Asthma in the Precision Medicine Era: Biologics and Probiotics
Abstract
:1. Pathophysiology of Asthma
2. Asthma Endotypes
3. Add-On Therapy
4. FDA-Approved Monoclonal Antibodies
5. Biological Therapies under Clinical Trials
6. Microbiota and Allergic Asthma
6.1. Hygiene Hypothesis
6.2. Gut–Lung Axis and Microbial Mechanisms
6.3. Relation of Microbial Taxa and Asthma
6.4. Contradictory Data—Take Ruminococcus gnavus as an Example
6.5. Therapeutic Potential of Microbiota
6.5.1. Probiotics
6.5.2. From Microbial Endotypes to Asthma Endotyping and Precision Medicine for Asthma
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Vos, T.; Abajobir, A.A.; Abate, K.H.; Abbafati, C.; Abbas, K.M.; Abd-Allah, F.; Abdulkader, R.S.; Abdulle, A.M.; Abebo, T.A.; Abera, S.F.; et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet 2017, 390, 1211–1259. [Google Scholar] [CrossRef] [Green Version]
- Pearce, N.; Aït-Khaled, N.; Beasley, R.; Mallol, J.; Keil, U.; Mitchell, E.; Robertson, C. Worldwide trends in the prevalence of asthma symptoms: Phase III of the International Study of Asthma and Allergies in Childhood (ISAAC). Thorax 2007, 62, 758–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lai, C.K.W.; Beasley, R.; Crane, J.; Foliaki, S.; Shah, J.; Weiland, S. Global variation in the prevalence and severity of asthma symptoms: Phase Three of the International Study of Asthma and Allergies in Childhood (ISAAC). Thorax 2009, 64, 476–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lambrecht, B.N.; Hammad, H. The immunology of asthma. Nat. Immunol. 2015, 16, 45–56. [Google Scholar] [CrossRef]
- Poulsen, L.K.; Hummelshoj, L. Triggers of IgE class switching and allergy development. Ann. Med. 2007, 39, 440–456. [Google Scholar] [CrossRef]
- Fulkerson, P.C.; Schollaert, K.L.; Bouffi, C.; Rothenberg, M.E. IL-5 triggers a cooperative cytokine network that promotes eosinophil precursor maturation. J. Immunol. 2014, 193, 4043–4052. [Google Scholar] [CrossRef] [Green Version]
- Roufosse, F. Targeting the Interleukin-5 Pathway for Treatment of Eosinophilic Conditions Other than Asthma. Front. Med. 2018, 5, 49. [Google Scholar] [CrossRef] [Green Version]
- Chakraborty, S.; Kubatzky, K.F.; Mitra, D.K. An Update on Interleukin-9: From Its Cellular Source and Signal Transduction to Its Role in Immunopathogenesis. Int. J. Mol. Sci. 2019, 20, 2113. [Google Scholar] [CrossRef] [Green Version]
- Angkasekwinai, P.; Dong, C. IL-9-producing T cells: Potential players in allergy and cancer. Nat. Rev. Immunol. 2021, 21, 37–48. [Google Scholar] [CrossRef]
- Ingram, J.L.; Kraft, M. IL-13 in asthma and allergic disease: Asthma phenotypes and targeted therapies. J. Allergy Clin. Immunol. 2012, 130, 829–842. [Google Scholar] [CrossRef]
- Marone, G.; Granata, F.; Pucino, V.; Pecoraro, A.; Heffler, E.; Loffredo, S.; Scadding, G.W.; Varricchi, G. The Intriguing Role of Interleukin 13 in the Pathophysiology of Asthma. Front. Pharmacol. 2019, 10, 1387. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Z.; Homer, R.J.; Wang, Z.; Chen, Q.; Geba, G.P.; Wang, J.; Zhang, Y.; Elias, J.A. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J. Clin. Investig. 1999, 103, 779–788. [Google Scholar] [CrossRef] [Green Version]
- Lambrecht, B.N.; Hammad, H.; Fahy, J.V. The Cytokines of Asthma. Immunity 2019, 50, 975–991. [Google Scholar] [CrossRef]
- Peters, S.P. Asthma phenotypes: Nonallergic (intrinsic) asthma. J. Allergy Clin. Immunol. Pract. 2014, 2, 650–652. [Google Scholar] [CrossRef]
- Corren, J. Asthma phenotypes and endotypes: An evolving paradigm for classification. Discov. Med. 2013, 15, 243–249. [Google Scholar]
- James, D.R.; Lyttle, M.D. British guideline on the management of asthma: SIGN Clinical Guideline 141, 2014. Arch. Dis. Child. Educ. Pract. Ed. 2016, 101, 319–322. [Google Scholar] [CrossRef] [Green Version]
- Robinson, D.; Humbert, M.; Buhl, R.; Cruz, A.A.; Inoue, H.; Korom, S.; Hanania, N.A.; Nair, P. Revisiting Type 2-high and Type 2-low airway inflammation in asthma: Current knowledge and therapeutic implications. Clin. Exp. Allergy 2017, 47, 161–175. [Google Scholar] [CrossRef]
- Baines, K.J.; Simpson, J.L.; Wood, L.G.; Scott, R.J.; Gibson, P.G. Transcriptional phenotypes of asthma defined by gene expression profiling of induced sputum samples. J. Allergy Clin. Immunol. 2011, 127, 153–160, 160.e151–159. [Google Scholar] [CrossRef]
- D’Silva, L.; Hassan, N.; Wang, H.Y.; Kjarsgaard, M.; Efthimiadis, A.; Hargreave, F.E.; Nair, P. Heterogeneity of bronchitis in airway diseases in tertiary care clinical practice. Can. Respir. J. 2011, 18, 144–148. [Google Scholar] [CrossRef] [Green Version]
- Simpson, J.L.; Scott, R.; Boyle, M.J.; Gibson, P.G. Inflammatory subtypes in asthma: Assessment and identification using induced sputum. Respirology 2006, 11, 54–61. [Google Scholar] [CrossRef]
- Crimi, E.; Spanevello, A.; Neri, M.; Ind, P.W.; Rossi, G.A.; Brusasco, V. Dissociation between airway inflammation and airway hyperresponsiveness in allergic asthma. Am. J. Respir. Crit. Care Med. 1998, 157, 4–9. [Google Scholar] [CrossRef] [PubMed]
- Wilson, N.M.; James, A.; Uasuf, C.; Payne, D.N.; Hablas, H.; Agrofioti, C.; Bush, A. Asthma severity and inflammation markers in children. Pediatr. Allergy Immunol. 2001, 12, 125–132. [Google Scholar] [CrossRef]
- Djukanovic, R.; Wilson, S.J.; Kraft, M.; Jarjour, N.N.; Steel, M.; Chung, K.F.; Bao, W.; Fowler-Taylor, A.; Matthews, J.; Busse, W.W.; et al. Effects of treatment with anti-immunoglobulin E antibody omalizumab on airway inflammation in allergic asthma. Am. J. Respir. Crit. Care Med. 2004, 170, 583–593. [Google Scholar] [CrossRef] [PubMed]
