Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHODS FOR TREATING NEUROLOGICAL CONDITIONS
Document Type and Number:
WIPO Patent Application WO/2011/159945
Kind Code:
A2
Abstract:
Provided herein are PAK inhibitors. Also provided herein are compositions and methods for treating an individual suffering from certain neurological conditions.

Inventors:
LICHTER JAY (US)
CAMPBELL DAVID (US)
DURON SERGIO G (US)
VOLLRATH BENEDIKT (US)
Application Number:
PCT/US2011/040769
Publication Date:
December 22, 2011
Filing Date:
June 16, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AFRAXIS INC (US)
LICHTER JAY (US)
CAMPBELL DAVID (US)
DURON SERGIO G (US)
VOLLRATH BENEDIKT (US)
International Classes:
A61K31/519; A61K31/38; A61K31/426; A61K31/505; A61P25/16; A61P25/18
Other References:
See references of EP 2582374A2
Attorney, Agent or Firm:
HOSTETLER, Michael J. et al. (650 Page Mill RoadPalo Alto, CA, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method of reversing, partially reversing, or delaying choreia associated with Huntington's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

2. A method of reversing, partially reversing or delaying onset of cognitive or memory deficits associated with Huntington's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

3. A method of treating substance abuse and/or substance addiction comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

4. The method of claim 3, wherein administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual in need thereof reverses or partially reverses neuro adaptation of the brain associated with substance abuse and/or substance addiction.

5. A method of treating Parkinson's disease comprising administration of a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

6. A method of reversing, partially reversing, or delaying symptoms of tremor, rigidity, bradykinesia and/or postural instability associated with Parkinson's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

7. A method of reversing, partially reversing or delaying onset of cognitive or memory deficits associated with Parkinson's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

8. A method of treating neurofibromatosis comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

9. The method of claim 8, wherein administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual in need thereof reduces the occurence of seizures associated with neurofibromatosis.

10. The method of claim 8, wherein administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual in need thereof reverses or partially reverses learning disability associated with neurofibromatosis.

11. The method of claim 8, wherein administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual in need thereof reduces the defects in language skills and/or cognitive performance associated with neurofibromatosis.

12. The method of claim 8, wherein the neurofibromatosis is type I or type II.

13. A method of treating clinical depression comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

14. A method of treating bipolar disorder comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

15. A method of treating anxiety and/or anxiety disorder comprising administering a

therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

16. A method of treating posttraumatic stress disorder (PTSD) comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

17. The method of any one of claims 1-16, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine morphology and/or synaptic function.

18. The method of claim 17, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine density.

19. The method of claim 17 or claim 18, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine length.

20. The method of any one of claims 17-19, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine neck diameter.

21. The method of any one of claims 17-20, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine shape.

22. The method of any one of claims 17-21, wherein the p21 -activated kinase (PAK) inhibitor increases the number of mushroom-shaped dendritic spines.

23. The method of any one of claims 17-22, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine head volume.

24. The method of any one of claims 17-22, wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine head diameter.

25. The method of claim 17, wherein the p21 -activated kinase (PAK) inhibitor modulates the ratio of the number of mature spines to the number of immature spines.

26. The method of claim 17, wherein the p21 -activated kinase (PAK) inhibitor modulates the ratio of the spine head volume to spine length.

27. The method of claim 17, wherein the p21 -activated kinase (PAK) inhibitor modulates synaptic function.

28. The method of any one of claims 1-27, wherein the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant baseline synaptic transmission associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD.

29. The method of any one of claims 1-27, wherein the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant synaptic plasticity associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD.

30. The method of any one of claims 1-27, wherein the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant long term depression (LTD) associated with

Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD.

31. The method of any one of claims 1-27, wherein the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant long term potentiation (LTP) associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD.

32. The method of claim 8, wherein administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual suffering from, suspected to be suffering from or pre-disposed to neurofibromatosis reverses, or reduces the incidence and/or severity of neurofibromas and/or optic nerve gliomas associated with neurofibromatosis.

33. The method of claim 8 wherein administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual suffering from, suspected to be suffering from or pre-disposed to neurofibromatosis reverse, or reduces the incidence and/or severity of hearing loss, paralysis, brain or spinal tumors, cataracts or retinal abnormalities associated with neurofibromatosis.

34. The method of any one of claims 1-33, wherein a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor causes substantially complete inhibition of one or more p21- activated kinases.

35. The method of any one of claims 1-33, wherein a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor causes partial inhibition of one or more p21 -activated kinases.

36. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits one or more of PAK1, PAK2, PAK3, PAK4, PAK5 or PAK6.

37. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor is a Group I PAK inhibitor.

38. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits one or more of PAK1, PAK2 or PAK3.

39. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits PAK1 and PAK3.

40. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits PAK1 and PAK2.

41. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits PAK2 and PAK3.

42. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits PAK1.

43. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits PAK2.

44. The method of any one of claims 1-33, wherein the p21-activated kinase (PAK) inhibitor inhibits PAK3.

45. The method of any one of claims 1-44, further comprising administration of a second therapeutic agent.

46. The method of claim 45, wherein the second therapeutic agent is an acetylcholinestrase inhibitor, an alpha7 nicotinic receptor agonist, an antioxidant, memantine or minocycline.

47. The method of any one of claims 1-46, wherein administration of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof improves, stabilizes, or lessens the deterioration of scores on the Mini-Mental State Exam (MMSE), HAM-D test, Unified Huntington's Disease Rating Scales (UHDRS) test, Wechsler Intelligence Scale, Wechsler Memory Scale, Dementia Rating Scale (DRS), Boston Naming Test, Stroop Color Word Test, Trail Making Test or Auditory Verbal Learning Test (AVLT) scale for the individual.

Description:
METHODS FOR TREATING NEUROLOGICAL CONDITIONS

CROSS-REFERENCE

[0001] This application claims the benefit of U.S. Provisional Application No. 61/355,278, filed

June 16, 2010, which is incorporated herein by reference in its entirety. BACKGROUND OF THE INVENTION

[0002] Neurological conditions are characterized by a variety of debilitating symptoms including cognitive impairment, seizures, or mood disorders.

SUMMARY OF THE INVENTION

[0003] Described herein are methods of treatment of neurological conditions including, and not limited to, epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, neurofibromatosis, tuberous sclerosis, bipolar disorder, Posttraumatic Stress Disorder, anxiety disorder and/or Down's syndrome that alleviate, halt or delay the progression of some or all symptoms of these conditions. In some embodiments, the methods of treatment described herein comprise administration of one or more PA inhibitors to an individual in need thereof.

[0004] In some instances, neurological conditions are associated with altered neuronal circuitry. In some instances, neurological conditions are associated with altered and/or defective dendritic spines. In some instances, p21 activated kinases modulate dendritic spine morphogenesis thereby modulating neural circuitry. In some instances, aberrant spine morphogenesis (e.g., abnormal spine density, length, thickness, shape or the like) is associated with pathogenesis of neurological conditions. In some instances, administration of a PAK inhibitor to individuals diagnosed with or suspected of having neurological conditions reduces, stabilizes or reverses abnormalities in dendritic spine morphology, density, and/or synaptic function, including but not limited to abnormal spine density, spine size, spine shape, spine plasticity, spine motility or the like.

[0005] In one aspect is a method of reversing, partially reversing, or delaying choreia associated with Huntington's disease comprising administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual in need thereof.

[0006] In another aspect is a method of reversing, partially reversing or delaying onset of cognitive or memory deficits associated with Huntington's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof. [0007] In a further aspect is a method of treating substance abuse and/or substance addiction comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0008] In one embodiment is a method of treating substance abuse and/or substance addiction wherein administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof reverses or partially reverses neuro adaptation of the brain associated with substance abuse and/or substance addiction.

[0009] In yet a further aspect is a method of treating Parkinson's disease comprising

administration of a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0010] In another aspect is a method of reversing, partially reversing, or delaying symptoms of tremor, rigidity, bradykinesia and/or postural instability associated with Parkinson's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0011] In one embodiment is a method of reversing, partially reversing or delaying onset of cognitive or memory deficits associated with Parkinson's disease comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0012] Also described herein is a method of treating neurofibromatosis comprising

administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof. In one embodiment, is a method of treating neurofibromatosis wherein administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof reduces the occurence of seizures associated with neurofibromatosis. In another embodiment, administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof reverses or partially reverses learning disability associated with neurofibromatosis. In a further embodiment, administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof reduces the defects in language skills and/or cognitive performance associated with neurofibromatosis. In yet a further embodiment, the neurofibromatosis is type I or type II.

[0013] In one aspect is a method of treating clinical depression comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof. [0014] In another aspect is a method of treating bipolar disorder comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0015] In another embodiment, is a method of treating anxiety and/or anxiety disorder comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0016] Also presented herein is a method of treating posttraumatic stress disorder (PTSD) comprising administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0017] Further provided herein is a method of reducing or halting incidence of abnormal neuronal activity in the brain associated with epilepsy comprising administering a

therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof.

[0018] In some embodiments, the epilepsy is localization related epilepsy, generalized epilepsy or epilepsy of unknown localization. In some embodiments, the epilepsy is idiopathic epilepsy, symptomatic epilepsy or cryptogenic epilepsy, or epilepsy of unknown origin.

[0019] In one embodiment, are methods described herein wherein the p21 -activated kinase (PAK) inhibitor modulates dendritic spine morphology and/or synaptic function. In another embodiment, the p21 -activated kinase (PAK) inhibitor modulates dendritic spine density. In a further embodiment, the p21 -activated kinase (PAK) inhibitor modulates dendritic spine length. In yet a further embodiment, the p21 -activated kinase (PAK) inhibitor modulates dendritic spine neck diameter. In another embodiment, the p21 -activated kinase (PAK) inhibitor modulates dendritic spine shape. In yet another embodiment, the p21 -activated kinase (PAK) inhibitor increases the number of mushroom-shaped dendritic spines. In one embodiment, the p21- activated kinase (PAK) inhibitor modulates dendritic spine head volume. In another

embodiment, the p21 -activated kinase (PAK) inhibitor modulates dendritic spine head diameter. In a further embodiment, the p21 -activated kinase (PAK) inhibitor modulates the ratio of the number of mature spines to the number of immature spines. In yet a further embodiment, the p21 -activated kinase (PAK) inhibitor modulates the ratio of the spine head volume to spine length. In one embodiment, the p21 -activated kinase (PAK) inhibitor modulates synaptic function. In another embodiment, the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant baseline synaptic transmission associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD. In yet another embodiment, the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant synaptic plasticity associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD. In a further embodiment, the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant long term depression (LTD) associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD. In yet a further embodiment, the p21 -activated kinase (PAK) inhibitor normalizes or partially normalizes aberrant long term potentiation (LTP) associated with Huntington's disease, substance abuse and/or substance dependency, Parkinson's disease, clinical depression, neurofibromatosis, bipolar disorder, anxiety syndrome, and PTSD.

[0020] In one embodiment, administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual suffering from, suspected to be suffering from or predisposed to neurofibromatosis reverses, or reduces the incidence and/or severity of

neurofibromas and/or optic nerve gliomas associated with neurofibromatosis.

[0021] In another embodiment, administering a therapeutically effective amount of a p21- activated kinase (PAK) inhibitor to an individual suffering from, suspected to be suffering from or pre-disposed to neurofibromatosis reverse, or reduces the incidence and/or severity of hearing loss, paralysis, brain or spinal tumors, cataracts or retinal abnormalities associated with neurofibromatosis. In yet another embodiment, administering a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor to an individual suffering from, suspected to be suffering from or pre-disposed to neurofibromatosis reverse, or reduces the incidence and/or severity of language disabilities, defects in visual-spatial skills or cognitive performance assocated with Neurofibromatosis. In yet another embodiment, a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor causes substantially complete inhibition of one or more p21 -activated kinases. In a further embodiment, a therapeutically effective amount of a p21 -activated kinase (PAK) inhibitor causes partial inhibition of one or more p21 -activated kinases.

[0022] In yet a further embodiment, the p21 -activated kinase (PAK) inhibitor inhibits one or more of PAK1, PAK2, PAK3, PAK4, PAK5 or PAK6. In one embodiment, the p21-activated kinase (PAK) inhibitor is a Group I PAK inhibitor. In another embodiment, the p21 -activated kinase (PAK) inhibitor inhibits one or more of PAK1, PAK2 or PAK3. In yet another embodiment, the p21 -activated kinase (PAK) inhibitor inhibits PAK1 and PAK3. In a further embodiment, the p21 -activated kinase (PAK) inhibitor inhibits PAK1 and PAK2. In yet a further embodiment, the p21 -activated kinase (PAK) inhibitor inhibits PAK2 and PAK3. In another embodiment, the p21 -activated kinase (PAK) inhibitor inhibits PAK1. In one embodiment, the p21 -activated kinase (PAK) inhibitor inhibits PAK2. In yet another embodiment, the p21 -activated kinase (PAK) inhibitor inhibits PAK3.

[0023] In one embodiment are the methods described herein further comprising administration of a second therapeutic agent. In a further embodiment, the second therapeutic agent is an acetylcholinestrase inhibitor, an alpha7 nicotinic receptor agonist, an antioxidant, memantine or minocycline.

[0024] In one embodiment, administration of a p21 -activated kinase (PAK) inhibitor to an individual in need thereof improves, stabilizes, or lessens the deterioration of scores on the Mini-Mental State Exam (MMSE), HAM-D test, Unified Huntington's Disease Rating Scales (UHDRS) test, Wechsler Intelligence Scale, Wechsler Memory Scale, Dementia Rating Scale (DRS), Boston Naming Test, Stroop Color Word Test, Trail Making Test or Auditory Verbal Learning Test (AVLT) scale for the individual.

BRIEF DESCRIPTION OF THE DRAWINGS

[0025] The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative

embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:

[0026] FIG. 1 describes illustrative shapes of dendritic spines.

[0027] FIG. 2 describes modulation of dendritic spine head diameter by a small molecule PAK inhibitor.

[0028] FIG. 3 describes modulation of dendritic spine length by a small molecule PAK inhibitor.

DETAILED DESCRIPTION OF THE INVENTION

[0029] Provided herein are methods for treatment of neurological conditions comprising administration of PAK inhibitors described herein. Examples of neurological conditions include and are not limited to epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, neurofibromatosis, tuberous sclerosis, bipolar disorder, Posttraumatic Stress Disorder, anxiety disorder, mania and/or Down's syndrome. In some instances, PAK inhibitors described herein improve cognition and/or memory deficits associated with a neurological condition, thereby improving overall quality of life and/or life expectancy of individuals suffering from neurological conditions. [0030] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) alleviate, halt or slow down progressive degeneration of neural tissue.

[0031] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) alleviate, halt or delay progressive atrophy of nervous tissue in the brain.

[0032] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) alleviate synaptic dysfunction in neurofibromatosis. [0033] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) reverse defects in synaptic function and/or plasticity in a patient diagnosed with a neurological condition such as, for example,clinical depression.

[0034] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) reverse neuroadapatation caused by an addiction (e.g., alcohol and/or drug abuse).

[0035] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) modulate dendritic spine length and/or spine head diameter. [0036] In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) modulate dendritic spine length and/or spine head diameter, thereby reversing or alleviating memory, vocabulary and/or cognitive deficits in individuals suffering from neurological conditions such as for example, Down's syndrome.

[0037] In some instances, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) reverse or reduce the incidence of abnormal neuronal activity in the brain thereby reducing or halting ocurrence of seizures in individuals suffering from neurological conditions such as, for example, epilepsy.

[0038] In some embodiments, the PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) alleviate, stabilize or reverse symptoms of neurological conditions in an individual that is non-responsive to other putative therapies for the neurological conditions (e.g., treatment with serotonin receptor antagonists for the treatment of clinical depression, or the like).

[0039] In some embodiments, treatment of a neurological conditions with (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) reduces side-effects compared to risk of side-effects with other putative therapies (e.g., treatment with PAK inhibitors reduces risk of suicide compared to treatment with other antidepressants).

[0040] In some instances, neurological conditions are associated with abnormal dendritic spine morphology, dendritic spine size, dendritic spine plasticity, dendritic spine motility, dendritic spine density and/or abnormal synaptic function. In some instances, PAK activation is implicated in defective spine morphogenesis, maturation, and maintenance. Described herein are methods for suppressing or reducing PAK activity by administering a PAK inhibitor (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) for rescue of defects in spine morphology, size, plasticity spine motility and/or density associated with neurological conditions as described herein. Neurological conditions

Epilepsy

[0041] Epilepsy is a brain disorder associated with episodes of abnormal neuronal activity in the brain. In some instances, the sporadic episodes of abnormal neuronal activity in the brain are manifested as unprovoked seizures. In some instances, brain function is altered due to abnormal neuronal activity and causes changes in attention or behavior. People with epilepsy often experience long-term cognitive dysfunction and other neurological deficits, including memory loss, learning disabilities and neurobehavioral disorders, which may exhibit a progressive course correlating with worsening seizure control.

[0042] In some instances, epilepsy arises from a small focused portion of the brain (localization related epilepsy). In other instances, epilepsy arises from neuronal circuits in the whole brain (generalized epilepsy). In other instances, epilepsy is of unknown localization. In some instances, epilepsy arises from genetic abnormalities (idiopathic epilepsy). In some instances, injury to the brain (e.g., a lesion, a tumor) leads to symptomatic epilepsy. In some instances, a lesion that is otherwise difficult to detect causes epilepsy (cryptogenic epilepsy).

[0043] As used herein, "epilepsy" includes epilepsy characterized by Absence seizures, atonic seizures, benign Rolandic epilepsy, childhood absence seizures, clonic seizures, complex partial seizures, frontal lobe epilepsy, Febrile seizures, Infantile spasms, Juvenile Myoclonic Epilepsy, Juvenile Absence Epilepsy, lennox-gastaut syndrom, Landau-Kleff er Syndrome , myoclonic seizures, Progressive Myoclonic Epilepsies, Psychogenic Seizures , Reflex Epilepsy,

Rasmussen's Syndrome, Simple Partial seizures, Secondarily Generalized Seizures, Temporal Lobe Epilepsy, Toni-clonic seizures, Tonic seizures, Psychomotor Seizures, Limbic Epilepsy, Partial-Onset Seizures, generalised-onset seizures, Status Epilepticus, Abdominal Epilepsy, Akinetic Seizures, Auto-nomic seizures, Massive Bilateral Myoclonus, Catamenial Epilepsy, Drop seizures, Emotional seizures, Focal seizures, Gelastic seizures, Jacksonian March, Lafora Disease, Motor seizures, Multifocal seizures, Neonatal seizures, Nocturnal seizures,

Photosensitive seizure, Pseudo seizures, Sensory seizures, Subtle seizures, Sylvan Seizures, Withdrawal seizures, Visual Reflex Seizures or the like.

[0044] In some instances, epilepsy patients have seizures that are intractable to treatments. Accordingly, provided herein are therapies for seizures and the neurological comorbidities of epilepsy.

[0045] In some instances, abnormal spurts of neuronal activity in the brain causes cellular changes in brain cells that contribute to epilepsy. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function contributes to the manifestation of overt symptoms of epilepsy (e.g., seizures or convulsions). In some instances, an abnormal dendritic spine density and/or dendritic morphology and/or synaptic function is associated with activation of p21 -activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function associated with epilepsy. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, or reduces abnormal neuronal activity in the brain thereby reversing or reducing occurrence of epileptic seizures. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces the severity and/or frequency of seizures in an individual that is refractory to treatment with other putative epilepsy treatments. Accordingly, provided herein are methods of treating epilepsy comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Huntington's disease

[0046] Huntington's disease is a genetic disorder that affects muscle coordination and cognitive function. A part of the Huntington's gene is a trinucleotide repeat, which varies in length between individuals and changes length between generations. When the length of the trinucleotide repeat exceeds a normal range, the defective Huntington's gene codes for a mutant form of the huntingtin protein (mHTT). In some instances, mutant huntingtin protein is responsible for symptoms of Huntington's disease including and not limited to uncoordinated, jerky body movements (choreia), a decline in mental abilities and behavioral and psychiatric problems.

[0047] In some instances, mHTT activity causes damage to certain areas in the brain and contributes to Huntington's disease. In some instances, Huntington's disease is associated with an abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function and subsequent manifestation of overt symptoms of Huntington's disease (e.g., seizures or convulsions). In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21 -activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function associated with Huntington's disease. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, reduces, reverses, or partially reverses choreia associated with Huntington's disease. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, reduces, reverses, or partially reverses subcortical dementia associated with Huntington's disease.

Accordingly, provided herein are methods of treating Huntington's disease comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Parkinson's disease

[0048] Parkinson's disease is a neurodegenerative disorder that affects motor skills, speech and other bodily functions. Parkinson's disease also causes neurological disturbances, including cognition deficits, and mood and behavior problems.

[0049] In some instances, cognition deficits and/or mood and behavior problems in Parkinson's disease are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function and subsequent manifestation of overt neurological symptoms of Parkinson's disease (e.g., mild cognitive impairment, memory problems, executive dysfunction or the like). In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21 -activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function associated with Parkinson's disease. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, reduces, reverses, or partially reverses cognitive impairment associated with Parkinson's disease. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, reduces, reverses, or partially reverses memory problems associated with Parkinson's disease.

[0050] In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces parkinsonism. As used herein, parkinsonism includes movement disorders associated with Parkinson's disease such as tremor, rigidity, bradykinesia, postural instability, or akinesia. In some instances, parkinsonism is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21-activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces tremor, rigidity, bradykinesia and/or postural instability associated with Parkinson's disease. Accordingly, provided herein are methods of treating Parkinson's disease comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Substance abuse and substance dependence

[0051] Drug addiction is characterized by long-term changes in behavior, such as craving, dependence and dysphoria during withdrawal. In some instances, alcohol and drugs of abuse such as, for example, cocaine, morphine, amphetamine or heroin, impact the human brain and cause long term changes in the neural circuitry in the brain. In some instances, excess alcohol consumption and/or drug abuse over a prolonged period of time triggers abnormalities in the release of neurotransmitters in the brain (e.g., suppression of neurotransmitter release, excess release of neurotransmitters) and initiates long-lasting changes in neuronal cells in the brain. In some instances, Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) and/or Diffusion Tensor Imaging (DTI) are useful in mapping such changes in brain circuitry. In some instances, addiction over a prolonged period of time changes neuronal plasticity in the brain . In some instances, withdrawal from addiction is associated with reversing neuro adaptation and/or re-engineering neural circuitry in the brain.

[0052] In some instances, alcohol or substance abuse is associated with an abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function and subsequent behavioral changes associated with addiction. In some instances, the defects in dendritic spine morphology and/or dendritic spine density affects Medium Spiny Neurons (MSN) in the nucleus accumbens. In some instances, the defects in dendritic spine morphology and/or dendritic spine density affects neurons in the "reward circuitry" of the brain. In some instances, the defects in dendritic spine morphology and/or dendritic spine density affects pyramidal neurons in the prefrontal cortex and/or the hippocampus. In some instances, the defects in dendritic spine morphology and/or dendritic spine density affects neurons in the Ventral Tegmental Area (VTA). In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21 -activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function associated with substance abuse and/or substance addiction. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, reduces, reverses, or partially reverses the neuro adaptation in the brain caused by substance abuse and/or substance addiction. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby re-engineering neural circuitry. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) reverses or reduces the neuro adaptation in an addict that is refractory to treatment with other putative regimens to break the addiction. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) reverses or reduces craving for the substance of abuse or alcohol. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) reverses or reduces symptoms associated with withdrawal from alcohol or substances of abuse, such as dysphoria.

Accordingly, provided herein are methods of treating addiction comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Clinical Depression

[0053] According to the Diagnostic and Statistical Manual of Mental Disorders (DSM-IV) (1994) classification, Clincal Depression or major depressive disorder is characterized by an all- encompassing low mood, low self-esteem, and loss of interest or pleasure in normally enjoyable activities. In some instances, altered neurotransmission contributes to clinical depression. In some instances, altered neurotransmission is associated with impairment of neuronal plasticity and/or neuronal resilience.

[0054] In some instances, clinical depression is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function and subsequent behavioral changes. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21-activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function associated with depression. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) inhibits, reduces, reverses, or partially reverses the changes in neuroplasticity in the brain associated with depression. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby enhancing neuroplasticity in the brain and reducing or alleviating depression. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces the severity and/or frequency of episodes of clinical depression in an individual. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces the severity and/or frequency of episodes of intractable clinical depression in an individual. In some embodiments, treatment with a PAK modulator (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces side effects (e.g., suicidal thoughts) in an individual that is refractory to treatment with other putative treatments for clinical depression. Accordingly, provided herein are methods of treating clinical depression comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Down's Syndrome

[0055] Down's syndrome is caused by the presence of an extra 21 st chromosome. Down's syndrome is associated with varying degrees of developmental disability and mental retardation. A number of individuals with Down syndrome have mental retardation in the mild (IQ 50-70) to moderate (IQ 35-50) range.

[0056] In some instances, developmental disability and/or mental retardation associated with Down's syndrome is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21- activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reversing or partially reversing developmental disability and/or mental retardation associated with Down's syndrome. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby re-engineering neural circuitry in the brain of an individual suffering from Down's syndrome. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces the impairment in cognitive function associated with Downs Syndrome. Accordingly, provided herein are methods of treating Down's syndrome comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Mental Retardation Sydromes other than Down's syndrome

[0057] Several other forms of mental retardation syndromes are known including and not limited to Fetal alcohol sydrome, Klinefelter's syndrome, congenital hypothyroidism, Williams syndrome, Smith-Lemli-Opitz syndrome, Prader-willi syndrome, Phelan-McDermid syndrome, Mowat- Wilson syndrome, Ciliopathy, Lowe syndrome and/or any other genetic mental retardation syndromes.

[0058] In some instances, developmental disability and/or mental retardation associated with the afroementioned syndromes is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21-activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reversing or partially reversing developmental disability and/or mental retardation associated with Down's syndrome. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby re-engineering neural circuitry in the brain of an individual suffering from mental retardation syndromes described above. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces the impairment in cognitive function associated with genetic mental retardation syndromes. Accordingly, provided herein are methods of treating any of the aforementioned mental retardation syndromes comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Tuberous Sclerosis

[0059] Tuberous Sclerosis (TS) is a genetically inherited disorder that causes formation of cortical tubers (hamartias) and tumors on other organs. A combination of symptoms includes seizures, developmental delay, and/or behavioral deficits. In some instances, Tuberous Sclerosis is associated with higher incidence of autism and/or obsessive compulsive disorder.

[0060] In some instances, developmental disability and/or behavioral problems associated with TS are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21 -activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reversing or partially reversing developmental disability and/or behavioral problems associated with TS. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reducing ocurrence of seizures in inviduals diagnosed with TS. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reducing or reversing learning disabilities associated with TS. In some instances, modulation of PAK activity reduces or reverses the learning disabilities associated with TS. In some instances, modulation of PAK activity reduces or reverses the learning autism and/or obsessive compulsive disorder associated with TS. In some instances, modulation of PAK activity reduces the occurence of seizures associated with TS. Accordingly, provided herein are methods of treating tuberous sclerosis comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

Neurofibromatosis

[0061] Neurofibromatosis (NF), also called von Recklinghaus disease, is an autosomal dominant genetically-inherited disorder in which the nerve tissue grows tumors (i.e., neurofibromas, ocular gliomas or the like). Patients with NF1 exhibit a number of different disease symptoms including increased risk of forming nervous system tumors and cognitive deficits such as defects in visual-spatial function, attention and motor coordination.

[0062] NF is of Type 1 or Type 2. As used herein, NF includes Type 1 NF and Type 2 NF. In some instances, Type 1 NF is inherited or results from spontaneous mutation of neurofibromin.

In some instances, NF Type 1 is associated with learning disabilities in individuals affected by the disease. In some instances the disease is associated with a partial absence seizure disorder.

In some instances NF Type 1 is associated with poor language, visual-spatial skills, learning disability (e.g., attention deficit hyperactivity disorder), headache, epilepsy or the like.

[0063] Type 2 NF is inherited or results from spontaneous mutation of merlin. In some instances, NF Type 2 causes symptoms of hearing loss, tinnitus, headaches, epilepsy, cataracts and/or retinal abnormalities, paralysis and/or learning disabilities.

[0064] Patients with NF1 and NF2 are at increased risk of forming nervous system tumors. In type 1 patients this includes dermal and plexiform neurofibromas, malignant peripheral nerve sheath tumors (MPNST) and other malignant tumors, while type 2 patients may develop multiple cranial and spinal tumors.

[0065] In one embodiment is a method of treating tumors in patients suffering from

neurofibromatosis comprising administering to the patient a PA inhibitor described herein. In one embodiment, the tumor is malignant peripheral nerve sheath tumor. In another embodiment, the tumor results from NF Type 1. In another embodiment, the tumor results from NF Type 2. In a further embodiment, the tumor is an eye tumor (glioma growing in the optic nerve.) In yet a further embodiment, the tumor is selected from a brain tumor, adrenal gland tumor, muscle tumor, spinal cord tumor and peripheral nerve sheath tumor. In one embodiment the PAK inhibitor used to treat the tumor resulting from neurofibromatosis is a compound of Formula I - XXIX.

[0066] In some instances, developmental disability and/or behavioral problems associated with NF are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function. In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function is associated with activation of p21 -activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reversing or partially reversing developmental disability and/or behavioral problems associated with NF. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reducing ocurrence of seizures in individuals diagnosed with NF. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reducing or reversing learning disabilities associated with NF. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces cognitive deficits associated with NF. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces learning disability and/or epilepsy and/or any other symptoms associated with NF. In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces the incidence of tumor development associated with NF. Accordingly, provided herein are methods of treating neurofibromatosis comprising

administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

[0067] In some embodiments, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK by a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of

Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), alleviates, reverses or reduces occurence of neurofibromas (tumors of the peripheral nerve) and/or optic nerve gliomas.

[0068] In some embodiments, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK by a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), alleviates, reverses or reduces occurence of hearing loss or tinnitus associated with NF.

[0069] In some embodiments, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK by a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), alleviates, reverses or reduces occurence of cataracts and/or retinal abnormalities.

[0070] In some embodiments, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK by a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), alleviates, reverses or reduces occurence of abnormal bone development associated with NF. Other diseases

[0071] Maple Syrup Urine Disease is an inherited metabolic disorder in which the body is unable to process certain protein building blocks (amino acids) properly. The disease is also associated with developmental delay and/or neurodegeneration. In some instances, untreated maple syrup urine disease leads to seizures. In some instances, developmental delay and/or neurodegeneration associated with Maple Syrun Urine Disease is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0072] Neimann Pick disease is an inherited lysosomal storage disease that affects multiple organs including the nervous system. In some instances, untreated disease is responsible for gradual loss of intellectual abilities causing dementia and seizures. In some instances, dementia and seizures associated with Neimann Pick disease are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0073] Bipolar disorder involves periods of excitability (mania) alternating with periods of depression. The "mood swings" between mania and depression are abrupt. In some instances, mood swings and/or mania and /or depression associated with Bipolar disorder are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0074] Mania, is a state of abnormally elevated or irritable mood, arousal, and/ or energy levels. In addition to mood disorders, manic behavior results from drug intoxication, medication side effects (notably steroids), or malignancy. Mania is also associated with bipolar disorder. In some instances, mood swings and/or mania are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0075] Anxiety disorder is a pattern of frequent, constant worry and anxiety over many different activities and events. Anxiety is defined as an unpleasant emotional state for which the cause is either not readily identified or perceived to be uncontrollable or unavoidable. Fear is an emotional and physiological response to a recognized external threat. As used herein, the term anxiety disorder includes fears (phobias) as well as anxieties. In some instances, anxiety disorders are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0076] Posttraumatic stress disorder (post-traumatic stress disorder, PTSD) is a severe anxiety disorder that in certain individuals develops after exposure to any event that results in psychological trauma. In some instances, PTSD is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function. [0077] Schizoaffective disorder is a psychiatric diagnosis characterized by recurring episodes of elevated or depressed mood, or of simultaneously elevated and depressed mood, that alternate with, or occur together with, distortions in perception. In some instances, schizoaffective disorder is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0078] Schizophreniform disorder is a mental disorder diagnosed when symptoms of schizophrenia are present for a significant portion of the time within a one-month period, but signs of disruption are not present for the full six months required for the diagnosis of schizophrenia. In some instances, schizophreniform disorder is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0079] In some instances, menopause is associated with cognitive decline. In some instances, such cognitive decline associated with menopause is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0080] Angelman syndrome (AS) is a neuro-genetic disorder characterized by intellectual and developmental delay, sleep disturbance, seizures, jerky movements (e.g., hand-flapping, frequent laughter or smiling). In some instances, such developmental delay and/or seizures are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0081] Obsessive-compulsive disorder (OCD) is a mental disorder characterized by intrusive thoughts that produce anxiety, by repetitive behaviors aimed at reducing anxiety, or by a combination thereof. In some instances, OCD is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0082] Panic disorder is an anxiety disorder and is characterized by unexpected and/or repeated episodes of intense fear accompanied by physical symptoms such as chest pain, heart palpitations, shortness of breath, dizziness, or abdominal distress. In some instances, panic disorder is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0083] A phobia is an intense and persistent fear of certain situations, activities, things, animals, or people. When the fear is uncontrolled, and/or if the fear is interfering with daily life, then a diagnosis under one of the anxiety disorders is made. In some instances, phobias are associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0084] Anorexia nervosa (AN) is an eating disorder characterized by refusal to maintain a healthy body weight, and an obsessive fear of gaining weight. In some instances, AN is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0085] Bulimia nervosa is an illness in which a person binges on food or has regular episodes of significant overeating and feels a loss of control. The affected person then uses various methods — such as vomiting or laxative abuse— to prevent weight gain. In some instances, bulimia nervosa is associated with abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function.

[0086] In some instances, abnormal dendritic spine morphology and/or dendritic spine density and/or synaptic function associated with any of the neurological conditions described above is associated with activation of p21-activated kinase (PAK). In some instances, modulation of PAK activity (e.g., inhibition or partial inhibition of PAK) alleviates, reverses or reduces abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function thereby reversing or partially reversing neuro degeneration and/or dementia and/or seizures associated with, for example, Maple Syrup Urine Disease and/or Neimann Pick disease.

[0087] Accordingly, provided herein are methods of treating Maple Syrup Urine Disease comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0088] Accordingly, also provided herein are methods of treating Neimann Pick disease comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof. [0089] Accordingly, also provided herein are methods of treating bipolar disorder comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0090] Accordingly, also provided herein are methods of treating mania comprising

administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0091] Accordingly, also provided herein are methods of treating anxiety disorder comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0092] Accordingly, also provided herein are methods of treating PTSD comprising

administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0093] Accordingly, also provided herein are methods of treating schizoaffective disorder comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0094] Accordingly, also provided herein are methods of treating schizophreniform comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0095] Accordingly, also provided herein are methods of treating cognitive decline associated with menopause comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0096] Accordingly, also provided herein are methods of treating Angelman Syndrome comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0097] Accordingly, also provided herein are methods of treating OCD comprising

administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of

Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[0098] Accordingly, also provided herein are methods of treating panic disorders comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of

Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof. [0099] Accordingly, also provided herein are methods of treating phobias comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[00100] Accordingly, also provided herein are methods of treating Anorexia Nervosa comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

[00101] Accordingly, also provided herein are methods of treating Bulimia Nervosa comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein), to an individual in need thereof.

Dendritic Spines

[00102] A dendritic spine is a small membranous protrusion from a neuron's dendrite that serves as a specialized structure for the formation, maintenance, and/or function of synapses. Dendritic spines vary in size and shape. In some instances, spines have a bulbous head (the spine head) of varying shape, and a thin neck that connects the head of the spine to the shaft of the dendrite. In some instances, spine numbers and shape are regulated by physiological and pathological events. In some instances, a dendritic spine head is a site of synaptic contact. In some instances, a dendritic spine shaft is a site of synaptic contact. Figure 1 shows examples of different shapes of dendritic spines. Dendritic spines are "plastic." In other words, spines are dynamic and continually change in shape, volume, and number. In some instances, spines change in shape, volume, length, thickness or number in a few hours. In some instances, spines change in shape, volume, length, thickness or number occurs within a few minutes. In some instances, spines change in shape, volume, length, thickness or number occurs in response to synaptic transmission and/or induction of synaptic plasticity. By way of example, dendritic spines are headless (filopodia as shown, for example, in Figure la), thin (for example, as shown in Figure lb), stubby (for example as shown in Figure lc), mushroom-shaped (have door-knob heads with thick necks, for example as shown in Figure Id), ellipsoid (have prolate spheroid heads with thin necks, for example as shown in Figure le), flattened (flattened heads with thin neck, for example as shown in Figure If) or branched (for example as shown in Figure lg).

[00103] In some instances, mature spines have variably-shaped bulbous tips or heads,

-0.5-2 μιη in diameter, connected to a parent dendrite by thin stalks 0.1-1 μιη long. In some instances, an immature dendritic spine is filopodia-like, with a length of 1.5 - 4 μιη and no detectable spine head. In some instances, average spine density ranges from 0.5 to 10 spines per micrometer length of dendrite, and varies with maturational stage of the spine and/or the neuronal cell. In some instances, dendritic spine density ranges from 1 to 40 spines per 10 micrometer in medium spiny neurons.

[00104] In some instances, the shape of the dendritic spine head determines synaptic function. Defects in dendritic spine morphology and/or function have been described in neurological diseases. As an example only, the density of dendritic spines has been shown to be reduced in pyramidal neurons from patients with schizophrenia (Glanz and Lewis, Arch Gen Psychiatry, 2000:51:65-13). In another example, neurons from patients with Fragile X mental retardation show a significant increase in the overall density of dendritic spines, together with an increase in the proportion of "immature", filopodia-like spines and a corresponding reduction of "mature", mushrooms-shaped spines (Irvin et al, Cerebral Cortex, 2000; 10: 1038-1044). In many cases, the dendritic spine defects found in samples from human brains have been recapitulated in rodent models of the disease and correlated to defective synapse funtion and/or plasticity. In some instances, dendritic spines with larger spine head diameter form more stable synapses compared with dendritic spines with smaller head diameter. In some instances, a mushroom-shaped spine head is associated with normal or partially normal synaptic function. In some instances, a mushroom-shaped spine head is a healthier (e.g., having normal or partially normal synapses) compared to a spine with a reduced spine head size, spine head volume and/or spine head diameter. In some instances, inhibition or partial inhibition of PAK activity (e.g., with a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) results in an increase in spine head diameter and/or spine head volume and/or reduction of spine length, thereby normalizing or partially normalizing synaptic function in individuals suffering or suspected to be suffering from or pre- disposed to neurological conditions.

p21-activated kinases (PAKs)

[00105] The PAKs constitute a family of serine-threonine kinases that are composed of

"conventional", or Group I PAKs, that includes PAK1, PAK2, andPAK3, and "non- conventional", or Group II PAKs, that includes PAK4, PAK5, and PAK6. See, e.g., Zhao et al. (2005), Biochem J, 386:201-214. These kinases function downstream of the small GTPases Rac and/or Cdc42 to regulate multiple cellular functions, including dendritic morphogenesis and maintenance (see, e.g., Ethell et al (2005), Prog in Neurobiol, 75: 161-205; Penzes et al (2003), Neuron, 37:263-274), motility, morphogenesis, angiogenesis, and apoptosis, (see, e.g., Bokoch et al., 2003, Annu. Rev. Biochem., 72:743; and Hofmann et al., 2004, J. Cell Sci., 117:4343;). GTP-bound Rac and/or Cdc42 bind to inactive PAK, releasing steric constraints imposed by a PAK autoinhibitory domain and/or permitting PAK phosphorylation and/or activation.

Numerous phosphorylation sites have been identified that serve as markers for activated PAK.

[00106] In some instances, upstream effectors of PAK include, but are not limited to,

TrkB receptors; NMDA receptors; adenosine receptors; estrogen receptors; integrins, EphB receptors; CDK5, FMRP; Rho-family GTPases, including Cdc42, Rac (including but not limited to Racl and Rac2), Chp, TC10, and Wrnch-1; guanine nucleotide exchange factors ("GEFs"), such as but not limited to GEFT, a-p-21 -activated kinase interacting exchange factor ( PIX), Kalirin-7, and Tiaml ; G protein-coupled receptor kinase-interacting protein 1 (GIT1), and sphingosine. [00107] In some instances, downstream effectors of PAK include, but are not limited to, substrates of PAK kinase, such as Myosin light chain kinase (MLCK), regulatory Myosin light chain (R-MLC), Myosins I heavy chain, myosin II heavy chain, Myosin VI, Caldesmon, Desmin, O l8/stathmin, Merlin, Filamin A, LIM kinase (LIMK), Ras, Raf, Mek, p47phox, BAD, caspase 3, estrogen and/or progesterone receptors, RhoGEF, GEF-H1, NET1, Gaz, phosphoglycerate mutase-B, RhoGDI, prolactin, p41Arc, cortactin and/or Aurora-A (See, e.g., Bokoch et al., 2003, Annu. Rev. Biochem., 72:743; and Hofmann et al., 2004, J. Cell Sci., 117:4343). Other substances that bind to PAK in cells include CIB; sphingo lipids;

lysophosphatidic acid, G-protein β and/or γ subunits; PIX/COOL; GIT/PKL; Nef; Paxillin; NESH; SH3 -containing proteins (e.g. Nek and/or Grb2); kinases (e.g. Akt, PDK1, PI 3- kinase/p85, Cdk5, Cdc2, Src kinases, Abl, and/or protein kinase A (PKA)); and/or phosphatases (e.g. phosphatase PP2A, POPX1, and/or POPX2).

