TiPs Tocris2 Asthma cs3 RESIZED · targets for asthma therapy. The table summarizes those that are...

1
Mast Cell-Directed Therapies Inhibition of mast cells is another potential asthma treatment. Mast cell activation may be inhibited by blocking IgE binding to the high-affinity IgE receptor (FcεRI), by inhibiting c-Kit, which is activated by stem cell factor (SCF), or by inhibiting Syk kinase. Mast cell stabilizers such as cromones and furosemide may work through specific ion channels for which novel modulators may be developed. Some specific targets and their inhibitors are IgE targeted by IgE antibody (Omalizumab), c-Kit targeted by Masitinib and Imatinib (approved for hematological malignancy and now explored for asthma), Syk kinase targeted by R-343 (in clinical development), and chloride ion channels targeted by cromones and furosemide. Mast cell stabilizers Lipid mediators Cyc-LTs, PGD 2 Degranulation Histamine Proteases Cytokines Cromones Furosemide Novel ion channel modulators? Anti-IgE FCεRI Sensitized mast cell Allergen IgE e.g., Omalizumab c-Kit antagonists e.g., Masitinib Blocking SCF antibody Syk kinase inhibitors e.g., R112 SCF SCF c-Kit c-Kit Syk kinase Syk kinase Lyn kinase Lyn kinase Anti-inflammatory Treatments Inhibition of signal transduction pathways that amplify inflamma- tory gene expression in asthmatic airways is another treatment option under investigation. Selective inhibitors have been developed for enzyme targets of phosphodiesterase-4 (PDE4), which degrades cAMP; inhibitor of inhibitor of NF-κB kinase (IKK2), which activates NF-κB; p38 mitogen-activated protein kinase (MAPK), which activates MAP kinase activated protein kinase 2 (MAPKAPK2); Jun kinase (JNK), which activates activator protein-1 (AP-1); and phosphoinositide-3 kinase (PI3K), which activates Akt. The table summarizes those that are currently in clinical development. INFLAMMATORY GENE EXPRESSION cAMP NF-κB MAPKAPK2 AP-1 AKT - - - - PDE4 IKK2 p38 MAPK JNK PI3Ky/δ Inflammatory stimuli - Selective inhibitors Cytokine Modulators Epithelial cells play an important role in orchestrating the inflammation of asthma through the release of multiple cytokines, including SCF (which maintains mast cells in the airways), TSLP (which acts on dendritic cells to release the Th2 chemoattractants CCL17 and CCL22, which act on CCR4), and several chemokines (particularly CCL11) that attract eosinophils by activating CCR3. Th2 cells orchestrate the inflammatory response in asthma through the release of IL-4 and IL-13 (stimulate B cells to synthesize IgE), IL-5 (necessary for eosinophilic inflammation), IL-9 (stimulates mast cell prolifera- tion), and IL-25, which further enhances cytokine release. Mast cells are thus orchestrated by several interacting cytokines and play an important role in asthma through the release of the bronchoconstrictor mediators histamine, cysteinyl-leukotrienes, and PGD 2 . Several of these cytokines (shown in purple) are now targets for asthma therapy. The table summarizes those that are currently in clinical development. Mast cell Histamine Cyc-LTs PGD 2 Broncho- constriction B-lymphocyte Target IL-4 IL-5 IL-13 CXCR2 Therapy Development status Eosinophils Inhaled allergens Dendritic cell Epithelial cells CCR3 IL-25 CCR4 TSLP Th2 cell CCL11, CCL24, CCL26, CCL5, CCL13 CCL11, CCL24, CCL26, CCL5, CCL13 CCL17, CCL22 CCL17, CCL22 SCF SCF IL-33 IL-33 IL-9 IL-9 IgE IgE IL-5 IL-5 IL-4, IL-13 IL-4, IL-13 Mutated IL-4 (pitrakinra); blocks IL-rRα and IL-13 Blocking antibody to IL-4Rα; also blocks IL-13 (AMG-317) Inhaled oligonucleotide against IL-4Rα; also blocks IL-13 (AIR645) Humanized blocking IL-5 antibody (mepolizumab) IL-5α receptor monoclonal antibody (MEDI-563) Blocking IL-13 monoclonal antibody (CAT-354 and others) Small-molecule antagonists (SCH527123) Reduced allergen responses in asthma; in clinical trials Clinical development Effective in allergen challenge; in clinical development Effective in severe asthma with sputum