Skip to main content

Main menu

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • COVID-19 submission information
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

User menu

  • Log in
  • Subscribe
  • Contact Us
  • My Cart

Search

  • Advanced search
  • ERS Publications
    • European Respiratory Journal
    • ERJ Open Research
    • European Respiratory Review
    • Breathe
    • ERS Books
    • ERS publications home

Login

European Respiratory Society

Advanced Search

  • Home
  • Current issue
  • Past issues
  • Authors/reviewers
    • Instructions for authors
    • Submit a manuscript
    • COVID-19 submission information
    • Institutional open access agreements
    • Peer reviewer login
  • Alerts
  • Subscriptions

Phosphodiesterase 4B inhibition: a potential novel strategy for treating pulmonary fibrosis

Martin Kolb, Bruno Crestani, Toby M. Maher
European Respiratory Review 2023 32: 220206; DOI: 10.1183/16000617.0206-2022
Martin Kolb
1Department of Respiratory Medicine, Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
2Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Martin Kolb
Bruno Crestani
3Service de Pneumologie A, Hôpital Bichat, APHP, Paris, France
4INSERM, Unité 1152, Université Paris Cité, Paris, France
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Bruno Crestani
Toby M. Maher
5Keck Medicine of USC, Los Angeles, CA, USA
6National Heart and Lung Institute, Imperial College London, London, UK
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: Toby.Maher@med.usc.edu
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Patients with interstitial lung disease can develop a progressive fibrosing phenotype characterised by an irreversible, progressive decline in lung function despite treatment. Current therapies slow, but do not reverse or stop, disease progression and are associated with side-effects that can cause treatment delay or discontinuation. Most crucially, mortality remains high. There is an unmet need for more efficacious and better-tolerated and -targeted treatments for pulmonary fibrosis. Pan-phosphodiesterase 4 (PDE4) inhibitors have been investigated in respiratory conditions. However, the use of oral inhibitors can be complicated due to class-related systemic adverse events, including diarrhoea and headaches. The PDE4B subtype, which has an important role in inflammation and fibrosis, has been identified in the lungs. Preferentially targeting PDE4B has the potential to drive anti-inflammatory and antifibrotic effects via a subsequent increase in cAMP, but with improved tolerability. Phase I and II trials of a novel PDE4B inhibitor in patients with idiopathic pulmonary fibrosis have shown promising results, stabilising pulmonary function measured by change in forced vital capacity from baseline, while maintaining an acceptable safety profile. Further research into the efficacy and safety of PDE4B inhibitors in larger patient populations and for a longer treatment period is needed.

Abstract

Preferential inhibition of phosphodiesterase 4 (PDE4) B in patients with pulmonary fibrosis could provide anti-inflammatory and antifibrotic effects with improved safety compared with pan-PDE4 inhibitors. https://bit.ly/3UE0CdU

Introduction

Interstitial lung diseases (ILDs) are a heterogeneous group of lung diseases that are characterised by pulmonary fibrosis, which causes lung damage, respiratory failure and early mortality [1–4]. Patients with ILD can develop a progressive fibrosing phenotype or progressive pulmonary fibrosis, both of which are characterised by an irreversible decline in lung function despite treatment, the prototype example being idiopathic pulmonary fibrosis (IPF) [3, 4].

There are currently two antifibrotic therapies approved and recommended [5] for the treatment of IPF, namely pirfenidone [6, 7] and nintedanib [8, 9]. Nintedanib, a tyrosine kinase inhibitor, is also approved for the treatment of systemic sclerosis (SSc)-associated ILD and other chronic fibrosing ILDs with a progressive phenotype [5, 8, 9]. Tocilizumab, an anti-interleukin (IL)-6-receptor biologic, is approved in the United States for treatment of SSc-associated ILD [10].

Current therapies slow, but do not reverse or stop, disease progression and are associated with side-effects that can cause treatment delay or discontinuation. Moreover, mortality remains high [11]. The only potentially curative treatment for IPF and other ILDs remains lung transplantation. As such, there is an unmet need for more efficacious and better-tolerated and -targeted treatments for pulmonary fibrosis [12].

Overview and introduction to phosphodiesterase (PDE) 4

PDE superfamily

PDEs are a large family of enzymes that mediate the hydrolysis of second messengers cAMP and cyclic guanosine monophosphate (cGMP) to 5′-AMP and 5′-GMP, respectively, during intracellular signalling [13]. PDEs have been reviewed extensively elsewhere [13–15]. Briefly, the PDE superfamily contains 11 gene families, coding for PDE1–11, with most of the subfamilies containing multiple subtypes (e.g. PDE1A, PDE1B and PDE1C) (table 1) and multiple variants (e.g. PDE1A1, PDE1A2, etc.). Although PDEs are ubiquitous, the distribution of PDE subfamilies varies between different cell and tissue types. Due to their role in intracellular signalling and the potential for precise targeting of subtypes, PDEs are attractive pharmacological targets in different diseases.

View this table:
  • View inline
  • View popup
TABLE 1

The phosphodiesterase (PDE) superfamily

PDE4 subfamily

The PDE4 subfamily contains four subtypes (PDE4A, B, C and D) (table 2), all of which hydrolyse cAMP [13, 14, 16]. PDE4 subtypes are highly expressed in the brain, cardiovascular tissue, smooth muscle and keratinocytes (figure 1) [13, 14, 16]. Inhibition of PDE4 subtypes increases intracellular levels of cAMP, affecting inflammatory and immune responses and fibrotic processes [17], including reducing the release of pro-inflammatory mediators and recruitment of inflammatory cells [18]. As such, it is a potential target for treating pulmonary fibrosis.

View this table:
  • View inline
  • View popup
TABLE 2

The phosphodiesterase 4 (PDE4) subfamily

FIGURE 1
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1

Role of pan-phosphodiesterase 4 (PDE4)/PDE4B in homeostasis and disease. AIP: aromatic hydrocarbon receptor-interacting protein; β-AR: β2 adrenergic receptor; GH: growth hormone; PKA: protein kinase A; SNP: single nucleotide polymorphism; Th2: T-helper 2 cell; TSHR: thyroid-stimulating hormone receptor.

PDE4A is ubiquitous, with relatively high expression in the testes and brain [19–21]. All PDE4A variants (with the exception of PDE4A1) have been identified in the lung, particularly in fibroblasts, inflammatory cells and pulmonary artery smooth muscle [22–25]. However, the pattern of expression within those tissues appears to depend on the subtype variant; for example, PDE4A5 has been identified in primary human airway epithelial cells [26], whereas PDE4A8 is not present in either alveolar or mesenchymal cells [23]. PDE4B is also widely distributed and has a tissue-specific distribution pattern, with high levels of expression, particularly in the lung, immune cells and the brain [14, 20, 27]. Except for the brain-specific PDE4B5, all PDE4B subtypes have been identified in the lungs [27, 28]. In immune cells, the PDE4B and PDE4D subtypes predominate compared with PDE4A and PDE4C [14, 29], and PDE4B1 and PDE4B2 variants have been identified in bronchoalveolar macrophages, peripheral blood monocytes and T-lymphocytes [22]. PDE4C expression, which has been reported in the lung, testes and other tissues [14, 21], is more restricted compared with other PDE4 subtypes, and there appears to be no expression in inflammatory cells [20, 21]. However, available data are limited and, in general, expression of PDE4C variants is not fully understood. PDE4D is widely distributed, with high levels in the brain and several other tissue types [14]. Human PDE4D haplotypes and single nucleotide polymorphisms are correlated with ischaemic stroke [30] and response to short-acting bronchodilators [31]. mRNA transcripts for all PDE4D subtypes have been identified in the lungs, although expression of PDE4D4 and PDE4D6 is comparatively low compared with the other subtypes [32]. PDE4D3 has been reported in primary human airway epithelial cells [26], whereas PDE4D5 is highly expressed in well-differentiated human bronchial epithelium cells [33]. There is an unmet need for cell-specific analyses of the differential expression of PDE4 variants in the normal and fibrotic lung, including fibroblasts, epithelial cells and endothelial cells, to fully appreciate the role of these enzymes in the fibrotic process.

PDE4B as a pharmacological target

Role of PDE4 inhibition in intracellular signalling

PDE4 regulates the production of pro- and anti-inflammatory cytokines via cAMP degradation [16]. PDE4 interacts with cyclic nucleotide modulators, including protein kinase A (PKA) and exchange factors directly activated by cAMP (EPAC1/2), forming cAMP signalosomes that have multiple downstream effects on immune response, cell proliferation and differentiation [16]. PDE4 inhibition leads to the elevation of intracellular cAMP levels and activation of PKA and EPAC1/2, reducing the release of pro-inflammatory cytokine and increasing the synthesis of anti-inflammatory cytokines (figure 2). As discussed later, the antifibrotic effects of PDE4 inhibition are not fully known; however, it is hypothesised to inhibit lung myofibroblast transformation and proliferation and expression of extracellular matrix proteins [34].