PAK Inhibitors

[00108] Described herein are PAK inhibitors that treat one or more symptoms associated with neurological conditions. Also described herein are pharmaceutical compositions comprising a PAK inhibitor (e.g., a PAK inhibitor compound of Formula I, compound of Formula II, compound of Formula III, compound of Formula IV, compound of Formula V, compound of Formula VI, compound of Formula VII, compound of Formula VIII, compound of Formula IX, compound of Formula X, compound of Formula XI, compound of Formula XII, compound of Formula XIII, compound of Formula XIV, compound of Formula XV, compound of Formula XVI, compound of Formula XVII, compound of Formula XVIII, compound of Formula XIX, compound of Formula XX, compound of Formula XXI, compound of Formula XXII, or a compound of Formula XXIII, as described herein) for treatment of one or more symptoms of neurological conditions. Also described herein is the use of a PAK inhibitor for manufacture of a medicament for treatment of one or more symptoms of neurological conditions. In some embodiments, PAK inhibitors and compositions thereof treat the cognitive impairments associated with neurological conditions (e.g., subcortical dementia, learning disability or the like). In some other embodiments, PAK inhibitors and compositions thereof delay or halt the progression of neurological conditions. In some embodiments, PAK inhibitors and compositions thereof reduce or reverse the occurence or severity of symptoms of neurological conditions (e.g., seizures).

[00109] In some embodiments, the PAK inhibitor is a Group I PAK inhibitor that inhibits, for example, one or more Group I PAK polypeptides, for example, PAK1, PAK2, and/or PAK3. In some embodiments, the PAK inhibitor is a PAK1 inhibitor. In some embodiments, the PAK inhibitor is a PAK2 inhibitor. In some embodiments, the PAK inhibitor is a PAK3 inhibitor. In some embodiments, the PAK inhibitor is a mixed PAK1/PAK3 inhibitor. In some embodiments, the PAK inhibitor inhibits all three Group I PAK isoforms (PAKl, 2 and PAK3) with equal or similar potency. In some embodiments, the PAK inhibitor is a Group II PAK inhibitor that inhibits one or more Group II PAK polypeptides, for example PAK4, PAK5, and/or PAK6. In some embodiments, the PAK inhibitor is a PAK4 inhibitor. In some embodiments, the PAK inhibitor is a PAK5 inhibitor. In some embodiments, the PAK inhibitor is a PAK6 inhibitor.

[00110] In some embodiments, a PAK inhibitor described herein reduces or inhibits the activity of one or more of PAKl, PAK2 and/or PAK3 while not affecting the activity of PAK4, PAK5 and/or PAK6. In some embodiments, a PAK inhibitor described herein reduces or inhibits the activity of one or more of PAKl, PAK2, PAK3, and/or PAK4. In some embodiments, a PAK inhibitor described herein reduces or inhibits the activity of one or more of PAKl, PAK2, PAK3, and/or one or more of PAK4, PAK5 and/or PAK6. In some embodiments, a PAK inhibitor described herein is a substantially complete inhibitor of one or more PAKs. As used herein, "substantially complete inhibition" means, for example, > 95% inhibition of one or more targeted PAKs. In other embodiments, "substantially complete inhibition" means, for example, > 90% inhibition of one or more targeted PAKs. In some other embodiments, "substantially complete inhibition" means, for example, > 80 % inhibition of one or more targeted PAKs. In some embodiments, a PAK inhibitor described herein is a partial inhibitor of one or more PAKs. As used herein, "partial inhibition" means, for example, between about 40% to about 60% inhibition of one or more targeted PAKs. In other embodiments, "partial inhibition" means, for example, between about 50% to about 70% inhibition of one or more targeted PAKs.

[00111] In some embodiments, a PAK inhibitor suitable for the methods described herein is a compound having the structure of Formula I or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond; R 6 is -CN, -OH, substituted or unsubstituted alkoxy, -N(R 10 )2, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is halogen, -CN, -OH, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , - N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted hetero arylalkyl, or substituted or unsubstituted cycloalkyl or heterocycloalkyl fused to ring A;

ring A is substituted or unsubstituted aryl or heteroaryl substituted with 0-4 R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , -

NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1( together with the atoms to which they are attached form a heterocycle;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8. [00112] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula II or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond;

R 6 is substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is H, halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted hetero arylalkyl, or substituted or unsubstituted cycloalkyl or heterocycloalkyl fused to ring A;

ring A is substituted or unsubstituted aryl or heteroaryl substituted with 0-4 R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1C together with the atoms to which they are attached form a heterocycle;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00113] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula III or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond;

R 6 is H, or halogen;

R 7 is acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl or substituted or unsubstituted heteroaryl;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted hetero arylalkyl, or substituted or unsubstituted cycloalkyl or heterocycloalkyl fused to ring A;

ring A is substituted or unsubstituted aryl or heteroaryl substituted with 0-4 R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 10 together with the atoms to which they are attached form a heterocycle;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00114] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula IV or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond;

R 6 is substituted or unsubstituted alkyl;

R 7 is substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted heterocycloalkyl;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted hetero arylalkyl, or substituted or unsubstituted cycloalkyl or heterocycloalkyl fused to ring A;

ring A is substituted or unsubstituted aryl or heteroaryl substituted with 0-4 R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , -

NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1( together with the atoms to which they are attached form a heterocycle;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00115] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula V or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond; R 6 is H, or halogen;

R 7 is H, halogen, CN, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , C0 2 R 10 , N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

Q is substituted or unsubstituted cycloalkyl or heterocycloalkyl fused to ring A;

ring A is substituted or unsubstituted aryl or heteroaryl substituted with 0-4 R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , -

NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1( together with the atoms to which they are attached form a heterocycle;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00116] In some embodiments, the compound of Formula V has the structure of Formula

VI:

wherein:

each of Y 3 , Y 4 and Y 5 are independently N-R la , CR ! R 2 , S0 2 , or C=0;

R la is H or substituted or unsubstituted alkyl;

R 1 and R 2 are each independently H or substituted or unsubstituted alkyl.

[00117] In some embodiments, the compound of Formula V has the structure of

Formula VIII:

wherein:

ring A is an aryl or heteroaryl substituted with R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 9 , NR 10 C(=O)OR 9 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroary, or two R 1( together with the nitrogen to which they are attached form a heterocycle;

each R 11 is independently H, halogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, or two R 11 together with the carbon atom to which they are attached form C=0;

s is 0-4;

k is 1-4;

z is 0 or 1 ; u is 1, 2 or 3;

provided that z + u≠ 1 ;

ring B is an aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , NR 10 C(=O)R 9 ,

NR 10 C(=O)OR 9 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8;

R 6 is H, or halogen;

R 7 is H, halogen, CN, OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , C0 2 R 10 , N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.

[00118] In some embodiments, ring A is a heteroaryl ring. In some embodiments, ring A is a phenyl ring.

[00119] In some embodiments, the compound of Formula VIII has a structure of Formula

VIIIA, Formula VIIIB, Formula VIIIC, Formula VIIID, Formula VIIIE, Formula VIIIF, Formula VIIICG or Formula VII1H:

[00120] In some embodiments, R 11 is H, halogen or substituted or unsubstituted alkyl. In some embodiments, R 11 is H. [00121] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula IX or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond;

R 6 is H;

R 7 is

ring T is aryl, heteroaryl, cycloalkyl or heterocycloalkyl substituted with R 3 and R 4 ;

R 3 is a substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl attached to ring T via a carbon atom;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, substituted or unsubstituted hetero arylalkyl, or substituted or unsubstituted cycloalkyl or heterocycloalkyl fused to ring A;

ring A is substituted or unsubstituted aryl or heteroaryl substituted with 0-4 R 4 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl; R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1( together with the atoms to which they are attached form a heterocycle;

s is 0-4;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00122] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula X or pharmaceutically acceptable salt or Noxide thereof:

W is a bond;

R 6 is H, halogen, -CN, -OH, substituted or unsubstituted alkoxy, -N(R 10 ) 2 , substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is H, halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

Q is

R 1 is H or substituted or unsubstituted alkyl;

R 2 is substituted or unsubstituted alkyl, or R 1 and R 2 together with the carbon to which they are attached form a C3-C6 cycloalkyl ring;

P is 1, 2 or 3;

ring A is aryl substituted with R 4 ;

R 3 is halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -CF 3 , -SR 8 , -S(=0)R 9 , S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 9 R 10 , NR 10 C(=O)OR 9 OR 9 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -CF 3 , -SR 8 , -S(=0)R 9 , - S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , - C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1C together with the atoms to which they are attached form a heterocycle;

s is 0-4;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8. [00123] In some embodiments, a compound of Formula X is a compound wherein

W is a bond;

R 6 is H, halogen, -CN, -OH, substituted or unsubstituted alkoxy, -N(R 10 )2, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is H, halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

Q is

R 1 is H or substituted or unsubstituted alkyl;

R 2 is substituted or unsubstituted alkyl, or R 1 and R 2 together with the carbon to which they are attached form a C3-C6 cycloalkyl ring;

P is 1, 2 or 3;

ring A is aryl substituted with R 3 and R 4 ;

R 3 is halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -CF 3 , -SR 8 , -S(=0)R 9 , S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 9 R 10 , NR 10 C(=O)OR 9 OR 9 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -CF 3 , -SR 8 , -S(=0)R 9 , - S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , - C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted

10 cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R together with the atoms to which they are attached form a heterocycle;

s is 0-4;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00124] In some embodiments, a compound of Formula X has the structure of Formula

XA or Formula XB:

[00125] In some embodiments, the compound of Formula X has the structure of Formula

XI:

wherein:

R is H or substituted or unsubstituted alkyl;

R 2 is substituted or unsubstituted alkyl; and

R 3 is halogen, alkyl, fluoroalkyl, alkoxy, fluoroalkoxy, or SR 8 . [00126] In some embodiments, the compound of Formula (XI) has the structure of

Formula (XIIA) or Formula (XIIB):

Formula (XIIB)

[00127] In some embodiments, a PAK inhibitor suitable for the methods described herein is a compound having the structure of Formula XIII or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is a bond;

R 6 is H, halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -N(R 10 ) 2 , substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is H, halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

Q is

R 1 and R 2 are each independently H or substituted or unsubstituted alkyl; or R 1 and R 2 together with the carbon to which they are attached form a C3-C6 cycloalkyl ring;

p is 1, 2 or 3;

ring A is aryl substituted with R 3 and R 4 ;

R 3 is a substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl attached to ring A via a carbon atom;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -CF 3 , -SR 8 , -S(=0)R 9 , -

S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -

C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , N R 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 1( together with the atoms to which they are attached form a heterocycle;

s is 0-4;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8.

[00128] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula XIV or pharmaceutically acceptable salt or Noxide thereof:

Formula XIV

wherein:

W is O;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted hetero arylalkyl;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8;

R 6 is halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, -N(R 10 ) 2 , substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is H, halogen, -CN, -OH, acyl, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -N(R 10 ) 2 , substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

[00129] In some embodiments, the compound of Formula XIV has the structure of

Formula XV:

wherein:

p is 0, 1, 2 or 3;

R 1 and R 2 are each independently H or substituted or unsubstituted alkyl; or R

together with the carbon to which they are attached form a C3-C6 cycloalkyl ring. [00130] In some embodiments, ring A is an aryl ring. In some embodiments, ring A is a phenyl or naphthyl ring. In some embodiments, ring A is a heteroaryl ring. In some

embodiments, ring A is a heterocycloalkyl ring. In some embodiments, ring A is a cycloalkyl ring.

[00131] In some embodiments, a PA inhibitor suitable for the methods described herein is a compound having the structure of Formula XVI or pharmaceutically acceptable salt or Noxide thereof:

wherein:

W is N-R la ;

R la is H or substituted or unsubstituted alkyl;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted hetero arylalkyl;

ring B is aryl or heteroaryl substituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , NR 10 C(=O)OR 10 , NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8;

R 6 is H, halogen, -CN, -OH, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, -N(R 10 ) 2 , substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 7 is H, halogen, -CN, -OH, acyl, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -N(R 10 ) 2 , substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

[00132] In some embodiments, the compound of Formula XVI has the structure of

Formula XVII:

wherein:

each of Y 3 , Y 4 and Y 5 are independently N-R la , CR ! R 2 , S0 2 , or C=0;

R la is H or substituted or unsubstituted alkyl;

R 1 and R 2 are each independently H or substituted or unsubstituted alkyl.

[00133] In some embodiments, a compound of Formula XVI has the structure of formula

XVIII:

[00134] In some embodiments, a compound of Formula XVI has the structure of formula

XIX:

p is 1, 2 or 3;

R ! and R 2 are each independently H or substituted or unsubstituted alkyl; or R 1 and R 2 together with the carbon to which they are attached form a C3-C6 cycloalkyl ring.

[00135] In some embodiments, ring A is a heteroaryl ring. In some embodiments, ring A is an aryl ring. In some embodiments, ring A is a heterocycloalkyl ring. In some embodiments, ring A is a cycloalkyl ring.

[00136] In some embodiments, the compound of Formula XVI has the structure of

Formula XX:

wherein:

each of Y 3 , Y 4 and Y 5 are independently N-R la , CR ! R 2 , S0 2 , or C=0;

R la is H or substituted or unsubstituted alkyl;

R 1 and R 2 are each independently H or substituted or unsubstituted alkyl.

[00137] In some embodiments, the compound of Formula XVI has the structure of

Formula XXI A Formula XXIB, Formula XXIC or Formula XXID:

Formula XXID

wherein:

each R 11 is independently H, halogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, or two R 11 together with the carbon atom to which they are attached form C=0; and

k is 1-4.

[00138] In some embodiments, for any of the compounds described above, ring B is

[00139] In one embodiment for any of the compounds described herein,

is selected from:

[00141] In some embodiments, a PAK inhibitor is a compound having the structure of

Formula XXII or a harmaceutically acceptable salt, solvate or N-oxide thereof:

wherein:

R 1 and ft 2 are each independently H or substituted or unsubstituted alkyl; or R 1 and R 2 together with the carbon to which they are attached form a substituted or unsubstituted C3-C6 cycloalkyl ring;

p is 1, 2 or 3; ring A is a heteroaryl comprising 1-4 heteroatoms selected from O, S and N;

R 3 is a substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroaryl attached to ring A via a carbon atom, or substituted or unsubstituted heterocycloalkyl attached to ring A via a carbon atom;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCHF 2 , -OCF 2 H, -CF 3 , -SR 8 , - S(=0)R 9 , -S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -N R 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted

heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or

unsubstituted heteroaryl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 10 together with the atoms to which they are attached form a substituted or unsubstituted heterocycle;

s is 0-4;

ring B is aryl or heteroaryl;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , -OR 10 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8; and

R 7 is H, halogen, -CN, substituted or unsubstituted alkyl, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -OR 10 , - N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl.

[00142] In some embodiments, a PA inhibitor is a compound having the structure of

Formula XXIII or pharmaceutically acceptable salt or N-oxide thereof:

Formula XXIII;

wherein:

wherein ring T is an aryl, or a heteroaryl ring;

R 3 is a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heteroaryl attached to ring T via a carbon atom of R 3 , or a substituted or unsubstituted heterocycloalkyl attached to ring T via a carbon atom of R 3 ;

Q is a substituted or unsubstituted alkyl, a substituted or unsubstituted heteroalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted cycloalkylalkyl, a substituted or unsubstituted heterocycloalkylalkyl, a substituted or unsubstituted aryl, a substituted or unsubstituted arylalkyl, a substituted or unsubstituted heteroaryl, or a substituted or unsubstituted heteroarylalkyl;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCH 2 F, -OCF 2 H, -CF 3 , -SR 8 , - NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - NR 10 C(=O)R 10 , -N R 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , a substituted or unsubstituted alkyl, a substituted or unsubstituted alkoxy, a substituted or unsubstituted heteroalkyl, a substituted or unsubstituted cycloalkyl, or a substituted or unsubstituted heterocycloalkyl;

R 8 is H or R 9 ;

R 9 is a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl;

each R 10 is independently H, a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or two R 10 , together with the atoms to which they are attached, form a heterocycle;

ring B is aryl or heteroaryl;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , -OR 10 , a substituted or unsubstituted alkyl, a substituted or unsubstituted alkoxy, a substituted or unsubstituted heteroalkyl, a substituted or unsubstituted cycloalkyl, or a substituted or unsubstituted heterocycloalkyl;

r is 0 to 8; and

s is 0 to 4.

[00143] In some embodiments, a PA inhibitor is a compound having the structure of

Formula XXIV or a pharmaceutically acceptable salt, solvate or N-oxide thereof:

Formula XXIV

wherein: each of Y 3 , Y 4 and Y 5 is independently a bond, O, N, N-R 1 , C-R 1 , C(R 1 ) 2 , S, S0 2 , or C=0;

provided that Y 3 is not a bond and at least one Y 3 , Y 4 or Y 5 is selected from O, N-R 1 , S, or S0 2 ; each Z is independently N or C-R 4 ;

each R 1 is independently selected from hydrogen, a substituted or unsubstituted alkyl, a substituted or unsubstituted alkoxy, a substituted or unsubstituted heteroalkyl, a substituted or unsubstituted cycloalkyl, or a substituted or unsubstituted heterocycloalkyl, -S(=0)R 9 , - S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , - C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -N R 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , or two R 1 together wiht the the atom to which they are attached form a ring;

R 2 is H or a substituted or unsubstituted alkyl;

each R 4 is independently hydrogen, halogen, -CN, -N0 2 , -OH, -OCFH 2 , -OCF 3 , -OCF 2 H, -CF 3 , - SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -N R 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , a substituted or unsubstituted alkyl, a substituted or unsubstituted alkoxy, a substituted or unsubstituted heteroalkyl, a substituted or unsubstituted cycloalkyl, or a substituted or unsubstituted hetero cyclo alkyl;

ring B is aryl or hetero aryl; each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , -OR 10 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

r is 0-8;

R 7 is H, halogen, -CN, substituted or unsubstituted alkyl, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -OR 10 , - N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 6 is H, halogen, -OR, -NR 10 R 10 , a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, a substituted or unsubstituted heteroaryl;

R 8 is H or R 9 ;

R 9 is a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroarylyl; and

each R 10 is independently H, a substituted or unsubstituted alkyl, a substituted or unsubstituted cycloalkyl, a substituted or unsubstituted heterocycloalkyl, a substituted or unsubstituted aryl, or a substituted or unsubstituted heteroaryl; or two R 10 , together with the atoms to which they are attached, form a heterocycle.

[00144] In one embodiment is a compound of Formula XXVI wherein X is selected from:

[00145] In some embodiments, a PA inhibitor is a compound having the structure of

Formula XXV or pharmaceuticall acceptable salt or N-oxide thereof:

Formula XXV:

wherein:

R 1 and R 2 are each independently H or substituted or unsubstituted alkyl; or R 1 and R 2 together with the carbon to which they are attached form a C3-C6 cycloalkyl ring;

p is 1, 2 or 3;

ring A is aryl or heteroaryl;

R 3 is S(=0)R 9 or -S(=0) 2 R 9 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -OCH 2 F, -CF 3 , -SR 8 , - S(=0)R 9 , -S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -OR 10 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -N R 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl; each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 10 together with the atoms to which they are attached form a heterocycle;

s is 0-4;

ring B is aryl or heteroaryl;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , -OR 10 , substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted hetero cyclo alkyl;

r is 0-8;

R 7 is H, halogen, -CN, substituted or unsubstituted alkyl, -C(=O)N(R 10 ) 2 , -C0 2 R 10 , -OR 10 , - N(R 10 ) 2 , acyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl

provided that the compound of Formula I is not 2-(4-(4-methylpiperazin-1-yl)phenylamino)-8- (2-(methylsulfonyl)benzyl)pyrido[2,3-d]pyrimidin-7(8H)-one, 8-(2-fluoro-6- (methylsulfinyl)benzyl)-2-(4-(4-methylpiperazin-l-yl)phenyla mino)pyrido[2,3-d]pyrimidin- 7(8H)-one, or 8-(2-fluoro-6-(methylsulfinyl)benzyl)-2-(4-(4-methylpiperazi n-1- yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H)-one.

[00146] In some embodiments, a PA inhibitor is a compound having the structure of

Formula XXVI or a pharmaceuticall acceptable salt, solvate, or N-oxide thereof:

Formula XXVI;

wherein:

ring T is aryl or heteroaryl;

R 3 is -S(=0)R 9 , (R) -S(=0)R 9 , (S) -S(=0)R 9 , or -S(=0) 2 R 9 ;

each R 4 is independently halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCF 2 H, -OCFH 2 , -CF 3 , -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 ,

-N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -N R 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, or substituted or unsubstituted cycloalkyl;

R 8 is H or R 9 ;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or two R 10 together with the atoms to which they are attached form a substituted or unsubstituted heterocycle;

s is 0-4;

Q is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkylalkyl, substituted or unsubstituted heterocycloalkylalkyl, substituted or unsubstituted aryl, substituted or unsubstituted arylalkyl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted hetero arylalkyl;

ring B is aryl or heteroaryl;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -OR 10 , -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , -NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted heterocycloalkyl; and

r is 0-8.

[00147] In some embodiments, a PA inhibitor is a compound having the structure of Formula XXVII or a pharmaceuticall acceptable salt, solvate or N-oxide thereof:

Formula XXVII;

wherein:

L 1 is O, NR 8 , or S;

ring B is an optionally substituted aryl or heteroaryl; R 7 is H, OR 10 , N(R 10 ) 2 , a substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

L 2 is C(R 1 R 2 ) P , O, NR 8 , or S and R 6 is alkyl, cycloalkyl, heterocycloalkyl, aryl or heteroaryl optionally substituted with at least one R 12 ; or

L 2 is a bond and R 6 is alkyl, cycloalkyl, aryl or heteroaryl optionally substituted with at least one R 12 ;

R 12 is halogen, -CN, -N0 2 , -OH, -OCF 3 , -OCHF 2 , -OCF 2 H, -CF 3 , -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , - NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=O)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , - OR 10 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl; substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -SR 8 , -S(=0)R 9 , -S(=0) 2 R 9 , NR 10 S(=O) 2 R 9 , -S(=O) 2 N(R 10 ) 2 , -C(=0)R 9 , -OC(=0)R 9 , -C0 2 R 10 , -N(R 10 ) 2 , -C(=O)N(R 10 ) 2 , -NR 10 C(=O)R 10 , -NR 10 C(=O)OR 10 , -NR 10 C(=O)N(R 10 ) 2 , -OR 10 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

R 8 is H or substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or

unsubstituted heteroaryl;

R 9 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

each R 10 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R 10 together with the atoms to which they are attached form a substituted or unsubstituted heterocycle;

p is 1, 2 or 3; and

r is 0-8.

[00148] In some embodiments, a PA inhibitor is a compound having the structure of

Formula XXVIII, or pharmaceutically acceptable salt or N-oxide thereof:

Formula XXVIII

wherein:

R 1 and R 2 are each independently H, halogen, CN, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy;

R 3 is H, -OH, -OR 6 , -SR 6 , -S(=0) 2 R 7 , -C0 2 R 8 , N(R 8 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl;

is H or substituted or unsubstituted alkyl;

R 7 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl;

each R 8 is independently H, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl or substituted or unsubstituted heteroaryl, or two R together with the nitrogen to which they are attached form a substituted or unsubstituted heterocycle;

each A is independently N or C-R 4 ;

each R 4 is independently H, halogen, CN, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy;

ring B is aryl or heteroaryl subsituted with R 5 ;

each R 5 is independently halogen, -CN, -N0 2 , -OH, -OR 6 , -SR 6 , -S(=0)R 7 , S(=0) 2 R 7 , - NHS(=0) 2 R 7 , -C(=0)R 7 , -OC(=0)R 7 , -C0 2 R 8 , N(R 8 ) 2 , C(=0)N(R 8 ) 2 , -NHC(=0)R 7 ,

NHC(=0)OR 7 , -NHC(=0)N(R 8 ) 2 , substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocycloalkyl;

n is 1-8;

R 9 and R 10 are each independently H, halogen, or substituted or unsubstituted alkyl;

p is 1-5; and

R 11 is H or substituted or unsubstituted alkyl.

[00149] In some embodiments, a PA inhibitor is a compound of Formula XXIX:

Formula XXIX

wherein:

R 6 is H, halo, hydroxy, cyano, substituted or unsubstituted alkyl, or substituted or unsubstituted alkoxy,

R 7 is substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted alkylamino, C(=O)-N(R 10 ) 2 , C(=0)-0(R 10 ), S(O) m -N(R 10 ) 2 , N(R 10 ) 2 C(=O)R 10 , OC(=0)(R 10 ), N(R 10 ) 2 S(O) m R 10 , substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted heterocycloalkyl; wherein each R 10 is independently H, substituted or unsubstituted alkyl; substituted or unsubstituted cycloalkyl, or substituted or unsubstituted alkylcyclo alkyl; and m is 1-2;

R 8 is H, halo, hydroxy, cyano, substituted or unsubstituted alkyl, substituted or unsubstituted alkoxy, substituted or unsubstituted alkylamino, C(=O)-N(R 10 ) 2 , C(=0)-0(R 10 ), S(O) m -N(R 10 ) 2 , N(R 10 ) 2 C(=O)R 10 , OC(=0)(R 10 ), N(R 10 ) 2 S(O) m R 10 ;

R 9 is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted heterocycloalkyl;

Q 7 , Q 8 are independently N or C-R 6 ;

X is O, N-R 11 or C(R n ) 2 , wherein each R 11 is independently H, hydroxy, substituted or unsubstituted alkyl; or two R 11 taken together are (=0) or (=NR 12 ); wherein R 12 is H, hydroxy, substituted or unsubstituted alkyl, or substituted or unsubstituted alkoxy;

8 7 7 8 6 9

provided that when Q is N, Q is CH, R , R are alkoxy, and R is cyano, R is not 2,4- dichloroanilino;

or a pharmaceutically acceptable salt thereof.

[00150] In some embodiments, PAK inhibitors suitable for methods described herein include compounds described in International Appl. No. PCT/US09/68855, U.S. Provisional Appl. Nos. 61/353,624, 61/353,622, 61/353,619, 61/353,085, 61/353,054, and 61/352,985; PAK inhibitor compounds described therein are incorporated herein by reference.

[00151] In some embodiments, PAK inhibitors described herein include, by way of example, N 1 -(5-(2-(3,4,5-trimethoxyphenylamino)pyrimidin-4-yl)pyr idin-2-yl)ethane-1,2- diamine, N 1 -(5 -(2-(4-(4-methylpiperazin- 1 -yl)phenylamino)pyrimidin-4-yl)pyridin-2-yl)ethane- 1 ,2-diamine, N-(4-(4-methylpiperazin- 1 -yl)phenyl)-4-(6-(2-(piperidin- 1 -yl)ethylamino)pyridin- 3-yl)pyrimidin-2-amine, 2-(4-(4-methylpiperazin- 1 -yl)phenylamino)-8-(2- (trifluoromethylthio)benzyl)pyrido[2,3- ]pyrimidin-7(SH)-one, 2-(4-(4-methylpiperazin-1- yl)phenylamino)-8-( 1 ,2,3 ,4-tetrahydronaphthalen- 1 -yl)pyrido [2,3- ]pyrimidin-7(SH)-one, 6- (2,6-dichlorophenyl)-8-methoxy-2-(4-(4-methylpiperazin-1-yl) phenylamino)pyrido[2,3- ]pyrimidin-7(SH)-one, 8-cyclopentyl-2-(4-(4-methylpiperazin-1-yl)phenylamino)pyrid o[2,3- ]pyrimidin-7(SH)-one, 4-(2-chloro-4-methylphenylamino)-6-methoxy-7-(3-(4-methylpip erazin- 1 -yl)propoxy)quinoline-3-carbonitrile, (5)-N-(2-(dimethylamino)- 1 -phenylethyl)-6,6-dimethyl- 3-(thieno[2,3-d]pyrimidin-4-ylamm^ 4- (2,4-dichlorophenylamino)-6-methoxy-7-(3 -(4-methylpiperazin- 1 -yl)propoxy)quino line-3 - carbonitrile or the like.

[00152] In some embodiments, PAK inhibitors include (5)-1-(4-benzyl-6-((5-cyclopropyl- iH-pyrazol-3-yl)methyl)pyrimidin-2-yl)azetidine-2-carboxamid e, (S)-2-(3,5-difluorophenyl)-4- (piperidin-3-ylamino)thieno[3,2-c]pyridine-7-carboxamide, or the like.

[00153] In certain instances, PAK inhibitors also include, e.g., compounds described in

U.S. Patents 5,863,532, 6,191,169, 6,248,549, and 6,498,163; U.S. Patent Applications

200200045564, 20020086390, 20020106690, 20020142325, 20030124107, 20030166623, 20040091992, 20040102623, 20040208880, 200500203114, 20050037965, 20050080002, and 20050233965, 20060088897; EP Patent Publication 1492871; PCT patent publication WO 9902701; PCT patent publication WO 2008/047307; Kumar et al, (2006), Nat. Rev. Cancer, 6:459; and Eswaran et al, (2007), Structure, 15:201-213, all of which are incorporated herein by reference for disclosure of kinase inhibitors and PAK inhibitors therein.

[00154] In certain instances, small molecule PAK inhibitors include BMS-387032; SNS- 032; CHI4-258; TKI-258; EKB-569; JNJ-7706621; PKC-412; staurosporine; SU-14813;

sunitinib; N-(3-chloro-4-fluoro-phenyl)-7-methoxy-6-(3-morpholin-4-ylpr opoxy)quinazolin-4- amine (gefitinib), VX-680; MK-0457; combinations thereof; or salts, prodrugs thereof.

[00155] In some embodiments, the PAK inhibitor is a polypeptide comprising an amino acid sequence about 80% to about 100% identical, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical the following amino acid sequence:

HT1HVGFDAVTGEFTGMPEQWARLLQTSNITKSEQKKNPQAVLDVLEFYNSKKTSNSQ KYMSFTDKS [00156] The above sequence corresponds to the PAK auto inhibitory domain (PAD) polypeptide amino acids 83-149 of PAKl polypeptide as described in, e.g., Zhao et al (1998). In some embodiments, the PAK inhibitor is a fusion protein comprising the above-described PAD amino acid sequence. In some embodiments, in order to facilitate cell penetration the fusion polypeptide (e.g., N-terminal or C-terminal) further comprises a polybasic protein transduction domain (PTD) amino acid sequence, e.g.: RKKR QR ; YARAAA QARA; THRLPRRRRRR; or GGRRARRRRRR.

[00157] In some embodiments, in order to enhance uptake into the brain, the fusion polypeptide further comprises a human insulin receptor antibody as described in U.S. Patent Application Serial No. 11/245,546.

[00158] In some embodiments, the PAK inhibitor is peptide inhibitor comprising a sequence at least 60% to 100%, e.g., 65%, 70%, 75%, 80%, 85%, 90%, 92%, 93%, 95%, 96%, 97%), 98%o, 99%), or any other percent from about 60%> to about 100% identical the following amino acid sequence: PPVIAPREHTKSVYTRS as described in, e.g., Zhao et al (2006), Nat Neurosci, 9(2):234-242. In some embodiments, the peptide sequence further comprises a PTD amino acid sequence as described above.

[00159] In some embodiments, the PAK inhibitor is a polypeptide comprising an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical to the FMRP1 protein (GenBank Accession No. Q06787), where the polypeptide is able to bind with a PAK (for example, PAKl, PAK2, PAK3, PAK4, PAK5and/or PAK6). In some embodiments, the PAK inhibitor is a polypeptide comprising an amino acid sequence at least 80%> to 100%), e.g., 85%, 90%>, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical to the FMRP1 protein (GenBank Accession No. Q06787), where the polypeptide is able to bind with a Group I PAK, such as, for example PAKl (see, e.g., Hayashi et al (2007), Proc Natl Acad Sci USA, 104(27): 11489-11494. In some embodiments, the PAK inhibitor is a polypeptide comprising a fragment of human FMRP1 protein with an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical to the sequence of amino acids 207-425 of the human FMRP1 protein (i.e., comprising the KHl and KH2 domains), where the polypeptide is able to bind to PAKl .

[00160] In some embodiments, the PAK inhibitor comprises a polypeptide comprising an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical to at least five, at least ten at least twenty, at least thirty, at least forty, at least fifty, at least sixty, at least seventy, at least eighty, at least ninety contiguous amino acids of the huntingtin (htt) protein (GenBank

Accession No. NP 002102, gi 90903231), where the polypeptide is able to bind to a Group 1 PAK (for example, PAK1, PAK2, and/or PAK3). In some embodiments, the PAK inhibitor comprises a polypeptide comprising an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical to at least a portion of the huntingtin (htt) protein (GenBank Accession No. NP 002102, gi 90903231), where the polypeptide is able to bind to PAKl . In some embodiments, the PAK inhibitor is a polypeptide comprising a fragment of human huntingtin protein with an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%), or any other percent from about 80%> to about 100% identical to a sequence of at least five, at least ten, at least twenty, at least thirty, at least forty, at least fifty, at least sixty, at least seventy, at least eighty, at least ninety, or at least 100 contiguous amino acids of the human huntingtin protein that is outside of the sequence encoded by exon 1 of the htt gene (i.e., a fragment that does not contain poly glutamate domains), where the polypeptide binds a PAK. In some embodiments, the PAK inhibitor is a polypeptide comprising a fragment of human huntingtin protein with an amino acid sequence at least 80% identical to a sequence of the human huntingtin protein that is outside of the sequence encoded by exon 1 of the htt gene (i.e., a fragment that does not contain poly glutamate domains), where the polypeptide binds PAKl .

Upstream regulators of p21 activated kinases

[00161] In certain embodiments, an indirect PAK modulator (e.g., an indirect PAK inhibitor) affects the activity of a molecule that acts in a signaling pathway upstream of PAK (upstream regulators of PAK). Upstream effectors of PAK include, but are not limited to: TrkB receptors; NMDA receptors; EphB receptors; adenosine receptors; estrogen receptors; integrins; FMRP; Rho-family GTPases, including Cdc42, Rac (including but not limited to Racl and Rac2), CDK5, PI3 kinases, NCK, PDK1, EKT, GRB2, Chp, TC10, Tel, and Wrch-1 ; guanine nucleotide exchange factors ("GEFs"), such as but not limited to GEFT, members of the Dbl family of GEFs, p21 -activated kinase interacting exchange factor (PIX), DEF6, Zizimin 1, Vavl, Vav2, Dbs, members of the DOCK180 family, Kalirin-7, and Tiaml ; G protein-coupled receptor kinase-interacting protein 1 (GIT1), CIB1, filamin A, Etk/Bmx, and sphingosine.

[00162] Modulators of NMDA receptor include, but are not limited to, 1- aminoadamantane, dextromethorphan, dextrorphan, ibogaine, ketamine, nitrous oxide, phencyclidine, riluzole, tiletamine, memantine, neramexane, dizocilpine, aptiganel, remacimide, 7-chlorokynurenate, DCKA (5,7-dichlorokynurenic acid), kynurenic acid, 1- aminocyclopropanecarboxylic acid (ACPC), AP7 (2-amino-7-phosphonoheptanoic acid), APV (R-2-amino-5-phosphonopentanoate), CPPene (3-[(R)-2-carboxypiperazin-4-yl]-prop-2-enyl- 1 - phosphonic acid); (+)-(lS, 2S)-1-(4-hydroxy-phenyl)-2-(4-hydroxy-4-phenylpiperidino)-1- pro- panol; (IS, 2S)- 1 -(4-hydroxy-3-methoxyphenyl)-2-(4-hydroxy-4-phenylpiperi-din o)- 1 -propanol; (3R, 4S)-3-(4-(4-fluorophenyl)-4-hydroxypiperidin-1-yl-)-chroman- 4,7-diol; (1R*, 2R*)-1-(4- hydroxy-3-methylphenyl)-2-(4-(4-fiuoro-phenyl)-4-hydroxypipe ridin- 1 -yl)-propan- 1 -ol- mesylate; and/or combinations thereof.

[00163] Modulators of estrogen receptors include, and are not limited to, PPT (4,4',4"-(4-

Propyl-[1H]-pyrazole-1,3 ,5-triyl)trisphenol); SKF-82958 (6-chloro-7,8-dihydroxy-3-allyl-1- phenyl-2,3,4,5-tetrahydro-1H-3-benzazepine); estrogen; estradiol; estradiol derivatives, including but not limited to 17-® estradiol, estrone, estriol, ERP-131, phytoestrogen, MK 101 (bioNovo); VG-1010 (bioNovo); DPN (diarylpropiolitrile); ERB-041; WAY-202196; WAY- 214156; genistein; estrogen; estradiol; estradiol derivatives, including but not limited to 17-® estradiol, estrone, estriol, benzopyrans and triazolo-tetrahydrofluorenones, disclosed in U.S. Patent No. 7,279,499, and Parker et al, Bioorg. & Med. Chem. Ltrs. 16: 4652-4656 (2006), each of which is incorporated herein by reference for such disclosure.

[00164] Modulators of TrkB include by way of example, neutorophic factors including

BDNF and GDNF. Modulators of EphB include XL647 (Exelixis), EphB modulator compounds described in WO/2006081418 and US Appl. Pub. No. 20080300245, incorporated herein by reference for such disclosure, or the like.

[00165] Modulators of integrins include by way of example, ATN-161, PF-04605412,

MEDI— 522, Volociximab, natalizumab, Volociximab, Ro 27-2771, Ro 27-2441, etaracizumab, CNTO-95, JSM6427, cilengitide, R411 (Roche), EMD 121974, integrin antagonist compounds described in J. Med. Chem., 2002, 45 (16), pp 3451-3457, incorporated herein by reference for such disclosure, or the like.

[00166] Adenosine receptor modulators include, by way of example, theophylline, 8-

Cyclopentyl-1,3-dimethylxanthine (CPX), 8-Cyclopentyl-1,3-dipropylxanthine (DPCPX), 8- Phenyl-1,3-dipropylxanthine, PSB 36, istradefylline, SCH-58261, SCH-442,416, ZM-241,385, CVT-6883, MRS-1706, MRS-1754, PSB-603, PSB-0788, PSB-1115, MRS-1191, MRS-1220, MRS-1334, MRS-1523, MRS-3777, MRE3008F20, PSB-10, PSB-11, VUF-5574, N6-

Cyclopentyladenosine, CCPA, 2'-MeCCPA, GR 79236, SDZ WAG 99, ATL-146e, CGS-21680, Regadenoson, 5 * -N-ethylcarboxamidoadenosine, BAY 60-6583, LUF-5835, LUF-5845, 2-(l- Hexynyl)-N-methyladenosine, CF-101 (IB-MECA), 2-Cl-IB-MECA, CP-532,903, MRS-3558, Rosuvastatin, KW-3902, SLV320, mefloquine, regadenoson, or the like. [00167] In some embodiments, compounds reducing PAK levels decrease PAK transcription or translation or reduce RNA or protein levels. In some embodiments, a compound that decreases PAK levels is an upstream effector of PAK. In some embodiments, exogenous expression of the activated forms of the Rho family GTPases Chp and cdc42 in cells leads to increased activation of PAK while at the same time increasing turnover of the PAK protein, significantly lowering its level in the cell (Hubsman et al. (2007) Biochem. J. 404: 487-497). PAK clearance agents include agents that increase expression of one or more Rho family GTPases and/or one or more guanine nucleotide exchange factors (GEFs) that regulate the activity of Rho family GTPases, in which overexpression of a Rho family GTPase and/or a GEF results in lower levels of PAK protein in cells. PAK clearance agents also include agonists of Rho family GTPases, as well as agonists of GTP exchange factors that activate Rho family GTPases, such as but not limited to agonists of GEFs of the Dbl family that activate Rho family GTPases.

[00168] Overexpression of a Rho family GTPase is optionally by means of introducing a nucleic acid expression construct into the cells or by administering a compound that induces transcription of the endogenous gene encoding the GTPase. In some embodiments, the Rho family GTPase is Rac (e.g., Racl, Rac2, or Rac3), cdc42, Chp, TC10, Tel, or Wrnch-1. For example, a Rho family GTPase includes Racl, Rac2, Rac3, or cdc42. A gene introduced into cells that encodes a Rho family GTPase optionally encodes a mutant form of the gene, for example, a more active form (for example, a constitutively active form, Hubsman et al. (2007) Biochem. J. 404: 487-497). In some embodiments, a PAK clearance agent is, for example, a nucleic acid encoding a Rho family GTPase, in which the Rho family GTPase is expressed from a constitutive or inducible promoter. PAK levels in some embodiments are reduced by a compound that directly or indirectly enhances expression of an endogenous gene encoding a Rho family GTPase.

[00169] In some embodiments, the PAK inhibitor is a compound that directly or indirectly decreases the activation or activity of the upstream effectors of PAK. For example, in some embodiments a compound that inhibits the GTPase activity of the small Rho-family GTPases such as Rac and cdc42 thereby reduce the activation of PAK kinase. In some embodiments, the compound that decreases PAK activation is by secramine that inhibits cdc42 activation, binding to membranes and GTP in the cell (Pelish et al. (2005) Nat. Chem. Biol. 2: 39-46). In some embodiments, PAK activation is decreased by EHT 1864, a small molecule that inhibits Racl, Rac lb, Rac2 and Rac3 function by preventing binding to guanine nucleotide association and engagement with downstream effectors (Shutes et al. (2007) J. Biol. Chem. 49: 35666-35678). In some embodiments, PAK activation is also decreased by the NSC23766 small molecule that binds directly to Racl and prevents its activation by Rac-specific RhoGEFs (Gao et al. (2004) Proc. Natl. Acad. Sci. U.S.A. 101 : 7618-7623). In some embodiments, PAK activation is also decreased by the 16 kDa fragment of prolactin (16k PRL), generated from the cleavage of the 23 kDa prolactin hormone by matrix metalloproteases and cathepsin D in various tissues and cell types. 16k PRL down-regulates the Ras-Tiaml-Racl-Pakl signaling pathway by reducing Racl activation in response to cell stimuli such as wounding (Lee et al. (2007) Cancer Res 67: 11045- 11053). In some embodiments, PAK activation is decreased by inhibition of NMD A and/or AMPA receptors. Examples of modulators of AMPA receptors include and are not limited to CNQX (6-cyano-7-nitroquinoxaline-2,3-dione); NBQX (2,3-dihydroxy-6-nitro-7-sulfamoyl- benzo[f]quinoxaline-2,3-dione); DNQX (6,7-dinitroquinoxaline-2,3-dione); kynurenic acid; 2,3- dihydroxy-6-nitro-7-sulfamoylbenzo-[f]quinoxaline quinoxaline or AMPAkines. Examples of modulators of NMD A receptors include and are not limited to ketamine, MK801, memantine, PCP or the like. In some embodiments, PAK activation is decreased by inhibition of TrkB activation. In some embodiments, PAK activation is decreased by inhibition of BDNF activation of TrkB. In some embodiments, the PAK inhibitor is an antibody to BDNF. In some

embodiments, PAK activation is decreased by inhibition of TrkB receptors; NMD A receptors; EphB receptors; adenosine receptors; estrogen receptors; integrins; Rho-family GTPases, including Cdc42, Rac (including but not limited to Racl and Rac2), CDK5, PI3 kinases, NCK, PDK1, EKT, GRB2, Chp, TC10, Tel, and Wrch-1 ; guanine nucleotide exchange factors

("GEFs"), such as but not limited to GEFT, members of the Dbl family of GEFs, p21 -activated kinase interacting exchange factor (PIX), DEF6, Zizimin 1, Vavl, Vav2, Dbs, members of the DOCK180 family, Kalirin-7, and Tiaml ; G protein-coupled receptor kinase-interacting protein 1 (GIT1), CIB1, filamin A, Etk/Bmx, and/or binding to FMRP and/or sphingosine.