eosinophilia and exacerbations; in clinical trials Effective in phase II studies; in clinical trials Clinical studies in severe asthmas so far negative Phase II studies; for severe asthma Target Phosphodiesterase-4 p38 MAP kinase Inhibitor of NF-κ B kinase-2 Phosphoinositide-3 kinase Roflumilast SB236063 AS602868 CAL-101 Inhibitor (example) Development Advanced development for COPD; early clinical studies in asthma Several in early clinical development, including inhaled drugs Early clinical development δ-isoenzyme inhibitor in clincal trials for asthma Lipid Mediator Inhibitors (A) Several lipid mediators are involved in asthma. Arachidonic acid is generated from membrane lipids by cytoplasmic or secretory phospholipase A 2 (cPLA 2 , sPLA 2 ). 5 Lipoxygenase (5 LO) generates leukotrienes (LT), including LTB 4 , which acts on BLT 1 receptors, and cysteinyl leukotrienes (LTC 4 , LTD 4 , LTE 4 ), which act on cysLT 1 receptors and to a lesser extent on cysLT 1 receptors. Cyclooxygenases (COX1 and COX2) generate prostaglandins (PGs) and thromboxane (Tx). PGD 2 acts on DP 1 , DP 2 (also known as CRTH2), and TP receptors. Inhibitors and receptor antagonists that may be beneficial in asthma are shown in white boxes. (B) PGD 2 may play an important role in asthma. It is synthe- sized mainly in mast cells by PDGD synthase (PGDS) and acts on DP 1 receptors on vessels to mediate vasodilatation and on dendritic cells to enhance their activation. PGD 2 also acts via DP 2 receptors (CRTH2) to attract Th2 lymphocytes and eosinophils and on TP receptors to cause bronchoconstriction. Antagonists of DP 1 , DP 2 receptors, and inhibitors of PGDS are now in clinical development for asthma therapy. DP 1 antagonist CyaLT 1 antagonist CyaLT 2 antagonist BLT 1 antagonist DP 2 antagonist (CRTH2 antagonist) TP antagonist EP 1-4 CysLT 1 CysLT 2 BLT 1 TP DP 2 DP 1 Membrane phospholipids PLA 2 PGDS 5 LO COX 2 Arachidonic acid PGD 2 TxA 2 PGH 2 LTA 2 PGE 2 CysLT LTB 4 B A Mast cell Th2 cell Eosinophil Airway smooth muscle cell Dendritic cell Bronchial vessel PGDS inhibitors CRTH2 antagonist TP antagonist DP 1 antagonist DP 1 DP 1 DP 2 DP 2 TP cPLA 2 /sPLA 2 inhibitors 5 LO inhibitors FLAP inhibitors New “Dissociated” Corticosteroids Development of “dissociated” steroids is based on the dissociation of the side-effect mechanisms from the anti-inflammatory mechanisms of corticosteroids. This is theoretically possible because side effects are largely mediated via transactivation and binding of glucocorticoid receptor (GR) dimers to glucocorticoid response elements (GREs) in the promoter region of target genes, resulting in gene activation. However, anti-inflammatory effects are largely mediated via transrepression of transcription factors through a nongenomic effect whereby GR monomers interact with coactivator molecules, such as NF-κB and CBP, with recruitment of histone deacetylase 2 (HDAC2). Dissociated steroids or selective glucocorticoid receptor activators (SEGRAs) have a greater effect on the transrepression than transactivation and may therefore have a better therapeutic ratio. However, some of the anti-inflammatory effects of corticosteroids may be due to transactivation of anti-inflammatory genes, such as MAP kinase phosphatase-1 (MKP-1), so SEGRAs may not be as efficacious as existing drugs. New corticosteroid options include flutica- sone furoate, characterized by longer duration of action due to a higher affinity for GR and longer receptor retention; cicleso- nide, a prodrug that is converted to the active form by esterases in the lower airways, giving reduced systemic absorption from the lungs; and AL-438 and ZK-216438, nonsteroidal SEGRAs with reduced gene activation effects in vitro, now in clinical development. Anti-inflammatory proteins (e.g., MKP-1) DNA binding trans-activation histone acetylation Side effects (metabolic, endocrine) Inflammatory proteins (cytokines, enzymes, adhesion molecules, etc.) Corticosteroids Dissociated steroid