[go: up one dir, main page]

Academia.eduAcademia.edu
+ MODEL ARTICLE IN PRESS JPT-05965; No of Pages 15 Pharmacology & Therapeutics xx (2007) xxx – xxx www.elsevier.com/locate/pharmthera Associate editor: Madhani Q11 Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease 3 James E. Clark, Negin Sarafraz, Michael S. Marber ⁎ 4 The Cardiovascular Division, Kings College London, The Rayne Institute, St Thomas' Hospital, London, SE1 7EH, United Kingdom OO F 2 Abstract 6 13 Chronic heart failure is debilitating, often fatal, expensive to treat and common. In most patients it is a late consequence of myocardial infarction (MI). The intracellular signals following infarction that lead to diminished contractility, apoptosis, fibrosis and ultimately heart failure are not fully understood but probably involve p38-mitogen activated protein kinases (p38), a family of serine/threonine kinases which, when activated, cause cardiomyocyte contractile dysfunction and death. Pharmacological inhibitors of p38 suppress inflammation and are undergoing clinical trials in rheumatoid arthritis, Chrohn's disease, psoriasis and surgery-induced tissue injury. In this review, we discuss the mechanisms, circumstances and consequences of p38 activation in the heart. The purpose is to evaluate p38 inhibition as a potential therapy for ischaemic heart disease. © 2007 Elsevier Inc. All rights reserved. 14 15 Keywords: p38; MAPK; Inhibitors; Heart failure; Ischaemia; Infarction 9 10 11 12 16 17 Contents 18 1. 2. 3. 4. 20 21 22 23 24 CO 25 26 27 31 32 33 34 35 UN 28 30 RR E Introduction . . . . . . . . . . . . . . . . . . . . . . . p38-Mitogen-activated protein kinase . . . . . . . . . . Structure and function of p38-MAPK . . . . . . . . . . Mechanisms of p38-MAPK activation. . . . . . . . . . 4.1. Mitogen-activated protein kinase kinases. . . . . 4.2. Autophosphorylation . . . . . . . . . . . . . . . 5. p38, myocardial ischaemia and ischaemic heart disease . 6. Pharmacological inhibitors of p38-MAPK. . . . . . . . 7. Clinical trials of p38-MAPK inhibition . . . . . . . . . 8. Summary and conclusions . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . 19 29 ED 8 CT 7 PR 5 1. Introduction Atherosclerotic vascular disease manifests predominantly as heart disease and stroke, which are the most frequent causes of death in the United Kingdom. Collectively atherosclerotic vascular disease was responsible for 40% of total mortality in the United Kingdom in 2004. Despite recent reductions in this high ⁎ Corresponding author. Tel.: +44 20 7188 1008. E-mail address: mike.marber@kcl.ac.uk (M.S. Marber). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 0 0 0 0 0 0 0 0 0 0 mortality, morbidity is increasing as more patients survive 36 myocardial infarction (MI) and stroke. For example, about 37 1.3 million people in the United Kingdom have survived acute 38 MI, 2 million have angina and 0.9 million have heart failure. The 39 extraordinarily high impact of atherosclerosis on the Nation's 40 health is reflected in its economic cost. Atherosclerotic disease 41 of just the coronary arteries costs the United Kingdom healthcare 42 system £3500 million with an estimated further £4400 million 43 lost to the economy through premature death, illness and infor- 44 mal care (The United Kingdom Heart Attack Study (UKHAS) 45 Collaborative Group, 1998; Office for National Statistics, 2000). 46 0163-7258/$ - see front matter © 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.pharmthera.2007.06.013 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 2 Study Model End point Inhibitor Outcome (Weinbrenner et al., 1997) p38 phosphorylation and cell viability (trypan blue) SB203580 5 μM during ischaemia Inhibition of p38 activation abolishes protection in preconditioned hearts and cardiomyocytes t1:4 (Wang et al., 1998a) Ex vivo: buffer perfused rabbit heart subjected to global ischaemia–reperfusion In vitro: isolated rabbit cardiomyocytes In vitro: neonatal rat cardiac myocytes cotransfected with p38α/β, active MKK3/MKK6 or dominant negative isoforms Cell survival, hypertrophic response, apoptosis The hypertrophic response in myocytes is mediated by p38β isoform, whereas over expression of p38α results in increase cell death t1:5 (Nagarkatti & Afi, 1998) In vitro: ischaemia in rat myoblast cell line H9C2 Cell viability (MTT) Modulation of p38α/β using overexpression of dominant negative isoforms or upstream activators SB203580 15 μM before/during ischaemia t1:6 (Meldrum et al., 1998) t1:7 LV function, coronary flow, CK release and tissue TNF LDH release t1:8 (Mackay & MochlyRosen, 1999) (Ma et al., 1999) t1:9 (Craig et al., 2000) t1:10 (Hoover et al., 2000) Ex vivo: buffer perfused rat heart exposed to H2O2 In vitro: simulated ischaemia in neonatal rat cardiomyocytes Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion In vitro: neonatal rat cardiac myocytes cotransfected with MKK6, TNF-α, TAK-1 and IL-6 In vitro: cardiac myocytes (transfected with MKK6) exposed to sorbitol t1:11 (Saurin et al., 2000) In vitro: simulated ischaemia in cultured rat neonatal cardiac myocytes t1:12 t1:13 t1:14 (Yue et al., 2000) In vitro: simulated ischaemia–reperfusion in rat neonatal cardiomyocytes In vitro: simulated ischaemia–reperfusion in rat neonatal cardiomyocytes In vivo: regional ischaemia–reperfusion in pig heart t1:15 (Mackay & MochlyRosen, 2000) (Barancik et al., 2000) t1:16 (Marais et al., 2001) t1:18 t1:19 (Gysembergh et al., 2001) (Schneider et al., 2001) t1:20 (Martin et al., 2001) t1:21 (Rakhit et al., 2001) t1:22 (Sanada et al., 2001) UN CO RR Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion In vitro: neonatal cardiac myocytes Ex vivo: buffer perfused rabbit heart subjected to focal ischaemia–reperfusion Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion In vitro: simulated ischaemia in neonatal/adult rat cardiocytes overexpressing wild-type p38α MAPK In vitro: simulated ichaemia–reperfusion injury in neonatal rat cardiomyocytes In vivo: regional ischaemic preconditioning in dog heart SB203580 1 mmol/min prior to insult SB203580 10 μM during ischaemia LV Function, apoptosis and CK release Apoptosis and IL-1 translation and transcription SB203580 10 μM before/during ischaemia SB203580 5 μM α-B crystallin expression and phosphorylation; MAPKAP-K2 and p38 activation and apoptosis p38 phosphorylation, CK and LDH release and cell viability (MTT) SB203580 5 μM during insult LV function and apoptosis (TUNEL) p38 phosphorylation, apoptosis and LDH release p38 phosphorylation and ATF-2 phosphorylation and Infarct size p38 phosphorylation, LV function and apoptosis SB242719 10 μM SB203580 10 μM SB203580 10 μM during ischaemia p38 activity and infarct size SB203580 1 μM during ischaemia LV function and necrosis SB202190 10 μM before ischaemia LDH release SB203580 10 μM during ischaemia p42/44 and p38 MAPK phosphorylation HSP27 phosphorylation, arterial blood pressure, infarct size and collateral flow SB203580 1 μM during ischaemia EC TE SB203580 10 μM during ischaemia DP Inhibition of p38 with SB203580 increased sorbitolmediated apoptosis Inhibition of p38 activation during prolonged ischaemia reduces injury and contributes to preconditioninginduced cardioprotection p38α is phosphorylated during ischaemia whereas p38β is deactivated Inhibition of p38/JNK leads to cardioprotection, whereas inhibition of ERK pathway exacerbates injury Incubation with SB203580 during ischaemia– reperfusion attenuates cell death Inhibition of p38 during ischaemia decrease infarct size and delays cell death RO SB203580 1 μM during ischaemia SB203580 1-10 μM during ischaemia/reperfusion SB203580 administered prior to ischaemia blocks preconditioning, but is protective during prolonged ischaemia p38 inhibition decreases myocardial TNF production, cardiomyocyte death and dysfunction p38 inhibition, by SB203580, during ischaemia protects against cardiomyocyte apoptosis Inhibition of p38 attenuates reperfusion injury by reducing apoptosis and improving cardiac function Activating both p38 and TNF-α augments myocardial survival during stress which is inhibited by SB203580 SB203580 1 μM during ischaemia/ reperfusion p38 inhibition by SB203580 during ischaemia and reperfusion is cardioprotective OF Inhibition of p38 activity during coronary artery occlusion is cardioprotective p38 inhibition reduces ischaemic injury and does not block protective effect of preconditioning SB203580 is cardioprotective through inhibition of p38 isoform and not due to inhibition or activation of other kinases SB203580 protected against injury, but p38β isoform does not contribute to survival SB203580 treatment during the preischemic and postischaemic periods had no significant effect on infarct size ARTICLE IN PRESS t1:1 t1:2 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 Table 1 Apoptosis and Fas-L cyclin D1 expression SB203580 10 μM during stress LV function SB202190 10 μM during rewarming/ reperfusion p38α activity, infarct size and LV function p38α+/− mice were used to knockdown p38 activity Inhibition of p38 activity attenuated stress-induced apoptosis and reversed changes in Fas-L and cyclin D1 expression SB202190, when present during reperfusion, improves recovery of LV function Inhibition of p38 did not protect against rewarming-induced injury Activation of p38α during ichaemia–reperfusion is detrimental; reduction in p38α expression results in protection The p38 activation that accompanies short-term hibernation does not appear to contribute to the contractile deficit Inhibition of p38 activation attenuates ischaemia– reperfusion injury in heart transplantation from nonheart-beating donors RWJ-67657 treatment post-MI had beneficial effects on LV remodelling and dysfunction FR167653-mediated inhibition of p38 activity during regional ichaemia–reperfusion injury reduces infarct size t1:26 (Sharov et al., 2003) t1:27 (Clanachan et al., 2003) t1:28 (Otsu et al., 2003) t1:29 (Gorog et al., 2004) Ex vivo: buffer perfused mouse heart subjected to low-flow global ischaemia–reperfusion LV function, Coronary flow and apoptosis SB203580 1 μM during ischaemia t1:30 (Koike et al., 2004) In vivo: canine heart transplantation from nonheart-beating donors Cardiac output (CO) and LV function FR167653 Dose not reported during cold storage t1:31 (See et al., 2004) In vivo: regional ischaemia in rat hearts t1:32 (Yada et al., 2004) In vivo: regional ichaemia–reperfusion in mouse hearts RWJ-67657 50 mg/day 7 days post MI for 21 days FR167653 2 mg/kg i.p. before ischaemia t1:33 (Kaiser et al., 2004) t1:35 (Aleshin et al., 2004) In vitro: simulated ischaemia–reperfusion in neonatal cardiomyocytes expressing dominantnegative p38 In vivo: regional ischaemia–reperfusion in dominant-negative MKK6 transgenic mice or dominant-negative p38α transgenic mice Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion LV function and postinfarction remodelling Protein kinase activation and kinase