AU2017200078B2 - Treatment of neurodegenerative diseases through inhibition of Hsp90 - Google Patents

Treatment of neurodegenerative diseases through inhibition of Hsp90 Download PDF

Info

Publication number
AU2017200078B2
AU2017200078B2 AU2017200078A AU2017200078A AU2017200078B2 AU 2017200078 B2 AU2017200078 B2 AU 2017200078B2 AU 2017200078 A AU2017200078 A AU 2017200078A AU 2017200078 A AU2017200078 A AU 2017200078A AU 2017200078 B2 AU2017200078 B2 AU 2017200078B2
Authority
AU
Australia
Prior art keywords
propyl
amino
ethyl
hsp90
tau
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2017200078A
Other versions
AU2017200078A1 (en
AU2017200078C1 (en
Inventor
Gabriela Chiosis
Fei Dou
Paul Greengard
Huazhong He
Wenjie Luo
Danuta Zatorska
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sloan Kettering Institute for Cancer Research
Rockefeller University
Original Assignee
Sloan Kettering Institute for Cancer Research
Rockefeller University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2007269144A external-priority patent/AU2007269144B2/en
Application filed by Sloan Kettering Institute for Cancer Research, Rockefeller University filed Critical Sloan Kettering Institute for Cancer Research
Priority to AU2017200078A priority Critical patent/AU2017200078C1/en
Publication of AU2017200078A1 publication Critical patent/AU2017200078A1/en
Publication of AU2017200078B2 publication Critical patent/AU2017200078B2/en
Application granted granted Critical
Publication of AU2017200078C1 publication Critical patent/AU2017200078C1/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

Treatment of neurodegenerative diseases is achieved using small molecule purine scaffold compounds that inhibit Hsp90 and that possess the ability to cross the blood-brain barrier or are otherwise delivered to the brain.

Description

Background of the Invention
This application relates to the treatment of neurodegenerative diseases through inhibition of heat shock protein 90 (HSP90).
The HSP90 family of proteins has four recognized members in mammalian cells: Hsp90 a and β, Grp94 and Trap-1. Hsp90 a and β exist in the cytosol and the nucleus in association with a number of other proteins. Hsp90 in its various forms is the most abundant cellular chaperone, and has been shown in experimental systems to be required for ATP-dependent refolding of denatured or “unfolded” proteins. It has therefore been proposed to function as part of the cellular defense against stress. When cells are exposed to heat or other environmental stresses, the aggregation of unfolded proteins is prevented by pathways that catalyze their refolding or degradation. This process depends on the association of the unfolded protein in an ordered fashion with multiple chaperons (Hsp 60, 90 and 70 and p23) forming a “refoldosome” and ultimately the ATP-dependent release of the chaperones from the refolded protein.
Hsp90 may also play a role in maintaining the stability and function of mutated proteins. It seems to be required for expression of mutated p53 and v-src to a much greater extent than for their wild-type counterparts. It has been suggested that this occurs as a result of Hsp90-mediated suppression of the phenotypes of mutations that lead to protein unfolding.
Hsp90 is also necessary to the conformational maturation of several key proteins involved in the growth response of the cell to extracellular factors. These include the steroid receptors as well as certain transmembrane kinases (i.e., Raf serine kinase, v-src and Her2). The mechanism whereby Hsp90 affects these proteins is not fully understood, but appears to
2017200078 06 Jan 2017
- 2 be similar to its role in protein refolding. In the case of the progesterone receptor, it has been shown that binding and release of Hsp90 from the receptor occurs in a cyclic fashion in concert with release of other chaperones and immunophilins and is required for high alfinily binding of the steroid to the receptor. Thus, Hsp90 could function as a physiologic regulator of signaling pathways, even in the absence of stress.
Hsp90 has been shown to be overexpressed in multiple tumor types and as a function of oncogenic transformation. Whether it plays a necessary role in maintaining transformation is unknown, but it could have at least three functions in this regard. Cancer cells grow in an environment of hypoxia, low pH and low nutrient concentration. They also rapidly adapt to or are selected to become resistant to radiation and cytotoxic chemotherapeutic agents. Thus, the general role of Hsp90 in maintaining the stability of proteins under stress may be necessary for cell viability under these conditions. Secondly, cancer cells harbor mutated oncogenic proteins. Some of these are gain-of-function mutations which are necessary for the transformed phenotype. Hsp90 may be required for maintaining the folded, I'unctionallyactivc conformation of these proteins. Thirdly, activation of signaling pathways mediated by steroid receptors, Raf and other Hsp90 targets is necessary for the growth and survival of many tumors which thus probably also require functional Hsp90.
Neurodegeneration, similar to cancer, is likely not the result of a single dysrcgulalory event, but rather a several-step process involving environmental, epigenetic and genetic events that lead to creation of a complex transformed phenotype manifested by abnormal expression, post-translational modification and processing of certain proteins. The functional maintenance of these dysregulated proteins in neurons may require, analogously to the cancer afflicted cell, the regulatory mechanism of molecular chaperones to evolve along with the transforming process.
In the context of neurodegenerative diseases, Hsp90 may play two roles. First, aberrantly activated kinases (such as cdk5/p35. gsk3beta) in neurodegenerative diseases may require Hsp90 for functioning. Thus, Hsp90 inhibition may restore damaged signaling networks in the diseased brain by alleviating aberrant phosphorylation, leading to reduced aberrant protein aggregation, and elimination or reduction of aggregates and of their associated toxicity. Second, pathogenic mutants (such as of APP or presenilins in AD or mtau in FTDP-17 or mutant androgen receptor in bulbar muscular atrophy) may require Hsp90 for
2017200078 06 Jan 2017
-3 correct folding and functioning, thus Hsp90 inhibition may lead to the elimination of these proteins and result in reduction of aggregates and consequent plaque or tangle formation.
