Next Article in Journal
Current Surgical Techniques in the Treatment of Adult Developmental Dysplasia of the Hip
Previous Article in Journal
Spanish Cross-Cultural Adaptation of the Australian Pelvic Floor Questionnaire
Previous Article in Special Issue
Treatment Results of Endoscopic Transnasal Orbital Decompression for Graves’ Orbitopathy—A Single-Center Retrospective Analysis in 28 Orbits of 16 Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

A Multidisciplinary Approach for Type 2 Allergic Diseases: What Do Biologics Teach Us?

by
Mauro Maniscalco
1,2,*,
Aikaterini Detoraki
3,
Giovanni Sarnelli
1,
Maria Nolano
4,
Amato De Paulis
3,
Giuseppe Spadaro
3 and
Elena Cantone
4,* on behalf of PATH-2 TASK FORCE
1
Department of Clinical Medicine and Surgery, Federico II University, 80138 Naples, Italy
2
Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
3
Department of Translational Medicine, Federico II University, 80138 Naples, Italy
4
Department of Neuroscience, Reproductive and Odontostomatological Sciences-ENT Section, University of Naples Federico II, 80131 Naples, Italy
*
Authors to whom correspondence should be addressed.
J. Pers. Med. 2023, 13(6), 941; https://doi.org/10.3390/jpm13060941
Submission received: 27 May 2023 / Accepted: 29 May 2023 / Published: 1 June 2023
(This article belongs to the Special Issue The Challenges and Prospects in Diagnostics of Otolaryngology)
Patients with atopic/allergic disorders, including atopic dermatitis (AD), allergic rhino-conjunctivitis (AR), chronic rhinosinusitis with/without nasal polyps (CRSwNP/CRSsNP), bronchial asthma, food allergy, and eosinophilic esophagitis (EoE), often share a common genetic background, a type Th2 polarized immune response, and several environmental factors [1,2].
Th2 immunity has evolved to ensure the integrity of the epithelial barrier and to protect against helminths [3]. Although the pathways underlying Th2-driven inflammation can differ and are related to age and ethnicity, they can share some common mediators [1]. The alarmins, namely, thymic stromal lymphopoietin (TSLP), interleukin (IL)-25 and IL-33, are released from injured epidermal and epithelial cells and play pivotal roles in Th2 cell development [4]. Other cytokines, including IL-4, IL-5, IL-9, IL-13, and IL-31, are also relevant in orchestrating a Th2-type immune response [3]. Naïve CD4+ T cells differentiate into Th2 cells under the influence of IL-4, which also stimulates isotype class switching of B-cells in order to produce immunoglobulin E (IgE) [5]. The additional signaling provided by IL-5 and IL-9 leads to eosinophil and mast cell recruitment to the inflammatory site, while IL-13 induces goblet cell hyperplasia, mucus hypersecretion, and airway hyperresponsiveness [6].
Atopic/allergic disorders affect different body systems and, in some cases, coexist simultaneously in the same patient. Conversely, in other cases, there is a progressive and predictable succession in the involvement of other systems (atopic march), with the progressive onset of AD, food allergy, AR, and allergic asthma [7]. Accordingly, asthma is diagnosed in up to 65% and up to 49.8% of patients with CRSwNP and AD, respectively [8]. Furthermore, some data seem to indicate the association of AD with the development of CRSwNP [9]. Asthma and EoE also frequently coexist and are both characterized by eosinophilic inflammation of the mucosa and submucosa [10]. Finally, recent studies have reported AD prevalence rates of between 7% and 55% among EoE patients [11].
When coexisting in the same patient, the severity of asthma, CRSwNP and AD is greater. In such instances, each of them should be treated concurrently to obtain the optimal treatment results [8,12].
The presence of several allergic disorders in the same individuals suggests that some subgroups of allergic diseases share the same subtype of health condition, namely the “endotype” variety, especially the Th2-driven endotype. Indeed, Th2-high inflammation is a feature of allergic asthma, AR, and CRSwNP [13]. A Th2-high, as opposed to Th2-low, immune response has been suggested for the endotyping of AD [13,14,15] and EoE [14,15]. A Th2-high profile has been identified in some types of food allergy and oral allergy syndrome [16].
