13.07.2015 Views

PTI-609: A Novel Analgesic that Binds Filamin A to Control Opioid ...

PTI-609: A Novel Analgesic that Binds Filamin A to Control Opioid ...

PTI-609: A Novel Analgesic that Binds Filamin A to Control Opioid ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>PTI</strong>-<strong>609</strong>, <strong>Novel</strong> <strong>Analgesic</strong> Recent Patents on CNS Drug Discovery, 2010, Vol. 5, No. 3 219xone, <strong>PTI</strong>-<strong>609</strong> avoids the second, lower-affinity binding siteon FLNA and therefore should maintain its beneficial effectswith increasing dose, allowing the picomolar site <strong>to</strong> saturatewith no interference. Although much work remains before<strong>PTI</strong>-<strong>609</strong> can enter clinical trials, the early research presentedhere shows <strong>PTI</strong>-<strong>609</strong> and related compounds <strong>to</strong> be efficaciousanalgesics <strong>that</strong> possess additional anti-inflamma<strong>to</strong>ry propertiesand, according <strong>to</strong> the standard conditioned placepreference paradigm, may not carry the abuse and addictivepotential of classical opioid drugs. The problems withcurrent opioid painkillers, from side effects <strong>to</strong> <strong>to</strong>lerance,dependence, abuse, diversion and addiction demonstrate agreat need for a better class of efficacious analgesics. <strong>PTI</strong>-<strong>609</strong> is a promising new drug candidate <strong>that</strong> seems wellpoised <strong>to</strong> fulfill <strong>that</strong> need.ACKNOWLEGEMENTSPain Therapeutics, Inc. acknowledges the followingcontracted services: pharmacophore modeling and in silicoscreening (Jian Wang); analgesia and conditioned placepreference testing (Ed Bilsky of AIV Testing, Biddeford,ME); medicinal chemistry, synthesis and CIA model (Medicilon,Inc., Shanghai, China). All in vitro screening for leadselection was performed by H.-Y. Wang at CUNY MedicalSchool under a research collaboration.CONFLICT OF INTERESTL.H. Burns is an employee of Pain Therapeutics Inc., thecompany <strong>that</strong> funded the development work on <strong>PTI</strong>-<strong>609</strong> aswell as earlier work on Oxytrex.REFERENCES[1] Crain SM, Shen K-F. Antagonists of excita<strong>to</strong>ry opioid recep<strong>to</strong>rfunctions enhance morphine's analgesic potency and attenuateopioid <strong>to</strong>lerance/dependence liability. Pain 2000; 84: 121-31.[2] Crain SM, Shen K-F. Ultra-low concentrations of naloxoneselectively antagonize excita<strong>to</strong>ry effects of morphine on sensoryneurons, thereby increasing its antinociceptive potency andattenuating <strong>to</strong>lerance/dependence during chronic cotreatment. ProcNatl Acad Sci USA 1995; 92: 10540-4.[3] Powell KJ, Abul-Husn NS, Jhamandas A, Olmstead MC, BeningerRJ, Jhamandas K. Paradoxical effects of the opioid antagonistnaltrexone on morphine analgesia, <strong>to</strong>lerance, and reward in rats. JPharmacol Exp Ther 2002; 300: 588-96.[4] Wang H-Y, Friedman E, Olmstead MC, Burns LH. Ultra-low-dosenaloxone suppresses opioid <strong>to</strong>lerance, dependence and associatedchanges in Mu opioid recep<strong>to</strong>r-G protein coupling and Gsignaling. Neuroscience 2005; 135: 247-61.[5] Crain SM, Shen K-F. Acute thermal hyperalgesia elicited by lowdosemorphine in normal mice is blocked by ultra-low-dosenaltrexone, unmasking potent opioid analgesia. Brain Res 2001;888: 75-82.[6] Olmstead MC, Burns LH. Ultra-low-dose naltrexone suppressesrewarding effects of opiates and aversive effects of opiatewithdrawal in rats. Psychopharmacol 2005; 181: 576-81.[7] Leri F, Burns LH. Ultra-low-dose naltrexone reduces the rewardingpotency of oxycodone and relapse vulnerability in rats. PharmacolBiochem Behav 2005; 82: 252-62.[8] Shen KF, Crain SM. Dual opioid modulation of the action potentialduration of mouse dorsal root ganglion neurons in culture. BrainRes 1989; 491: 227-42.[9] Crain SM, Shen K-F. <strong>Opioid</strong>s can evoke direct recep<strong>to</strong>r-mediatedexcita<strong>to</strong>ry effects on sensory neurons. Trends Pharmacol Sci 1990;11: 77-81.[10] Chakrabarti S, Regec A, Gintzler AR. Biochemical demonstrationof mu-opioid recep<strong>to</strong>r association with Gs: Enhancementfollowing morphine exposure. Mol Brain Res 2005; 135: 217-24.