[00170] In some embodiments a compound that decreases PAK levels in the cell is a compound that directly or indirectly increases the activity of a guanine exchange factor (GEF) that promotes the active state of a Rho family GTPase, such as an agonist of a GEF that activates a Rho family GTPase, such as but not limited to, Rac or cdc42. Activation of GEFs is also effected by compounds that activate TrkB, NMD A, or EphB receptors.

[00171] In some embodiments, a PAK clearance agent is a nucleic acid encoding a GEF that activates a Rho family GTPase, in which the GEF is expressed from a constitutive or inducible promoter. In some embodiments, a guanine nucleotide exchange factor (GEF), such as but not limited to a GEF that activates a Rho family GTPase is overexpressed in cells to increase the activation level of one or more Rho family GTPases and thereby lower the level of PAK in cells. GEFs include, for example, members of the Dbl family of GTPases, such as but not limited to, GEFT, PIX (e.g., alphaPIX, betaPIX), DEF6, Zizimin 1, Vavl, Vav2, Dbs, members of the DOCK180 family, hPEM-2, FLJ00018, kalirin, Tiaml, STEF, DOCK2, DOCK6, DOCK7, DOCK9, Asf, EhGEF3, or GEF-1. In some embodiments, PAK levels are also reduced by a compound that directly or indirectly enhances expression of an endogenous gene encoding a GEF. A GEF expressed from a nucleic acid construct introduced into cells is in some embodiments a mutant GEF, for example a mutant having enhanced activity with respect to wild type.

[00172] The clearance agent is optionally a bacterial toxin such as Salmonella

typhinmurium toxin SpoE that acts as a GEF to promote cdc42 nucleotide exchange (Buchwald et al. (2002) EMBO J. 21 : 3286-3295; Schlumberger et al. (2003) J. Biological Chem. 278: 27149-27159). Toxins such as SopE, fragments thereof, or peptides or polypeptides having an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%), or any other percent from about 80%> to about 100%) identical to a sequence of at least five, at least ten, at least twenty, at least thirty, at least forty, at least fifty, at least sixty, at least seventy, at least eighty, at least ninety, or at least 100 contiguous amino acids of the toxin are also optionally used as downregulators of PAK activity. The toxin is optionally produced in cells from nucleic acid constructs introduced into cells.

Modulators of upstream regulators of PAKs

[00173] In some embodiments, a modulator of an upstream regulator of PAKs is an indirect inhibitor of PAK. In certain instances, a modulator of an upstream regulator of PAKs is a modulator of PDK1. In some instances, a modulator of PDK1 reduces or inhibits the activity of PDK1. In some instances a PDK1 inhibitor is an antisense compound (e.g., any PDK1 inhibitor described in U.S. Patent No. 6,124,272, which PDK1 inhibitor is incorporated herein by reference). In some instances, a PDK1 inhibitor is a compound described in e.g., U.S. Patent Nos. 7,344,870, and 7,041,687, which PDK1 inhibitors are incorporated herein by reference. In some embodiments, an indirect inhibitor of PAK is a modulator of a PI3 kinase. In some instances a modulator of a P13 kinase is a PI3 kinase inhibitor. In some instances, a PI3 kinase inhibitor is an antisense compound (e.g., any PI3 kinase inhibitor described in WO

2001/018023, which PI3 kinase inhibitors are incorporated herein by reference). In some instances, an inhibitor of a PI3 kinase is 3-morpholino-5-phenylnaphthalen-l(4H)-one

(LY294002), or a peptide based covalent conjugate of LY294002, (e.g., SF1126, Semaphore pharmaceuticals). In certain embodiments, an indirect inhibitor of PAK is a modulator of Cdc42. In certain embodiments, a modulator of Cdc42 is an inhibitor of Cdc42. In certain embodiments, a Cdc42 inhibitor is an antisense compound (e.g., any Cdc42 inhibitor described in U.S. Patent No. 6,410,323, which Cdc42 inhibitors are incorporated herein by reference). In some instances, an indirect inhibitor of PAK is a modulator of GRB2. In some instances, a modulator of GRB2 is an inhibitor of GRB2. In some instances a GRB2 inhibitor is a GRb2 inhibitor described in e.g., U.S. Patent No. 7,229,960, which GRB2 inhibitor is incorporated by reference herein. In certain embodiments, an indirect inhibitor of PAK is a modulator of NCK. In certain

embodiments, an indirect inhibitor of PAK is a modulator of ETK In some instances, a modulator of ETK is an inhibitor of ETK. In some instances an ETK inhibitor is a compound e.g., a-Cyano-(3,5-di-t-butyl-4-hydroxy)thiocinnamide (AG 879).

[00174] In some embodiments the PAK inhibitors, binding molecules, and clearance agents provided herein are administered to an individual suffering from a neurological condition to alleviate, halt or delay the loss of dendritic spine density in an individual. A pharmacological composition comprising a therapeutically effective amount of at least one of the compounds disclosed herein, including: a PAK transcription inhibitor, a PAK clearance agent, an agent that binds PAK to prevent its interaction with one or more cellular or extracellular proteins, and a PAK antagonist. In some specific embodiments, the pharmacological composition comprises a therapeutically effective amount of at least one of the compounds chosen from the group consisting of: a PAK transcription inhibitor, PAK clearance agent, an agent that binds a PAK to prevent its interaction with one or more cellular proteins, and a PAK antagonist. An individual is an animal, and is preferably a mammal, preferably human.

[00175] In other methods PAK inhibitors, binding molecules, and clearance agents provided herein are administered to an individual suffering from a neurological condition to reverse some or all defects in dendritic spine morphology, spine size, spine motility and/or spine plasticity in a subject having, or suspected of having a neurological condition. The method includes: administering to an individual a pharmacological composition comprising a therapeutically effective amount of at least one of the compounds chosen from the group consisting of: a PAK transcription inhibitor, a PAK clearance agent, an agent that binds PAK to prevent its interaction with one or more cellular or extracellular proteins, and a PAK antagonist. In some specific embodiments, the pharmacological composition comprises a therapeutically effective amount of at least one of the compounds chosen from the group consisting of: a Group 1 PAK transcription inhibitor, a Group 1 PAK clearance agent, an agent that binds a Group 1 PAK to prevent its interaction with one or more cellular proteins, and a Group 1 PAK

antagonist. An individual is an animal, and is preferably a mammal, preferably human. [00176] In some embodiments, indirect PAK inhibitors act by decreasing transcription and/or translation of PAK. A PAK inhibitor, in some embodiments, decreases transcription and/or translation of a PAK. For example, in some embodiments, modulation of PAK transcription or translation occurs through the administration of specific or non-specific inhibitors of PAK transcription or translation. In some embodiments, proteins or non-protein factors that bind the upstream region of the PAK gene or the 5 ' UTR of a PAK mRNA are assayed for their affect on transcription or translation using transcription and translation assays (see, for example, Baker, et al. (2003) J. Biol. Chem. 278: 17876-17884; Jiang et al. (2006) J. Chromatography A 1133: 83-94; Novoa et al. (1997) Biochemistry 36: 7802-7809; Brandi et al. (2007) Methods Enzymol. 431 : 229-267). PAK inhibitors include DNA or RNA binding proteins or factors that reduce the level of transcription or translation or modified versions thereof. In other embodiments, a PAK inhibitor is a modified form (e.g., mutant form or chemically modified form) of a protein or other compound that positively regulates transcription or translation of PAK, in which the modified form reduces transcription or translation of PAK. In yet other embodiments, a transcription or translation inhibitor is an antagonist of a protein or compound that positively regulates transcription or translation of PAK, or is an agonist of a protein that represses transcription or translation.

[00177] Regions of a gene other than those upstream of the transcriptional start site and regions of an mRNA other than the 5' UTR (such as but not limited to regions 3' of the gene or in the 3 ' UTR of an mRNA, or regions within intron sequences of either a gene or mRNA) also include sequences to which effectors of transcription, translation, mRNA processing, mRNA transport, and mRNA stability bind. In some embodiments, a PAK inhibitor is a clearance agent comprising a polypeptide having homology to an endogenous protein that affects mRNA processing, transport, or stability, or is an antagonist or agonist of one or more proteins that affect mRNA processing, transport, or turnover, such that the inhibitor reduces the expression of PAK protein by interfering with PAK mRNA transport or processing, or by reducing the half- life of PAK mRNA. In some embodiments, PAK clearance agents interfere with transport or processing of a PAK mRNA, or by reducing the half- life of a PAK mRNA.

[00178] For example, PAK clearance agents decrease RNA and/or protein half-life of a PAK isoform, for example, by directly affecting mRNA and/or protein stability. In certain embodiments, PAK clearance agents cause PAK mRNA and/or protein to be more accessible and/or susceptible to nucleases, proteases, and/or the proteasome. In some embodiments, PAK inhibitors decrease the processing of PAK mRNA thereby reducing PAK activity. For example, PAK inhibitors function at the level of pre-mRNA splicing, 5' end formation (e.g. capping), 3' end processing (e.g. cleavage and/or polyadenylation), nuclear export, and/or association with the translational machinery and/or ribosomes in the cytoplasm. In some embodiments, PAK inhibitors cause a decrease in the level of PAK mRNA and/or protein, the half-life of PAK mRNA and/or protein by at least about 5%, at least about 10%, at least about 20%, at least about 30%), at least about 40%>, at least about 50%>, at least about 60%>, at least about 80%>, at least about 90%), at least about 95%, or substantially 100%.

[00179] In some embodiments, the PAK inhibitor is a clearance agent that comprises one or more RNAi or antisense oligonucleotides directed against one or more PAK isoform RNAs. In some embodiments, the PAK inhibitor comprises one or more ribozymes directed against one or more PAK isoform RNAs. The design, synthesis, and use of RNAi constructs, antisense oligonucleotides, and ribozymes are found, for example, in Dykxhoorn et al. (2003) Nat. Rev. Mol. Cell. Biol. 4: 457-467; Hannon et al. (2004) Nature 431 : 371-378; Sarver et al. (1990) Science 247: 1222-1225; Been et al. (1986) Cell 47:207-216) . In some embodiments, nucleic acid constructs that induce triple helical structures are also introduced into cells to inhibit transcription of the PAK gene (Helene (1991) Anticancer Drug Des. 6:569-584).

[00180] For example, a PAK inhibitor that is a clearance agent is in some embodiments an RNAi molecule or a nucleic acid construct that produces an RNAi molecule. An RNAi molecule comprises a double-stranded RNA of at least about seventeen bases having a 2-3 nucleotide single-stranded overhangs on each end of the double-stranded structure, in which one strand of the double-stranded RNA is substantially complementary to the target PAK RNA molecule whose downregulation is desired. "Substantially complementary" means that one or more nucleotides within the double-stranded region are not complementary to the opposite strand nucleotide(s). Tolerance of mismatches is optionally assessed for individual RNAi structures based on their ability to downregulate the target RNA or protein. In some

embodiments, RNAi is introduced into the cells as one or more short hairpin RNAs ("shRNAs") or as one or more DNA constructs that are transcribed to produce one or more shRNAs, in which the shRNAs are processed within the cell to produce one or more RNAi molecules.

[00181] Nucleic acid constructs for the expression of siRNA, shRNA, antisense RNA, ribozymes, or nucleic acids for generating triple helical structures are optionally introduced as RNA molecules or as recombinant DNA constructs. DNA constructs for reducing gene expression are optionally designed so that the desired RNA molecules are expressed in the cell from a promoter that is transcriptionally active in mammalian cells, such as, for example, the SV40 promoter, the human cytomegalovirus immediate-early promoter (CMV promoter), or the pol III and/or pol II promoter using known methods. For some purposes, it is desirable to use viral or plasmid-based nucleic acid constructs. Viral constructs include but are not limited to retroviral constructs, lentiviral constructs, or based on a pox virus, a herpes simplex virus, an adenovirus, or an adeno-associated virus (AAV).

[00182] In other embodiments, a PAK inhibitor is a polypeptide that decreases the activity of PAK. In some embodiments, a PAK inhibitor is a polypeptide that decreases the activity of a PAK. Protein and peptide inhibitors of PAK are optionally based on natural substrates of PAK, e.g., Myosin light chain kinase (MLCK), regulatory Myosin light chain (R-MLC), Myosins I heavy chain, myosin II heavy chain, Myosin VI, Caldesmon, Desmin, Opl8/stathmin, Merlin, Filamin A, LIM kinase (LIMK), cortactin, cofilin, Ras, Raf, Mek, p47(phox), BAD, caspase 3, estrogen and/or progesterone receptors, NET1, Gaz, phosphoglycerate mutase-B, RhoGDI, prolactin, p41Arc, cortactin and/or Aurora- A. In some embodiments, a PAK inhibitor is based on a sequence of PAK itself, for example, the autoinhibitory domain in the N-terminal portion of the PAK protein that binds the catalytic domain of a partner PAK molecule when the PAK molecule is in its homodimeric state (Zhao et al. (1998) Mol. Cell Biol. 18:2153-2163; Knaus et al. (1998) J. Biol. Chem. 273: 21512-21518; Hofman et al. (2004) J. Cell Sci. 117: 4343-4354). In some embodiments, polypeptide inhibitors of PAK comprise peptide mimetics, in which the peptide has binding characteristics similar to a natural binding partner or substrate of PAK.

[00183] In some embodiments, provided herein are compounds that downregulate PAK protein level. In some embodiments, the compounds described herein activate or increase the activity of an upstream regulator or downstream target of PAK. In some embodiments, compounds described herein downregulate protein level of a PAK. In some instances compounds described herein reduce at least one of the symptoms related a neurological condition by reducing the amount of PAK in a cell. In some embodiments a compound that decreases PAK protein levels in cells also decreases the activity of PAK in the cells. In some embodiments a compound that decreases PAK protein levels does not have a substantial impact on PAK activity in cells. In some embodiments a compound that increases PAK activity in cells decreases PAK protein levels in the cells.

[00184] In some embodiments, a compound that decreases the amount of PAK protein in cells decreases transcription and/or translation of PAK or increases the turnover rate of PAK mRNA or protein by modulating the activity of an upstream effector or downstream regulator of PAK. In some embodiments, PAK expression or PAK levels are influenced by feedback regulation based on the conformation, chemical modification, binding status, or activity of PAK itself. In some embodiments, PAK expression or PAK levels are influenced by feedback regulation based on the conformation, chemical modification, binding status, or activity of molecules directly or indirectly acted on by PAK signaling pathways. As used herein "binding status" refers to any or a combination of whether PAK, an upstream regulator of PAK, or a downstream effector of PAK is in a monomeric state or in an oligomeric complex with itself, or whether it is bound to other polypeptides or molecules. For example, a downstream target of PAK, when phosphorylated by PAK, in some embodiments directly or indirectly downregulates PAK expression or decrease the half- life of PAK mRNA or protein. Downstream targets of PAK include but are not limited to: Myosin light chain kinase (MLCK), regulatory Myosin light chain (R-MLC), Myosins I heavy chain, myosin II heavy chain, Myosin VI, Caldesmon, Desmin, Opl8/stathmin, Merlin, Filamin A, LIM kinase (LIMK), Ras, Raf, Mek, p47 phox , BAD, caspase 3, estrogen and/or progesterone receptors, NET1, Gaz, phosphoglycerate mutase-B, RhoGDI, prolactin, p41 Arc , cortactin and/or Aurora-A. Downregulators of PAK levels include downstream targets of PAK or fragments thereof in a phosphorylated state and downstream targets of PAK or fragments thereof in a hyperphosphorylated state.

[00185] A fragment of a downstream target of PAK includes any fragment with an amino acid sequence at least 80% to 100%, e.g., 85%, 90%, 92%, 93%, 95%, 96%, 97%, 98%, 99%, or any other percent from about 80% to about 100% identical to a sequence of at least five, at least ten, at least twenty, at least thirty, at least forty, at least fifty, at least sixty, at least seventy, at least eighty, at least ninety, or at least 100 contiguous amino acids of the downstream regulator, in which the fragment of the downstream target of PAK is able to downregulate PAK mRNA or protein expression or increase turnover of PAK mRNA or protein. In some embodiments, the fragment of a downstream regulator of PAK comprises a sequence that includes a

phosphorylation site recognized by PAK, in which the site is phosphorylated.

[00186] In some embodiments, a compound that decreases the level of PAK includes a peptide, polypeptide, or small molecule that inhibits dephosphorylation of a downstream target of PAK, such that phosphorylation of the downstream target remains at a level that leads to downregulation of PAK levels.

[00187] In some embodiments, PAK activity is reduced or inhibited via activation and/or inhibition of an upstream regulator and/or downstream target of PAK. In some embodiments, the protein expression of a PAK is downregulated. In some embodiments, the amount of PAK in a cell is decreased. In some embodiments a compound that decreases PAK protein levels in cells also decreases the activity of PAK in the cells. In some embodiments a compound that decreases PAK protein levels does not decrease PAK activity in cells. In some embodiments a compound that increases PAK activity in cells decreases PAK protein levels in the cells. [00188] In some embodiments, a PA inhibitor is a small molecule. As referred to herein, a "small molecule" is an organic molecule that is less than about 5 kilodaltons (kDa) in size. In some embodiments, the small molecule is less than about 4 kDa, 3 kDa, about 2 kDa, or about 1 kDa. In some embodiments, the small molecule is less than about 800 daltons (Da), about 600 Da, about 500 Da, about 400 Da, about 300 Da, about 200 Da, or about 100 Da. In some embodiments, a small molecule is less than about 4000 g/mol, less than about 3000g/mol, 2000 g/mol, less than about 1500 g/mol, less than about 1000 g/mol, less than about 800 g/mol, or less than about 500 g/mol. In some embodiments, small molecules are non-polymeric. Typically, small molecules are not proteins, polypeptides, polynucleotides, oligonucleotides,

polysaccharides, glycoproteins, or proteoglycans, but include peptides of up to about 40 amino acids. A derivative of a small molecule refers to a molecule that shares the same structural core as the original small molecule, but which is prepared by a series of chemical reactions from the original small molecule. As one example, a pro-drug of a small molecule is a derivative of that small molecule. An analog of a small molecule refers to a molecule that shares the same or similar structural core as the original small molecule, and which is synthesized by a similar or related route, or art-recognized variation, as the original small molecule.

[00189] In certain embodiments, compounds described herein have one or more chiral centers. As such, all stereoisomers are envisioned herein. In various embodiments, compounds described herein are present in optically active or racemic forms. It is to be understood that the compounds described herein encompass racemic, optically-active, regioisomeric and

stereoisomeric forms, or combinations thereof that possess the therapeutically useful properties described herein. Preparation of optically active forms is achieve in any suitable manner, including by way of non-limiting example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically- active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase. In some embodiments, mixtures of one or more isomer are utilized as the therapeutic compound described herein. In certain embodiments, compounds described herein contain one or more chiral centers. These

compounds are prepared by any means, including enantioselective synthesis and/or separation of a mixture of enantiomers and/or diastereomers. Resolution of compounds and isomers thereof is achieved by any means including, by way of non-limiting example, chemical processes, enzymatic processes, fractional crystallization, distillation, chromatography, and the like.

[00190] In various embodiments, pharmaceutically acceptable salts described herein include, by way of non-limiting example, a nitrate, chloride, bromide, phosphate, sulfate, acetate, hexafluorophosphate, citrate, gluconate, benzoate, propionate, butyrate, subsalicylate, maleate, laurate, malate, fumarate, succinate, tartrate, amsonate, pamoate, p-tolunenesulfonate, mesylate and the like. Furthermore, pharmaceutically acceptable salts include, by way of non- limiting example, alkaline earth metal salts (e.g., calcium or magnesium), alkali metal salts (e.g., sodium-dependent or potassium), ammonium salts and the like.

[00191] The compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein and as described, for example, in Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplemental (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, ADVANCED ORGANIC CHEMISTRY 4 th Ed., (Wiley 1992); Carey and Sundberg, ADVANCED ORGANIC CHEMISTRY 4 th Ed., Vols. A and B (Plenum 2000, 2001), and Green and Wuts, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS 3 rd Ed., (Wiley 1999) (all of which are

incorporated by reference for such disclosure). General methods for the preparation of compound as described herein are modified by the use of appropriate reagents and conditions, for the introduction of the various moieties found in the formulae as provided herein. As a guide the following synthetic methods are utilized.

[00192] Compounds described herein are synthesized starting from compounds that are available from commercial sources or that are prepared using procedures outlined herein.

Formation of Covalent Linkages by Reaction of an Electrophile with a Nucleophile

[00193] The compounds described herein are modified using various electrophiles and/or nucleophiles to form new functional groups or substituents. Table A entitled "Examples of Covalent Linkages and Precursors Thereof lists selected non-limiting examples of covalent linkages and precursor functional groups which yield the covalent linkages. Table A is used as guidance toward the variety of electrophiles and nucleophiles combinations available that provide covalent linakges. Precursor functional groups are shown as electrophilic groups and nucleophilic groups. Table A: Examples of Covalent Linkages and Precursors Thereof

Co ilcni l .inkimc Product Uccirophilo Vi loophile

Amidines imido esters amines/anilines

Ureas Isocyanates amines/anilines

Urethanes Isocyanates alcohols/phenols

Thioureas isothiocyanates amines/anilines

Thio ethers Maleimides Thiols

Phosphite esters pho sphoramidites Alcohols

Silyl ethers silyl halides Alcohols

Alkyl amines sulfonate esters amines/anilines

Thio ethers sulfonate esters Thiols

Esters sulfonate esters carboxylic acids

Ethers sulfonate esters Alcohols

Sulfonamides sulfonyl halides amines/anilines

Sulfonate esters sulfonyl halides phenols/alcohols

Use of Protecting Groups

[00194] In the reactions described, it is necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, in order to avoid their unwanted participation in reactions. Protecting groups are used to block some or all of the reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed. In some embodiments it is

contemplated that each protective group be removable by a different means. Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal.

[00195] In some embodiments, protective groups are removed by acid, base, reducing conditions (such as, for example, hydro geno lysis), and/or oxidative conditions. Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and Fmoc groups, which are base labile.

Carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl in the presence of amines blocked with acid labile groups such as t-butyl carbamate or with carbamates that are both acid and base stable but hydro lytically removable. [00196] In some embodiments carboxylic acid and hydroxy reactive moieties are blocked with hydro lytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc. Carboxylic acid reactive moieties are protected by conversion to simple ester compounds as exemplified herein, which include conversion to alkyl esters, or are blocked with oxidatively- removable protective groups such as 2,4-dimethoxybenzyl, while co-existing amino groups are blocked with fluoride labile silyl carbamates.

[00197] Allyl blocking groups are useful in the presence of acid- and base- protecting groups since the former are stable and are subsequently removed by metal or pi-acid catalysts. For example, an allyl-blocked carboxylic acid is deprotected with a Pd°-catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups. Yet another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and does not react. Once released from the resin, the functional group is available to react.

[00198] Typically blocking/protecting groups are selected from:

PMB trityl acetyl Wmm

[00199] Other protecting groups, plus a detailed description of techniques applicable to the creation of protecting groups and their removal are described in Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, NY, 1999, and

Kocienski, Protective Groups, Thieme Verlag, New York, NY, 1994, which are incorporated herein by reference for such disclosure. Certain Definitions

[00200] As used herein the term "Treatment", "treat", or "treating" includes achieving a therapeutic benefit and/or a prophylactic benefit. Therapeutic benefit is meant to include eradication or amelioration of the underlying disorder or condition being treated. For example, in an individual with Huntington's disease, therapeutic benefit includes alleviation or partial and/or complete halting of the progression of the disease, or partial or complete reversal of the disease. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological or psychological symptoms associated with the underlying condition such that an improvement is observed in the patient, notwithstanding the fact that the patient is still affected by the condition. For example, in an individual suffering from epilepsy, therapeutic benefit includes alleviation or partial and/or complete halting of seizures, or reduction in frequency of seizures. A prophylactic benefit of treatment includes prevention of a condition, retarding the progress of a condition, or decreasing the likelihood of occurrence of a condition. As used herein, "treat", "treating" or "treatment" includes prophylaxis.

[00201] As used herein, the phrase "neurological condition" refers to any condition that has its origin in some part of an individual's nervous system and results in chronic impairment in cognition, affect, and/or motor function. Example of neurological conditions include and are not limited to Parkinson's disease, Huntington's disease, substance abuse and substance dependency, Neurofibromatosis I, Neurofibromatosis II, tuberous sclerosis, depression, Maple Syrup Disease, Niemann Picks disease, encephalitis, Down's syndrome, mental retardation syndromes other than Down's syndrome such as Fetal alcohol syndrome, Klinefelter's syndrome, congenital hypothyroidism, Williams syndrome, Smith-Lemli-Opitz syndrome, Prader-willi syndrome, Phelan-McDermid syndrome, Mowat- Wilson syndrome, Ciliopathy, Lowe syndrome, epilepsy, bipolar disorder, mania, anxiety disorder, postratumatic stress disorder, schizoaffective disorder, schizophreniform, cognitive decline associated with menopause, Angelman syndrome, obsessive compulsive disorder, panic disorder, phobias, aneroxia nervosa, bulimia nervosa, or any other neurological condition.

[00202] As used herein, the phrase "abnormal spine size" refers to dendritic spine volumes or dendritic spine surface areas (e.g., volumes or surface areas of the spine heads and/or spine necks) associated with neurological conditions that deviate significantly relative to spine volumes or surface areas in the same brain region (e.g., the CA1 region, the prefrontal cortex) in a normal individual (e.g., a mouse, rat, or human) of the same age; such abnormalities are determined as appropriate, by methods including, e.g., tissue samples, relevant animal models, post-mortem analyses, or other model systems. [00203] The phrase "defective dendritic spine morphology" or "abnormal dendritic spine morphology" or "aberrant dendritic spine morphology" refers to abnormal dendritic spine shapes, volumes, surface areas, length, width (e.g., diameter of the neck), spine head diameter, spine head volume, spine head surface area, spine density, ratio of mature to immature spines, ratio of spine volume to spine length, or the like that is associated with neurological conditions relative to the dendritic spine shapes, volumes, surface areas, length, width (e.g., diameter of the neck), spine density, ratio of mature to immature spines, ratio of spine volume to spine length, or the like observed in the same brain region in a normal individual (e.g., a mouse, rat, or human) of the same age; such abnormalities or defects are determined as appropriate, by methods including, e.g., tissue samples, relevant animal models, post-mortem analyses, or other model systems.

[00204] The phrase "abnormal spine function" or "defective spine function" or "aberrant spine function" refers to a defect of dendritic spines to undergo stimulus-dependent

morphological or functional changes (e.g., following activation of AMPA and/or NMDA receptors, LTP, LTD, etc) associated with neurological conditions as compared to dendritic spines in the same brain region in a normal individual of the same age. The "defect" in spine function includes, e.g., a reduction in dendritic spine plasticity, (e.g., an abnormally small change in dendritic spine morphology or actin re-arrangement in the dendritic spine), or an excess level of dendritic plasticity, (e.g., an abnormally large change in dendritic spine morphology or actin re-arrangement in the dendritic spine). Such abnormalities or defects are determined as appropriate, by methods including, e.g., tissue samples, relevant animal models, post-mortem analyses, or other model systems.

[00205] The phrase "abnormal spine motility" refers to a significant low or high movement of dendritic spines associated with neurological conditions as compared to dendritic spines in the same brain region in a normal individual of the same age. Any defect in spine morphology (e.g., spine length, density or the like) or synaptic plasticity or synaptic function (e.g., LTP, LTD or the like) or spine motility occurs in any region of the brain, including, for example, the frontal cortex, the hippocampus, the amygdala, the CA1 region, the prefrontal cortex or the like. Such abnormalities or defects are determined as appropriate, by methods including, e.g., tissue samples, relevant animal models, post-mortem analyses, or other model systems.

[00206] As used herein, the phrase "biologically active" refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological effect on that organism is considered to be biologically active. In particular embodiments, where a protein or polypeptide is biologically active, a portion of that protein or polypeptide that shares at least one biological activity of the protein or polypeptide is typically referred to as a "biologically active" portion.

[00207] As used herein, the term "subcortical dementia" refers to symptoms related to Huntington's disease (e.g., deficits in executive functions such as planning, cognitive flexibility, abstract thinking, rule acquisition, initiating appropriate actions, inhibiting inappropriate actions; memory deficits such as short-term memory deficits, long-term memory difficulties, deficits in episodic (memory of one's life), procedural (memory of the body of how to perform an activity) and working memory, and the like). In some instances, "progression toward dementia" is identified, monitored or diagnosed by neuropsychological or behavioral testing. In other instances, "progression toward dementia" is identified, monitored or diagnosed by neuroimaging or brain scans.

[00208] As used herein, the term "effective amount" is an amount, which when administered systemically, is sufficient to effect beneficial or desired results, such as beneficial or desired clinical results, or enhanced cognition, memory, mood, or other desired effects. An effective amount is also an amount that produces a prophylactic effect, e.g., an amount that delays, reduces, or eliminates the appearance of a pathological or undesired condition associated with neurological conditions. An effective amount is optionally administered in one or more administrations. In terms of treatment, an "effective amount" of a composition described herein is an amount that is sufficient to palliate, alleviate, ameliorate, stabilize, reverse or slow the progression of neurological conditions, e.g., cognitive decline toward dementia, mental retardation or the like. An "effective amount" includes any PAK inhibitor used alone or in conjunction with one or more agents used to treat a disease or disorder. An "effective amount" of a therapeutic agent as described herein will be determined by a patient's attending physician or other medical care provider. Factors which influence what a therapeutically effective amount will be include, the absorption profile (e.g., its rate of uptake into the brain) of the PAK inhibitor, time elapsed since the initiation of disease, and the age, physical condition, existence of other disease states, and nutritional status of an individual being treated. Additionally, other medication the patient is receiving, e.g., antidepressant drugs used in combination with a PAK inhibitor, will typically affect the determination of the therapeutically effective amount of the therapeutic agent to be administered.

[00209] As used herein, the term "inhibitor" refers to a molecule which is capable of inhibiting (including partially inhibiting or allosteric inhibition) one or more of the biological activities of a target molecule, e.g., a p21-activated kinase. Inhibitors, for example, act by reducing or suppressing the activity of a target molecule and/or reducing or suppressing signal transduction. In some embodiments, a PAK inhibitor described herein causes substantially complete inhibition of one or more PAKs. In some embodiments, the phrase "partial inhibitor" refers to a molecule which can induce a partial response for example, by partially reducing or suppressing the activity of a target molecule and/or partially reducing or suppressing signal transduction. In some instances, a partial inhibitor mimics the spatial arrangement, electronic properties, or some other physico chemical and/or biological property of the inhibitor. In some instances, in the presence of elevated levels of an inhibitor, a partial inhibitor competes with the inhibitor for occupancy of the target molecule and provides a reduction in efficacy, relative to the inhibitor alone. In some embodiments, a PAK inhibitor described herein is a partial inhibitor of one or more PAKs. In some embodiments, a PAK inhibitor described herein is an allosteric modulator of PAK. In some embodiments, a PAK inhibitor described herein blocks the p21 binding domain of PAK. In some embodiments, a PAK inhibitor described herein blocks the ATP binding site of PAK. In some embodiments, a PAK inhibitor is a "Type Π" kinase inhibitor. In some embodiment a PAK inhibitor stabilizes PAK in its inactive conformation. In some embodiments, a PAK inhibitor stabilizes the "DFG-out" conformation of PAK.

[00210] In some embodiments, PAK inhibitors reduce, abolish, and/or remove the binding between PAK and at least one of its natural binding partners (e.g., Cdc42 or Rac). In some instances, binding between PAK and at least one of its natural binding partners is stronger in the absence of a PAK inhibitor (by e.g., 90%, 80%, 70%, 60%, 50%, 40%, 30% or 20%) than in the presence of a PAK inhibitor. Alternatively or additionally, PAK inhibitors inhibit the phosphotransferase activity of PAK, e.g., by binding directly to the catalytic site or by altering the conformation of PAK such that the catalytic site becomes inaccessible to substrates. In some embodiments, PAK inhibitors inhibit the ability of PAK to phosphorylate at least one of its target substrates, e.g., LIM kinase 1 (LIMK1), myosin light chain kinase (MLCK), myosin light chain, cortactin; or itself. PAK inhibitors include inorganic and/or organic compounds.

[00211] In some embodiments, PAK inhibitors described herein (e.g., compounds of

Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase dendritic spine length. In some embodiments, PA inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) decrease dendritic spine length. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase dendritic neck diameter. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) decrease dendritic neck diameter. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase dendritic spine head diameter. In some

embodiments, PA inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase dendritic spine head volume. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula

XXIX) decrease dendritic spine head volume. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase dendritic spine surface area. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula

IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) decrease dendritic spine surface area. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula

XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase dendritic spine density. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula

X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) decrease dendritic spine density. In some embodiments, PA inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) increase the number of mushroom shaped spines. In some embodiments, PAK inhibitors described herein (e.g., compounds of Formula I, compounds of Formula II, compounds of Formula III, compounds of Formula IV, compounds of Formula V, compounds of Formula VI, compounds of Formula VII, compounds of Formula VIII, compounds of Formula IX, compounds of Formula X, compounds of Formula XI, compounds of Formula XII, compounds of Formula XIII, compounds of Formula XIV, compounds of Formula XV, compounds of Formula XVI, compounds of Formula XVII, compounds of Formula XVIII, compounds of Formula XIX, compounds of Formula XX, compounds of Formula XXI, compounds of Formula XXII, compounds of Formula XXIII, compounds of Formula XXIV, compounds of Formula XXV, compounds of Formula XXVI, compounds of Formula XXVII, compounds of Formula XXVIII, or compounds of Formula XXIX) decrease the number of mushroom shaped spines.

[00212] In some embodiments, a PAK inhibitor suitable for the methods described herein is a direct PAK inhibitor. In some embodiments, a PAK inhibitor suitable for the methods described herein is an indirect PAK inhibitor. In some embodiments, a PAK inhibitor suitable for the methods described herein decreases PAK activity relative to a basal level of PAK activity by about 1.1 fold to about 100 fold, e.g., to about 1.2 fold, 1.5 fold, 1.6 fold, 1.7 fold, 2.0 fold,

3.0 fold, 5.0 fold, 6.0 fold, 7.0 fold, 8.5 fold, 9.7 fold, 10 fold, 12 fold, 14 fold, 15 fold, 20 fold, 30 fold, 40 fold, 50 fold, 60 fold, 70 fold, 90 fold, 95 fold, or by any other amount from about

1.1 fold to about 100 fold relative to basal PAK activity. In some embodiments, the PAK inhibitor is a reversible PAK inhibitor. In other embodiments, the PAK inhibitor is an irreversible PAK inhibitor. Direct PA inhibitors are optionally used for the manufacture of a medicament for treating a neurological condition.

[00213] In some embodiments, a PAK inhibitor used for the methods described herein has in vitro ED 50 for PAK activation of less than 100 μΜ (e.g., less than 10 μΜ, less than 5 μΜ, less than 4 μΜ, less than 3 μΜ, less than 1 μΜ, less than 0.8 μΜ, less than 0.6 μΜ, less than 0.5 μΜ, less than 0.4 μΜ, less than 0.3 μΜ, less than less than 0.2 μΜ, less than 0.1 μΜ, less than 0.08 μΜ, less than 0.06 μΜ, less than 0.05 μΜ, less than 0.04 μΜ, less than 0.03 μΜ, less than less than 0.02 μΜ, less than 0.01 μΜ, less than 0.0099 μΜ, less than 0.0098 μΜ, less than 0.0097 μΜ, less than 0.0096 μΜ, less than 0.0095 μΜ, less than 0.0094 μΜ, less than 0.0093 μΜ, less than 0.00092, or less than 0.0090 μΜ).

[00214] As used herein, synaptic function refers to synaptic transmission and/or synaptic plasticity, including stabilization of synaptic plasticity. As used herein, "defect in synaptic plasticity" or "aberrant synaptic plasticity" refers to abnormal synaptic plasticity following stimulation of that synapse. In some embodiments, a defect in synaptic plasticity is a decrease in LTP. In some embodiments, a defect in synaptic plasticity is an increase in LTD. In some embodiments, a defect in synaptic plasticity is erratic (e.g., fluctuating, randomly increasing or decreasing) synaptic plasticity. In some instances, measures of synaptic plasticity are LTP and/or LTD (induced, for example, by theta-burst stimulation, high-frequency stimulation for LTP, low- frequency (1 Hz) stimulation for LTD) and LTP and/or LTD after stabilization. In some embodiments, stabilization of LTP and/or LTD occurs in any region of the brain including the frontal cortex, the hippocampus, the prefrontal cortex, the amygdala or any combination thereof.

[00215] As used herein "stabilization of synaptic plasticity" refers to stable LTP or LTD following induction (e.g., by theta-burst stimulation, high-frequency stimulation for LTP, low- frequency (1 Hz) stimulation for LTD).

[00216] "Aberrant stabilization of synaptic transmission" (for example, aberrant stabilization of LTP or LTD), refers to failure to establish a stable baseline of synaptic transmission following an induction paradigm (e.g., by theta-burst stimulation high-frequency stimulation for LTP, low- frequency (1 Hz) stimulation for LTD) or an extended period of vulnerability to disruption by pharmacological or electrophysiological means

[00217] As used herein "synaptic transmission" or "baseline synaptic transmission" refers to the EPSP and/or IPSP amplitude and frequency, neuronal excitability or population spike thresholds of a normal individual (e.g., an individual not suffering from a neurological condition) or that predicted for an animal model for a normal individual. As used herein "aberrant synaptic transmission" or "defective synaptic transmission" refers to any deviation in synaptic transmission compared to synaptic transmission of a normal individual or that predicted for an animal model for a normal individual. In some embodiments, an individual suffering from a neurological condition has a defect in baseline synaptic transmission that is a decrease in baseline synaptic transmission compared to the baseline synaptic transmission in a normal individual or that predicted for an animal model for a normal individual. In some embodiments, an individual suffering from a neurological condition has a defect in baseline synaptic transmission that is an increase in baseline synaptic transmission compared to the baseline synaptic transmission in a normal individual or that predicted for an animal model for a normal individual.

[00218] As used herein, "normalization of aberrant synaptic plasticity" refers to a change in aberrant synaptic plasticity in an individual suffering from, suspected of having, or predisposed to a neurological condition to a level of synaptic plasticity that is substantially the same as the synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual. As used herein, substantially the same means, for example, about 90% to about 110% of the measured synaptic plasticity in a normal individual or to that predicted from an animal model for a normal individual. In other embodiments, substantially the same means, for example, about 80% to about 120% of the measured synaptic plasticity in a normal individual or to that predicted from an animal model for a normal individual. In yet other embodiments, substantially the same means, for example, about 70% to about 130% of the synaptic plasticity in a normal individual or to that predicted from an animal model for a normal individual. As used herein, "partial normalization of aberrant synaptic plasticity" refers to any change in aberrant synaptic plasticity in an individual suffering from, suspected of having, or pre-disposed to a neurological condition that trends towards synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual. As used herein "partially normalized synaptic plasticity" or "partially normal synaptic plasticity" is, for example, ± about 25%, ± about 35%, ± about 45%, ± about 55%, ± about 65%, or ± about 75% of the synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant synaptic plasticity in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is lowering of aberrant synaptic plasticity where the aberrant synaptic plasticity is higher than the synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant synaptic plasticity in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is an increase in aberrant synaptic plasticity where the aberrant synaptic plasticity is lower than the synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of synaptic plasticity in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from an erratic (e.g., fluctuating, randomly increasing or decreasing) synaptic plasticity to a normal (e.g. stable) or partially normal (e.g., less fluctuating) synaptic plasticity compared to the synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of synaptic plasticity in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from a non-stabilizing synaptic plasticity to a normal (e.g., stable) or partially normal (e.g., partially stable) synaptic plasticity compared to the synaptic plasticity of a normal individual or to that predicted from an animal model for a normal individual.

[00219] As used herein, "normalization of aberrant baseline synaptic transmission" refers to a change in aberrant baseline synaptic transmission in an individual suffering from, suspected of having, or pre-disposed to a neurological condition to a level of baseline synaptic

transmission that is substantially the same as the baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual. As used herein, substantially the same means, for example, about 90% to about 110% of the measured baseline synaptic transmission in a normal individual or to that predicted from an animal model for a normal individual. In other embodiments, substantially the same means, for example, about 80% to about 120% of the measured baseline synaptic transmission in a normal individual or to that predicted from an animal model for a normal individual. In yet other embodiments, substantially the same means, for example, about 70% to about 130% of the measured baseline synaptic transmission in a normal individual or to that predicted from an animal model for a normal individual. As used herein, "partial normalization of aberrant baseline synaptic transmission" refers to any change in aberrant baseline synaptic transmission in an individual suffering from, suspected of having, or pre-disposed to a neurological condition that trends towards baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual. As used herein "partially normalized baseline synaptic transmission" or

"partially normal baseline synaptic transmission" is, for example, ± about 25%, ± about 35%, ± about 45%), ± about 55%, ± about 65%, or ± about 75% of the measured baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant baseline synaptic transmission in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is lowering of aberrant baseline synaptic transmission where the aberrant baseline synaptic transmission is higher than the baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual. In some

embodiments, normalization or partial normalization of aberrant baseline synaptic transmission in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is an increase in aberrant baseline synaptic transmission where the aberrant baseline synaptic transmission is lower than the baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of baseline synaptic transmission in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from an erratic (e.g., fluctuating, randomly increasing or decreasing) baseline synaptic transmission to a normal (e.g. stable) or partially normal (e.g., less fluctuating) baseline synaptic transmission compared to the baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant baseline synaptic transmission in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from a non-stabilizing baseline synaptic transmission to a normal (e.g., stable) or partially normal (e.g., partially stable) baseline synaptic transmission compared to the baseline synaptic transmission of a normal individual or to that predicted from an animal model for a normal individual.