Recently, new data have indicated that Th2 effector cells are not homogeneous. Indeed, the scientific community has identified a subgroup of pathogenic Th2 cells (Tpath2), which express large amounts of chemoattractant receptor-homologous molecules that are expressed on Th2 cells (CRTH2), CD49d, and CD161. Additionally, researchers have come to identify alarmins, which also produce large amounts of IL-5 and IL-9 [17]. In this regard, IL-33 seems to be required to obtain the optimal effector function of human Tpath2 cells [17], which exacerbate eosinophilic inflammation [18] through the production of a large quantity of IL-5 [19]. The identification of Tpath2 cells has improved the understanding of the pathogenesis of atopic/allergic diseases [20].
In line with a growing body of evidence, if some Th2-driven pathophysiological mechanisms are shared across multiple atopic/allergic disorders, it is possible to hypothesize that, by treating a single clinical condition in an individual with multiple Th2 diseases, a simultaneous improvement can be obtained. Recently the increasing availability of new biological drugs that possess the capacity to interfere, whether directly and indirectly, with the action of single cytokines has allowed effective treatment for several Th2 diseases and related comorbidities.
Omalizumab, a monoclonal antibody targeting the high-affinity receptor binding site of IgE, has demonstrated clinical efficacy in subjects with CRSwNP [21], food allergies [22], severe allergic asthma [23], and moderate-to-severe chronic idiopathic urticaria [24]. Although IgE seems to be implicated in AD, its targeting is clinically ineffective [25], mepolizumab, a humanized monoclonal antibody capable of directly binding free IL-5, has shown clinical efficacy in treating patients with CRSwNP, although its use is ineffective in patients with AD [26]. By contrast, the anti-IL-5Rα Benralizumab has shown remarkable therapeutic effects in various types of eosinophilic diseases, including eosinophilic CRS and asthma [27]. Tralokinumab, a monoclonal antibody inhibiting IL-13, is relatively effective in patients with AD, but not in asthmatic subjects, suggesting that IL-13 is clinically relevant in AD but that its role in the pathophysiology of asthma is smaller [3]. Dupilumab, a humanized monoclonal antibody inhibiting both IL-4 and IL-13 signaling, has been approved for patients with Th2-driven inflammatory disorders, including AD [28], asthma [29] and CRSwNP [30], supporting the key pathogenic role of IL-4 and IL-13 in these conditions. Similar and encouraging results are also emerging in the management of EoE [31]. Finally, the use of Tezepelumab, which targets TSLP, is clinically useful in asthma but has not produced clinically relevant results in AD [3].
The use of biologics to treat severe uncontrolled CRSwNP is quite recent compared to their use in cases of asthma and atopic dermatitis. In fact, there are still few real-life studies, and those in existence are limited to discussions of dupilumab [32]. Furthermore, there are no comparative studies evaluating the indication of one biologic agent versus another for the treatment of different CRS endotypes.
The clinical results obtained from biological drugs directed against single receptors and/or mediators in Th2 inflammatory diseases have allowed researchers to better define their pathogenetic role in each of them. Conversely, and on the other hand, has stimulated the research inro additional biologics directed against other relevant cytokines. However, although the positive clinical response to biologics confirms the association between Th2-driven diseases, no biomarker with high specificity can anticipate the therapeutic response in either single or multiple Th2 conditions.
Although Th2-driven immune responses are multifaceted, the use of biologics has allowed significant therapeutic advances and has clarified that the underlying pathophysiological mechanisms are substantially limited in number. However, many issues still remain to be adequately addressed.
In the context of precision medicine and individualized approaches, future research should focus on the identification of biomarkers that can accurately indicate the specific endotype for a specific patient and, therefore, the most suitable biological drug. Furthermore, it is important to establish the correct timing for the administration of biologics in the clinical course of a Th2-driven disorder. This matter is far from insignificant. In fact, considering that allergic/atopic disorders often coexist and/or follow each other chronologically, it is essential to know whether their early use for the treatment of one of these conditions possesses the capacity to prevent “atopic march”.