Most neurodegenerative diseases are probably characterized by both mutants and aberrant signaling, and Hsp90 can play a role with respect to pathogenic mutants as well. Tau mutations cause autosomal dominant frontal temporal dementia. Pathologies linked to mutations of the androgen receptor include the complete androgen insensitivity syndrome (CAIS) and the spinal and bulbar muscular atrophy (SBMA or Kennedy's disease). (4) Mutations in the presenilin genes are the major cause of familial AD. Analysis of conditional knockout mice has shown that inactivation of presenilins results in progressive memory impairment and age-dependent neurodegeneration, suggesting that reduced presenilin activity might represent an important pathogenic mechanism. Presenilins positively regulate the transcription of cAMP response element (CRE)-containing genes, some of which arc known to be important for memory formation and neuronal survival. (5) Alzheimer's Disease (AD) is characterized both by NFTs (tau aggregates) and plaques (Αβ deposits). In Alzheimer's disease, mutations in amyloid precursor protein or in the presenilins cause autosomal dominant disease. These are the substrate and proteases responsible for the production of the deposited peptide Αβ. Prion mutations cause Gerstmann Strausslcr syndrome and hereditary Creutzfeldt-Jakob disease, alpha-synuclein mutations cause autosomal dominant Parkinson's disease. In these cases, the pathogenic mutation is in the protein that is deposited in the diseased tissue and the whole protein is deposited. Huntington D results from a mutant huntingtin. (9) Thus, in all the cases, the mutations lead to the disease by a mechanism that involves the deposition process.
These characteristics of Hsp90 make it a viable target for therapeutic agents. HSP90 family members possess a unique pocket in their N-terminal region that is specific to and conserved among all Hsp90s from bacteria to mammals, but which is not present in other molecular chaperones. The endogenous ligand for this pocket is not known, but it binds ATP and ADP with low affinity and has weak ATPase activity. The ansamycin antibiotics geldanamycin (GM) and herbimycin (HA) have been shown to hind to this conserved pocket, and this binding affinity has been shown for all members of the HSP90 family. International Patent Publication No. WO98/51702 discloses the use of ansamycin antibiotics coupled to a targeting moiety to provide targeted delivery of the ansamycin leading to the degradation of
2017200078 06 Jan 2017
-4 proteins in and death of the targeted cells. International Patent Publication No. WOOO/6I57S relates to bifunctional molecules having two moieties which interact with the chaperone protein Hsp90, including in particular homo- and heterodimers of ansamycin antibiotics.
These bifunctional molecules act to promote degradation and/or inhibition of HER-family tyrosine kinases and are effective for treatment of cancers which overexpress Hcr-kinascs.
Exemplary small molecule therapeutics that bind to the same binding pocket of Hsp90 as ATP and the ansamycin antibiotics are disclosed in PCT Publication No. W002/36075. PCT Application No. PCT/US06/03676 and US Patent Publications 2005-0113339. 20050004026, 2005-0049263, 2005-0256183, 2005-0119292, 2005-0113340 and 2005-0107343. all of which are incorporated herein by reference.
In aged organisms, chaperone overload leads to a significant decrease in the robustness of cellular networks shifting their function towards a more stochastic behavior. Unbalanced chaperone requirement and chaperone capacity helps the accumulation of misfolded and aggregated proteins especially in the nervous system, due to the very limited proliferation potential of neurons. In addition, damaged signaling networks lose their original stringency, and irregular protein phosphorylation occurs. An appealing approach to alleviating and reversing such damaging effects is by modulating Hsp90 activity. Inhibitors of Hsp90 activity release HSFI from Hsp90 complexes and conect the defective regulation of HSF1 activity after heat stress leading to an increase in cellular levels of chaperones, such as Hsp70 and Hsp40. Overexpression of these chaperones has been shown to represent a general way of reinstating proper folding and alleviating misfolded proteins' toxic effects. In addition to their effects on reinstating correct folding, Hsp90 inhibitors may regulate proteins involved in signaling networks of diseased neurons.
The usefulness of Hsp90 inhibitors as clinical agents in the treatment of neurodegenerative diseases, however, will depend on whether their effects occur at concentrations of drug that are tolerable to the patient and on whether the drugs can be administered in such a fashion so as to achieve these concentrations in the brain. Unfortunately, known Hsp90 inhibitors such as geldanamycin and J7AAG. its derivative in Phase I clinical trial for cancer, and the unrelated compound radicicol have significant limitations. They are poorly soluble, difficult to formulate and do not cross the blood-brain barrier. Thus, in order to realize the potential for treatment of neurodegenerative diseases.
2017200078 06 Jan 2017
- 5 therapeutic agents that inhibit Hsp90, and that have sufficient solubility and the ability to cross the blood-brain barrier or otherwise be delivered to the brain are needed.
Summary of the Invention
In accordance with the present invention, treatment of neurodegeneralive diseases is achieved using small molecule purine scaffold compounds that inhibit Hsp90 and that possess the ability to cross the blood-brain barrier. Thus, in accordance with the present invention, there is provided a method for treatment of neurodegenerative disease comprising the step of administering to an individual in need of such treatment an effective amount of a punnescaffold compound that inhibits Hsp90, and that crosses the blood-brain barrier or is otherwise delivered to the brain.
In one embodiment, the purine scaffold compound used in the method of the invention has a purine moiety connected at the 8- or 9-position via a linker to a monocyclic substituent group. Such compounds are described in PCT Publication No. W002/36075,
PCT Application No. PCT/US06/03676 and US Patent Publications 2005-0113339. 20050004026. 2005-0049263. 2005-0256183, 2005-0119292, 2005-0113340 and 2005-0107343.
In one embodiment, the method of the invention makes use of a small molecule purine scaffold compound has the general structure:
Figure AU2017200078B2_D0001
R 1-1 right side, ar/l left side, adenine
-62017200078 06 Jan 2017 wherein R is hydrogen, a C, to C,o alkyl, alkenyl, alkynyl, or an alkoxyalkyl group, optionally including heteroatoms such as N or O, optionally connected to the 2'-position to form an 8 to 10 member ring:
Y and Y, are independently C, N, S or O, with the proviso that when Y, and/or Y, is O the double bonds are missing or reananged to retain the aryl nature of the ring X4 is hydrogen, halogen, for example F or Cl, or Br;
X, is CH,, CF, S, SO, SO,, Ο, NH, or NR2, wherein R2 is alkyl; and
X, is halogen, alkyl, halogenated alkyl, alkoxy, halogenated alkoxy, hydroxyalkyl, pyrollyl. optionally substituted aryloxy, alkylamino, dialkylamino, carbamyl. amido, alkylamido dialkylamido, acylamino, alkylsulfonylamido, trihulomethoxy, trihalocarbon, thioalkyl, SO,, alkyl, COO-alkyl, NH, OH, or CN or part of a ring formed by R; and
X, represents one more substituents on the aryl group, with the proviso that X, represents at least one substituent in the 5'-position said substituent in the 5'-position being selected from the same choices as X,. C, to C6alkyl or alkoxy; or wherein X, has the formula -O-(CH,),,-0-. wherein n is 1 or 2, and one of the oxygens is bonded at the 5'-position of the aryl ring and the other is bonded to the 4' position.