[00220] As used herein, "normalization of aberrant synaptic function" refers to a change in aberrant synaptic function in an individual suffering from, suspected of having, or predisposed to a neurological condition to a level of synaptic function that is substantially the same as the synaptic function of a normal individual or to that predicted from an animal model for a normal individual. As used herein, substantially the same means, for example, about 90% to about 110% of the synaptic function in a normal individual or to that predicted from an animal model for a normal individual. In other embodiments, substantially the same means, for example, about 80% to about 120% of the synaptic function in a normal individual or to that predicted from an animal model for a normal individual. In yet other embodiments, substantially the same means, for example, about 70% to about 130% of the synaptic function in a normal individual or to that predicted from an animal model for a normal individual. As used herein, "partial normalization of aberrant synaptic function" refers to any change in aberrant synaptic function in an individual suffering from, suspected of having, or pre-disposed to a neurological condition that trends towards synaptic function of a normal individual or to that predicted from an animal model for a normal individual. As used herein "partially normalized synaptic function" or "partially normal synaptic function" is, for example, ± about 25%, ± about 35%, ± about 45%), ± about 55%, ± about 65%, or ± about 75% of the measured synaptic function of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant synaptic function in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is lowering of aberrant synaptic function where the aberrant synaptic function is higher than the synaptic function of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant synaptic function in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is an increase in aberrant synaptic function where the aberrant synaptic function is lower than the synaptic function of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of synaptic function in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from an erratic (e.g., fluctuating, randomly increasing or decreasing) synaptic function to a normal (e.g. stable) or partially normal (e.g., less fluctuating) synaptic function compared to the synaptic function of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant synaptic function in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from a non-stabilizing synaptic function to a normal (e.g., stable) or partially normal (e.g., partially stable) synaptic function compared to the synaptic function of a normal individual or to that predicted from an animal model for a normal individual.

[00221] As used herein, "normalization of aberrant long term potentiation (LTP)" refers to a change in aberrant LTP in an individual suffering from, suspected of having, or predisposed to a neurological condition to a level of LTP that is substantially the same as the LTP of a normal individual or to that predicted from an animal model for a normal individual. As used herein, substantially the same means, for example, about 90% to about 110% of the LTP in a normal individual or to that predicted from an animal model for a normal individual. In other embodiments, substantially the same means, for example, about 80% to about 120% of the LTP in a normal individual or to that predicted from an animal model for a normal individual. In yet other embodiments, substantially the same means, for example, about 70% to about 130% of the LTP in a normal individual or to that predicted from an animal model for a normal individual. As used herein, "partial normalization of aberrant LTP" refers to any change in aberrant LTP in an individual suffering from, suspected of having, or pre-disposed to a neurological condition that trends towards LTP of a normal individual or to that predicted from an animal model for a normal individual. As used herein "partially normalized LTP" or "partially normal LTP" is, for example, ± about 25%, ± about 35%, ± about 45%, ± about 55%, ± about 65%, or ± about 75% of the measured LTP of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant LTP in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is lowering of aberrant LTP where the aberrant LTP is higher than the LTP of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant LTP in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is an increase in aberrant LTP where the aberrant LTP is lower than the LTP of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of LTP in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from an erratic (e.g., fluctuating, randomly increasing or decreasing) LTP to a normal (e.g. stable) or partially normal (e.g., less fluctuating) LTP compared to the LTP of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant LTP in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from a non-stabilizing LTP to a normal (e.g., stable) or partially normal (e.g., partially stable) LTP compared to the LTP of a normal individual or to that predicted from an animal model for a normal individual.

[00222] As used herein, "normalization of aberrant long term depression (LTD)" refers to a change in aberrant LTD in an individual suffering from, suspected of having, or pre-disposed to a neurological condition to a level of LTD that is substantially the same as the LTD of a normal individual or to that predicted from an animal model for a normal individual. As used herein, substantially the same means, for example, about 90% to about 110% of the LTD in a normal individual or to that predicted from an animal model for a normal individual. In other embodiments, substantially the same means, for example, about 80% to about 120% of the LTD in a normal individual or to that predicted from an animal model for a normal individual. In yet other embodiments, substantially the same means, for example, about 70% to about 130% of the LTD in a normal individual or to that predicted from an animal model for a normal individual. As used herein, "partial normalization of aberrant LTD" refers to any change in aberrant LTD in an individual suffering from, suspected of having, or pre-disposed to a neurological condition that trends towards LTD of a normal individual or to that predicted from an animal model for a normal individual. As used herein "partially normalized LTD" or "partially normal LTD" is, for example, ± about 25%, ± about 35%, ± about 45%, ± about 55%, ± about 65%, or ± about 75% of the measured LTD of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant LTD in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is lowering of aberrant LTD where the aberrant LTD is higher than the LTD of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant LTD in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is an increase in aberrant LTD where the aberrant LTD is lower than the LTD of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of LTD in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from an erratic (e.g., fluctuating, randomly increasing or decreasing) LTD to a normal (e.g. stable) or partially normal (e.g., less fluctuating) LTD compared to the LTD of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant LTD in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from a non-stabilizing LTD to a normal (e.g., stable) or partially normal (e.g., partially stable) LTD compared to the LTD of a normal individual or to that predicted from an animal model for a normal individual.

[00223] As used herein, "normalization of aberrant sensorimotor gating" refers to a change in aberrant sensorimotor gating in an individual suffering from, suspected of having, or pre-disposed to a neurological condition to a level of sensorimotor gating that is substantially the same as the sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual. As used herein, substantially the same means, for example, about 90% to about 110% of the sensorimotor gating in a normal individual or to that predicted from an animal model for a normal individual. In other embodiments, substantially the same means, for example, about 80% to about 120% of the sensorimotor gating in a normal individual or to that predicted from an animal model for a normal individual. In yet other embodiments, substantially the same means, for example, about 70% to about 130% of the sensorimotor gating in a normal individual or to that predicted from an animal model for a normal individual. As used herein, "partial normalization of aberrant sensorimotor gating" refers to any change in aberrant sensorimotor gating in an individual suffering from, suspected of having, or pre- disposed to a neurological condition that trends towards sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual. As used herein "partially normalized sensorimotor gating" or "partially normal sensorimotor gating" is, for example, ± about 25%, ± about 35%, ± about 45%, ± about 55%, ± about 65%, or ± about 75%, of the measured sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant sensorimotor gating in an individual suffering from, suspected of having, or predisposed to a neurological condition is lowering of aberrant sensorimotor gating where the aberrant sensorimotor gating is higher than the sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments,

normalization or partial normalization of aberrant sensorimotor gating in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is an increase in aberrant sensorimotor gating where the aberrant sensorimotor gating is lower than the sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of sensorimotor gating in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from an erratic (e.g., fluctuating, randomly increasing or decreasing) sensorimotor gating to a normal (e.g. stable) or partially normal (e.g., less fluctuating) sensorimotor gating compared to the sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual. In some embodiments, normalization or partial normalization of aberrant sensorimotor gating in an individual suffering from, suspected of having, or pre-disposed to a neurological condition is a change from a non-stabilizing sensorimotor gating to a normal (e.g., stable) or partially normal (e.g., partially stable) sensorimotor gating compared to the sensorimotor gating of a normal individual or to that predicted from an animal model for a normal individual.

[00224] As used herein, "expression" of a nucleic acid sequence refers to one or more of the following events: (1) production of an R A template from a DNA sequence (e.g., by transcription); (2) processing of an RNA transcript (e.g., by splicing, editing, 5' cap formation, and/or 3' end formation); (3) translation of an RNA into a polypeptide or protein; (4) post- translational modification of a polypeptide or protein.

[00225] As used herein the term "PAK polypeptide" or "PAK protein" or "PAK" refers to a protein that belongs in the family of p21 -activated serine/threonine protein kinases. These include mammalian iso forms of PAK, e.g., the Group I PAK proteins (sometimes referred to as Group A PAK proteins), including PAK1, PAK2, PAK3, as well as the Group II PAK proteins (sometimes referred to as Group B PAK proteins), including PAK4, PAK5, and/or PAK6 Also included as PAK polypeptides or PAK proteins are lower eukaryotic isoforms, such as the yeast Ste20 (Leberter et al, 1992, EMBO J., 11 :4805; incorporated herein by reference) and/or the Dictyostelium single-headed myosin I heavy chain kinases (Wu et al, 1996, J. Biol. Chem., 271 :31787; incorporated herein by reference). Representative examples of PAK amino acid sequences include, but are not limited to, human PAK1 (GenBank Accession Number

AAA65441), human PAK2 (GenBank Accession Number AAA65442), human PAK3 (GenBank Accession Number AAC36097), human PAK 4 (GenBank Accession Numbers NP 005875 and CAA09820), human PAK5 (GenBank Accession Numbers CAC 18720 and BAA94194), human PAK6 (GenBank Accession Numbers NP 064553 and AAF82800), human PAK7 (GenBank Accession Number Q9P286), C. elegans PAK (GenBank Accession Number BAA11844), D. melanogaster PAK (GenBank Accession Number AAC47094), and rat PAKl (GenBank Accession Number AAB95646). In some embodiments, a PAK polypeptide comprises an amino acid sequence that is at least 70% to 100% identical, e.g., at least 75%, 80%, 85%, 86%, 87%, 88%, 90%, 91%, 92%, 94%, 95%, 96%, 97%, 98%, or any other percent from about 70% to about 100% identical to sequences of GenBank Accession Numbers AAA65441, AAA65442, AAC36097, NP_005875, CAA09820, CAC18720, BAA94194, NP_064553, AAF82800, Q9P286, BAA11844, AAC47094, and/or AAB95646. In some embodiments, a Group I PAK polypeptide comprises an amino acid sequence that is at least 70%> to 100% identical, e.g., at least 75%, 80%, 85%, 86%, 87%, 88%, 90%, 91%, 92%, 94%, 95%, 96%, 97%, 98%, or any other percent from about 70% to about 100% identical to sequences of GenBank Accession Numbers AAA65441, AAA65442, and/or AAC36097.

[00226] Representative examples of PAK genes encoding PAK proteins include, but are not limited to, human PAKl (GenBank Accession Number U24152), human PAK2 (GenBank Accession Number U24153), human PAK3 (GenBank Accession Number AF068864), human PAK4 (GenBank Accession Number AJOl 1855), human PAK5 (GenBank Accession Number AB040812), and human PAK6 (GenBank Accession Number AF276893). In some

embodiments, a PAK gene comprises a nucleotide sequence that is at least 70%> to 100% identical, e.g., at least 75%, 80%, 85%, 86%, 87%, 88%, 90%, 91%, 92%, 94%, 95%, 96%, 97%), 98%o, or any other percent from about 70%> to about 100%) identical to sequences of GenBank Accession Numbers U24152, U24153, AF068864, AJOl 1855, AB040812, and/or AF276893. In some embodiments, a Group I PAK gene comprises a nucleotide sequence that is at least 70% to 100% identical, e.g., at least 75%, 80%, 85%, 86%, 87%, 88%, 90%, 91%, 92%, 94%, 95%, 96%, 97%, 98%, or any other percent from about 70% to about 100% identical to sequences of GenBank Accession Numbers U24152, U24153, and/or AF068864.

[00227] To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the

corresponding position in the second sequence, then the molecules are identical at that position. The percent homology between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = # of identical positions/total # of positions (e.g., overlapping positions) x 100). In one embodiment the two sequences are the same length.

[00228] To determine percent homology between two sequences, the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-2268, modified as in Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877 is used. Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990) J. Mol. Biol. 215:403-410. BLAST nucleotide searches are performed with the NBLAST program, score=100, wordlength=12 to obtain nucleotide sequences homologous to a nucleic acid molecules described or disclose herein. BLAST protein searches are performed with the

XBLAST program, score=50, wordlength=3. To obtain gapped alignments for comparison purposes, Gapped BLAST is utilized as described in Altschul et al. (1997) Nucleic Acids Res. 25:3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) are used. See the website of the National Center for Biotechnology Information for further details (on the World Wide Web at

ncbi.nlm.nih.gov). Proteins suitable for use in the methods described herein also includes proteins having between 1 to 15 amino acid changes, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acid substitutions, deletions, or additions, compared to the amino acid sequence of any protein PAK inhibitor described herein. In other embodiments, the altered amino acid sequence is at least 75% identical, e.g., 77%, 80%, 82%, 85%, 88%, 90%, 92%, 95%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of any protein PAK inhibitor described herein. Such sequence-variant proteins are suitable for the methods described herein as long as the altered amino acid sequence retains sufficient biological activity to be functional in the compositions and methods described herein. Where amino acid substitutions are made, the substitutions should be conservative amino acid substitutions. Among the common amino acids, for example, a "conservative amino acid substitution" is illustrated by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine. The

BLOSUM62 table is an amino acid substitution matrix derived from about 2,000 local multiple alignments of protein sequence segments, representing highly conserved regions of more than 500 groups of related proteins (Henikoff ei al (1992), Proc. Natl Acad. Sci. USA, 89: 10915- 10919). Accordingly, the BLOSUM62 substitution frequencies are used to define conservative amino acid substitutions that may be introduced into the amino acid sequences described or described herein. Although it is possible to design amino acid substitutions based solely upon chemical properties (as discussed above), the language "conservative amino acid substitution" preferably refers to a substitution represented by a BLOSUM62 value of greater than -1. For example, an amino acid substitution is conservative if the substitution is characterized by a BLOSUM62 value of 0, 1, 2, or 3. According to this system, preferred conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 1 (e.g., 1, 2 or 3), while more preferred conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 2 (e.g., 2 or 3).

[00229] As used herein, the term "PAK activity," unless otherwise specified, includes, but is not limited to, at least one of PAK protein-protein interactions, PAK phosphotransferase activity (intermolecular or intermolecular), translocation, etc. of one or more PAK iso forms.

[00230] As used herein, a "PAK inhibitor" refers to any molecule, compound, or composition that directly or indirectly decreases the PAK activity. In some embodiments, PAK inhibitors inhibit, decrease, and/or abolish the level of a PAK mR A and/or protein or the half- life of PAK mRNA and/or protein, such inhibitors are referred to as "clearance agents". In some embodiments, a PAK inhibitor is a PAK antagonist that inhibits, decreases, and/or abolishes an activity of PAK. In some embodiments, a PAK inhibitor also disrupts, inhibits, or abolishes the interaction between PAK and its natural binding partners (e.g., a substrate for a PAK kinase, a Rac protein, a cdc42 protein, LIM kinase) or a protein that is a binding partner of PAK in a pathological condition, as measured using standard methods. In some embodiments, the PAK inhibitor is a Group I PAK inhibitor that inhibits, for example, one or more Group I PAK polypeptides, for example, PAK1, PAK2, and/or PAK3. In some embodiments, the PAK inhibitor is a PAK1 inhibitor. In some embodiments, the PAK inhibitor is a PAK2 inhibitor. In some embodiments, the PAK inhibitor is a PAK3 inhibitor. In some embodiments, the PAK inhibitor is a mixed PAK1/PAK3 inhibitor. In some embodiments, the PAK inhibitor inhibits all three Group I PAK iso forms (PAK1, PAK2 and PAK3) with equal or similar potency. In some embodiments, the PAK inhibitor is a Group II PAK inhibitor that inhibits one or more Group II PAK polypeptides, for example PAK4, PAK5, and/or PAK6. In some embodiments, the PAK inhibitor is a PAK4 inhibitor. In some embodiments, the PAK inhibitor is a PAK5 inhibitor. In some embodiments, the PAK inhibitor is a PAK6 inhibitor. In some embodiments, the PAK inhibitor is a PAK7 inhibitor. As used herein, a PAK5 polypeptide is substantially homologous to a PAK7 polypeptide.

[00231] In some embodiments, PAK inhibitors reduce, abolish, and/or remove the binding between PAK and at least one of its natural binding partners (e.g., Cdc42 or Rac). In some instances, binding between PAK and at least one of its natural binding partners is stronger in the absence of a PAK inhibitor (by e.g., 90%, 80%, 70%, 60%, 50%, 40%, 30% or 20%) than in the presence of a PAK inhibitor. In some embodiments, PAK inhibitors prevent, reduce, or abolish binding between PAK and a protein that abnormally accumulates or aggregates in cells or tissue in a disease state. In some instances, binding between PAK and at least one of the proteins that aggregates or accumulates in a cell or tissue is stronger in the absence of a PAK inhibitor (by e.g., 90%, 80%, 70%, 60%, 50%, 40%, 30% or 20%) than in the presence of an inhibitor.

[00232] An "individual" or an "individual," as used herein, is a mammal. In some embodiments, an individual is an animal, for example, a rat, a mouse, a dog or a monkey. In some embodiments, an individual is a human patient. In some embodiments an "individual" or an "individual" is a human. In some embodiments, an individual suffers from a neurological condition or is suspected to be suffering from a neurological condition or is pre-disposed to a neurological condition.

[00233] In some embodiments, a pharmacological composition comprising a PAK inhibitor is "administered peripherally" or "peripherally administered." As used herein, these terms refer to any form of administration of an agent, e.g., a therapeutic agent, to an individual that is not direct administration to the CNS, i.e., that brings the agent in contact with the non- brain side of the blood-brain barrier. "Peripheral administration," as used herein, includes intravenous, intra-arterial, subcutaneous, intramuscular, intraperitoneal, transdermal, by inhalation, transbuccal, intranasal, rectal, oral, parenteral, sublingual, or trans-nasal. In some embodiments, a PAK inhibitor is administered by an intracerebral route.

[00234] The terms "polypeptide," and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. That is, a description directed to a polypeptide applies equally to a description of a protein, and vice versa. The terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally occurring amino acid, e.g., an amino acid analog. As used herein, the terms encompass amino acid chains of any length, including full length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds.

[00235] The term "amino acid" refers to naturally occurring and non-naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally encoded amino acids are the 20 common amino acids (alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine) and pyrolysine and selenocysteine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.

[00236] Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.

[00237] The term "nucleic acid" refers to deoxyribonucleotides, deoxyribonucleosides, ribonucleosides, or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers to oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like). Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al, Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al, J. Biol. Chem. 260:2605-2608 (1985); and Cassol et al. (1992); Rossolini et al, Mol. Cell. Probes 8:91-98 (1994)). [00238] The terms "isolated" and "purified" refer to a material that is substantially or essentially removed from or concentrated in its natural environment. For example, an isolated nucleic acid is one that is separated from the nucleic acids that normally flank it or other nucleic acids or components (proteins, lipids, etc.) in a sample. In another example, a polypeptide is purified if it is substantially removed from or concentrated in its natural environment. Methods for purification and isolation of nucleic acids and proteins are documented methodologies.

[00239] The term "antibody" describes an immunoglobulin whether natural or partly or wholly synthetically produced. The term also covers any polypeptide or protein having a binding domain which is, or is homologous to, an antigen-binding domain. CDR grafted antibodies are also contemplated by this term.

[00240] The term antibody as used herein will also be understood to mean one or more fragments of an antibody that retain the ability to specifically bind to an antigen, (see generally, Holliger et al., Nature Biotech. 23 (9) 1126-1129 (2005)). Non-limiting examples of such antibodies include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CHI domains; (ii) a F(ab')2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341 :544 546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR). Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they are optionally joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al. (1988) Science 242:423 426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879 5883; and Osbourn et al. (1998) Nat. Biotechnol. 16:778). Such single chain antibodies are also intended to be encompassed within the term antibody. Any VH and VL sequences of specific scFv is optionally linked to human immunoglobulin constant region cDNA or genomic sequences, in order to generate expression vectors encoding complete IgG molecules or other isotypes. VH and VL are also optionally used in the generation of Fab, Fv or other fragments of immunoglobulins using either protein chemistry or recombinant DNA technology. Other forms of single chain antibodies, such as diabodies are also encompassed.

[00241] "F(ab')2" and "Fab"' moieties are optionally produced by treating

immunoglobulin (monoclonal antibody) with a protease such as pepsin and papain, and includes an antibody fragment generated by digesting immunoglobulin near the disulfide bonds existing between the hinge regions in each of the two H chains. For example, papain cleaves IgG upstream of the disulfide bonds existing between the hinge regions in each of the two H chains to generate two homologous antibody fragments in which an L chain composed of VL (L chain variable region) and CL (L chain constant region), and an H chain fragment composed of VH (H chain variable region) and CHyl (γΐ region in the constant region of H chain) are connected at their C terminal regions through a disulfide bond. Each of these two homologous antibody fragments is called Fab'. Pepsin also cleaves IgG downstream of the disulfide bonds existing between the hinge regions in each of the two H chains to generate an antibody fragment slightly larger than the fragment in which the two above-mentioned Fab' are connected at the hinge region. This antibody fragment is called F(ab')2.

[00242] The Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CHI domain including one or more cysteine(s) from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are documented.

[00243] "Fv" is the minimum antibody fragment which contains a complete antigen- recognition and antigen-binding site. This region consists of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer antigen- binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.

[00244] "Single-chain Fv" or "sFv" antibody fragments comprise a VH, a VL, or both a

VH and VL domain of an antibody, wherein both domains are present in a single polypeptide chain. In some embodiments, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the sFv to form the desired structure for antigen binding. For a review of sFv see, e.g., Pluckthun in The Pharmacology of Monoclonal Antibodies, Vol. 113, Rosenburg and Moore eds. Springer- Verlag, New York, pp. 269 315 (1994).

[00245] A "chimeric" antibody includes an antibody derived from a combination of different mammals. The mammal is, for example, a rabbit, a mouse, a rat, a goat, or a human. The combination of different mammals includes combinations of fragments from human and mouse sources.

[00246] In some embodiments, an antibody described or described herein is a monoclonal antibody (MAb), typically a chimeric human-mouse antibody derived by humanization of a mouse monoclonal antibody. Such antibodies are obtained from, e.g., transgenic mice that have been "engineered" to produce specific human antibodies in response to antigenic challenge. In this technique, elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci. In some embodiments, the transgenic mice synthesize human antibodies specific for human antigens, and the mice are used to produce human antibody- secreting hybridomas.

[00247] The term "optionally substituted" or "substituted" means that the referenced group substituted with one or more additional group(s). In certain embodiments, the one or more additional group(s) are individually and independently selected from amide, ester, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl, heteroalicyclic, hydroxy, alkoxy, aryloxy, alkylthio, arylthio, alkylsulfoxide, arylsulfoxide, ester, alkylsulfone, arylsulfone, cyano, halogen, alkoyl, alkoyloxo, isocyanato, thiocyanato, isothiocyanato, nitro, haloalkyl, haloalkoxy, fluoroalkyl, amino, alkyl-amino, dialkyl-amino, amido.

[00248] An "alkyl" group refers to an aliphatic hydrocarbon group. Reference to an alkyl group includes "saturated alkyl" and/or "unsaturated alkyl". The alkyl group, whether saturated or unsaturated, includes branched, straight chain, or cyclic groups. By way of example only, alkyl includes methyl, ethyl, propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, t-butyl, pentyl, iso- pentyl, neo-pentyl, and hexyl. In some embodiments, alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tertiary butyl, pentyl, hexyl, ethenyl, propenyl, butenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. A "lower alkyl" is a Ci-C 6 alkyl. A "heteroalkyl" group substitutes any one of the carbons of the alkyl group with a heteroatom having the appropriate number of hydrogen atoms attached (e.g., a CH 2 group to an NH group or an O group).

[00249] An "alkoxy" group refers to a (alkyl)0- group, where alkyl is as defined herein.

[00250] The term "alkylamine" refers to the -N(alkyl) x H y group, wherein alkyl is as defined herein and x and y are selected from the group x=l, y=l and x=2, y=0. When x=2, the alkyl groups, taken together with the nitrogen to which they are attached, optionally form a cyclic ring system. [00251] An "amide" is a chemical moiety with formula C(0)NHR or NHC(0)R, where R is selected from alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon).

[00252] The term "ester" refers to a chemical moiety with formula -C(=0)OR, where R is selected from the group consisting of alkyl, cycloalkyl, aryl, heteroaryl and heteroalicyclic.

[00253] As used herein, the term "aryl" refers to an aromatic ring wherein each of the atoms forming the ring is a carbon atom. Aryl rings described herein include rings having five, six, seven, eight, nine, or more than nine carbon atoms. Aryl groups are optionally substituted. Examples of aryl groups include, but are not limited to phenyl, and naphthalenyl.

[00254] The term "cycloalkyl" refers to a monocyclic or polycyclic non-aromatic radical, wherein each of the atoms forming the ring (i.e. skeletal atoms) is a carbon atom. In various embodiments, cycloalkyls are saturated, or partially unsaturated. In some embodiments, cycloalkyls are fused with an aromatic ring. Cycloalkyl groups include groups having from 3 to 10 ring atoms. Illustrative examples of cycloalkyl groups include, but are not limited to, the f llowing moieties:

and the like. Monocyclic cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. Dicylclic cycloalkyls include, but are not limited to tetrahydronaphthyl, indanyl, tetrahydropentalene or the like. Polycyclic cycloalkyls include admantane, norbornane or the like. The term cycloalkyl includes "unsaturated nonaromatic carbocyclyl" or "nonaromatic unsaturated carbocyclyl" groups both of which refer to a nonaromatic carbocycle, as defined herein, that contains at least one carbon carbon double bond or one carbon carbon triple bond.

[00255] The term "heterocyclo" refers to hetero aromatic and heteroalicyclic groups containing one to four ring heteroatoms each selected from O, S and N. In certain instances, each heterocyclic group has from 4 to 10 atoms in its ring system, and with the proviso that the ring of said group does not contain two adjacent O or S atoms. Non-aromatic heterocyclic groups include groups having 3 atoms in their ring system, but aromatic heterocyclic groups must have at least 5 atoms in their ring system. The heterocyclic groups include benzo-fused ring systems. An example of a 3-membered heterocyclic group is aziridinyl (derived from aziridine). An example of a 4-membered heterocyclic group is azetidinyl (derived from azetidine). An example of a 5-membered heterocyclic group is thiazolyl. An example of a 6- membered heterocyclic group is pyridyl, and an example of a 10-membered heterocyclic group is quinolinyl. Examples of non-aromatic heterocyclic groups are pyrrolidinyl, tetrahydroiuranyl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidino, morpholino, thiomorpholino, thioxanyl, piperazinyl, aziridinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, 1,2,3,6- tetrahydropyridinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxanyl, 1,3-dioxolanyl, pyrazolinyl, dithianyl, dithiolanyl, dihydropyranyl, dihydrothienyl,

dihydrofuranyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 3-azabicyclo[3.1.0]hexanyl, 3- azabicyclo[4.1.0]heptanyl, 3H-indolyl and quinolizinyl. Examples of aromatic heterocyclic groups are pyridinyl, imidazolyl, pyrimidinyl, pyrazolyl, triazolyl, pyrazinyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, isothiazolyl, pyrrolyl, quinolinyl, iso quinolinyl, indolyl, benzimidazolyl, benzofuranyl, cinnolinyl, indazolyl, indolizinyl, phthalazinyl, pyridazinyl, triazinyl, isoindolyl, pteridinyl, purinyl, oxadiazolyl, thiadiazolyl, furazanyl, benzofurazanyl, benzothiophenyl, benzothiazolyl, benzoxazolyl, quinazolinyl, quinoxalinyl, naphthyridinyl, and furopyridinyl.

[00256] The terms "heteroaryl" or, alternatively, "hetero aromatic" refers to an aryl group that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur. An N- containing "heteroaromatic" or "heteroaryl" moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom. In certain embodiments, heteroaryl groups are monocyclic or polycyclic. Examples of monocyclic heteroaryl groups include and are not limited to

Examples of bicyclic heteroaryl groups include and are not limited to

or the like.

[00258] A "heteroalicyclic" group or "heterocyclo" group or "heterocycloalkyl" group or

"heterocyclyl" group refers to a cycloalkyl group, wherein at least one skeletal ring atom is a heteroatom selected from nitrogen, oxygen and sulfur. In some embodiments, the radicals are fused with an aryl or heteroaryl. Example of saturated heterocyloalkyl groups include

[00260] Other illustrative examples of heterocyclo groups, also referred to as non- aromatic heterocycles, include:

[00261] The term heteroalicyclic also includes all ring forms of the carbohydrates, including but not limited to the monosaccharides, the disaccharides and the oligosaccharides.

[00262] The term "halo" or, alternatively, "halogen" means fluoro, chloro, bromo and iodo.

[00263] The terms "haloalkyl," and "haloalkoxy" include alkyl and alkoxy structures that are substituted with one or more halogens. In embodiments, where more than one halogen is included in the group, the halogens are the same or they are different. The terms "fluoroalkyl" and "fluoroalkoxy" include haloalkyl and haloalkoxy groups, respectively, in which the halo is fluorine.

[00264] The term "heteroalkyl" include optionally substituted alkyl, alkenyl and alkynyl radicals which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus, silicon, or combinations thereof. In certain embodiments, the heteroatom(s) is placed at any interior position of the heteroalkyl group.

Examples include, but are not limited to, -CH 2 -0-CH 3 , -CH 2 -CH 2 -0-CH 3 , -CH 2 -NH-CH 3 , -CH 2 - CH 2 -NH-CH 3 , -CH 2 -N(CH 3 )-CH 3 , -CH 2 -CH 2 -NH-CH 3 , -CH 2 -CH 2 -N(CH 3 )-CH 3 , -CH 2 -S-CH 2 - CH 3 , -CH 2 -CH 2 ,-S(0)-CH 3 , -CH 2 -CH 2 -S(0) 2 -CH 3 , -CH=CH-0-CH 3 , -Si(CH 3 ) 3 , -CH 2 -CH=N- OCH 3 , and -CH=CH-N(CH 3 )-CH 3 . In some embodiments, up to two heteroatoms are consecutive, such as, by way of example, -CH 2 -NH-OCH 3 and -CH 2 -0-Si(CH 3 ) 3 .

[00265] A "cyano" group refers to a CN group.

[00266] An "isocyanato" group refers to a NCO group.

[00267] A "thiocyanato" group refers to a CNS group.

[00268] An "isothiocyanato" group refers to a NCS group.

[00269] "Alkoyloxy" refers to a RC(=0)0- group.

[00270] "Alkoyl" refers to a RC(=0)- group.

Methods

[00271] Provided herein are methods of treating one or more symptoms of a neurological or neurological condition comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof.

[00272] Provided herein, in some embodiments, are methods of treating one or more symptoms of epilepsy comprising administration of a therapeutically effective amount of a p21- activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments of the methods provided herein,

administration of a p21 -activated kinase inhibitor to an individual in need thereof reduces or reverses incidence of abnormal neuronal activity in the brain. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having epilepsy) reduces or reverses incidence of abnormal electricity in the brain that is associated with epilepsy.

Abnormal electrical activity in the brain is determined, for example, by techniques such as

Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) and/or Diffusion Tensor Imaging (DTI).

[00273] Provided herein, in some embodiments, are methods of reversing or delaying one or more symptoms of Huntington's disease comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Huntington's disease) stabilizes, alleviates or reverses one or more behavioral symptoms (e.g., deficits in executive functions such as planning, cognitive flexibility, abstract thinking, rule acquisition, initiating appropriate actions, inhibiting inappropriate actions; memory deficits such as short-term memory deficits, long-term memory difficulties, deficits in episodic (memory of one's life), procedural (memory of the body of how to perform an activity) and working memory, and the like) associated with subcortical dementia. In some embodiments of the methods provided herein, administration of a p21-activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Huntington's disease) halts or delays progression of subcortical dementia to dementia. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Huntington's disease) reverses or partially reverses choreia associated with Huntington's disease.

[00274] Provided herein, in some embodiments, are methods of reversing or delaying one or more symptoms of Parkinson's disease comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Huntington's disease) stabilizes, alleviates or reverses one or more behavioral symptoms (e.g., deficits in executive functions such as planning, cognitive flexibility, abstract thinking, rule acquisition, initiating appropriate actions, inhibiting inappropriate actions; memory deficits such as short-term memory deficits, long-term memory difficulties, deficits in episodic (memory of one's life), procedural (memory of the body of how to perform an activity) and working memory, and the like) associated with Parkinson's disease. In some embodiments of the methods provided herein, administration of a p21-activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Parkinson's disease) reverses or partially reverses deficits in motor skills and speech. In some of such embodiments, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Parkinson's disease) reverses or partially reverses muscle rigidity, tremor, slowing of physical movement (bradykinesia) and/or loss of physical movement (akinesia). In some embodiments, administration of a p21-activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of having Parkinson's disease) delays disease progression.

[00275] Provided herein, in some embodiments, are methods of treating substance abuse and substance dependency comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments of the methods provided herein, administration of a p21-activated kinase inhibitor to an individual in need thereof (e.g., an individual addicted to, for example, alcohol, narcotic drugs, pain killers, gambling, video games or the like) reverses or partially reverses the changes in neuronal plasticity associated with addiction. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual diagnosed with or suspected of being an addict) reverses, partially reverses, halts or delays progression of neuro adaptation associated with addiction in the brain. Neuronal activity in the brain associated with addiction is determined, for example, by techniques such as Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) and/or Diffusion Tensor Imaging (DTI).

[00276] Provided herein, in some embodiments, are methods of treating clinical depression comprising administration of a therapeutically effective amount of a p21 -activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from clinical depression) reverses, partially reverses, halts or delays progression of neuro adaptation associated with clinical depression in the brain. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from clinical depression) normalizes or partially normalizes aberrant neuroplasticity associated with clinical depression in the brain. Neuronal activity in the brain associated with clinical depression is determined, for example, by techniques such as Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) and/or Diffusion Tensor Imaging (DTI).

[00277] Provided herein, in some embodiments, are methods of treating Down's syndrome comprising administration of a therapeutically effective amount of a p21 -activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual suffering from Down's syndrome) reverses, or partially reverses mental retardation associated with Down's syndrome. In some embodiments of the methods provided herein, administration of a p21 -activated kinase inhibitor to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from clinical depression) normalizes or partially normalizes aberrant neuroplasticity associated with clinical depression in the brain. Neuronal activity in the brain associated with clinical depression is determined, for example, by techniques such as Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET) and/or Diffusion Tensor Imaging (DTI).

[00278] Provided herein are methods for treating mental retardation other than Down's syndrome (including and not limited to Fetal alcohol sydrome, Klinefelter's syndrome, congenital hypothyroidism, Williams syndrome, Smith-Lemli-Opitz syndrome, Prader-willi syndrome, Phelan-McDermid syndrome, Mowat- Wilson syndrome, Ciliopathy, Lowe syndrome and/or any other genetic mental retardation syndromes) comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from the aforementioned mental retardation syndromes reduces or reverses developmental disability and/or loss of cognitive ability associated with such mental retardation.

[00279] Provided herein are methods for treating neurofibromatosis (including

Neurofibromatosis I and Neurofibromatosis II) comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from Neurofibromatosis) reduces or reverses neurofibromatoses and or optical nerve gliomas. In some embodiments, administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from Neurofibromatosis) reduces or reverses neurofibromatoses and or optical nerve gliomas. In some embodiments, administration of a therapeutically effective amount of a p21 -activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from Neurofibromatosis) reduces or reverses learning disabilities and/or cognition deficits and/or memory deficits associated with neurofibromatosis.

[00280] Provided herein are methods for treating tuberous sclerosis comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some embodiments, administration of a therapeutically effective amount of a p21 -activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from tuberous sclerosis) reduces or reverses occurence of tubers in the individual. In some embodiments, administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected to be suffering from tuberous sclerosis) reduces or reverses learning disabilities and/or cognition deficits and/or memory deficits associated with tuberous sclerosis.

[00281] Provided herein are methods for treating Maple Syrup Disease comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from Maple Syrup Disease reduces or reverses the neurological deterioration associated with Maple Syrup Disease.

[00282] Provided herein are methods for treating Nieman Pick disease comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from Nieman Pick Disease reduces or reverses the neurological deterioration associated with Nieman Pick Disease.

[00283] Provided herein are methods for treating encephalitis comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from encephalitis reduces or reverses brain inflammation associated with encephalitis. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from encephalitis halts or reduces the occurence and/or severity of seizures associated with encephalitis.

[00284] Provided herein are methods for treating bipolar disorder comprising

administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from bipolar disorder reduces or reverses behavioral symptoms associated with bipolar disorder.

[00285] Provided herein are methods for treating mania comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of

Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from mania reduces or reverses behavioral symptoms associated with mania.

[00286] Provided herein are methods for treating Anxiety Disorder comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from anxiety disorder reduces or reverses behavioural symptoms associated with anxiety disorder.

[00287] Provided herein are methods for treating PTSD comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from PTSD reduces or reverses behavioral symptoms associated with PTSD.

[00288] Provided herein are methods for treating schizoaffective disorder comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from schizoaffective disorder reduces or reverses behavioral symptoms associated with scizo affective disorder.

[00289] Provided herein are methods for treating schizophreniform comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from schizophreniform reduces or reverses behavioral symptoms associated with schizophreniform.

[00290] Provided herein are methods for treating age-related cognitive decline associated with menopause comprising administration of a therapeutically effective amount of a p21- activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from age-related cognitive decline associated with menopause reduces or reverses memory loss symptoms associated with age- related cognitive decline associated with menopause.

[00291] Provided herein are methods for treating Angelman Syndrome comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from Angelman Syndrom reduces or reverses behavioral symptoms associated with Angelman Syndrome.

[00292] Provided herein are methods for treating OCD comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of

Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from OCD reduces or reverses behavioral symptoms associated with OCD.

[00293] Provided herein are methods for treating panic disorder comprising

administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from panic disorder reduces or reverses behavioral symptoms associated with panic disorder.

[00294] Provided herein are methods for treating phobias comprising administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of

Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from a phobia reduces or reverses behavioral symptoms associated with the phobia.

[00295] Provided herein are methods for treating aneroxia nervosa comprising

administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from aneroxia nervosa reduces or reverses behavioral symptoms associated with aneroxia nervosa.

[00296] Provided herein are methods for treating bulimia nervosa comprising

administration of a therapeutically effective amount of a p21-activated kinase inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof. In some of such embodiments, administration of a PAK inhibitor to an individual suffering from or suspected to be suffering from bulimia nervosa reduces or reverses behavioral symptoms associated with bulimia nervosa.

[00297] Also provided herein are methods for modulation of dendritic spine morphology and/or dendritic spine density and/or synaptic function comprising administering to an individual in need thereof (e.g., an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression or the like) a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). In some embodiments, modulation of dendritic spine morphology and/or synaptic function stabilizes, alleviates or reverses memory and/or cognitive impairment associated with a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, neurofibromatoses, tuberous sclerosis, Down's syndrome or the like. In some embodiments, modulation of dendritic spine morphology and/or synaptic function halts or delays progression of memory and/or cognitive impairment associated with any neurological condition described herein.

[00298] Provided herein are methods for modulation of synaptic function or synaptic plasticity comprising administering to an individual in need thereof (e.g., an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, NF, TS, or the like) a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Modulation of synaptic function or plasticity includes, for example, stabilization, alleviation or reversal of defects in LTP, LTD or the like.

[00299] Defects in LTP include, for example, an increase in LTP or a decrease in LTP in any region of the brain in an individual suffering from a neurological condition. Defects in LTD include for example a decrease in LTD or an increase in LTD in any region of the brain (e.g., the temporal lobe, parietal lobe, the frontal cortex, the cingulate gyrus, the prefrontal cortex, the cortex, or the hippocampus or any other region in the brain or a combination thereof) in an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, NF, TS, or any other neurological condition described herein.

[00300] In some embodiments of the methods, administration of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) modulates synaptic function (e.g., synaptic transmission and/or plasticity) by increasing long term potentiation (LTP) in an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, NF, TS, or any other neurological condition described herein. In some embodiments of the methods described herein, administration of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of

Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof modulates synaptic function (e.g., synaptic transmission and/or plasticity) by increasing long term potentiation (LTP) in the prefrontal cortex, or the cortex, or the hippocampus or any other region in the brain or a combination thereof. In some embodiments of the methods described herein, administration of a PAK inhibitor modulates synaptic function (e.g., synaptic transmission and/or plasticity) by decreasing long term depression (LTD) in an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, NF, TS, or any other neurological condition described herein. In some embodiments of the methods described herein, administration of a PAK inhibitor to an individual in need thereof modulates synaptic function (e.g., synaptic transmission and/or plasticity) by decreasing long term depression (LTD) in the temporal lobe, parietal lobe, the frontal cortex, the cingulate gyrus, the prefrontal cortex, the cortex, or the hippocampus or any other region in the brain or a combination thereof.

[00301] Provided herein are methods for stabilization and/or normalization and/or partial normalization of synaptic plasticity comprising administering to an individual in need thereof (e.g., an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, NF, TS, or any other neurological condition described herein, a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). In some embodiments of the methods described herein, administration of a PAK inhibitor stabilizes LTP or LTD following induction (e.g., by theta-burst stimulation, high-frequency stimulation).

[00302] Provided herein are methods for stabilization and/or normalization and/or partial normalization of synaptic transmission comprising administering to an individual in need thereof (e.g., in an individual suffering from a neurological condition such as epilepsy, Huntington's disease, Parkinson's disease, addiction, clinical depression, NF, TS, or any other neurological condition described herein, a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). In some embodiments of the methods described herein, administration of a PAK inhibitor stabilizes LTP or LTD following induction (e.g., by theta-burst stimulation, high-frequency stimulation). [00303] Also provided herein are methods for stabilization, alleviation or reversal of neuro adaptation associated with neurological conditions such as addiction and/or clinical depression comprising administering to an individual in need thereof (e.g., in an individual suffering from addiction, clinical depression or the like) a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein).