GLUCOCORTICOID RECEPTORS GRα Inhibition of transcription factors NF-κB, AP-1, etc. trans-repression histone deacetylation NF-κB GR dimer GRE GR monomer HDAC2 Bronchodilators Activation of β 2 adrenoceptors (β 2 AR), vasoactive intestinal peptide (VIP), and prostaglandin E2 (PGE 2 ) receptors results in activation of adenylyl cyclase (AC) via a stimulatory G protein (G s ) and an increase in 3 -5 cyclic adenosine monophosphate (cAMP). This activates protein kinase A (PKA), which then phosphorylates several target proteins, resulting in the opening of calcium-activated potassium channels (KCa ) or Maxi-K channels, decrease in phosphoinositide (PI) hydrolysis, increase in sodium/calcium ion (Na + /Ca 2+ ) exchange, increase in Na + /K + ATPase activity, and decrease in myosin light chain kinase (MLCK) activity, leading to relaxation of airway smooth muscle. β 2 AR may also be coupled directly to K Ca via G s . cAMP is degraded by phosphodiesterases (PDEs) that are inhibited by theophylline and selective PDE3 inhibitors. Current bronchodilators are inhaled β 2 AR agonists, both short acting (salbutamol, terbutaline) and long acting (formoterol, salmeterol), and theophylline. New bronchodilators are once-inhaled daily β 2 AR agonists (indacaterol, carmoterol, GW-642444, and BI-1744), once-daily anticholinergics (tiotropium, glycopyrrolate, daratropium, and aclidinium), PGE 2 , potassium channel openers, vasoactive intestinal peptide analogues (Ro 25-1553), and PDE3 inhibitors. Smooth muscle myosin inhibitors are currently in development. K + Ca 2+ channel K + channel PI/Ca 2+ ATP cAMP AMP PDE MLCK G S PKA PGD 2 AC K + channel openers β 2 Agonist VIP PGE 2 β 2 AR Theophylline PDE3 inhibitors Bronchodilatation Smooth muscle myosin inhibitors Adenosine receptors Fexofenadine Amthamine, Tiotidine Imetit, Thioperamide Histamine receptors H 1 receptor H 2 receptor H 3 receptor 4-Methylhistamine, JNJ 10191584 H 4 receptor β 2 Adrenoceptors Glucocorticoid receptors Chemokine receptors Prostanoid receptors VIP receptors Signal Transduction Signal Transduction API-2, 10-DEBC Celecoxib, SC 560 AKT COX MG 132, Celastrol, IKK 16 NF-κ B MAPK SB 239063, Anisomycin, SP 600125 740 Y-P, LY 294002 PI3K AACOCF3, YM 26734 PLA 2 PDE Cilostamide, Zardaverine, Theophylline, Rolipram Nitric Oxide N ω -Propyl-L-arginine, 1400W, L-NAME, BYK 191023 Leukotriene receptors K + channels Cytokines and cytokine receptors Compounds Acting on Asthma Pathways Available from CGS 21680, 2-CI-IB-MECA, SCH 58261, ZM 241385, MRS 1754 ICI 118,551, Salmeterol, Formoterol Corticosterone, Fluticasone RS 504393, C 021, SB 328437, UCB 35625, SB 265610 Gabexate, Thalidomide, Pirfenidone, GIT 27, YM 90709 MK 886, BAY-u 9773, MK 571, Leukotriene B 4 SC 51089, L-161,982, Prostaglandin E 2 , U 46619, MK 571 VIP (6-28), VIP Iberiotoxin, 1-EBIO Asthma is now one of the most common chronic diseases in the world, affecting over 300 million people, and its prevalence is rising, particularly in develop- ing countries. Chronic inflammation of the airways results in airway hyperresponsiveness (AHR), which makes the airways “twitchy” and causes them to constrict more easily than normal in response to different triggers, such as exercise, cold air, and exposure to allergens and irritants. The inflammation may be worsened by exposure to allergens and by upper respiratory tract virus infections, and it may activate sensory nerves of the airways, resulting in coughing and chest tightness. Asthma symptoms are intermittent and rarely progress. Patients may have differing degrees of severity, ranging from mild intermittent symptoms to severe symptoms and disability. In the past, asthma was viewed as a disease of bronchoconstriction and treated predominantly with bronchodilators. Current therapy for asthma depends on the severity of the condition and is increased in a stepwise manner until asthma control is achieved. Inhaled corticosteroids (ICS) are the mainstay of treat- ment in all patients with