The ride-side aryl group may be phenyl, or may include one or more hetcroatoms. For example, the right-side aryl group may be a nitrogen-containing aromatic heterocycle such as pyrimidine.
In specific embodiments of the composition of the invention, the right-side aryl group is substituted at the 2' and 5' position only. In other embodiment, the right side aryl group is substituted at the 2', 4'. and 5’ positions. In yet other embodiments, the right side aryl group is substituted at the 4' and 5' positions only. As will be appreciated by persons skilled in the art. the numbering is based on the structure as drawn, and variations in the structure such as the insertion of a heteroatom may alter the numbering for purposes of formal nomenclature.
In other specific embodiments of the composition of the invention, the right side aryl group has a substituent at the 2'- position and X, has the formula -X-Y-Z- with X and Z connected at the 4’ and 5' positions to the right side aryl, wherein X, Y and Z arc independently C. N, S or O, connected by single or double bonds and with appropriate hydrogen, alkyl or other substitution to satisfy valence. In some embodiments, at least one of X, Y and Z is a carbon atom. In one specific embodiment. X, is -O-(CH,),,-0-. wherein n is I
-72017200078 06 Jan 2017 or 2, and one of the oxygen atoms is bonded at the 5'-position of the aryl ring and the other at the 4' position. Additional examples of compounds of this type are shown in the Fig 4.
In accordance with specific embodiments of the invention, the purine scaffold composition has a formula as shown in Fig 5.
The composition of the invention may also be a homodimer or heterodimer of these compounds having the formula:
Figure AU2017200078B2_D0002
provided that the compound retains the ability to inhibit hsp90 and also to cross the blood brain barrier.
Embodiments of the invention are defined by each of the accompanying claims. For example, in one embodiment, the invention relates to a compound as defined by claim 1.
Brief Description of the Drawings
Fig. 1A shows tau phosphorylation activity in mouse brain following short term administration of PU24FC1.
Fig. IB shows concentration of PU24FC1 in mouse brain following short term administration.
1001572712
-82017200078 06 Jan 2017
Fig. 2 shows the effects of long-term Hsp90 inhibition with PU24FCI on tau phosphorylation and expression of other proteins.
Fia. 3 shows the effects of long-term Hsp90 inhibition with PU-DZ8 on tan phosphorylation.
Fig. 4 shows compounds useful in the method of the invention.
Fig. 5 shows compounds useful in the method of the invention.
Fia. 6 shows a synthetic scheme for making compounds useful in the invention.
Fig. 7 shows a synthetic scheme for making compounds useful in the invention.
Figs. 8A and B shows levels of various proteins in the brains of mice treated in accordance with the invention by intraperitoneal administration of a purine scaffold compound.
Fig. 9 shows degradation of the mutant protein, mtau (HT7) after one dose administration of PU-DZ8. It also shows the change in chaperone levels (hsp70 increase) and kinase expression (p35 levels).
Fig. 10 shows the dependency of mutant tau protein on hsp90 chaperoning.
Figs. 11 A and B show hsp90 binding and hsp70 induction by purine scaffold compounds in neuroblastoma cells.
Figs. 12 shows the binding affinity of PU-DZ8, PU24FCI and 17AAG to hsp90 in JNPL3 brain extracts.
Fig. 13 shows that PH-DZ8 reaches pharmacologically relevant concentrations in JNPL3 transgenic mouse brain following administration of one dose of 75 mg/kg PU-DZ8 administered i.p .
Fig. 14A shows the effects of one dose, short term administration of PU-DZ8 on the levels of soluble mutant tau in the JNPL3 mouse brain The subcortical brain region of 2.5 to 4-month old mice is presented. Human Tau levels were normalized to those of Hsp90.
Fig. 14B shows the effect of one dose, short-term administration of PU-DZ8 on the levels of insoluble mutant tau in the JNPL3 mouse brain. Analysis of the insoluble tau (P3) fractions extracted from the subcortical brain region of 6-month old mice treated with PU-DZ8 (75 mg/kg) for 4, 8. 12 and 24h is presented.
Fig. 15 shows the effect of long term PU-DZ8 administration on hyperphosphorylaicd tau in toxic tau aggregates.
-92017200078 06 Jan 2017
Fig. 16A shows the effect of PU-DZ8 on p35 in the htau mice that express pathogenically hyperphosphorylated WT tau similarly to Alzheimer's patients
Fig. 16B shows the effect of PU-DZ8 tau phosphorylation in the htau mice that express pathogenically hyperphosphorylated WT tau similarly to Alzheimer's patients
Detailed Description of the Invention
Reference to any prior art in the specification is not, and should not be taken as, an acknowledgment, or any form of suggestion, that this prior art forms part of the common general knowledge in Australia or any other jurisdiction or that this prior art could reasonably be expected to be ascertained, understood and regarded as relevant by a person skilled in the art.
As used herein, except where the context requires otherwise, the term comprise and variations of the term, such as comprising, comprises and comprised, are not intended to exclude other additives, components, integers or steps.
The present invention provides a method for treatment of neurodegenerative disease, comprising the step of administering to an individual in need of such treatment a therapeutically effective amount of a purine scaffold compound that inhibits Hsp90 and that crosses the bloodbrain barrier or is otherwise delivered to the brain.
As used in this application, the term treatment refers to delaying the onset of symptoms, reducing the severity or delaying the symptomatic progression of neurodegenerative disease in the individual. A cure of the disease is not required to fall within the scope of treatment. Further, it will be appreciated that the specific results of these treatment goals will vary from individual to individual, and that some individuals may obtain greater or lesser benefits than the statistical average for a representative population. Thus, treatment refers to administration of composition to an individual in need, with the expectation that they will obtain a therapeutic benefit.