- Leckie, M.J.; Brinke, A.t.; Khan, J.; Diamant, Z.; O’Connor, B.J.; Walls, C.M.; Mathur, A.K.; Cowley, H.C.; Chung, K.F.; Djukanovic, R.; et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsìveness, and the late asthmatic response. Lancet 2000, 356, 2144–2148. [Google Scholar] [CrossRef]
- Flood-Page, P.T.; Menzies-Gow, A.N.; Kay, A.B.; Robinson, D.S. Eosinophil’s role remains uncertain as anti–Interleukin-5 only partially depletes numbers in asthmatic airway. Am. J. Respir. Crit. Care Med. 2003, 167, 199–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berry, M.A.; Hargadon, B.; Shelley, M.; Parker, D.; Shaw, D.E.; Green, R.H.; Bradding, P.; Brightling, C.E.; Wardlaw, A.J.; Pavord, I.D. Evidence of a role of tumor necrosis factor alpha in refractory asthma. N. Engl. J. Med. 2006, 354, 697–708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Price, D.B.; Rigazio, A.; Campbell, J.D.; Bleecker, E.R.; Corrigan, C.J.; Thomas, M.; Wenzel, S.E.; Wilson, A.M.; Small, M.B.; Gopalan, G.; et al. Blood eosinophil count and prospective annual asthma disease burden: A UK cohort study. Lancet Respir. Med. 2015, 3, 849–858. [Google Scholar] [CrossRef]
- Price, D.; Wilson, A.M.; Chisholm, A.; Rigazio, A.; Burden, A.; Thomas, M.; King, C. Predicting frequent asthma exacerbations using blood eosinophil count and other patient data routinely available in clinical practice. J. Asthma Allergy 2016, 9, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Woodruff, P.G.; Modrek, B.; Choy, D.F.; Jia, G.; Abbas, A.R.; Ellwanger, A.; Arron, J.R.; Koth, L.L.; Fahy, J.V. T-helper type 2–driven inflammation defines major subphenotypes of asthma. Am. J. Respir. Crit. Care Med. 2009, 180, 388–395. [Google Scholar] [CrossRef]
- Fahy, J.V. Type 2 inflammation in asthma—present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef]
- Kuruvilla, M.E.; Lee, F.E.-H.; Lee, G.B. Understanding asthma phenotypes, endotypes, and mechanisms of disease. Clin. Rev. Allergy Immunol. 2019, 56, 219–233. [Google Scholar] [CrossRef]
- Hammad, H.; Lambrecht, B.N. The basic immunology of asthma. Cell 2021, 184, 1469–1485. [Google Scholar] [CrossRef]
- Stokes, J.R.; Casale, T.B. Characterization of asthma endotypes: Implications for therapy. Ann. Allergy Asthma Immunol. Off. Publ. Am. Coll. Allergy Asthma Immunol. 2016, 117, 121–125. [Google Scholar] [CrossRef]
- Boulet, L.P.; Reddel, H.K.; Bateman, E.; Pedersen, S.; FitzGerald, J.M.; O’Byrne, P.M. The Global Initiative for Asthma (GINA): 25 years later. Eur. Respir. J. 2019, 54. [Google Scholar] [CrossRef]
- Wenzel, S.E. Asthma: Defining of the persistent adult phenotypes. Lancet 2006, 368, 804–813. [Google Scholar] [CrossRef]
- Smith, A.D.; Cowan, J.O.; Brassett, K.P.; Herbison, G.P.; Taylor, D.R. Use of exhaled nitric oxide measurements to guide treatment in chronic asthma. N. Engl. J. Med. 2005, 352, 2163–2173. [Google Scholar] [CrossRef] [Green Version]
- Pifferi, M.; Bush, A.; Pioggia, G.; Di Cicco, M.; Chinellato, I.; Bodini, A.; Macchia, P.; Boner, A.L. Monitoring asthma control in children with allergies by soft computing of lung function and exhaled nitric oxide. Chest 2011, 139, 319–327. [Google Scholar] [CrossRef]
- Dweik, R.A.; Sorkness, R.L.; Wenzel, S.; Hammel, J.; Curran-Everett, D.; Comhair, S.A.; Bleecker, E.; Busse, W.; Calhoun, W.J.; Castro, M.; et al. Use of exhaled nitric oxide measurement to identify a reactive, at-risk phenotype among patients with asthma. Am. J. Respir. Crit. Care Med. 2010, 181, 1033–1041. [Google Scholar] [CrossRef] [Green Version]
- Kuo, C.-H.S.; Pavlidis, S.; Loza, M.; Baribaud, F.; Rowe, A.; Pandis, I.; Hoda, U.; Rossios, C.; Sousa, A.; Wilson, S.J.; et al. A Transcriptome-driven analysis of epithelial brushings and bronchial biopsies to define asthma phenotypes in U-BIOPRED. Am. J. Respir. Crit. Care Med. 2017, 195, 443–455. [Google Scholar] [CrossRef] [Green Version]
- Sur, S.; Crotty, T.B.; Kephart, G.M.; Hyma, B.A.; Colby, T.V.; Reed, C.E.; Hunt, L.W.; Gleich, G.J. Sudden-onset fatal asthma. A distinct entity with few eosinophils and relatively more neutrophils in the airway submucosa? Am. Rev. Respir. Dis. 1993, 148, 713–719. [Google Scholar] [CrossRef]
- Wenzel, S.E.; Szefler, S.J.; Leung, D.Y.; Sloan, S.I.; Rex, M.D.; Martin, R.J. Bronchoscopic evaluation of severe asthma. Persistent inflammation associated with high dose glucocorticoids. Am. J. Respir. Crit. Care Med. 1997, 156, 737–743. [Google Scholar] [CrossRef] [PubMed]
- Huang, M.-T.; Chen, Y.-L.; Lien, C.-I.; Liu, W.-L.; Hsu, L.-C.; Yagita, H.; Chiang, B.-L. Notch ligand DLL4 alleviates allergic airway inflammation via induction of a homeostatic regulatory pathway. Sci. Rep. 2017, 7, 43535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, M.-T.; Chiu, C.-J.; Chiang, B.-L. Multi-faceted Notch in allergic airway inflammation. Int. J. Mol. Sci. 2019, 20, 3508. [Google Scholar] [CrossRef] [Green Version]
- Raita, Y.; Camargo, C.A.; Bochkov, Y.A.; Celedón, J.C.; Gern, J.E.; Mansbach, J.M.; Rhee, E.P.; Freishtat, R.J.; Hasegawa, K. Integrated-omics endotyping of infants with rhinovirus bronchiolitis and risk of childhood asthma. J. Allergy Clin. Immunol. 2020. [Google Scholar] [CrossRef] [PubMed]
- Tyler, S.R.; Bunyavanich, S. Leveraging-omics for asthma endotyping. J. Allergy Clin. Immunol. 2019, 144, 13–23. [Google Scholar] [CrossRef] [PubMed]
- Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention; Global Initiative for Asthma—GINA: Fontana, WI, USA, 2020. [Google Scholar]
- Wenzel, S.E.; Busse, W.W. Severe asthma: Lessons from the Severe Asthma Research Program. J. Allergy Clin. Immunol. 2007, 119, 14–21. [Google Scholar] [CrossRef] [PubMed]