Author Contributions

Conceptualization, M.M. and E.C.; software, M.M., M.N. and E.C.; writing—original draft preparation, M.M., A.D., G.S. (Giovanni Sarnelli), M.N., G.S. (Giuseppe Spadaro), A.D.P. and E.C.; writing—review and editing, M.M. and E.C.; visualization, A.D.; supervision, M.M., G.S.(Giovanni Sarnelli), A.D.P. and E.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by the “Ricerca Corrente” funding scheme of the Ministry of Health, Italy.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hassoun, D.; Malard, O.; Barbarot, S.; Magnan, A.; Colas, L. Type 2 immunity-driven diseases: Towards a multidisciplinary approach. Clin. Exp. Allergy 2021, 51, 1538–1552. [Google Scholar] [CrossRef] [PubMed]
  2. Maggi, E.; Parronchi, P.; Azzarone, B.G.; Moretta, L. A pathogenic integrated view explaining the different endotypes of asthma and allergic disorders. Allergy 2022, 77, 3267–3292. [Google Scholar] [CrossRef] [PubMed]
  3. Haddad, E.B.; Cyr, S.L.; Arima, K.; McDonald, R.A.; Levit, N.A.; Nestle, F.O. Current and Emerging Strategies to Inhibit Type 2 Inflammation in Atopic Dermatitis. Dermatol. Ther. 2022, 12, 1501–1533. [Google Scholar] [CrossRef]
  4. Saenz, S.A.; Taylor, B.C.; Artis, D. Welcome to the neighborhood: Epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites. Immunol. Rev. 2008, 226, 172–190. [Google Scholar] [CrossRef] [PubMed]
  5. Nakayama, T.; Hirahara, K.; Onodera, A.; Endo, Y.; Hosokawa, H.; Shinoda, K.; Tumes, D.J.; Okamoto, Y. Th2 Cells in Health and Disease. Annu. Rev. Immunol. 2017, 35, 53–84. [Google Scholar] [CrossRef]
  6. Caminati, M.; Pham, D.L.; Bagnasco, D.; Canonica, G.W. Type 2 immunity in asthma. World Allergy Organ. J. 2018, 11, 13. [Google Scholar] [CrossRef]
  7. Maiello, N.; Giannetti, A.; Ricci, G.; Cinicola, B.; Carello, R.; Indolfi, C.; Caffarelli, C.; Marseglia, A.; Calvani, M.; Miraglia Del Giudice, M.; et al. Atopic dermatitis and atopic march: Which link? Acta Biomed. 2021, 92, e2021525. [Google Scholar]
  8. De Prins, L.; Raap, U.; Mueller, T.; Schmid-Grendelmeier, P.; Haase, C.H.; Backer, V.; Fokkens, W.; Benoist, L.B.; Prokopakis, E.; Hopkins, C.; et al. White Paper on European Patient Needs and Suggestions on Chronic Type 2 Inflammation of Airways and Skin by EUFOREA. Front. Allergy 2022, 3, 889221. [Google Scholar] [CrossRef]
  9. Chandra, R.K.; Lin, D.; Tan, B.; Tudor, R.S.; Conley, D.B.; Peters, A.T.; Grammer, L.C.; Schleimer, R.P.; Kern, R.C. Chronic rhinosinusitis in the setting of other chronic inflammatory diseases. Am. J. Otolaryngol. 2011, 32, 388–391. [Google Scholar] [CrossRef]
  10. Virchow, J.C. Eosinophilic esophagitis: Asthma of the esophagus? Dig. Dis. 2014, 32, 54–60. [Google Scholar] [CrossRef]
  11. McCormick, J.P.; Lee, J.T. Insights into the Implications of Coexisting Type 2 Inflammatory Diseases. J. Inflamm. Res. 2021, 14, 4259–4266. [Google Scholar] [CrossRef] [PubMed]
  12. Van Bulck, P.; Cools, L.; Soumya, M.S.; Nyembue, D.T.; Kabobo, P.; Zhang, L.; Scadding, G.K.; Toskala, E.; Fokkens, W.J.; Steelant, B.; et al. A multicenter real-life study on the multiple reasons for uncontrolled allergic rhinitis. Int. Forum. Allergy Rhinol. 2021, 11, 1452–1460. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, M.; Bu, X.; Luan, G.; Lin, L.; Wang, Y.; Jin, J.; Zhang, L.; Wang, C. Distinct type 2-high inflammation associated molecular signatures of chronic rhinosinusitis with nasal polyps with comorbid asthma. Clin. Transl. Allergy 2020, 10, 26. [Google Scholar] [CrossRef] [PubMed]