The term neurodegenerative disease refers to disease characterized by abnormalities in signaling pathways, for example aberrant phosphorylation due to dysregulated kinase activity, mutant proteins (mutant tau, mutant APP) and chaperone unbalance leading to misfolding and
1001572712
2017200078 06 Jan 2017
-9Aincreased apoptosis. In a specific embodiment, the neurodegenerative disease is a tauopathy, i.e. neurodegenerative disease characterized by tau protein abnormalities that share the feature of hyper-phosphorylated tau protein, and intracellular neurofibrillary tangle (NFT) formation. Without limitation, the term neurodegnerative diesease as used in this application refers to and encompasses Alcohol-induced neurodegeneration (10)): Alzheimer's disease (11): Amyotrophic lateral sclerosis (13: 14 ); Brain ischemia (15: 20): Cocaine addiction (21): Diffuse Lewy body disease (22): Electroconvulsive seizures (23 ): Fetal alcohol syndrome (10); Focal cortical dysplasia (24): Hereditary canine spinal muscular atrophy (25): Inclusion body myositis (26); Multiple system atrophy (27: 28): Niemann-Pick type C; Parkinson's disease (22); and Peripheral nerve injury (71).
1001572712
- 102017200078 06 Jan 2017
The term administering refers to the act of introducing into the individual the therapeutic compound. In general, any route of administration can be used. Because the compounds used in the method of the invention may be capable of crossing the blood-brain barrier, systemic administration can be used. Thus, in certain embodiment of the invention, administration by oral, intravenous, intramuscular or parenteral injection is appropriate. Administration may also be done to the brain by inhalation because there is a compartment at the upper side of the nose that connects with the brain without having the BBB capillaries. Compounds that cross the blood brain barrier are preferred for this mode of administration as well, although this characteristics is not strictly required.
The term therapeutically effective amount encompasses both the amount of the compound administered and the schedule of administration that on a statistical basis obtains the result of preventing, reducing the severity or delaying the progression of neurodegenerative disease in the individual. As will be appreciated, preferred amounts will vary from compound to compound in order to balance toxicity/tolerancc with therapeutic efficacy and the mode of administration. Determination of maximum tolerated dose and of the treatment regime in terms of number and frequency of dosing is a routine part of early clinical evaluation of a compound.
The term crosses the blood brain barrier as used herein refers to the ability of the compound to transit to the brain in detectable amounts following systemic administration.
The ability of a compound to cross the blood brain barrier can be assessed using animal models such as mice As illustrated in the examples below, a single dose administration, for example at 50 to 200 mg/kg, can be employed, with animals sacrificed at intervals and the brain concentration of the compound determined. Il will be appreciated that the extent to which a compound does transit to the brain will also have an impact on the amount of the therapeutic compound that is necessary. In general, however, compounds that cross the blood brain barrier will have molecular weights of less than 400 daltons, a degree of lipid solubility, preferably comparable to the compounds disclosed herein, the absence of restrictive plasma protein bindings and the absence of significant affinity for any of the several BBB active efflux transporters such as p-glycoprotein. In this regard, it is noted that 17-AAG docs not effectively cross the blood brain barrier and is a P-glycoprotein substrate.
- ii 2017200078 06 Jan 2017
The therapeutic compound employed in the method of the present invention is suitably a small molecule purine scaffold compounds that inhibit Hsp90 and that possess the ability to cross the blood-brain barrier. The term purine scaffold compound refers to a compound that has a purine moiety that to which is bonded an additional aryl or hctcroaryl ring at the 8- or 9-position, wherein the compound as a whole possesses the necessary flexibility and substituent groups to be received within the N-terminal pocket of Hsp90.
These general requirements are discussed in PCT Publication No. W002/36075.
In one embodiment, the method of the invention makes use of a small molecule purine scaffold compound has the general structure:
Figure AU2017200078B2_D0003
R 1-1 right side, aryl left side, adenine wherein R is hydrogen, a C, to Cl0 alkyl, alkenyl, alkynyl, or an alkoxyalkyl group, optionally including heteroatoms such as N or O, optionally connected to the 2'-position to form an 8 io 10 member ring,
Y, and Y, are independently C. N, S or O, with the proviso that when Y, and/or Y, is O the double bonds are missing or rearranged to retain the aryl nature of the ring X4is hydrogen, halogen, for example F or Cl. or Br;
X, isCH,, CF, S, SO, SO,, Ο, NH. or NR2, wherein R2is alkyl; and
X, is halogen, alkyl, halogenated alkyl, alkoxy, halogenated alkoxy, hydroxyalkyl, pyrollyl, optionally substituted aryloxy, alkylamino, dialylamino, carbamyl, amido, alkylamido
- 12 2017200078 06 Jan 2017 dialkylamido, acylamino, alkylsulfonylamido, trihalomethoxy, trihalocarbon, thioalkyl, SO2. alkyl, COO-alkyl. NH2 OH, or CN or pan of a ring formed by R; and
X, represents one more substituents on the aryl group, with the proviso that X, represents at least one substituent in the 5'-position said substituent in the 5'-position being selected from the same choices as X:. C, to C6alkyl or alkoxy; or wherein X, has the formula -O-(CH2)„-O-. wherein n is 1 or 2, and one of the oxygens is bonded at the 5'-position of the aryl ring and the other is bonded to the 4' position.
The ride-side aryl group may be phenyl, or may include one or more hclcroatoms. For example, the right-side aryl group may be a nitrogen-containing aromatic hctcrocyclc such as pyrimidine.
In specific embodiments of the composition of the invention, the right-side aryl group is substituted at the 2' and 5’ position only. In other embodiment, the right side aryl group is substituted at the 2’, 4’, and 5' positions. In yet other embodiments, the right side aryl group is substituted at the 4’ and 5' positions only. As will be appreciated by persons skilled in the art, the numbering is based on the structure as drawn, and variations in the structure such as the insertion of a heteroatom may alter the numbering for purposes of formal nomenclature.
In other specific embodiments of the composition of the invention, the right side aryl group has a substituent at the 2'- position and X, has the formula -X-Y-Z- with X and Z connected at the 4' and 5' positions to the right side aryl, wherein X, Y and Z are independently C. N, S or O, connected by single or double bonds and with appropriate hydrogen, alkyl or other substitution to satisfy valence. In some embodiments, al least one of X, Y and Z is a carbon atom. Y in -X-Y-Z may also he -(CH2)2 such that the X-Y-Z group forms a six-membered ring. In one specific embodiment, X, is -O-(CH2)„-O-, wherein n is I or 2 from 0 to 2, and one of the oxygen atoms is bonded at the 5'-position of the aryl ring and the other at the 4' position. Additional examples of compounds of this type arc shown in the Fig. 4.