activity, Nuclear factor κB activity, inflammatory cytokines and infarct size Cell death, apoptosis, DNA fragmentation and Infarct size TNFα mRNA expression; LV function and CK release FR167653 1.0 mg/kg i.p. before I/R, and 1.0 mg/L during perfusion t1:36 (Martindale et al., 2005) (Kabir et al., 2005) LV dimensions (by echo); αB-crystallin expression; DNA fragmentation p38 and HSP27 phosphorylation and infarct size p38 activity was modified by cardiac overexpression of MKK6 t1:37 In vivo: regional ischaemia–reperfusion in hearts from mice overexpressing cardiacrestricted wild type MKK6 Ex vivo: buffer perfused mouse heart subjected to global ischaemia–reperfusion in presence/ absence of antimycin A t1:38 (Wang et al., 2005) Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion p38, activation and cytokine expression, activation of caspases and LV function SB203580 20 μM before ichaemia t1:39 (Okada et al., 2005) In vitro: hypoxia-reoxygenation in neonatal rat cardiomyocyte MAPKAP phosphorylation; Cyt. C release from mitochondria, caspase-3 activation and LDH release SB203580 10 μM throughout the experiment UN CO RR EC TE Reduction of endogenous p38 using overexpression of dominant negative p38 isoform or upstream activation DP p38α functions as a prodeath signalling effector in both cultured myocytes as well as in the intact heart RO SB203580 1 μM at the same time as antimycin A FR167653 inhibited ichaemia–reperfusion-mediated myocardial TNFα production and p38 activation and improved functional recovery Overexpression of MKK6 resulted in less myocardial damage following ischaemia–reperfusion and enhanced functional recovery Cardioprotection initiated by antimycin A is dependant upon p38 activation but independent of the upstream kinase MKK3; however, during lethal ischaemia, inhibition of p38 activity was protective Inhibition of p38 activation during ischaemia resulted in less MAPKAPK2, caspase-1, caspase-3 and caspase-11 activation, and TNF, IL-1beta, IL-6 production after myocardial ichaemia as well as increasing functional recovery of the heart SB203580 abrogated activation of p38 MAPK, translocation of HSP27, and F-actin reorganization, prevented cytochrome C release, caspase-3 activation, and DNA fragmentation OF 3 (continued on next page) ARTICLE IN PRESS SB203580 sensitive ischaemic activation of p38 by TAB1-associated autophosphorylation contributes to myocardial injury J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx SB203580 1 μM during ischaemia (Tanno et al., 2003) Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 Ex vivo: buffer perfused mouse heart subjected to global ischaemia–reperfusion (mkk3−/− and mkk3+/+) In vitro: global ischaemia in H9c2 myoblasts expressing wild-type and drug-resistant p38α In vitro: cardiomyocytes isolated from dogs with heart failure simulated by hypoxia, Angiotensin-II or nor-epinephrine Ex vivo: buffer perfused rat heart subjected to hypothermia-rewarming and global ischaemia– reperfusion In vivo: regional Ichaemia–reperfusion in p38α+/+ and p38α+/− mice Infarction/risk volume, p38, TAB1 and HSP27 phosphorylation t1:24 4 t1:41 Study Model t1:42 (Sumida et al., 2005) Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion t1:43 (Liu et al., 2005) In vivo: mouse hearts subjected to regional ischaemia–reperfusion t1:44 (House et al., 2005) t1:45 (Li et al., 2005) t1:47 (Gupta et al., 2005) Ex vivo: buffer perfused mouse heart subjected to global low-flow ischaemia–reperfusion In vitro: rat neonatal cardiomyocytes overexpressing MKK6 In vivo: hearts from MKK6bE transgenic mice subjected to ischaemia–reperfusion In vitro: adult rat ventricular myocytes (ARVMs) during sepsis t1:48 (Kim et al., 2006) In vitro:Hypoxia–re–oxygination in neonatal rat cardiomyocyte t1:49 (Khan et al., 2006) Ex vivo: buffer perfused rat heart subjected to global ischaemia–reperfusion t1:50 (Bellahcene et al., 2006) Ex vivo: buffer perfused mouse hearts from mkk3−/− mice subjected to TNF-α t1:51 (Engel et al., 2006) In vivo: regional ischaemia in rat hearts t1:52 (Li et al., 2006) In vivo: angiotensin and L-NAME-mediated cardiac hypertrophy in rats t1:53 (Vahebi et al., 2007) t1:54 t1:55 UN CO End point Inhibitor Outcome p38 and JNK activities LV contractility, CK release, mitochondrial ATP generation and infarct size LV function, cardiac remodelling SB203580 10 μM during reperfusion p38 inhibition exerts cardioprotection only when contractile force-induced necrosis is prevented SC-409 30 mg/kg/day given after MI for 12 weeks Infarct size and p38 and HSP27 phosphorylation LV remodelling, cytokine release and LV function SB203580 2 μM during ischaemia/ reperfusion SB239068 20 μM and 1200 ppm in drinking water or culture medium during exposure Inhibition of p38 MAPK attenuates cardiac remodelling and improves cardiac function in mice with heart failure after infarction Inhibition of p38 during ichaemia–reperfusion injury protects against myocardial cell death p38 inhibition prevents induction of inflammatory cytokines in cardiomyocytes and extracellular remodelling in heart RR EC Cell fractional shortening, cell viability (MTT), caspase-3 activity Cell viability (trypan blue), apoptosis (TUNEL), necrosis, ROS generation Coronary flow, LV function, LDH release, infarct size and apoptosis (TUNNEL) LV function, p38 and HSP27 phosphorylation SB203580 10 μM before insult SB203580 1 μM during ischaemia/ TE reperfusion SB203580 10 μM before TNF-α DP SB203580 1 μM before ischaemia Left ventricular remodelling, fractional shortening and neovascularisation LV function, arterial inflammatory cell infiltration, and cardiomyocyte apoptosis SB203580 2 mg/kg at time of surgery In vitro: isolated skinned cardiac muscle fibre bundles Cardiac myofilament function, Phosphorylation of α-Tm and TNI Reduction of endogenous p38 using overexpression of dominant negative p38α isoform or upstream activation (Riad et al., 2007) In vivo: diabetic mellitus induced by a single injection of streptozotocin SB239063 40 mg/kg/day for 43 days after induction of diabetes mellitus (Clark et al., 2007) In vivo: regional ischaemia in hearts from mkk3−/− and mkk3+/+ mice LV function, p38 phosphorylation and peripheral ICAM-1 and VCAM-1 LV function, LV remodelling, p38 and HSP27 phosphorylation SB203580 pretreatment followed by bigET-1 administration decreased p38 phosphorylation and downregulated ET(B) receptor expression in sepsis group Inhibition of p38α prevented hypoxia re–oxygenation induced apoptosis of cardiomyocytes Inhibition of p38 MAPK improved cardiac function after reperfusion and attenuated ischaemic reperfusioninduced myocardial apoptosis and necrosis Activation of p38 contributes to TNF-α induced contractile depression in intact heart and in isolated cardiac myocytes through MKK3; inhibition of p38 abolished contractile depression caused by TNFα SB203580 and FGF-1 induces cardiomyocyte mitosis, reduces scarring, and rescues function after MI RO SD-282 60 mg/kg administered for 4 days during treatment Knockout of MKK3 SD-282, reduces inflammatory response and apoptosis, resulting in a reduction of myocardial damage, which, in turn, improves cardiac function following angiotensin II and L-NAME treatment Activation of p38α directly depresses saromeric function by decreased phosphorylation of α-tropomyosin, which is reversed by inhibition of p38α activity or by over expressing dominant negative isoform Inhibition of p38MAPK reduced cardiac cell adhesion molecules expression indicating both antiinflammatory and vasculoprotective effects in the diabetic heart Postinfarction LV remodelling continues in the absence of MKK3 as does p38 activation OF ARTICLE IN PRESS Table 1 (continued ) J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 t1:40 ARTICLE IN PRESS J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 92 93 94 95 96 97 98 99 100 101 OF 53 EC 52 CO RR 51 UN 50 RO 2. p38-Mitogen-activated protein kinase 49 surprisingly these cellular effects have clear consequence(s), translating into involvement in complex pathophysiologies, such as wound healing (Lim et al., 1998), inflammatory arthritis (Badger et al., 1996), sepsis (Kotlyarov et al., 1999), acute respiratory distress syndrome (Carter et al., 1999), and malignant hypertension (Behr et al., 2001). Four p38 isoforms (α, β, δ and γ) exist, which have preserved structure but variable sensitivity to pharmacological inhibition. All 4 isoforms have a Thr180-Gly181 -Tyr182 (TGY) dual phosphorylation motif which is used by investigators to infer activation. p38α and β have high sequence homology and share sensitivity to pharmacological inhibition by prydinyl imidazole molecules (such as SB203580) but have only 60% homology with p38γ and δ, which are resistant to SB203580 (SB) inhibition (Eyers et al., 1999). Of the SB-sensitive isoforms, p38α is the predominant form in human and rodent myocardium (Lemke et al., 2001; Rakhit et al., 2001; Sanada et al., 2001; Braz et al., 2003). Studies with knockout mice and cells have shown that p38α is essential for embryonic development as knockout of the α isoform results in embryonal lethality, but mice lacking p38β, p38γ, and p38δ are viable (Allen et al., 2000; Adams et al., 2000; Tamura et al., 2000; Brancho et al., 2003). DP 91 48 3. Structure and function of p38-MAPK TE 90 Atherosclerotic plaque rupture/erosion results in MI, which is characterised by necrosis and apoptosis of cardiomyocytes. Although the acute condition alone may result in death due to ventricular arrhythmias or pump failure, in the patients with substantial infarction that survive, a chronic phase of ventricular remodelling occurs. Remodelling, is a maladaptive process characterised by cardiomyocyte apoptosis, fibrosis, thinning of the ventricular wall at the site of infarction, ventricular chamber enlargement and hypertrophy of surviving cardiomyocytes (Pfeffer & Braunwald, 1990; Swynghedauw, 1999; Udelson et al., 2003). These events may, eventually, lead to heart failure which is frequently lethal despite current best care. Therefore, intervention to minimise pathological cardiac remodelling is highly desirable to reduce the mortality and the incidence and severity of congestive heart failure after MI. Various intracellular signalling pathways are thought to play a critical role in the myocardial response to ischaemia and consequent pathological remodelling. Multiple mitogen-activated protein kinase (MAPK) are activated during ischaemia and may contribute to the structural and functional changes. MAPK are highly conserved serine/threonine kinases that are activated by a dual phosphorylation of a Thr-X-Tyr motif, in response to wide a variety of stimuli such as cytokines, osmotic and other environmental stresses and consequently play a role in numerous cell functions including growth and proliferation (English et al., 1999; Pearson et al., 2001). Three of the five major MAPK cascades have been extensively studied in the heart: extracellular signal-regulated kinase (ERK1 and ERK2), c-Jun N-terminal kinases (JNK1 and JNK2) and p38 kinases. It has been shown that JNK and p38 contribute to, whereas ERK/ ERK2 protect against, apoptotic cell death. Although the mechanisms by which p38 and JNK induce apoptosis may be cell and stimulus specific, there is overwhelming evidence that the activation of p38-MAPK (or p38) that occurs during prolonged ischaemia accelerates injury since its inhibition by pharmacological or genetic means slows the rate of infarction/death (Saurin et al., 2000; Martin et al., 2001; see Table 1). Although this evidence is based on animal data, it seems likely similar mechanisms operate in the human heart since p38 is identically activated by ischaemia (Han et al., 1995; Cain et al., 1999; Cook et al., 1999; Lee et al., 2000; Lemke et al., 2001) and early clinical trials indicate a potential benefit (de Winter et al., 2005). Thus, superficially at least, inhibitors of p38 have therapeutic potential in ischemic heart disease (Force et al., 2004). 