persistent asthma, and long-acting β 2 agonists (LABAs) are added in moderate cases. In severe asthma, other add-on therapies may be necessary, and in very severe asthma, anti-IgE and immosuppressants may be used. Additionally, combination inhalers (corticosteroid + LABA) are now commonly prescribed to control asthma since they are more convenient for patients, simplify treatments, improve compliance, and, very importantly, prevent the use of LABAs alone, which is potentially dangerous. Nonetheless, it is now clear that asthma is a complex chronic inflammatory disease of the airways with many potential therapeutic targets, and treatment of the inflammation is now considered to be the key to successful control of symptoms. There is also a real need for new asthma treatments due to poor compliance with current treatments, side effects outside the lungs for the current inhaled treatments, patients’ preference for oral therapy, difficulty in treating severe asthma, and current unavailability of disease-modifying and curative treatments. Here, the focus is placed on new asthma treatment options that are being developed as the understanding of cellular pathways involved in the disease increases. INTRODUCTION Current therapy for asthma with ICS and inhaled LABA is highly effective, safe, and relatively inexpensive, but many patients’ symptoms remain poorly controlled, and severe asthma remains difficult to control with current treatments. Some innovative strategies and cutting-edge issues in asthma research and therapy include the following: developing selective therapies for subphenotypes via yet-to-be-identified discriminatory biomarkers and genetic profiling; treating patients with severe asthma who are relatively corticosteroid resistant by restoring HDAC2 activity via low doses of theophylline; developing an effective oral therapy for patients with mild and moderate disease; and using genetic and pharmacogenomic information to improve understanding of the biology of asthma to develop treatments. None of the currently available treatments for asthma have long-term effects on airway inflammation or remodeling, and therefore they are not disease modifying or curative. The prospects for a cure seem remote until the molecular and genetic causes of asthma are better understood, but there is a possibility that vaccination approaches have the potential to reverse the abnormal inflammation found in asthma. However, the long-term consequences of these approaches need to be carefully evaluated, particularly as they would probably need to be applied in children at the onset of disease. CONCLUSION AND FUTURE DIRECTIONS Further Reading Adcock, I.M., et al. (2006). Kinase inhibitors and airway inflammation. Eur. J. Pharmacol. 533, 118–132. Adcock, I.M., et al. (2008). New targets for drug development in asthma. Lancet 372, 1073–1087. Barnes, P.J. (2006). How corticosteroids control inflammation. Br. J. Pharmacol. 148, 245–254. Barnes, P.J. (2010). New therapies for asthma: Is there any progress? Trends Pharmacol. Sci. doi:10.1016/j.tips.2010.04.009. Barnes, P.J. (2008). Cytokine networks in asthma and chronic obstructive pulmonary disease. J. Clin. Invest. 118, 3546–3556. Barnes, P.J., et al. (2009). Glucocorticoid resistance in inflamma- tory diseases. Lancet 342, 1905–1917. Broide, D.H. (2009). Immunomodulation of allergic disease. Annu. Rev. Med. 60, 279–291. Cazzola, M., et al. (2009). Emerging inhaled bronchodilators: An update. Eur. Respir. J. 34, 757–769. Chung, K.F. (2006). Phosphodiesterase inhibitors in airways disease. Eur. J. Pharmacol. 533, 110–117. Holgate, S.T., et al. (2008). Treatment strategies for allergy and asthma. Nat. Rev. Immunol. 8, 218–230. New Therapies, Challenges, and Breakthroughs in Asthma Peter J. Barnes National Heart and Lung Institute, Imperial College London School of Medicine, Dovehouse Street, London SW3 6LY, UK [email protected] To obtain copies of this poster, please visit TOCRIS at www.tocris.com