[11] Laugwitz KL, Offermanns S, Spicher K, Schultz G. Mu and deltaopioid recep<strong>to</strong>rs differentially couple <strong>to</strong> G protein subtypes inmembranes of human neuroblas<strong>to</strong>ma SH-SY5Y cells. Neuron 1993;10: 233-42.[12] Gintzler AR, Chakrabarti S. <strong>Opioid</strong> <strong>to</strong>lerance and the emergence ofnew opioid recep<strong>to</strong>r-coupled signaling. Mol Neurobiol 2001; 21:21-33.[13] Wang H-Y, Burns LH. G <strong>that</strong> interacts with adenylyl cyclase inopioid <strong>to</strong>lerance originates from a Gs protein. J Neurbiol 2006; 66:1302-10.[14] Wang H-Y, Frankfurt M, Burns LH. High-affinity naloxone binding<strong>to</strong> filamin A prevents mu opioid recep<strong>to</strong>r - Gs coupling underlyingopioid <strong>to</strong>lerance and dependence. PLoS One 2008; 3(2): e1554.[15] Feng Y, Walsh C. The many faces of filamin: A versatile molecularscaffold for cell motility and signalling. Nat Cell Biol 2004; 6:1034-8.[16] S<strong>to</strong>ssel T, Condeelis J, Cooley L, Hartwig J, Noegel A, SchleicherM, et al. <strong>Filamin</strong>s as integra<strong>to</strong>rs of cell mechanics and signalling.Nature 2001; 2: 138-45.[17] Onoprishvili I, Andria M, Kramer H, Ancevska-Taneva N, Hiller J,Simon E. Interaction between the μ opioid recep<strong>to</strong>r and fliamin A isinvolved in recep<strong>to</strong>r regulation and trafficking. Mol Pharmacol2003; 64: 1092-100.[18] Burns LH. Ultra-low-dose opioid antagonists enhance opioidanalgesia while reducing <strong>to</strong>lerance, dependence and addictiveproperties. In: Capasso A, Ed. Recent Developments in PainResearch. Kerala, India: Research Signpost 2005; 115-36.[19] Shen K-F, Crain SM, Moate P, Bos<strong>to</strong>n R, de Kater AW,Schoenhard GL. <strong>PTI</strong>-555, reverses and prevents morphine-induced<strong>to</strong>lerance and naloxone-precipitated withdrawal in mice chronicallytreated with morphine. J Pain 2002; 3: 50.[20] Shen K-F, Crain SM, Moate P, Bos<strong>to</strong>n R, de Kater AW,Schoenhard GL. <strong>PTI</strong>-801, a novel formulation of oxycodone, showsabsence of <strong>to</strong>lerance, physical dependence and naloxoneprecipitatedwithdrawal effects in mice. J Pain 2002; 3: 49.[21] Kest B, Sar<strong>to</strong>n E, Dahan A. Gender differences in opioid-mediatedanalgesia. Anesthesiology 2000; 93: 539-47.[22] Terner JM, Barrett AC, Lomas LM, Negus SS, Picker MJ.Influence of low doses of naltrexone on morphine antinociceptionand morphine <strong>to</strong>lerance in male and female rats of four strains. Pain2006; 122: 90-101.[23] Crain SM, Shen K-F. Enhanced analgesic potency and reduced<strong>to</strong>lerance of morphine in 129/SvEv mice: evidence for a deficiencyin GM1 ganglioside-regulated excita<strong>to</strong>ry opioid recep<strong>to</strong>r functions.Brain Res 2000; 856: 227-35.[24] Hamman SR, Malik H, Sloan JW, Wala EP. Interactions of "ultralow"doses of naltrexone and morphine in mature and young maleand female rats. Recept Chan 2004; 10: 73-81.[25] Largent-Milnes TM, Guo W, Wang H-Y, Burns LH, Vanderah T.Oxycodone plus ultra-low-dose naltrexone attenuates neuropathicpain and associated μ opioid recep<strong>to</strong>r - Gs coupling. J Pain 2008; 9:700-13.[26] Gan TJ, Ginsberg B, Glass PSA, Fortney J, Jhaveri R, Perno R.<strong>Opioid</strong>-sparing effects of a low-dose infusion of naloxone inpatient-administered morphine sulfate. Anesthesiology 1997; 87:1075-81.[27] Cruciani RA, Lussier D, Miller-Saultz D, Arbuck DM. Ultra-lowdose oral naltrexone decreases side effects and potentiates the effec<strong>to</strong>f methadone. J Pain Symp<strong>to</strong>m Manage 2003; 25: 491-4.[28] Cepeda MS, Africano JM, Manrique AM, Fragoso W, Carr DB.The combination of low dose naloxone and morphine in PCA doesnot decrease opioid requirements in the pos<strong>to</strong>perative period. Pain2002; 96: 73-9.[29] Cepeda MS, Alvarez H, Morales O, Carr DB. Addition of ultralowdose naloxone <strong>to</strong> pos<strong>to</strong>perative morphine PCA: Unchangedanalgesia and opioid requirement but decreased incidence of opioidside effects. Pain 2004; 107: 41-6.[30] Joshi GP, Duffy L, Chehade J, Wesevich J, Gajraj N, Johnson ER.Effects of prophylactic nalmefene on the incidence of morphinerelatedside effects in patients receiving intravenous patientcontrolledanalgesia. Anesthesiology 1999; 90: 1007-11.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!