- Allegra, L.; Cremonesi, G.; Girbino, G.; Ingrassia, E.; Marsico, S.; Nicolini, G.; Terzano, C. Real-life prospective study on asthma control in Italy: Cross-sectional phase results. Respir. Med. 2012, 106, 205–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beasley, R.; Harper, J.; Bird, G.; Maijers, I.; Weatherall, M.; Pavord, I.D. Inhaled Corticosteroid Therapy in Adult Asthma. Time for a New Therapeutic Dose Terminology. Am. J. Respir. Crit. Care Med. 2019, 199, 1471–1477. [Google Scholar] [CrossRef]
- Jackson, D.J.; Bacharier, L.B.; Mauger, D.T.; Boehmer, S.; Beigelman, A.; Chmiel, J.F.; Fitzpatrick, A.M.; Gaffin, J.M.; Morgan, W.J.; Peters, S.P.; et al. Quintupling Inhaled Glucocorticoids to Prevent Childhood Asthma Exacerbations. N. Engl. J. Med. 2018, 378, 891–901. [Google Scholar] [CrossRef]
- Lemanske, R.F., Jr.; Mauger, D.T.; Sorkness, C.A.; Jackson, D.J.; Boehmer, S.J.; Martinez, F.D.; Strunk, R.C.; Szefler, S.J.; Zeiger, R.S.; Bacharier, L.B.; et al. Step-up therapy for children with uncontrolled asthma receiving inhaled corticosteroids. N. Engl. J. Med. 2010, 362, 975–985. [Google Scholar] [CrossRef] [Green Version]
- Schulman, E.S. Development of a monoclonal anti-immunoglobulin E antibody (omalizumab) for the treatment of allergic respiratory disorders. Am. J. Respir. Crit. Care Med. 2001, 164, S6–S11. [Google Scholar] [CrossRef]
- Chang, T.W.; Wu, P.C.; Hsu, C.L.; Hung, A.F. Anti-IgE antibodies for the treatment of IgE-mediated allergic diseases. Adv. Immunol. 2007, 93, 63–119. [Google Scholar] [CrossRef]
- Johansson, M.W. Eosinophil Activation Status in Separate Compartments and Association with Asthma. Front. Med. 2017, 4, 75. [Google Scholar] [CrossRef]
- Walsh, G.M. Reslizumab, a humanized anti-IL-5 mAb for the treatment of eosinophil-mediated inflammatory conditions. Curr. Opin. Mol. Ther. 2009, 11, 329–336. [Google Scholar]
- Castro, M.; Mathur, S.; Hargreave, F.; Boulet, L.P.; Xie, F.; Young, J.; Wilkins, H.J.; Henkel, T.; Nair, P. Reslizumab for poorly controlled, eosinophilic asthma: A randomized, placebo-controlled study. Am. J. Respir. Crit. Care Med. 2011, 184, 1125–1132. [Google Scholar] [CrossRef]
- Pham, T.H.; Damera, G.; Newbold, P.; Ranade, K. Reductions in eosinophil biomarkers by benralizumab in patients with asthma. Respir. Med. 2016, 111, 21–29. [Google Scholar] [CrossRef] [Green Version]
- Fitzgerald, J.M.; Bleecker, E.R.; Nair, P.; Korn, S.; Ohta, K.; Lommatzsch, M.; Ferguson, G.T.; Busse, W.W.; Barker, P.; Sproule, S.; et al. Benralizumab, an anti-interleukin-5 receptor alpha monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet 2016, 388, 2128–2141. [Google Scholar] [CrossRef]
- O’Quinn, S.; Xu, X.; Hirsch, I. Rescue medication use reduction with Benralizumab for patients with severe, uncontrolled eosinophilic asthma. Ann. Allergy Asthma Immunol. 2018, 121, S18. [Google Scholar] [CrossRef]
- Deeks, E.D. Dupilumab: A Review in Moderate to Severe Asthma. Drugs 2019, 79, 1885–1895. [Google Scholar] [CrossRef]
- Varricchi, G.; Pecoraro, A.; Marone, G.; Criscuolo, G.; Spadaro, G.; Genovese, A.; Marone, G. Thymic Stromal Lymphopoietin Isoforms, Inflammatory Disorders, and Cancer. Front. Immunol. 2018, 9, 1595. [Google Scholar] [CrossRef] [Green Version]
- Menzies-Gow, A.; Colice, G.; Griffiths, J.M.; Almqvist, G.; Ponnarambil, S.; Kaur, P.; Ruberto, G.; Bowen, K.; Hellqvist, A.; Mo, M.; et al. NAVIGATOR: A phase 3 multicentre, randomized, double-blind, placebo-controlled, parallel-group trial to evaluate the efficacy and safety of tezepelumab in adults and adolescents with severe, uncontrolled asthma. Respir. Res. 2020, 21, 266. [Google Scholar] [CrossRef] [PubMed]
- Corren, J.; Parnes, J.R.; Wang, L.; Mo, M.; Roseti, S.L.; Griffiths, J.M.; van der Merwe, R. Tezepelumab in Adults with Uncontrolled Asthma. N. Engl. J. Med. 2019, 380, 2082. [Google Scholar] [CrossRef]
- Emson, C.; Diver, S.; Chachi, L.; Megally, A.; Small, C.; Downie, J.; Parnes, J.R.; Bowen, K.; Colice, G.; Brightling, C.E. CASCADE: A phase 2, randomized, double-blind, placebo-controlled, parallel-group trial to evaluate the effect of tezepelumab on airway inflammation in patients with uncontrolled asthma. Respir. Res. 2020, 21, 265. [Google Scholar] [CrossRef] [PubMed]
- Chinthrajah, S.; Cao, S.; Liu, C.; Lyu, S.C.; Sindher, S.B.; Long, A.; Sampath, V.; Petroni, D.; Londei, M.; Nadeau, K.C. Phase 2a randomized, placebo-controlled study of anti-IL-33 in peanut allergy. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corren, J.; Lemanske, R.F.; Hanania, N.A.; Korenblat, P.E.; Parsey, M.V.; Arron, J.R.; Harris, J.M.; Scheerens, H.; Wu, L.C.; Su, Z.; et al. Lebrikizumab treatment in adults with asthma. N. Engl. J. Med. 2011, 365, 1088–1098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ultsch, M.; Bevers, J.; Nakamura, G.; Vandlen, R.; Kelley, R.F.; Wu, L.C.; Eigenbrot, C. Structural basis of signaling blockade by anti-IL-13 antibody Lebrikizumab. J. Mol. Biol. 2013, 425, 1330–1339. [Google Scholar] [CrossRef] [PubMed]
- Korenblat, P.; Kerwin, E.; Leshchenko, I.; Yen, K.; Holweg, C.T.J.; Anzures-Cabrera, J.; Martin, C.; Putnam, W.S.; Governale, L.; Olsson, J.; et al. Efficacy and safety of lebrikizumab in adult patients with mild-to-moderate asthma not receiving inhaled corticosteroids. Respir. Med. 2018, 134, 143–149. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Casale, T.B.; Chipps, B.E.; Rosen, K.; Trzaskoma, B.; Haselkorn, T.; Omachi, T.A.; Greenberg, S.; Hanania, N.A. Response to omalizumab using patient enrichment criteria from trials of novel biologics in asthma. Allergy 2018, 73, 490–497. [Google Scholar] [CrossRef]
- Hanania, N.A.; Alpan, O.; Hamilos, D.L.; Condemi, J.J.; Reyes-Rivera, I.; Zhu, J.; Rosen, K.E.; Eisner, M.D.; Wong, D.A.; Busse, W. Omalizumab in severe allergic asthma inadequately controlled with standard therapy: A randomized trial. Ann. Intern. Med. 2011, 154, 573–582. [Google Scholar] [CrossRef]
