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52): results from two multicentre, randomised, double-blind, placebo-controlled, parallel-group phase 3 trials. Lancet. 2019;394(10209):1638–50.
165.
Bachert C, Zhang N, Cavaliere C, Weiping W, Gevaert E, Krysko O. Biologics for chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol. 2020;145(3):725–39.
166.
Tanosaki T, Kabata H, Matsusaka M, Miyata J, Masaki K, Mochimaru T, et al. Clinical characteristics of patients with not well-controlled severe asthma in Japan: analysis of the Keio Severe Asthma Research Program in Japanese population (KEIO-SARP) registry. Allergol Int. 2021;70(1):61–7.
167.
Diamant Z, Vijverberg S, Alving K, Bakirtas A, Bjermer L, Custovic A, et al. Toward clinically applicable biomarkers for asthma: an EAACI position paper. Allergy. 2019;74(10):1835–51.
168.
Xie Y, Abel PW, Casale TB, Tu Y. TH17 cells and corticosteroid insensitivity in severe asthma. J Allergy Clin Immunol. 2022;149(2):467–79.
169.
Busse WW, Holgate S, Kerwin E, Chon Y, Feng J, Lin J, et al. Randomized, double-blind, placebo-controlled study of brodalumab, a human anti-IL-17 receptor monoclonal antibody, in moderate to severe asthma. Am J Respir Crit Care Med. 2013;188(11):1294–302.
170.
Chen S, Chen Z, Deng Y, Zha S, Yu L, Li D, et al. Prevention of IL-6 signaling ameliorates toluene diisocyanate-induced steroid-resistant asthma. Allergol Int. 2022;71(1):73–82.
171.
Chu DK, Al-Garawi A, Llop-Guevara A, Pillai RA, Radford K, Shen P, et al. Therapeutic potential of anti-IL-6 therapies for granulocytic airway inflammation in asthma. Allergy Asthma Clin Immunol. 2015;11(1):14.
172.
McDowell PJ, Heaney LG. Different endotypes and phenotypes drive the heterogeneity in severe asthma. Allergy. 2020;75(2):302–10.
173.
Choy DF, Arron JR. Beyond type 2 cytokines in asthma - new insights from old clinical trials. Expert Opin Ther Targets. 2020;24(5):463–75.
174.
Esteban-Gorgojo I, Antolín-Amérigo D, Domínguez-Ortega J, Quirce S. Non-eosinophilic asthma: current perspectives. J Asthma Allergy. 2018;11:267–81.
175.
Nair P, Gaga M, Zervas E, Alagha K, Hargreave FE, O'Byrne PM, et al. Safety and efficacy of a CXCR2 antagonist in patients with severe asthma and sputum neutrophils: a randomized, placebo-controlled clinical trial. Clin Exp Allergy. 2012;42(7):1097–103.
176.
O’Byrne PM, Metev H, Puu M, Richter K, Keen C, Uddin M, et al. Efficacy and safety of a CXCR2 antagonist, AZD5069, in patients with uncontrolled persistent asthma: a randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2016;4(10):797–806.
177.
Gauvreau GM, Sehmi R, Ambrose CS, Griffiths JM. Thymic stromal lymphopoietin: its role and potential as a therapeutic target in asthma. Expert Opin Ther Targets. 2020;24(8):777–92.
178.
Maun HR, Jackman JK, Choy DF, Loyet KM, Staton TL, Jia G, et al. An allosteric anti-tryptase antibody for the treatment of mast cell-mediated severe asthma. Cell. 2019;179(2):417–31.e19.
179.
Jutel M, Torres MJ, Palomares O, Akdis CA, Eiwegger T, Untersmayr E, et al. COVID-19 vaccination in patients receiving Allergen Immunotherapy (AIT) or biologicals-EAACI recommendations. Allergy. 2022;77(8):2313–36.
180.
Hsu J, Avila PC, Kern RC, Hayes MG, Schleimer RP, Pinto JM. Genetics of chronic rhinosinusitis: state of the field and directions forward. J Allergy Clin Immunol. 2013;131(4):977–93.e5.
181.
Stevens WW, Ocampo CJ, Berdnikovs S, Sakashita M, Mahdavinia M, Suh L, et al. Cytokines in chronic rhinosinusitis. Role in eosinophilia and aspirin-exacerbated respiratory disease. Am J Respir Crit Care Med. 2015;192(6):682–94.
182.
Hong SN, Kim JK, Kim JA, Cha H, Kim JY, Lim HS, et al. Viral stimulation modulates endotype-related ACE2 expression in eosinophilic chronic rhinosinusitis. Rhinology. 2021;59(5):460–9.

Additional information

Edited by: H.-U. Simon, Bern.