47 p38-MAPK are activated by a wide range of extracellular influences, including radiation, ultraviolet light, heat shock, osmotic stress, proinflammatory cytokines such as interleukin (IL)-1 and tumour necrosis factor (TNF)-α, and certain mitogens (Sugden & Clerk, 1998) in addition to myocardial ischaemia (Bogoyevitch et al., 1996; Saurin et al., 2000; Luss et al., 2000; Ping & Murphy, 2000). Furthermore, the consequent activation of p38-MAPK is intimately involved in multiple cellular responses, including growth, proliferation, differentiation, and death (English et al., 1999; Ono & Han, 2000). Perhaps not 5 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 The catalytic site of p38 lies in a pocket between the N- and 125 C-terminal domains. These domains are connected by a single 126 hinge and the L16 loop of the C-terminal domain which wraps 127 back around the N-terminal domain and controls the relationship 128 between the relatively rigid domains (see Fig. 2). In addition, in 129 the inhibitor bound nonphosphorylated state, there is a mis- 130 alignment between the N- and C-lobes which prevents the 131 cooperation between a lysine residue (Lys53) in the N-terminal 132 lobe and aspartic acid residue (Asp168) in the C-terminal lobe, 133 imperative to binding and stabilization of the α phosphate group 134 and adjacent ribose of ATP, respectively (Wilson et al., 1996; 135 Gum et al., 1998). Therefore, it is widely thought that the 136 nondual phospho- form of p38 is inactive as a result of steric 137 obstruction of the peptide-binding channel and low ATP affinity. 138 Thr180 and Tyr182 are located on a flexible “activation loop” 139 that guards the active site. Dual phosphorylation of these 2 140 amino acids in response to exotoxin, cytokines, physical stress 141 (such as hyperosmolarity), and chemical oxidant stress, such as 142 hydrogen peroxide (Han et al., 1994; Freshney et al., 1994; 143 Rouse et al., 1994; Raingeaud et al., 1995) is thought to cause the 144 activation loop to refold and move out of the peptide-binding 145 channel. This movement is then thought to exert a “crank- 146 handle” effect on the overall tertiary structure of the kinase 147 reorienting the N- and C-, terminal lobes so that Lys53 and 148 Asp168 move towards one another by 2.5–5 Å. This alters the 149 conformation of the catalytic site enabling the cooperation 150 necessary for ATP binding and allowing substrate access 151 (Wilson et al., 1996; Diskin et al., 2007). The docking grooves 152 used by substrates and activators consist of 2 regions (see Fig. 2), 153 the CD region and the ED region (Tanoue et al., 2000). The CD 154 region is part of a shallow groove formed by the acidic residues 155 Asp313, Asp316 , Glu81, and the aromatic residues Phe129 and 156 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 ARTICLE IN PRESS 6 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx 4. Mechanisms of p38-MAPK activation 164 4.1. Mitogen-activated protein kinase kinases 167 168 169 170 171 DP 166 Although the intracellular activation cascade for p38 under most physiological conditions is still unclear, several upstream MAPK kinases (MKK) have been identified from in vitro analysis, including MKK3 and MKK6 (Derijard et al., 1995; Han et al., 1996). MKK4 is predominately involved in JNK activation but is able to activate p38-MAPK, at least, in vitro (Deacon & Blank, 1997). Using MKK-targeted mouse lines, it TE 165 EC 161 CO RR 160 UN 159 has been shown that, in response to most stress stimuli, MKK3 and MKK6 are the principal MKK activating p38α and β, respectively (see Fig. 1). MKK3 and MKK6 are in turn activated by phosphorylation by a MKK kinase (MKKK). The MKKK, being responsible for activation of the p38 cascade, appears to be cell type and stimulus specific, and several have been implicated (Yamaguchi et al., 1995; Moriguchi et al., 1996; Ichijo et al., 1997; Hutchison et al., 1998; Gallo & Johnson, 2002; Ge et al., 2002; Cheung et al., 2003). However, p38 activation is not limited to this traditional phospho-relay signalling cascade. Since SB203508 (the most widely used p38 kinase inhibitor) occupies the catalytic site, without inhibiting upstream MKK, it should only inhibit the phosphorylation events downstream of p38 without inhibiting the dual-phosphorylation of p38 itself (Young et al., 1997). However, certain conditions, such as myocardial ischaemia, cause a SB-sensitive form of p38 dual phosphorylation. Two mutually exclusive explanations for these observations are OF 163 158 RO 162 Tyr311. The ED region is part of a deeper groove formed by residues 159–163 at one side and residues Gln120 , His126 and Phe129 at the opposite side (Haar et al., 2007). It is believed that these 2 binding regions facilitate activator (MAPK kinase 3, MKK3) and substrate (MK2 and MEF2) binding (Chang et al., 2002; Haar et al., 2007). 157 Fig. 1. Mechanisms of p38-MAPK activation. Classical activation by MKK3/MKK6 is depicted as mechanism ①. TAB1-induced autoactivation is depicted as mechanism ②. TCR-mediated Tyr323 phorphorylation by ZAP70 is depicted as mechanism ③. TAB1 activates TAK1, which in turn activates MKK3/MKK6. In addition, TAB1 is a p38 substrate. PhosphoTAB1 is less able to activate TAK1 (★). Pharmacological inhibition of p38-MAPK diminishes p38 dual phosphorylation and phosphoTAB-1. Heavy lines represent an interaction; dotted lines represent a modification (phosphorylation); open arrows represent a multi-element pathway. Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 ARTICLE IN PRESS J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 TE 197 EC 196 CO RR 195 Ge and co-workers elegantly reported that auto-phosphorylation of p38 can occur, facilitated by an interaction with the nonenzymatic adaptor protein transforming growth factor-βactivated protein kinase-1 (TAK1) binding protein-1 (TAB1; Ge et al., 2002). TAB1 is known to perform a similar function by inducing the autophosphorylation of TAK1, which in turn activates MKK3/MKK6. In vitro co-expression experiments have shown that the interaction of TAB1 and p38α leads to phosphorylation of the TGY activation motif. TAB1-dependent p38α activation appears to play a role in the injury response during myocardial ischaemia (Tanno et al., 2003; Fiedler et al., 2006), myocyte-derived dendritic cell maturation (Matsuyama et al., 2003), and peripheral T-cell anergy maintenance (OhkusuTsukada et al., 2005). The interpretation of the TAB1-p38 interaction was, however, complicated by Cohen's group who demonstrated that the phosphorylation of TAB1 on Ser423 and Tyr431 was p38-MAPK-dependent and hence prevented by SB203580. The authors proposed a feedback control mechanism of TAK1 activity, whereby p38 activity inhibits TAK1, through the phosphorylation of TAB1. Inhibition of p38 activity (by SB203580) abolishes this feedback control of TAK1, causing unopposed activation of the parallel JNK pathway and consequently IKK (Cheung et al., 2003). This is depicted in Fig. 1. Although TAB1 and MKK3/MKK6 mechanisms interact through the potential modulation of TAK1, other possibilities also exist. For example there is some evidence to suggest that TAB1 causes p38 redistribution to the cytoplasm and may restrict access to downstream targets, such as MAPK-activated protein kinase 2 (MAPKAPK2). This is in direct contrast to the pattern seen with MKK3/MKK6 (Lu et al., 2005). Using MKK3/MKK6 double knockout and MKK4/MKK7 double knock out mouse embryonic fibroblasts (MEF), Kang et al. have shown that peroxynitrite-induced phosphorylation of p38α is associated with an ∼85 kDa disulfide complex in wild type MEF (Kang et al., 2006). This association was diminished in MKK3/MKK6 knockout MEF (Kang et al., 2006). The authors suggested that phosphorylation of p38 mediated by TAB-1 can be modulated by a yet unknown binding partner(s) in a manner dependent on a disulfide complex (Kang et al., 2006). In addition to TAB-1-mediated activation, p38 can also autophosphorylate through an alternative pathway in response to T-cell antigen receptor (TCR) activation (Dong et al., 2002; Rincon & Pedraza-Alva, 2003). In this pathway, activation of TCR leads to recruitment of a Syk family kinase, ZAP-70, which directly phosphorylates p38 on Tyr323 (Rouse et al., 1994). In a recent study it was shown that the phosphorylation of p38 on Try323 can be blocked in the presence of the DNA damage inducible gene Gadd45a, an autoimmune suppressor. Absence of Gadd45a has been shown to result in chronic phosphorylation of p38, T-cell hyperproliferation and autoimmunity (Salvador et al., 2005a, 2005b). Following ZAP-70 UN 194 OF 4.2. Autophosphorylation phosphorylation of p38, an autophosphorylation event similar to that induced by interaction with TAB-1 occurs resulting in dual phosphorylation, and activation of the kinase. However, regulation of p38 kinase activity in vitro, at least is not solely dependent on upstream kinases and binding partners. Diskin and co-workers, using a in vitro mutation approach, made intrinsically active p38 isoforms based on activating mutations previously found in the yeast MAPK kinase p38/Hog1 (Bell et al., 2001). Single and multiple point mutations of human p38α resulted in high intrinsic activity independent of activation by dual phosphorylation. Structural analysis of these p38 mutants has identified a hydrophobic core stabilised by 3 aromatic residues, Tyr69, Phe327 and Trp337, in the vicinity of the L16 Loop region. It is believed that the hydrophobic core is an inherent stabiliser that maintains the low basal activity level of unphosphorylated p38 (Diskin et al., 2004). Upon activation, however, a segment of the L16 Loop, including Phe327 becomes disordered allowing ATP and substrate binding. The mutation of these amino acids involved in the hydrophobic core results in the conformational changes imposed naturally by dual phosphorylation, namely destabilising the hydrophobic core and locking the kinase in a constitutively active state. In addition, in this active state, p38 is able to autophosphorylate in an invitro kinase assay (Diskin et al., 2004). More recently p38β, p38γ and p38δ mutants were similarly constructed (Askari et al., 2007). In these mutants, a highly conserved aspartic acid located in the activation loop (Asp170 in Hog-1; Asp176 in p38α, p38β and p38δ; and Asp179 in p38) was mutated. The spontaneous kinase activity of p38β, p38γ and p38δ appeared to be lower than the dual phosphorylated wild-type isoforms whereas the p38α isoform presented the highest spontaneous activity. Therefore, it is apparent that modifications of the amino acids in the hydrophobic core along with the mutations in Asp176 are capable of activating p38s (Askari et al., 2007), the former likely explaining the mechanism by which ZAP-70 induces autoactivation (Mittelstadt et al., 2005). In addition, these mutants provide a tool to dissect isoform-specific downstream signalling (Askari et al., 2007). RO 193 191 DP 192 (i) that p38 is able to autophosphorylate its activation loop or (ii) that SB203580 inhibits a kinase upstream of p38 involved in its activation by trans-phosphorylation during ischaemia. 