Transcript of TiPs Tocris2 Asthma cs3 RESIZED · targets for asthma therapy. The table summarizes those that are...

Page 1: TiPs Tocris2 Asthma cs3 RESIZED · targets for asthma therapy. The table summarizes those that are ... COPD; early clinical studies in asthma Several in early clinical development,

Mast Cell-Directed TherapiesInhibition of mast cells is another potential asthma treatment. Mast cell activation may be inhibited by blocking IgE binding to the high-affinity IgE receptor (FcεRI), by inhibiting c-Kit, which is activated by stem cell factor (SCF), or by inhibiting Syk kinase. Mast cell stabilizers such as cromones and furosemide may work through specific ion channels for which novel modulators may be developed. Some specific targets and their inhibitors are IgE targeted by IgE antibody (Omalizumab), c-Kit targeted by Masitinib and Imatinib (approved for hematological malignancy and now explored for asthma), Syk kinase targeted by R-343 (in clinical development), and chloride ion channels targeted by cromones and furosemide.

Mast cell stabilizers

Lipid mediatorsCyc-LTs, PGD2

DegranulationHistamineProteases

Cytokines

CromonesFurosemideNovel ion channelmodulators?

Anti-IgE

FCεRI

Sensitizedmast cell

AllergenIgE

e.g., Omalizumab

c-Kit antagonistse.g., Masitinib

Blocking SCFantibody

Syk kinaseinhibitorse.g., R112

SCFSCF

c-Kitc-Kit

Syk kinaseSyk kinase

Lyn kinaseLyn kinase

Anti-inflammatory TreatmentsInhibition of signal transduction pathways that amplify inflamma-tory gene expression in asthmatic airways is another treatment option under investigation. Selective inhibitors have been developed for enzyme targets of phosphodiesterase-4 (PDE4), which degrades cAMP; inhibitor of inhibitor of NF-κB kinase (IKK2), which activates NF-κB; p38 mitogen-activated protein kinase (MAPK), which activates MAP kinase activated protein kinase 2 (MAPKAPK2); Jun kinase (JNK), which activates activator protein-1 (AP-1); and phosphoinositide-3 kinase (PI3K), which activates Akt. The table summarizes those that are currently in clinical development.

INFLAMMATORY GENE EXPRESSION

cAMP NF-κB MAPKAPK2 AP-1 AKT

-

-

-

-

PDE4 IKK2 p38 MAPK JNK PI3Ky/δ

Inflammatory stimuli

-

Selectiveinhibitors

Cytokine ModulatorsEpithelial cells play an important role in orchestrating the inflammation of asthma through the release of multiple cytokines, including SCF (which maintains mast cells in the airways), TSLP (which acts on dendritic cells to release the Th2 chemoattractants CCL17 and CCL22, which act on CCR4), and several chemokines (particularly CCL11) that attract eosinophils by activating CCR3. Th2 cells orchestrate the inflammatory response in asthma through the release of IL-4 and IL-13 (stimulate B cells to synthesize IgE), IL-5 (necessary for eosinophilic inflammation), IL-9 (stimulates mast cell prolifera-tion), and IL-25, which further enhances cytokine release. Mast cells are thus orchestrated by several interacting cytokines and play an important role in asthma through the release of the bronchoconstrictor mediators histamine, cysteinyl-leukotrienes, and PGD2. Several of these cytokines (shown in purple) are now targets for asthma therapy. The table summarizes those that are currently in clinical development.

Mast cell

HistamineCyc-LTsPGD2

Broncho-constriction

B-lymphocyte

Target

IL-4

IL-5

IL-13

CXCR2

Therapy Development status

Eosinophils

Inhaled allergens

Dendritic cell

Epithelialcells

CCR3IL-25

CCR4

TSLP

Th2 cell

CCL11, CCL24,CCL26, CCL5,

CCL13

CCL11, CCL24,CCL26, CCL5,

CCL13

CCL17, CCL22CCL17, CCL22

SCFSCFIL-33IL-33

IL-9IL-9

IgEIgE

IL-5IL-5IL-4,IL-13IL-4,IL-13

Mutated IL-4 (pitrakinra); blocks IL-rRα and IL-13

Blocking antibody to IL-4Rα; also blocks IL-13 (AMG-317)

Inhaled oligonucleotide against IL-4Rα; also blocks IL-13 (AIR645)

Humanized blocking IL-5 antibody (mepolizumab)

IL-5α receptor monoclonal antibody (MEDI-563)

Blocking IL-13 monoclonal antibody (CAT-354 and others)

Small-molecule antagonists (SCH527123)

Reduced allergen responses in asthma; in clinical trials

Clinical development

Effective in allergen challenge; in clinical development

Effective in severe asthma with sputum eosinophilia and exacerbations; in clinical trials