[00304] Also provided herein are methods for stabilization, alleviation or reversal of mental retardation associated with neurological conditions such as Down's syndrome comprising administering to an individual in need thereof a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). In some of such embodiments, administration of a PAK inhibitor to an individual suffering from a neurological condition stabilizes, or improves scores in tests such as the Mini-Mental State Examination

(MMSE), HAM-D test, UHDRS test, MATRICS cognitive battery, BACS score, Mild Cognitive Impairment Assessment Scale - Cognitive Subscale (ADAS-Cog), Hopkins Verbal Learning Test-Revised, Wechsler Intelligence Scale-Revised, Wechsler Memory Scale-Revised, Dementia Rating Scale (DRS), Boston Naming Test, Stroop Color Word Test, Trail Making Test or Auditory Verbal Learning Test (AVLT) scale or the like.

[00305] Also provided herein are methods for stabilization, alleviation or reversal of choreia associated with neurological conditions such as Huntington's disease comprising administering to an individual in need thereof a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Further provided herein are methods for stabilization, alleviation or reversal of parkinsonism (e.g., muscle rigidity, bradykinesia, akinesia, tremor) associated with neurological conditions such as Parkinson's disease comprising administering to an individual in need thereof a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Provided herein are methods for stabilization, alleviation or reversal of neurofibromatoses associated with conditions such as Neurofibromatosis comprising administering to an individual in need thereof a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Provided herein are methods for stabilization, alleviation or reversal of hearing loss associated with conditions such as neurofibromatosis comprising administering to an individual in need thereof a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Provided herein are methods for

stabilization, alleviation or reversal of tubers associated with conditions such as tuberous sclerosis comprising administering to an individual in need thereof a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein).

[00306] Provided herein are methods for stabilizing, reducing or reversing abnormalities in dendritic spine morphology and/or dendritic spine density and/or synaptic function that are caused by increased activation of PAK at the synapse, comprising administration of a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected of having epilepsy, Huntington's disease, suffering from addiction, clinical depression or the like). In some embodiments of the methods described herein, increased activation of PAK at the synapse is caused by aberrant protein (e.g., mHTT, hamartin, tuberin or the like). In some instances, increased activation of PAK at the synapse is caused by redistribution of PAK from the cytosol to the synapse. In some embodiments of the methods described herein,

administration of a effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) to an individual in need thereof (e.g., an individual suffering from or suspected of having a neurological condition) reduces or prevents redistribution of PAK from the cytosol to the synapse in neurons, thereby stabilizing, reducing or reversing abnormalities in dendritic spine morphology or synaptic function that are caused by increased activation of PAK at the synapse.

[00307] Provided herein are methods for stabilizing, reducing or reversing neuronal withering and/or atrophy or nervous tissue and/or degeneration of nervous tissue that is associated with a neurological condition. In some embodiments of the methods described herein, administration of a PAK inhibitor to an individual suffering from a neurological condition stabilizes, alleviates or reverses neuronal withering and/or atrophy and/or degeneration in the temporal lobe, parietal lobe, the frontal cortex, the cingulate gyrus or the like. In some embodiments of the methods described herein, administration of a PAK inhibitor to an individual suffering from a neurological condition stabilizes, reduces or reverses deficits in memory and/or cognition associated with subcortical dementia. In some embodiments of the methods described herein, administration of a PAK inhibitor to an individual suffering from a neurological condition stabilizes, reduces or reverses progressive deterioration of subcortical dementia toward dementia.

[00308] Provided herein are methods for halting or delaying the onset of a neurological condition comprising administering to an individual in need thereof (e.g. an individiual having mutations in the huntingtin gene, mutations on chromosome 17ql 1.2 (gene product

Neurofibromin), mutation on chromosome 22q (gene product Merlin), mutations on

chromosome 9 q34 (gene product hamartin), mutations on chromosome 16 pi 3.3 (gene product tuberin), or any other high-risk allele) a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Provided herein are methods for delaying the loss of dendritic spine density comprising administering to an individual in need thereof (e.g. mutations in the huntingtin gene, mutations on chromosome 17ql 1.2 (gene product

Neurofibromin), mutation on chromosome 22q (gene product Merlin), mutations on

chromosome 9 q34 (gene product hamartin), mutations on chromosome 16 pi 3.3 (gene product tuberin), or any other high-risk allele) a therapeutically effective amount of a PAK inhibitor. In some embodiments of the methods described herein, prophylactic administration of a PAK inhibitor to an individual at a high risk for developing a neurological condition reverses abnormalities in dendritic spine morphology and/or synaptic function and prevents development of a neurological condition. In some embodiments of the methods described herein, prophylactic administration of a PAK inhibitor to an individual at a high risk for developing a neurological condition (e.g., an individual with a mutation in huntingtin gene or a high-risk allele that pre- disposes the individual to Huntington's disease) delays, reduces or prevents symptoms of the neurological condition (e.g., subcortical dementia, choreia).

[00309] Provided herein are methods for modulation of dendritic spine density, dendritic spine shape, dendritic spine length, dendritic spine head volume, or dendritic spine neck diameter or the like comprising administering to an individual in need thereof (e.g., an individual suffering from a neurological condition such as addiction, clinical depression) a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). Provided herein are methods of modulating the ratio of mature dendritic spines to immature dendritic spines comprising administering to an individual in need thereof (e.g., an individual suffering from a neurological condition such as Huntington's disease) a therapeutically effective amount of a PAK inhibitor. Provided herein are methods of modulating the ratio of dendritic spines head volume to dendritic spines length comprising administering to an individual in need thereof (e.g., an individual suffering from a neurological condition described herein) a therapeutically effective amount of a PAK inhibitor (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein). In some embodiments, administration of a PAK inhibitor results in (i) Altering/restructuring dendritic spine morphology and/or (ii) improving synaptic function.

[00310] In some embodiments of the methods described herein, administration of a PAK inhibitor (e.g., a maintenance dose of a PAK inhibitor) halts or delays the progression of symptoms or pathologies of a neurological condition in an individual. In some embodiments of the methods described herein, administration of a PAK inhibitor causes substantially complete inhibition of PAK and restores dendritic spine morphology and/or synaptic function to normal or partially normal levels. In some embodiments of the methods described herein, administration of a PAK inhibitor causes partial inhibition of PAK and restores dendritic spine morphology and/or dendritic spine density and/or synaptic function to normal or partially normal levels.

[00311] In some instances, neuro adaptation, subcortical dementia, memory deficits, deficits in cognitive ability and/or mental retardation are associated with a decrease in dendritic spine density. In some embodiments of the methods described herein, administration of a PAK inhibitor increases dendritic spine density. In some instances, neuro adaptation, subcortical dementia, memory deficits, deficits in cognitive ability and/or mental retardation are associated with an increase in dendritic spine length. In some embodiments of the methods described herein, administration of a PAK inhibitor decreases dendritic spine length. In some instances, neuro adaptation, subcortical dementia, memory deficits, deficits in cognitive ability and/or mental retardation are associated with a decrease in dendritic spine neck diameter. In some embodiments of the methods described herein, administration of a PAK inhibitor increases dendritic spine neck diameter. In some instances, neuro adaptation, subcortical dementia, memory deficits, deficits in cognitive ability and/or mental retardation are associated with a decrease in dendritic spine head volume and/or dendritic spine head surface area. In some embodiments of the methods described herein, administration of a PAK inhibitor increases dendritic spine head volume and/or dendritic spine head surface area. [00312] In some instances, neuro adaptation, subcortical dementia, memory deficits, deficits in cognitive ability and/or mental retardation are associated with an increase in immature spines and a decrease in mature spines. In some embodiments of the methods described herein, administration of a PAK inhibitor modulates the ratio of immature spines to mature spines. In some instances, neuro adaptation, subcortical dementia, memory deficits, deficits in cognitive ability and/or mental retardation are associated with an increase in stubby spines and a decrease in mushroom-shaped spines. In some embodiments of the methods described herein,

administration of a PAK inhibitor modulates the ratio of stubby spines to mushroom-shaped spines.

[00313] In some embodiments of the methods described herein, administration of a PAK inhibitor modulates a spine:head ratio, e.g., ratio of the volume of the spine to the volume of the head, ratio of the length of a spine to the length of a head of the spine, ratio of the surface area of a spine to the surface area of the head of a spine, or the like, compared to a spine:head ratio in the absence of a PAK inhibitor. In certain embodiments, a PAK inhibitor suitable for the methods described herein modulates the volume of the spine head, the width of the spine head, the surface area of the spine head, the length of the spine shaft, the diameter of the spine shaft, or a combination thereof. In some embodiments, provided herein is a method of modulating the volume of a spine head, the width of a spine head, the surface area of a spine head, the length of a spine shaft, the diameter of a spine shaft, or a combination thereof, by contacting a neuron comprising the dendritic spine with an effective amount of a PAK inhibitor described herein. In specific embodiments, the neuron is contacted with the PAK inhibitor in vivo.

[00314] In certain embodiments, a compound or a composition comprising a compound described herein is administered for prophylactic and/or therapeutic treatments. In therapeutic applications, the compositions are administered to an individual already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest the symptoms of the disease or condition. In various instances, amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, an individual's health status, weight, and response to the drugs, and the judgment of the treating physician.

[00315] In some embodiments, a composition containing a therapeutically effective amount of a PAK inhibitor is administered prophylactically to an individual that, while not overtly manifesting symptoms of a neurological condition, has been identified as having a high risk of developing a neurological condition, e.g., an individual is identified as being a carrier of a mutation or polymorphism associated with a higher risk to develop a neurological condition, or an individual that is from a family that has a high incidence of a neurological condition. In some embodiments, MRI is used to detect brain morphological changes in the brain prior to the onset of a neurological condition.

[00316] In prophylactic applications, compounds or compositions containing compounds described herein are administered to an individual susceptible to or otherwise at risk of a particular disease, disorder or condition. In certain embodiments of this use, the precise amounts of compound administered depend on an individual's state of health, weight, and the like.

Furthermore, in some instances, when a compound or composition described herein is administered to an individual, effective amounts for this use depend on the severity and course of the disease, disorder or condition, previous therapy, an individual's health status and response to the drugs, and the judgment of the treating physician.

[00317] In certain instances, wherein following administration of a selected dose of a compound or composition described herein, an individual's condition does not improve, upon the doctor's discretion the administration of a compound or composition described herein is optionally administered chronically, that is, for an extended period of time, including throughout the duration of an individual's life in order to ameliorate or otherwise control or limit the symptoms of an individual's disorder, disease or condition.

[00318] In certain embodiments, an effective amount of a given agent varies depending upon one or more of a number of factors such as the particular compound, disease or condition and its severity, the identity (e.g., weight) of an individual or host in need of treatment, and is determined according to the particular circumstances surrounding the case, including, e.g., the specific agent being administered, the route of administration, the condition being treated, and an individual or host being treated. In some embodiments, doses administered include those up to the maximum tolerable dose. In certain embodiments, about 0.02-5000 mg per day, from about 1-1500 mg per day, about 1 to about 100 mg/day, about 1 to about 50 mg/day, or about 1 to about 30 mg/day, or about 5 to about 25 mg/day of a compound described herein is administered. In various embodiments, the desired dose is conveniently be presented in a single dose or in divided doses administered simultaneously (or over a short period of time) or at appropriate intervals, for example as two, three, four or more sub-doses per day.

[00319] In certain instances, there are a large number of variables in regard to an individual treatment regime, and considerable excursions from these recommended values are considered within the scope described herein. Dosages described herein are optionally altered depending on a number of variables such as, by way of non-limiting example, the activity of the compound used, the disease or condition to be treated, the mode of administration, the requirements of an individual, the severity of the disease or condition being treated, and the judgment of the practitioner.

[00320] Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined by pharmaceutical procedures in cell cultures or experimental animals, including, but not limited to, the determination of the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50%> of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD 50 and ED 50 . Compounds exhibiting high therapeutic indices are preferred. In certain embodiments, data obtained from cell culture assays and animal studies are used in formulating a range of dosage for use in human. In specific embodiments, the dosage of compounds described herein lies within a range of circulating concentrations that include the ED 50 with minimal toxicity. The dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.

Combination Therapy

[00321] In some embodiments, one or more PAK inhibitors are used in combination with one or more other therapeutic agents to treat an individual suffering from a neurological condition. The combination of PAK inhibitors with a second therapeutic agent (e.g., an antidepressant agent) allows a reduced dose of both agents to be used thereby reducing the likelihood of side effects associated with higher dose monotherapies. In one embodiment, the dose of a second active agent (e.g., an anticholinergic) is reduced in the combination therapy by at least 50% relative to the corresponding monotherapy dose, whereas the PAK inhibitor dose is not reduced relative to the monotherapy dose; in further embodiments, the reduction in dose of a second active agent is at least 75%; in yet a further embodiment, the reduction in dose of a second active agent is at least 90%. In some embodiments, the second therapeutic agent is administered at the same dose as a monotherapy dose, and the addition of a PAK inhibitor to the treatment regimen alleviates symptoms of a neurological condition that are not treated by monotherapy with the second therapeutic agent.

[00322] In some embodiments, the combination of a PAK inhibitor and a second therapeutic agent is synergistic (e.g., the effect of the combination is better than the effect of each agent alone). In some embodiments, the combination of a PAK inhibitor and a second therapeutic agent is additive (e.g., the effect of the combination of active agents is about the same as the effect of each agent alone). In some embodiments, an additive effect is due to the PAK inhibitor and the second therapeutic agent modulating the same regulatory pathway. In some embodiments, an additive effect is due to the PAK inhibitor and the second therapeutic agent modulating different regulatory pathways. In some embodiments, an additive effect is due to the PAK inhibitor and the second therapeutic agent treating different symptom groups of a neurological condition (e.g., a PAK inhibitor treats learning disorders and the second therapeutic agent treats seizures). In some embodiments, administration of a second therapeutic agent treats the remainder of the same or different symptoms or groups of symptoms that are not treated by administration of a PAK inhibitor alone.

[00323] In some embodiments, administration of a combination of a PAK inhibitor and a second therapeutic agent alleviates side effects that are caused by the second therapeutic agent (e.g., side effects caused by an antidepressant agent). In some embodiments, administration of the second therapeutic agent inhibits metabolism of an administered PAK inhibitor (e.g., the second therapeutic agent blocks a liver enzyme that degrades the PAK inhibitor) thereby increasing efficacy of a PAK inhibitor. In some embodiments, administration of a combination of a PAK inhibitor and a second therapeutic agent (e.g. a second agent that modulates dendritic spine morphology (e.g., minocyline)) improves the therapeutic index of a PAK inhibitor.

Antidepressant agents

[00324] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed an antidepressant. In some embodiments, administration of a PAK inhibitor in combination with an antidepressant agent has a synergistic effect and provides an improved therapeutic outcome compared to monotherapy with antidepressant agent or monotherapy with PAK inhibitor. Alternatively, a PAK inhibitor composition described herein is administered to an individual who is non-responsive to, or being unsatisfactorily treated with an antidepressant agent. Examples of antidepressant agents include, and are not limited to, selective serotonin reuptake inhibitors (SSRIs) such Escitalopram (Lexapro, Cipralex), Fluoxetine (Prozac), Fluvoxamine (Luvox), Paroxetine (Paxil), Sertraline (Zoloft) or the like, Serotonin- norepinephrine reuptake inhibitors (SNRIs) such as Desvenlafaxine (Pristiq), Duloxetine (Cymbalta), Milnacipram (Ixel), Venlafaxine (Effexor) or the like, Noradrenergic and specific serotonergic antidepressants (NaSSAs) such as Mianserin (Tolvon), Mirtazapine (Remeron, Avanza, Zispin) or the like; Norepinephrine (noradrenaline) reuptake inhibitors (NRIs) such as Atomoxetine (Strattera), Mazindol (Mazanor, Sanorex), Reboxetine (Edronax), Viloxazine (Vivalan) or the like, Norepinephrine-dopamine reuptake inhibitors (NDRIs) such as Bupropion (Wellbutrin, Zyban) or the like, Selective serotonin reuptake enhancers (SSREs) such as Tianeptine (Stablon, Coaxil, Tatinol) or the like, Melatonergic agonists such as Agomelatine (Valdoxan, Melitor, Thymanax) or the like, Tricyclic antidepressants (TCAs) such as

Amitriptyline (Elavil, Endep), Clomipramine (Anafranil), Doxepin (Adapin, Sinequan),

Imipramine (Tofranil), Trimipramine (Surmontil), Desipramine (Norpramin), Nortriptyline (Pamelor, Aventyl), Protriptyline (Vivactil) or the like, Monoamine oxidase inhibitors (MAOIs) such as Isocarboxazid (Marplan), Moclobemide (Aurorix, Manerix), Phenelzine (Nardil), Selegiline (Eldepryl, Emsam), Tranylcypromine (Parnate) or the like.

Anticholinergic agents

[00325] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed an anticholinergic agent. In some embodiments, administration of a PAK inhibitor in combination with an anticholinergic agent has a synergistic effect and provides an improved therapeutic outcome compared to monotherapy with anticholinergic agent or monotherapy with PAK inhibitor. Alternatively, a PAK inhibitor composition described herein is administered to an individual who is non-responsive to, or being unsatisfactorily treated with an anticholinergic agent. Example of anticholinergic drugs include ipratropium bromide

(Atrovent), oxitropium bromide (Oxivent), tiotropium (Spiriva), donepezil (Aricept),

galantamine (Razadyne), rivastigmine (Exelon and Exelon Patch), physostigimine, scopolamine, orphenadrine, dicycloverine/dicyclomine or the like.

Anxiolytics

[00326] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed an anxiolytic.

[00327] Examples of anxiolytics include, but are not limited to, benzodiazepines, e.g.,

Alprazolam, Chlordiazepoxide, Clonazepam, Diazepam, Lorazepam; azapirones, e.g., buspirone, tandospirone, Gepirone; barbituarates, hydroxyzine, pregabalin, or the like.

Antipsychotics

[00328] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed an antipsychotic.

[00329] Examples of antipsychotics include, but are not limited to, any of the following: typical antipsychotics, e.g., Chlorpromazine (Largactil, Thorazine), Fluphenazine (Prolixin), Haloperidol (Haldol, Serenace), Molindone, Thiothixene (Navane), Thioridazine (Mellaril),

Trifluoperazine (Stelazine), Loxapine, Perphenazine, Prochlorperazine (Compazine, Buccastem, Stemetil), Pimozide (Orap), Zuclopenthixol; and atypical antipsychotics, e.g., LY2140023, Clozapine, Risperidone, Olanzapine, Quetiapine, Ziprasidone, Aripiprazole, Paliperidone, Asenapine, Iloperidone, Sertindole, Zotepine, Amisulpride, Bifeprunox, and Melperone.

Alpha 7 nicotinic receptor agonists

[00330] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed an alpha 7 nicotinic receptor agonist.

[00331] Examples of alpha7 nicotinic receptor agonists include and are not limited to (+)-

N-(l-azabicyclo[2.2.2]oct-3-yl)benzo[b]furan- 2-carboxamide, PHA-709829, PNU-282,987, A- 582941, TC-1698, TC-5619, GTS-21, SSR180711, tropisetron or the like. Examples of alpha7 nicotinic receptor antagonists include a-conotoxin, quinolizidine or the like. Alpha7 nicotinic receptor allosteric potentiators include PNU-120596, NS-1738, XY4083, A-867744, EVP-6124 (Envivo), or the like.

GABA agonists

[00332] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed a GABA agonist.

[00333] Examples of GABA agonists include and are not limited to picamilon, benzodiazepines, Zolpidem, Zopiclone, Zaleplon, barbiturates, methaqualone, baclofen, muscimol, progabide, tiagabine, or the like.

Stimulants

[00334] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed a stimulant.

[00335] Examples of stimulants include and are not limited to ritalin, adderall or the like.

NMDA receptor antagonists

[00336] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed an NMDA receptor antagonist. Examples of NMDA receptor antagonists useful in the methods and compositions described herein include and are not limited to amantadine, memantine, tramadol (Ultracet) or the like.

Dopamine Receptor agonists

[00337] In some embodiments, a PAK inhibitor composition described herein is administered to a patient in combination with a dopamine receptor agonist bromocriptine (Parlodel), cabergoline (Dostinex), piribedil (Trivastal), pramipexole (Mirapex), ropinirole (Requip), apomorphine (Apokyn), rotigotine (Neupro) or the like.

Antioxidants

[00338] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who is taking or has been prescribed an antioxidant. Examples of antioxidants useful in the methods and compositions described herein include and are not limited to ubiquinone, aged garlic extract, curcumin, lipoic acid, beta-carotene, melatonin, resveratrol, Ginkgo biloba extract, vitamin C, viatmin E or the like.

Neuroprotectants

[00339] In some embodiments, a PAK inhibitor or a composition thereof described herein is administered in combination with a neuroprotectant such as, for example, minocycline, resveratrol or the like.

Trophic factors

[00340] In some embodiments, a PAK inhibitor or a composition thereof described herein is administered in combination with a trophic agent including, by way of example, glial derived nerve factor (GDNF), brain derived nerve factor (BDNF) or the like. Trkb agonists

[00341] Where a subject is suffering from or at risk of suffering from a neurological condition, a PAK inhibitor composition described herein is optionally used together with one or more agents or methods for treating a neurological condition in any combination. In some embodiments, a PAK inhibitor composition described herein is administered to a patient who has been prescribed a Trkb agonist.

[00342] Examples of Trkb agonists include and are not limited to BDNF, N-

Acetylserotonin, Amitriptyline or the like.

Other Agents

[00343] In some embodiments, one or more PAK inhibitors are used in combination with one or more agents that modulate dendritic spine morphology or synaptic function. Examples of agents that modulate dendritic spine morphology include minocycline, trophic factors (e.g., brain derived neutrophic factor, glial cell-derived neurtrophic factor), or anesthetics that modulate spine motility, or the like. In some embodiments, one or more PAK inhibitors are used in combination with one or more agents that modulate cognition. In some embodiments, a second therapeutic agent is a nootropic agent that enhances cognition. Examples of nootropic agents include and are not limited to piracetam, pramiracetam, oxiracetam, and aniracetam.

Blood Brain Barrier facilitators

[00344] In some instances, a PAK inhibitor is optionally administered in combination with a blood brain barrier facilitator. In certain embodiments, an agent that facilitates the transport of a PAK inhibitor is covalently attached to the PAK inhibitor. In some instances, PAK inhibitors described herein are modified by covalent attachment to a lipophilic carrier or co- formulation with a lipophilic carrier. In some embodiments, a PAK inhibitor is covalently attached to a lipophilic carrier, such as e.g., DHA, or a fatty acid. In some embodiments, a PAK inhibitor is covalently attached to artificial low density lipoprotein particles. In some instances, carrier systems facilitate the passage of PAK inhibitors described herein across the blood-brain barrier and include but are not limited to, the use of a dihydropyridine pyridinium salt carrier redox system for delivery of drug species across the blood brain barrier. In some instances a PAK inhibitor described herein is coupled to a lipophilic phosphonate derivative. In certain instances, PAK inhibitors described herein are conjugated to PEG-oligomers/polymers or aprotinin derivatives and analogs. In some instances, an increase in influx of a PAK inhibitor described herein across the blood brain barrier is achieved by modifying A PAK inhibitor described herein (e.g., by reducing or increasing the number of charged groups on the compound) and enhancing affinity for a blood brain barrier transporter. In certain instances, a PAK inhibitor is co-administered with an an agent that reduces or inhibits efflux across the blood brain barrier, e.g. an inhibitor of P-glycoprotein pump (PGP) mediated efflux (e.g., cyclosporin, SCH66336 (lonafarnib, Schering)).

[00345] In some instances, a PAK inhibitor polypeptide is delivered to one or more brain regions of an individual by administration of a viral expression vector, e.g., an AAV vector, a lentiviral vector, an adenoviral vector, or a HSV vector. A number of viral vectors for delivery of therapeutic proteins are described in, e.g., U.S. Patent Nos., 7,244,423, 6,780,409, 5,661,033. In some embodiments, the PAK inhibitor polypeptide to be expressed is under the control of an inducible promoter (e.g., a promoter containing a tet-operator). Inducible viral expression vectors include, for example, those described in U.S. Patent No. 6,953,575. Inducible expression of a PAK inhibitor polypeptide allows for tightly controlled and reversible increases of PAK inhibitor polypeptide expression by varying the dose of an inducing agent (e.g., tetracycline) administered to an individual.

[00346] Any combination of one or more PAK inhibitors and a second therapeutic agent is compatible with any method described herein. The PAK inhibitor compositions described herein are also optionally used in combination with other therapeutic reagents that are selected for their therapeutic value for the condition to be treated. In general, the compositions described herein and, in embodiments where combinational therapy is employed, other agents do not have to be administered in the same pharmaceutical composition, and, because of different physical and chemical characteristics, are optionally administered by different routes. The initial administration is generally made according to established protocols, and then, based upon the observed effects, the dosage, modes of administration and times of administration subsequently modified.

[00347] In certain instances, it is appropriate to administer at least one PAK inhibitor composition described herein in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving one of the PAK inhibitor compositions described herein is nausea, then it is appropriate to administer an anti-nausea agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of a PAK inhibitor is enhanced by administration of an adjuvant (i.e., by itself the adjuvant has minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, by way of example only, the benefit experienced by a patient is increased by administering a PAK inhibitor with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit. In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient is either simply additive of the two therapeutic agents or the patient experiences a synergistic benefit.

[00348] Therapeutically-effective dosages vary when the drugs are used in treatment combinations. Suitable methods for experimentally determining therapeutically-effective dosages of drugs and other agents include, e.g., the use of metronomic dosing, i.e., providing more frequent, lower doses in order to minimize toxic side effects. Combination treatment further includes periodic treatments that start and stop at various times to assist with the clinical management of the patient.

[00349] In any case, the multiple therapeutic agents (one of which is a PAK inhibitor described herein) are administered in any order, or even simultaneously. If simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). In some embodiments, one of the therapeutic agents is given in multiple doses, or both are given as multiple doses. If not simultaneous, the timing between the multiple doses optionally varies from more than zero weeks to less than four weeks. In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations is also envisioned.

[00350] The pharmaceutical agents which make up the combination therapy disclosed herein are optionally a combined dosage form or in separate dosage forms intended for substantially simultaneous administration. The pharmaceutical agents that make up the combination therapy are optionally also be administered sequentially, with either therapeutic compound being administered by a regimen calling for two-step administration. The two-step administration regimen optionally calls for sequential administration of the active agents or spaced-apart administration of the separate active agents. The time period between the multiple administration steps ranges from, a few minutes to several hours, depending upon the properties of each pharmaceutical agent, such as potency, solubility, bioavailability, plasma half-life and kinetic profile of the pharmaceutical agent. Circadian variation of the target molecule concentration can optionally be used to determine the optimal dose interval.

[00351] In addition, a PAK inhibitor is optionally used in combination with procedures that provide additional or synergistic benefit to the patient. By way of example only, patients are expected to find therapeutic and/or prophylactic benefit in the methods described herein, wherein pharmaceutical composition of a PAK inhibitor and /or combinations with other therapeutics are combined with genetic testing to determine whether that individual is a carrier of a mutant gene that is correlated with certain diseases or conditions. [00352] A PAK inhibitor and the additional therapy(ies) are optionally administered before, during or after the occurrence of a disease or condition, and the timing of administering the composition containing a PAK inhibitor varies in some embodiments. Thus, for example, the PAK inhibitor is used as a prophylactic and administered continuously to individuals with a propensity to develop conditions or diseases in order to prevent the occurrence of the disease or condition. The PAK inhibitors and compositions are optionally administered to a individual during or as soon as possible after the onset of the symptoms. The administration of the compounds are optionally initiated within the first 48 hours of the onset of the symptoms, preferably within the first 48 hours of the onset of the symptoms, more preferably within the first 6 hours of the onset of the symptoms, and most preferably within 3 hours of the onset of the symptoms. The initial administration is optionally via any route practical, such as, for example, an intravenous injection, a bolus injection, infusion over 5 minutes to about 5 hours, a pill, a capsule, transdermal patch, buccal delivery, and the like, or combination thereof. A PAK inhibitor is optionally administered as soon as is practicable after the onset of a disease or condition is detected or suspected, and for a length of time necessary for the treatment of the disease, such as, for example, from about 1 month to about 3 months. The length of treatment optionally varies for each individual, and the length is then determined using the known criteria. For example, the PAK inhibitor or a formulation containing the PAK inhibitor is administered for at least 2 weeks, preferably about 1 month to about 5 years, and more preferably from about 1 month to about 3 years.

[00353] In some embodiments, the particular choice of compounds depends upon the diagnosis of the attending physicians and their judgment of the condition of an individual and the appropriate treatment protocol. The compounds are optionally administered concurrently (e.g., simultaneously, essentially simultaneously or within the same treatment protocol) or sequentially, depending upon the nature of the disease, disorder, or condition, the condition of an individual, and the actual choice of compounds used. In certain instances, the determination of the order of administration, and the number of repetitions of administration of each therapeutic agent during a treatment protocol, is based on an evaluation of the disease being treated and the condition of an individual.

[00354] In some embodiments, therapeutically-effective dosages vary when the drugs are used in treatment combinations. Methods for experimentally determining therapeutically- effective dosages of drugs and other agents for use in combination treatment regimens are described in the literature. [00355] In some embodiments of the combination therapies described herein, dosages of the co-administered compounds vary depending on the type of co-drug employed, on the specific drug employed, on the disease or condition being treated and so forth. In addition, when co-administered with one or more biologically active agents, the compound provided herein is optionally administered either simultaneously with the biologically active agent(s), or sequentially. In certain instances, if administered sequentially, the attending physician will decide on the appropriate sequence of therapeutic compound described herein in combination with the additional therapeutic agent.

[00356] The multiple therapeutic agents (at least one of which is a therapeutic compound described herein) are optionally administered in any order or even simultaneously. If

simultaneously, the multiple therapeutic agents are optionally provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). In certain instances, one of the therapeutic agents is optionally given in multiple doses. In other instances, both are optionally given as multiple doses. If not simultaneous, the timing between the multiple doses is any suitable timing, e.g., from more than zero weeks to less than four weeks. In some embodiments, the additional therapeutic agent is utilized to achieve reversal or amelioration of a neurological condition, whereupon the therapeutic agent described herein (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) is subsequently administered. In addition, the combination methods, compositions and formulations are not to be limited to the use of only two agents; the use of multiple therapeutic combinations are also envisioned (including two or more compounds described herein).

[00357] In certain embodiments, a dosage regimen to treat, prevent, or ameliorate the condition(s) for which relief is sought, is modified in accordance with a variety of factors. These factors include the disorder from which an individual suffers, as well as the age, weight, sex, diet, and medical condition of an individual. Thus, in various embodiments, the dosage regimen actually employed varies and deviates from the dosage regimens set forth herein. Examples of Pharmaceutical Compositions and Methods of Administration

[00358] Provided herein, in certain embodiments, are compositions comprising a therapeutically effective amount of any compound described herein (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein).

[00359] Pharmaceutical compositions are formulated using one or more physiologically acceptable carriers including excipients and auxiliaries which facilitate processing of the active compounds into preparations which are used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. A summary of pharmaceutical compositions is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Eahston, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins, 1999).

[00360] Provided herein are pharmaceutical compositions that include one or more PA inhibitors and a pharmaceutically acceptable diluent(s), excipient(s), or carrier(s). In addition, the PAK inhibitor is optionally administered as pharmaceutical compositions in which it is mixed with other active ingredients, as in combination therapy. In some embodiments, the pharmaceutical compositions includes other medicinal or pharmaceutical agents, carriers, adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure, and/or buffers. In addition, the pharmaceutical compositions also contain other therapeutically valuable substances.

[00361] A pharmaceutical composition, as used herein, refers to a mixture of a PAK inhibitor with other chemical components, such as carriers, stabilizers, diluents, dispersing agents, suspending agents, thickening agents, and/or excipients. The pharmaceutical

composition facilitates administration of the PAK inhibitor to an organism. In practicing the methods of treatment or use provided herein, therapeutically effective amounts of a PAK inhibitor are administered in a pharmaceutical composition to a mammal having a condition, disease, or disorder to be treated. Preferably, the mammal is a human. A therapeutically effective amount varies depending on the severity and stage of the condition, the age and relative health of an individual, the potency of the PAK inhibitor used and other factors. The PA inhibitor is optionally used singly or in combination with one or more therapeutic agents as components of mixtures.

[00362] The pharmaceutical formulations described herein are optionally administered to a individual by multiple administration routes, including but not limited to, oral, parenteral (e.g., intravenous, subcutaneous, intramuscular), intranasal, buccal, topical, rectal, or transdermal administration routes. The pharmaceutical formulations described herein include, but are not limited to, aqueous liquid dispersions, self-emulsifying dispersions, solid solutions, liposomal dispersions, aerosols, solid dosage forms, powders, immediate release formulations, controlled release formulations, fast melt formulations, tablets, capsules, pills, delayed release

formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate and controlled release formulations.

[00363] The pharmaceutical compositions will include at least one PAK inhibitor, as an active ingredient in free-acid or free-base form, or in a pharmaceutically acceptable salt form. In addition, the methods and pharmaceutical compositions described herein include the use of N- oxides, crystalline forms (also known as polymorphs), as well as active metabolites of these PAK inhibitors having the same type of activity. In some situations, PAK inhibitors exist as tautomers. All tautomers are included within the scope of the compounds presented herein. Additionally, the PAK inhibitor exists in unsolvated as well as solvated forms with

pharmaceutically acceptable solvents such as water, ethanol, and the like. The solvated forms of the PAK inhibitors presented herein are also considered to be disclosed herein.

[00364] "Carrier materials" include any commonly used excipients in pharmaceutics and should be selected on the basis of compatibility with compounds disclosed herein, such as, a PAK inhibitor, and the release profile properties of the desired dosage form. Exemplary carrier materials include, e.g., binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, diluents, and the like.

[00365] Moreover, the pharmaceutical compositions described herein, which include a PAK inhibitor, are formulated into any suitable dosage form, including but not limited to, aqueous oral dispersions, liquids, gels, syrups, elixirs, slurries, suspensions and the like, for oral ingestion by a patient to be treated, solid oral dosage forms, aerosols, controlled release formulations, fast melt formulations, effervescent formulations, lyophilized formulations, tablets, powders, pills, dragees, capsules, delayed release formulations, extended release formulations, pulsatile release formulations, multiparticulate formulations, and mixed immediate release and controlled release formulations. In some embodiments, a formulation comprising a PAK inhibitor is a solid drug dispersion. A solid dispersion is a dispersion of one or more active ingredients in an inert carrier or matrix at solid state prepared by the melting (or fusion), solvent, or melting-solvent methods. (Chiou and Riegelman, Journal of Pharmaceutical Sciences, 60, 1281 (1971)). The dispersion of one or more active agents in a solid diluent is achieved without mechanical mixing. Solid dispersions are also called solid-state dispersions. In some

embodiments, any compound described herein (e.g., a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, as described herein) is formulated as a spray dried dispersion (SDD). An SDD is a single phase amorphous molecular dispersion of a drug in a polymer matrix. It is a solid solution prepared by dissolving the drug and a polymer in a solvent (e.g., acetone, methanol or the like) and spray drying the solution. The solvent rapidly evaporates from droplets which rapidly solidifies the polymer and drug mixture trapping the drug in amorphous form as an amorphous molecular dispersion. In some embodiments, such amorphous dispersions are filled in capsules and/or constituted into oral powders for reconstitution. Solubility of an SDD comprising a drug is higher than the solubility of a crystalline form of a drug or a non-SDD amorphous form of a drug. In some embodiments of the methods described herein, PAK inhibitors are administered as SDDs constituted into appropriate dosage forms described herein.

[00366] Pharmaceutical preparations for oral use are optionally obtained by mixing one or more solid excipient with a PAK inhibitor, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients include, for example, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methylcellulose,

micro crystalline cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose; or others such as: polyvinylpyrrolidone (PVP or povidone) or calcium phosphate. If desired, disintegrating agents are added, such as the cross linked croscarmellose sodium, polyvinylpyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.

[00367] Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions are generally used, which optionally contain gum arabic, talc,

polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments are optionally added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.

[00368] In some embodiments, the solid dosage forms disclosed herein are in the form of a tablet, (including a suspension tablet, a fast-melt tablet, a bite-disintegration tablet, a rapid- disintegration tablet, an effervescent tablet, or a caplet), a pill, a powder (including a sterile packaged powder, a dispensable powder, or an effervescent powder) a capsule (including both soft or hard capsules, e.g., capsules made from animal-derived gelatin or plant-derived HPMC, or "sprinkle capsules"), solid dispersion, solid solution, bioerodible dosage form, controlled release formulations, pulsatile release dosage forms, multiparticulate dosage forms, pellets, granules, or an aerosol. In other embodiments, the pharmaceutical formulation is in the form of a powder. In still other embodiments, the pharmaceutical formulation is in the form of a tablet, including but not limited to, a fast-melt tablet. Additionally, pharmaceutical formulations of a PAK inhibitor are optionally administered as a single capsule or in multiple capsule dosage form. In some embodiments, the pharmaceutical formulation is administered in two, or three, or four, capsules or tablets.

[00369] In another aspect, dosage forms include microencapsulated formulations. In some embodiments, one or more other compatible materials are present in the microencapsulation material. Exemplary materials include, but are not limited to, pH modifiers, erosion facilitators, anti-foaming agents, antioxidants, flavoring agents, and carrier materials such as binders, suspending agents, disintegration agents, filling agents, surfactants, solubilizers, stabilizers, lubricants, wetting agents, and diluents.

[00370] Exemplary microencapsulation materials useful for delaying the release of the formulations including a PAK inhibitor, include, but are not limited to, hydroxypropyl cellulose ethers (HPC) such as Klucel® or Nisso HPC, low-substituted hydroxypropyl cellulose ethers (L- HPC), hydroxypropyl methyl cellulose ethers (HPMC) such as Seppifilm-LC, Pharmacoat®, Metolose SR, Methocel®-E, Opadry YS, PrimaFlo, Benecel MP824, and Benecel MP843, methylcellulose polymers such as Methocel®-A, hydroxypropylmethylcellulose acetate stearate Aqoat (HF-LS, HF-LG,HF-MS) and Metolose®, Ethylcelluloses (EC) and mixtures thereof such as E461, Ethocel®, Aqualon®-EC, Surelease®, Polyvinyl alcohol (PVA) such as Opadry AMB, hydroxyethylcelluloses such as Natrosol®, carboxymethylcelluloses and salts of

carboxymethylcelluloses (CMC) such as Aqualon®-CMC, polyvinyl alcohol and polyethylene glycol co-polymers such as Kollicoat IR®, monoglycerides (Myverol), triglycerides (KLX), polyethylene glycols, modified food starch, acrylic polymers and mixtures of acrylic polymers with cellulose ethers such as Eudragit® EPO, Eudragit® L30D-55, Eudragit® FS 30D

Eudragit® L100-55, Eudragit® L100, Eudragit® S100, Eudragit® RD100, Eudragit® E100, Eudragit® L12.5, Eudragit® S12.5, Eudragit® NE30D, and Eudragit® NE 40D, cellulose acetate phthalate, sepifilms such as mixtures of HPMC and stearic acid, cyclodextrins, and mixtures of these materials.

[00371] The pharmaceutical solid oral dosage forms including formulations described herein, which include a PAK inhibitor, are optionally further formulated to provide a controlled release of the PAK inhibitor. Controlled release refers to the release of the PAK inhibitor from a dosage form in which it is incorporated according to a desired profile over an extended period of time. Controlled release profiles include, for example, sustained release, prolonged release, pulsatile release, and delayed release profiles. In contrast to immediate release compositions, controlled release compositions allow delivery of an agent to a individual over an extended period of time according to a predetermined profile. Such release rates provide therapeutically effective levels of agent for an extended period of time and thereby provide a longer period of pharmacologic response while minimizing side effects as compared to conventional rapid release dosage forms. Such longer periods of response provide for many inherent benefits that are not achieved with the corresponding short acting, immediate release preparations.

[00372] In other embodiments, the formulations described herein, which include a PAK inhibitor, are delivered using a pulsatile dosage form. A pulsatile dosage form is capable of providing one or more immediate release pulses at predetermined time points after a controlled lag time or at specific sites. Pulsatile dosage forms including the formulations described herein, which include a PAK inhibitor, are optionally administered using a variety of pulsatile formulations that include, but are not limited to, those described in U.S. Pat. Nos. 5,011,692, 5,017,381, 5,229,135, and 5,840,329. Other pulsatile release dosage forms suitable for use with the present formulations include, but are not limited to, for example, U.S. Pat. Nos. 4,871,549, 5,260,068, 5,260,069, 5,508,040, 5,567,441 and 5,837,284.

[00373] Liquid formulation dosage forms for oral administration are optionally aqueous suspensions selected from the group including, but not limited to, pharmaceutically acceptable aqueous oral dispersions, emulsions, solutions, elixirs, gels, and syrups. See, e.g., Singh et al, Encyclopedia of Pharmaceutical Technology, 2nd Ed., pp. 754-757 (2002). In addition to the PAK inhibitor, the liquid dosage forms optionally include additives, such as: (a) disintegrating agents; (b) dispersing agents; (c) wetting agents; (d) at least one preservative, (e) viscosity enhancing agents, (f) at least one sweetening agent, and (g) at least one flavoring agent. In some embodiments, the aqueous dispersions further includes a crystal- forming inhibitor.

[00374] In some embodiments, the pharmaceutical formulations described herein are elf- emulsifying drug delivery systems (SEDDS). Emulsions are dispersions of one immiscible phase in another, usually in the form of droplets. Generally, emulsions are created by vigorous mechanical dispersion. SEDDS, as opposed to emulsions or microemulsions, spontaneously form emulsions when added to an excess of water without any external mechanical dispersion or agitation. An advantage of SEDDS is that only gentle mixing is required to distribute the droplets throughout the solution. Additionally, water or the aqueous phase is optionally added just prior to administration, which ensures stability of an unstable or hydrophobic active ingredient. Thus, the SEDDS provides an effective delivery system for oral and parenteral delivery of hydrophobic active ingredients. In some embodiments, SEDDS provides

improvements in the bioavailability of hydrophobic active ingredients. Methods of producing self-emulsifying dosage forms include, but are not limited to, for example, U.S. Pat. Nos.

5,858,401, 6,667,048, and 6,960,563.