  14. Agache, I.; Akdis, C.A. Endotypes of allergic diseases and asthma: An important step in building blocks for the future of precision medicine. Allergol. Int. 2016, 65, 243–252. [Google Scholar] [CrossRef]
  15. Ruffner, M.A.; Cianferoni, A. Phenotypes and endotypes in eosinophilic esophagitis. Ann. Allergy Asthma Immunol. 2020, 124, 233–239. [Google Scholar] [CrossRef]
  16. Chinthrajah, R.S.; Hernandez, J.D.; Boyd, S.D.; Galli, S.J.; Nadeau, K.C. Molecular and cellular mechanisms of food allergy and food tolerance. J. Allergy Clin. Immunol. 2016, 137, 984–997. [Google Scholar] [CrossRef]
  17. Calise, J.; Garabatos, N.; Bajzik, V.; Farrington, M.; Robinson, D.; Jeong, D.; Londei, M.; Wambre, E. Optimal human pathogenic TH2 cell effector function requires local epithelial cytokine signaling. J. Allergy Clin. Immunol. 2021, 148, 867–875.e4. [Google Scholar] [CrossRef]
  18. Mitson-Salazar, A.; Yin, Y.; Wansley, D.L.; Young, M.; Bolan, H.; Arceo, S.; Ho, N.; Koh, C.; Milner, J.D.; Stone, K.D.; et al. Hematopoietic prostaglandin D synthase defines a proeosinophilic pathogenic effector human T(H)2 cell subpopulation with enhanced function. J. Allergy Clin. Immunol. 2016, 137, 907–918.e9. [Google Scholar] [CrossRef]
  19. Hirahara, K.; Shinoda, K.; Morimoto, Y.; Kiuchi, M.; Aoki, A.; Kumagai, J.; Kokubo, K.; Nakayama, T. Immune Cell-Epithelial/Mesenchymal Interaction Contributing to Allergic Airway Inflammation Associated Pathology. Front. Immunol. 2019, 10, 570. [Google Scholar] [CrossRef]
  20. Kokubo, K.; Onodera, A.; Kiuchi, M.; Tsuji, K.; Hirahara, K.; Nakayama, T. Conventional and pathogenic Th2 cells in inflammation, tissue repair, and fibrosis. Front. Immunol. 2022, 13, 945063. [Google Scholar] [CrossRef]
  21. Gevaert, P.; Omachi, T.A.; Corren, J.; Mullol, J.; Han, J.; Lee, S.E.; Kaufman, D.; Ligueros-Saylan, M.; Howard, M.; Zhu, R.; et al. Efficacy and safety of omalizumab in nasal polyposis: 2 randomized phase 3 trials. J. Allergy Clin. Immunol. 2020, 146, 595–605. [Google Scholar] [CrossRef]
  22. Fiocchi, A.; Artesani, M.C.; Riccardi, C.; Mennini, M.; Pecora, V.; Fierro, V.; Calandrelli, V.; Dahdah, L.; Valluzzi, R.L. Impact of Omalizumab on Food Allergy in Patients Treated for Asthma: A Real-Life Study. J. Allergy Clin. Immunol. Pract. 2019, 7, 1901–1909.e5. [Google Scholar] [CrossRef] [PubMed]
  23. Holgate, S.T.; Chuchalin, A.G.; Hebert, J.; Lotvall, J.; Persson, G.B.; Chung, K.F.; Bousquet, J.; Kerstjens, H.A.; Fox, H.; Thirlwell, J.; et al. Efficacy and safety of a recombinant anti-immunoglobulin E antibody (omalizumab) in severe allergic asthma. Clin. Exp. Allergy 2004, 34, 632–638. [Google Scholar] [CrossRef] [PubMed]
  24. Maurer, M.; Rosen, K.; Hsieh, H.J.; Saini, S.; Grattan, C.; Gimenez-Arnau, A.; Agarwal, S.; Doyle, R.; Canvin, J.; Kaplan, A.; et al. Omalizumab for the treatment of chronic idiopathic or spontaneous urticaria. N. Engl. J. Med. 2013, 368, 924–935. [Google Scholar] [CrossRef] [PubMed]