FIGURE 2
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2

Hypothesised role of pan-phosphodiesterase 4 (PDE4)/PDE4B inhibition in treating lung fibrosis. Dotted lines indicated hypothesised role of PDE4B preferential inhibition on inflammatory and fibrotic pathways. ECM: extracellular matrix; EPAC1/2: exchange protein directly activated by cAMP 1/2; GPCR: G protein-coupled receptor; PKA: protein kinase A.

Pre-clinical evidence for targeting PDE4 in lung fibrosis

In a mouse model of lung fibrosis, PDE4 inhibitor administration demonstrated antifibrotic activity that was equivalent to that exhibited by the antifibrotic treatments pirfenidone and nintedanib [12]. PDE4 inhibitor treatment was also associated with a decrease in plasma surfactant protein D concentration, a reduction in the plasma levels of several chemokines implicated in lung fibrosis and an in vitro inhibition of fibroblast profibrotic gene expression [12]. In a canine model, a mangostanin-derived PDE4 inhibitor had comparable antifibrotic effects to pirfenidone in a bleomycin-induced rat model of pulmonary fibrosis [35] and did not result in emesis, unlike the nonselective PDE4 inhibitor roflumilast. Unlike nintedanib, the PDE4 inhibitor AA6216 was found to suppress pathogenic segregated-nucleus-containing atypical monocytes in the lung [36]. These produce tumour necrosis factor (TNF)-α and are involved in murine lung fibrosis. In another study, AA6216 significantly inhibited transforming growth factor (TGF)-β1 production by THP-1 cells, a human monocytic cell line, and suppressed TNF-α production by alveolar macrophages from patients with IPF [37]. In the same study, AA6216 reduced fibrosis scores, collagen-stained areas and TGF-β1 in bronchoalveolar lavage fluid in a mouse model of bleomycin-induced IPF [37]. In terms of inflammation, there was evidence of increased PDE4 expression in the lungs associated with airway inflammation in an allergy rat model [38]. These studies indicate the potential for PDE4 inhibitors in treating pulmonary fibrosis.

Pre-clinical evidence for targeting PDE4B in lung fibrosis

PDE4B has an important role in inflammation and fibrosis in vitro. Gene deletion studies have demonstrated that PDE4B knockout mice fail to develop airway inflammation (i.e. T-helper 2 (Th2) cytokine production and eosinophil infiltration) and airway hyperactivity in response to antigen-induced challenge with methacholine is absent in both PDE4B and PDE4D knockout mice [39, 40]. Deletion of PDE4B, but not PDE4A or PDE4D, significantly suppresses lipopolysaccharide (LPS)-induced TNF-α production in circulating monocytes and macrophages in bronchoalveolar lavage fluid and disrupts Th2-cell proliferation and cytokine production in cultured bronchial lymph node cells [39, 40]. PDE4B knockdown also inhibits cytokine-induced proliferation and myofibroblast conversion in human lung fibroblasts [41].

PDE4-selective inhibitors

PDE4 inhibitors are associated with anti-inflammatory and antifibrotic effects, and have the potential to reduce inflammation and fibrotic remodelling in lung diseases, as described above (“Pre-clinical evidence for targeting PDE4 in lung fibrosis”).

PDE4 has also been targeted for various inflammatory conditions, including asthma, COPD, psoriasis, atopic dermatitis, inflammatory bowel diseases, rheumatic arthritis, lupus and neuroinflammation. To date, three PDE4 inhibitors – roflumilast [42, 43], apremilast [44, 45] and crisaborole [46] – have been approved for various indications. Roflumilast and apremilast have an oral administration, whereas crisaborole is topical.

Pre-clinical evidence

In a mouse model of pulmonary fibrosis, roflumilast prevented the development of lung injury and alleviated the pulmonary fibrotic and vascular remodelling response to bleomycin in a therapeutic protocol [47]. In pre-clinical models of SSc, apremilast reduced inflammatory cell activity and the release of profibrotic cytokines from M2 macrophages, leading to decreased fibroblast activation and collagen release [48]. Apremilast has also suppressed production of inflammatory mediators, reduced oxidative stresses and fibrosis, and inhibited infiltration of immune cells into inflamed tissues in a mouse model of ulcerative colitis [49]. Piclamilast, an agent that was evaluated in early animal models but has not been developed clinically to date, reduced expression of profibrotic genes, including collagen 1A1, in the gastrocnemius and diaphragm in a mouse model of Duchenne muscular dystrophy [50]. PDE4 inhibition with either piclamilast or rolipram prolonged survival by inhibiting inflammation and reducing alveolar fibrin deposition in pre-term rat pups with neonatal hyperoxic lung injury [51].

Clinical evidence

Rolipram is a well-characterised, first-generation oral PDE4 inhibitor that was initially developed because of its antidepressant effects, but which has subsequently shown poor tolerability in clinical studies [52, 53]. A clinical trial of rolipram in patients with multiple sclerosis was terminated due to poor tolerability and safety concerns after an increase, rather than decrease, in brain inflammatory activity, measured by contrast-enhancing lesions on magnetic resonance imaging, was observed [52]. Adverse events included nausea, vomiting and insomnia.

Cilomilast is a potent oral inhibitor of PDE4 that has been investigated as a potential therapy in patients with COPD [53, 54]. However, there were mixed results, with only two of five pivotal phase III studies showing a significant reduction in COPD exacerbations following cilomilast treatment for 24 weeks [53]. There was also no effect on the incidence of COPD exacerbations in a study specifically powered for exacerbations; in addition, there was no significant effect on anti-inflammatory activity, assessed by sputum neutrophil levels [53, 55]. In all studies, gastrointestinal adverse events, including those that interfered with daily activities, were reported in those who took cilomilast, mostly occurring in the first 2 weeks after treatment initiation [53, 54]. Inconsistent results and the drug's adverse event profile led to the clinical development of cilomilast being terminated.

Inhaled PDE4 inhibitors have also been investigated, on the basis that direct administration to the lungs may reduce systemic effects [56]. Although most have been discontinued in clinical development, tanimilast has been investigated both in patients with asthma and COPD [57]. Tanimilast inhibited the allergen-induced late-asthmatic response and reduced biomarkers of airway inflammation in COPD, with a similar gastrointestinal adverse event profile to placebo; phase III trials in patients with COPD and chronic bronchitis are ongoing (clinicaltrials.gov NCT04636801; NCT04636814).

Marketed PDE4 inhibitors

Roflumilast is approved in the United States and European Union for the treatment of severe COPD associated with chronic bronchitis and a history of exacerbations [42, 43], and is currently the only PDE4 inhibitor approved for a pulmonary indication. In a pooled analysis of two phase III trials, roflumilast increased pre-bronchodilator forced expiratory volume in 1 s by 48 mL compared with a placebo [58]. The rate of exacerbations that were moderate or severe per patient per year was 1.14 with roflumilast and 1.37 with placebo (reduction 17%; 95% CI 8–25) [58]. Common adverse events seen in trials of roflumilast include headache and gastrointestinal symptoms, such as diarrhoea, weight loss and nausea [58, 59]. Concerns have also been raised regarding the potential for suicidal ideation following treatment with roflumilast [59]. Apremilast is a small-molecule PDE4 inhibitor approved for the treatment of psoriatic arthritis, plaque psoriasis and oral ulcers associated with Behçet's disease [44, 45]. The most frequently reported adverse events across the clinical trials were diarrhoea, nausea and headache [60–62]. Psychiatric adverse events were also reported, including depression and attempted suicide [60, 61]. Crisaborole is approved in the United States for the topical treatment of mild-to-moderate atopic dermatitis [46, 63, 64]. The mechanism via which PDE4 inhibition by crisaborole exerts a therapeutic effect is not well defined [46]. In summary, PDE4 inhibition thus offers a variety of potential therapeutic effects, but the use of oral PDE4 inhibitors can be complicated due to class-related systemic adverse events, including gastrointestinal and psychiatric symptoms [16, 37, 42–45, 56, 65].

Emesis as an adverse event

A potential strategy to reduce the adverse events associated with PDE4 inhibitors is to preferentially target the PDE4B subtype. As mentioned above, gastrointestinal side-effects, including vomiting, are known effects limiting the use of PDE4 inhibitors in humans. Both rolipram and cilomilast failed in clinical trials due to lack of efficacy, as well as dose-limiting nausea- and emesis-associated effects caused by the inhibition of PDE4 subtypes in the emetic centre in the brain [66]. A model of emesis in mice found that inhibition of PDE4D, but not PDE4B, may be responsible for the emetic effects of PDE4 inhibition [67]. In addition, cilomilast has tenfold greater inhibition for PDE4D than for PDE4A, B and C [68].

Pre-clinical evidence supporting the use of BI 1015550, a novel PDE4B preferential inhibitor, in pulmonary fibrosis

BI 1015550 is a novel PDE4 inhibitor that preferentially inhibits PDE4B, with roughly tenfold selectivity for PDE4B versus PDE4D compared with the pan-PDE4 inhibitor roflumilast [34]. Like roflumilast [69], BI 1015550 demonstrates anti-inflammatory activity, inhibiting TNF-α and IL-2 in human peripheral blood mononuclear cells, as well as inhibiting LPS-induced TNF-α synthesis in human and rat whole blood [34]. This anti-inflammatory activity was reflected in animal models, where BI 1015550 inhibited LPS-induced TNF-α synthesis ex vivo and in house musk shrews (Suncus murinus) by inhibiting neutrophil influx into bronchoalveolar lavage fluid stimulated by nebulised LPS [34].

In lung fibroblasts from patients with IPF, BI 1015550 inhibited TGF-β1-stimulated myofibroblast transformation, IL-1β-induced cell proliferation, mRNA expression of extracellular matrix proteins, as well as fibroblast growth factor [34]. BI 1015550 also reversed a decrease in pulmonary function in a therapeutic protocol in a mouse model of bleomycin-induced pulmonary fibrosis (41% improvement in forced vital capacity (FVC) versus vehicle; p<0.05). In addition, co-administration with the antifibrotic nintedanib appeared to be synergistic, resulting in a tenfold shift of the concentration–response curve to the left. The combination of BI 1015550 and pirfenidone did not appear to have additional inhibitory effects in vitro [34].

Pre-clinical studies with BI 1015550, have indicated improved tolerability compared with roflumilast. Suncus murinus is an insectivore used as a model of emesis [70, 71] and has previously been used to investigate emesis with the PDE4 inhibitors rolipram and piclamast [72]. Using the S. murinus model, the therapeutic ratio for BI 1015550 appeared to be substantially improved compared with roflumilast, a pan-PDE4 inhibitor (0.3 versus 0.7 mean emetic events per animal, respectively) [34].

Taken together, these findings suggest that treatment with a preferential inhibitor of the subtype PDE4B, which is highly expressed in the lungs, should retain many beneficial anti-inflammatory and antifibrotic effects, with reduced adverse events compared with pan-PDE4 inhibitors [14, 41, 56, 73].

Clinical evidence for targeting PDE4B

Phase I trials with BI 1015550 in healthy male volunteers (n=42) and patients with IPF (n=15) found most adverse events to be either mild or moderate [74]. Gastrointestinal adverse events were the most common events and were more frequent in patients treated with BI 1015550 than placebo (80% versus 40%, respectively) [74]. One patient with IPF had a severe adverse event of insomnia that was considered treatment-related and was the only adverse event leading to discontinuation of BI 1015550 in the trial. In terms of pharmacokinetics, exposure to BI 1015550 increased proportionally with dose administered. 95% of the steady-state concentration was reached after approximately five administrations at 18 mg twice a day, and there was no difference in pharmacokinetic parameters between healthy volunteers and patients with IPF [74].

In a phase II trial conducted in patients with IPF (n=147), BI 1015550 stabilised lung function compared with a decline in the placebo arm, following 12 weeks of treatment [75]. Among patients without background antifibrotic use, FVC median change was +5.7 mL (95% credible interval −39.1–50.5) in the active treatment arm compared with −81.7 mL (95% credible interval −133.5–−44.8) in the placebo arm. Similar effects were also observed in patients receiving background antifibrotic treatment. In terms of safety, adverse events were more frequent in patients treated with BI 1015550 compared with placebo for those with and without background antifibrotics. The most common adverse event was diarrhoea of mild intensity. Phase III studies to further characterise the efficacy, safety and tolerability profile of this preferential PDE4B inhibitor in patients with IPF (clinicaltrials.gov NCT05321069) and progressive fibrosing ILD (NCT05321082) started in Q3 2022.

Questions for future research

Currently, there are no long-term clinical data for preferential PDE4B inhibitors, although phase III trials are underway in patients with IPF and other types of progressive pulmonary fibrosis. Additional information is required to evaluate the suitability of preferential PDE4B inhibitors either alone or in combination with background antifibrotics as treatments for pulmonary fibrosis.

Conclusions

In conclusion, pre-clinical and early-phase clinical evidence supports the rationale for targeting PDE4B in pulmonary fibrosis. Preferential inhibition of PDE4B results in anti-inflammatory and antifibrotic effects with the anticipation of an improved safety profile compared with pan-PDE4 inhibitors. Combination therapy with a preferential PDE4B inhibitor and existing treatments, such as nintedanib and pirfenidone, has the potential to improve clinical outcomes for patients with IPF and other forms of progressive pulmonary fibrosis. Further investigation of preferential PDE4B inhibition in a larger patient population and for a longer treatment period is warranted.

Acknowledgements

The authors meet criteria for authorship as recommended by the International Committee of Medical Journal Editors (ICMJE). The authors did not receive payment related to the development of the review. Helen Keyworth, PhD, CMPP, of Nucleus Global provided writing, editorial support and formatting assistance, which was contracted and funded by Boehringer Ingelheim International GmbH (BI). BI was given the opportunity to review the manuscript for medical and scientific accuracy as well as intellectual property considerations.

Footnotes

  • Provenance: Submitted article, peer reviewed.

  • A graphical abstract and video to accompany this article is available from www.globalmedcomms.com/respiratory/Fibrosis_Review_Kolb

  • Conflict of interest: M. Kolb declares research funding for pre-clinical work from Boehringer Ingelheim and Pieris, and research funding for clinical work from Roche; consulting fees from Boehringer Ingelheim, Roche, Horizon, Cipla, AbbVie, Bellerophon, Algernon, CSL Behring, United Therapeutics and LabCorp; payment or honoraria from Roche, Novartis and Boehringer Ingelheim; payment for expert testimony from Roche; participation on a Data Safety Monitoring Board or Advisory Board for United Therapeutics and LabCorp; and received an allowance as the chief editor of the European Respiratory Journal.

  • Conflict of interest: B. Crestani declares grants or contracts from Bristol-Myers Squibb, Boehringer Ingelheim and Roche; personal consulting fees from Apellis; payment or honoraria from Boehringer Ingelheim, Bristol-Myers Squibb, Roche, Sanofi, Novartis, AstraZeneca and Chiesi; and receipt of equipment, materials, drugs or other services from Translate Bio.

  • Conflict of interest: T.M. Maher declares consulting fees from Boehringer Ingelheim, Roche/Genentech, AstraZeneca, Bayer, Blade Therapeutics, Bristol-Myers Squibb, Galapagos, Galecto, GlaxoSmithKline, IQVIA, Pliant, Respivant, Theravance and Veracyte; and payment or honoraria from Boehringer Ingelheim and Roche/Genentech.

  • Support statement: Funding for medical writing support was provided by Boehringer Ingelheim International GmbH. Funding information for this article has been deposited with the Crossref Funder Registry.

  • Received October 24, 2022.
  • Accepted December 4, 2022.
  • Copyright ©The authors 2023
http://creativecommons.org/licenses/by/4.0/

This version is distributed under the terms of the Creative Commons Attribution Licence 4.0.

References

  1. ↵
    1. Westra IM,
    2. Pham BT,
    3. Groothuis GM, et al.
    Evaluation of fibrosis in precision-cut tissue slices. Xenobiotica 2013; 43: 98–112. doi:10.3109/00498254.2012.723151
    OpenUrlCrossRefPubMed
    1. Wynn TA
    . Integrating mechanisms of pulmonary fibrosis. J Exp Med 2011; 208: 1339–1350. doi:10.1084/jem.20110551
    OpenUrlAbstract/FREE Full Text
  2. ↵
    1. Ley B,
    2. Collard HR,
    3. King TE, Jr.
    . Clinical course and prediction of survival in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2011; 183: 431–440. doi:10.1164/rccm.201006-0894CI
    OpenUrlCrossRefPubMed
  3. ↵
    1. Wong AW,
    2. Ryerson CJ,
    3. Guler SA
    . Progression of fibrosing interstitial lung disease. Respir Res 2020; 21: 32. doi:10.1186/s12931-020-1296-3
    OpenUrlCrossRefPubMed
  4. ↵
    1. Raghu G,
    2. Remy-Jardin M,
    3. Richeldi L, et al.
    Idiopathic pulmonary fibrosis (an update) and progressive pulmonary fibrosis in adults: an official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med 2022; 205: e18–e47. doi:10.1164/rccm.202202-0399ST
    OpenUrlCrossRefPubMed
  5. ↵
    1. European Medicines Agency
    . Esbriet® (pirfenidone): summary of product characteristics. 2022. Available from: www.ema.europa.eu/en/documents/product-information/esbriet-epar-product-information_en.pdf Date last accessed: 5 August 2022.
  6. ↵
    1. US Food and Drug Administration
    . Esbriet® (pirfenidone): prescribing information. 2022. Available from: www.accessdata.fda.gov/drugsatfda_docs/label/2019/022535s012,208780s002lbl.pdf Date last accessed: 12 August 2022. Date last updated: July 2019.
  7. ↵
    1. European Medicines Agency
    . Ofev® (nintedanib): summary of product characteristics. 2021. Available from: www.ema.europa.eu/en/documents/product-information/ofev-epar-product-information_en.pdf Date last accessed: 12 August 2022. Date last updated: September 2021.
  8. ↵
    1. US Food and Drug Administration
    . Ofev® (nintedanib): prescribing information. 2020. Available from: www.accessdata.fda.gov/drugsatfda_docs/label/2020/205832s014lbl.pdf Date last accessed: 17 August 2022. Date last updated: October 2020.
  9. ↵
    1. US Food and Drug Administration
    . Actemra® (tocilizumab): prescribing information. 2021. Available from: www.accessdata.fda.gov/drugsatfda_docs/label/2021/125276s131lbl.pdf Date last accessed: 14 June 2022. Date last updated: March 2021.
  10. ↵
    1. Ghumman M,
    2. Dhamecha D,
    3. Gonsalves A, et al.
    Emerging drug delivery strategies for idiopathic pulmonary fibrosis treatment. Eur J Pharm Biopharm 2021; 164: 1–12. doi:10.1016/j.ejpb.2021.03.017
    OpenUrl
  11. ↵
    1. Sisson TH,
    2. Christensen PJ,
    3. Muraki Y, et al.
    Phosphodiesterase 4 inhibition reduces lung fibrosis following targeted type II alveolar epithelial cell injury. Physiol Rep 2018; 6: e13753. doi:10.14814/phy2.13753
    OpenUrl
  12. ↵
    1. Azevedo MF,
    2. Faucz FR,
    3. Bimpaki E, et al.
    Clinical and molecular genetics of the phosphodiesterases (PDEs). Endocr Rev 2014; 35: 195–233. doi:10.1210/er.2013-1053
    OpenUrlCrossRefPubMed
  13. ↵
    1. Bender AT,
    2. Beavo JA
    . Cyclic nucleotide phosphodiesterases: molecular regulation to clinical use. Pharmacol Rev 2006; 58: 488–520. doi:10.1124/pr.58.3.5
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Francis SH,
    2. Blount MA,
    3. Corbin JD
    . Mammalian cyclic nucleotide phosphodiesterases: molecular mechanisms and physiological functions. Physiol Rev 2011; 91: 651–690. doi:10.1152/physrev.00030.2010
    OpenUrlCrossRefPubMed
  15. ↵
    1. Li H,
    2. Zuo J,
    3. Tang W
    . Phosphodiesterase-4 inhibitors for the treatment of inflammatory diseases. Front Pharmacol 2018; 9: 1048. doi:10.3389/fphar.2018.01048
    OpenUrl
  16. ↵
    1. Maurice DH,
    2. Ke H,
    3. Ahmad F, et al.
    Advances in targeting cyclic nucleotide phosphodiesterases. Nat Rev Drug Discov 2014; 13: 290–314. doi:10.1038/nrd4228
    OpenUrlCrossRefPubMed
  17. ↵
    1. Zuo H,
    2. Cattani-Cavalieri I,
    3. Musheshe N, et al.
    Phosphodiesterases as therapeutic targets for respiratory diseases. Pharmacol Ther 2019; 197: 225–242. doi:10.1016/j.pharmthera.2019.02.002
    OpenUrl
  18. ↵
    1. Bolger G,
    2. Michaeli T,
    3. Martins T, et al.
    A family of human phosphodiesterases homologous to the dunce learning and memory gene product of Drosophila melanogaster are potential targets for antidepressant drugs. Mol Cell Biol 1993; 13: 6558–6571.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Engels P,
    2. Fichtel K,
    3. Lubbert H
    . Expression and regulation of human and rat phosphodiesterase type IV isogenes. FEBS Lett 1994; 350: 291–295. doi:10.1016/0014-5793(94)00788-8
    OpenUrlCrossRefPubMed
  20. ↵
    1. Engels P,
    2. Sullivan M,
    3. Muller T, et al.
    Molecular cloning and functional expression in yeast of a human cAMP-specific phosphodiesterase subtype (PDE IV-C). FEBS Lett 1995; 358: 305–310. doi:10.1016/0014-5793(94)01460-I
    OpenUrlCrossRefPubMed
  21. ↵
    1. Barber R,
    2. Baillie GS,
    3. Bergmann R, et al.
    Differential expression of PDE4 cAMP phosphodiesterase isoforms in inflammatory cells of smokers with COPD, smokers without COPD, and nonsmokers. Am J Physiol Lung Cell Mol Physiol 2004; 287: L332–L343. doi:10.1152/ajplung.00384.2003
    OpenUrlCrossRefPubMed
  22. ↵
    1. Mackenzie KF,
    2. Topping EC,
    3. Bugaj-Gaweda B, et al.
    Human PDE4A8, a novel brain-expressed PDE4 cAMP-specific phosphodiesterase that has undergone rapid evolutionary change. Biochem J 2008; 411: 361–369. doi:10.1042/BJ20071251
    OpenUrlAbstract/FREE Full Text
    1. Millen J,
    2. MacLean MR,
    3. Houslay MD
    . Hypoxia-induced remodelling of PDE4 isoform expression and cAMP handling in human pulmonary artery smooth muscle cells. Eur J Cell Biol 2006; 85: 679–691. doi:10.1016/j.ejcb.2006.01.006
    OpenUrlCrossRefPubMed
  23. ↵
    1. Sachs BD,
    2. Baillie GS,
    3. McCall JR, et al.
    p75 neurotrophin receptor regulates tissue fibrosis through inhibition of plasminogen activation via a PDE4/cAMP/PKA pathway. J Cell Biol 2007; 177: 1119–1132. doi:10.1083/jcb.200701040
    OpenUrlAbstract/FREE Full Text
  24. ↵
    1. Wright LC,
    2. Seybold J,
    3. Robichaud A, et al.
    Phosphodiesterase expression in human epithelial cells. Am J Physiol 1998; 275: L694–L700. doi:10.1152/ajplung.1998.275.4.L694
    OpenUrl
  25. ↵
    1. Shepherd M,
    2. McSorley T,
    3. Olsen AE, et al.
    Molecular cloning and subcellular distribution of the novel PDE4B4 cAMP-specific phosphodiesterase isoform. Biochem J 2003; 370: 429–438. doi:10.1042/bj20021082
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Cheung YF,
    2. Kan Z,
    3. Garrett-Engele P, et al.
    PDE4B5, a novel, super-short, brain-specific cAMP phosphodiesterase-4 variant whose isoform-specifying N-terminal region is identical to that of cAMP phosphodiesterase-4D6 (PDE4D6). J Pharmacol Exp Ther 2007; 322: 600–609. doi:10.1124/jpet.107.122218
    OpenUrlAbstract/FREE Full Text
  27. ↵
    1. Wang P,
    2. Wu P,
    3. Ohleth KM, et al.
    Phosphodiesterase 4B2 is the predominant phosphodiesterase species and undergoes differential regulation of gene expression in human monocytes and neutrophils. Mol Pharmacol 1999; 56: 170–174. doi:10.1124/mol.56.1.170
    OpenUrlAbstract/FREE Full Text
  28. ↵
    1. Gretarsdottir S,
    2. Thorleifsson G,
    3. Reynisdottir ST, et al.
    The gene encoding phosphodiesterase 4D confers risk of ischemic stroke. Nat Genet 2003; 35: 131–138. doi:10.1038/ng1245
    OpenUrlCrossRefPubMed
  29. ↵
    1. Labuda M,
    2. Laberge S,
    3. Briere J, et al.
    Phosphodiesterase type 4D gene polymorphism: association with the response to short-acting bronchodilators in paediatric asthma patients. Mediators Inflamm 2011; 2011: 301695. doi:10.1155/2011/301695
    OpenUrlPubMed
  30. ↵
    1. Richter W,
    2. Jin SL,
    3. Conti M
    . Splice variants of the cyclic nucleotide phosphodiesterase PDE4D are differentially expressed and regulated in rat tissue. Biochem J 2005; 388: 803–811. doi:10.1042/BJ20050030
    OpenUrlAbstract/FREE Full Text
  31. ↵
    1. Barnes AP,
    2. Livera G,
    3. Huang P, et al.
    Phosphodiesterase 4D forms a cAMP diffusion barrier at the apical membrane of the airway epithelium. J Biol Chem 2005; 280: 7997–8003. doi:10.1074/jbc.M407521200
    OpenUrlAbstract/FREE Full Text
  32. ↵
    1. Herrmann FE,
    2. Hesslinger C,
    3. Wollin L, et al.
    BI 1015550 is a PDE4B inhibitor and a clinical drug candidate for the oral treatment of idiopathic pulmonary fibrosis. Front Pharmacol 2022; 13: 838449. doi:10.3389/fphar.2022.838449
    OpenUrl
  33. ↵
    1. Huang YY,
    2. Deng J,
    3. Tian YJ, et al.
    Mangostanin derivatives as novel and orally active phosphodiesterase 4 inhibitors for the treatment of idiopathic pulmonary fibrosis with improved safety. J Med Chem 2021; 64: 13736–13751. doi:10.1021/acs.jmedchem.1c01085
    OpenUrl
  34. ↵
    1. Matsuhira T,
    2. Nishiyama O,
    3. Tabata Y, et al.
    The phosphodiesterase 4 inhibitor AA6216 suppresses activity of fibrosis-specific macrophages. Biochem Biophys Rep 2021; 28: 101118. doi:10.1016/j.bbrep.2021.101118
    OpenUrl
  35. ↵
    1. Matsuhira T,
    2. Nishiyama O,
    3. Tabata Y, et al.
    A novel phosphodiesterase 4 inhibitor, AA6216, reduces macrophage activity and fibrosis in the lung. Eur J Pharmacol 2020; 885: 173508. doi:10.1016/j.ejphar.2020.173508
    OpenUrl
  36. ↵
    1. Tang HF,
    2. Chen JQ,
    3. Xie QM, et al.
    The role of PDE4 in pulmonary inflammation and goblet cell hyperplasia in allergic rats. Biochim Biophys Acta 2006; 1762: 525–532. doi:10.1016/j.bbadis.2005.12.009
    OpenUrlPubMed
  37. ↵
    1. Jin SL,
    2. Goya S,
    3. Nakae S, et al.
    Phosphodiesterase 4B is essential for T(H)2-cell function and development of airway hyperresponsiveness in allergic asthma. J Allergy Clin Immunol 2010; 126: 1252–1259.e1212. doi:10.1016/j.jaci.2010.08.014
    OpenUrlCrossRefPubMed
  38. ↵
    1. Hansen G,
    2. Jin S,
    3. Umetsu DT, et al.
    Absence of muscarinic cholinergic airway responses in mice deficient in the cyclic nucleotide phosphodiesterase PDE4D. Proc Natl Acad Sci USA 2000; 97: 6751–6756. doi:10.1073/pnas.97.12.6751
    OpenUrlAbstract/FREE Full Text
  39. ↵
    1. Selige J,
    2. Hatzelmann A,
    3. Dunkern T
    . The differential impact of PDE4 subtypes in human lung fibroblasts on cytokine-induced proliferation and myofibroblast conversion. J Cell Physiol 2011; 226: 1970–1980. doi:10.1002/jcp.22529
    OpenUrlCrossRefPubMed
  40. ↵
    1. European Medicines Agency
    . Daxas® (roflumilast): summary of product characteristics. 2022. Available from: www.ema.europa.eu/en/documents/product-information/daxas-epar-product-information_en.pdf Date last accessed: 2 August 2022. Date last updated: June 2022.
  41. ↵
    1. US Food and Drug Administration
    . Daliresp® (roflumilast): prescribing information. 2018. Available from: www.accessdata.fda.gov/drugsatfda_docs/label/2018/022522s009lbl.pdf Date last accessed: 28 January 2022. Date last updated: January 2018.
  42. ↵
    1. European Medicines Agency
    . Otezla® (apremilast): summary of product characteristics. 2022. Available from: www.ema.europa.eu/en/documents/product-information/otezla-epar-product-information_en.pdf Date last accessed: 2 August 2022. Date last updated: July 2022.
  43. ↵
    1. US Food and Drug Administration
    . Otezla® (apremilast): prescribing information. 2021. Available from: www.accessdata.fda.gov/drugsatfda_docs/label/2021/205437s011lbl.pdf Date last accessed: 23 February 2022. Date last updated: December 2021.
  44. ↵
    1. US Food and Drug Administration
    . EucrisaTM (crisaborole): prescribing information. 2021. Available from: www.accessdata.fda.gov/drugsatfda_docs/label/2020/207695s007s009s010lbl.pdf Date last accessed: 28 January 2022. Date last updated: March 2020.
  45. ↵
    1. Cortijo J,
    2. Iranzo A,
    3. Milara X, et al.
    Roflumilast, a phosphodiesterase 4 inhibitor, alleviates bleomycin-induced lung injury. Br J Pharmacol 2009; 156: 534–544. doi:10.1111/j.1476-5381.2008.00041.x
    OpenUrlCrossRefPubMed
  46. ↵
    1. Maier C,
    2. Ramming A,
    3. Bergmann C, et al.
    Inhibition of phosphodiesterase 4 (PDE4) reduces dermal fibrosis by interfering with the release of interleukin-6 from M2 macrophages. Ann Rheum Dis 2017; 76: 1133–1141. doi:10.1136/annrheumdis-2016-210189
    OpenUrlAbstract/FREE Full Text
  47. ↵
    1. Li H,
    2. Fan C,
    3. Feng C, et al.
    Inhibition of phosphodiesterase-4 attenuates murine ulcerative colitis through interference with mucosal immunity. Br J Pharmacol 2019; 176: 2209–2226. doi:10.1111/bph.14667
    OpenUrlPubMed
  48. ↵
    1. Nio Y,
    2. Tanaka M,
    3. Hirozane Y, et al.
    Phosphodiesterase 4 inhibitor and phosphodiesterase 5 inhibitor combination therapy has antifibrotic and anti-inflammatory effects in mdx mice with Duchenne muscular dystrophy. FASEB J 2017; 31: 5307–5320. doi:10.1096/fj.201700249R
    OpenUrlCrossRefPubMed
  49. ↵
    1. de Visser YP,
    2. Walther FJ,
    3. Laghmani EH, et al.
    Phosphodiesterase-4 inhibition attenuates pulmonary inflammation in neonatal lung injury. Eur Respir J 2008; 31: 633–644. doi:10.1183/09031936.00071307
    OpenUrlAbstract/FREE Full Text
  50. ↵
    1. Bielekova B,
    2. Richert N,
    3. Howard T, et al.
    Treatment with the phosphodiesterase type-4 inhibitor rolipram fails to inhibit blood–brain barrier disruption in multiple sclerosis. Mult Scler 2009; 15: 1206–1214. doi:10.1177/1352458509345903
    OpenUrlCrossRefPubMed
  51. ↵
    1. Rennard S,
    2. Knobil K,
    3. Rabe KF, et al.
    The efficacy and safety of cilomilast in COPD. Drugs 2008; 68: Suppl. 2, 3–57. doi:10.2165/0003495-200868002-00002
    OpenUrlPubMed
  52. ↵
    1. Rennard SI,
    2. Schachter N,
    3. Strek M, et al.
    Cilomilast for COPD: results of a 6-month, placebo-controlled study of a potent, selective inhibitor of phosphodiesterase 4. Chest 2006; 129: 56–66. doi:10.1378/chest.129.1.56
    OpenUrlCrossRefPubMed
  53. ↵
    1. GlaxoSmithKline
    . A randomised, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of oral cilomilast (15 mg bd) when given as maintenance treatment for 12 months to subjects with chronic obstructive pulmonary disease (COPD). 2018. Available from: www.gsk-studyregister.com/en/trial-details/?id=207499/157 Date last accessed: 21 March 2022. Date last updated: 4 November 2018.
  54. ↵
    1. Phillips JE
    . Inhaled phosphodiesterase 4 (PDE4) inhibitors for inflammatory respiratory diseases. Front Pharmacol 2020; 11: 259. doi:10.3389/fphar.2020.00259
    OpenUrl
  55. ↵
    1. Facchinetti F,
    2. Civelli M,
    3. Singh D, et al.
    Tanimilast, a novel inhaled PDE4 inhibitor for the treatment of asthma and chronic obstructive pulmonary disease. Front Pharmacol 2021; 12: 740803. doi:10.3389/fphar.2021.740803
    OpenUrl
  56. ↵
    1. Calverley PM,
    2. Rabe KF,
    3. Goehring UM, et al.
    Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet 2009; 374: 685–694. doi:10.1016/S0140-6736(09)61255-1
    OpenUrlCrossRefPubMed
  57. ↵
    1. Rabe KF
    . Update on roflumilast, a phosphodiesterase 4 inhibitor for the treatment of chronic obstructive pulmonary disease. Br J Pharmacol 2011; 163: 53–67. doi:10.1111/j.1476-5381.2011.01218.x
    OpenUrlCrossRefPubMed
  58. ↵
    1. Kavanaugh A,
    2. Gladman DD,
    3. Edwards CJ, et al.
    Long-term experience with apremilast in patients with psoriatic arthritis: 5-year results from a PALACE 1–3 pooled analysis. Arthritis Res Ther 2019; 21: 118. doi:10.1186/s13075-019-1901-3
    OpenUrl
  59. ↵
    1. Crowley J,
    2. Thaci D,
    3. Joly P, et al.
    Long-term safety and tolerability of apremilast in patients with psoriasis: pooled safety analysis for ≥156 weeks from 2 phase 3, randomized, controlled trials (ESTEEM 1 and 2). J Am Acad Dermatol 2017; 77: 310–317.e1. doi:10.1016/j.jaad.2017.01.052
    OpenUrlPubMed
  60. ↵
    1. Hatemi G,
    2. Mahr A,
    3. Takeno M, et al.
    Apremilast for oral ulcers associated with active Behcet's syndrome over 68 weeks: long-term results from a phase 3 randomised clinical trial. Clin Exp Rheumatol 2021; 39: Suppl. 132, 80–87. doi:10.55563/clinexprheumatol/ra8ize
    OpenUrl
  61. ↵
    1. Paller AS,
    2. Tom WL,
    3. Lebwohl MG, et al.
    Efficacy and safety of crisaborole ointment, a novel, nonsteroidal phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in children and adults. J Am Acad Dermatol 2016; 75: 494–503.e6. doi:10.1016/j.jaad.2016.05.046
    OpenUrlCrossRefPubMed
  62. ↵
    1. Simpson EL,
    2. Paller AS,
    3. Boguniewicz M, et al.
    Crisaborole ointment improves quality of life of patients with mild to moderate atopic dermatitis and their families. Dermatol Ther 2018; 8: 605–619. doi:10.1007/s13555-018-0263-0
    OpenUrl
  63. ↵
    1. Cazzola M,
    2. Calzetta L,
    3. Rogliani P, et al.
    The discovery of roflumilast for the treatment of chronic obstructive pulmonary disease. Expert Opin Drug Discov 2016; 11: 733–744. doi:10.1080/17460441.2016.1184642
    OpenUrl
  64. ↵
    1. Giembycz MA
    . 4D or not 4D – the emetogenic basis of PDE4 inhibitors uncovered? Trends Pharmacol Sci 2002; 23: 548. doi:10.1016/S0165-6147(02)02089-8
    OpenUrlPubMed
  65. ↵
    1. Robichaud A,
    2. Stamatiou PB,
    3. Jin SL, et al.
    Deletion of phosphodiesterase 4D in mice shortens alpha(2)-adrenoceptor-mediated anesthesia, a behavioral correlate of emesis. J Clin Invest 2002; 110: 1045–1052. doi:10.1172/JCI0215506
    OpenUrlCrossRefPubMed
  66. ↵
    1. Giembycz MA
    . Cilomilast: a second generation phosphodiesterase 4 inhibitor for asthma and chronic obstructive pulmonary disease. Expert Opin Investig Drugs 2001; 10: 1361–1379. doi:10.1517/13543784.10.7.1361
    OpenUrlCrossRefPubMed
  67. ↵
    1. Hatzelmann A,
    2. Schudt C
    . Anti-inflammatory and immunomodulatory potential of the novel PDE4 inhibitor roflumilast in vitro. J Pharmacol Exp Ther 2001; 297: 267–279.
    OpenUrlAbstract/FREE Full Text
  68. ↵
    1. Ueno S,
    2. Matsuki N,
    3. Saito H
    . Suncus murinus: a new experimental model in emesis research. Life Sci 1987; 41: 513–518. doi:10.1016/0024-3205(87)90229-3
    OpenUrlCrossRefPubMed
  69. ↵
    1. Ebukuro S,
    2. Wakana S,
    3. Hioki K, et al.
    Selective breeding of house musk shrew (Suncus murinus) lines in relation to emesis induced by veratrine sulfate. Comp Med 2000; 50: 281–283.
    OpenUrlPubMed
  70. ↵
    1. Hirose R,
    2. Manabe H,
    3. Nonaka H, et al.
    Correlation between emetic effect of phosphodiesterase 4 inhibitors and their occupation of the high-affinity rolipram binding site in Suncus murinus brain. Eur J Pharmacol 2007; 573: 93–99. doi:10.1016/j.ejphar.2007.06.045
    OpenUrlCrossRefPubMed
  71. ↵
    1. Ti H,
    2. Zhou Y,
    3. Liang X, et al.
    Targeted treatments for chronic obstructive pulmonary disease (COPD) using low-molecular-weight drugs (LMWDs). J Med Chem 2019; 62: 5944–5978. doi:10.1021/acs.jmedchem.8b01520
    OpenUrl
  72. ↵
    1. Maher TM,
    2. Schlecker C,
    3. Luedtke D, et al.
    Phase I studies of BI 1015550, a preferential phosphodiesterase 4B inhibitor, in healthy males and patients with idiopathic pulmonary fibrosis. ERJ Open Res 2022; 8: 00240–2022. doi:10.1183/23120541.00240-2022
    OpenUrl
  73. ↵
    1. Richeldi L,
    2. Azuma A,
    3. Cottin V, et al.
    Trial of a preferential phosphodiesterase 4b inhibitor for idiopathic pulmonary fibrosis. N Engl J Med 2022; 386: 2178–2187. doi:10.1056/NEJMoa2201737
    OpenUrl
    1. Lefievre L,
    2. de Lamirande E,
    3. Gagnon C
    . Presence of cyclic nucleotide phosphodiesterases PDE1A, existing as a stable complex with calmodulin, and PDE3A in human spermatozoa. Biol Reprod 2002; 67: 423–430. doi:10.1095/biolreprod67.2.423
    OpenUrlCrossRefPubMed
    1. Kanda N,
    2. Watanabe S
    . Regulatory roles of adenylate cyclase and cyclic nucleotide phosphodiesterases 1 and 4 in interleukin-13 production by activated human T cells. Biochem Pharmacol 2001; 62: 495–507. doi:10.1016/S0006-2952(01)00688-8
    OpenUrlCrossRefPubMed
    1. Reed TM,
    2. Repaske DR,
    3. Snyder GL, et al.
    Phosphodiesterase 1B knock-out mice exhibit exaggerated locomotor hyperactivity and DARPP-32 phosphorylation in response to dopamine agonists and display impaired spatial learning. J Neurosci 2002; 22: 5188–5197. doi:10.1523/JNEUROSCI.22-12-05188.2002
    OpenUrlAbstract/FREE Full Text
    1. Deng C,
    2. Wang D,
    3. Bugaj-Gaweda B, et al.
    Assays for cyclic nucleotide-specific phosphodiesterases (PDEs) in the central nervous system (PDE1, PDE2, PDE4, and PDE10). Curr Protoc Neurosci 2007; Chapter 7: Unit 7.21. doi:10.1002/0471142301.ns0721s38
    OpenUrl
    1. Rybalkin SD,
    2. Rybalkina I,
    3. Beavo JA, et al.
    Cyclic nucleotide phosphodiesterase 1C promotes human arterial smooth muscle cell proliferation. Circ Res 2002; 90: 151–157. doi:10.1161/hh0202.104108
    OpenUrlAbstract/FREE Full Text
    1. Nagel DJ,
    2. Aizawa T,
    3. Jeon KI, et al.
    Role of nuclear Ca2+/calmodulin-stimulated phosphodiesterase 1A in vascular smooth muscle cell growth and survival. Circ Res 2006; 98: 777–784. doi:10.1161/01.RES.0000215576.27615.fd
    OpenUrlAbstract/FREE Full Text
    1. Mongillo M,
    2. Tocchetti CG,
    3. Terrin A, et al.
    Compartmentalized phosphodiesterase-2 activity blunts beta-adrenergic cardiac inotropy via an NO/cGMP-dependent pathway. Circ Res 2006; 98: 226–234. doi:10.1161/01.RES.0000200178.34179.93
    OpenUrlAbstract/FREE Full Text
    1. Sadhu K,
    2. Hensley K,
    3. Florio VA, et al.
    Differential expression of the cyclic GMP-stimulated phosphodiesterase PDE2A in human venous and capillary endothelial cells. J Histochem Cytochem 1999; 47: 895–906. doi:10.1177/002215549904700707
    OpenUrlCrossRefPubMed
    1. Spiessberger B,
    2. Bernhard D,
    3. Herrmann S, et al.
    cGMP-dependent protein kinase II and aldosterone secretion. FEBS J 2009; 276: 1007–1013. doi:10.1111/j.1742-4658.2008.06839.x
    OpenUrlCrossRefPubMed
    1. Degerman E,
    2. Belfrage P,
    3. Manganiello VC
    . Structure, localization, and regulation of cGMP-inhibited phosphodiesterase (PDE3). J Biol Chem 1997; 272: 6823–6826. doi:10.1074/jbc.272.11.6823
    OpenUrlFREE Full Text
    1. Ahmad F,
    2. Degerman E,
    3. Manganiello VC
    . Cyclic nucleotide phosphodiesterase 3 signaling complexes. Horm Metab Res 2012; 44: 776–785. doi:10.1055/s-0032-1312646
    OpenUrlCrossRefPubMed
    1. Begum N,
    2. Shen W,
    3. Manganiello V
    . Role of PDE3A in regulation of cell cycle progression in mouse vascular smooth muscle cells and oocytes: implications in cardiovascular diseases and infertility. Curr Opin Pharmacol 2011; 11: 725–729. doi:10.1016/j.coph.2011.10.006
    OpenUrlCrossRefPubMed
    1. Omar B,
    2. Banke E,
    3. Guirguis E, et al.
    Regulation of the pro-inflammatory cytokine osteopontin by GIP in adipocytes–a role for the transcription factor NFAT and phosphodiesterase 3B. Biochem Biophys Res Commun 2012; 425: 812–817. doi:10.1016/j.bbrc.2012.07.157
    OpenUrlCrossRefPubMed
    1. Sallstrom J,
    2. Jensen BL,
    3. Skott O, et al.
    Neuronal nitric oxide synthase supports Renin release during sodium restriction through inhibition of phosphodiesterase 3. Am J Hypertens 2010; 23: 1241–1246. doi:10.1038/ajh.2010.153
    OpenUrlCrossRefPubMed
    1. Beca S,
    2. Ahmad F,
    3. Shen W, et al.
    Phosphodiesterase type 3A regulates basal myocardial contractility through interacting with sarcoplasmic reticulum calcium ATPase type 2a signaling complexes in mouse heart. Circ Res 2013; 112: 289–297. doi:10.1161/CIRCRESAHA.111.300003
    OpenUrlAbstract/FREE Full Text
    1. Fujita M,
    2. Hines CS,
    3. Zoghbi SS, et al.
    Downregulation of brain phosphodiesterase type IV measured with 11C-(R)-rolipram positron emission tomography in major depressive disorder. Biol Psychiatry 2012; 72: 548–554. doi:10.1016/j.biopsych.2012.04.030
    OpenUrlCrossRefPubMed
    1. Houslay MD
    . The long and short of vascular smooth muscle phosphodiesterase-4 as a putative therapeutic target. Mol Pharmacol 2005; 68: 563–567. doi:10.1124/mol.105.015719
    OpenUrlAbstract/FREE Full Text
    1. McKenna SD,
    2. Pietropaolo M,
    3. Tos EG, et al.
    Pharmacological inhibition of phosphodiesterase 4 triggers ovulation in follicle-stimulating hormone-primed rats. Endocrinology 2005; 146: 208–214. doi:10.1210/en.2004-0562
    OpenUrlCrossRefPubMed
    1. Santos-Silva AJ,
    2. Cairrao E,
    3. Morgado M, et al.
    PDE4 and PDE5 regulate cyclic nucleotides relaxing effects in human umbilical arteries. Eur J Pharmacol 2008; 582: 102–109. doi:10.1016/j.ejphar.2007.12.017
    OpenUrlCrossRefPubMed
    1. Luedders DW,
    2. Muenz BM,
    3. Li F, et al.
    Role of cGMP in sildenafil-induced activation of endothelial Ca2+-activated K+ channels. J Cardiovasc Pharmacol 2006; 47: 365–370. doi:10.1097/01.fjc.0000206438.35477.f2
    OpenUrlPubMed
    1. Reneerkens OA,
    2. Rutten K,
    3. Akkerman S, et al.
    Phosphodiesterase type 5 (PDE5) inhibition improves object recognition memory: indications for central and peripheral mechanisms. Neurobiol Learn Mem 2012; 97: 370–379. doi:10.1016/j.nlm.2012.02.008
    OpenUrlCrossRefPubMed
    1. Lin CS,
    2. Lau A,
    3. Tu R, et al.
    Expression of three isoforms of cGMP-binding cGMP-specific phosphodiesterase (PDE5) in human penile cavernosum. Biochem Biophys Res Commun 2000; 268: 628–635. doi:10.1006/bbrc.2000.2187
    OpenUrlCrossRefPubMed
    1. Christiansen JR,
    2. Ramamurthy V
    . Greasing the protein biosynthesis machinery of photoreceptor neurons: role for postprenylation processing of proteins. Cell Logist 2012; 2: 15–19. doi:10.4161/cl.19804
    OpenUrlPubMed
    1. Muradov KG,
    2. Granovsky AE,
    3. Artemyev NO
    . Mutation in rod PDE6 linked to congenital stationary night blindness impairs the enzyme inhibition by its gamma-subunit. Biochemistry 2003; 42: 3305–3310. doi:10.1021/bi027095x
    OpenUrlCrossRefPubMed
    1. Ekstrom L,
    2. Schulze JJ,
    3. Guillemette C, et al.
    Bioavailability of testosterone enanthate dependent on genetic variation in the phosphodiesterase 7B but not on the uridine 5ʹ-diphospho-glucuronosyltransferase (UGT2B17) gene. Pharmacogenet Genomics 2011; 21: 325–332. doi:10.1097/FPC.0b013e328344c5c6
    OpenUrlCrossRefPubMed
    1. Lipina TV,
    2. Palomo V,
    3. Gil C, et al.
    Dual inhibitor of PDE7 and GSK-3-VP1.15 acts as antipsychotic and cognitive enhancer in C57BL/6J mice. Neuropharmacology 2013; 64: 205–214. doi:10.1016/j.neuropharm.2012.06.032
    OpenUrlCrossRefPubMed
    1. Li L,
    2. Yee C,
    3. Beavo JA
    . CD3- and CD28-dependent induction of PDE7 required for T cell activation. Science 1999; 283: 848–851. doi:10.1126/science.283.5403.848
    OpenUrlAbstract/FREE Full Text
    1. Dong H,
    2. Osmanova V,
    3. Epstein PM, et al.
    Phosphodiesterase 8 (PDE8) regulates chemotaxis of activated lymphocytes. Biochem Biophys Res Commun 2006; 345: 713–719. doi:10.1016/j.bbrc.2006.04.143
    OpenUrlCrossRefPubMed
    1. Vasta V,
    2. Shimizu-Albergine M,
    3. Beavo JA
    . Modulation of Leydig cell function by cyclic nucleotide phosphodiesterase 8A. Proc Natl Acad Sci USA 2006; 103: 19925–19930. doi:10.1073/pnas.0609483103
    OpenUrlAbstract/FREE Full Text
    1. Arnaud-Lopez L,
    2. Usala G,
    3. Ceresini G, et al.
    Phosphodiesterase 8B gene variants are associated with serum TSH levels and thyroid function. Am J Hum Genet 2008; 82: 1270–1280. doi:10.1016/j.ajhg.2008.04.019
    OpenUrlCrossRefPubMed
    1. de Vente J,
    2. Abildayeva K,
    3. van de Waarenburg M, et al.
    NO-mediated cGMP synthesis in cultured cholinergic neurons from the basal forebrain of the fetal rat. Brain Res 2008; 1217: 25–36. doi:10.1016/j.brainres.2008.03.089
    OpenUrlPubMed
    1. Andreeva SG,
    2. Dikkes P,
    3. Epstein PM, et al.
    Expression of cGMP-specific phosphodiesterase 9A mRNA in the rat brain. J Neurosci 2001; 21: 9068–9076. doi:10.1523/JNEUROSCI.21-22-09068.2001
    OpenUrlAbstract/FREE Full Text
    1. Mishra S,
    2. Sadagopan N,
    3. Dunkerly-Eyring B, et al.
    Inhibition of phosphodiesterase type 9 reduces obesity and cardiometabolic syndrome in mice. J Clin Invest 2021; 131: e148798. doi:10.1172/JCI148798
    OpenUrl
    1. Reneerkens OAH,
    2. Rutten K,
    3. Bollen E, et al.
    Inhibition of phoshodiesterase type 2 or type 10 reverses object memory deficits induced by scopolamine or MK-801. Behav Brain Res 2013; 236: 16–22. doi:10.1016/j.bbr.2012.08.019
    OpenUrlCrossRefPubMed
    1. Rodefer JS,
    2. Murphy ER,
    3. Baxter MG
    . PDE10A inhibition reverses subchronic PCP-induced deficits in attentional set-shifting in rats. Eur J Neurosci 2005; 21: 1070–1076. doi:10.1111/j.1460-9568.2005.03937.x
    OpenUrlCrossRefPubMed
    1. Jager R,
    2. Russwurm C,
    3. Schwede F, et al.
    Activation of PDE10 and PDE11 phosphodiesterases. J Biol Chem 2012; 287: 1210–1219. doi:10.1074/jbc.M111.263806
    OpenUrlAbstract/FREE Full Text
    1. Pomara G,
    2. Morelli G
    . Inhibition of phosphodiesterase 11 (PDE11) impacts on sperm quality. Int J Impot Res 2005; 17: 385–386. doi:10.1038/sj.ijir.3901304
    OpenUrlCrossRefPubMed
    1. Rena G,
    2. Begg F,
    3. Ross A, et al.
    Molecular cloning, genomic positioning, promoter identification, and characterization of the novel cyclic amp-specific phosphodiesterase PDE4A10. Mol Pharmacol 2001; 59: 996–1011. doi:10.1124/mol.59.5.996
    OpenUrlAbstract/FREE Full Text
    1. Wallace DA,
    2. Johnston LA,
    3. Huston E, et al.
    Identification and characterization of PDE4A11, a novel, widely expressed long isoform encoded by the human PDE4A cAMP phosphodiesterase gene. Mol Pharmacol 2005; 67: 1920–1934. doi:10.1124/mol.104.009423
    OpenUrlAbstract/FREE Full Text
    1. Hansen RT, III.,
    2. Conti M,
    3. Zhang HT
    . Mice deficient in phosphodiesterase-4A display anxiogenic-like behavior. Psychopharmacology (Berl) 2014; 231: 2941–2954. doi:10.1007/s00213-014-3480-y
    OpenUrlCrossRefPubMed
    1. McLaughlin MM,
    2. Cieslinski LB,
    3. Burman M, et al.
    A low-Km, rolipram-sensitive, cAMP-specific phosphodiesterase from human brain. Cloning and expression of cDNA, biochemical characterization of recombinant protein, and tissue distribution of mRNA. J Biol Chem 1993; 268: 6470–6476. doi:10.1016/S0021-9258(18)53275-0
    OpenUrlAbstract/FREE Full Text
    1. Ariga M,
    2. Neitzert B,
    3. Nakae S, et al.
    Nonredundant function of phosphodiesterases 4D and 4B in neutrophil recruitment to the site of inflammation. J Immunol 2004; 173: 7531–7538. doi:10.4049/jimmunol.173.12.7531
    OpenUrlAbstract/FREE Full Text
    1. Jin SL,
    2. Conti M
    . Induction of the cyclic nucleotide phosphodiesterase PDE4B is essential for LPS-activated TNF-α responses. Proc Natl Acad Sci USA 2002; 99: 7628–7633. doi:10.1073/pnas.122041599
    OpenUrlAbstract/FREE Full Text
    1. Jin SL,
    2. Lan L,
    3. Zoudilova M, et al.
    Specific role of phosphodiesterase 4B in lipopolysaccharide-induced signaling in mouse macrophages. J Immunol 2005; 175: 1523–1531. doi:10.4049/jimmunol.175.3.1523
    OpenUrlAbstract/FREE Full Text
    1. Zhang HT,
    2. Huang Y,
    3. Masood A, et al.
    Anxiogenic-like behavioral phenotype of mice deficient in phosphodiesterase 4B (PDE4B). Neuropsychopharmacology 2008; 33: 1611–1623. doi:10.1038/sj.npp.1301537
    OpenUrlCrossRefPubMed
    1. Siuciak JA,
    2. McCarthy SA,
    3. Chapin DS, et al.
    Behavioral and neurochemical characterization of mice deficient in the phosphodiesterase-4B (PDE4B) enzyme. Psychopharmacology 2008; 197: 115–126. doi:10.1007/s00213-007-1014-6
    OpenUrlCrossRefPubMed
    1. Smith PG,
    2. Wang F,
    3. Wilkinson KN, et al.
    The phosphodiesterase PDE4B limits cAMP-associated PI3K/AKT-dependent apoptosis in diffuse large B-cell lymphoma. Blood 2005; 105: 308–316. doi:10.1182/blood-2004-01-0240
    OpenUrlAbstract/FREE Full Text
    1. Lobban M,
    2. Shakur Y,
    3. Beattie J, et al.
    Identification of two splice variant forms of type-IVB cyclic AMP phosphodiesterase, DPD (rPDE-IVB1) and PDE-4 (rPDE-IVB2) in brain: selective localization in membrane and cytosolic compartments and differential expression in various brain regions. Biochem J 1994; 304: 399–406. doi:10.1042/bj3040399
    OpenUrlAbstract/FREE Full Text
    1. Omori K,
    2. Kotera J
    . Overview of PDEs and their regulation. Circ Res 2007; 100: 309–327. doi:10.1161/01.RES.0000256354.95791.f1
    OpenUrlAbstract/FREE Full Text
    1. Beard MB,
    2. O'Connell JC,
    3. Bolger GB, et al.
    The unique N-terminal domain of the cAMP phosphodiesterase PDE4D4 allows for interaction with specific SH3 domains. FEBS Lett 1999; 460: 173–177. doi:10.1016/S0014-5793(99)01335-6
    OpenUrlCrossRefPubMed
    1. Wang D,
    2. Deng C,
    3. Bugaj-Gaweda B, et al.
    Cloning and characterization of novel PDE4D isoforms PDE4D6 and PDE4D7. Cell Signal 2003; 15: 883–891. doi:10.1016/S0898-6568(03)00042-1
    OpenUrlCrossRefPubMed
    1. Li YF,
    2. Cheng YF,
    3. Huang Y, et al.
    Phosphodiesterase-4D knock-out and RNA interference-mediated knock-down enhance memory and increase hippocampal neurogenesis via increased cAMP signaling. J Neurosci 2011; 31: 172–183. doi:10.1523/JNEUROSCI.5236-10.2011
    OpenUrlAbstract/FREE Full Text
    1. Xiang Y,
    2. Naro F,
    3. Zoudilova M, et al.
    Phosphodiesterase 4D is required for β2 adrenoceptor subtype-specific signaling in cardiac myocytes. Proc Natl Acad Sci USA 2005; 102: 909–914. doi:10.1073/pnas.0405263102
    OpenUrlAbstract/FREE Full Text
    1. Mehats C,
    2. Jin SL,
    3. Wahlstrom J, et al.
    PDE4D plays a critical role in the control of airway smooth muscle contraction. FASEB J 2003; 17: 1831–1841. doi:10.1096/fj.03-0274com
    OpenUrlCrossRefPubMed
    1. Conti M
    . Phosphodiesterases and regulation of female reproductive function. Curr Opin Pharmacol 2011; 11: 665–669. doi:10.1016/j.coph.2011.10.004
    OpenUrlCrossRefPubMed
PreviousNext
Back to top
View this article with LENS
Vol 32 Issue 167 Table of Contents
European Respiratory Review: 32 (167)
  • Table of Contents
  • Index by author
Email

Thank you for your interest in spreading the word on European Respiratory Society .

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Phosphodiesterase 4B inhibition: a potential novel strategy for treating pulmonary fibrosis
(Your Name) has sent you a message from European Respiratory Society
(Your Name) thought you would like to see the European Respiratory Society web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Print
Citation Tools
Phosphodiesterase 4B inhibition: a potential novel strategy for treating pulmonary fibrosis
Martin Kolb, Bruno Crestani, Toby M. Maher
European Respiratory Review Mar 2023, 32 (167) 220206; DOI: 10.1183/16000617.0206-2022

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero

Share
Phosphodiesterase 4B inhibition: a potential novel strategy for treating pulmonary fibrosis
Martin Kolb, Bruno Crestani, Toby M. Maher
European Respiratory Review Mar 2023, 32 (167) 220206; DOI: 10.1183/16000617.0206-2022
Reddit logo Technorati logo Twitter logo Connotea logo Facebook logo Mendeley logo
Full Text (PDF)

Jump To

  • Article
    • Abstract
    • Abstract
    • Introduction
    • Overview and introduction to phosphodiesterase (PDE) 4
    • PDE4B as a pharmacological target
    • PDE4-selective inhibitors
    • Clinical evidence for targeting PDE4B
    • Conclusions
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Subjects

  • Interstitial and orphan lung disease
  • Pulmonary pharmacology and therapeutics
  • Tweet Widget
  • Facebook Like
  • Google Plus One

More in this TOC Section

  • Strength of association between comorbidities and asthma
  • The role of smoking on COVID-19 progression: a meta-analysis
  • PAP therapy for post-stroke sleep disordered breathing
Show more Reviews

Related Articles

Navigate

  • Home
  • Current issue
  • Archive

About the ERR

  • Journal information
  • Editorial board
  • Press
  • Permissions and reprints
  • Advertising
  • Sponsorship

The European Respiratory Society

  • Society home
  • myERS
  • Privacy policy
  • Accessibility

ERS publications

  • European Respiratory Journal
  • ERJ Open Research
  • European Respiratory Review
  • Breathe
  • ERS books online
  • ERS Bookshop

Help

  • Feedback

For authors

  • Instructions for authors
  • Publication ethics and malpractice
  • Submit a manuscript

For readers

  • Alerts
  • Subjects
  • RSS

Subscriptions

  • Accessing the ERS publications

Contact us

European Respiratory Society
442 Glossop Road
Sheffield S10 2PX
United Kingdom
Tel: +44 114 2672860
Email: journals@ersnet.org

ISSN

Print ISSN: 0905-9180
Online ISSN: 1600-0617

Copyright © 2023 by the European Respiratory Society