[00375] Suitable intranasal formulations include those described in, for example, U.S. Pat. Nos. 4,476,116, 5,116,817 and 6,391,452. Nasal dosage forms generally contain large amounts of water in addition to the active ingredient. Minor amounts of other ingredients such as pH adjusters, emulsifiers or dispersing agents, preservatives, surfactants, gelling agents, or buffering and other stabilizing and solubilizing agents are optionally present.

[00376] For administration by inhalation, the PAK inhibitor is optionally in a form such as an aerosol, a mist or a powder. Pharmaceutical compositions described herein are

conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane,

trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit is determined by providing a valve to deliver a metered amount. Capsules and cartridges of, such as, by way of example only, gelatin for use in an inhaler or insufflator are formulated containing a powder mix of the PAK inhibitor and a suitable powder base such as lactose or starch.

[00377] Buccal formulations that include a PAK inhibitor include, but are not limited to,

U.S. Pat. Nos. 4,229,447, 4,596,795, 4,755,386, and 5,739,136. In addition, the buccal dosage forms described herein optionally further include a bioerodible (hydro lysable) polymeric carrier that also serves to adhere the dosage form to the buccal mucosa. The buccal dosage form is fabricated so as to erode gradually over a predetermined time period, wherein the delivery of the PAK inhibitor, is provided essentially throughout. Buccal drug delivery avoids the

disadvantages encountered with oral drug administration, e.g., slow absorption, degradation of the active agent by fluids present in the gastrointestinal tract and/or first-pass inactivation in the liver. The bioerodible (hydrolysable) polymeric carrier generally comprises hydrophilic (water- soluble and water-swellable) polymers that adhere to the wet surface of the buccal mucosa. Examples of polymeric carriers useful herein include acrylic acid polymers and co, e.g., those known as "carbomers" (Carbopol®, which may be obtained from B.F. Goodrich, is one such polymer). Other components also be incorporated into the buccal dosage forms described herein include, but are not limited to, disintegrants, diluents, binders, lubricants, flavoring, colorants, preservatives, and the like. For buccal or sublingual administration, the compositions optionally take the form of tablets, lozenges, or gels formulated in a conventional manner.

[00378] Transdermal formulations of a PAK inhibitor are administered for example by those described in U.S. Pat. Nos. 3,598,122, 3,598,123, 3,710,795, 3,731,683, 3,742,951, 3,814,097, 3,921,636, 3,972,995, 3,993,072, 3,993,073, 3,996,934, 4,031,894, 4,060,084, 4,069,307, 4,077,407, 4,201,211, 4,230,105, 4,292,299, 4,292,303, 5,336,168, 5,665,378, 5,837,280, 5,869,090, 6,923,983, 6,929,801 and 6,946,144.

[00379] The transdermal formulations described herein include at least three components:

(1) a formulation of a PAK inhibitor; (2) a penetration enhancer; and (3) an aqueous adjuvant. In addition, transdermal formulations include components such as, but not limited to, gelling agents, creams and ointment bases, and the like. In some embodiments, the transdermal formulation further includes a woven or non- woven backing material to enhance absorption and prevent the removal of the transdermal formulation from the skin. In other embodiments, the transdermal formulations described herein maintain a saturated or supersaturated state to promote diffusion into the skin.

[00380] In some embodiments, formulations suitable for transdermal administration of a

PAK inhibitor employ transdermal delivery devices and transdermal delivery patches and are lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive. Such patches are optionally constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents. Still further, transdermal delivery of the PAK inhibitor is optionally accomplished by means of iontophoretic patches and the like. Additionally, transdermal patches provide controlled delivery of the PAK inhibitor. The rate of absorption is optionally slowed by using rate-controlling membranes or by trapping the PAK inhibitor within a polymer matrix or gel. Conversely, absorption enhancers are used to increase absorption. An absorption enhancer or carrier includes absorbable pharmaceutically acceptable solvents to assist passage through the skin. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the PAK inhibitor optionally with carriers, optionally a rate controlling barrier to deliver the PAK inhibitor to the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.

[00381] Formulations that include a PAK inhibitor suitable for intramuscular,

subcutaneous, or intravenous injection include physiologically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and non-aqueous carriers, diluents, solvents, or vehicles including water, ethanol, polyols

(propyleneglycol, polyethylene-glycol, glycerol, cremophor and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.

Proper fluidity is maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. Formulations suitable for subcutaneous injection also contain optional additives such as preserving, wetting, emulsifying, and dispensing agents.

[00382] For intravenous injections, a PAK inhibitor is optionally formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. For other parenteral injections, appropriate formulations include aqueous or nonaqueous solutions, preferably with

physiologically compatible buffers or excipients.

[00383] Parenteral injections optionally involve bolus injection or continuous infusion.

Formulations for injection are optionally presented in unit dosage form, e.g., in ampoules or in multi dose containers, with an added preservative. In some embodiments, the pharmaceutical composition described herein are in a form suitable for parenteral injection as a sterile suspensions, solutions or emulsions in oily or aqueous vehicles, and contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the PAK inhibitor in water soluble form. Additionally, suspensions of the PAK inhibitor are optionally prepared as appropriate oily injection suspensions. [00384] In some embodiments, the PAK inhibitor is administered topically and formulated into a variety of topically administrable compositions, such as solutions,

suspensions, lotions, gels, pastes, medicated sticks, balms, creams or ointments. Such pharmaceutical compositions optionally contain solubilizers, stabilizers, tonicity enhancing agents, buffers and preservatives.

[00385] The PAK inhibitor is also optionally formulated in rectal compositions such as enemas, rectal gels, rectal foams, rectal aerosols, suppositories, jelly suppositories, or retention enemas, containing conventional suppository bases such as cocoa butter or other glycerides, as well as synthetic polymers such as polyvinylpyrrolidone, PEG, and the like. In suppository forms of the compositions, a low-melting wax such as, but not limited to, a mixture of fatty acid glycerides, optionally in combination with cocoa butter is first melted.

Examples of Methods of Dosing and Treatment Regimens

[00386] The PAK inhibitor is optionally used in the preparation of medicaments for the prophylactic and/or therapeutic treatment of a neurological condition that would benefit, at least in part, from amelioration of symptoms. In addition, a method for treating any of the diseases or conditions described herein in a individual in need of such treatment, involves administration of pharmaceutical compositions containing at least one PAK inhibitor described herein, or a pharmaceutically acceptable salt, pharmaceutically acceptable N-oxide, pharmaceutically active metabolite, pharmaceutically acceptable prodrug, or pharmaceutically acceptable solvate thereof, in therapeutically effective amounts to said individual.

[00387] In the case wherein the patient's condition does not improve, upon the doctor's discretion the administration of the PAK inhibitor is optionally administered chronically, that is, for an extended period of time, including throughout the duration of the patient's life in order to ameliorate or otherwise control or limit the symptoms of the patient's disease or condition.

[00388] In the case wherein the patient's status does improve, upon the doctor's discretion the administration of the PAK inhibitor is optionally given continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a "drug holiday"). The length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days. The dose reduction during a drug holiday includes from 10%- 100%, including, by way of example only, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%. [00389] Once improvement of the patient's conditions has occurred, a maintenance dose is administered if necessary. Subsequently, the dosage or the frequency of administration, or both, is reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained. In some embodiments, patients require intermittent treatment on a long- term basis upon any recurrence of symptoms.

[00390] In some embodiments, the pharmaceutical compositions described herein are in unit dosage forms suitable for single administration of precise dosages. In unit dosage form, the formulation is divided into unit doses containing appropriate quantities of one or more PAK inhibitor. In some embodiments, the unit dosage is in the form of a package containing discrete quantities of the formulation. Non-limiting examples are packaged tablets or capsules, and powders in vials or ampoules. In some embodiments, aqueous suspension compositions are packaged in single-dose non-reclosable containers. Alternatively, multiple-dose reclosable containers are used, in which case it is typical to include a preservative in the composition. By way of example only, formulations for parenteral injection are presented in unit dosage form, which include, but are not limited to ampoules, or in multi dose containers, with an added preservative.

[00391] The daily dosages appropriate for the PAK inhibitor are from about 0.01 to 2.5 mg/kg per body weight. An indicated daily dosage in the larger mammal, including, but not limited to, humans, is in the range from about 0.5 mg to about 1000 mg, conveniently administered in divided doses, including, but not limited to, up to four times a day or in extended release form. Suitable unit dosage forms for oral administration include from about 1 to 500 mg active ingredient, from about 1 to 250 mg of active ingredient, or from about 1 to about 100 mg active ingredient. The foregoing ranges are merely suggestive, as the number of variables in regard to an individual treatment regime is large, and considerable excursions from these recommended values are not uncommon. Such dosages are optionally altered depending on a number of variables, not limited to the activity of the PAK inhibitor used, the disease or condition to be treated, the mode of administration, the requirements of an individual individual, the severity of the disease or condition being treated, and the judgment of the practitioner.

[00392] Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the

determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD50 and ED50. PAK inhibitors exhibiting high therapeutic indices are preferred. The data obtained from cell culture assays and animal studies is optionally used in formulating a range of dosage for use in human. The dosage of such PAK inhibitors lies preferably within a range of circulating concentrations that include the ED50 with minimal toxicity. The dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized.

Assays for identification and characterization of PAK inhibitors

[00393] Small molecule PAK inhibitors are optionally identified in high-throughput in vitro or cellular assays as described in, e.g., Yu et al (2001), J Biochem (Tokyo); 129(2):243- 251; Rininsland et al (2005), BMC Biotechnol, 5: 16; and Allen et al (2006), ACS Chem Biol; l(6):371-376. PAK inhibitors suitable for the methods described herein are available from a variety of sources including both natural (e.g., plant extracts) and synthetic. For example, candidate PAK inhibitors are isolated from a combinatorial library, i.e., a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical "building blocks." For example, a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be

synthesized through such combinatorial mixing of chemical building blocks, as desired.

Theoretically, the systematic, combinatorial mixing of 100 interchangeable chemical building blocks results in the synthesis of 100 million tetrameric compounds or 10 billion pentameric compounds. See Gallop et al. (1994), J. Med. Chem. 37(9), 1233. Each member of a library may be singular and/or may be part of a mixture (e.g. a "compressed library"). The library may comprise purified compounds and/or may be "dirty" (i.e., containing a quantity of impurities). Preparation and screening of combinatorial chemical libraries are documented methodologies. See Cabilly, ed., Methods in Molecular Biology, Humana Press, Totowa, NJ, (1998).

Combinatorial chemical libraries include, but are not limited to: diversomers such as hydantoins, benzodiazepines, and dipeptides, as described in, e.g., Hobbs et al. (1993), Proc. Natl. Acad. Sci. U.S.A. 90, 6909; analogous organic syntheses of small compound libraries, as described in Chen et al. (1994), J. Amer. Chem. Soc, 116: 2661; Oligocarbamates, as described in Cho, et al.

(1993), Science 261, 1303; peptidyl phosphonates, as described in Campbell et al. (1994), J. Org. Chem., 59: 658; and small organic molecule libraries containing, e.g., thiazolidinones and metathiazanones (U.S. Pat. No. 5,549,974), pyrrolidines (U.S. Pat. Nos. 5,525,735 and

5,519,134), benzodiazepines (U.S. Pat. No. 5,288,514). In addition, numerous combinatorial libraries are commercially available from, e.g., ComGenex (Princeton, NX); Asinex (Moscow, Russia); Tripos, Inc. (St. Louis, MO); ChemStar, Ltd. (Moscow, Russia); 3D Pharmaceuticals (Exton, PA); and Martek Biosciences (Columbia, MD).

[00394] Devices for the preparation of combinatorial libraries are commercially available

(see, e.g., 357 MPS, 390 MPS from Advanced Chem Tech, Louisville, KY; Symphony from Rainin, Woburn, MA; 433A from Applied Biosystems, Foster City, CA; and 9050 Plus from Millipore, Bedford, MA). A number of robotic systems have also been developed for solution phase chemistries. These systems include automated workstations like the automated synthesis apparatus developed by Takeda Chemical Industries, LTD (Osaka, Japan), and many robotic systems utilizing robotic arms (Zymate II). Any of the above devices are optionally used to generate combinatorial libraries for identification and characterization of PAK inhibitors which mimic the manual synthetic operations performed by small molecule PAK inhibitors suitable for the methods described herein. Any of the above devices are optionally used to identify and characterize small molecule PAK inhibitors suitable for the methods disclosed herein. In many of the embodiments disclosed herein, PAK inhibitors, PAK binding molecules, and PAK clearance agents are disclosed as polypeptides or proteins (where polypeptides comprise two or more amino acids). In these embodiments, the inventors also contemplate that PAK inhibitors, binding molecules, and clearance agents also include peptide mimetics based on the

polypeptides, in which the peptide mimetics interact with PAK or its upstream or downstream regulators by replicating the binding or substrate interaction properties of PAK or its regulators. Nucleic acid aptmers are also contemplated as PAK inhibitors, binding molecules, and clearance agents, as are small molecules other than peptides or nucleic acids. For example, in some embodiments small molecule PAK binding partners, inhibitors, or clearance agents, or small molecule agonists or antagonists of PAK modulators or targets, are designed or selected based on analysis of the structure of PAK or its modulators or targets and binding interactions with interacting molecules, using "rational drug design" (see, for example Jacobsen et al. (2004)

Molecular Interventions 4:337-347; Shi et al. (2007) Bioorg. Med. Chem. Lett. 17:6744-6749).

[00395] The identification of potential PAK inhibitors is determined by, for example, assaying the in vitro kinase activity of PAK in the presence of candidate inhibitors. In such assays, PAK and/or a characteristic PAK fragment produced by recombinant means is contacted with a substrate in the presence of a phosphate donor (e.g., ATP) containing radiolabeled phosphate, and PAK-dependent incorporation is measured. "Substrate" includes any substance containing a suitable hydroxyl moiety that can accept the γ-phosphate group from a donor molecule such as ATP in a reaction catalyzed by PAK. The substrate may be an endogenous substrate of PAK, i.e. a naturally occurring substance that is phosphorylated in unmodified cells by naturally-occurring PAK or any other substance that is not normally phosphorylated by PAK in physiological conditions, but may be phosphorylated in the employed conditions. The substrate may be a protein or a peptide, and the phosphrylation reaction may occur on a serine and/or threonine residue of the substrate. For example, specific substrates, which are commonly employed in such assays include, but are not limited to, histone proteins and myelin basic protein. In some embodiments, PAK inhibitors are identified using IMAP® technology.

[00396] Detection of PAK dependent phosphorylation of a substrate can be quantified by a number of means other than measurement of radiolabeled phosphate incorporation. For example, incorporation of phosphate groups may affect physio chemical properties of the substrate such as electrophoretic mobility, chromatographic properties, light absorbance, fluorescence, and phosphorescence. Alternatively, monoclonal or polyclonal antibodies can be generated which selectively recognize phosphorylated forms of the substrate from non- phosphorylated forms whereby allowing antibodies to function as an indicator of PAK kinase activity.

[00397] High-throughput PAK kinase assays can be performed in, for example, microtiter plates with each well containing PAK kinase or an active fragment thereof, substrate covalently linked to each well, P32 radio labled ATP and a potential PAK inhibitor candidate. Microtiter plates can contain 96 wells or 1536 wells for large scale screening of combinatorial library compounds. After the phosphorylation reaction has completed, the plates are washed leaving the bound substrate. The plates are then detected for phosphate group incorporation via

autoradiography or antibody detection. Candidate PAK inhibitors are identified by their ability to decease the amount of PAK phosphotransferase ability upon a substrate in comparison with PAK phosphotransferase ability alone.

[00398] The identification of potential PAK inhibitors may also be determined, for example, via in vitro competitive binding assays on the catalytic sites of PAK such as the ATP binding site and/or the substrate binding site. For binding assays on the ATP binding site, a known protein kinase inhibitor with high affinity to the ATP binding site is used such as staurosporine. Staurosporine is immobolized and may be fluorescently labeled, radiolabeled or in any manner that allows detection. The labeled staurosporine is introduced to recombinantly expressed PAK protein or a fragment thereof along with potential PAK inhibitor candidates. The candidate is tested for its ability to compete, in a concentration-dependant manner, with the immobolized staurosporine for binding to the PAK protein. The amount of staurosporine bound PAK is inversely proportional to the affinity of the candidate inhibitor for PAK. Potential inhibitors would decrease the quantifiable binding of staurosporine to PAK. See e.g., Fabian et al (2005) Nat. Biotech., 23:329. Candidates identified from this competitive binding assay for the ATP binding site for PAK would then be further screened for selectivity against other kinases for PAK specificity.

[00399] The identification of potential PAK inhibitors may also be determined, for example, by in cyto assays of PAK activity in the presence of the inhibitor candidate. Various cell lines and tissues may be used, including cells specifically engineered for this purpose. In cyto screening of inhibitor candidates may assay PAK activity by monitoring the downstream effects of PAK activity. Such effects include, but are not limited to, the formation of peripheral actin microspikes and or associated loss of stress fibers as well as other cellular responses such as growth, growth arrest, differentiation, or apoptosis. See e.g., Zhao et al, (1998) Mol. Cell.

Biol. 18:2153. For example in a PAK yeast assay, yeast cells grow normally in glucose medium. Upon exposure to galactose however, intracellular PAK expression is induced, and in turn, the yeast cells die. Candidate compounds that inhibit PAK activity are identified by their ability to prevent the yeast cells from dying from PAK activation.

[00400] Alternatively, PAK-mediated phosphorylation of a downstream target of PAK can be observed in cell based assays by first treating various cell lines or tissues with PAK inhibitor candidates followed by lysis of the cells and detection of PAK mediated events. Cell lines used in this experiment may include cells specifically engineered for this purpose. PAK mediated events include, but are not limited to, PAK mediated phosphorylation of downstream PAK mediators. For example, phosphorylation of downstream PAK mediators can be detected using antibodies that specifically recognize the phosphorylated PAK mediator but not the unphosphorylated form. These antibodies have been described in the literature and have been extensively used in kinase screening campaigns. In some instances a phospho LIMK antibody is used after treatment of HeLa cells stimulated with EGF or sphingosine to detect downstream PAK signaling events.

[00401] The identification of potential PAK inhibitors may also be determined, for example, by in vivo assays involving the use of animal models, including transgenic animals that have been engineered to have specific defects or carry markers that can be used to measure the ability of a candidate substance to reach and/or affect different cells within the organism. In certain embodiments, primate models of Huntington's disease (for example transgenic rhesus macaques described in Chan et al, Biology and Nature, May, 2008) can be used to model Huntington's disease. Thus, identification of PAK inhibitors can comprise administering a candidate PAK inhibitor to an organism and observing for delayed progression or improvement in working memory scores, for example as assessed by evaluation in a morris water maze as a readout for PA inhibition. Cognitive function can be assessed using behavioral paradigms in rodents, for example in the Novel Object Recognition Task or using a "win-shift" paradigm in an 8-arm radial maze. In some embodiments, a mouse model described in Menalled et al. J Comp Neurol. 2003; 465(1): 11-26 is used to model early motor and neuropathological anomalies in a knock-in mouse model of Huntington's disease with 140 CAG repeats. In other embodiments, mouse models of neurofibromatosis (e.g., mice mice that carry linked germ line mutations in Nfl and p53 as described in Chichowski Science. 1999, 286(5447):2172-6) are used. In other embodiments, the mouse model of neurofibromatosis is a chimeric mouse compased in part from Nfl-/- cells. Thus, identification of PAK inhibitors can comprise administering a candidate PAK inhibitor to an organism and observing for development of peripheral nerve sheath tumors, as a readout for PAK inhibition.

[00402] Administration of the candidate to the animal is via any clinical or non-clinical route, including but not limited to oral, nasal, buccal and/or topical administrations. Additionally or alternatively, administration may be intratracheal instillation, bronchial instillation, intradermal, subcutaneous, intramuscular, intraperitoneal, inhalation, and/or intravenous injection.

[00403] Changes in spine morphology are detected using any suitable method, e.g., by use of 3D and/or 4D real time interactive imaging and visualization. In some instances, the Imaris suite of products (available from Bitplane Scientific Solutions) provides functionality for visualization, segmentation and interpretation of 3D and 4D microscopy datasets obtained from confocal and wide field microscopy data.

EXAMPLES

[00404] The following examples are to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever.

[00405] All synthetic chemistry was performed in standard laboratory glassware unless indicated otherwise in the examples. Commercial reagents were used as received. Analytical

LC/MS was performed on an Agilent 1200 system with a variable wavelength detector and

Agilent 6140 Single quadrupole mass spectrometer, alternating positive and negative ion scans.

Retention times were determined from the extracted 220 nm chromatogram. 1H NMR was performed on a Bruker DRX-400 at 400 MHz. Microwave reactions were performed in a

Biotage Initiator using the instrument software to control heating time and pressure.

Hydrogenation reactions were performed on a H-Cube using the commercially available catalyst cartridges. Silica gel chromatography was performed manually. [00406] Preparative HPLC was performed on a Waters 1525/2487 with UV detection at

220 nm and manual collection.

[00407] Analytical LC/MS method A:

[00408] HPLC column: Zorbax SB-C18, 3.5 μιη, 2.1 mm x 30 mm, maintained at 40 °C.

[00409] HPLC Gradient: 0.4 mL/min, 95:5:0.1 water :acetonitrile: formic acid for 0.1 min then to 5:95:0.1 water :acetonitrile: formic acid in 3.9 min, maintaining for 0.5 min.

[00410] Analytical LC/MS method B:

[00411] HPLC column: Kinetex, 2.6 μιη, CI 8, 50 x 2.1mm, maintained at 40 °C.

[00412] HPLC Gradient: 1.0 mL/min, 95 :5 :0.1 water :acetonitrile: formic acid to 5 :95 :0.1 water :acetonitrile: formic acid in 2.5 min, maintaining for 0.5 min.

[00413] Analytical LC/MS method C was performed on a Shimadzu system with an attached API 165 single quadrupole mass spectrometer. Retention times were determined from the 220 nm chromatogram.

[00414] HPLC column: Phenomenex, CI 8, 2.5 μιη, 20 x 2mm, maintained at 25 °C.

[00415] HPLC Gradient: 0.5 mL/min, 95 :5 :0.02 water:acetonitrile:CF 3 COOH to

5:95:0.02 water:acetonitrile:CF 3 COOH in 2.9 min, maintaining for 0.9 min.

[00416] Preparative HPLC method A: Preparative HPLC was performed on a Waters

1525/2487 with UV detection at 220 nm and manual collection.

[00417] HPLC column: Zorbax SB-C18 21.2 x 100 mm.

[00418] HPLC Gradient: 20 mL/min, 95:5:0.1 water:methanol:formic acid to 5:95:0.1 water:methanol:formic acid; the gradient shape was optimized for individual separations.

[00419] Preparative HPLC method B:

[00420] HPLC column: Reprosil-Pur CI 8-AQ 250 x 20 mm.

[00421] HPLC Gradient: 25 mL/min, 25:75:0.02 acetonitrile:water:trifluoroacetic acid to

100:0:0.02 acetonitrile:water:trifluoroacetic acid; the gradient shape was optimized for individual separations. Example 1: Synthesis of 8-(7-methoxy-2,3-dihydro-1H-inden-1-yl)-2-(4-(4- methylpiperazin-1-yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H )-one (Cmpd 5).

Preparation of Intermediate compounds:

[00422] Intermediate 1 : Synthesis of 3-bromo-2-chloromethyl-thiophene.

[00423] Step 1 : Synthesis of (3-bromo-thiophen-2-yl -methanol. To a solution of 3- bromothiophene-2-carbaldehyde (500 mg, 2.62 mmol) in methanol (10 mL) was added sodium borohydride (169 mg, 4.47 mmol) in small portions at 0 °C and the reaction was stirred for 2 hrs The solvent was evaporated and the residue partitioned between ethyl acetate (20 mL) and 10% ammonium chloride solution (10 mL). The organic layer was washed with water (10 mL), dried over sodium sulfate and evaporated. The title compound (505 mg, 2.62 mmol, 100%) was obtained as a yellow oil.

[00424] Step 2: Synthesis of 3-bromo-2-chloromethyl-thiophene. (3-Bromo-thiophen-2- yl)-methanol (505 mg, 2.62 mmol) was dissolved in dichloromethane (20 mL) and thionyl chloride (357 μί, 4.89 mmol) was added. The mixture was heated at reflux for 4 hrs, then evaporated. The crude product was purified by column chromatography on silica, eluting with dichloromethane. The title compound (466 mg, 2.20 mmol, 84%) was obtained as a pale yellow oil.

[00425] Intermediate 2: Synthesis of 4-Bromo-5-chloromethyl-thiazole. [00426] The following intermediate was made by the method of Intermediate 1 using the appropriate bromo-substituted heterocycle: 4-Bromo-5-chloromethyl-thiazole.

Synthesis of 8-(3-Cvclopropyl-thiophen-2-ylmethyl)-2-[4-(4-methyl-piperaz in-1-yl)- phenylaminol -8H-pyrido [2,3-dl pyrimidin-7-one.

[00427] Step 1 : Synthesis of 8-(3-bromo-thiophen-2-ylmethyl)-2-methylsulfanyl-8H- pyrido[2,3-d]-pyrimidin-7-one. To a solution of 2-methylsulfanyl-8H-pyrido[2,3-d]pyrimidin-7- one (300 mg, 1.55 mmol) in anhydrous dimethylformamide (10 mL) sodium hydride (60% in mineral oil, 93 mg, 2.33 mmol) was added and stirred at room temperature. After 30 min, a solution of 3-bromo-2-chloromethyl-thiophene (426 mg, 2.01 mmol) in anhydrous

dimethylformamide (1 mL) was added slowly and the stirring was continued for 18 h. The resulting mixture was poured into ice water (50 g) then extracted with ethyl acetate (3 x 30 mL). The combined organic layers was dried over sodium sulfate, filtered and evaporated. The title compound (686 mg, 1.86 mmol, quant.) was obtained as a light brown solid. ESMS m/z 368 (M+H)+.

[00428] Step 2: Synthesis of 8-(3-cyclopropyl-thiophen-2-ylmethyl)-2-methylsulfanyl-

8H-pyrido[2,3-d]pyrimidin-7-one. 8-(3-Bromo-thiophen-2-ylmethyl)-2-methylsulfanyl-8H- pyrido[2,3-d]pyrimidin-7-one (350 mg, 0.95 mmol), cyclopropylboronic acid (245 mg, 2.85 mmol), K3P04 (605 g, 2.85 mmol) and PdC12(Pcy3)2 (70 mg, 0.09 mmol) were mixed under argon in a degassed mixture of toluene and water (20: 1, 8 mL). The resulting suspension was refluxed for 14 h, cooled and diluted with water (8 mL). The two phases were separated, the aqueous layer was washed extracted with toluene (2 x 10 mL), and the combined organic layers were dried over sodium sulfate, filtered and evaporated. The residue was purified by column chromatography using dichloromethane: ethyl acetate (100:3) as the eluent. The title compound (156 mg, 0.47 mmol, 49%) was obtained as a solid. ESMS m/z 330 (M+H)+.

[00429] Step 3: Synthesis of 8-(3-cyclopropyl-thiophen-2-ylmethyl)-2-methanesulfinyl-

8H-pyrido[2,3-d]-pyrimidin-7-one. To a solution of 8-(3-cyclopropyl-thiophen-2-ylmethyl)-2- methylsulfanyl-8H-pyrido[2,3-d]pyrimidin-7-one (155 mg, 0.47 mmol) in dichloromethane (5 mL) was added 3-chloroperbenzoic acid (77%, 106 mg, 0.47 mmol) in dichloromethane (5 mL) at 0-5 °C and the mixture was stirred at 0-5 °C for 1 h. The reaction mixture was washed with saturated sodium bicarbonate solution (1 x 10 mL) then water (1 x 10 mL). The organic layer was dried over sodium sulfate, filtered and evaporated to dryness. The crude compound was purified by column chromatography using dichloromethane: methanol (100:3) to yield 8-(3- cyclopropyl-thiophen-2-ylmethyl)-2-methanesulfinyl-8Hpyrido[ 2,3-d]pyrimidin-7-one as a white solid (118 mg, 0.34 mmol, 72%). ESMS m z 346 (M+H)+. [00430] Step 4: Synthesis of 8-(3-Cyclopropyl-thiophen-2-ylmethyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one . 8-(3-Cyclopropyl-thiophen-2- ylmethyl)-2-methanesulfinyl-8Hpyrido[2,3-d]pyrimidin-7-one (118 mg, 0.34 mmol) and 4-(4- methylpiperazino)aniline (65 g, 0.34 mmol) were stirred at 140 °C for 6 h. The reaction mixture was purified by column chromatography using dichloromethane: methanol (9: 1) and the solid product was suspended in diisopropyl ether and the solid was collected to give the title compound (52 mg, 0.11 mmol, 32%) as a light yellow solid. ESMS m/z 473 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.49 (s, 1H) 7.49 (d, J = 9.3Hz, 1H) 7.46 (d, J = 8.5Hz, 2H) 7.18 (s, 1H) 7.00 (d, J = 5.3Hz, 1H) 6.92 (d, J= 8.5Hz, 2H) 6.40 - 6.49 (m, 2H) 5.78 (s, 2H) 3.12 - 3.25 (m, 4H) 2.60 (d, J= 4.5Hz, 4H) 2.30 - 2.39 (m, 4H) 0.75 - 0.91 (m, 2H) 0.54 - 0.65 (m, 2H).

Example 2: Synthesis of 8-((4-cyclopropylthiazol-5-yl)methyl)-2-(4-(4-methylpiperazi n-1- yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (Compd 6).

[00431] The following compound was made by the method of Example 1 using the appropriate aryl methyl halide at Step 1 and aniline at Step 4. The aryl methyl halide was synthesized by the method used for Intermediate 1.

Example 3: Synthesis of 8-(5-cyclopropyl-thiazol-4-ylmethyl)-2-[4-(4-methyl-piperazi n-1- yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one (Compd 9).

Preparation of Intermediate compounds:

[00432] Intermediate 3 : Synthesis of 4-chloromethyl-5-cyclopropyl-thiazole.

[00433] Step 1 : Synthesis of 5-cyclopropyl-thiazole-4-carboxylic acid methyl ester. 5-

Bromothiazole-4-carboxylic acid methyl ester (325 mg, 1.46 mmol), cyclopropylboronic acid (282 mg, 3.28 mmol), K3P04 (697 g, 3.28 mmol) and Pd(PPh3)4 (110 mg, 0.09 mmol) were mixed under argon in a degassed mixture of toluene and water (20: 1, 9 mL). The resulting suspension was irradiated for 2 h at 120 °C in a microwave reactor. The reaction mixture was diluted with water (8 mL), the two phases were separated, the aqueous layer was washed with dichloromethane (3 x 10 mL), and the combined organic layers was dried over sodium sulfate, filtered and evaporated. The residue was purified by column chromatography using n- hexane:ethyl acetate (2: 1) as eluent. The title compound (137 mg, 0.75 mmol, 51%) was obtained as a yellow oil. ESMS m/z 184 (M+H)+.

[00434] Step 2: Synthesis of (5-cyclopropyl-thiazol-4-yl)-methanol. To a solution of 5- cyclopropyl-thiazole-4-carboxylic acid methyl ester (219 mg, 1.19 mmol) in anhydrous tetrahydrofuran (5 mL) at 0 °C lithium aluminum hydride (45 mg, 1.19 mmol) was added in small portions and the mixture was stirred for 2 h. Water (5 mL) was added dropwise, then the mixture was allowed to reach room temperature. The mixture was diluted with ethyl acetate (20 mL) and brine (10 mL), the two phases were separated, and the aqueous layer was extracted with ethyl acetate (10 mL). The combined organic layers were dried over sodium sulfate, filtered and evaporated. The title compound (127 mg, 0.82 mmol, 69%) was obtained as a yellow oil. ESMS m/z 156 (M+H)+.

[00435] Step 3: Synthesis of 4-chloromethyl-5-cyclopropyl-thiazole. (5-Cyclopropyl- thiazol-4-yl)-methanol (127 mg, 0.82 mmol) was dissolved in anhydrous dichloromethane (5 mL) and thionyl chloride (300 μί, 4.09 mmol) was added. After refluxing for 4 h the reaction mixture was evaporated. The title compound (140 mg, 0.81 mmol, 99%) was obtained as a brown oil.

[00436] Intermediate 4: Synthesis of 5 -Chloromethyl-4-cyclopropyl-l -methyl- 1H- imidazole

[00437] The following intermediate was made by the method of Intermediate 3 using the appropriate bromo-substituted heterocycle: 5 -Chloromethyl-4-cyclopropyl-l -methyl- 1H- imidazole.

Synthesis of 8-(5-cyclopropyl-thiazol-4-ylmethyl -2-[4-(4-methyl-piperazin- 1 -yl - phenylamino]-8H-pyrido[2.3-d]pyrimidin-7-one.

[00438] Step 1 : Synthesis of 8-(5-cyclopropyl-thiazol-4-ylmethyl)-2-methylsulfanyl-8H- pyrido[2,3-d]-pyrimidin-7-one. To a solution of 2-methylsulfanyl-8H-pyrido[2,3-d]pyrimidin-7- one (120 mg, 0.62 mmol) in anhydrous dimethylformamide (2 mL), sodium hydride (60% in mineral oil, 37 mg, 0.93 mmol) was added and stirred at room temperature. After 30 min, a solution of 4-chloromethyl-5-cyclopropylthiazole (140 mg, 0.81 mmol) in anhydrous dimethylformamide (2 mL) was added slowly and the stirring was continued for 18 h. The resulting mixture was poured into ice water (20 g), and extracted with dichloromethane (3 x 20 mL). The combined organic layers were dried over sodium sulfate, filtered and evaporated. The residue was purified by column chromatography using dichloromethane: ethyl acetate (1 : 1) as eluent. The title compound (100 mg, 0.30 mmol, 48%) was obtained as a beige foam. ESMS m/z 331 (M+H)+.

[00439] Step 2: Synthesis of 8-(5-cyclopropyl-thiazol-4-ylmethyl)-2-methanesulfinyl-8H- pyrido[2,3-d]pyrimidin-7-one. To a solution of 8-(5-cyclopropyl-thiazol-4-ylmethyl)-2- methylsulfanyl-8H-pyrido[2,3-d]-pyrimidin-7-one (100 mg, 0.30 mmol) in dichloromethane (10 mL) was added 3-chloroperbenzoic acid (77%, 61 mg, 0.27 mmol) in dichloromethane (2 mL) at 0-5 °C and the mixture was stirred at 0-5 °C for 1 h. The reaction mixture was washed with saturated sodium bicarbonate solution (1 x 10 mL), and the aqueous layer was back extracted with dichloromethane (15 mL). The combined organic layers were washed with water (10 mL), dried over sodium sulfate, filtered and evaporated. The crude compound was purified by column chromatography using dichloromethane: methanol (100:3) to yield the title compound as a beige solid (70 mg, 0.20 mmol, 67%). ESMS m/z 347 (M+H)+.

[00440] Step 3: Synthesis of 8-(5-cyclopropyl-thiazol-4-ylmethyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one . 8-(5-Cyclopropyl-thiazol-4- ylmethyl)-2-methanesulfinyl-8H-pyrido[2,3-d]pyrimidin-7-one (70 mg, 0.20 mmol) and 4-(4- methylpiperazino)aniline (38 mg, 0.20 mmol) were stirred at 140 °C for 6 h. The reaction mixture was purified by column chromatography eluting with dichloromethane: methanol (95 :5) and the solid product was suspended in diisopropyl ether and collected by filtration to give the title compound (10 mg, 0.02 mmol, 10%) as a light yellow solid. ESMS m/z 474 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.50 (s, 1H) 8.39 (s, 1H) 7.53 (d, J = 9.3Hz, 1H) 7.37 (d, J = 8.8Hz, 2H) 7.14 (s, 1H) 6.86 (d, J = 8.8Hz, 2H) 6.47 (d, J = 9.3Hz, 1H) 5.71 (s, 2H) 3.12 - 3.22 (m, 4H) 2.54 - 2.63 (m, 4H) 2.36 (s, 3H) 2.11 - 2.22 (m, 1H) 0.99 - 1.07 (m, 2H) 0.68 - 0.76 (m, 2H).

[00441] Examples 4-5: Synthesis of 8-((4-cyclopropyl-1-methyl-1H-imidazol-5- yl)methyl)-2-(4-(4-methylpiperazin-1-yl)phenylamino)pyrido[2 ,3-d]pyrimidin-7(8H)-one (Compd 10) and 8-((l-cyclopentyl-1H-imidazol-5-yl)methyl)-2-(4-(4-methylpip erazin-1- yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (Compd 11).

[00442] The following compounds were made by the method of Example 3 using the appropriate aryl methyl halide at Step 1 and aniline at Step 3. If necessary, the aryl methyl halide was synthesized by the method used for Intermediate 3. Examples containing secondary amines on the aniline were synthesized using the appropriate Boc protected amino aniline and in the final step were treated with a solution of hydrogen chloride in an organic solvent to produce the example compound, usually isolated as the hydrochloride salt.

Example 6: Synthesis of 6-(2-chloro-4-[1,3,4]oxadiazol-2-yl-phenyl)-8-ethyl-2-[4-(4- methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one (Compd 17).

[00443] Preparation of Intermediate compounds:

[00444] Intermediate 1 : Synthesis of 6-bromo-8-ethyl-2-(methylthio)pyrido[2,3- d]pyrimidin-7(8H)-one.

[00445] Step 1 : Synthesis of 6-bromo-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one.

[00446] To a solution of 2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (1.00 g, 5.18 mmol) in anhydrous dimethylformamide (25 mL) was added N-bromosuccinimide (0.99 g, 5.59 mmol) portionwise at room temperature, and the reaction mixture was stirred for 18 h. The mixture was concentrated, and the solid was triturated with hot water (1 x 20 mL), filtered, and washed with isopropanol to give title compound as a pale yellow solid (0.68 g, 2.50 mmol, 48%). ESMS m/z 272 (M+H)+; 1H NMR (400 MHz, DMSO-d6) δ ppm 12.88 (br. s., 1H), 8.84 (s, 1H), 8.47 (s, 1H), 2.57 (s, 3H).

[00447] Step 2: Synthesis of 6-bromo-8-ethyl-2-(methylthio)pyrido[2,3-d]pyrimidin-

7(8H)-one.

[00448] To a suspension of NaH (60%, 0.15 g, 3.75 mmol) in anhydrous

dimethylformamide (10 mL) was added 6-bromo-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)- one (0.68 g, 2.50 mmol) at room temperature and the reaction was stirred at 50 °C for 0.5 h. The reaction mixture was cooled to room temperature, ethyl bromide (0.22 mL, 0.32 g, 2.93 mmol) was added, and the reaction was stirred at 50 °C for 1.5 h. The mixture was poured onto ice water (10 g), and the white precipitate was collected to give 6-bromo-8-ethyl-2- (methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (0.57 g, 1.90 mmol, 76%). ESMS m/z 300 (M+H) + . The material was used without any further purification.

[00449] Synthesis of 6-(2-chloro-4-[1.3.41oxadiazol-2-yl-phenylV8-ethyl-2-[4-(4-m ethyl- piperazin-1-yl -phenylamino]-8H-pyrido[2.3-d]pyrimidin-7-one.

1 7

[00450] Step 3: Synthesis of 6-bromo-8-ethyl-2-methanesulfmyl-8H-pyrido[2,3- d]pyrimidin-7-one.

[00451] To a solution of 6-bromo-8-ethyl-2-methylsulfanyl-8H-pyrido[2,3-d]pyrimidin-7 - one (0.96 g, 3.19 mmol) in dichloromethane (40 mL) was added 3-chloroperbenzoic acid (77 %, 0.68 g, 3.04 mmol) in dichloromethane (10 mL) at 0-5 °C and the mixture was stirred at 0-5 °C for lh. The reaction mixture was washed with 10% sodium bicarbonate solution (1 x 20 mL) and water (1 x 20 mL). The organic layer was dried over sodium sulfate, filtered and evaporated. The title compound was obtained as a pale yellowish solid (0.98 g, 3.10 mmol, 97 %>). ESMS m/z 316 (M+H)+. [00452] Step 4: Synthesis of 6-bromo-8-ethyl-2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one.

[00453] 6-Bromo-8-ethyl-2-methanesulfinyl-8H-pyrido[2,3-d]pyrimidin- 7-one (600 mg,

1.90 mmol) and 4-(4-methylpiperazino)aniline (363 mg, 1.90 mmol) were stirred at 120 °C for 3 h. The reaction mixture was purified by column chromatography using

dichloromethane: methanol (100:3→100:5) to give the title compound (340 mg, 0.77 mmol, 40 %) as a yellow solid. ESMS m/z 443 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.47 (s, 1H) 7.92 (s, 1H) 7.51 (d, J = 8.8Hz, 2H) 7.24 (br. s., 1H) 6.96 (d, J = 8.8Hz, 2H) 4.48 (q, J = 7.0Hz, 2H) 3.13 - 3.29 (m, 4H) 2.53 - 2.64 (m, 4H) 2.36 (s, 3H) 1.35 (t, J = 7.0 Hz, 3H).

[00454] Step 5: Synthesis of 3-chloro-4-{8-ethyl-2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-7-oxo-7,8-dihydropyrido[2,3-d]pyrimidin-6-yl}be nzoic acid methyl ester.

[00455] 6-Bromo-8-ethyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]- 8H-pyrido[2,3- d]pyrimidin-7-one (110 mg, 0.25 mmol), 2-chloro-4-(methoxycarbonyl)benzene boronic acid (58 mg, 0.27 mmol), K3P04 (58 mg, 0.27 mmol) and PdC12dppf (20 mg, 0.02 mmol) were mixed under argon in a degassed mixture of dimethylformamide and water (20: 1 , 4.5 mL). The resulting suspension was irradiated for 30 min at 140 °C in a microwave reactor. The reaction mixture was evaporated and the residue was purified by column chromatography, eluting with dichloromethane: methanol (95:5). The title compound (78 mg, 0.15 mmol, 60%) was obtained as a yellow solid. ESMS m/z 533 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.55 (s, 1H) 8.15 (d, J = 1.5Hz, 1H) 7.97 (dd, J = 7.9, 1.5Hz, 1H) 7.51 - 7.64 (m, 3H) 7.48 (d, J = 7.8Hz, 1H) 7.27 (br. s., 1H) 6.97 (d, J = 9.0Hz, 2H) 4.49 (q, J = 7.3Hz, 2H) 3.95 (s, 3H) 3.18 - 3.35 (m, 4H) 2.67 (br. s., 4H) 2.42 (br. s., 3H) 1.38 (t, J = 7.3Hz, 3H).