Effective in phase II studies; in clinical trials

Clinical studies in severe asthmas so far negative

Phase II studies; for severe asthma

Target

Phosphodiesterase-4

p38 MAP kinase

Inhibitor of NF-κBkinase-2

Phosphoinositide-3 kinase

Roflumilast

SB236063

AS602868

CAL-101

Inhibitor (example) Development

Advanced development for COPD; early clinical studies in asthma

Several in early clinical development, including inhaled drugs

Early clinical development

δ-isoenzyme inhibitor in clincal trials for asthma

Lipid Mediator Inhibitors(A) Several lipid mediators are involved in asthma. Arachidonic acid is generated from membrane lipids by cytoplasmic or secretory phospholipase A2 (cPLA2, sPLA2). 5′ Lipoxygenase (5′ LO) generates leukotrienes (LT), including LTB4, which acts on BLT1 receptors, and cysteinyl leukotrienes (LTC4, LTD4, LTE4), which act on cysLT1 receptors and to a lesser extent on cysLT1 receptors. Cyclooxygenases (COX1 and COX2) generate prostaglandins (PGs) and thromboxane (Tx). PGD2 acts on DP1, DP2 (also known as CRTH2), and TP receptors. Inhibitors and receptor antagonists that may be beneficial in asthma are shown in white boxes.(B) PGD2 may play an important role in asthma. It is synthe-sized mainly in mast cells by PDGD synthase (PGDS) and acts on DP1 receptors on vessels to mediate vasodilatation and on dendritic cells to enhance their activation. PGD2 also acts via DP2 receptors (CRTH2) to attract Th2 lymphocytes and eosinophils and on TP receptors to cause bronchoconstriction. Antagonists of DP1, DP2 receptors, and inhibitors of PGDS are now in clinical development for asthma therapy.

DP

1 antagonist

CyaLT

1 antagonistC

yaLT2 antagonist

BLT1 antagonist

DP

2 antagonist

(CRTH

2 antagonist)TP

antagonist

EP1-4 CysLT1 CysLT2 BLT1TPDP2DP1

Membrane phospholipids

PLA2

PGDS

5 LOCOX2

Arachidonic acid

PGD2 TxA2

PGH2 LTA2

PGE2 CysLT LTB4

B

A

Mast cell

Th2 cell Eosinophil Airway smoothmuscle cell

Dendriticcell

Bronchialvessel

PGDS inhibitors

CRTH2antagonist TP antagonistDP1 antagonist

DP1 DP1 DP2 DP2 TP

cPLA2/sPLA2 inhibitors

5′ LO inhibitorsFLAP inhibitors

New “Dissociated” CorticosteroidsDevelopment of “dissociated” steroids is based on the dissociation of the side-effect mechanisms from the anti-inflammatory mechanisms of corticosteroids. This is theoretically possible because side effects are largely mediated via transactivation and binding of glucocorticoid receptor (GR) dimers to glucocorticoid response elements (GREs) in the promoter region of target genes, resulting in gene activation. However, anti-inflammatory effects are largely mediated via transrepression of transcription factors through a nongenomic effect whereby GR monomers interact with coactivator molecules, such as NF-κB and CBP, with recruitment of histone deacetylase 2 (HDAC2). Dissociated steroids or selective glucocorticoid receptor activators (SEGRAs) have a greater effect on the transrepression than transactivation and may therefore have a better therapeutic ratio. However, some of the anti-inflammatory effects of corticosteroids may be due to transactivation of anti-inflammatory genes, such as MAP kinase phosphatase-1 (MKP-1), so SEGRAs may not be as efficacious as existing drugs. New corticosteroid options include flutica-sone furoate, characterized by longer duration of action due to a higher affinity for GR and longer receptor retention; cicleso-nide, a prodrug that is converted to the active form by esterases in the lower airways, giving reduced systemic absorption from the lungs; and AL-438 and ZK-216438, nonsteroidal SEGRAs with reduced gene activation effects in vitro, now in clinical development.

Anti-inflammatoryproteins

(e.g., MKP-1)

DNA bindingtrans-activation

histone acetylation

Side effects(metabolic,endocrine)

Inflammatory proteins(cytokines, enzymes,

adhesion molecules, etc.)

CorticosteroidsDissociated

steroidGLUCOCORTICOID RECEPTORS

GRα

Inhibition oftranscription factorsNF-κB, AP-1, etc.trans-repression

histone deacetylation

NF-κBGR dimer

GRE GR monomer

HDAC2

BronchodilatorsActivation of β2 adrenoceptors (β2AR), vasoactive intestinal peptide (VIP), and prostaglandin E2 (PGE2) receptors results in activation of adenylyl cyclase (AC) via a stimulatory G protein (Gs) and an increase in 3′-5′ cyclic adenosine monophosphate (cAMP). This activates protein kinase A (PKA), which then phosphorylates several target proteins, resulting in the opening of calcium-activated potassium channels (KCa) or Maxi-K channels, decrease in phosphoinositide (PI) hydrolysis, increase in sodium/calcium ion (Na+/Ca2+) exchange, increase in Na+/K+ ATPase activity, and decrease in myosin light chain kinase (MLCK) activity, leading to relaxation of airway smooth muscle. β2AR may also be coupled directly to KCa via Gs. cAMP is degraded by phosphodiesterases (PDEs) that are inhibited by theophylline and selective PDE3 inhibitors.