- MacDonald, K.M.; Kavati, A.; Ortiz, B.; Alhossan, A.; Lee, C.S.; Abraham, I. Short- and long-term real-world effectiveness of omalizumab in severe allergic asthma: Systematic review of 42 studies published 2008–2018. Expert Rev. Clin. Immunol. 2019, 15, 553–569. [Google Scholar] [CrossRef] [Green Version]
- Yancey, S.W.; Ortega, H.G.; Keene, O.N.; Mayer, B.; Gunsoy, N.B.; Brightling, C.E.; Bleecker, E.R.; Haldar, P.; Pavord, I.D. Meta-analysis of asthma-related hospitalization in mepolizumab studies of severe eosinophilic asthma. J. Allergy Clin. Immunol. 2017, 139, 1167–1175.e1162. [Google Scholar] [CrossRef] [Green Version]
- Gupta, A.; Ikeda, M.; Geng, B.; Azmi, J.; Price, R.G.; Bradford, E.S.; Yancey, S.W.; Steinfeld, J. Long-term safety and pharmacodynamics of mepolizumab in children with severe asthma with an eosinophilic phenotype. J. Allergy Clin. Immunol. 2019, 144, 1336–1342.e1337. [Google Scholar] [CrossRef] [Green Version]
- Gupta, A.; Pouliquen, I.; Austin, D.; Price, R.G.; Kempsford, R.; Steinfeld, J.; Bradford, E.S.; Yancey, S.W. Subcutaneous mepolizumab in children aged 6 to 11 years with severe eosinophilic asthma. Pediatric Pulmonol. 2019, 54, 1957–1967. [Google Scholar] [CrossRef]
- Bel, E.H.; Wenzel, S.E.; Thompson, P.J.; Prazma, C.M.; Keene, O.N.; Yancey, S.W.; Ortega, H.G.; Pavord, I.D.; Investigators, S. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1189–1197. [Google Scholar] [CrossRef]
- Corren, J.; Weinstein, S.; Janka, L.; Zangrilli, J.; Garin, M. Phase 3 Study of Reslizumab in Patients With Poorly Controlled Asthma: Effects Across a Broad Range of Eosinophil Counts. Chest 2016, 150, 799–810. [Google Scholar] [CrossRef] [Green Version]
- Bjermer, L.; Lemiere, C.; Maspero, J.; Weiss, S.; Zangrilli, J.; Germinaro, M. Reslizumab for Inadequately Controlled Asthma With Elevated Blood Eosinophil Levels: A Randomized Phase 3 Study. Chest 2016, 150, 789–798. [Google Scholar] [CrossRef] [Green Version]
- Rogliani, P.; Calzetta, L.; Matera, M.G.; Laitano, R.; Ritondo, B.L.; Hanania, N.A.; Cazzola, M. Severe asthma and biological therapy: When, which, and for whom. Pulm. Ther. 2020, 6, 47–66. [Google Scholar] [CrossRef] [Green Version]
- Casale, T.B.; Pacou, M.; Mesana, L.; Farge, G.; Sun, S.X.; Castro, M. Reslizumab Compared with Benralizumab in Patients with Eosinophilic Asthma: A Systematic Literature Review and Network Meta-Analysis. J. Allergy Clin. Immunol. Pract. 2019, 7, 122–130.e121. [Google Scholar] [CrossRef]
- Rodrigues, M.A.; Nogueira, M.; Torres, T. Dupilumab for atopic dermatitis: Evidence to date. G Ital. Dermatol. Venereol. 2019, 154, 696–713. [Google Scholar] [CrossRef]
- Zayed, Y.; Kheiri, B.; Banifadel, M.; Hicks, M.; Aburahma, A.; Hamid, K.; Bachuwa, G.; Chandran, A. Dupilumab safety and efficacy in uncontrolled asthma: A systematic review and meta-analysis of randomized clinical trials. J. Asthma 2019, 56, 1110–1119. [Google Scholar] [CrossRef]
- Brooks, G.D. Updated Evaluation of Dupilumab in the Treatment of Asthma: Patient Selection and Reported Outcomes. Ther. Clin. Risk Manag. 2020, 16, 181–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Just, J.; Deschildre, A.; Lejeune, S.; Amat, F. New perspectives of childhood asthma treatment with biologics. Pediatric Allergy Immunol. 2019, 30, 159–171. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Lambrecht, B.N.; Hammad, H. The airway epithelium in asthma. Nat. Med. 2012, 18, 684–692. [Google Scholar] [CrossRef]
- Viana, F. TRPA1 channels: Molecular sentinels of cellular stress and tissue damage. J. Physiol. 2016, 594, 4151–4169. [Google Scholar] [CrossRef] [Green Version]
- Caceres, A.I.; Brackmann, M.; Elia, M.D.; Bessac, B.F.; del Camino, D.; D’Amours, M.; Witek, J.S.; Fanger, C.M.; Chong, J.A.; Hayward, N.J.; et al. A sensory neuronal ion channel essential for airway inflammation and hyperreactivity in asthma. Proc. Natl. Acad. Sci. USA 2009, 106, 9099–9104. [Google Scholar] [CrossRef] [Green Version]
- Balestrini, A.; Joseph, V.; Dourado, M.; Reese, R.M.; Shields, S.D.; Rouge, L.; Bravo, D.D.; Chernov-Rogan, T.; Austin, C.D.; Chen, H.; et al. A TRPA1 inhibitor suppresses neurogenic inflammation and airway contraction for asthma treatment. J. Exp. Med. 2021, 218, e20201637. [Google Scholar] [CrossRef]
- Strachan, D.P. Hay fever, hygiene, and household size. BMJ 1989, 299, 1259–1260. [Google Scholar] [CrossRef] [Green Version]
- Rautava, S.; Ruuskanen, O.; Ouwehand, A.; Salminen, S.; Isolauri, E. The hygiene hypothesis of atopic disease-an extended version. J. Pediatr. Gastroenterol. Nutr. 2004, 38, 378–388. [Google Scholar] [CrossRef]
- Wold, A.E. The hygiene hypothesis revised: Is the rising frequency of allergy due to changes in the intestinal flora? Allergy 1998, 53, 20–25. [Google Scholar] [CrossRef]
- Fujimura, K.E.; Lynch, S.V. Microbiota in allergy and asthma and the emerging relationship with the gut microbiome. Cell Host. Microbe 2015, 17, 592–602. [Google Scholar] [CrossRef] [Green Version]
- Kirjavainen, P.V.; Karvonen, A.M.; Adams, R.I.; Täubel, M.; Roponen, M.; Tuoresmäki, P.; Loss, G.; Jayaprakash, B.; Depner, M.; Ege, M.J.; et al. Farm-like indoor microbiota in non-farm homes protects children from asthma development. Nat. Med. 2019, 25, 1089–1095. [Google Scholar] [CrossRef]
- Kozik, A.; Huang, Y.J. Ecological interactions in asthma: From environment to microbiota and immune responses. Curr. Opin. Pulm. Med. 2020, 26, 27–32. [Google Scholar] [CrossRef]
- Karvonen, A.M.; Kirjavainen, P.V.; Täubel, M.; Jayaprakash, B.; Adams, R.I.; Sordillo, J.E.; Gold, D.R.; Hyvärinen, A.; Remes, S.; von Mutius, E.; et al. Indoor bacterial microbiota and development of asthma by 10.5 years of age. J. Allergy Clin. Immunol. 2019, 144, 1402–1410. [Google Scholar] [CrossRef] [Green Version]
- Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T.; Creasy, H.H.; Earl, A.M.; Fitzgerald, M.G.; Fulton, R.S.; et al. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar]
- Velasquez-Manoff, M. Gut Microbiome: The peacekeepers. Nature 2015, 518, S3–S11. [Google Scholar] [CrossRef] [Green Version]
- Frati, F.; Salvatori, C.; Incorvaia, C.; Bellucci, A.; Di Cara, G.; Marcucci, F.; Esposito, S. The role of the microbiome in asthma: The gut-lung axis. Int. J. Mol. Sci. 2018, 20, 123. [Google Scholar] [CrossRef] [Green Version]
- Dzidic, M.; Abrahamsson, T.R.; Artacho, A.; Björkstén, B.; Collado, M.C.; Mira, A.; Jenmalm, M.C. Aberrant IgA responses to the gut microbiota during infancy precede asthma and allergy development. J. Allergy Clin. Immunol. 2017, 139, 1017–1025.e1014. [Google Scholar] [CrossRef] [Green Version]
- Ivanov, I.I.; Atarashi, K.; Manel, N.; Brodie, E.L.; Shima, T.; Karaoz, U.; Wei, D.; Goldfarb, K.C.; Santee, C.A.; Lynch, S.V.; et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009, 139, 485–498. [Google Scholar] [CrossRef] [Green Version]
- Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [Green Version]
- Round, J.L.; Mazmanian, S.K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 2010, 107, 12204–12209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atarashi, K.; Tanoue, T.; Shima, T.; Imaoka, A.; Kuwahara, T.; Momose, Y.; Cheng, G.; Yamasaki, S.; Saito, T.; Ohba, Y.; et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 2010, 331, 337–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strauch, U.G.; Obermeier, F.; Grunwald, N.; Gurster, S.; Dunger, N.; Schultz, M.; Griese, D.P.; Maehler, M.; Scholmerich, J.; Rath, H.C. Influence of intestinal bacteria on induction of regulatory T cells: Lessons from a transfer model of colitis. Gut 2005, 54, 1546–1552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ostman, S.; Rask, C.; Wold, A.E.; Hultkrantz, S.; Telemo, E. Impaired regulatory T cell function in germ-free mice. Eur. J. Immunol. 2006, 36, 2336–2346. [Google Scholar] [CrossRef] [PubMed]
- Mazmanian, S.K.; Round, J.L.; Kasper, D.L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008, 453, 620–625. [Google Scholar] [CrossRef] [Green Version]
- Tsuda, M.; Hosono, A.; Yanagibashi, T.; Kihara-Fujioka, M.; Hachimura, S.; Itoh, K.; Hirayama, K.; Takahashi, K.; Kaminogawa, S. Intestinal commensal bacteria promote T cell hyporesponsiveness and down-regulate the serum antibody responses induced by dietary antigen. Immunol. Lett. 2010, 132, 45–52. [Google Scholar] [CrossRef]
- Chinen, T.; Volchkov, P.Y.; Chervonsky, A.V.; Rudensky, A.Y. A critical role for regulatory T cell-mediated control of inflammation in the absence of commensal microbiota. J. Exp. Med. 2010, 207, 2323–2330. [Google Scholar] [CrossRef] [Green Version]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; de Roos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Zeng, H.; Chi, H. Metabolic control of regulatory T cell development and function. Trends Immunol. 2015, 36, 3–12. [Google Scholar] [CrossRef] [Green Version]
- Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef]
- Lewis, G.; Wang, B.; Shafiei Jahani, P.; Hurrell, B.P.; Banie, H.; Aleman Muench, G.R.; Maazi, H.; Helou, D.G.; Howard, E.; Galle-Treger, L.; et al. Dietary fiber-induced microbial short chain fatty acids suppress ILC2-dependent airway inflammation. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [Green Version]
- den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.-J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef] [Green Version]
- Gensollen, T.; Iyer, S.S.; Kasper, D.L.; Blumberg, R.S. How colonization by microbiota in early life shapes the immune system. Science 2016, 352, 539–544. [Google Scholar] [CrossRef] [Green Version]
- Thorsen, J.; Rasmussen, M.A.; Waage, J.; Mortensen, M.; Brejnrod, A.; Bønnelykke, K.; Chawes, B.L.; Brix, S.; Sørensen, S.J.; Stokholm, J.; et al. Infant airway microbiota and topical immune perturbations in the origins of childhood asthma. Nat. Commun. 2019, 10, 5001. [Google Scholar] [CrossRef] [Green Version]
- Hilty, M.; Burke, C.; Pedro, H.; Cardenas, P.; Bush, A.; Bossley, C.; Davies, J.; Ervine, A.; Poulter, L.; Pachter, L.; et al. Disordered microbial communities in asthmatic airways. PLoS ONE 2010, 5, e8578. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.J.; Nariya, S.; Harris, J.M.; Lynch, S.V.; Choy, D.F.; Arron, J.R.; Boushey, H. The airway microbiome in patients with severe asthma: Associations with disease features and severity. J. Allergy Clin. Immunol. 2015, 136, 874–884. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.; Jackson, D.; Bacharier, L.B.; Mauger, D.; Boushey, H.; Castro, M.; Durack, J.; Huang, Y.; Lemanske, R.F., Jr.; Storch, G.A.; et al. The upper-airway microbiota and loss of asthma control among asthmatic children. Nat. Commun. 2019, 10, 5714. [Google Scholar] [CrossRef] [Green Version]
- Arrieta, M.-C.; Arévalo, A.; Stiemsma, L.; Dimitriu, P.; Chico, M.E.; Loor, S.; Vaca, M.; Boutin, R.C.T.; Morien, E.; Jin, M.; et al. Associations between infant fungal and bacterial dysbiosis and childhood atopic wheeze in a nonindustrialized setting. J. Allergy Clin. Immunol. 2018, 142, 424–434.e410. [Google Scholar] [CrossRef] [Green Version]
- Arrieta, M.-C.; Stiemsma, L.T.; Dimitriu, P.A.; Thorson, L.; Russell, S.; Yurist-Doutsch, S.; Kuzeljevic, B.; Gold, M.J.; Britton, H.M.; Lefebvre, D.L.; et al. Early infancy microbial and metabolic alterations affect risk of childhood asthma. Sci. Transl. Med. 2015, 7, 307ra152. [Google Scholar] [CrossRef]