[00456] Step 6: Synthesis of 3-chloro-4-{8-ethyl-2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-7-oxo-7,8-dihydro-pyrido[2,3-d]pyrimidin-6-yl}- benzoic acid hydrazide.

[00457] 3-Chloro-4-{8-ethyl-2-[4-(4-methyl-piperazin-1-yl)-phenylami no]-7-oxo-7,8- dihydro-pyrido [2, 3-d]pyrimidin-6-yl} benzoic acid methyl ester (77 mg, 0.14 mmol) in the mixture of ethanol (4 mL) and hydrazine hydrate (1 mL) was heated at reflux for 2 h. After completion, the reaction mixture was cooled and the yellow precipitate was collected and washed with 2-propanol and diethyl ether to afford the title compound (40 mg, 0.08 mmol, 57 %) as a yellow solid. ESMS m z 533 (M+H)+; 1H NMR (400 MHz, DMSO-d6) δ ppm 9.94 (br. s., 2H) 8.77 (s, 1H) 7.95 (d, J = 1.5Hz, 1H) 7.87 (s, 1H) 7.83 (dd, J = 7.8, 1.5Hz, 1H) 7.66 (d, J = 9.0Hz, 2H) 7.51 (d, J = 7.8Hz, 1H) 6.94 (d, J = 9.0Hz, 2H) 4.56 (br. s., 2H) 4.36 (q, J = 7.0Hz, 2H) 3.05 - 3.15 (m, 4H) 2.42 - 2.48 (m, 4H) 2.22 (s, 3H) 1.28 (t, J = 7.0Hz, 3H). [00458] Step 7: Synthesis of 6-(2-chloro-4-[1,3 ,4]oxadiazol-2-yl-phenyl)-8-ethyl-2-[4-(4- methyl-piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidi n-7-one.

[00459] 3-Chloro-4-{8-ethyl-2-[4-(4-methyl-piperazin-1-yl)-phenylami no]-7-oxo-7,8- dihydro-pyrido [2, 3-d]pyrimidin-6-yl} benzoic acid hydrazide (30 mg, 0.06 mmol) was suspended in triethyl orthoformate (5 mL) to which was added trifluoro acetic acid (1 mL). The resulting reaction mixture was heated at 130 °C for 2 h. The volatiles were removed and the residue was taken up in dichloromethane (1 x 20 mL) and washed with 10% sodium hydroxide solution (2 x 10 mL). The organic layer was dried over sodium sulfate, filtered and evaporated. The residue was purified by column chromatography using dichloromethane:methanol (95:5). The title compound (18 mg, 0.03 mmol, 50 %) was obtained as a yellow solid. ESMS m/z 543 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.57 (s, 1H) 8.50 (s, 1H) 8.21 (d, J = 1.5Hz, 1H) 8.04 (dd, J = 8.0, 1.5Hz, 1H) 7.62 (s, 1H) 7.52 - 7.60 (m, 3H) 7.29 (br. s., 1H) 6.98 (d, J = 9.0Hz, 2H) 4.50 (q, J= 6.8Hz, 2H) 3.16 - 3.27 (m, 4H) 2.56 - 2.65 (m, 4H) 2.37 (s, 3H) 1.39 (d, J= 6.8Hz, 3H).

Example 7: Synthesis of 6-[2-chloro-4-(thiophen-2-yl)phenyl]-8-ethyl-2-(4-(4- methylpiperazin-1-yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H )-one (Compd 22).

[00460] Preparation of Intermediate Compounds:

[00461] Intermediate 2: Synthesis of ethyl 4-bromo-2-chlorophenylacetate.

[00462] Step 1 : Synthesis of (4-Bromo-2-chlorophenyl)methanol.

[00463] 4-Bromo-2-chlorobenzoic acid (92.0 g, 0.391 mol) was dissolved in dry tetrahydrofuran (920 mL) and cooled to -15 °C. Isobutyryl chloro formate (51.0 mL, 0.390 mol) was added followed by N-methylmorpholine (43.5 mL, 0.391 mol). The resulting mixture was stirred for 10 minutes at -15 °C, cooled to -25 °C and the precipitated N-methylmorpholine hydrochloride salt was filtered off. The filtrate was warmed to -5 °C and a solution of sodium borohydride (22.19 g, 0.586 mol) in water (190 mL) was added dropwise to the mixture keeping the temperature below 0 °C. After stirring for 1 h at 0 °C, the volatiles were evaporated, and the residue was diluted with water (500 mL) and dichloromethane (450 mL). The layers were separated and the aqueous layer was extracted with dichloromethane (150 mL). The combined organic layers were washed with water (150 mL), dried over sodium sulfate and evaporated. The product (86.08 g, 0.389 mol, 99%) was obtained as a white crystalline solid.

[00464] Step 2: Synthesis of 4-Bromo-1-bromomethyl-2-chlorobenzene.

[00465] Phosphorus tribromide (40.5 mL, 0.431 mol) was added dropwise to a solution of

(4-bromo-2-chloro-phenyl)-methanol (86.08 g, 0.386 mol) in dichloro ethane (430 mL) at 0 °C. The reaction mixture was stirred for 10 minutes at this temperature then for 0.5 h at 10 °C. The mixture was cooled to 0 °C and a sodium hydroxide solution (600 mL, 2N) was added dropwise. The two layers were separated and the aqueous layer was extracted with dichloro ethane (200 mL). The combined organic layers were washed with water (200 mL), dried over sodium sulfate and evaporated in vacuo. The crude product (90.92 g) was distilled under reduced pressure (7 mmHg), to give 4-bromo-1-bromomethyl-2-chlorobenzene (62.51 g, 0.220 mol, 57 %) as a colorless oil.

[00466] Step 3: Synthesis of (4-Bromo-2-chlorophenyl)acetonitrile.

[00467] To a stirred solution of 4-bromo-1-bromomethyl-2-chlorobenzene (62.51 g, 0.220 mol) in dichloro ethane (522 mL) and water (480 mL) was added tetrabutylammonium chloride (5.05 g), followed by a solution of potassium cyanide (43.22 g, 75.8 mmol) in water (523 mL). and the solution was stirred for 4 h at room temperature. The layers were separated and the aqueous layer was extracted with dichloro ethane (100 mL). The combined organic layers were washed with water (100 mL), dried over sodium sulfate filtered and evaporated. The crude product (52.34 g) was distilled under reduced pressure (1 mmHg), affording (4-bromo-2-chloro- phenyl)-acetonitrile (45.49 g, 0.220 mol, 90 %).

[00468] Step 4: Synthesis of (4-bromo-2-chloro-phenyl)acetic acid.

[00469] (4-Bromo-2-chlorophenyl)acetonitrile (45.49 g, 0.220 mol) was added to 675 mL sodium hydroxide solution (8.2 %) and heated at reflux for 4 h. The homogeneous solution was cooled to room temperature and concentrated hydrochloric acid (117 mL) was added. The mixture was extracted with dichloromethane (500, 200 mL). The combined organic layers were washed with water (100 mL), dried over sodium sulfate and filtered. The filtrate was treated with charcoal (4.5 g), filtered and evaporated. The residue was triturated with hexane (200 mL) and the solid was collected to give (4-Bromo-2-chloro-phenyl)-acetic acid (44.59 g, 0.179 mol, 81 %) as a white crystalline solid. ESMS m/z 497 [2M-1]-.

[00470] Step 5: Synthesis of (4-bromo-2-chlorophenyl)acetic acid ethyl ester.

[00471] (4-Bromo-2-chloro-phenyl)-acetic acid (44.03 g, 0.177 mol) was dissolved in methanol (440 mL) and thionyl chloride (44.0 mL, 0.606 mol) was added dropwise. The mixture was refluxed for 1 h, and evaporated. The residue was taken up in toluene and evaporated (2 x 100 mL). The crude oily product was dissolved in dichloromethane (300 mL) and washed with water (2 x 100 mL), dried over sodium sulfate, filtered and evaporated. The residue was dried in high vacuum (0.2 mmHg) at room temperature, solidifying to a light yellow low melting crystalline solid (45.50 g, 0.164 mol, 93 %) ESMS m/z 294 [M+H+NH 3 ] + ;1H NMR (300 MHz, CDC1 3 ): δ 7.55 (1H,d, J= 1.8 Hz), 7.37 (1H, dd, J= 8.2, 1.8 Hz), 7.16 (1H, d, J= 8.2 Hz), 4.18

[00472] Synthesis of 6-[2-chloro-4-(thiophen-2-vnphenyll-8-ethyl-2-(4-(4- methylpiperazin-1-yl phenylamino pyrido[2.3-d]pyrimidin-7(8H -one. [00473] Step 1 : Synthesis of 6-(4-bromo-2-chlorophenyl)-8-ethyl-2-

(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one.

[00474] To a solution of 4-ethylamino-2-(methylthio)pyrimidine-5-carbaldehyde (1.00 g,

5.07 mmol) in anhydrous dimethylacetamide (10 mL) was added ethyl 4-bromo-2- chlorophenylacetate (1.70 g, 6.12 mmol) and cesium carbonate (3.30 g, 10.13 mmol). The reaction mixture was stirred at 100 °C for 2 h. The mixture was poured into ice-water and the orange solid was collected, washed with water, dried and purified by Teledyne-Isco using a hexane: ethyl acetate gradient (l :0→4: l) to afford the title compound as a white solid (0.30 g, 0.73 mmol, 14%). ESMS m/z 410 (M+H) + ; 1H NMR (400 MHz, CDCh) δ ppm 8.65 (s, 1H), 7.66 (d, J= 1.5Hz, 1H), 7.63 (s, 1H), 7.47 (dd, J= 8.3, 1.5Hz, 1H), 7.26 (d, J= 8.3Hz, 1H), 4.55 (q, J= 7.0Hz, 2H), 2.66 (s, 3H), 1.37 (t, J= 7.0Hz, 3H).

[00475] Step 2: Synthesis of 6-(4-bromo-2-chlorophenyl)-8-ethyl-2-

(methylsulfinyl)pyrido[2,3-d]pyrimidin-7(8H)-one.

[00476] To a solution of 6-(4-bromo-2-chlorophenyl)-8-ethyl-2-(methylthio)-pyrido[2,3 - d]pyrimidin-7(8H)-one (1.30 g, 3.16 mmol) in dichloromethane (20 mL) was added dropwise a solution of 3-chloroperbenzoic acid (77 %, 0.57 g, 2.54 mmol) in dichloromethane (5 mL) at 0- 5 °C and the mixture was stirred for 5 h. The reaction mixture was washed with saturated sodium bicarbonate solution (2 x 20 mL) and water (10 mL), and the organic layer was dried over sodium sulfate, filtered and evaporated. The crude product was purified by silica gel column chromatography using dichloromethane: ethyl acetate (5: 1→5:2→2: 1→1 : 1) to give the title compound as an off-white solid (0.96 g, 2.25 mmol, 71%). ESMS m/z 426 (M+H)+;

[00477] Step 3: Synthesis of 6-(4-bromo-2-chlorophenyl)-8-ethyl-2-(4-(4- methylpiperazin-1-yl)-phenylamino)pyrido[2,3-d]pyrimidin-7(8 H)-one

[00478] 6-(4-Bromo-2-chlorophenyl)-8-ethyl-2-(methylsulfinyl)pyrido[ 2,3-d]pyrimidin- 7(8H)-one (0.60 g, 1.41 mmol) and 4-(4-methylpiperazino)aniline (0.27 g, 1.41 mmol) were stirred at 150 °C for 4 h. The cooled reaction mixture was taken up in dichloromethane (50 mL) and washed with 10% NaOH (1 x 25 mL) then with water (1 x 20 mL). The organic layer was dried over sodium sulfate, filtered and evaporated. The residue was purified by column chromatography using chloroform: methanol (100:3) as eluent to give the title compound as a yellow solid (0.32 g, 0.58 mmol, 41%). ESMS m z 553 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.53 (s, 1H), 7.65 (d, J = 1.8Hz, 1H), 7.52 - 7.59 (m, 3H), 7.45 (dd, J = 8.2, 1.9Hz, 1H), 7.28 (d, J = 8.2Hz, 1H), 6.97 (d, J = 8.8Hz, 2H), 4.48 (q, J = 7.0Hz, 2H), 3.17 - 3.26 (m, 4H), 2.55 - 2.70 (m, 4H), 2.37 (s, 3H), 1.37 (t, J = 7.0Hz, 3H). [00479] Step 4: Synthesis of 6-[2-chloro-4-(thiophen-2-yl)phenyl]-8-ethyl-2-(4-(4- methylpiperazin-1-yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H )-one

[00480] 6-(4-Bromo-2-chlorophenyl)-8-ethyl-2-(4-(4-methylpiperazin- 1 - yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (50 mg, 0.09 mmol), thiophene-2-boronic acid (35 mg, 0.27 mmol), K3P04 (57 mg, 0.27 mmol) and PdC12dppf (7 mg, 0.01 mmol) were mixed as solids and placed under argon. Argon was bubbled through a mixture of

dimethylformamide: water (20: 1, 2.0 mL) for 20 min. The solvent was added to the solid and the suspension was heated under microwave irradiation at 140 °C for 30 min. The reaction mixture was evaporated and the crude product was purified by column chromatography, eluting with dichloromethane: methanol (100:3). The product was triturated with refluxing acetonitrile to yield the title compound as a yellow solid (48 mg, 0.09 mmol, 100 %). ESMS m/z 557 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.54 (s, 1H), 7.72 (s, 1H), 7.51 - 7.60 (m, 4H), 7.40 (d, J = 8.0Hz, 1H), 7.30 - 7.36 (m, 2H), 7.27 (s, 1H), 7.08 - 7.13 (m, 1H), 6.97 (d, J = 8.8Hz, 2H), 4.50 (q, J = 7.0Hz, 2H), 3.16 - 3.28 (m, 4H), 2.54 - 2.69 (m, 4H), 1.39 (t, J = 7.0Hz, 3H).

Examples 8-14 (Compds 23-29):

[00481] The following compounds were made by the method of Example 7 using the appropriate arylacetic acid at Step 1, aniline at Step 3 and boronic acid or ester at Step 4.

Examples containing secondary amines on the aniline were synthesized using the appropriate Boc protected amino aniline and in the final step were treated with a solution of hydrogen chloride in an organic solvent to produce the example compound, usually isolated

hydrochloride salt.

LCMS

Compd. MW Rt

Structure Ion

23 522.7 523 1.47

H ^

24 557.1 557 1.47

H ^ Example 15: Synthesis of 8-(2-ethanesulfonyl-benzyl)-2-[4-(4-methyl-piperazin-1-yl)- phenylami -8H-pyrido[2,3-d]pyrimidin-7-one (Compd 32).

32

[00482] Preparation of Intermediate compounds:

[00483] Intermediate 1 : Synthesis of l-chloromethyl-2-ethanesulfonyl-benzene.

[00484] Step 1 : Synthesis of 2-ethylsulfanyl-benzoic acid methyl ester.

[00485] To a solution of 2-mercaptobenzoic acid methyl ester (1.00 g, 5.95 mmol) in anhydrous dimethylformamide (6 mL) was added K 2 CO 3 (1.00 g, 7.24 mmol). The reaction mixture was stirred for 15 min at room temperature, ethyl bromide (662 μΐ, 8.93 mmol) was added and the reaction was stirred for 1 h. The mixture was poured onto ice (25 g) and extracted with diethyl ether (2 x 25 mL). The organic layer was dried over sodium sulfate, filtered and evaporated to afford the title compound (1.11 g, 95%) as a yellow oil.

[00486] Step 2: Synthesis of (2-ethylsulfanyl-phenyl)-methanol.

[00487] To a suspension of lithium aluminum hydride (0.22 g, 5.66 mmol) in anhydrous tetrahydroiuran (22 mL) was added a solution of 2-ethylsulfanyl-benzoic acid methyl ester (2, 1.11 g, 5.65 mmol) in anhydrous tetrahydrofuran (20 mL) at 0 °C and the reaction was stirred for 1 h. Brine (30 mL) was added dropwise to the reaction mixture at 0 °C, and the mixture was allowed to warm to room temperature. The two phases were separated and the aqueous layer was extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over sodium sulfate, filtered, and evaporated. The title compound (0.92 g, 5.47 mmol, 97%) was obtained as a yellow oil. ESMS m/z 151 (M+H-H20)+.

[00488] Step 3: Synthesis of l-chloromethyl-2-ethylsulfanyl-benzene.

[00489] (2-Ethylsulfanyl-phenyl)-methanol (0.92 g, 5.47 mmol) was dissolved in anhydrous dichloromethane (15 mL) and thionyl chloride (400 μί, 5.48 mmol) was added. The reaction mixture was heated at reflux for 4 h and evaporated. The title compound (0.99 g, 5.27 mmol, 96%) was obtained as a yellow oil.

[00490] Step 4: Synthesis of l-chloromethyl-2-ethanesulfonyl-benzene.

[00491] To a solution of l-chloromethyl-2-ethylsulfanyl-benzene (0.99 g, 5.27 mmol) in methanol (25 mL) was added a solution of Oxone (3.73 g, 6.07 mmol) in water (27 mL) at room temperature. The reaction mixture was stirred for 18 h. The reaction was evaporated to dryness and the residue taken up in ethyl acetate (30 mL) and water (15 mL). The two phases were separated and the aqueous layer was extracted with ethyl acetate (2 x 15 mL). The combined organic layers were dried over sodium sulfate, filtered and evaporated to give the title compound (1.09 g, 4.98 mmol, 94%) as a yellow oil. ESMS m/z 236 (M+H+NH3)+; 1H NMR (400 MHz, CDC13) δ ppm 8.04 (d, J = 7.8Hz, 1H) 7.61 - 7.71 (m, 2H) 7.54 (td, J = 7.8, 1.8Hz, 1H) 5.14 (s, 2H) 3.34 (q, J = 7.4Hz, 2H) 1.30 (t, J = 7.4Hz, 3H).

[00492] Intermediates 2 & 3: Synthesis of l-Chloromethyl-2-cyclopentanesulfonyl- benzene & l-Chloromethyl-2-cyclohexanesulfonyl-benzene.

[00493] The following intermediates were made by the method of Intermediate 1 using the appropriate alkyl halide at Step 1 : l-Chloromethyl-2-cyclopentanesulfonyl-benzene & 1- Chloromethyl-2-cyclohexanesulfonyl-benzene.

[00494] Synthesis of 8-(2-ethanesulfonyl-benzyl)-2-[4-(4-methyl-piperazin- 1 -yl)- phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one.

[00495] Step 1 : Synthesis of 8-(2-ethanesulfonyl-benzyl)-2-methylsulfanyl-8H- pyrido [2 , 3 -d] -pyrimidin-7-one .

[00496] To a suspension of sodium hydride (60% in mineral oil, 204 mg, 5.1 mmol) in anhydrous dimethylformamide (12 mL) was added a solution of 2-methylsulfanyl-8H- pyrido[2,3-d]pyrimidin-7-one (500 mg, 2.59 mmol) in anhydrous dimethylformamide (5 mL) then lithium bromide (590 mg, 6.79 mmol) at 0-5 °C. After 30 min, a solution of 1- chloromethyl-2-ethanesulfonyl-benzene (743 mg, 3.39 mmol) in anhydrous dimethylformamide (7 mL) was added slowly and stirring was continued at room temperature for 18 h. The resulting mixture was poured into ice water (150 g) and extracted with ethyl acetate (5 x 20 mL). The combined organic layers were dried over sodium sulfate, filtered and evaporated. The residue was purified by ISCO using n-hexane: ethyl acetate (1 :0→1 : 1) as eluent. The title compound (188 mg, 0.50 mmol, 19%) was obtained as a light yellow solid. ESMS m/z 376 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.69 (s, 1H) 8.02 - 8.08 (m, 1H) 7.72 (d, J = 9.5Hz, 1H) 7.39 - 7.48 (m, 2H) 6.79 - 6.88 (m, 1H) 6.69 (d, J = 9.5Hz, 1H) 6.03 (s, 2H) 3.57 (q, J = 7.4Hz, 2H) 2.47 (s, 3H) 1.40 (t, J = 7.4Hz, 3H).

[00497] Step 2: Synthesis of 8-(2-ethanesulfonyl-benzyl)-2-methanesulfinyl-8H- pyrido [2 , 3 -d] -pyrimidin-7-one .

[00498] To a solution of 8-(2-ethanesulfonyl-benzyl)-2-methylsulfanyl-8H-pyrido[2,3- d]pyrimidin-7-one (183 mg, 0.49 mmol) in dichloromethane (5 mL) was added 3- chloroperbenzoic acid (77%, 101 mg, 0.46 mmol) in dichloromethane (5 mL) at 0-5 °C and the mixture was stirred for 1 h. The reaction mixture was washed with saturated sodium

bicarbonate solution (1 x 10 mL) then with water (1 x 10 mL). The organic layer was dried over sodium sulfate, filtered and evaporated to yield the title compound as an off-white solid (191 mg, 0.49 mmol, 100%). ESMS m/z 392 (M+H)+.

[00499] Step 3: Synthesis of 8-(2-ethanesulfonyl-benzyl)-2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one.

[00500] 8-(2-Ethanesulfonyl-benzyl)-2-methanesulfinyl-8H-pyrido[2,3- d]pyrimidin-7-one (181 mg, 0.46 mmol) and 4-(4-methylpiperazino)aniline (80 mg, 0.90 mmol) were stirred at 120 °C for 4 h. The reaction mixture was purified by column chromatography using

dichloromethane: methanol (95:5) as eluent and the solid product was suspended in diisopropyl ether, filtered, and collected to give the title compound (50 mg, 0.10 mmol, 22%) as a light yellow solid. ESMS m/z 519 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.58 (s, 1H) 8.05 (dd, J = 7.4, 1.9Hz, 1H) 7.62 (d, J = 9.3Hz, 1H) 7.39 - 7.47 (m, 2H) 7.31 (d, J= 8.5Hz, 2H) 7.20 (s, 1H) 6.90 (d, J= 8.5Hz, 1H) 6.82 (d, J= 8.5Hz, 2H) 6.46 (d, J= 9.3Hz, 1H) 5.98 (s, 2H) 3.48 (q, J= 7.4Hz, 2H) 3.11 - 3.21 (m, 4H) 2.51 - 2.62 (m, 4H) 2.34 (s, 3H) 1.37 (t, J= 7.4Hz, 3H). Examples 16 - 17: Synthesis of 8-(2-(cyclopentylsulfonyl)benzyl)-2-(4-(4-methylpiperazin- l-yl)phenylamino)pyrido[2,3-d]pyrimidin-7(8H)-one (Compd 33) and 8-(2-

(cyclohexylsulfonyl)benzyl)-2-(4-(4-methylpiperazin-1-yl) phenylamino)pyrido[2,3- d]pyrimidin-7(8H)-one (Compd 34).

[00501] The following compounds were made by the method of Example 15 using the appropriate aryl methyl halide at Step 1 and aniline at Step 3. The aryl methyl halides were synthesized by the method used for Intermediate 1. Examples containing secondary amines on the aniline were synthesized using the appropriate Boc protected aminoaniline and in the final step were treated with a solution of hydrogen chloride in an organic solvent to produce the example compound, usually isolated as the hydrochloride salt.

LCMS

Compd. MW Rt

Structure Ion

33 558.7 559 1.18 ο ο

34 572.7 573 1.24 ο ο

Example 18: Synthesis of 8-(2-Ethanesulfonylbenzyl)-2-[4-(4-methyl-piperazin-1-yl)- phenylamino] -6-phenyl-8H-pyrido [2,3-d] pyrimidin-7-one (Compd 40).

37 38

[00502] Step 1 : Synthesis of 6-bromo-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one.

[00503] To a solution of 2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (1.00 g, 5.2 mmol) in anhydrous dimethylformamide (25 mL) was added N-bromosuccinimide (0.99 g, 5.6 mmol) in small portions at room temperature, and the reaction mixture was stirred for 18 h. The mixture was concentrated, and the solid was triturated with hot water (1 x 20 mL), filtered, and washed with isopropanol to give title compound as a pale yellow solid (0.68 g, 2.5 mmol, 48%). ESMS m/z 272 (M+H)+; 1H NMR (400 MHz, DMSO-d6) δ ppm 12.88 (br. s., 1H), 8.84 (s, 1H), 8.47 (s, 1H), 2.57 (s, 3H).

[00504] Step 2: Synthesis of 6-Bromo-8-(2-ethanesulfonyl-benzyl)-2-methylsulfanyl-8H- pyrido[2,3-] pyrimidin-7-one.

[00505] To a suspension of sodium hydride (60% on mineral oil, 1.17 g, 29.2 mmol) in anhydrous dimethylformamide (40 mL) was added a solution of 6-bromo-2- (methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (3.97 g, 14.6 mmol) in anhydrous

dimethylformamide (20 mL) then lithium bromide (3.29 g, 37.9 mmol) at 0-5 °C. The reaction mixture was stirred for 30 min at 0-5 °C and a solution of l-chloromethyl-2-ethanesulfonyl- benzene (4.47 g, 20.4 mmol) in anhydrous dimethylformamide (20 mL) was added slowly. The reaction was allowed to warm to room temperature and stirred for 18 h. The mixture was poured onto ice water (250 g), extracted with ethyl acetate (4 x 80 mL) and the combined organic layers were dried over sodium sulfate, filtered and evaporated. The residue was purified by ISCO using a gradient of n-hexane: ethyl acetate (1 :0→0: 1). The title compound (1.06 mg, 2.29 mmol, 16 %) was obtained as an off white powder. ESMS m z 454 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.71 (s, 1H) 8.17 (s, 1H) 8.03 - 8.10 (m, 1H) 7.43 - 7.48 (m, 2H) 6.74 - 6.81 (m, 1H) 6.10 (s, 2H) 3.56 (q, J = 7.4Hz, 2H) 2.49 (s, 3H) 1.42 (t, J = 7.4Hz, 3H).

[00506] Step 3: Synthesis of 6-Bromo-8-(2-ethanesulfonylbenzyl)-2-methanesulfinyl-8H- pyrido[2,3-] pyrimidin-7-one

[00507] To a solution of 6-Bromo-8-(2-ethanesulfonyl-benzyl)-2-methylsulfanyl-8H- pyrido[2,3-] pyrimidin-7-one (1.04 g, 2.3 mmol) in dichloromethane (30 mL) was added 3- chloroperbenzoic acid (77 %, 462 mg, 2.06 mmol) in dichloromethane (10 mL) at 0-5 °C and the mixture was stirred at 0-5 °C for 1 h. The reaction mixture was washed with saturated sodium bicarbonate solution (1 x 40 mL) and water (1 x 40 mL), and the organic layer was dried over sodium sulfate, filtered, evaporated, and recrystallized from ethyl acetate to yield the title compound as a white solid (790 mg, 1.63 mmol, 73 %). ESMS m/z 470 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 9.07 (s, 1H) 8.35 (s, 1H) 8.04 - 8.11 (m, 1H) 7.41 - 7.51 (m, 2H) 6.69 - 6.76 (m, 1H) 6.17 (d, J = 16.8Hz, 1H) 6.12 (d, J = 16.8Hz, 1H) 3.54 (q, J = 7.3Hz, 2H) 2.88 (s, 3H) 1.42 (t, J = 7.3Hz, 3H).

[00508] Step 4: Synthesis of 6-Bromo-8-(2-ethanesulfonylbenzyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one [00509] 6-Bromo-8-(2-ethanesulfonylbenzyl)-2-methanesulfinyl-8H-pyri do[2,3-] pyrimidin-7-one (760 mg, 1.6 mmol) and 4-(4-methylpiperazino)aniline (309 mg, 1.6 mmol) were stirred at 120 °C for 4 h. The reaction mixture was triturated in boiling ethyl acetate, and the solid was collected and washed with diethyl ether to give the title compound (720 mg, 1.21 mmol, 74 %) as a yellow solid. ESMS m/z 597 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.59 (s, 1H) 8.04 - 8.09 (m, 2H) 7.41 - 7.48 (m, 2H) 7.31 (br. s., 3H) 6.85 - 6.90 (m, 3H) 6.04 (s, 2H) 3.48 (q, J = 7.4Hz, 2H) 3.14 - 3.20 (m, 4H) 2.54 - 2.60 (m, 4H) 2.35 (s, 3H) 1.40 (t, J = 7.4Hz, 3H).

[00510] Step 5: Synthesis of 8-(2-Ethanesulfonylbenzyl)-2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-6-phenyl-8H-pyrido[2,3-d]pyrimidin-7-one.

[00511] 6-Bromo-8-(2-ethanesulfonylbenzyl)-2-[4-(4-methyl-piperazin- 1 -yl)- phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one (100 mg, 0.17 mmol), phenylboronic acid (22 mg, 0.18 mmol), K3P04 (38 mg, 0.18 mmol) and PdC12(dppf) (15 mg, 0.02 mmol) were mixed under argon in a degassed mixture of dimethylformamide and water (20: 1, 4 mL). The resulting suspension was irradiated for 30 min at 140 °C in a microwave reactor. The reaction mixture was evaporated and the residue was purified by preparative HPLC, and the crude product (50 mg) was recrystallized from ethyl acetate. The title compound (15 mg, 0.025 mmol, 15 %) was obtained as a light yellow solid. ESMS m z 519 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.66 (s, 1H) 8.07 (dd, J = 7.5, 1.8Hz, 1H) 7.77 (s, 1H) 7.66 (d, J = 7.0Hz, 2H) 7.29 - 7.52 (m, 7H) 7.26 (d, J = 8.2Hz, 1H) 6.94 (d, J = 7.0Hz, 1H) 6.84 (d, J = 8.8Hz, 2H) 6.07 (s, 2H) 3.47 (q, J = 7.5Hz, 2H) 3.23 (br. s., 4H) 2.66 (br. s., 4H) 2.42 (br. s., 3H) 1.38 (t, J = 7.5Hz, 3H).

Example 19: Synthesis of 6-(2-chloro-4-[1,3,4]oxadiazol-2-yl-phenyl)-8-ethyl-2-[4-(4- meth l-piperazin- l-yl)-phenylamino] -8H-pyrido [2,3-d] pyrimidin-7-one (Compd 19) .

14 41

[00512] Preparation of Intermediate compounds: [00513] Intermediate 1 : Synthesis 6-bromo-8-ethyl-2-(methylthio)pyrido[2, 3- d]pyrimidin-7(8H)-one.

[00514] Step 1 : Synthesis of 6-bromo-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one. To a solution of 2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (1.00 g, 5.18 mmol) in anhydrous dimethylformamide (25 mL) was added N-bromosuccinimide (0.99 g, 5.59 mmol) portionwise at room temperature, and the reaction mixture was stirred for 18 h. The mixture was concentrated, and the solid was triturated with hot water (1 x 20 mL), filtered, and washed with isopropanol to give title compound as a pale yellow solid (0.68 g, 2.50 mmol, 48%). ESMS m/z 272 (M+H)+; 1H NMR (400 MHz, DMSO-d6) δ ppm 12.88 (br. s., 1H), 8.84 (s, 1H), 8.47 (s, 1H), 2.57 (s, 3H).

[00515] Step 2: Synthesis of 6-bromo-8-ethyl-2-(methylthio)pyrido[2,3-d]pyrimidin-

7(8H)-one. To a suspension of NaH (60%>, 0.15 g, 3.75 mmol) in anhydrous dimethylformamide (10 mL) was added 6-bromo-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (0.68 g, 2.50 mmol) at room temperature and the reaction was stirred at 50 °C for 0.5 h. The reaction mixture was cooled to room temperature, ethyl bromide (0.22 mL, 0.32 g, 2.93 mmol) was added, and the reaction was stirred at 50 °C for 1.5 h. The mixture was poured onto ice water (10 g), and the white precipitate was collected to give 6-bromo-8-ethyl-2-(methylthio)pyrido[2,3-d]pyrimidin- 7(8H)-one (0.57 g, 1.90 mmol, 76%). ESMS m z 300 (M+H)+. The material was used without any further purification.

[00516] Synthesis of 6-(2-chloro-4-[1.3.41oxadiazol-2-yl-phenylV8-ethyl-2-[4-(4-m ethyl- piperazin-1-yl -phenylamino]-8H-pyrido[2.3-d]pyrimidin-7-one.

14 41

[00517] Step 1 : Synthesis of 6-bromo-8-ethyl-2-methanesulfinyl-8H-pyrido[2, 3- d]pyrimidin-7-one. To a solution of 6-bromo-8-ethyl-2-methylsulfanyl-8H-pyrido[2,3- d]pyrimidin-7-one (0.96 g, 3.19 mmol) in dichloromethane (40 mL) was added 3- chloroperbenzoic acid (77 %, 0.68 g, 3.04 mmol) in dichloromethane (10 mL) at 0-5 °C and the mixture was stirred at 0-5 °C for lh. The reaction mixture was washed with 10% sodium bicarbonate solution (1 x 20 mL) and water (1 x 20 mL). The organic layer was dried over sodium sulfate, filtered and evaporated. The title compound was obtained as a pale yellow solid (0.98 g, 3.10 mmol, 97 %). ESMS m/z 316 (M+H)+.

[00518] Step 2: Synthesis of 6-bromo-8-ethyl-2-[4-(4-methyl-piperazin-1-yl)- phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one. 6-Bromo-8-ethyl-2-methanesulfinyl-8H- pyrido[2,3-d]pyrimidin-7-one (600 mg, 1.90 mmol) and 4-(4-methylpiperazino)aniline (363 mg, 1.90 mmol) were stirred at 120 °C for 3 h. The reaction mixture was purified by column chromatography using dichloromethane: methanol (100:3→100:5) to give the title compound (340 mg, 0.77 mmol, 40 %) as a yellow solid. ESMS m/z 443 (M+H)+; 1H NMR (400 MHz, CDC13) δ ppm 8.47 (s, 1H) 7.92 (s, 1H) 7.51 (d, J = 8.8Hz, 2H) 7.24 (br. s., 1H) 6.96 (d, J = 8.8Hz, 2H) 4.48 (q, J = 7.0Hz, 2H) 3.13 - 3.29 (m, 4H) 2.53 - 2.64 (m, 4H) 2.36 (s, 3H) 1.35 (t, J = 7.0 Hz, 3H).

[00519] Step 3 : Synthesis of 8-ethyl-6-(4-methanesulfonyl-phenyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one . 6-Bromo-8-ethyl-2-[4-(4- methyl-piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidi n-7-one (120 mg, 0.27 mmol), 4-methylsulfonylphenylboronic acid (60 mg, 0.30 mmol), K3P04 (64 mg, 0.30 mmol) and PdC12dppf (25 mg, 0.03 mmol) were mixed under argon in a degassed mixture of

dimethylformamide and water (20: 1, 4 mL). The resulting suspension was irradiated for 30 min at 140 °C in a microwave reactor. After completion, the reaction mixture was evaporated and the residue purified by ISCO eluting with chloroform: methanol (100:0→98:2). The partially purified product (50 mg) was recrystallized from ethyl acetate. The title compound (21 mg, 0.04 mmol, 15 %) was obtained as a yellow solid. ESMS m z 519 (M+H)+; 1H NMR (400 MHz,

CDC13) δ ppm 8.60 (s, 1H) 7.99 (d, J = 8.3Hz, 2H) 7.90 (d, J = 8.3Hz, 2H) 7.71 (s, 1H) 7.55 (d, J = 8.8Hz, 2H) 7.30 (br. s, 1H) 6.98 (d, J = 8.8Hz, 2H) 4.51 (q, J = 7.0Hz, 2H) 3.18 - 3.27 (m, 4H) 3.07 (s, 3H) 2.54 - 2.67 (m, 4H) 2.37 (s, 3H) 1.39 (t, J = 7.0Hz, 3H).

Examples 20-24 (Compds 42-46):

[00520] The following compounds were made by the method of Example 19 using the appropriate intermediate at Step 1, aniline at Step 2 and boronic acid or ester at Step 3.

Examples containing secondary amines on the aniline were synthesized using the appropriate Boc protected amino aniline and in the final step were treated with a solution of hydrogen chloride in an organic solvent to produce the example compound, usually isolated as the hydrochloride salt.

Example 25: Synthesis of 6-(2-chloro-4-methanesulfonyl-phenyl)-8-ethyl-2-[4-(4-methyl - piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one . (Compd 49).

[00521] Preparation of Intermediate compounds:

[00522] Intermediate 2: S nthesis l-bromo-2-chloro-4-methylsulfonylbenzene.

[00523] Step 1 : Synthesis of l-bromo-2-chloro-4-methylsulfanylbenzene.

[00524] A solution of l-bromo-2-chloro-4-fluorobenzene (7, 0.50 g, 2.39 mmol) and sodium thiomethoxide (0.17 g, 2.42 mmol) in DMSO (2.5 mL) was stirred at 100 °C for 1 h. The reaction mixture was added to 20 mL water with stirring, the aqueous mixture was extracted with ethyl acetate (2 x 20 mL) and the combined organic layers were dried over sodium sulfate, filtered and evaporated in vacuo. The title compound (0.62 g) was obtained as a colorless oil and was used in the next step without further purification.

[00525] Step 2: Synthesis of l-bromo-2-chloro-4-methylsulfonyl-benzene.

[00526] To a solution of l-bromo-2-chloro-4-methylsulfanylbenzene (0.62 g, ~2.4 mmol) in dichloromethane (20 mL) was added 3-chloroperbenzoic acid (77 %, 1.07 g, 4.80 mmol) in small portions at room temperature and the reaction mixture was stirred for 15 h. The suspension was filtered and the filtrate was diluted with dichloromethane (30 mL) and washed with 10% aqueous sodium bicarbonate (30 mL), then water (20 mL). The organic layer was dried over sodium sulfate, filtered and evaporated. The residue was purified by column chromatography using dichloromethane as eluent. The title compound (0.30 g, 1.11 mmol, 46%> over two steps) was obtained as a white solid. 1H NMR (400 MHz, CDC1 3 ) δ ppm 8.04 (d, J= 2.0Hz, 1H) 7.86 (d, J= 8.3Hz, 1H) 7.69 (dd, J= 8.3, 2.0Hz, 1H) 3.08 (s, 3H).

[00527] Synthesis of 6-(2-chloro-4-methanesulfonyl-phenyl -8-ethyl-2-[4-(4-methyl- piperazin-1-yl -phenylamino]-8H-pyrido[2 -d]pyrimidin-7-one.

[00528] Step 1 : Synthesis of 8-ethyl-2-[4-(4-methyl-piperazin- 1 -yl)-phenylamino]-7-oxo-

7,8-dihydro-pyrido[2,3-d]pyrimidine-6-boronic acid.

[00529] 6-Bromo-8-ethyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]- 8H-pyrido[2,3- d]pyrimidin-7-one (100 mg, 0.22 mmol), bis(pinacolato)diboron (63 mg, 0.25 mmol), potassium acetate (66 mg, 0.68 mmol) and PdCl 2 (PPh 3 ) 2 (16 mg, 0.02 mmol) were mixed under argon in degassed toluene (3 mL). The resulting suspension was irradiated for 30 min at 120 °C in a microwave reactor. After completion, the reaction mixture was evaporated and the residue was purified by column chromatography using dichloromethane:methanol: triethylamine

(4: 1 :0→1 : 1 :0→0:95:5) as eluent. The crude product was dissolved in dichloromethane (10 mL), washed with water (5 mL), and the organic layer was dried over sodium sulfate, filtered and evaporated. The title compound (15 mg, 0.04 mmol, 18 %) was obtained as a yellow solid. ESMS m/z 409 (M+H) + ; 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 10.07 (br. s., 1H) 8.85 (s, 1H) 8.52 (s, 2H) 8.28 (s, 1H) 7.64 (br. s., 2H) 6.94 (d, J= 9.0Hz, 2H) 4.33 (q, J= 6.9Hz, 2H) 3.07 - 3.14 (m, 4H) 2.43 - 2.47 (m, 4H) 2.22 (s, 3H) 1.26 (t, J= 6.9Hz, 3H).

[00530] Synthesis of 6-(2-chloro-4-methanesulfonyl-phenyl -8-ethyl-2-[4-(4-methyl- piperazin-1-yl -phenylamino]-8H-pyrido[2.3-d]pyrimidin-7-one.

[00531] 8-Ethyl-2-[4-(4-methyl-piperazin-1-yl)-phenylamino]-7-oxo-7, 8-dihydro- pyrido[2,3-d]pyrimidine-6-boronic acid (5, 100 mg, 0.24 mmol), l-bromo-2-chloro-4- methylsulfonylbenzene (75 mg, 0.28 mmol), sodium carbonate (80 mg, 0.75 mmol) and Pd(PPh 3 ) 4 (30 mg, 0.026 mmol) were mixed under argon in a degassed mixture of dimethoxyethane: water (10: 1, 2.5 mL). The resulting suspension was irradiated for 30 min at 120 °C in a microwave reactor. After completion, the reaction mixture was evaporated and the residue was purified by column chromatography using dichloromethane: methanol (100:3) as eluent, The crude product was triturated with isopropyl ether (10 mL) and collected. The title compound (49 mg, 0.088 mmol, 37 %) was obtained as a yellow solid. ESMS m/z 553 (M+H) + ; 1H NMR (400 MHz, CDC1 3 ) δ ppm 8.04 (d, J= 2.0Hz, 1H) 7.86 (d, J= 8.3Hz, 1H) 7.69 (dd, J = 8.3, 2.0Hz, 1H) 3.08 (s, 3H).

Example 26: Synthesis of 8-(2-cyclopropyl-6-fluorobenzyl)-2-[4-(4-methyl-piperazin-1- yl)- phenylamino]-6-prop-1-ynyl-8H-pyrido[2,3-d]pyrimidin-7-one (Compd 53)

[00532] Preparation of Intermediate compounds:

[00533] Intermediate 1 : Synthesis of 3-bromo-2-chloromethyl-thiophene.

[00534] Step 1 : Synthesis of methyl 2-bromo-6-fluorobenzoate.

[00535] 2-Bromo-6-fluorobenzoic acid (12.50 g, 57 mmol) was dissolved in a mixture of methanol (60 mL) and cone, sulfuric acid (65 mL). The solution was heated to 80 °C and stirred for 12 h. The reaction mixture was cooled and 20% sodium carbonate solution (500 mL) was added slowly to reach pH = 8. The mixture was extracted with dichloromethane (3 x 180 mL), and the combined organic layers were dried over magnesium sulfate and evaporated. The title compound (11.29 g, 48.5 mmol, 85%) was obtained as a brown oil.