In specific embodiments of the invention, R is 3-isopropylaminopiopyl, 3-(isopropyl(methyIlamino)propyl, 3-(isopropyl(ethyl)amino)propyl, 3-((2-hydroxyethyl)(isopropyl)amino)piOpyl. 3-(methyl(prop-2-ynyl)amino)propyl. 3-(allyl(methyl)amino)propyl, 3-(ethyl(melhyl)amino)propyl. 3-(cyclopiOpyl(propyl)amino)piOpyl, 3-(cyclohexyl(2-hydroxyethyl)amino)piopyl,
- 13 2017200078 06 Jan 2017
3-(2-methylaziridin-l-yl)propyl, 3-(pi peridin-1 -yl )propyk
3-(4-(2-hydiOxyethyl)piperazin-l-yl)propyl. 3-morpholinopiOpyl,
3-(trimethylammonio)propyl, 2-(isopropylamino)ethyl, 2-(isobutylamino)ethyl, 2-(neopentylamino)ethyl, 2-(cyclopropylmethylamino)ethyl, 2-(ethyl(rnethyl)amino)cthyl. 2-(isobutyl(methyl)amino)ethyl. or 2-(methyl(prop-2-ynyl)amino)ethyl.
In accordance with specific embodiments of the invention, the purine scaffold composition has a formula as shown in Fig. 5.
The composition of the invention may also be a homodimer or heterodimcr of these compounds having the formula:
Figure AU2017200078B2_D0004
provided that the compound retains the ability to inhibit hsp90 and also to cross the blood brain barrier.
Where the active compound in vivo is the dimeric form, the compound retains the ability to inhibit hsp90 and also to cross the blood brain barrier. In this case, the linker may be any generally linear group of atoms that provides the two parts of the dimer with sufficient rotation freedom to allow both to interact independently with an N-tcrminal pocket of HSP90.
- 142017200078 06 Jan 2017
Non-limiting examples of suitable linkers include C4 to C,„ alkyl, alkenyl or alkynyl groups, and secondary amines having a total length of 4 to 10 atoms.
Compounds of this type may also be provided with a degradable or clcavablc linker, such that monomeric agents are provided in vivo. In this embodiment, the dimeric form need not retain activity or the ability to cross the blood brain barrier, and the nature of the linker therefore is not relevant to activity, only to the ability to form active monomeric species.
In general, moderately lipophilic drugs such as PUs cross the BBB by passive diffusion. A good structural understanding for the BBB permeability is still lacking, but several parameters are believed to facilitate such behavior. Lipophilicity was the first of the descriptors to be identified as important for CNS penetration. For several classes of CNS active substances, Hansch and Leo (89) found that blood-brain barrier penetration is optimal when the LogP values are in the range of 1.5-2.7, with the mean value of 2.1. The mean value for ClogP for the marketed CNS drugs is 2.5. PU-DZ8 has a calculated logP value of 1.73 (using Molinspiration) and an experimentally determined value of 1.53 (using RP-HPLC). CNS drugs have significantly reduced molecular weights (MW) compared with other therapeutics. The rules for molecular weight in CNS drugs have been reviewed, where small molecules may undergo significant passive lipid-mediated transport through the blood brain barrier, when the molecular mass is kept in or below a 400- to 600-Da range (90). PU-DZ8 has a MW of 512. All the QSAR equations emphasize the importance of hydrogen bonding CNS penetration requires 5 or less hydrogen bond acceptors (91). PU-DZ8 has 4. PSA has been shown to be a very good descriptor characterizing drug absoiption, including intestinal absorption, bioavuilabi 1 ity, Caco-2 permeability and BBB penetration. PSA has been used as a predictor for BBB penetration by many investigators (92). In general, drugs aimed al the CNS tend to have lower polar surface areas than other classes (93,94). PSA for CNS drugs is significantly less than for other therapeutics with PSA for CNS penetration estimated at 60-70 A2 through 90 A2 (95,96). The upper limit for PSA for a molecule to penetrate the brain is around 90 A2, DZ8 has a PSA of 104 A2. Changing the nature of the chain attached to the 9N position from a secondary to a tertiary amine drops the PSA to 90 A2. Number of rotatable bonds has been shown to be a very good descriptor of oral bioavailability of drugs (97-99). It is suggested that compounds which meet only the two criteria of (1) 10 or fewer rotatable bonds and (2) polar surface area equal to or less than 140 A (or 12 or fewer H-bond donors
- 15 2017200078 06 Jan 2017 and acceptors) will have a high probability of good oral bioavailability in the rat (99). Many CNS drugs are basic and exist in equilibrium between their charged and neutral states under physiological conditions or are amphiphilic if they also possess an acidic group. Possession of a positive charge at pH 7-8 tends to favor brain permeation (100). Additionally, compounds possessing a tertiary nitrogen (a feature of many CNS drugs) show a higher degree of brain permeation. All these characteristics are modeled into purine scaffold compounds as described herein.
Another characteristic which is indicative of the ability to cross the hlood brain barrier is protein binding. Drug-protein interaction is a reversible process and a successful CNS drug should not be an efficient P-glycoprotein substrate (in vivo) (102). It is not sufficient for a potential neurotherapeutic agent to move across the BBB-it also has to stay in the brain long enough to exert its desired action. This means that it also has to avoid being a substrate for a variety of transport proteins that work to extrude compounds from the brain. The Hsp90 inhibitor 17AAG is a P-gp substrate, however the purine scaffold therapeutic PU-DZ8 is not a substrate of P-pg and thus is not readily extruded from the brain by this mechanism.
Synthetic methods for making compounds useful in the method of the invention are described in PCT Publication No. W002/36075. PCT Application No. PCT/US06/03676 and US Patent Publications 2005-0113339, 2005-0004026, 2005-0049263, 2005-0256183, 20050119292, 2005-0113340 and 2005-0107343. Figs 6 and 7 shows synthetic schemes for making compounds with the structures as shown in Fig. 4. In the case of a carbon linker, phenylacetic acids are first generated by replacing the methylenedioxy bridge with the metabolically stable isosters depicted in Fig. 6. Synthesis commences by coupling 2,4.5,6-tetraaminopyrimidine with the acid fluoride of the corresponding carboxylic acid. The acid fluoride is generated by treating the phenylacetic acid with cyanuric fluoride and pyridine in CH;CL Following a quick water wash, the resulted acid fluoride is used in the next step without further purification. The amide resulted from the pyrimidine -acid fluoride coupling is cyclized to by heating in alcoholic NaOMe. Transformation of the C2-amino group to fluorine (NH; to F) is conducted by a modified Schiemann diazoiizalion-fluorodcdiaz.oniation of the amino derivative in HF/pyridine in the presence of NuNCF. We and others have previously determined that fluorine in this position in general augmented the potency of the resulting purines, likely by increasing the hydrogen donor ability of C6 NH2. Further
- 162017200078 06 Jan 2017 selective halogenation using either NIS or NBS leads to the corresponding iodo- or bromoderivatives. These are alkylated first with 1,3-dibromopropane or 1,2-dibromobulanc in the presence of Cs-CO,. Formation of dimer is not detected in this reaction. The resulted bromine is further alkylated in the presence of excess R,R2NH to give the final product.