- Chua, H.-H.; Chou, H.-C.; Tung, Y.-L.; Chiang, B.-L.; Liao, C.-C.; Liu, H.-H.; Ni, Y.-H. Intestinal dysbiosis featuring abundance of Ruminococcus gnavus associates with allergic diseases in infants. Gastroenterology 2018, 154, 154–167. [Google Scholar] [CrossRef]
- Budden, K.F.; Gellatly, S.L.; Wood, D.L.A.; Cooper, M.A.; Morrison, M.; Hugenholtz, P.; Hansbro, P.M. Emerging pathogenic links between microbiota and the gut–lung axis. Nat. Rev. Microbiol. 2017, 15, 55–63. [Google Scholar] [CrossRef] [PubMed]
- Wopereis, H.; Oozeer, R.; Knipping, K.; Belzer, C.; Knol, J. The first thousand days—intestinal microbiology of early life: Establishing a symbiosis. Pediatric Allergy Immunol. 2014, 25, 428–438. [Google Scholar] [CrossRef] [Green Version]
- Johnson, C.C.; Ownby, D.R. The infant gut bacterial microbiota and risk of pediatric asthma and allergic diseases. Transl. Res. 2017, 179, 60–70. [Google Scholar] [CrossRef] [Green Version]
- Zimmermann, P.; Messina, N.; Mohn, W.W.; Finlay, B.B.; Curtis, N. Association between the intestinal microbiota and allergic sensitization, eczema, and asthma: A systematic review. J. Allergy Clin. Immunol. 2019, 143, 467–485. [Google Scholar] [CrossRef]
- Galazzo, G.; van Best, N.; Bervoets, L.; Dapaah, I.O.; Savelkoul, P.H.; Hornef, M.W.; Lau, S.; Hamelmann, E.; Penders, J. Development of the microbiota and associations with birth mode, diet, and atopic disorders in a longitudinal analysis of stool samples, collected from infancy through early childhood. Gastroenterology 2020, 158, 1584–1596. [Google Scholar] [CrossRef]
- Fujimura, K.E.; Sitarik, A.R.; Havstad, S.; Lin, D.L.; Levan, S.; Fadrosh, D.; Panzer, A.R.; LaMere, B.; Rackaityte, E.; Lukacs, N.W.; et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 2016, 22, 1187–1191. [Google Scholar] [CrossRef] [Green Version]
- Goleva, E.; Jackson, L.P.; Harris, J.K.; Robertson, C.E.; Sutherland, E.R.; Hall, C.F.; Good, J.T., Jr.; Gelfand, E.W.; Martin, R.J.; Leung, D.Y. The effects of airway microbiome on corticosteroid responsiveness in asthma. Am. J. Respir. Crit. Care Med. 2013, 188, 1193–1201. [Google Scholar] [CrossRef] [Green Version]
- Durack, J.; Lynch, S.V.; Nariya, S.; Bhakta, N.R.; Beigelman, A.; Castro, M.; Dyer, A.-M.; Israel, E.; Kraft, M.; Martin, R.J.; et al. Features of the bronchial bacterial microbiome associated with atopy, asthma, and responsiveness to inhaled corticosteroid treatment. J. Allergy Clin. Immunol. 2017, 140, 63–75. [Google Scholar] [CrossRef] [Green Version]
- Song, X.; Sun, X.; Oh, S.F.; Wu, M.; Zhang, Y.; Zheng, W.; Geva-Zatorsky, N.; Jupp, R.; Mathis, D.; Benoist, C.; et al. Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis. Nature 2020, 577, 410–415. [Google Scholar] [CrossRef] [PubMed]
- Campbell, C.; McKenney, P.T.; Konstantinovsky, D.; Isaeva, O.I.; Schizas, M.; Verter, J.; Mai, C.; Jin, W.-B.; Guo, C.-J.; Violante, S.; et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 2020, 581, 475–479. [Google Scholar] [CrossRef] [PubMed]
- Barcik, W.; Boutin, R.C.T.; Sokolowska, M.; Finlay, B.B. The role of lung and gut microbiota in the pathology of asthma. Immunity 2020, 52, 241–255. [Google Scholar] [CrossRef] [Green Version]
- Azad, M.B.; Coneys, J.G.; Kozyrskyj, A.L.; Field, C.J.; Ramsey, C.D.; Becker, A.B.; Friesen, C.; Abou-Setta, A.M.; Zarychanski, R. Probiotic supplementation during pregnancy or infancy for the prevention of asthma and wheeze: Systematic review and meta-analysis. BMJ 2013, 347, f6471. [Google Scholar] [CrossRef] [Green Version]
- Elazab, N.; Mendy, A.; Gasana, J.; Vieira, E.R.; Quizon, A.; Forno, E. Probiotic administration in early life, atopy, and asthma: A meta-analysis of clinical trials. Pediatrics 2013, 132, e666–e676. [Google Scholar] [CrossRef] [Green Version]
- Zuccotti, G.; Meneghin, F.; Aceti, A.; Barone, G.; Callegari, M.L.; Di Mauro, A.; Fantini, M.P.; Gori, D.; Indrio, F.; Maggio, L.; et al. Probiotics for prevention of atopic diseases in infants: Systematic review and meta-analysis. Allergy 2015, 70, 1356–1371. [Google Scholar] [CrossRef] [Green Version]
- Wei, X.; Jiang, P.; Liu, J.; Sun, R.; Zhu, L. Association between probiotic supplementation and asthma incidence in infants: A meta-analysis of randomized controlled trials. J. Asthma Off. J. Assoc. Care Asthma 2020, 57, 167–178. [Google Scholar] [CrossRef]
- Wickens, K.; Black, P.; Stanley, T.V.; Mitchell, E.; Barthow, C.; Fitzharris, P.; Purdie, G. A protective effect of Lactobacillus rhamnosus HN001 against eczema in the first 2 years of life persists to age 4 years. Clin. Exp. Allergy 2012, 42, 1071–1079. [Google Scholar] [CrossRef]
- Wickens, K.; Stanley, T.V.; Mitchell, E.A.; Barthow, C.; Fitzharris, P.; Purdie, G.; Siebers, R. Early supplementation with Lactobacillus rhamnosus HN001 reduces eczema prevalence to 6 years: Does it also reduce atopic sensitization? Clin. Exp. Allergy 2013, 43, 1048–1057. [Google Scholar] [CrossRef]
- Kalliomäki, M.; Salminen, S.; Poussa, T.; Isolauri, E. Probiotics during the first 7 years of life: A cumulative risk reduction of eczema in a randomized, placebo-controlled trial. J. Allergy Clin. Immunol. 2007, 119, 1019–1021. [Google Scholar] [CrossRef]
- Kuitunen, M.; Kukkonen, K.; Juntunen-Backman, K.; Korpela, R.; Poussa, T.; Tuure, T.; Haahtela, T. Probiotics prevent IgE-associated allergy until age 5 years in cesarean-delivered children but not in the total cohort. J. Allergy Clin. Immunol. 2009, 123, 335–341. [Google Scholar] [CrossRef]
- Dotterud, C.K.; Storrø, O.; Johnsen, R.; Oien, T. Probiotics in pregnant women to prevent allergic disease: A randomized, double-blind trial. Br. J. Dermatol. 2010, 163, 616–623. [Google Scholar] [CrossRef]