Current bronchodilators are inhaled β2AR agonists, both short acting (salbutamol, terbutaline) and long acting (formoterol, salmeterol), and theophylline. New bronchodilators are once-inhaled daily β2AR agonists (indacaterol, carmoterol, GW-642444, and BI-1744), once-daily anticholinergics (tiotropium, glycopyrrolate, daratropium, and aclidinium), PGE2, potassium channel openers, vasoactive intestinal peptide analogues (Ro 25-1553), and PDE3 inhibitors. Smooth muscle myosin inhibitors are currently in development.

K+

Ca2+ channelK+ channel

PI/Ca2+

ATPcAMPAMPPDE

MLCK

GS

PKA

PGD2

AC

K+ channelopeners

β2 AgonistVIP

PGE2

β2AR

TheophyllinePDE3 inhibitors

Bronchodilatation

Smooth musclemyosin inhibitors

Asthma Poster:Compounds Available

from Tocris

API-2, 1EBIO

Celecoxib, SC 560

Adenosinereceptors

Adrenergic b2 receptors

Fexofenadine

Amthamine, Tiotidine

Imetit, Thioperamide

Glucocorticoidreceptors

Chemokinereceptors

Histaminereceptors

H1 receptor

H2 receptor

H3 receptor

4-Methylhistamine,JNJ 10191584

H4 receptor

SignalTransduction

AKT

COX

740 Y-P, LY 294002PI3K

AACOCF3, YM 26734PLA2

MG 132, Celastrol, IKK 16NF-kB

MAPK

CGS 21680, 2-CI-IB-MECA,SCH 58261, ZM 241385, MRS 1754

ICI 118,551, Salmeterol,Formoterol

Corticosterone,Fluticasone

RS 504393, C 021 dihydro-chloride, SB 328437,UCB 35625, SB 265610

Prostanoidreceptors

SC 51089, L-161,982, Prostaglandin E2, U 46619, MK 571

VIP receptors VIP (6-28) (Human, rat, porcine, bovine)

VIP (Human, rat, mouse, rabbit, canine, porcine)

SB 239063, Anisomycin,SP 600125

PDE Cilostamide, Zardaverine,Theophpylline, Rolipram

Nitric Oxide Nw-Propyl-L-arginine,1400W, L-NAME,BYK 181023

Leukotriene receptors

MK 886, BAY-u 9773,MK 571, Leukotriene B4

K+ channels Iberiotoxin, 1-EBIO

Cytokines and Cytokinereceptors

Gabexate, Thalidomide, Pirfenidone, GIT 27,YM 90709

Adenosinereceptors Fexofenadine

Amthamine, TiotidineImetit, Thioperamide

Histamine receptorsH1 receptor

H2 receptor

H3 receptor

4-Methylhistamine,JNJ 10191584

H4 receptor β2 Adrenoceptors

Glucocorticoidreceptors

Chemokinereceptors

Prostanoidreceptors

VIP receptorsSignal Transduction

Signal Transduction

API-2, 10-DEBCCelecoxib, SC 560

AKT

COX

MG 132, Celastrol, IKK 16NF-κB

MAPK SB 239063, Anisomycin,SP 600125

740 Y-P, LY 294002PI3K

AACOCF3, YM 26734PLA2

PDE Cilostamide, Zardaverine,Theophylline, Rolipram

Nitric Oxide Nω-Propyl-L-arginine,1400W, L-NAME,BYK 191023

Leukotrienereceptors

K+ channels

Cytokines andcytokine

receptors

Compounds Acting on Asthma Pathways Available fromCGS 21680, 2-CI-IB-MECA,SCH 58261, ZM 241385,MRS 1754

ICI 118,551, Salmeterol,Formoterol

Corticosterone,Fluticasone

RS 504393, C 021,SB 328437, UCB 35625, SB 265610

Gabexate, Thalidomide, Pirfenidone, GIT 27,YM 90709

MK 886, BAY-u 9773,MK 571, Leukotriene B4

SC 51089, L-161,982,Prostaglandin E2, U 46619,MK 571

VIP (6-28), VIP

Iberiotoxin, 1-EBIO

Asthma is now one of the most common chronic diseases in the world, affecting over 300 million people, and its prevalence is rising, particularly in develop-ing countries. Chronic inflammation of the airways results in airway hyperresponsiveness (AHR), which makes the airways “twitchy” and causes them to constrict more easily than normal in response to different triggers, such as exercise, cold air, and exposure to allergens and irritants. The inflammation may be worsened by exposure to allergens and by upper respiratory tract virus infections, and it may activate sensory nerves of the airways, resulting in coughing and chest tightness. Asthma symptoms are intermittent and rarely progress. Patients may have differing degrees of severity, ranging from mild intermittent symptoms to severe symptoms and disability.