For derivatives containing a sulfur linker, synthesis is earned out using a method previously described by He et al (1) and employs the copper catalyzed coupling of 8-mercaptoadcnine with the aryliodide (Fig. 7). The reaction occurs in anhydrous DMF at 110 °C under nitrogen. The 8-arylsulfanyl adenine is further iodinated selectively at position 2 of the aryl moiety using NIS as a source of electrophylic iodine and TFA as a catalyst. This is further alkylated at 9N in the presence of excess R,R2NH to give the final product.
Application of the Invention to Tauopathies
Alzheimer’s disease (AD) is the most common neurodegenerative disorder characterized by the progressive deterioration of cognition and memory in association with the presence of senile plaques, neurofibrillary tangles, and massive loss of neurons, primarily in the cerebral cortex and hippocampus. Senile plaques are extracellular deposits composed of β-amyloid (Αβ) fibrils, surrounded by dystrophic neurites, reactive microglia and astrocytes. Filamentous Tau inclusions are increasingly recognized as the hallmark of tauopathies, a growing family of neurodegenerative diseases including AD, Down’s syndrome (DS), several variants of prion diseases, progressive supranuclear palsy (PSP), amyotrophic lateral sclerosis/ parkinsonism-dementia complex of Guam (ALS/PDC), sporadic frontotemporal dementia with parkinsonism(FTDP), Pick's disease and familial FTDP-17 syndromes. Tau is a critical component of the neuronal cytoskclcton. Some of the moiphological changes associated with neuronal apoptosis involve a significant modification of the cytoskeletal network, likely to contribute to the subsequent degeneration of neurons, indicating disruption of cytoskeletal network can cause neurodegencration. In axons, tau protein is one of the predominant microtubule associated proteins (30). It stabilizes microtubules and promotes neurite outgrowth. This apparently beneficial role of tau contrasts with its anomalous behavior in several neurodegenerative diseases, most prominently AD. where it occurs in a highly phosphorylated form, detaches from microtubules, and aggregates. Pathogenic tau mutations or abnormal tau phosphorylation (which occurs in AD and
- 17 2017200078 06 Jan 2017 frontotemporal dementias) result in a more rapid development of NFTs and neurologic disease, a feature consistent with the view that these diseases result from tau aggregation (31).
Several mutations in human tau isoforms on chromosome 17 result in a cluster of neurodegenerative diseases, termed frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) and are characterized by the accumulation of neurofibrillary tansies similar to those in AD, in affected brain regions. Biochemical studies of these tau mutants reveal that they are less stable than normal tau and tend to form fibrillar aggregates (32). consistent with the view that tauopathies are diseases related to protein folding and stability. The tau proteins in AD are not mutated, yet nevertheless comprise NFTs. In AD. lau becomes hyperphosphorylated, and it has been hypothesized that this impairs the microtubule stabilizing role of tau
Hyperphosphorylated tau is believed to misfold, undergo net dissociation from microtubules, form abnormal filamentous aggregates (paired helical filaments. PHFs) and polymerize into NFTs (33). The central role of protein misfolding in this process is illustrated by observations that the different tau mutations linked to FDTP-17 differ in their levels of phosphorylation and in their effects on microtubules (34). Wc have shown an inverse relationship between aggregated tau and the levels of heat shock protein (Hsp)70/90 in tau transgenic mice and Alzheimer’s disease brains. In various cellular models, increased levels of Hsp70 and Hsp90 promoted tau solubility and tau binding to microtubules, reduced insoluble tau and caused reduced tau phosphorylation. Conversely, lowered levels of Hsp70 and Hsp90 resulted in the opposite effects. We have also demonstrated a direct association of the chaperones with tau proteins. Our results suggested that up-rcgulation of molecular chaperones may suppress formation of neurofibrillary tangles by partitioning lau into a productive folding pathway and thereby preventing tau aggregation (12).
Hsp90 inhibitors were found to beneficially increase levels of Hsp70 chaperone in other neurodegenerative systems. Induction of chaperones, especially Hsp70 and Hsp40. was found to delay the onset or to diminish the symptoms in folding diseases (3). GM was found to activate a heat shock response and inhibit huntingtin aggregation in a cell culture model of Huntington’s disease (16). GM was reported to restore function to a defective heal shock response in scrapie-infected cells (17, 18). Auluck et al (19) reported that treatment of a fly model of Parkinson’s disease with GM fully protected against α-synuclcin toxicity. These
- 18 2017200078 06 Jan 2017 effects were seen without altering the microscopic appearance of neuronal inclusions, suggesting that chaperones detoxify” the proteins aggregates in a more subtle way than just preventing the formation of protein aggregates. Auluck also suggested that only a modest change or redistribution of chaperones might be sufficient for ncuroprotcction (19).
These effects of the Hsp90 inhibitors occur by their modulation of the HSFl-hsp90 complexes. In normal cells, the presence of misfolded or aggregated proteins triggers a complex biological response, referred to as the heat shock response (6) This involves the expression of heat shock proteins (HSPs, molecular chaperones) and of proteins involved in the ubiquitin-proteasome pathway. The evolution of such complex machinery testifies to the fact that is necessary for cells to isolate and rapidly clear unfolded proteins as soon as they appear. In unstressed cells, HSF1 forms a dynamic complex with Hsp90 (7). When protein unfolding increases, these non-native proteins compete with HSF1 for Hsp90 binding resulting in an increase in unbound HSF1 and induction of HSPs. When stress-induced synthesis of chaperones is impaired folding diseases are possible (8). As suggested by its regulation of HSF1 activity, interference with Hsp90 activity by Hsp90 inhibitors triggers a heat shock response. The activity of neuronal disease-activated kinases is regulated by Hsp90.