- Boyle, R.J.; Ismail, I.H.; Kivivuori, S.; Licciardi, P.V.; Robins-Browne, R.M.; Mah, L.J.; Axelrad, C. Lactobacillus GG treatment during pregnancy for the prevention of eczema: A randomized controlled trial. Allergy 2011, 66, 509–516. [Google Scholar] [CrossRef]
- Jensen, M.P.; Meldrum, S.; Taylor, A.L.; Dunstan, J.A.; Prescott, S.L. Early probiotic supplementation for allergy prevention: Long-term outcomes. J. Allergy Clin. Immunol. 2012, 130, 1209–1211. [Google Scholar] [CrossRef]
- West, C.E.; Hammarström, M.L.; Hernell, O. Probiotics in primary prevention of allergic disease-follow-up at 8–9 years of age. Allergy 2013, 68, 1015–1020. [Google Scholar] [CrossRef]
- Abrahamsson, T.R.; Jakobsson, T.; Björkstén, B.; Oldaeus, G.; Jenmalm, M.C. No effect of probiotics on respiratory allergies: A seven-year follow-up of a randomized controlled trial in infancy. Pediatr. Allergy Immunol. 2013, 24, 556–561. [Google Scholar] [CrossRef] [Green Version]
- Kauppi, P.K.; Kuokkanen, M.K.; Kukkonen, K.K.; Laitinen, T.H.; Kuitunen, M.K. Interaction of NPSR1 genotypes and probiotics in the manifestation of atopic eczema in early childhood. Allergol. Immunopathol. 2014, 42, 560–567. [Google Scholar]
- Loo, E.X.; Llanora, G.V.; Lu, Q.; Aw, M.M.; Lee, B.W.; Shek, L.P. Supplementation with probiotics in the first 6 months of life did not protect against eczema and allergy in at-risk Asian infants: A 5-year follow-up. Int. Arch. Allergy Immunol. 2014, 163, 25–28. [Google Scholar] [CrossRef] [PubMed]
- Gorissen, D.M.W.; Rutten, N.; Oostermeijer, C.M.J.; Niers, L.E.M.; Hoekstra, M.O.; Rijkers, G.T.; van der Ent, C.K. Preventive effects of selected probiotic strains on the development of asthma and allergic rhinitis in childhood. Clin. Exp. Allergy 2014, 44, 1431–1433. [Google Scholar] [CrossRef] [PubMed]
- Berni Canani, R.; Di Costanzo, M.; Bedogni, G.; Amoroso, A.; Cosenza, L.; Di Scala, C.; Granata, V. Extensively hydrolyzed casein formula containing Lactobacillus rhamnosus GG reduces the occurrence of other allergic manifestations in children with cow’s milk allergy: 3-year randomized controlled trial. J. Allergy Clin. Immunol. 2017, 139, 1906–1913.e1904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cabana, M.D.; McKean, M.; Caughey, A.B.; Fong, L.; Lynch, S.; Wong, A. Early probiotic supplementation for eczema and asthma prevention: A randomized controlled trial. Pediatrics 2017, 140, 1–9. [Google Scholar] [CrossRef] [Green Version]
- Hol, J.; van Leer, E.H.G.; Elink Schuurman, B.E.E.; de Ruiter, L.F.; Samsom, J.N.; Hop, W.; Neijens, H.J. The acquisition of tolerance toward cow’s milk through probiotic supplementation: A randomized, controlled trial. J. Allergy Clin. Immunol. 2008, 121, 1448–1454. [Google Scholar] [CrossRef]
- Kopp, M.V.; Hennemuth, I.; Heinzmann, A.; Urbanek, R. Randomized, double-blind, placebo-controlled trial of probiotics for primary prevention: No clinical effects of Lactobacillus GG supplementation. Pediatrics 2008, 121, e850. [Google Scholar] [CrossRef]
- Wickens, K.; Black, P.N.; Stanley, T.V.; Mitchell, E.; Fitzharris, P.; Tannock, G.W.; Purdie, G. A differential effect of 2 probiotics in the prevention of eczema and atopy: A double-blind, randomized, placebo-controlled trial. J. Allergy Clin. Immunol. 2008, 122, 788–794. [Google Scholar] [CrossRef]
- West, C.E.; Hammarström, M.L.; Hernell, O. Probiotics during weaning reduce the incidence of eczema. Pediatr. Allergy Immunol. 2009, 20, 430–437. [Google Scholar] [CrossRef]
- Van der Aa, L.B.; van Aalderen, W.M.C.; Heymans, H.S.A.; Henk Sillevis Smitt, J.; Nauta, A.J.; Knippels, L.M.J.; Ben Amor, K. Synbiotics prevent asthma-like symptoms in infants with atopic dermatitis. Allergy 2011, 66, 170–177. [Google Scholar] [CrossRef]
- Gore, C.; Custovic, A.; Tannock, G.W.; Munro, K.; Kerry, G.; Johnson, K.; Peterson, C. Treatment and secondary prevention effects of the probiotics Lactobacillus paracasei or Bifidobacterium lactis on early infant eczema: Randomized controlled trial with follow-up until age 3 years. Clin. Exp. Allergy 2012, 42, 112–122. [Google Scholar] [CrossRef]
- Ou, C.Y.; Kuo, H.C.; Wang, L.; Hsu, T.Y.; Chuang, H.; Liu, C.A.; Chang, J.C. Prenatal and postnatal probiotics reduces maternal but not childhood allergic diseases: A randomized, double-blind, placebo-controlled trial. Clin. Exp. Allergy 2012, 42, 1386–1396. [Google Scholar] [CrossRef]
- Simpson, M.R.; Dotterud, C.K.; Storrø, O.; Johnsen, R.; Øien, T. Perinatal probiotic supplementation in the prevention of allergy related disease: 6 year follow up of a randomised controlled trial. BMC Dermatol. 2015, 15, 13. [Google Scholar] [CrossRef] [Green Version]
- Wickens, K.; Barthow, C.; Mitchell, E.A.; Stanley, T.V.; Purdie, G.; Rowden, J.; Kang, J.; Hood, F.; van den Elsen, L.; Forbes-Blom, E.; et al. Maternal supplementation alone with Lactobacillus rhamnosus HN001 during pregnancy and breastfeeding does not reduce infant eczema. Pediatr. Allergy Immunol. 2018, 29, 296–302. [Google Scholar] [CrossRef]
- Patnode, M.L.; Beller, Z.W.; Han, N.D.; Cheng, J.; Peters, S.L.; Terrapon, N.; Henrissat, B.; Le Gall, S.; Saulnier, L.; Hayashi, D.K.; et al. Interspecies competition impacts targeted manipulation of human gut bacteria by fiber-derived glycans. Cell 2019, 179, 59–73.e13. [Google Scholar] [CrossRef] [PubMed]
- Delzenne, N.M.; Bindels, L.B. Food for thought about manipulating gut bacteria. Nature 2020, 577, 32–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deehan, E.C.; Yang, C.; Perez-Muñoz, M.E.; Nguyen, N.K.; Cheng, C.C.; Triador, L.; Zhang, Z.; Bakal, J.A.; Walter, J. Precision microbiome modulation with discrete dietary fiber structures directs short-chain fatty acid production. Cell Host Microbe 2020. [Google Scholar] [CrossRef] [PubMed]