190 7 5. p38, myocardial ischaemia and ischaemic heart disease 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 Ischaemic heart disease remains the leading cause of death, 284 accounting for approximately 1 quarter of all deaths in the 285 United Kingdom. Currently, the most effective method of 286 reducing mortality in such patients is to achieve rapid 287 reperfusion by lysis or mechanical disruption of the occlusive 288 coronary thrombus and plaque. The mortality from acute MI 289 under these circumstances is inversely related to the amount of 290 myocardial salvage achieved by reperfusion. 291 There is increasing evidence from preclinical investigations 292 that inhibition of p38 during prolonged ischaemia slows the rate 293 of infarction/death and inhibits the production of inflammatory 294 cytokines, such as TNF-α, IL-1 and IL-8, which aggravate 295 ischaemic injury (Young et al., 1997; see Table 1 for summary). 296 It was fist demonstrated as early as 1996 that p38α and β 297 are activated in response to ischaemia and reperfusion in the 298 heart (Bogoyevitch et al., 1996). Since then, using gene transfer 299 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 ARTICLE IN PRESS 8 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 OF 307 308 RO 306 DP 305 punctate distribution (Court NW et al., 2002), and p38δ mRNA 357 is broadly expressed in a wide variety of mouse and human 358 tissues including the heart (Wang et al., 1997; Beardmore et al., 359 2005). The C-terminal tail of p38γ allows its interaction with 360 PDZ domains of its substrate protein(s) and association with 361 α1-syntrophin and SAP90/PSD95 in skeletal muscle and 362 neuronal synapses, respectively (Zhang et al., 2003; Hasegawa 363 & Cahill, 2004; Sabio et al., 2004). p38γ-catalysed phosphory- 364 lation of hDlg (the mammalian homologue of the Drosophila 365 tumour suppressor Dlg) triggers its dissociation from the 366 cytoskeleton, indicating that this may regulate the integrity of 367 intercellular-junctional complexes, cell shape, volume and cell 368 polarity in response to many kinds of external stimuli. In support 369 of these findings, Parker et al. identified a novel p38δ substrate as 370 stathmin, a cytoplasmic protein that was previously reported to be 371 a substrate of several intercellular signalling kinases which have 372 been linked to regulation of microtubule (MT) dynamics in a 373 phosphorylation-dependent manner (Belmont & Mitchison, 374 1996). This may suggest that a common theme in p38 pathway 375 activation may be the re-organisation of the cytoskeletal frame- 376 work to enhance cell survival in times of stress such as ischaemia 377 (Parker et al., 1998). Moreover, both p38γ and p38δ phos- 378 phorylate the MT-associated protein Tau in neurons in vivo. 379 Hyperphosphorylated Tau is the major component of the paired 380 helical filaments, which constitute one of the main neuropatho- 381 logical hallmarks of many neurodegenerative disorders (Sabio 382 et al., 2005). So, although there is only circumstantial evidence to 383 support a role for γ and δ isoforms in the heart, this is likely an 384 emerging area of research. In the absence of isoform-selective 385 pharmacological inhibitors, this is, to some extent, aided by the 386 availability of a number of p38 isoform-targeted mouse lines 387 (Beardmore et al., 2005; Sabio et al., 2005) and spontaneously 388 active mutants (Askari et al., 2007). 389 TE 304 EC 302 303 techniques, the α isoform that has been implicated in myocyte apoptosis, consistent with the findings that this isoform alone contributes to cell death following ischaemia (Saurin et al., 2000; Martin et al., 2001). p38s phosphorylate a number of known transcription factors to alter their transactivating potential influencing gene expression. However, the immediate downstream targets of p38 that aggravate myocardial injury are still largely unknown. One downstream substrate of p38α is MAPKAP2, which can, in turn, phosphorylate HSP27, a heat shock protein, which is thought to confer a number of protective effects (Kim et al., 2005). In addition, phosphorylation of MAPKAP2 can also result in phosphorylation of factors that transactivate cytokine genes, such as TNF-α, a cytokine implicated in chronic heart failure. Interestingly, TNF-α also activates p38 and thus p38 has been considered as the keystone in an autoamplifying cytokine cascade by most investigators and an attractive target for antiinflammatory drug development (Lee et al., 2000; Kuma et al., 2005). Furthermore, a proapoptotic role for p38α and/or p38β during myocardial ischaemia is suggested by protection of cardiac myocytes from ischaemic damage using a selective p38α/ p38β isoform inhibitor, SB203580 (Wang et al., 1998a). Using adenoviral-mediated expression of p38α and p38β in rat neonatal cardiomyocytes our group have previously shown that after 2.5 hr simulated ischaemia p38α was activated, whereas p38β activation was significantly inhibited (Saurin et al., 2000). Inhibition of p38α activation during prolonged ischaemia, but not β, resulted in an increase in cell viability (Saurin et al., 2000). This strongly supported Wang et al. who suggested that p38α activation in cardiac myocytes is sufficient to cause apoptosis whereas activation of the β isoforms leads to protection and hypertrophy (Wang et al., 1998a). However, there is some evidence to suggest that p38 isoforms may have potential protective function and suggest a possible adverse effect of prolonged p38 inhibition in the heart. Glembotski's laboratory have demonstrated chronic activation of p38 through overexpression of MKK6 in the heart can result in improved functional recovery from ischaemia and MI (Martindale et al., 2005). The protective role of p38β has also been investigated in a recent study by Kim and co-workers who have shown that activation of p38β by carbon monoxide promotes the nuclear translocation of heat shock factor-1 (HSF-1), which regulates the expression of cytoprotective HSP70 in cells and tissues (Kim et al., 2005). HSF-1 can also serve as a negative regulator of proinflammatory genes, including IL-1β, and TNF-α (Xie et al., 2002). The role of p38s (the α isoform predominantly) in myocardial ischaemic injury has been studied extensively since the findings of (Wang et al. 1998a). These studies, which in the main suggest p38 activation during ischaemia worsens injury and depresses LV function, are too numerous to review and appear in Table 1. There is little information in the literature regarding the roles of either the γ and δ isoform of p38 in the myocardium during ischaemia. Conserved cardiac expression of p38γ amongst several different species suggests that this isoform may play an important role in the heart and therefore is unlikely to be functionally redundant (Court NW et al., 2002). p38γ is localised in the cytoplasm of the cardiac myocyte and is reported to have a CO RR 301 UN 300 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx 6. Pharmacological inhibitors of p38-MAPK Early efforts in drug discovery of small molecule inhibitors of kinases were met with scepticism that selectivity could ever be accomplished, due to the high degree of structural similarity in the adenosine binding pocket among the entire kinome. Thus, it was somewhat of a surprise when SB203580, the first reported p38 inhibitor, emerged showing selectivity over the closely related JNK and ERK MAPK families (Lantos et al., 1984). The pyridinyl imidazole antiinflammatory agents were soon shown to be highly selective p38 inhibitors and the bi-cyclic pyridinyl imidazole SKF-86002 was the first compound reported to inhibit LPS-stimulated cytokine production (Lee et al., 1988, 1994). It was not long before investigators explored dual 5-lipooxygenase/cyclooxygenase (LO/COX) and cytokine inhibition as potential mechanisms for the potent anti-inflammatory activity of these compounds (Lee et al., 1993), and subsequently, SB203580 was used as a pharmacological inhibitor to study the cascade of kinases (via p38) involved in cytokine production (Gallagher et al., 1997). The structures of representative classes of p38 inhibitors are shown in Fig. 3. The crystal structures of pyridinyl imidazole-p38α complexes have recently become available and suggest that SB203580 binds Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 Q2 ARTICLE IN PRESS J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 OF 418 RO 417 DP 416 ketones, indole amides, diamides, quinazolinones, and pyridylaino-quinazolines (Cirillo et al., 2002). Unlike the imidazolebased p38α inhibitors, the urea-containing inhibitors act in a noncompetitive manner (Kulkarni et al., 2007). Crystallographic studies of urea-containing p38α inhibitors, such as BIRB-796, have revealed that these compounds bind to at a site remote from ATP pocket, and induce a significant movement of Phe169, such that this residue fills the ATP pocket, preventing ATP binding (Pargellis et al., 2002). Thus, inhibitors of p38 can be divided into 2 groups dependent upon their mode of binding to p38; these are (i) active site or “gatekeeping” inhibitors (such as SB203580) and (ii) those which bind remotely and interfere with ATP binding indirectly (such as BIRB-796). Enthusiasm for “blanket” pharmacological inhibition of p38 is tempered by the fact that this kinase is involved in innumerable biological processes and therefore not surprising that under many circumstances its activation leads to myocardial protection rather than injury (Weinbrenner et al., 1997; Craig et al., 2000; Hoover et al., 2000; Communal et al., 2000; Force et al., 2004; Zheng & Zuo, 