We have also shown that tau phosphorylation levels at pathological sites was reduced after treatment with the Hsp90 inhibitor geldanamycin (GM) in AD cellular models. Cdk5. Gsk3 and MAPK are three major kinases that can phosphorylate tau at the pathological sites. Because phosphorylation releases tau from microtubules and because tau in the PHF is highly phosphorylated, kinases have been viewed suspiciously for a possible role in pathogenesis. There is increasing evidence that CDK5 and GSK3a may be involved in the pathogenesis of several neurodegenerative disorders. In neurons that no longer divide, deregulation of Cdks, especially Cdk5, occurs in many neurological disorders, including Alzheimer’s disease (AD) and Parkinson’s disease (PD). Fath et al. has shown that replacement of certain amino acids at known sites of phosphorylation with a charged amino acid created ‘pseudohyperphosphorylated’ tau that can mimic structural and functional aspects of hyperphosphorylated tau (35). In vivo evidence for an interaction with tau exists for Cdk5 and Gsk3. Over-expression of human p25 (an activator of Cdk5) in mice induced tau hyperphosphorylation and cytoskeletal disruptions reminiscent of AD, but no filamentous
- 192017200078 06 Jan 2017 deposits (36). Noble et al. crossed transgenic mice over-expressing the Cdk5 activator p25, with transgenic mice over-expressing mutant (P301L) human tau. Tau was hyperphosphorylated at several sites in the double trunsgenics, and a highly significant accumulation of aggregated tau occurred in the brainstem and cortex. Increased numbers of silver-stained neurofibrillary tangles (NFTs) accompanied these changes as well as an association of active GSK with insoluble tau (37). Over-expression of GSK-3 under ihe control of a tetracycline sensitive transactivator also induced tau hypcrphosphorylation, somatodendritic mislocalization of tau, and neuronal apoptosis (38). Recent studies have shown that the β-amyloid peptide (Αβ) induces a deregulation of Cdk5 in cultured brain cells and raises the question on the possible roles of this tauphosphorylating protein kinase in the sequence of molecular events leading to neuronal death triggered by Αβ. In this context, then is evidence that Cdk5 is involved in tau hyperphosphorylation promoted by Αβ in its oligomeric form (42). Cdk5 inhibitors protect hippocampal neurons against both tau anomalous phosphorylations and neuronal death. The links between the studies on the Cdk5/p35 system in normal neurogenesis and its claimed participation in ncurodegeneration, provide the framework to understand the regulatory relevance of this kinase system, and changes in its regulation that may be implicated in disturbances such as those occurring in
Alzheimer disease (70). Overall these studies implicate tau hyper-phosphorylation in tau-related neurodegeneration and allude to Cdk5, Gsk3 and MAPK as major players in the process.
As demonstrated in the examples set forth below, small molecule purine scaffold compounds are able to inactive the kinases involved in tau phosphorylation and when the appropriate substitution patterns are selected are able to cross the blood brain barrier.
Further, addition of PU24FCI Hsp90 inhibitor to a panel of transformed cells led to a dosedependent induction of Hsp70 and Hsp40. This phenomenon occurred in all the tested cell lines irrespective of their tissue of provenance and was duplicated in rat cortical primary neurons. Doses of PU24FC1 and PU29FCI (another early PU-class compound) that induce a stress response were not toxic against normal cells, as demonstrated in a panel of normal epithelial and fibroblast cells.
-202017200078 06 Jan 2017
Application of (he Invention to Other Neurodegenerative Diseases
Amyotrophic lateral sclerosis is a neurological disorder that selectively affects motor neurons of bruin and spinal cord. Amyotrophic lateral sclerosis (ALS) is characterized by a progressive degeneration of motor neurons that results in severe weakness and skeletal muscle atrophy. The disease is progressive and patients usually succumb to bulbar paralysis, cachexia or respiratory failure within 2-5 years of onset (44 ). A distinguishing feature of ALS is the accumulation of neurofilaments in the perikarya and axons of the spinal motor neurons (for review see Julien 2001, 45). NF-H and NF-M are substrates of CDK5, and the motor neuron inclusion bodies that occur in ALS cases contain hyperphosphoryktlcd NF-H (for review see Julien 1999,47). Emerging evidence indicates an involvement of the serine/threonine cyclin-dependenl kinase 5 (Cdk5) in the pathogenesis. Deregulation of Cdk5 by its truncated coactivators, p25 and p29, contributes to neurodegcncration by altering the phosphorylation state of cytosolic and cytoskeletal proteins and, possibly, through the induction of cell cycle regulators.
Parkinson's disease is characterized by bradykincsia in most patients and many patients may develop a resting tremor (for review see Fahn 2003, 48 ). Classic pathological findings include loss of neuromelanincontaining neurons within the substantia nigra and the presence Lewy bodies (48). The Lewy body is an eosinophilic cytoplasmic neuronal inclusion (for review see Fahn 2003, 48), and CDK5 immunoreactivity occurs in Lewy bodies in the midbrain of Parkinson's disease patients (22), In rats, induction of apoptosis in neurons of the substantia nigra resulted in increased CDK5 levels and activity at the later stages of apoptosis (49 ). Further, CDK5 and p35 immunoreactivity was observed in the perikaryon and nuclei of apoptotic neurons, whereas immunoreactivity in healthy neurons was confined to the axons (49 ).
Other kinases that are also deregulated in PD, and for which pathogenic mutations have been identified in sporadic PD patients are strong candidates as HSP90 clients. These include leucine-rich repeat kinase-2 (LRRK2) gene were pathogenic mutations cause autosomal-dominant and certain cases of sporadic Parkinson disease. The G2019S substitution in LRRK2 is the most common genetic determinant of Parkinson disease identified so far, and maps to a specific region of the kinase domain called the activation segment. Here we show that autophosphorylation of LRRK2 is an intcrmolecular reaction and
- 21 2017200078 06 Jan 2017 targets two residues within the activation segment. The prominent pathogenic G20I9S mutation in LRRK2 results in altered autophosphorylation, and increased autophosphorylation and substrate phosphorylation, through a process that seems to involve reorganization of the activation segment. Another mutant kinase in the PTEN induced putative kinase 1 (PINK1) gene. These mutations were originally discovered in three pedigrees with recessively inherited PD. Two homozygous PINK1 mutations were initially identified: a truncating nonsense mutation (W437X) and a G309D missense mutation. Subsequently, multiple additional types of PD-linked mutations or truncations in PINK 1 have been reported, making PINK1 the second most common causative gene of recessive PD. Interestingly, despite autosomal recessive transmission of PINKl-linked early-onsct PD. a number of heterozygous mutations affecting only one PINK1 allele have been associated with late-onsei PD. The pathogenic mechanisms by which PINK1 mutations lead to neurodegeneration are unknown.