- Sharma, A.; Laxman, B.; Naureckas, E.T.; Hogarth, D.K.; Sperling, A.I.; Solway, J.; Ober, C.; Gilbert, J.A.; White, S.R. Associations between fungal and bacterial microbiota of airways and asthma endotypes. J. Allergy Clin. Immunol. 2019, 144, 1214–1227.e1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vandenborght, L.-E.; Enaud, R.; Urien, C.; Coron, N.; Girodet, P.-O.; Ferreira, S.; Berger, P.; Delhaes, L. Type 2–high asthma is associated with a specific indoor mycobiome and microbiome. J. Allergy Clin. Immunol. 2021, 147, 1296–1305.e1296. [Google Scholar] [CrossRef] [PubMed]
- Denner, D.R.; Sangwan, N.; Becker, J.B.; Hogarth, D.K.; Oldham, J.; Castillo, J.; Sperling, A.I.; Solway, J.; Naureckas, E.T.; Gilbert, J.A.; et al. Corticosteroid therapy and airflow obstruction influence the bronchial microbiome, which is distinct from that of bronchoalveolar lavage in asthmatic airways. J. Allergy Clin. Immunol. 2016, 137, 1398–1405.e1393. [Google Scholar] [CrossRef] [Green Version]
- McCauley, K.; Durack, J.; Valladares, R.; Fadrosh, D.W.; Lin, D.L.; Calatroni, A.; LeBeau, P.K.; Tran, H.T.; Fujimura, K.E.; LaMere, B.; et al. Distinct nasal airway bacterial microbiotas differentially relate to exacerbation in pediatric patients with asthma. J. Allergy Clin. Immunol. 2019, 144, 1187–1197. [Google Scholar] [CrossRef]
Allergic Asthma (Th2-High) | Non-Allergic Asthma (Th2-Low or Non-Th2) | |
---|---|---|
Prevalence [14] | 60% | 10–33% |
Age of onset | Occurs early in life | Mostly occurs later in life |
Triggers | House dust mites, pollen, pet dander and cockroaches, etc. | More diverse. Cold air, smoke, obesity, occupational exposure, and exercise |
Inflammatory mediators | IL-4, IL-5, IL-13, IL-25, IL-33 and TSLP | IL-1β, IL-6, IFN-γ, TNF-α and IL-17 |
Severity | Milder than Th2-low | More severe than allergic asthma |
Treatment | Responds well to ICS | Require higher doses of ICS or non-responsive to ICS [15,16] |
Recruiting cells in the airway | Eosinophilic inflammation [17] | neutrophilic or pauci-granulocytic inflammation [18,19,20] |
Serum total IgE | High | Normal |
Skin prick test | Positive | Negative |
FDA-Approved Monoclonal Antibodies | ||||||||
---|---|---|---|---|---|---|---|---|
Name | Target | Biological Effects | Dosing | Indication | Sponsor | |||
Omalizumab (Xolair®) | FcεRI binding site of IgE [52,53] | Decrease circulating total IgE Decreased expression of FcεRI on inflammatory cells Decreased mediator release. | According to IgE levels Every 2–4 wks (s.c.) | Moderate-severe allergic asthma IgE ≥ 30 IU/mL + skin prick test | Genentech | |||
Mepolizumab (Nucala®) | IL-5 | inhibiting the bioactivity of IL-5 by blocking its binding to IL-5Rα complex expressed on the eosinophil cell surface [54] Reduces the production and survival of eosinophils | 100 mg Every 4 wks (s.c.) | Severe eosinophilic asthma blood eosinophils ≥ 400/uL | GlaxoSmithKline | |||
Reslizumab (Cinqair®) | IL-5 | Inhibiting IL-5 signaling [55] Decreased eosinophils in blood and sputum [56] | 3 mg/kg Every 4 wks (i.v.) | Severe eosinophilic asthma blood eosinophils ≥ 400/uL | Teva Pharmaceuticals | |||
Benralizumab (Fasenta®) | IL-5R⍺ | Decreased eosinophils and basophils though ADCC [57] | 30 mg Every 8 wks (s.c.) [58,59] | Severe eosinophilic asthma blood eosinophils ≥ 300/uL | AstraZeneca | |||
Dupilumab (Dupixent®) | IL-4R⍺ | Blockade IL-4/IL-4R⍺ binding Blockade IL-13/IL-4R⍺ binding | 300 mg Every 2 wks (s.c.) [60] | blood eosinophils ≥ 150/uL FeNO > 25 ppb | Sanofi and Regeneron | |||
Biological Therapies under Clinical Trials | ||||||||
Name | Target | Biological Effects | Dosing | Indication | Sponsor/Development Status | |||
Tezepelumab | TSLP | TSLP blockade [61,62,63] | 210 mg/kg Every 4 wks (s.c.) [64] | Patients with high (≥300 cells/µL) or low (<300 cells/µL) blood eosinophil counts Adults with oral corticosteroid-dependent asthma | AstraZeneca and Amgen/Phase III (NCT03406078) https://clinicaltrials.gov/ct2/show/NCT03406078 (accessed on 20 April 2021) | |||
Etokimab (ANB020) | IL-33 | IL-33 blockade | 300 mg single dose (i.v.) | Adults with severe eosinophilic asthma blood eosinophil counts ≥ 300 cells/µL Stably maintained on ICS/LABA dose for at least 3 months [65] | AnaptysBio/Phase IIa(NCT03469934) https://clinicaltrials.gov/ct2/show/NCT03469934 (accessed on 20 April 2021) | |||
Lebrikizumab (RO5490255) | IL-13 | Binds to soluble IL-13 and blocks downstream signaling [66,67] | 125 mg Every 4 wks (s.c.) [68] | Adults with uncontrolled asthma On ICS and a second controller medication | Hoffmann-La Roche/Phase II (NCT02099656) https://clinicaltrials.gov/ct2/show/NCT02099656 (accessed on 20 April 2021) | |||
GDC-0334 (small molecule) | TRPA1 | TRPA1 inhibitor | Orally with dose escalation between cohorts | Phase I study in healthy adult subjects | Genentech/Phase I (NCT03381144) https://clinicaltrials.gov/ct2/show/NCT03381144 (accessed on 20 April 2021) |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Chiu, C.-J.; Huang, M.-T. Asthma in the Precision Medicine Era: Biologics and Probiotics. Int. J. Mol. Sci. 2021, 22, 4528. https://doi.org/10.3390/ijms22094528
Chiu C-J, Huang M-T. Asthma in the Precision Medicine Era: Biologics and Probiotics. International Journal of Molecular Sciences. 2021; 22(9):4528. https://doi.org/10.3390/ijms22094528
Chicago/Turabian StyleChiu, Chiao-Juno, and Miao-Tzu Huang. 2021. "Asthma in the Precision Medicine Era: Biologics and Probiotics" International Journal of Molecular Sciences 22, no. 9: 4528. https://doi.org/10.3390/ijms22094528
APA StyleChiu, C. -J., & Huang, M. -T. (2021). Asthma in the Precision Medicine Era: Biologics and Probiotics. International Journal of Molecular Sciences, 22(9), 4528. https://doi.org/10.3390/ijms22094528