2004; Martindale et al., 2005). This particularly seems to be the case when p38 activation occurs as a consequence of an intervention that precedes lethal myocardial ischaemia, such as ischaemic or pharmacological preconditioning (Nagarkatti & Afi, 1998; Marais et al., 2001; Sanada et al., 2001). However, in these studies the same inhibitor, at the same concentration, reduces injury if present solely during lethal ischaemic injury (Nagarkatti & Afi, 1998; Marais et al., 2001; Sanada et al., 2001; Tanno et al., 2003). The cause of this apparent paradoxical observation may relate to an attenuation of p38 activation during lethal ischaemia by its prior transient activation. Thus there is ample evidence from cardiac as well as other research fields that p38 activation can have beneficial consequences whilst it is also incontrovertible that restricting p38 inhibition to the activation that accompanies lethal myocardial ischaemia reduces infarction (Nagarkatti & Afi, 1998; Marais et al., 2001; Sanada et al., 2001; Marais et al., 2005; Table 1). However, if we are to consider currently available p38 inhibitors with the aim of treating chronic conditions, it is likely greater levels of selectivity will be required to avoid the inhibition of beneficial forms of activation. As more kinases are investigated, a better understanding of selectivity over the kinome has followed. With expanded kinase panels, it has now emerged that “classic” p38α inhibitors like SB203580, which were once described as being selective over certain kinases, now have been shown to have similar, and sometimes lower IC50s. These actions are a result of similarities in the inhibitory binding site or in particular the hydrophobic pocket equivalent to that formed by Thr106 (Fabian et al., 2005). So if broad inhibition of p38 is not the answer, what is? At present, the majority of pharmacological inhibitors of p38 are selective for the α and β isoforms of the kinase. It is clear from the published data that the during prolonged ischaemia the α isoform plays an important role in the progression of dysfunction. Perhaps a more rational approach to inhibit p38 in a site- and condition-specific manner might be to target the activation of the kinase pathway upstream of p38 itself, such as TAB1 or MKK3/MKK6. Using a model of TE 415 EC 414 to the active site of both phosphorylated (active) and unphosphorylated (inactive) p38 in an ATP-competitive manner (see Figs. 2 and 3). These inhibitors bind to an aryl-specificity pocket behind the site, which is normally occupied by the adenine ring of ATP. The interaction occurs between the 4-pyridinyl group (analogous to the N-1 adenine of ATP) and the N-H of Met109. In addition, studies have also have implicated Thr106 as a key residue conferring selectivity (Wilson et al., 1996; Gum et al., 1998). The 2 adjacent residues, His107 and Leu108, along with Thr106 lie at the back of the ATP pocket and are identical in p38α and p38β, but are different in p38γ and p38δ (Met106, Pro107 and Phe108, respectively) which are insensitive to SB203580 inhibition. Using a mutagenesis approach it has been shown that if these 3 residues in p38α and p38β are changed to Met-Pro-Phe (as found in p38γ and p38δ) the mutant kinase is no longer inhibited by SB203580. By contrast, introduction of the Thr-His-Leu sequence of p38α into p38γ or p38δ confers sensitivity to SB203580. Taken together, these studies have identified Thr106 as the key residue forming the aryl-specificity pocket (Saccani et al., 2002). In addition to novel prydinyl imidazole compounds, a new group of selective p38 inhibitors are the arly-pyridinyle-heterocylces. In these compounds, the imidazole core is replaced by other heteroaryl scaffolding, and consequently these compounds generally exhibit in vitro potency similar to prydinyl imidazoles (Boehm et al., 2001). Other structurally diverse p38 inhibitors include a subset of novel non-aryl-pyridinyls such as triazanapthalenones, N,N′-diary ureas, benzzophenones, pyrazole CO RR 413 UN 412 Fig. 2. Crystal structure of p38 with SB203580 occupying the ATP binding site. The Thr106 residue (4), which is important for binding of pyridinyl imidazole inhibitors, and the 2 residues within the activation loop that are phosphorylated (Thr180 (2) and Try182 (1)) are highlighted. Tyr323 (3), which has been implicated in TCR-mediated activation of p38 is also shown. SB203580 is shown in green. The activation loop is shown in orange. ED and CD activator/substrate binding regions are highlighted. The C-terminal extension that forms the L16 loop bridging the domains is also indicated (created in JenaLib Jmol; PDB entry 1a9u; Wang et al., 1998b). 9 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 ARTICLE IN PRESS 10 UN CO RR EC TE DP RO OF J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx Fig. 3. Structure of representative classes of p38 MAPK inhibitors. p38 inhibitors can be divided into 2 groups dependant upon their mode of binding to p38; active site inhibitors, such as SB203580 and RJW-67657, bind competitively to the ATP site of the enzyme whereas others bind remotely and interfere with ATP binding indirectly (such as BIRB-796). 496 497 498 coronary artery ligation in a mkk3-targetted mouse line, we have recently demonstrated that removing MKK3 does not alter pathological remodelling and progression to ventricular dys- function after MI (Clark et al., 2007). Maybe this is not sur- 499 prising considering the multitude of pathways involved in 500 ischaemia and inflammation but it does, perhaps, highlight the 501 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 ARTICLE IN PRESS J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx 7. Clinical trials of p38-MAPK inhibition 552 8. Summary and conclusions 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 553 554 EC 508 CO RR 507 UN 506 TE 551 Despite the apparent dichotomy in preclinical research of the consequences of p38 inhibition, 2 types of molecular inhibitors of p38, aryl-prydinyl heterocycles (namely SB242235 and RWJ67657) and non-aryl-prydinyl heterocycles (VX-745, BIRB-796 and RO3201195), have so far advanced to clinical trials. Results of phase I and early phase II trials were also promising. In one study, orally administered SB242235 (1–500 mg), which has been shown to have potent antiinflammatory effects in a rat model of arthritis (Badger et al., 2000), was well tolerated and suppressed production of TNF-α, IL-1β, IL-6, and IL-8 in a dose-dependent manner within 3 hr (Adams et al., 2001). A randomised clinical study to examine the efficacy of RWJ-67657 to combat the effects of endotoxin on normal healthy volunteer was recently carried out. Development of flu-like symptoms, which were associated with raised serum levels of TNF-α, IL-6 and IL-8, were reduced in a dose dependent manner by RWJ67657 (Fijen et al., 2001). However, clinical studies have not been limited to healthy controls; VX-745 (Vertex) has been given to patients with active rheumatoid arthritis (Haddad, 2001; Weisman, 2002), and although the drug is well tolerated, it was associated with adverse effects, such as elevation in liver transaminases. Preclinical safety evaluations of this drug in animals have revealed that at that concentrations VX-745 can cross the blood–brain barrier and exert adverse neurological side effects (Weisman, 2002). For these reasons, further investigation on VX-745 has been suspended. The rise in the level of liver transaminases has also been observed in a series of double-blinded, randomised, placebo-controlled studies of BIRB-796 in healthy volunteers. This compound also suppressed neutrophil activation ex vivo, but no inhibition of LPSinduced TNF-α production was observed (Wood et al., 2002). Currently, the most clinically advanced p38 inhibitors are the Scios compound (now Johnson & Johnson) SCIO-323 for treatment of stroke and the Vertex compound VX-702, which has been tested in patients with acute coronary syndromes in whom percutaneous coronary intervention (PCI) is planned. There were no adverse side effects and the serum level of C-reactive protein (CRP), which is considered a risk factor in MI patients, was suppressed (de Winter et al., 2005). However, although many p38 inhibitors have advanced to phase I, II or III clinical trial, many of these studies have been stopped prematurely due to adverse side effects. One reason for this might be the crossreactivity against other kinases or other cellular signalling molecules. An alternative explanation is that p38-dependant signalling is vital to normal cell function necessitating a greater understanding of mechanisms of activation in the hope that this will reveal circumstance-specific targets. 505 OF 504 biological processes, such as cell growth, apoptosis and inflammation. In the heart p38 is activated by various pathological conditions, such as ischaemia and pressure overload. Studies using genetic and pharmacological inhibitors of p38 suggests that this kinase plays a key role in left ventricular matrix remodelling (Petrich & Wang, 2004), cell survival following ischaemia–reperfusion (Ma et al., 1999; Shao et al., 2006) and possibly, in post MI remodelling (Ren et al., 2005). Investigators have shown that multiple isoforms of p38 are activated in response to specific stimuli and take part in distinct signalling pathways, which result in activation of specific downstream substrates. From the existing evidence, it appears that p38α and p38β are differentially regulated during myocardial stresses and that the consequences of activation of each isoform may differ by cell type. This highlights the likelihood that different members within a single kinase family can play distinct roles in the heart during ischaemia. Despite continued interest in the p38 pathway few studies to date have addressed the role of p38 isoforms other than p38α during ischaemia. Furthermore, since there are no isoform-specific pharmacological inhibitors of p38 activity, the contribution of each isoform remains unclear. Understanding the physiological roles of each p38 isoform and identifying their mechanism(s) of activation and potential substrates are important avenues that may lead to pharmacological inhibitors with greater circumstance selectivity thereby avoiding the potential pitfalls of chronic systemic inhibition. RO potential importance of other pathways which warrant further investigation. DP 503 502 11 The p38 kinase pathway has been studied intensely since its discovery in the early 1990s. It has been implicated in various 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 References 581 Adams, R. H., Porras, A., Alonso, G., Jones, M., Vintersten, K., Panelli, S., et al. (2000). Essential role of p38[alpha] MAP kinase in placental but not embryonic cardiovascular development. Mol Cell 6(1), 109−116. Adams, J. L., Badger, A. M., Kumar, S., & Lee, J. C. (2001). p38 MAP kinase: molecular target for the inhibition