In the past, asthma was viewed as a disease of bronchoconstriction and treated predominantly with bronchodilators. Current therapy for asthma depends on the severity of the condition and is increased in a stepwise manner until asthma control is achieved. Inhaled corticosteroids (ICS) are the mainstay of treat-ment in all patients with persistent asthma, and long-acting β2 agonists (LABAs) are added in moderate cases. In severe asthma, other add-on therapies may be necessary, and in very severe asthma, anti-IgE and immosuppressants may be used. Additionally, combination inhalers (corticosteroid + LABA) are now commonly prescribed to control asthma since they are more convenient for patients, simplify treatments, improve compliance, and, very importantly, prevent the use of LABAs alone, which is potentially dangerous.

Nonetheless, it is now clear that asthma is a complex chronic inflammatory disease of the airways with many potential therapeutic targets, and treatment of the inflammation is now considered to be the key to successful control of symptoms. There is also a real need for new asthma treatments due to poor compliance with current treatments, side effects outside the lungs for the current inhaled treatments, patients’ preference for oral therapy, difficulty in treating severe asthma, and current unavailability of disease-modifying and curative treatments. Here, the focus is placed on new asthma treatment options that are being developed as the understanding of cellular pathways involved in the disease increases.

INTRODUCTION

Current therapy for asthma with ICS and inhaled LABA is highly effective, safe, and relatively inexpensive, but many patients’ symptoms remain poorly controlled, and severe asthma remains difficult to control with current treatments. Some innovative strategies and cutting-edge issues in asthma research and therapy include the following: developing selective therapies for subphenotypes via yet-to-be-identified discriminatory biomarkers and genetic profiling; treating patients with severe asthma who are relatively corticosteroid resistant by restoring HDAC2 activity via low doses of theophylline; developing an effective oral therapy for patients with mild and moderate disease; and using genetic and pharmacogenomic information to improve understanding of the biology of asthma to develop treatments.

None of the currently available treatments for asthma have long-term effects on airway inflammation or remodeling, and therefore they are not disease modifying or curative. The prospects for a cure seem remote until the molecular and genetic causes of asthma are better understood, but there is a possibility that vaccination approaches have the potential to reverse the abnormal inflammation found in asthma. However, the long-term consequences of these approaches need to be carefully evaluated, particularly as they would probably need to be applied in children at the onset of disease.

CONCLUSION AND FUTURE DIRECTIONS

Further ReadingAdcock, I.M., et al. (2006). Kinase inhibitors and airway inflammation. Eur. J. Pharmacol. 533, 118–132.

Adcock, I.M., et al. (2008). New targets for drug development in asthma. Lancet 372, 1073–1087.

Barnes, P.J. (2006). How corticosteroids control inflammation. Br. J. Pharmacol. 148, 245–254.

Barnes, P.J. (2010). New therapies for asthma: Is there any progress? Trends Pharmacol. Sci. doi:10.1016/j.tips.2010.04.009.

Barnes, P.J. (2008). Cytokine networks in asthma and chronic obstructive pulmonary disease. J. Clin. Invest. 118, 3546–3556.

Barnes, P.J., et al. (2009). Glucocorticoid resistance in inflamma-tory diseases. Lancet 342, 1905–1917.

Broide, D.H. (2009). Immunomodulation of allergic disease. Annu. Rev. Med. 60, 279–291.

Cazzola, M., et al. (2009). Emerging inhaled bronchodilators: An update. Eur. Respir. J. 34, 757–769.

Chung, K.F. (2006). Phosphodiesterase inhibitors in airways disease. Eur. J. Pharmacol. 533, 110–117.

Holgate, S.T., et al. (2008). Treatment strategies for allergy and asthma. Nat. Rev. Immunol. 8, 218–230.

New Therapies, Challenges,and Breakthroughs in Asthma

Peter J. BarnesNational Heart and Lung Institute, Imperial College London School of Medicine, Dovehouse Street, London SW3 6LY, UK

[email protected]

To obtain copies of this poster, please visit TOCRIS at www.tocris.com

C

M

Y

CM

MY

CY

CMY

K

TiPs_Tocris2_Asthma_cs3_RESIZED.pdf 6/2/2010 4:56:53 PM