[00536] Step 2: Synthesis of methyl 2-cyclopropyl-6-fluorobenzoate.

[00537] Methyl 2-bromo-6-fluorobenzoate (11.29 g, 48.5 mmol), cyclopropylboronic acid

(6.24 g, 72.7 mmol), K 3 P0 4 (30.85 g, 145.4 mmol) and Pd(PPh 3 ) 4 (2.80 g, 2.4 mmol) were mixed under argon in a degassed mixture of toluene and water (20: 1, 160 mL). The resulting brown suspension was heated at reflux for 2.5 h, cooled, filtered through Celite, and evaporated. The residue was partitioned between ethyl acetate (150 mL) and brine (150 mL). The layers were separated and organic layer was dried over magnesium sulfate and evaporated. The oily residue was suspended in hexane and the solid was removed by filtration. Evaporation of the filtrate afforded the title compound (9.30 g, 47.9 mmol, 99%) as a light brown oil.

[00538] Step 3 : Synthesis of (2-cyclopropyl-6-fluorophenyl)-methanol.

[00539] To a solution of methyl 2-cyclopropyl-6-fluorobenzoate (4.54 g, 23.4 mmol) in anhydrous tetrahydroiuran (30 mL) at room temperature was added lithium aluminium hydride (1.42 g, 37.4 mmol) in small portions and the mixture was stirred overnight. The reaction mixture was cooled to 0 °C and water (3 mL) was added dropwise, the mixture was warmed to room temperature and an additional portion of water (30 mL) was added. The solids were removed by filtration, and the aqueous filtrate was extracted with ethyl acetate (2 x 30 mL). The combined organic layers were dried over magnesium sulfate and evaporated. Purification by column chromatography over silica using n-hexane: ethyl acetate (8: 1) as eluent afforded the title compound (3.28 g, 19.8 mmol, 84%) as a pale yellow oil.

[00540] Step 4: Synthesis of 2-chloromethyl-1-cyclopropyl-3-fluorobenzene.

[00541] (2-Cyclopropyl-6-fluorophenyl)-methanol (5.56 g, 34 mmol) was dissolved in dichloromethane (100 mL) and thionyl chloride (2.49 mL, 34 mmol) was added. The mixture was refluxed for 4 h and evaporated. The crude product was purified by column chromatography over silica, eluting with n-hexane: ethyl acetate (95:5). The title compound (3.41 g, 18.4 mmol, 54%) was isolated as a pale yellow oil.

[00542] Synthesis of 8-(2-cyclopropyl-6-fluorobenzyl -2-[4-(4-methyl-piperazin- 1 -yl)- phenylamino]-6-prop-1-ynyl-8H-pyrido[2.3-d]pyrimidin-7-one.

[00543] Step 1 : Synthesis of 6-bromo-2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one.

[00544] To a solution of 2-(methylthio)pyrido[2,3-d]pyrimidin-7(8H)-one (1.00 g, 5.2 mmol) in anhydrous dimethylformamide (25 mL) was added N-bromosuccinimide (0.99 g, 5.6 mmol) portionwise at room temperature, and the reaction mixture was stirred for 18 h. The mixture was concentrated, and the solid was triturated with hot water (1 x 20 mL), filtered, and washed with isopropanol to give title compound as a pale yellow solid (0.68 g, 2.5 mmol, 48%). ESMS m/z 272 (M+H) + ; 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 12.88 (br. s, 1H), 8.84 (s, 1H), 8.47 (s, 1H), 2.57 (s, 3H).

[00545] Step 2: Synthesis of 6-bromo-8-(2-cyclopropyl-6-fluoro-benzyl)-2- methylsulfanyl-8H-pyrido[2,3-d]pyrimidin-7-one.

[00546] To a solution of 6-bromo-2-methylsulfanyl-8H-pyrido[2,3-d]pyrimidin-7-one

(2.12 g, 7.8 mmol) in anhydrous dimethylformamide (40 mL) sodium hydride (60% on mineral oil, 467 mg, 11.7 mmol) was added and the reaction was stirred at room temperature for 30 min. A solution of 2-chloromethyl-1-cyclopropyl-3-fluorobenzene (1.58 g, 8.6 mmol) in anhydrous dimethylformamide (10 mL) was added slowly and the reaction was stirred for 18 h. The mixture was poured onto ice water (250 g) and extracted with dichloromethane (3 x 80 mL). The combined organic layers were dried over magnesium sulfate, filtered and evaporated. The residue was triturated with n-hexane, the precipitated product was collected, washed with methanol, then diethyl ether. The crude product was recrystallized from ethyl acetate to give the title compound (1.31 g, 3.1 mmol, 54%) as a pale yellow solid. ESMS m/z 420 (M+H) + .

[00547] Step 3 : Synthesis of 6-bromo-8-(2-cyclopropyl-6-fluorobenzyl)-2- methanesulfinyl-8H-pyrido[2,3-d]pyrimidin-7-one.

[00548] To a solution of 6-bromo-8-(2-cyclopropyl-6-fluorobenzyl)-2-methylsulfanyl-

8H-pyrido[2,3-d]pyrimidin-7-one (500 mg, 1.2 mmol) in dichloromethane (20 mL) was added 3-chloroperbenzoic acid (77%, 253 mg, 1.1 mmol) in dichloromethane (15 mL) at 0-5 °C and the mixture was stirred at 0-5 °C for 1 h. The reaction mixture was washed with saturated sodium bicarbonate solution (2 x 30 mL) then with brine (30 mL). The organic layer was dried over sodium sulfate, filtered and evaporated. The crude product was purified by column chromatography eluting with chloroform to yield the title compound as a white solid (385 mg, 0.9 mmol, 74%). ESMS m/z 436 (M+H) + .

[00549] Step 4: Synthesis of 6-bromo-8-(2-cyclopropyl-6-fluorobenzyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one .

[00550] 6-Bromo-8-(2-cyclopropyl-6-fluorobenzyl)-2-methanesulfinyl-8 H-pyrido[2,3- d]pyrimidin-7-one (385 mg, 0.9 mmol) and 4-(4-methylpiperazino)aniline (169 mg, 0.9 mmol) were stirred at 140 °C for 6 h. The reaction mixture was purified by column chromatography using dichloromethane: methanol (100: 1→100:3→100:5) to yield the title compound as a yellow solid (272 mg, 0.48 mmol, 55%). ESMS m z 563 (M+H) + .

[00551] Step 5 : Synthesis of 8-(2-cyclopropyl-6-fluorobenzyl)-2-[4-(4-methyl-piperazin-

1 -yl)-phenylamino]-6-prop- 1 -ynyl-8H-pyrido [2,3-d]pyrimidin-7-one.

[00552] To the solution of 6-bromo-8-(2-cyclopropyl-6-fluorobenzyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one (130 mg, 0.23 mmol) in degassed dioxane (5 mL) tributyl(l-propynyl)-tin (105 μΐ, 0.34 mmol) and Pd(PPh 3 ) 4 (27 mg, 0.02 mmol) was added. The resulting mixture was irradiated at 100 °C for 90 min and evaporated. The solid residue was dissolved in dichloromethane (15 mL) and was extracted with saturated sodium bicarbonate (2 x 15 mL). The combined aqueous layers were back-extracted with dichloromethane (15 mL). The combined organic layers were dried over sodium sulfate and evaporated. The residue was dissolved in dichloromethane and filtered through a silica pad which was washed with dichloromethane: methanol (9: 1). The crude product was purified by preparative HPLC to give the title compound (25 mg, 0.05 mmol, 22%>) as a yellow solid. ESMS m/z 522 (M+H) + ; 1H NMR (400 MHz, CDC1 3 ) δ ppm 8.48 (s, 1H) 7.67 (s, 1H) 7.37 (d, J = 9.0Hz, 2H) 7.15 (br. s, 1H) 7.07 - 7.14 (m, 1H) 6.99 (s, 1H) 6.83 - 6.91 (m, 3H) 6.75 - 6.82 (m, 1H) 5.81 (s, 2H) 3.26 (br. s, 4H) 2.73 (br. s, 4H) 2.47 (br. s, 3H) 2.19 - 2.27 (m, 1H) 2.09 (s, 3H) 0.81 - 0.87 (m, 2H) 0.62 - 0.69 (m, 2H).

Example 27: Synthesis of 8-(2-cyclopropyl-6-fluoro-benzyl)-6-ethynyl-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one (Compd 54).

[00553] Step 1 : Synthesis of 8-(2-cyclopropyl-6-fluorobenzyl)-6-ethynyl-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one .

[00554] The mixture of 6-bromo-8-(2-cyclopropyl-6-fluorobenzyl)-2-[4-(4-methyl- piperazin-1-yl)-phenylamino]-8H-pyrido[2,3-d]pyrimidin-7-one (120 mg, 0.21 mmol), ethynyltrimethylsilane (74 μΐ, 0.53 mmol), PdCl 2 (PPh 3 ) 2 (15 mg, 0.02 mmol) and Cul (3 mg,

0.02 mmol) in triethylamine (1.5 mL) was irradiated for 90 min at 80 °C in a microwave reactor. The reaction mixture was cooled, filtered through Celite (washed with triethylamine and dichloromethane) and evaporated to dryness. The crude material was purified by column chromatography over silica eluting with chloroform. The isolated material (87 mg) was suspended in 5 mL methanol, potassium carbonate (38 mg, 0.28 mmol) was added and the yellow suspension was stirred for 30 min at room temperature and evaporated. The solid residue was partitioned between water (10 mL) and dichloromethane (2 x 15 mL). The combined organic layers were washed with brine (15 mL), dried over sodium sulfate, filtered, evaporated and purified by filtration through silica with chloroform:methanol (95:5) followed by

preparative HPLC. The title compound (19 mg, 0.04 mmol, 17%) was obtained as a yellow powder. ESMS m/z 509 (M+H) + ; 1H NMR (400 MHz, CDC1 3 ) δ ppm 8.50 (s, 1H) 7.79 (s, 1H) 7.34 (d, J= 8.5Hz, 2H) 7.19 (br. s, 1H) 7.07 - 7.14 (m, 1H) 6.82 - 6.92 (m, 3H) 6.78 (dd, J = 10.5, 8.8Hz, 1H) 5.80 (s, 2H) 3.28 (s, 1H) 3.16 - 3.25 (m, 4H) 2.60 - 2.69 (m, 4H) 2.39 (s, 3H) 2.16 - 2.27 (m, 1H) 0.80 - 0.87 (m, 2H) 0.61 - 0.68 (m, 2H).

Example 28 (Compd 55):

[00555] The following compound was made by the method of Example 26 using the appropriate aryl methyl halide at Step 2, aniline at Step 4 and alkyne at Step 5. If necessary, the aryl methyl halide was synthesized by the method used for Intermediate 1. Examples containing secondary amines on the aniline were synthesized using the appropriate Boc protected amino aniline and in the final step were treated with a solution of hydrogen chloride in an organic solvent to produce the example compound, usually isolated as the hydrochloride salt.

Example 29: Identification of Compounds Having High Affinity for PAK Active Sites

[00556] A fluorescence-based assay format is used to determine IC50 values of test compounds in vitro. Purified PAK kinase is incubated with ATP, and a test compound at various concentrations and a substrate peptide containing two fluorophores. In a second step, the reaction mix is incubated with a site-specific protease that cleaves non-phosphorylated but not phosphorylated substrate peptide, disrupting the FRET signal generated by the two fluorophores in the cleaved peptide (Z'Lyte™ Kinase assay platform; Life Technologies).

[00557] Reagents: 50 mM HEPES, pH 7.5; 0.01% BRIJ-35; 10 mM MgCl 2 ; 1 mM

EGTA, 2 uM substrate peptide Ser/Thr20 (proprietary Life Technologies Sequence), PAK enzyme [2.42-30.8 ng for PAK1, 0.29 - 6 ng for PAK2, 1.5 - 20 ng for PAK3 and 0.1 - 0.86 ng for PAK4; actual enzyme amounts depend on lot activity of the enzyme preparation]

[00558] Test compounds are dissolved in DMSO at various concentrations; the final

DMSO concentration in the assay reaction is 1%.

[00559] ATP concentration at Km apparent is used in the assay [50 ΓΜ ATP for PAKl assay, 75 ΓΜ ATP for PAK2 assay, 100 μΜ ATP for PAK3 assay, 5 μΜ ATP for PAK4 assay] in a total assay volume of 10 μΐ. Assay reactions are incubated at room temperature for 1 fir. Following the kinase reaction, 5 μΐ of 1 :256 dilution of development solution A (Life

Technologies) is added and the reaction mix is incubated for an additional 1 fir at room temperature.

[00560] Plates are analyzed in a standard fluorescence plate reader (Tecan or equivalent) using an excitation wavelength of 400 nm and emission wavelengths of 445 nm and 520 nm. Inhibition of kinase reaction is determined by

emission ratio = emission@ 445 nm / emission@ 520 nm [00561] Based on these data, specific compounds have been identified that have relatively high affinity for the catalytic domain of at least one PAK isoform, and are therefore useful inhibitors, as described herein.

Table 1

A: IC50 < ΙμΜ; B: IC50 >1 μΜ and < 10 μΜ; C: IC50 > 10 μΜ

Example 30: Slice electrophysiology assay for determination of PAK inhibitory activity

[00562] Materials: coronal cortical slices (400 μηι) containing temporal cortex from 2- to

3-month-old C57-Black-6 mice male littermates (from Elevage Janvier, FRANCE) are prepared and allowed to recover in oxygenated (95% 02 and 5% C02) warm (30°C) artificial cerebrospinal fluid (ACSF) containing 124 mM NaCl, 5 mM KCl, 1.25 mM, NaH 2 P0 4 , 1 mM MgCl 2 , 2 mM CaCl 2 , 26 mM NaHC0 3 , and 10 mM dextrose.

[00563] Compound dilution: a 10 mM DMSO stock solution is prepared for each test compound and 100 aliquots are stored at -20°C. On the day of experiment an aliquot is thawed and vortexed for fresh solutions preparation. The final concentration of DMSO is adjusted to 0.1 % in all solutions, including control ACSF solution.

[00564] Perfusion: Artificial Cerebro-Spinal Fluid (ACSF) is perfused at 3 mL/min. The recording chamber has a volume of 1 mL. Then the chamber medium is renewed every 20 s. The perfusion liquid is maintained at 30 ± 0.1 °C.

[00565] Data acquisition: evoked-responses are sampled at 5 kHz before recording on the harddisk of the computer

[00566] Recording in cortical layer II/III: The recording is carried out on a Multi

Electrode Array. Responses (field portentials) in layer II/III are evoked by layer IV stimulation between one MEA electrode and the GND electrode. I/O curve is first performed to define evoked responses for stimulation intensities between 100 and 800 μΑ, by 100 μΑ steps. The stimulus consists in a monopolar biphasic current pulse (negative for 60 μβ and then positive for 60 μβ) which is applied every 30 s to evoke "responses" (field Excitatory Post Synaptic

Potentials; (fEPSP) in cortical layer II/III. [00567] Basal synaptic transmission: a monopolar stimulation (a bi-phasic stimulus:± 300 mA for 120 ms between one MEA electrode and the GND) is applied every 30 s on the MPP fibres to evoke "responses" (field potentials: fEPSP) in the DG region. The basal stimulation intensity will be set to evoke 40% of maximal amplitude response. The same stimulation intensity will be used in the 100 Hz stimulation protocol.

[00568] LTP: a stimulus is applied every 30 s with an intensity settled at 40 % of the maximal amplitude responses. LTP is then induced by TBS, which consists of eight brief bursts (each with four pulses at 100 Hz) of stimuli delivered every 200 ms. Potentiation of synaptic transmission is then monitored for an additional 40 minutes period. Since fEPSP result from glutamatergic synaptic transmission consecutive to afferent pathway stimulation, 10 μΜ NBQX are perfused on the slice, at the end of each experiment, to validate the glutamatergic nature of synaptic transmission as well as to subtract background noise at individual electrode level.

[00569] Compound evaluation: following a 10 minutes control recording period (to verify baseline stability), the compound is perfused for 20 minutes. Then, LTP is triggered and the fEPSP amplitude will be recorded for an additional 40 minutes period in the presence of compound.

[00570] Data analysis: fEPSP amplitudes are measured as the difference between the baseline (before stimulation) and the maximal peak amplitude. The fEPSP are normalized as a percent of the meanaveraged amplitude recorded over a 10 min control period, before compound application. Normalized fEPSP values are then averaged for each experiment carried out in control conditions and with the test compound. The fEPSP mean values (+/- SEM) are expressed as a function of time before and after LTP induction.

[00571] An increased LTP, indicates an increase in synaptic plasticity mediated by inhibition of PAK. Compounds are tested using the procedure described above to determine the effect of PAK inhibitors on synaptic plasticity.

Example 31 In Vivo Monitoring of Changes in the Dendritic Spines in NF1+/- heterozygote animals or R6/2 transgenic animals Treated with a PAK Inhibitor

[00572] In the following experiment, changes in the dendritic spines of double transgenic

GFP/NF1 +/- or GFP/R6/2 mice treated with a PAK inhibitor or a placebo are directly monitored in vivo by two photon laser scanning microscopy (TPLSM). Mice expressing GFP in a subset of cortical layer 5 neurons (transgenic line GFP-M described in Feng et al., 2000, Neuron 28:41- 51) are crossed with either NF1 +/- heterozygote animals {Jacks et al, 1994, Nat Genet 7:353- 361) or R6/2 transgenic animals {Mangiarini et al, 1996, Cell 87(3): 493-506) to obtain double transgenic mice.

[00573] GFP/NF1 +/- or GFP/R6/2 animals aged 60-90 d are anesthetized using avertin (16 μΐ/g body weight; Sigma, St. Louis, MO). The skull is exposed, scrubbed, and cleaned with ethanol. Primary visual, somatosensory, auditory, and motor cortices are identified based on stereotaxic coordinates, and their location is confirmed with tracer injections (see below).

[00574] Long-term imaging experiments are started at P40. The skull is thinned over the imaging area as described in Grutzendler et al, (2002), Nature, 420:812-816. A small metal bar is affixed to the skull. The metal bar is then screwed into a plate that connected directly to the microscope stage for stability during imaging. The metal bar also allows for maintaining head angle and position during different imaging sessions. At the end of the imaging session, animals are sutured and returned to their cage. Thirty animals previously imaged at P40 are then divided into a control group receiving a 1% sugar solution (oral gavage once per day) and a treatment group administered a PAK inhibitor, in 0.1% DMSO (oral gavage. 1 mg/kg, once per day). During the subsequent imaging sessions (at P45, P50, P55, or P70), animals are reanesthetized and the skull is rethinned. The same imaging area is identified based on the blood vessel pattern and gross dendritic pattern, which generally remains stable over this time period.

[00575] At the end of the last imaging session, injections of cholera toxin subunit B coupled to Alexa Fluor 594 are made adjacent to imaged areas to facilitate identification of imaged cells and cortical areas after fixation. Mice are transcardially perfused and fixed with paraformaldehyde, and coronal sections are cut to verify the location of imaged cells. Sections are then mounted in buffer, coverslipped, and sealed. Images are collected using a Fluoview confocal microscope (Olympus Optical, Melville, NY).

[00576] For in vivo two photon imaging, a two-photon laser scanning microscope is used as described in Majewska et al, (2000), Pfliigers Arch, 441 :398-408. The microscope consists of a modified Fluoview confocal scan head (Olympus Optical) and a titanium/sulphur laser providing 100 fs pulses at 80 MHz at a wavelength of 920 nm (Tsunami; Spectra-Physics, Menlo Park, CA) pumped by a 10 W solid-state source (Millenia; Spectra-Physics).

Fluorescence is detected using photomultiplier tubes (HCl25-02; Hamamatsu, Shizouka, Japan) in whole-field detection mode. The craniotomy over the visual cortex is initially identified under whole-field fluorescence illumination, and areas with superficial dendrites are identified using a 20x, 0.95 numerical aperture lens (IR2; Olympus Optical). Spiny dendrites are further identified under digital zoom (7-1 Ox) using two-photon imaging, and spines 50-200 μιη below the pial surface are studied. Image acquisition is accomplished using Fluoview software. Dendrites and axons located in layers 1-3 are studied. Although both layer 5 and layer 6 neurons are labeled in the mice used in this study, only layer 5 neurons send a clear apical dendrite close to the pial surface thus, the data will come from spines on the apical tuft of layer 5 neurons and axons in superficial cortical layers.

[00577] Images are exported to Matlab (Math Works, Natick, MA) in which they are processed using custom- written algorithms for image enhancement and alignment of the time series. For spine motility measurements (see Majewska et al, (2003), Proc Natl Acad Sci USA, 100: 16024-16029) spines are analyzed on two-dimensional projections containing between 5 and 30 individual images; therefore, movements in the z dimension are not analyzed. Spine motility is defined as the average change in length per unit time (micrometers per minute). Lengths are measured from the base of the protrusion to its tip. The position of spines are compared on different imaging days. Spines that are farther than 0.5 μιη laterally from their previous location are considered to be different spines. Values for stable spines are defined as the percentage of the original spine population present on the second day of imaging. Only areas that show high signal-to-noise ratio in all imaging sessions will be considered for analysis. Analysis is performed blind with respect to animal age and sensory cortical area. Spine motility, spine morphology and spine density are then compared between control and treatment groups. It is expected that treatment with the PAK inhibitor will rescue defective spine morphology relative to that observed in untreated control animals.

Examples 32-34 Compounds described herein are tested using the procedure described above in Example 31.

Example 35 Treatment of Clinical depression by Administration of a PAK Inhibitor in an Animal Model

[00578] A rat olfactory bulbectomy (OBX) model of clinical depression (see, e.g., van

Riezen et al (1990), Pharmacol Ther, 47(l):21-34) is used to evaluate treatment of clinical depression with a PAK inhibitor described herein. Dendritic spine density and morphology are compared in treated and untreated groups of animals as described below. It is expected that treatment of OBX animals with a PAK inhibitor described herein will cause an increase in spine density relative to that observed in untreated OBX animals.

[00579] All experiments are performed in strict accordance with N1H standards for laboratory animal use. The study uses 48 adult male Sprague-Dawley rats (230-280 g) housed in groups of four animals (two sham and two OBX), as indicated in van Riezen et al supra, in a controlled environment with food and water available ad libitum. Half of the experimental animals (n = 24) undergo bilateral olfactory bulbectomy (OBX) while the other half undergot sham surgery (n = 24). Upon completion of surgery, animals are allowed to recover for 2 weeks prior to behavioral testing. This is necessary to: 1) allow for the recovery of animal body weight which is reduced following surgery, 2) allow complete healing of superficial surgical sites, and ) "bulbectomy syndrome" develops during the first 2 weeks postsurgery.

[00580] Two weeks after surgery, OBX and sham-operated animals are subdivided into one of four experimental conditions. One group of OBX animals is administered daily injections of 0.9% saline (n = 6 for each surgical condition) or a compound described herein (5 mg/kg) (n = 6 for each surgical condition). These groups are included to examine the effect of chronic administration of a PAK inhibitor on olfactory bulbectomized animals (2 weeks postsurgical recovery + 2 weeks PAK inhibitor described herein treatment). Injections are given at the same time each day and in the home cage of each animal. Groups of OBX and sham-operated animals receive no treatment during this 2-week period and serve as unhandled controls. These groups are necessary to examine the persistence of observed effects of OBX on dendritic spine density (4 weeks postsurgery). Animals receiving postsurgery drug treatment are sacrificed 24 h after the last injection.

[00581] Animals are perfused transcardially with 4% formaldehyde (in 0.1 M sodium phosphate buffer, pH = 7.4) under deep anesthesia with sodium pentobarbital (60 mg/kg) at the completion of experimental procedures. Following fixation, brains are removed and placed in 4% formaldehyde (freshly depolymerized from para- formaldehyde) overnight. Brains are then sectioned at 100 μιη on a vibratome and prepared for Golgi impregnation using a protocol adapted from previously described methods (Izzo et al, 1987). In brief, tissue sections are postfixed in 1% Os0 4 for 30 min and then washed in 0.1 M phosphate buffer (3 X 15 min). Sections are free-floated in 3.5% K 2 Cr 2 07 solution for 90 min, mounted between two microscope slides in a "sandwich" assembly, and rapidly immersed in a 1% AgN0 3 solution. The following day, sections are rinsed in ddH 2 0, dehydrated in 70%> and 100% ethanol, cleared with Histoclear™, and mounted on microscope slides with DPX.

[00582] Dendritic spines are counted on 125 OX camera lucida images that include all spines observable in each focal plane occupied by the dendrite. Cells are analyzed only if they are fully impregnated (CA1: primary apical dendrites extended into stratum lacunosum moleculare and basilar dendrites extended into stratum oriens; CA3: primary apical dendrites extended into stratum lacunosum moleculare and basilar dendrites extended into stratum oriens; dentate gyrus: secondary dendrites extended from primary dendrite within the molecular layer), intact, and occurring in regions of the section that are free of blood vessels, precipitate, and/or other imperfections. Dendritic spines are counted along the entire length of secondary oblique dendritic processes (50-100 μηι) extending from the primary apical dendrite within stratum radiatum of area CA1 and CA3. In CA1 and CA3, secondary dendrites are defined as those branches projecting directly from the primary apical dendrite exclusive of tertiary daughter branches. In addition, spines are counted along the length of secondary dendrites of granule cells in the dentate gyrus to determine if effects are limited to CA1 and CA3. In dentate gyrus, secondary dendrites are analyzed in the glutamatergic entorhinal input zone in the outer two- thirds of the molecular layer. Approximately 20 dendritic segments (10 in each cerebral hemisphere; 50-100 μιη in length) in each hippocampal subregion (CA1, CA3, and dentate gyrus) are examined for each experimental animal. Treatment conditions are coded throughout the entire process of cell identification, spine counting, dendritic length analysis, and subsequent data analysis. Analysis of variance and Tukey post-hoc pairwise comparisons are used to assess differences between experimental groups.

[00583] When significant changes in dendritic spine density are observed, camera lucida images and the Zeiss CLSM measurement program are used to quantify the number and length of secondary dendrites. This analysis is necessary as apparent changes in dendritic spine density can result from an increase or decrease in the length of dendrites and not the formation or loss of spines per se. Photomicrographs are obtained with a helium-neon 633 laser and Zeiss 410 confocal laser scanning microscope.

[00584] Examples 36-42 Compounds described herein are tested using the procedure described in Example 35.

Example 43 Treatment of Epilepsy by Administration of a PAK Inhibitor in an Animal Model

[00585] A rat tetanus toxin model of epilepsy is used to evaluate treatment of epilepsy with a PAK inhibitor (Compound 102).

[00586] Wistar rat pups (Harlan Sprague Dawley, Indianapolis, IN), 10 d of age, are anesthetized with an intraperitoneal injection of ketamine and xylazine (33 and 1.5 mg/kg, respectively). When necessary, this is supplemented by inhalation of methoxyflurane

(Metofane). Tetanus toxin solution to be injected is generated by dissolving 2.5 or 5 ng of tetanus toxin in 20 or 40 nl of sterile saline solution. Afterwards, the tetanus toxin solution is coinjected into the right hippocampus along with a solution (DMSO + water) of a PAK inhibitor described herein.

[00587] To inject tetanus toxin and a PAK inhibitor described herein, the pups are placed in an infant rat stereotaxic head holder, a midline incision is made, and a small hole is drilled in the skull. The stereotaxic coordinates for injection are: anteroposterior, -2.1 mm; medio lateral, 3.0 mm from the bregma; and dorsoventral,—2.95 mm from the dural surface. The toxin and Compound 102 solution are slowly injected at 4 nl/min. After injection, the needle is left in place for 15 min to reduce reflux up the needle track. During injections, the body temperature of rat pups is maintained by a warmed (electrically regulated) metal plate. Littermates,

stereotaxically injected with sterile saline, or untreated rats serve as controls.

[00588] The frequency of behavioral seizures is monitored for 1 hr/day for 10 consecutive days after tetanus toxin or a PAK inhibitor injections. The types and duration of seizures are scored. Wild running seizures are most easily identified.

[00589] After seizure scoring on the 10 th day animals are perfused transcardially and dendritic spines in the CA3 region are counted and analyzed as described above.

[00590] The t test for comparison of two independent means is used in comparing the number of seizures in treated vs untreated rats and in comparing dendritic and axon arbors in experimental and control rats. When data are not normally distributed, a Mann- Whitney U test is used. Sigma Stat is used to perform all statistical tests.

Example 44 Clinical Trial: Treatment of Down's Syndrome with a PAK Inhibitor

[00591] The following human clinical trial is performed to determine the safety and efficacy of the PAK inhibitor Compound 4 for the treatment of Down's syndrome. This is a one year study to assess whether PAK inhibitor described herein is effective and safe in preventing age related cognitive deterioration and dementia in people with Down's syndrome (DS) age 40 and over. Participants will be assessed on memory skills, attention and problem solving abilities. Quality of life and abilities for everyday living skills will also be regularly checked.

[00592] Study Type: Interventional

[00593] Study Design: Treatment, Randomized, Double-Blind, Placebo Control, Parallel

Assignment, Safety/Efficacy Study

Primary Aims

[00594] Clinical: To determine the clinical efficacy of PAK inhibitor described herein versus placebo in preventing cognitive decline in people with DS; to compare the safety and tolerability of PAK inhibitor versus placebo in people with Down's syndrome (DS).

[00595] Biochemical and pathological: To examine the ability of PAK inhibitor to alter markers of disease progression in DS patients.

Secondary Aims

[00596] Clinical: To determine whether PAK inhibitor described herein has, as compared with placebo, a significant positive impact on: level of independent functioning as measured by the carer-rated adaptive behavioural scale, (ABS) in adults with DS; quality of life in adults with DS.

Inclusion criteria

[00597] Participants with learning disabilities due to Down's syndrome (DS) confirmed by karyotype. A clinical diagnosis (provided by the participant's general practitioner or hospital specialist) will be accepted if karyotype is not known and participant does not agree to have it tested. Ages 40 years and over or any age if a diagnosis of dementia is established In participants with dementia, the diagnosis will be consistent with the 10th version of the

International Classification of Diseases (ICD-10) (World Health Organization [WHO], 1992) diagnostic criteria. Level of speech and comprehension of verbal commands are sufficient to understand and to answer simple requests. Resident in care facility or community living with a carer who is willing to accept responsibility for supervising the treatment and will provide input to efficacy parameters in accordance with protocol requirements

Exclulsion criteria

[00598] Severe, unstable or uncontrolled medical or psychiatric conditions apparent from history, physical examination or investigations. A current diagnosis of primary

neurodegenerative disorder other than dementia such as Huntington's disease, etc. Uncontrolled epilepsy. Presence of severe motor or sensory impairment (severe deafness or blindness) that renders the participant as untestable with the battery of tests used in the study. Current evidence of delirium. Severe renal impairment. Low probability of treatment compliance

[00599] The effects of Compound 4 on Neuropsychological Test Battery and

Neurological Inventory (NPI) are analyzed (separately) by 2 (Treatment: a PA inhibitor described herein, placebo) x 3 (Time: baseline, 12 weeks, 26 weeks, 52 weeks) analysis of variance (ANOVA). Patients are divided into two groups, a placebo group and a a PAK inhibitor group. Each patient receives two daily doses of placebo or a PAK inhibitor. Patients are monitored for one year.

[00600] All cognitive variables are first examined for their distribution properties, i.e., to ensure normality. The cognitive effects of a PAK inhibitor over time are then evaluated by Treatment x Time ANOVA, performed separately for each variable, with Time as a within- individuals factor and Treatment as a between-individuals factor, followed by post-hoc mean comparisons wherever appropriate. All cognitive effects are then re-evaluated using ANOVA performed separately on change scores computed for each variable (12 weeks data minus baseline data, 26 weeks, 52 weeks data minus baseline data). Alpha level for testing significance of effects is p = 0.05. Primary Outcome Measures:

[00601] Down's Attention Memory and Executive Function Scale. Part I of the Adaptive

Behaviour Scale

Secondary Outcome Measures:

[00602] Clinician's Global Impression of Change

Example 45 Clinical Trial: Treatment of Clinical Depression with a

PAK1/PAK2/PAK3 Inhibitor

[00603] The purpose of the study is to examine the effectiveness of treatment with a

PAK1/PAK2/PAK3 inhibitor described herein in patients with refractory depression. Refractory depression is defined as clinical depression that is unimproved after treatment with at least 2 different antidepressants of adequate dose and time trial.

[00604] This is a 40-week, randomized, double blind, parallel groups designed, study of an oral PAK1/PAK2/PAK3 inhibitor described herein in symptomatic patients with a diagnosis of refractory clinical depression.

[00605] Study Type: Interventional

[00606] Study Design: Treatment, Randomized, Double-Blind, Placebo Control, Single

Group Assignment, Safety/Efficacy Study

Primary Outcome Measures:

[00607] Change in Ham-D scores to assess mood response to a PA 1/PAK2/PAK3 inhibitor.

Secondary Outcome Measures:

[00608] Change in stress hormone levels pre and post treatment relative to mood response

Inclusion Criteria:

[00609] 21 -item HAM-D score of 20 or above. If currently taking antipsychotic, antidepressant, anticonvulsant, and/or mood-stabilizing medications, must be stable on the medication for at least three weeks prior to entering the study. At least 2 failed antidepressant medication trials of adequate dose and duration. Between 18 and 75 years of age. Not currently pregnant or trying to become pregnant.

Exclusion criteria:

[00610] History of schizophrenia or other psychotic disorders. Transcranial magnetic stimulation treatment or ECT in the 3 months prior to starting the study. History of vagus nerve stimulation treatment. No unstable or untreated cardiovascular disease, hypertension, or endocrine disorder. Experimental Design

[00611] Patients are divided into two groups, a placebo group and a PAK1/PAK2/PAK3 inhibitor group. Each patient receives a daily dose of placebo or a PA 1/PAK2/PAK3 inhibitor. Patients are monitored for a period of 40 weeks with experimental sessions every 4 weeks.

[00612] Groups are compared using ANOVA. Single variable differences are analyzed using an independent samples t-test. A Pearson's coefficient is used to determine relationship between depression and medication dose. Clinical Global Impressions (CGI) score, and change in the Hamilton Rating Scale for Depression D (Ham-D) scores are recorded at each visit.

Example 46 Clinical Trial of a PAK Inhibitor in reducing choreia in patients with

Huntington's Disease

[00613] The purpose of the study is to examine the effectiveness of treatment with a PAK inhibitor in reducing choreia in patients with Huntington's Disease.

[00614] This is a 24-week, randomized, double blind, parallel groups designed, study of an oral PAK inhibitor in symptomatic patients with a diagnosis of refractory clinical depression.

[00615] Study Type: Interventional

[00616] Study Design: Randomized, Double-blind, Placebo-controlled, Parallel-group,

Multiple Oral Dose Titration Proof of Concept Study in Patients with Huntington's Disease to assess the efficacy, safety and tolerability of a PAK inhibitor described herein.

Primary Outcome Measures:

[00617] Efficacy of a PAK inhibitor on the severity of choreia in Huntington's disease measured by Unified Huntington's Disease Rating Scale (UHDRS) Maximal Choreia score. Secondary Outcome Measures:

[00618] Safety and tolerability of a PAK inhibitor in Huntington's disease patients.

Potential effect of a PAK inhibitor described herein on the motor, cognitive, behavioral and functional assessments using UHDRS. Potential effect of a PAK inhibitor described herein on functional and quality of life scales, neurological assessments and cognitive assessments in Huntington's Disease patients

Inclusion Criteria:

[00619] Huntington's disease (based on DNA testing polyQ >36) with a UHDRS maximal choreia score of >10. Patients with concomitant Huntington's medication (anti-depressants, neuroleptics, benzodiazepines) are allowed but the total daily dose and dosing regimen has to be stable for at least one months prior to randomization. Female patients without childbearing potential (post-menopausal or surgically sterilized). Exclusion Criteria:

[00620] Patients with marked cognitive impairment (MMSE less than 18), with presence of psychosis and/or confusional states. Patients with a history or presence of renal impairment and/or liver disease Other protocol-defined inclusion/exclusion criteria may apply

Experimental Design

[00621] Patients are divided into two groups, a placebo group and a a PAK inhibitor treatment group. Each patient receives a daily dose of placebo or a PAK inhibitor. Patients are monitored for a period of 24 weeks with experimental sessions every 4 weeks.

[00622] Groups are compared using ANOVA. Single variable differences are analyzed using an independent samples t-test. A Pearson's coefficient is used to determine relationship between depression and medication dose. Clinical Global Impressions (CGI) score, and change in the Unified Huntington's Disease Rating Scale (UHDRS) Maximal Choreia scores are recorded at each visit.

Example 47 Clinical Trial of a PAK Inhibitor for Treatment of Patients with Plexiform Neurofibromas that Cannot be Removed by Surgery

[00623] The purpose of this 30 month study is to examine the effectiveness of treatment with a PAK inhibitor in treating patients with neurofibromatosis type 1 plexiform neurofibromas that cannot be removed by surgery.

[00624] Study Type: Interventional

[00625] Study Design: Open Label

Experimental Design

[00626] Patients are divided into two groups, a placebo group and a a PAK inhibitor described herein treatment group. Each patient receives a daily oral dose of placebo or a PAK inhibitor for days 1-7. The treatment is repeated every 28 days for up to 30 courses.

Primary Outcome Measures:

[00627] Time to tumor progression as assessed by volumetric MRI analysis at baseline, after courses 3, 6, 9, and 15, after every 6 courses, and then at the end of treatment; Objective radiographic response as assessed by volumetric MRI analysis at baseline, after courses 3, 6, 9, and 15, after every 6 courses, and then at the end of treatment.

Secondary Outcome Measures:

[00628] Comparison of volumetric 3-D MRI measurements with conventional 2-D and 1-

D MRI measurements in determining tumor response; Clinical response, defined as

improvement in function and performance status or decrease in plexiform neurofibroma-related pain. Example 48: Pharmaceutical Compositions

Example 48a: Parenteral Composition

[00629] To prepare a parenteral pharmaceutical composition suitable for administration by injection, 100 mg of a water-soluble salt of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is dissolved in DMSO and then mixed with 10 mL of 0.9% sterile saline. The mixture is incorporated into a dosage unit form suitable for administration by injection.

Example 48b: Oral Composition

[00630] To prepare a pharmaceutical composition for oral delivery, 100 mg of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is mixed with 750 mg of starch. The mixture is incorporated into an oral dosage unit for, such as a hard gelatin capsule, which is suitable for oral administration. Example 48c: Sublingual (Hard Lozenge) Composition

[00631] To prepare a pharmaceutical composition for buccal delivery, such as a hard lozenge, mix 100 mg of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, with 420 mg of powdered sugar mixed, with 1.6 mL of light corn syrup, 2.4 mL distilled water, and 0.42 mL mint extract. The mixture is gently blended and poured into a mold to form a lozenge suitable for buccal administration. Example 48d: Fast-Disintegrating Sublingual Tablet

[00632] A fast-disintegrating sublingual tablet is prepared by mixing 48.5% by weigh of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, 44.5%> by weight of micro crystalline cellulose (KG-802), 5%> by weight of low-substituted hydro xypropyl cellulose (50 μιη), and 2%> by weight of magnesium stearate. Tablets are prepared by direct compression {AAPS PharmSciTech. 2006;7(2):E41). The total weight of the compressed tablets is maintained at 150 mg. The formulation is prepared by mixing the amount of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula

VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, with the total quantity of micro crystalline cellulose (MCC) and two-thirds of the quantity of low-substituted hydro xypropyl cellulose (L- HPC) by using a three dimensional manual mixer (lnversina ®, Bioengineering AG,

Switzerland) for 4.5 minutes. All of the magnesium stearate (MS) and the remaining one-third of the quantity of L-HPC are added 30 seconds before the end of mixing.

Example 48e: Inhalation Composition

[00633] To prepare a pharmaceutical composition for inhalation delivery, 20 mg of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula

VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is mixed with 50 mg of anhydrous citric acid and 100 mL of 0.9% sodium chloride solution. The mixture is incorporated into an inhalation delivery unit, such as a nebulizer, which is suitable for inhalation administration.

Example 48f: Rectal Gel Composition

[00634] To prepare a pharmaceutical composition for rectal delivery, 100 mg of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is mixed with 2.5 g of methylcelluose (1500 mPa), 100 mg of methylparapen, 5 g of glycerin and 100 mL of purified water. The resulting gel mixture is then incorporated into rectal delivery units, such as syringes, which are suitable for rectal

administration.

Example 48g: Topical Gel Composition

[00635] To prepare a pharmaceutical topical gel composition, 100 mg of a compound of

Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is mixed with 1.75 g of hydro xypropyl celluose, 10 mL of propylene glycol, 10 mL of isopropyl myristate and 100 mL of purified alcohol USP. The resulting gel mixture is then incorporated into containers, such as tubes, which are suitable for topicl administration.

Example 48h: Ophthalmic Solution Composition

[00636] To prepare a pharmaceutical opthalmic solution composition, 100 mg of a compound of Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is mixed with 0.9 g of NaCl in 100 mL of purified water and filterd using a 0.2 micron filter. The resulting isotonic solution is then incorporated into ophthalmic delivery units, such as eye drop containers, which are suitable for ophthalmic administration.

Example 48i: Nasal spray solution

[00637] To prepare a pharmaceutical nasal spray solution, 10 g of a compound of

Formula I, a compound of Formula II, a compound of Formula III, a compound of Formula IV, a compound of Formula V, a compound of Formula VI, a compound of Formula VII, a compound of Formula VIII, a compound of Formula IX, a compound of Formula X, a compound of Formula XI, a compound of Formula XII, a compound of Formula XIII, a compound of Formula XIV, a compound of Formula XV, a compound of Formula XVI, a compound of Formula XVII, a compound of Formula XVIII, a compound of Formula XIX, a compound of Formula XX, a compound of Formula XXI, a compound of Formula XXII, or a compound of Formula XXIII, is mixed with 30 mL of a 0.05M phosphate buffer solution (pH 4.4). The solution is placed in a nasal administrator designed to deliver 100 μΐ of spray for each application.

[00638] While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.