of pro-inflammatory cytokines. Prog Med Chem 38, 1−60. Aleshin, A., Sawa, Y., Ono, M., Funatsu, T., Miyagawa, S., & Matsuda, H. (2004). Myocardial protective effect of FR167653; a novel cytokine inhibitor in ischemic-reperfused rat heart. Eur J Cardio-Thorac Surg 26(5), 974−980. Allen, M., Svensson, L., Roach, M., Hambor, J., McNeish, J., & Gabel, C. A. (2000). Deficiency of the stress kinase p38{alpha} results in embryonic lethality: characterization of the kinase dependence of stress responses of enzyme-deficient embryonic stem cells. J Exp Med 191(5), 859−870. Askari, N., Diskin, R., Avitzour, M., Capone, R., Livnah, O., & Engelberg, D. (2007). Hyperactive variants of p38alpha induce, whereas hyperactive variants of p38gamma suppress, activating protein 1-mediated transcription. J Biol Chem 282(1), 91−99. Badger, A. M., Bradbeer, J. N., Votta, B., Lee, J. C., Adams, J. L., & Griswold, D. E. (1996). Pharmacological profile of SB 203580, a selective inhibitor of cytokine suppressive binding protein/p38 kinase, in animal models of arthritis, bone resorption, endotoxin shock and immune function. J Pharmacol Exp Ther 279(3), 1453−1461. Badger, A. M., Griswold, D. E., Kapadia, R., Blake, S., Swift, B. A., Hoffman, S. J., et al. (2000). Disease-modifying activity of SB 242235, a selective inhibitor of p38 mitogen-activated protein kinase, in rat adjuvant-induced arthritis. Arthritis Rheum 43(1), 175−183. Barancik, M., Htun, P., Strohm, C., Kilian, S., & Schaper, W. (2000). Inhibition of the cardiac p38-MAPK pathway by SB203580 delays ischemic cell death. J Cardiovasc Pharmacol 35(3), 474−483. Beardmore, V. A., Hinton, H. J., Eftychi, C., Apostolaki, M., Armaka, M., Darragh, J., et al. (2005). Generation and characterization of p38beta (MAPK11) gene-targeted mice. Mol Cell Biol 25(23), 10454−10464. Behr, T. M., Nerurkar, S. S., Nelson, A. H., Coatney, R. W., Woods, T. N., Sulpizio, A., et al. (2001). Hypertensive end-organ damage and premature 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 ARTICLE IN PRESS 12 DP RO OF de Winter, R. J., Tijssen, J. G. P., Windhausen, F., Krasznai, K., Michels, H. R., Ziekenhuis, C., et al. (2005). A major determinant of C-reactive protein production in patients with acute coronary syndromes undergoing PCI: the p38 mitogen activated protein kinase signalling pathway. American Heart Association Scientific Sessions. Deacon, K., & Blank, J. L. (1997). Characterization of the mitogen-activated protein kinase kinase 4(MKK4)/c-Jun NH2-terminal kinase 1 and MKK3/ p38 pathways regulated by MEK kinases 2á and 3. MEK kinase 3 activates MKK3 but does not cause activation of p38 kinase in vivo. J Biol Chem 272(22), 14489−14496. Derijard, B., Raingeaud, J., Barrett, T., Wu, I. H., Han, J., Ulevitch, R. J., et al. (1995). Independent human MAP-kinase signal transduction pathways defined by MEK and MKK isoforms. Science 267(5198), 682−685. Diskin, R., Askari, N., Capone, R., Engelberg, D., & Livnah, O. (2004). Active mutants of the human p38alpha mitogen-activated protein kinase. J Biol Chem 279(45), 47040−47049. Diskin, R., Lebendiker, M., Engelberg, D., & Livnah, O. (2007). Structures of p38alpha active mutants reveal conformational changes in L16 loop that induce autophosphorylation and activation. J Mol Biol 365(1), 66−76. Dong, C., Davis, R. J., & Flavell, R. A. (2002). MAP kinases in the immune response. Annu Rev Immunol 20, 55−72. Engel, F. B., Hsieh, P. C., Lee, R. T., & Keating, M. T. (2006). FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction. Proc Natl Acad Sci U S A 103(42), 15546−15551. English, J., Pearson, G., Wilsbacher, J., Swantek, J., Karandikar, M., Xu, S., et al. (1999). New insights into the control of MAP kinase pathways. Exp Cell Res 253(1), 255−270. Eyers, P. A., van, d. I., Quinlan, R. A., Goedert, M., & Cohen, P. (1999). Use of a drug-resistant mutant of stress-activated protein kinase 2a/p38 to validate the in vivo specificity of SB 203580. FEBS Lett 451(2), 191−196. Fabian, M. A., Biggs, W. H., III, Treiber, D. K., Atteridge, C. E., Azimioara, M. D., Benedetti, M. G., et al. (2005). A small molecule-kinase interaction map for clinical kinase inhibitors. Nat Biotechnol 23(3), 329−336. Fiedler, B., Feil, R., Hofmann, F., Willenbockel, C., Drexler, H., Smolenski, A., et al. (2006). cGMP-dependent protein kinase type I inhibits TAB1-p38 mitogen-activated protein kinase apoptosis signaling in cardiac myocytes. J Biol Chem 281(43), 32831−32840. Fijen, J. W., Zijlstra, J. G., De, B. P., Spanjersberg, R., Tervaert, J. W., van der Werf, T. S., et al. (2001). Suppression of the clinical and cytokine response to endotoxin by RWJ-67657, a p38 mitogen-activated protein-kinase inhibitor, in healthy human volunteers. Clin Exp Immunol 124(1), 16−20. Force, T., Kuida, K., Namchuk, M., Parang, K., & Kyriakis, J. M. (2004). Inhibitors of protein kinase signaling pathways: emerging therapies for cardiovascular disease. Circulation 109(10), 1196−1205. Freshney, N. W., Rawlinson, L., Guesdon, F., Jones, E., Cowley, S., Hsuan, J., et al. (1994). Interleukin-1 activates a novel protein kinase cascade that results in the phosphorylation of Hsp27. Cell 78(6), 1039−1049. Gallagher, T. F., Seibel, G. L., Kassis, S., Laydon, J. T., Blumenthal, M. J., Lee, J. C., et al. (1997). Regulation of stress-induced cytokine production by pyridinylimidazoles; inhibition of CSBP kinase. Bioorg Med Chem 5(1), 49−64. Gallo, K. A., & Johnson, G. L. (2002). Mixed-lineage kinase control of JNK and p38 MAPK pathways. Nat Rev Mol Cell Biol 3(9), 663−672. Ge, B., Gram, H., Di, P. F., Huang, B., New, L., Ulevitch, R. J., et al. (2002). MAPKK-independent activation of p38alpha mediated by TAB1-dependent autophosphorylation of p38alpha. Science 295(5558), 1291−1294. Gorog, D. A., Tanno, M., Cao, X., Bellahcene, M., Bassi, R., Kabir, A. M., et al. (2004). Inhibition of p38 MAPK activity fails to attenuate contractile dysfunction in a mouse model of low-flow ischemia. Cardiovasc Res 61(1), 123−131. Gum, R. J., McLaughlin, M. M., Kumar, S., Wang, Z., Bower, M. J., Lee, J. C., et al. (1998). Acquisition of sensitivity of stress-activated protein kinases to the p38 inhibitor, SB 203580, by alteration of one or more amino acids within the ATP binding pocket. J Biol Chem 273(25), 15605−15610. Gupta, A., Aberle, N. S., Kapoor, R., Ren, J., & Sharma, A. C. (2005). Bigendothelin-1 via p38-MAPK-dependent mechanism regulates adult rat ventricular myocyte contractility in sepsis. Biochim Biophys Acta 1741(1-2), 127−139. CO RR EC TE mortality are p38 mitogen-activated protein kinase-dependent in a rat model of cardiac hypertrophy and dysfunction. Circulation 104(11), 1292−1298. Bell, M., Capone, R., Pashtan, I., Levitzki, A., & Engelberg, D. (2001). Isolation of hyperactive mutants of the MAPK p38/Hog1 that are independent of MAPK kinase activation. J Biol Chem 276(27), 25351−25358. Bellahcene, M., Jacquet, S., Cao, X. B., Tanno, M., Haworth, R. S., Layland, J., et al. (2006). Activation of p38 mitogen-activated protein kinase contributes to the early cardiodepressant action of tumor necrosis factor. J Am Coll Cardiol 48, 545−555. Belmont, L. D., & Mitchison, T. J. (1996). Identification of a protein that interacts with tubulin dimers and increases the catastrophe rate of microtubules. Cell 84(4), 623−631. Boehm, J. C., Bower, M. J., Gallagher, T. F., Kassis, S., Johnson, S. R., & Adams, J. L. (2001). Phenoxypyrimidine inhibitors of p38alpha kinase: synthesis and statistical evaluation of the p38 inhibitory potencies of a series of 1-(piperidin-4-yl)-4-(4-fluorophenyl)-5-(2-phenoxypyrimidin-4-yl) imidazoles. Bioorg Med Chem Lett 11(9), 1123−1126. Bogoyevitch, M. A., Gillespie-Brown, J., Ketterman, A. J., Fuller, S. J., BenLevy, R., Ashworth, A., et al. (1996). Stimulation of the stress-activated mitogen-activated protein kinase subfamilies in perfused heart. p38/RK mitogen-activated protein kinases and c-Jun N-terminal kinases are activated by ischemia/reperfusion. Circ Res 79(2), 162−173. Brancho, D., Tanaka, N., Jaeschke, A., Ventura, J. J., Kelkar, N., Tanaka, Y., et al. (2003). Mechanism of p38 MAP kinase activation in vivo. Genes Dev 17(16), 1969−1978. Braz, J. C., Bueno, O. F., Liang, Q., Wilkins, B. J., Dai, Y. S., Parsons, S., et al. (2003). Targeted inhibition of p38 MAPK promotes hypertrophic cardiomyopathy through upregulation of calcineurin-NFAT signaling. J Clin Invest 111(10), 1475−1486. Cain, B. S., Meldrum, D. R., Meng, X., Dinarello, C. A., Shames, B. D., Banerjee, A., et al. (1999). p38 MAPK inhibition decreases TNF-[alpha] production and enhances postischemic human myocardial function. J Surg Res 83(1), 7−12. Carter, A. B., Monick, M. M., & Hunninghake, G. W. (1999). Both Erk and p38 kinases are necessary for cytokine gene transcription. Am J Respir Cell Mol Biol 20(4), 751−758. Chang, C. I., Xu, B. E., Akella, R., Cobb, M. H., & Goldsmith, E. J. (2002). Crystal structures of MAP kinase p38 complexed to the docking sites on its nuclear substrate MEF2A and activator MKK3b. Mol Cell 9(6), 1241−1249. Cheung, P. C., Campbell, D. G., Nebreda, A. R., & Cohen, P. (2003). Feedback control of the protein kinase TAK1 by SAPK2a/p38alpha. EMBO J 22(21), 5793−5805. Cirillo, P. F., Pargellis, C., & Regan, J. (2002). The non-diaryl heterocycle classes of p38 MAP kinase inhibitors. Curr Top Med Chem 2(9), 1021−1035. Clanachan, A. S., Jaswal, J. S., Gandhi, M., Bottorff, D. A., Coughlin, J., Finegan, B. A., et al. (2003). Effects of inhibition of myocardial extracellularresponsive kinase and P38 mitogen-activated protein kinase on mechanical function of rat hearts after prolonged hypothermic ischemia. Transplantation 75(2), 173−180. Clark, J. E., Flavell, R. A., Faircloth, M. E., Davis, R. J., Heads, R. J., & Marber, M. S. (2007). Post-infarction remodeling is independent of mitogenactivated protein kinase kinase 3 (MKK3). Cardiovasc Res. Communal, C., Colucci, W. S., & Singh, K. (2000). p38 mitogen-activated protein kinase pathway protects adult rat ventricular myocytes against betaadrenergic receptor-stimulated apoptosis. Evidence for Gi-dependent activation. J Biol Chem 275(25), 19395−19400. Cook, S. A., Sugden, P. H., & Clerk, A. (1999). Activation of c-Jun N-terminal kinases and p38-mitogen-activated protein kinases in human heart failure secondary to ischaemic heart disease. J Mol Cell Cardiol 31(8), 1429−1434. Court, N. W., dos Remedios, C. G., Cordell, J., & Bogoyevitch, M. A. (2002). Cardiac expression and subcellular localization of the p38 mitogen-activated protein kinase member, stress-activated protein kinase-3 (SAPK3). J Mol Cell Cardiol 34(4), 413−426. Craig, R., Larkin, A., Mingo, A. M., Thuerauf, D. J., Andrews, C., McDonough, P. M., et al. (2000). p38 MAPK and NF-kappa B collaborate to induce interleukin-6 gene expression and release. Evidence for a cytoprotective autocrine signaling pathway in a cardiac myocyte model system. J Biol Chem 275(31), 23814−23824. UN 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 Q3667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 ARTICLE IN PRESS J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx DP RO OF Kuma, Y., Sabio, G., Bain, J., Shpiro, N., Marquez, R., & Cuenda, A. (2005). BIRB796 inhibits all p38 MAPK isoforms in vitro and in vivo. J Biol Chem 280(20), 19472−19479. Lantos, I., Bender, P. E., Razgaitis, K. A., Sutton, B. M., DiMartino, M. J., Griswold, D. E., et al. (1984). Antiinflammatory activity of 5,6-diaryl-2,3dihydroimidazo[2,1-b]thiazoles. Isomeric 4-pyridyl and 4-substituted phenyl derivatives. J Med Chem 27(1), 72−75. Lee, J. C., Griswold, D. E., Votta, B., & Hanna, N. (1988). Inhibition of monocyte IL-1 production by the anti-inflammatory compound, SK&F 86002. Int J Immunopharmacol 10(7), 835−843. Lee, J. C., Badger, A. M., Griswold, D. E., Dunnington, D., Truneh, A., Votta, B., et al. (1993). Bicyclic imidazoles as a novel class of cytokine biosynthesis inhibitors. Ann N Y Acad Sci 696, 149−170. Lee, J. C., Laydon, J. T., McDonnell, P. C., Gallagher, T. F., Kumar, S., Green, D., et al. (1994). A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature 372(6508), 739−746. Lee, J. C., Kumar, S., Griswold, D. E., Underwood, D. C., Votta, B. J., & Adams, J. L. (2000). Inhibition of p38 MAP kinase as a therapeutic strategy. Immunopharmacology 47(2-3), 185−201. Lemke, L. E., Bloem, L. J., Fouts, R., Esterman, M., Sandusky, G., & Vlahos, C. J. (2001). Decreased p38 MAPK activity in end-stage failing human myocardium: p38 MAPK alpha is the predominant isoform expressed in human heart. J Mol Cell Cardiol 33(8), 1527−1540. Li, M., Georgakopoulos, D., Lu, G., Hester, L., Kass, D. A., Hasday, J., et al. (2005). p38 MAP kinase mediates inflammatory cytokine induction in cardiomyocytes and extracellular matrix remodeling in heart. Circulation 111(19), 2494−2502. Li, Z., Ma, J. Y., Kerr, I., Chakravarty, S., Dugar, S., Schreiner, G., et al. (2006). Selective inhibition of p38alpha MAPK improves cardiac function and reduces myocardial apoptosis in rat model of myocardial injury. Am J Physiol Heart Circ Physiol 291(4), H1972−H1977. Lim, M., Martinez, T., Jablons, D., Cameron, R., Guo, H., Toole, B., et al. (1998). Tumor-derived EMMPRIN (extracellular matrix metalloproteinase inducer) stimulates collagenase transcription through MAPK p38. FEBS Lett 441(1), 88−92. Liu, Y. H., Wang, D., Rhaleb, N. E., Yang, X. P., Xu, J., Sankey, S. S., et al. (2005). Inhibition of p38 mitogen-activated protein kinase protects the heart against cardiac remodeling in mice with heart failure resulting from myocardial infarction. J Card Fail 11(1), 74−81. Lu, G., Kang, Y. J., Han, J., Herschman, H. R., Stefani, E., & Wang, Y. (2005). TAB-1 modulates intracellular localization of p38 MAP kinase and downstream signaling. J Biol Chem, M507610200. Luss, H., Neumann, J., Schmitz, W., Schulz, R., & Heusch, G. (2000). The stress-responsive MAP kinase p38 is activated by low-flow ischemia in the in situ porcine heart. J Mol Cell Cardiol 32(10), 1787−1794. Ma, X. L., Kumar, S., Gao, F., Louden, C. S., Lopez, B. L., Christopher, T. A., et al. (1999). Inhibition of p38 mitogen-activated protein kinase decreases cardiomyocyte apoptosis and improves cardiac function after myocardial ischemia and reperfusion. Circulation 99(13), 1685−1691. Mackay, K., & Mochly-Rosen, D. (1999). An inhibitor of p38 mitogen-activated protein kinase protects neonatal cardiac myocytes from ischemia. J Biol Chem 274(10), 6272−6279. Mackay, K., & Mochly-Rosen, D. (2000). Involvement of a p38 mitogenactivated protein kinase phosphatase in protecting neonatal rat cardiac myocytes from ischemia. J Mol Cell Cardiol 32(8), 1585−1588. Marais, E., Genade, S., Huisamen, B., Strijdom, J. G., Moolman, J. A., & Lochner, A. (2001). Activation of p38 MAPK induced by a multi-cycle ischaemic preconditioning protocol is associated with attenuated p38 MAPK activity during sustained ischaemia and reperfusion. J Mol Cell Cardiol 33(4), 769−778. Marais, E., Genade, S., Salie, R., Huisamen, B., Maritz, S., Moolman, J. A., et al. (2005). The temporal relationship between p38 MAPK and HSP27 activation in ischaemic and pharmacological preconditioning. Basic Res Cardiol 100(1), 35−47. Martin, J. L., Avkiran, M., Quinlan, R. A., Cohen, P., & Marber, M. S. (2001). Antiischemic effects of SB203580 are mediated through the inhibition of p38{alpha} mitogen-activated protein kinase: evidence from ectopic expression of an inhibition-resistant kinase. Circ Res 89(9), 750−752. CO RR EC TE Gysembergh, A., Simkhovich, B. Z., Kloner, R. A., & Przyklenk, K. (2001). p38 MAPK activity is not increased early during sustained coronary artery occlusion in preconditioned versus control rabbit heart. J Mol Cell Cardiol 33(4), 681−690. Haar, E. T., Prabakhar, P., Liu, X., & Lepre, C. (2007). Crystal structure of the P38 {alpha}-MAPKAP kinase 2 heterodimer. J Biol Chem 282(13), 9733−9739. Haddad, J. J. (2001). VX-745. Vertex Pharmaceuticals. Curr Opin Investig Drugs 2(8), 1070−1076. Han, J., Lee, J. D., Bibbs, L., & Ulevitch, R. J. (1994). A MAP kinase targeted by endotoxin and hyperosmolarity in mammalian cells. Science 265(5173), 808−811. Han, J., Richter, B., Li, Z., Kravchenko, V., & Ulevitch, R. J. (1995). Molecular cloning of human p38 MAP kinase. Biochim Biophys Acta 1265(2-3), 224−227. Han, J., Lee, J. D., Jiang, Y., Li, Z., Feng, L., & Ulevitch, R. J. (1996). Characterization of the structure and function of a novel MAP kinase kinase (MKK6). J Biol Chem 271(6), 2886−2891. Hasegawa, M., & Cahill, G. M. (2004). Regulation of the circadian oscillator in Xenopus retinal photoreceptors by protein kinases sensitive to the stressactivated protein kinase inhibitor, SB 203580. J Biol Chem 279(21), 22738−22746. Hoover, H. E., Thuerauf, D. J., Martindale, J. J., & Glembotski, C. C. (2000). alpha B-crystallin gene induction and phosphorylation by MKK6-activated p38. A potential role for alpha B-crystallin as a target of the p38 branch of the cardiac stress response. J Biol Chem 275(31), 23825−23833. House, S. L., Branch, K., Newman, G., Doetschman, T., & Schultz, J. J. (2005). Cardioprotection induced by cardiac-specific overexpression of fibroblast growth factor-2 is mediated by the MAPK cascade. Am J Physiol Heart Circ Physiol 289(5), H2167−H2175. Hutchison, M., Berman, K. S., & Cobb, M. H. (1998). Isolation of TAO1, a protein kinase that activates MEKs in stress-activated protein kinase cascades. J Biol Chem 273(44), 28625−28632. Ichijo, H., Nishida, E., Irie, K., ten, D. P., Saitoh, M., Moriguchi, T., et al. (1997). Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science 275(5296), 90−94. Kabir, A. M., Cao, X., Gorog, D. A., Tanno, M., Bassi, R., Bellahcene, M., et al. (2005). Antimycin A induced cardioprotection is dependent on pre-ischemic p38-MAPK activation but independent of MKK3. J Mol Cell Cardiol 39(4), 709−717. Kaiser, R. A., Bueno, O. F., Lips, D. J., Doevendans, P. A., Jones, F., Kimball, T. F., et al. (2004). Targeted inhibition of p38 mitogen-activated protein kinase antagonizes cardiac injury and cell death following ischemia-reperfusion in vivo. J Biol Chem 279(15), 15524−15530. Kang, Y. J., Seit-Nebi, A., Davis, R. J., & Han, J. (2006). Multiple activation mechanisms of p38alpha mitogen-activated protein kinase. J Biol Chem 281 (36), 26225−26234. Khan, M., Varadharaj, S., Ganesan, L. P., Shobha, J. C., Naidu, M. U., Parinandi, N. L., et al. (2006). C-phycocyanin protects against ischemia–reperfusion injury of heart through involvement of p38 MAPK and ERK signaling. Am J Physiol Heart Circ Physiol 290(5), H2136−H2145. Kim, H. P., Wang, X., Zhang, J., Suh, G. Y., Benjamin, I. J., Ryter, S. W., et al. (2005). Heat shock protein-70 mediates the cytoprotective effect of carbon monoxide: involvement of p38{beta} MAPK and heat shock factor-1. J Immunol 175(4), 2622−2629. Kim, J. K., Pedram, A., Razandi, M., & Levin, E. R. (2006). Estrogen prevents cardiomyocyte apoptosis through inhibition of reactive oxygen species and differential regulation of p38 kinase isoforms. J Biol Chem 281(10), 6760−6767. Koike, N., Takeyoshi, I., Ohki, S., Tokumine, M., Matsumoto, K., & Morishita, Y. (2004). Effects of adding P38 mitogen-activated protein-kinase inhibitor to celsior solution in canine heart transplantation from non-heart-beating donors. Transplantation 77(2), 286−292. Kotlyarov, A., Neininger, A., Schubert, C., Eckert, R., Birchmeier, C., Volk, H. D., et al. (1999). MAPKAP kinase 2 is essential for LPS-induced TNF-alpha biosynthesis. Nat Cell Biol 1(2), 94−97. Kulkarni, R. G., Srivani, P., Achaiah, G., & Sastry, G. N. (2007). Strategies to design pyrazolyl urea derivatives for p38 kinase inhibition: a molecular modeling study. J Comput Aided Mol Des. UN 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 Q4819 13 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 ARTICLE IN PRESS 14 DP RO OF Rincon, M., & Pedraza-Alva, G. (2003). JNK and p38 MAP kinases in CD4+ and CD8+T cells. Immunol Rev 192, 131−142. Rouse, J., Cohen, P., Trigon, S., Morange, M., Onso-Llamazares, A., Zamanillo, D., et al. (1994). A novel kinase cascade triggered by stress and heat shock that stimulates MAPKAP kinase-2 and phosphorylation of the small heat shock proteins. Cell 78(6), 1027−1037. Sabio, G., Reuver, S., Feijoo, C., Hasegawa, M., Thomas, G. M., Centeno, F., et al. (2004). Stress- and mitogen-induced phosphorylation of the synapse-associated protein SAP90/PSD-95 by activation of SAPK3/p38gamma and ERK1/ERK2. Biochem J 380(Pt 1), 19−30. Sabio, G., Arthur, J. S., Kuma, Y., Peggie, M., Carr, J., Murray-Tait, V., et al. (2005). p38gamma regulates the localisation of SAP97 in the cytoskeleton by modulating its interaction with GKAP. EMBO J 24(6), 1134−1145. Saccani, S., Pantano, S., & Natoli, G. (2002). p38-Dependent marking of inflammatory genes for increased NF-kappa B recruitment. Nat Immunol 3(1), 69−75. Salvador, J. M., Mittelstadt, P. R., Belova, G. I., Fornace, A. J., Jr., & Ashwell, J. D. (2005). The autoimmune suppressor Gadd45alpha inhibits the T cell alternative p38 activation pathway. Nat Immunol 6(4), 396−402. Salvador, J. M., Mittelstadt, P. R., Guszczynski, T., Copeland, T. D., Yamaguchi, H., Appella, E., et al. (2005). Alternative p38 activation pathway mediated by T cell receptor-proximal tyrosine kinases. Nat Immunol 6(4), 390−395. Sanada, S., Kitakaze, M., Papst, P. J., Hatanaka, K., Asanuma, H., Aki, T., et al. (2001). Role of Phasic Dynamism of p38 Mitogen-Activated Protein Kinase Activation in Ischemic Preconditioning of the Canine Heart. Circ Res 88(2), 175−180. Saurin, A. T., Martin, J. L., Heads, R. J., Foley, C. L. A. I., Mockridge, J. W., Wright, M. J., et al. (2000). The role of differential activation of p38mitogen-activated protein kinase in preconditioned ventricular myocytes. FASEB J 14(14), 2237−2246. Schneider, S., Chen, W., Hou, J., Steenbergen, C., & Murphy, E. (2001). Inhibition of p38 MAPK {alpha}/{beta} reduces ischemic injury and does not block protective effects of preconditioning. Am J Physiol Heart Circ Physiol 280(2), H499−H508. See, F., Thomas, W., Way, K., Tzanidis, A., Kompa, A., Lewis, D., et al. (2004). p38 mitogen-activated protein kinase inhibition improves cardiac function and attenuates left ventricular remodeling following myocardial infarction in the rat. J Am Coll Cardiol 44(8), 1679−1689. Shao, Z., Bhattacharya, K., Hsich, E., Park, L., Walters, B., Germann, U., et al. (2006). c-Jun N-Terminal Kinases Mediate Reactivation of Akt and Cardiomyocyte Survival After Hypoxic Injury In Vitro and In Vivo. Circ Res 98(1), 111−118. Sharov, V. G., Todor, A., Suzuki, G., Morita, H., Tanhehco, E. J., & Sabbah, H. N. (2003). Hypoxia, angiotensin-II, and norepinephrine mediated apoptosis is stimulus specific in canine failed cardiomyocytes: a role for p38 MAPK, Fas-L and cyclin D1. Eur J Heart Fail 5(2), 121−129. Sugden, P. H., & Clerk, A. (1998). “Stress-responsive” mitogen-activated protein kinases (c-Jun N-terminal kinases and p38 mitogen-activated protein kinases) in the myocardium. Circ Res 83(4), 345−352. Sumida, T., Otani, H., Kyoi, S., Okada, T., Fujiwara, H., Nakao, Y., et al. (2005). Temporary blockade of contractility during reperfusion elicits a cardioprotective effect of the p38 MAP kinase inhibitor SB-203580. Am J Physiol Heart Circ Physiol 288(6), H2726−H2734. Swynghedauw, B. (1999). Molecular mechanisms of myocardial remodeling. Physiol Rev 79(1), 215−262. Tamura, K., Sudo, T., Senftleben, U., Dadak, A. M., Johnson, R., & Karin, M. (2000). Requirement for p38alpha in erythropoietin expression: a role for stress kinases in erythropoiesis. Cell 102(2), 221−231. Tanno, M., Bassi, R., Gorog, D. A., Saurin, A. T., Jiang, J., Heads, R. J., et al. (2003). Diverse mechanisms of myocardial p38 mitogen-activated protein kinase activation: evidence for MKK-independent activation by a TAB1associated mechanism contributing to injury during myocardial ischemia. Circ Res 93(3), 254−261. Tanoue, T., Adachi, M., Moriguchi, T., & Nishida, E. (2000). A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat Cell Biol 2(2), 110−116. The United Kingdom Heart Attack Study (UKHAS) Collaborative Group. (1998). The falling mortality from coronary heart disease: a clinic-pathological CO RR EC TE Martindale, J. J., Wall, J. A., Martinez-Longoria, D. M., Aryal, P., Rockman, H. A., Guo, Y., et al. (2005). Overexpression of mitogen-activated protein kinase kinase 6 in the heart improves functional recovery from ischemia in vitro and protects against myocardial infarction in vivo. J Biol Chem 280(1), 669−676. Matsuyama, W., Faure, M., & Yoshimura, T. (2003). Activation of discoidin domain receptor 1 facilitates the maturation of human monocyte-derived dendritic cells through the TNF receptor associated factor 6/TGF-betaactivated protein kinase 1 binding protein 1 beta/p38 alpha mitogenactivated protein kinase signaling cascade. J Immunol 171(7), 3520−3532. Meldrum, D. R., Dinarello, C. A., Cleveland, J. C., Jr., Cain, B. S., Shames, B. D., Meng, X., et al. (1998). Hydrogen peroxide induces tumor necrosis factor alpha-mediated cardiac injury by a P38 mitogen-activated protein kinasedependent mechanism. Surgery 124(2), 291−296. Mittelstadt, P. R., Salvador, J. M., Fornace, A. J., Jr., & Ashwell, J. D. (2005). Activating p38 MAPK: new tricks for an old kinase. Cell Cycle 4(9), 1189−1192. Moriguchi, T., Kuroyanagi, N., Yamaguchi, K., Gotoh, Y., Irie, K., Kano, T., et al. (1996). A novel kinase cascade mediated by mitogen-activated protein kinase kinase 6 and MKK3. J Biol Chem 271(23), 13675−13679. Nagarkatti, D. S., & Afi, R. I. (1998). Role of p38 MAP kinase in myocardial stress. J Mol Cell Cardiol 30(8), 1651−1664. Office for National Statistics (2000). Key Health Statistics from General Practice. London: The Stationery Office. Ohkusu-Tsukada, K., Tominaga, N., Udono, H., & Yui, K. (2005). Regulation of the maintenance of peripheral T-cell anergy by TAB1-1 mediated p38alpha activation. Mol Cell Biol 25(19), 8763. Okada, T., Otani, H., Wu, Y., Kyoi, S., Enoki, C., Fujiwara, H., et al. (2005). Role of F-actin organization in p38 MAP kinase-mediated apoptosis and necrosis in neonatal rat cardiomyocytes subjected to simulated ischemia and reoxygenation. Am J Physiol Heart Circ Physiol 289(6), H2310−H2318. Ono, K., & Han, J. (2000). The p38 signal transduction pathway: activation and function. Cell Signal 12(1), 1−13. Otsu, K., Yamashita, N., Nishida, K., Hirotani, S., Yamaguchi, O., Watanabe, T., et al. (2003). Disruption of a single copy of the p38alpha MAP kinase gene leads to cardioprotection against ischemia–reperfusion. Biochem Biophys Res Commun 302(1), 56−60. Pargellis, C., Tong, L., Churchill, L., Cirillo, P. F., Gilmore, T., Graham, A. G., et al. (2002). Inhibition of p38 MAP kinase by utilizing a novel allosteric binding site. Nat Struct Biol 9(4), 268−272. Parker, C. G., Hunt, J., Diener, K., McGinley, M., Soriano, B., Keesler, G. A., et al. (1998). Identification of stathmin as a novel substrate for p38 delta. Biochem Biophys Res Commun 249(3), 791−796. Pearson, G., Robinson, F., Beers, G. T., Xu, B. E., Karandikar, M., Berman, K., et al. (2001). Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev 22(2), 153−183. Petrich, B. G., & Wang, Y. (2004). Stress-activated MAP kinases in cardiac remodeling and heart failure: new insights from transgenic studies. Trends Cardiovasc Med 14(2), 50−55. Pfeffer, M. A., & Braunwald, E. (1990). Ventricular remodeling after myocardial infarction. Experimental observations and clinical implications. Circulation 81(4), 1161−1172. Ping, P., & Murphy, E. (2000). Role of p38 mitogen-activated protein kinases in preconditioning: a detrimental factor or a protective kinase? Circ Res 86(9), 921−922. Raingeaud, J., Gupta, S., Rogers, J. S., Dickens, M., Han, J., Ulevitch, R. J., et al. (1995). Pro-inflammatory cytokines and environmental stress cause p38 mitogen-activated protein kinase activation by dual phosphorylation on tyrosine and threonine. J Biol Chem 270(13), 7420−7426. Rakhit, R. D., Kabir, A. N. M., Mockridge, J. W., Saurin, A., & Marber, M. S. (2001). Role of G proteins and modulation of p38 MAPK activation in the protection by nitric oxide against ischemia-reoxygenation injury. Biochem Biophys Res Commun 286(5), 995−1002. Ren, J., Zhang, S., Kovacs, A., Wang, Y., & Muslin, A. J. (2005). Role of p38alpha MAPK in cardiac apoptosis and remodeling after myocardial infarction. J Mol Cell Cardiol 38(4), 617−623. Riad, A., Unger, D., Du, J., Westermann, D., Mohr, Z., Sobirey, M., et al. (2007). Chronic inhibition of p38MAPK improves cardiac and endothelial function in experimental diabetes mellitus. Eur J Pharmacol 554(1), 40−45. UN 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 ARTICLE IN PRESS J.E. Clark et al. / Pharmacology & Therapeutics xx (2007) xxx–xxx RO OF Wood, C. C., Yong, C. L., Madwed, J. B., & Gupta, A. (2002). Pharmacokinetics and Pharmacodynamics of an Oral p38 MAP Kinase Inhibitor (BIRB 768 BS), Administered once Daily for 7 Days (pp. A993). Xie, Y., Chen, C., Stevenson, M. A., Auron, P. E., & Calderwood, S. K. (2002). Heat shock factor 1 represses transcription of the IL-1beta gene through physical interaction with the nuclear factor of interleukin 6. J Biol Chem 277(14), 11802−11810. Yada, M., Shimamoto, A., Hampton, C. R., Chong, A. J., Takayama, H., Rothnie, C. L., et al. (2004). FR167653 diminishes infarct size in a murine model of myocardial ischemia–reperfusion injury. J Thorac Cardiovasc Surg 128(4), 588−594. Yamaguchi, K., Shirakabe, K., Shibuya, H., Irie, K., Oishi, I., Ueno, N., et al. (1995). Identification of a member of the MAPKKK family as a potential mediator of TGF-beta signal transduction. Science 270(5244), 2008−2011. Young, P. R., McLaughlin, M. M., Kumar, S., Kassis, S., Doyle, M. L., McNulty, D., et al. (1997). Pyridinyl imidazole inhibitors of p38 mitogen-activated protein kinase bind in the ATP site. J Biol Chem 272(18), 12116−12121. Yue, T. L., Wang, C., Gu, J. L., Ma, X. L., Kumar, S., Lee, J. C., et al. (2000). Inhibition of extracellular signal-regulated kinase enhances ischemia/ reoxygenation-induced apoptosis in cultured cardiac myocytes and exaggerates reperfusion injury in isolated perfused heart. Circ Res 86(6), 692−699. Zhang, S., Weinheimer, C., Courtois, M., Kovacs, A., Zhang, C. E., Cheng, A. M., et al. (2003). The role of the Grb2-p38 MAPK signaling pathway in cardiac hypertrophy and fibrosis. J Clin Invest 111(6), 833−841. Zheng, S., & Zuo, Z. (2004). Isoflurane preconditioning induces neuroprotection against ischemia via activation of P38 mitogen-activated protein kinases. Mol Pharmacol 65(5), 1172−1180. DP perspective. Heart 80, 121−126 BHF Coronary heart disease statistics at www.heartstats.org Udelson, J. E., Patten, R. D., & Konstam, M. A. (2003). New concepts in postinfarction ventricular remodeling. Rev Cardiovasc Med 4(Suppl 3), S3−S12. Vahebi, S., Ota, A., Li, M., Warren, C. M., de Tombe, P. P., Wang, Y., et al. (2007). p38-MAPK induced dephosphorylation of alpha-tropomyosin is associated with depression of myocardial sarcomeric tension and ATPase activity. Circ Res 100(3), 408−415. Wang, R., Wang, Z., & Wu, L. (1997). Carbon monoxide-induced vasorelaxation and the underlying mechanisms. Br J Pharmacol 121(5), 927−934. Wang, Y., Huang, S., Sah, V. P., Ross, J., Jr., Brown, J. H., Han, J., et al. (1998). Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. J Biol Chem 273(4), 2161−2168. Wang, Z., Canagarajah, B. J., Boehm, J. C., Kassisa, S., Cobb, M. H., Young, P. R., et al. (1998). Structural basis of inhibitor selectivity in MAP kinases. Structure 6(9), 1117−1128. Wang, M., Tsai, B. M., Turrentine, M. W., Mahomed, Y., Brown, J. W., & Meldrum, D. R. (2005). p38 mitogen activated protein kinase mediates both death signaling and functional depression in the heart. Ann Thorac Surg 80(6), 2235−2241. Weinbrenner, C., Liu, G. S., Cohen, M. V., & Downey, J. M. (1997). Phosphorylation of tyrosine 182 of p38 mitogen-activated protein kinase correlates with the protection of preconditioning in the rabbit heart. J Mol Cell Cardiol 29(9), 2383−2391. Weisman, M. (2002). Double-blind, placebo-controlled trial of VX-745, an oral p38 mitogen activated protein kinase inhibitor, in patients with rheumatoid arthritis (RA). Ann. European Congress Rheumatol. Wilson, K. P., Fitzgibbon, M. J., Caron, P. R., Griffith, J. P., Chen, W., McCaffrey, P. G., et al. (1996). Crystal structure of p38 mitogen-activated protein kinase. J Biol Chem 271(44), 27696−27700. TE 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 15 UN CO RR EC 1083 Please cite this article as: Clark, J. E. et al. Potential of p38-MAPK inhibitors in the treatment of ischaemic heart disease. Pharmacol Ther (2007), doi:10.1016/j. pharmthera.2007.06.013 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082