  25. Siegels, D.; Heratizadeh, A.; Abraham, S.; Binnmyr, J.; Brockow, K.; Irvine, A.D.; Halken, S.; Mortz, C.G.; Flohr, C.; Schmid-Grendelmeier, P.; et al. Systemic treatments in the management of atopic dermatitis: A systematic review and meta-analysis. Allergy 2021, 76, 1053–1076. [Google Scholar] [CrossRef]
  26. Oldhoff, J.M.; Darsow, U.; Werfel, T.; Katzer, K.; Wulf, A.; Laifaoui, J.; Hijnen, D.J.; Plotz, S.; Knol, E.F.; Kapp, A.; et al. Anti-IL-5 recombinant humanized monoclonal antibody (mepolizumab) for the treatment of atopic dermatitis. Allergy 2005, 60, 693–696. [Google Scholar] [CrossRef]
  27. Nagase, H.; Ueki, S.; Fujieda, S. The roles of IL-5 and anti-IL-5 treatment in eosinophilic diseases: Asthma, eosinophilic granulomatosis with polyangiitis, and eosinophilic chronic rhinosinusitis. Allergol. Int. 2020, 69, 178–186. [Google Scholar] [CrossRef]
  28. Simpson, E.L.; Bieber, T.; Guttman-Yassky, E.; Beck, L.A.; Blauvelt, A.; Cork, M.J.; Silverberg, J.I.; Deleuran, M.; Kataoka, Y.; Lacour, J.P.; et al. Two Phase 3 Trials of Dupilumab versus Placebo in Atopic Dermatitis. N. Engl. J. Med. 2016, 375, 2335–2348. [Google Scholar] [CrossRef] [PubMed]
  29. Castro, M.; Corren, J.; Pavord, I.D.; Maspero, J.; Wenzel, S.; Rabe, K.F.; Busse, W.W.; Ford, L.; Sher, L.; FitzGerald, J.M.; et al. Dupilumab Efficacy and Safety in Moderate-to-Severe Uncontrolled Asthma. N. Engl. J. Med. 2018, 378, 2486–2496. [Google Scholar] [CrossRef] [PubMed]
  30. Bachert, C.; Han, J.K.; Desrosiers, M.; Hellings, P.W.; Amin, N.; Lee, S.E.; Mullol, J.; Greos, L.S.; Bosso, J.V.; Laidlaw, T.M.; 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, 1638–1650. [Google Scholar] [CrossRef] [PubMed]
  31. Hirano, I.; Dellon, E.S.; Hamilton, J.D.; Collins, M.H.; Peterson, K.; Chehade, M.; Schoepfer, A.M.; Safroneeva, E.; Rothenberg, M.E.; Falk, G.W.; et al. Efficacy of Dupilumab in a Phase 2 Randomized Trial of Adults with Active Eosinophilic Esophagitis. Gastroenterology 2020, 158, 111–122.e10. [Google Scholar] [CrossRef] [PubMed]
  32. Gevaert, P.; Lee, S.E.; Settipane, R.A.; Wagenmann, M.; Msihid, J.; Siddiqui, S.; Nash, S.; Jacob-Nara, J.A.; Khan, A.H.; Kamat, S.; et al. Dupilumab provides early and durable improvement of symptoms in patients with chronic rhinosinusitis with nasal polyps. Clin. Transl. Immunol. 2023, 12, e1433. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Maniscalco, M.; Detoraki, A.; Sarnelli, G.; Nolano, M.; De Paulis, A.; Spadaro, G.; Cantone, E., on behalf of PATH-2 TASK FORCE. A Multidisciplinary Approach for Type 2 Allergic Diseases: What Do Biologics Teach Us? J. Pers. Med. 2023, 13, 941. https://doi.org/10.3390/jpm13060941

AMA Style

Maniscalco M, Detoraki A, Sarnelli G, Nolano M, De Paulis A, Spadaro G, Cantone E on behalf of PATH-2 TASK FORCE. A Multidisciplinary Approach for Type 2 Allergic Diseases: What Do Biologics Teach Us? Journal of Personalized Medicine. 2023; 13(6):941. https://doi.org/10.3390/jpm13060941

Chicago/Turabian Style

Maniscalco, Mauro, Aikaterini Detoraki, Giovanni Sarnelli, Maria Nolano, Amato De Paulis, Giuseppe Spadaro, and Elena Cantone on behalf of PATH-2 TASK FORCE. 2023. "A Multidisciplinary Approach for Type 2 Allergic Diseases: What Do Biologics Teach Us?" Journal of Personalized Medicine 13, no. 6: 941. https://doi.org/10.3390/jpm13060941

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop