We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

PDE4 inhibitors for disease therapy: advances and future perspective

    Baochan Du‡

    Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Min Luo‡

    Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Changyu Ren

    *Author for correspondence:

    E-mail Address: 654076835@qq.com

    Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, 611130, China

    &
    Jifa Zhang

    **Author for correspondence:

    E-mail Address: zjf298257@163.com

    Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China

    Published Online:https://doi.org/10.4155/fmc-2023-0101

    The PDE4 enzyme family is specifically responsible for hydrolyzing cAMP and plays a vital role in regulating the balance of second messengers. As a crucial regulator in signal transduction, PDE4 has displayed promising pharmacological targets in a variety of diseases, for which its inhibitors have been used as a therapeutic strategy. This review provides a comprehensive summary of the development of PDE4 inhibitors in the past few years, along with the structure, clinical and research progress of multiple inhibitors of PDE4, focusing on the research and development strategies of PDE4 inhibitors. We hope our analysis will provide a significant reference for the future development of new PDE4 inhibitors.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Maurice DH, Ke H, Ahmad F, Wang Y, Chung J, Manganiello VC. Advances in targeting cyclic nucleotide phosphodiesterases. Nat. Rev. Drug Discov. 13(4), 290–314 (2014).
    • 2. Schick MA, SchlegelN. Clinical Implication of Phosphodiesterase-4-Inhibition. Int. J. Mol. Sci. 23(3), 1209 (2022).
    • 3. Tibbo AJ, Baillie GS. Phosphodiesterase 4B: master regulator of brain signaling. Cells 9(5), 1254 (2020).
    • 4. Conti M, Richter W, Mehats C, Livera G, Park JY, Jin C. Cyclic AMP-specific PDE4 phosphodiesterases as critical components of cyclic AMP signaling. J. Biol. Chem. 278(8), 5493–5496 (2003).
    • 5. Peng T, Qi B, He J, Ke H, Shi J. Advances in the development of phosphodiesterase-4 inhibitors. J. Med. Chem. 63(19), 10594–10617 (2020). •• PDE4 inhibitor development.
    • 6. Peng T, Gong J, Jin Y et al. Inhibitors of phosphodiesterase as cancer therapeutics. Eur. J. Med. Chem. 150, 742–756 (2018).
    • 7. Jansen C, Kooistra AJ, Kanev GK, Leurs R, de Esch IJ, de Graaf C. PDEStrIAn: a phosphodiesterase structure and ligand interaction annotated database as a tool for structure-based drug design. J. Med. Chem. 59(15), 7029–7065 (2016).
    • 8. Feng X, Wang H, Ye M et al. Identification of a PDE4-specific pocket for the design of selective inhibitors. Biochemistry 57(30), 4518–4525 (2018).
    • 9. Bjørgo E, Moltu K, Taskén K. Phosphodiesterases as targets for modulating T-cell responses. Handb. Exp. Pharmacol. 204, 345–363 (2011).
    • 10. Cedervall P, Aulabaugh A, Geoghegan KF, Mclellan TJ, Pandit J. Engineered stabilization and structural analysis of the autoinhibited conformation of PDE4. Proc. Natl Acad. Sci. USA 112(12), E1414–1422 (2015).
    • 11. Picchianti-Diamanti A, Spinelli FR, Rosado MM, Conti F, Laganà B. Inhibition of phosphodiesterase-4 in psoriatic arthritis and inflammatory bowel diseases. Int. J. Mol. Sci. 22(5), 2638 (2021).
    • 12. Jones NA, Boswell-Smith V, Lever R, Page CP. The effect of selective phosphodiesterase isoenzyme inhibition on neutrophil function in vitro. Pulm. Pharmacol. Ther. 18(2), 93–101 (2005). •• New studies on inhaled PDE4 inhibitors.
    • 13. Facchinetti F, Civelli M, Singh D, Papi A, Emirova A, Govoni M. Tanimilast, a novel inhaled PDE4 inhibitor for the treatment of asthma and chronic obstructive pulmonary disease. Front. Pharmacol. 12, 740803 (2021).
    • 14. Wedzicha JA. Oral phosphodiesterase-4 inhibitors for chronic obstructive pulmonary disease ‘super exacerbators’. Am. J. Respir. Crit. Care Med. 194(5), 527–528 (2016).
    • 15. Kroegel C, Foerster M. Phosphodiesterase-4 inhibitors as a novel approach for the treatment of respiratory disease: cilomilast. Expert Opin. Investig. Drugs 16(1), 109–124 (2007).
    • 16. Yang X, Kambe N, Takimoto-Ito R, Kabashima K. Advances in the pathophysiology of atopic dermatitis revealed by novel therapeutics and clinical trials. Pharmacol. Ther. 224, 107830 (2021).
    • 17. Bissonnette R, Pavel AB, Diaz A et al. Crisaborole and atopic dermatitis skin biomarkers: an intrapatient randomized trial. J. Allergy Clin. Immunol. 144(5), 1274–1289 (2019).
    • 18. Papp KA, Leonardi C, Menter A et al. Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis. N. Engl. J. Med. 366(13), 1181–1189 (2012).
    • 19. Schafer PH, Parton A, Gandhi AK et al. Apremilast, a cAMP phosphodiesterase-4 inhibitor, demonstrates anti-inflammatory activity in vitro and in a model of psoriasis. Br. J. Pharmacol. 159(4), 842–855 (2010).
    • 20. Strober B, Thaçi D, Sofen H et al. Deucravacitinib versus placebo and apremilast in moderate to severe plaque psoriasis: efficacy and safety results from the 52-week, randomized, double-blinded, phase 3 program for evaluation of TYK2 inhibitor psoriasis second trial. J. Am. Acad. Dermatol. 88(1), 40–51 (2023).
    • 21. Joshi TP, Tschen J. Apremilast in the management of disseminated granuloma annulare. Cureus 13(5), e14918 (2021).
    • 22. Gadgil A, Duncan SR. Role of T-lymphocytes and pro-inflammatory mediators in the pathogenesis of chronic obstructive pulmonary disease. Int. J. Chron. Obstruct. Pulmon. Dis. 3(4), 531–541 (2008).
    • 23. Zhang W, Nie Y, Chong L et al. PI3K and Notch signal pathways coordinately regulate the activation and proliferation of T lymphocytes in asthma. Life Sci. 92(17–19), 890–895 (2013).
    • 24. Crocetti L, Floresta G, Cilibrizzi A, Giovannoni MP. An Overview of PDE4 Inhibitors in Clinical Trials: 2010 to Early 2022. Molecules. 27(15), 4964 (2022).
    • 25. Spina D. PDE4 inhibitors: current status. Br. J. Pharmacol. 155(3), 308–315 (2008).
    • 26. Breijyeh Z, Karaman R. Comprehensive review on Alzheimer’s disease: causes and treatment. Molecules 25(24), 5789 (2020).
    • 27. Bhat A, Ray B, Mahalakshmi AM et al. Phosphodiesterase-4 enzyme as a therapeutic target in neurological disorders. Pharmacol. Res. 160, 105078 (2020).
    • 28. Duarte-Silva E, Araújo S, Oliveira WH et al. Sildenafil ameliorates EAE by decreasing apoptosis in the spinal cord of C57BL/6 mice. J. Neuroimmunol. 321, 125–137 (2018).
    • 29. Kumar N, Goldminz AM, Kim N, Gottlieb AB. Phosphodiesterase 4-targeted treatments for autoimmune diseases. BMC Med. 11, 96 (2013).
    • 30. Scheltens P, Blennow K, Breteler MM et al. Alzheimer’s disease. Lancet 388(10043), 505–517 (2016).
    • 31. Pérez-Torres S, Cortés R, Tolnay M, Probst A, Palacios JM, Mengod G. Alterations on phosphodiesterase type 7 and 8 isozyme mRNA expression in Alzheimer’s disease brains examined by in situ hybridization. Exp. Neurol. 182(2), 322–334 (2003).
    • 32. Vitali E, Cambiaghi V, Spada A et al. cAMP effects in neuroendocrine tumors: The role of Epac and PKA in cell proliferation and adhesion. Exp. Cell Res. 339(2), 241–251 (2015).
    • 33. Massimi M, Ragusa F, Cardarelli S, Giorgi M. Targeting cyclic AMP signalling in hepatocellular carcinoma. Cells 8(12), 1511 (2019).
    • 34. Hsien Lai S, Zervoudakis G, Chou J, Gurney ME, Quesnelle KM. PDE4 subtypes in cancer. Oncogene 39(19), 3791–3802 (2020).
    • 35. Goldhoff P, Warrington NM, Limbrick DD Jr et al. Targeted inhibition of cyclic AMP phosphodiesterase-4 promotes brain tumor regression. Clin. Cancer Res. 14(23), 7717–7725 (2008).
    • 36. Pullamsetti SS, Banat GA, Schmall A et al. Phosphodiesterase-4 promotes proliferation and angiogenesis of lung cancer by crosstalk with HIF. Oncogene 32(9), 1121–1134 (2013).
    • 37. Kim DU, Nam J, Cha MD, Kim SW. Inhibition of phosphodiesterase 4D decreases the malignant properties of DLD-1 colorectal cancer cells by repressing the AKT/mTOR/Myc signaling pathway. Oncol. Lett. 17(3), 3589–3598 (2019).
    • 38. Dwivedi Y, Conley RR, Roberts RC, Tamminga CA, Pandey GN. [(3)H]cAMP binding sites and protein kinase A activity in the prefrontal cortex of suicide victims. Am. J. Psychiatry 159(1), 66–73 (2002).
    • 39. Lonze BE, Ginty DD. Function and regulation of CREB family transcription factors in the nervous system. Neuron 35(4), 605–623 (2002).
    • 40. Zhang HT, Frith SA, Wilkins J, O'Donnell JM. Comparison of the effects of isoproterenol administered into the hippocampus, frontal cortex, or amygdala on behavior of rats maintained by differential reinforcement of low response rate. Psychopharmacology 159(1), 89–97 (2001).
    • 41. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ. COVID-19: consider cytokine storm syndromes and immunosuppression. Lancet 395(10229), 1033–1034 (2020).
    • 42. Bridgewood C, Damiani G, Sharif K et al. Rationale for evaluating PDE4 inhibition for mitigating against severe inflammation in COVID-19 pneumonia and beyond. Isr. Med. Assoc. J. 22(6), 335–339 (2020).
    • 43. Palacios JM, Beleta J, Segarra V. Second messenger systems as targets for new therapeutic agents: focus on selective phosphodiesterase inhibitors. Farmaco. 50(12), 819–827 (1995).
    • 44. Cazzola M, Ora J, Puxeddu E. Dual bronchodilation and exacerbations of COPD. J. Thorac. Dis. 8(9), 2383–2386 (2016).
    • 45. Riahi A, Lam JM. Crisaborole 2% ointment for mild-to-moderate atopic dermatitis. Skin Therapy Lett. 26(1), 1–4 (2021).
    • 46. Dozier L, Bartos G, Kerdel F. Apremilast and psoriasis in the real world: a retrospective case series. J. Am. Acad. Dermatol. 83(1), 221–222 (2020).
    • 47. Rolan P, Hutchinson M, Johnson K. Ibudilast: a review of its pharmacology, efficacy and safety in respiratory and neurological disease. Expert Opin. Pharmacother. 10(17), 2897–2904 (2009).
    • 48. García-Osta A, Cuadrado-Tejedor M, García-Barroso C, Oyarzábal J, Franco R. Phosphodiesterases as therapeutic targets for Alzheimer’s disease. ACS Chem. Neurosci. 3(11), 832–844 (2012).
    • 49. Wedzicha JA, Calverley PM, Rabe KF. Roflumilast: a review of its use in the treatment of COPD. Int. J. Chron. Obstruct. Pulmon. Dis. 11, 81–90 (2016).
    • 50. Strand V, Fiorentino D, Hu C, Day RM, Stevens RM, Papp KA. Improvements in patient-reported outcomes with apremilast, an oral phosphodiesterase 4 inhibitor, in the treatment of moderate to severe psoriasis: results from a phase IIb randomized, controlled study. Health Qual. Life Outcomes 11, 82 (2013). •• Introduction of PDE4 subtypes and drug application.
    • 51. Paes D, Schepers M, Rombaut B, van den Hove D, Vanmierlo T, Prickaerts J. The molecular biology of phosphodiesterase 4 enzymes as pharmacological targets: an interplay of isoforms, conformational states, and inhibitors. Pharmacol. Rev. 73(3), 1016–1049 (2021).
    • 52. Xia C, He JP, Feng KW et al. Discovery of Novel 3-Amino-4-alkoxyphenylketones as PDE4 Inhibitors with Improved Oral Bioavailability and Safety against Spatial Memory Impairments. ACS Chem. Neurosci. 13(3), 390–405 (2022).
    • 53. Zhou ZZ, Cheng YF, Zou ZQ et al. Discovery of N-alkyl catecholamides as selective phosphodiesterase-4 inhibitors with anti-neuroinflammation potential exhibiting antidepressant-like effects at non-emetic doses. ACS Chem. Neurosci. 8(1), 135–146 (2017).
    • 54. Thirupataiah B, Bhuktar H, Mounika G et al. Fe(III)-catalyzed regioselective and faster synthesis of isocoumarins with 3-oxoalkyl moiety at C-4: identification of new inhibitors of PDE4. Bioorg. Chem. 121, 105667 (2022).
    • 55. Zhong J, Xie J, Xiao J et al. Inhibition of PDE4 by FCPR16 induces AMPK-dependent autophagy and confers neuroprotection in SH-SY5Y cells and neurons exposed to MPP(+)-induced oxidative insult. Free Radic. Biol. Med. 135, 87–101 (2019).
    • 56. Tang L, Huang C, Zhong J et al. Discovery of arylbenzylamines as PDE4 inhibitors with potential neuroprotective effect. Eur. J. Med. Chem. 168, 221–231 (2019).
    • 57. Xu B, Wang T, Xiao J et al. FCPR03, a novel phosphodiesterase 4 inhibitor, alleviates cerebral ischemia/reperfusion injury through activation of the AKT/GSK3β/ β-catenin signaling pathway. Biochem. Pharmacol. 163 234–249 (2019).
    • 58. Zhou ZZ, Ge BC, Zhong QP et al. Development of highly potent phosphodiesterase 4 inhibitors with anti-neuroinflammation potential: Design, synthesis, and structure-activity relationship study of catecholamides bearing aromatic rings. Eur. J. Med. Chem. 124, 372–379 (2016).
    • 59. Gurney ME, Nugent RA, Mo X et al. Design and synthesis of selective phosphodiesterase 4D (PDE4D) allosteric inhibitors for the treatment of fragile X syndrome and other brain disorders. J. Med. Chem. 62(10), 4884–4901 (2019).
    • 60. Woo TE, Kuzel P. Crisaborole 2% ointment (eucrisa) for atopic dermatitis. Skin Therapy Lett. 24(2), 4–6 (2019).
    • 61. Chu Z, Xu Q, Zhu Q et al. Design, synthesis and biological evaluation of novel benzoxaborole derivatives as potent PDE4 inhibitors for topical treatment of atopic dermatitis. Eur. J. Med. Chem. 213, 113171 (2021).
    • 62. Barberot C, Moniot A, Allart-Simon I et al. Synthesis and biological evaluation of pyridazinone derivatives as potential anti-inflammatory agents. Eur. J. Med. Chem. 146, 139–146 (2018).
    • 63. Allart-Simon I, Moniot A, Bisi N et al. Pyridazinone derivatives as potential anti-inflammatory agents: synthesis and biological evaluation as PDE4 inhibitors. RSC Med. Chem. 12(4), 584–592 (2021).
    • 64. Moniot A, Braux J, Bour C et al. Pyridazinone derivatives limit osteosarcoma-cells growth in vitro and in vivo. Cancers (Basel) 13(23), 5992 (2021).
    • 65. Moniot A, Braux J, Siboni R et al. Inhibition of recruitment and activation of neutrophils by pyridazinone-scaffold-based compounds. Int. J. Mol. Sci. 23(13), 7226 (2022).
    • 66. Li YS, Hu DK, Zhao DS et al. Design, synthesis and biological evaluation of 2,4-disubstituted oxazole derivatives as potential PDE4 inhibitors. Bioorg. Med. Chem. 25(6), 1852–1859 (2017).
    • 67. Hu DK, Zhao DS, He M et al. Synthesis and bioactivity of 3,5-dimethylpyrazole derivatives as potential PDE4 inhibitors. Bioorg. Med. Chem. Lett. 28(19), 3276–3280 (2018).
    • 68. Lin Y, Ahmed W, He M, Xiang X, Tang R, Cui ZN. Synthesis and bioactivity of phenyl substituted furan and oxazole carboxylic acid derivatives as potential PDE4 inhibitors. Eur. J. Med. Chem. 207, 112795 (2020).
    • 69. Liao Y, Jia X, Tang Y et al. Discovery of novel inhibitors of phosphodiesterase 4 with 1-phenyl-3,4-dihydroisoquinoline scaffold: structure-based drug design and fragment identification. Bioorg. Med. Chem. Lett. 29(22), 126720 (2019).
    • 70. Zhang X, Dong G, Li H et al. Structure-aided identification and optimization of tetrahydro-isoquinolines as novel PDE4 inhibitors leading to discovery of an effective antipsoriasis agent. J. Med. Chem. 62(11), 5579–5593 (2019).
    • 71. Zhang R, Li H, Zhang X et al. Design, synthesis, and biological evaluation of tetrahydroisoquinolines derivatives as novel, selective PDE4 inhibitors for antipsoriasis treatment. Eur. J. Med. Chem. 211, 113004 (2021).
    • 72. Thirupataiah B, Mounika G, Sujeevan Reddy G et al. CuCl2-catalyzed inexpensive, faster and ligand/additive free synthesis of isoquinolin-1(2H)-one derivatives via the coupling-cyclization strategy: evaluation of a new class of compounds as potential PDE4 inhibitors. Bioorg. Chem. 115, 105265 (2021).
    • 73. Zagórska A, Partyka A, Bucki A et al. Characteristics of metabolic stability and the cell permeability of 2-pyrimidinyl-piperazinyl-alkyl derivatives of 1H-imidazo[2,1-f]purine-2,4(3H,8H)-dione with antidepressant- and anxiolytic-like activities. Chem. Biol. Drug Des. 93(4), 511–521 (2019).
    • 74. Purushothaman B, Arumugam P, Kulsi G, Song JM. Design, synthesis, and biological evaluation of novel catecholopyrimidine based PDE4 inhibitor for the treatment of atopic dermatitis. Eur. J. Med. Chem. 145, 673–690 (2018).
    • 75. Purushothaman B, Arumugam P, Song JM. A novel catecholopyrimidine based small molecule PDE4B inhibitor suppresses inflammatory cytokines in atopic mice. Front. Pharmacol. 9, 485 (2018).
    • 77. Yougbare I, Keravis T, Lugnier C. NCS 613, a PDE4 inhibitor, by increasing cAMP level suppresses systemic inflammation and immune complexes deposition in kidney of MRL/lpr lupus-prone mice. Biochim. Biophys. Acta Mol. Basis Dis. 1867(3), 166019 (2021).
    • 78. Sahin Z, Biltekin SN, Yurttas L et al. Novel cyanothiouracil and cyanothiocytosine derivatives as concentration-dependent selective inhibitors of U87MG glioblastomas: adenosine receptor binding and potent PDE4 inhibition. Eur. J. Med. Chem. 212, 113125 (2021).
    • 79. Gewald R, Grunwald C, Egerland U. Discovery of triazines as potent, selective and orally active PDE4 inhibitors. Bioorg. Med. Chem. Lett. 23(15), 4308–4314 (2013).
    • 80. Nunes I, de Souza ET, Martins IRR et al. Discovery of sulfonyl hydrazone derivative as a new selective PDE4A and PDE4D inhibitor by lead-optimization approach on the prototype LASSBio-448: in vitro and in vivo preclinical studies. Eur. J. Med. Chem. 204, 112492 (2020).
    • 81. Brullo C, Rapetti F, Abbate S et al. Design, synthesis, biological evaluation and structural characterization of novel GEBR library PDE4D inhibitors. Eur. J. Med. Chem. 223, 113638 (2021).
    • 82. Ookawara M, Nio Y, Yamasaki M et al. Protective effect of a novel phosphodiesterase 4 selective inhibitor, compound A, in diabetic nephropathy model mice. Eur. J. Pharmacol. 894, 173852 (2021).
    • 83. Bastos ITS, Pinheiro PSM, Costa FN et al. Design, synthesis, experimental and theoretical characterization of a new multitarget 2-thienyl-N-acylhydrazone derivative. Pharmaceuticals (Basel) 11(4), 119 (2018).
    • 84. Guo H, Cheng Y, Wang C et al. FFPM, a PDE4 inhibitor, reverses learning and memory deficits in APP/PS1 transgenic mice via cAMP/PKA/CREB signaling and anti-inflammatory effects. Neuropharmacology 116, 260–269 (2017).
    • 85. Liang L, Chen H, Mao P et al. ZL-n-91, a specific phosphodiesterase-4 inhibitor, suppresses the growth of triple-negative breast cancer. Invest. New Drugs 40(5), 875–883 (2022).
    • 86. Gràcia J, Buil MA, Castro J et al. Biphenyl pyridazinone derivatives as inhaled PDE4 inhibitors: structural biology and structure–activity relationships. J. Med. Chem. 59(23), 10479–10497 (2016).
    • 87. Moretto N, Caruso P, Bosco R et al. CHF6001 I: a novel highly potent and selective phosphodiesterase 4 inhibitor with robust anti-inflammatory activity and suitable for topical pulmonary administration. J. Pharmacol. Exp. Ther. 352(3), 559–567 (2015).
    • 88. Schioppa T, Nguyen HO, Salvi V et al. The PDE4 inhibitor tanimilast restrains the tissue-damaging properties of human neutrophils. Int. J. Mol. Sci. 23(9), 4982 (2022).
    • 89. Burkovetskaya ME, Liu Q, Vadukoot AK et al. KVA-D-88, a novel preferable phosphodiesterase 4B inhibitor, decreases cocaine-mediated reward properties in vivo. ACS Chem. Neurosci. 11(15), 2231–2242 (2020).
    • 90. Furue M, Kitahara Y, Akama H, Hojo S, Hayashi N, Nakagawa H. Safety and efficacy of topical E6005, a phosphodiesterase 4 inhibitor, in Japanese adult patients with atopic dermatitis: results of a randomized, vehicle-controlled, multicenter clinical trial. J. Dermatol. 41(7), 577–585 (2014).
    • 91. Tsai YF, Chu TC, Chang WY et al. 6-Hydroxy-5,7-dimethoxy-flavone suppresses the neutrophil respiratory burst via selective PDE4 inhibition to ameliorate acute lung injury. Free Radic. Biol. Med. 106, 379–392 (2017).
    • 92. Cazzola M, Calzetta L, Rogliani P, Matera MG. Ensifentrine (RPL554): an investigational PDE3/4 inhibitor for the treatment of COPD. Expert Opin. Investig. Drugs 28(10), 827–833 (2019).
    • 93. Matera MG, Ora J, Cavalli F, Rogliani P, Cazzola M. New avenues for phosphodiesterase inhibitors in asthma. J. Exp. Pharmacol. 13, 291–302 (2021).
    • 94. Bjermer L, Abbott-Banner K, Newman K. Efficacy and safety of a first-in-class inhaled PDE3/4 inhibitor (ensifentrine) vs salbutamol in asthma. Pulm. Pharmacol. Ther. 58, 101814 (2019).
    • 95. Świerczek A, Pociecha K, Plutecka H, Ślusarczyk M, Chłoń-Rzepa G, Wyska E. Pharmacokinetic/pharmacodynamic evaluation of a new purine-2,6-dione derivative in rodents with experimental autoimmune diseases. Pharmaceutics 14(5), 1090 (2022).
    • 96. Dąbrowska M, Starek M, Chłoń-Rzepa G et al. Estimation of the lipophilicity of purine-2,6-dione-based TRPA1 antagonists and PDE4/7 inhibitors with analgesic activity. Bioorg. Med. Chem. Lett. 49, 128318 (2021). • Latest breakthrough of dual PDE4/7 inhibitors.
    • 97. Zygmunt M, Ślusarczyk M, Jankowska A et al. Evaluation of analgesic and anti-inflammatory activity of purine-2,6-dione-based TRPA1 antagonists with PDE4/7 inhibitory activity. Pharmacol. Rep. 74(5), 982–997 (2022).
    • 98. Ručilová V, Świerczek A, Vanda D et al. New imidazopyridines with phosphodiesterase 4 and 7 inhibitory activity and their efficacy in animal models of inflammatory and autoimmune diseases. Eur. J. Med. Chem. 209, 112854 ((.2021).
    • 99. Martinez A, Gil C. cAMP-specific phosphodiesterase inhibitors: promising drugs for inflammatory and neurological diseases. Expert Opin. Ther. Pat. 24(12), 1311–1321 (2014).
    • 100. Kita T, Fujimura M, Myou S, Watanabe K, Waseda Y, Nakao S. Effects of KF19514, a phosphodiesterase 4 and 1 inhibitor, on bronchial inflammation and remodeling in a murine model of chronic asthma. Allergol. Int. 58(2), 267–275 (2009).
    • 101. Blöcher R, Wagner KM, Gopireddy RR et al. Orally available soluble epoxide hydrolase/phosphodiesterase 4 dual inhibitor treats inflammatory pain. J. Med. Chem. 61(8), 3541–3550 (2018).
    • 102. Armani E, Rizzi A, Capaldi C et al. Discovery of M(3) antagonist-PDE4 inhibitor dual pharmacology molecules for the treatment of chronic obstructive pulmonary disease. J. Med. Chem. 64(13), 9100–9119 (2021).
    • 103. Liu J, Ye W, Xu JP et al. Discovery of novel trimethoxyphenylbenzo[d]oxazoles as dual tubulin/PDE4 inhibitors capable of inducing apoptosis at G2/M phase arrest in glioma and lung cancer cells. Eur. J. Med. Chem. 224, 113700 (2021).
    • 104. Guo YQ, Tang GH, Lou LL et al. Prenylated flavonoids as potent phosphodiesterase-4 inhibitors from Morus alba: Isolation, modification, and structure-activity relationship study. Eur. J. Med. Chem. 144, 758–766 (2018).
    • 105. Reinboth M, Wolffram S, Abraham G, Ungemach FR, Cermak R. Oral bioavailability of quercetin from different quercetin glycosides in dogs. Br. J. Nutr. 104(2), 198–203 (2010).
    • 106. Nishibe S, Mitsui-Saitoh K, Sakai J, Fujikawa T. The biological effects of Forsythia leaves containing the cyclic AMP phosphodiesterase 4 inhibitor phillyrin. Molecules 26(8), 2362 (2021).
    • 107. Guo Z, Abulaizi A, Huang L et al. Discovery of p-terphenyl metabolites as potential phosphodiesterase PDE4D inhibitors from the coral-associated fungus Aspergillus sp. ITBBc1. Mar. Drugs 20(11), 679 (2022).
    • 108. Yu Y, Wang J, Huang X. The anti-depressant effects of a novel PDE4 inhibitor derived from resveratrol. Pharm. Biol. 59(1), 418–423 (2021). • Discovery and structural optimization of a new PDE4 natural product.
    • 109. Song Z, Huang YY, Hou KQ et al. Discovery and structural optimization of toddacoumalone derivatives as novel PDE4 inhibitors for the topical treatment of psoriasis. J. Med. Chem. 65(5), 4238–4254 (2022).
    • 110. Zhou F, Huang Y, Liu L et al. Structure-based optimization of toddacoumalone as highly potent and selective PDE4 inhibitors with anti-inflammatory effects. Biochem. Pharmacol. 202, 115123 (2022).
    • 111. Liang J, Huang YY, Zhou Q et al. Discovery and Optimization of α-Mangostin Derivatives as Novel PDE4 Inhibitors for the Treatment of Vascular Dementia. J. Med. Chem. 63(6), 3370–3380 (2020).
    • 112. Liu H, Wang Q, Huang Y et al. Discovery of novel PDE4 inhibitors targeting the M-pocket from natural mangostanin with improved safety for the treatment of Inflammatory Bowel Diseases. Eur. J. Med. Chem. 242, 114631 (2022).
    • 113. Thirupataiah B, Mounika G, Reddy GS et al. PdCl2-catalyzed synthesis of a new class of isocoumarin derivatives containing aminosulfonyl / aminocarboxamide moiety: first identification of a isocoumarin based PDE4 inhibitor. Eur. J. Med. Chem. 221, 113514 (2021).