P1NK1 encodes a 581-amino-acid protein with a predicted N-tcrminal mitochondrial targeting sequence and a conserved serine/threonine kinase domain. P1NK1 protein has been shown to localize in the mitochondria and exhibit autophosphorylation activity in vitro. The in vivo substrate(s) and biochemical function of P1NK1 remain unknown. In cultured mammalian cells, overexpression of wild-type P1NKI protects cells against apoptolic stimuli . whereas small interfering RNA (siRNA)-mediated depletion of P1NK1 increases the susceptibility to apoptotic cell death. In Drosophila, loss of PINK1 leads to mitochondrial defects and degeneration of muscle and dopaminergic neurons. Despite ample evidence indicating an essential role of P1NK1 in cytoprotection, the mechanism by which PINK I protects against apoptosis is not understood.
Our results showed that at least Cdk5 and P35 are client proteins of Hsp90. Inhibition of Hsp90 could decrease Cdk5/P35 protein level in vitro and P35 level in vivo. Since accumulated evidence implicate that Cdk5/P35 is related to those neurodegenerative diseases. Hsp90 inhibitor can also be used in the treatment of those diseases.
The invention will now be further described with reference to the following, nonlimiting examples.
2017200078 06 Jan 2017
- 22 Example I
Juvenile mice: Four- to six-week old nu/nu athymic female mice were obtained from the National Cancer Institute-Frederick Cancer Center and maintained in ventilated caging. Experiments were carried out under an Institutional Animal Care and Use
Committee-approved protocol, and institutional guidelines for the proper and humane use of animals in research were followed. Before administration, a solution of PU24FC) was prepared at desired concentration in 50 /zL vehicle (PBS:DMSO:EtOH at 1:1:1 ratio). In experiments designed to define the short-term effects of PU24FCI on tau phosphorylation, mice (2 per time point) were treated with 200 mg/kg PU24FCI or with vehicle alone. At the time of sacrifice, brains were collected and immediately flash frozen. For protein analysis brains were homogenized in SDS lysis buffer (50 mM Tris pH 7.4, 2% SDS). For long-term administration studies, mice (n = 5) were treated every other day for 30 days with the indicated doses of PU24FC1. Weight and behavior changes were monitored for all animals. Mice were sacrificed by CO; euthanasia at 8 h post-last PU24FCI injection. Brains were collected and processed as mentioned above. Proteins were further analyzed by Western blot.
Phosphorylation of tau in juvenile and embryonic brains is enhanced (50) and similar to AD afflicted brain (5T. 52). Further, nude athymic mice 4-6 weeks of age may express tau phosphorylated at relevant disease epitopes In a first in vivo experiment, the short term modulation of Hsp90 in the brains of these animals was evaluated. One dose of PU24FCI (200 mg/kg) was administered intraperitoneally to these mice and animals were sacrificed at 0, 6, 12, 24, 36 and 48 hours. Whole brains were homogenized in lysing buffer and lau phosphorylation at S202/T205 was evaluated by Western blot. A burst in tau phosphorylation at this epitope was observed 12 h post-administration, with a decline to basal levels shortly after (Fig. 1 A). Drug levels in the brain tissue were analyzed by LC-MS and showed the presence in brain tissue at therapeutically relevant levels with a spike at around 24 hours (Fig. IB). In these same mice, PU24FC1 was quickly cleared from the liver, scrum and uterus.
In a second experiment, we analyzed the effects of long-term Hsp90 inhibition on tau phosphorylation Mice were treated on alternate days for 30 days with PU24FCI without observing remarkable toxicity or weight loss in these animals. As seen in Fig. 2, a significant decrease in tau phosphorylation at S202/T205 was evident in all treated mice. Such difference in effects between short and long term modulation of Hsp90 has been documented for other
- 23 2017200078 06 Jan 2017 proteins chaperoned by Hsp90. Treatment of cells with Hsp90 inhibitors caused degradation of Raf-1 over a long time course, while inducing a transient burst of Raf-1 activity when administered for a short time (53). Similar evidence has been demonstrated for the activity of the RNAdependent kinase PK.R, which becomes active upon short treatment with GM (54). These observations suggest that Hsp90 may act to restrain the basal signaling of these kinases. Additional examples are found in the regulation of steroid hormone receptors. Hsp90 masks dimerization and inhibits DNA binding of steroid hormone receptors until chaperone interactions are interrupted, typically as a consequence of hormone binding. Thus, steroid hormone receptors stripped from chaperones are competent for dimerization and DNA binding in the absence of hormone (55). While this function of Hsp90 may not hold true for all its client proteins, in the case of p35/cdk5, Hsp90 may undertake a similar role restraining the intrinsic activity of the complex, while retaining it in a primed conformation, ready for interaction with tau.
Reduction in tau phosphorylation in the long-term treatment experiment was associated with a 60 to 70% decrease in p35 expression (Fig. 2). In addition, an increase in the expression of the inducible Hsp70 was observed in these mice (Fig. 2). Expression of cdk5 in the whole brain was not affected. The cdk5 protein is widely distributed in mammalian tissues and in cultured cell lines and is complexed with an array of other proteins, with each association serving a diverse cellular role. The cdk5/p35 associated kinase activity has been demonstrated only in the cerebral cortex (56. 57). When immunoprecipilaicd cdk5 activity was examined in AD brains it was found to be elevated in the prefrontal cortex (58). The limited localization of p35/cdk5 in the cortex may explain why total cdk5 expression in the whole brain was unchanged upon Hsp90 inhibition. Very likely, the high background caused by cdk5 localized to other compartments made impossible monitoring a small change in cdk5 steady-states by Western blot. These results may also suggest that management of cdk5 by Hsp90 in the brain is likely limited to regulating the activity of the p35/cdk5 complex.
Example 2
Transgenic mice: Transgenic mice, JNPL3 line (59) overexpressing niulanl human tau (P301L. 4R0N) were used in this study. Mice were heterozygous and on a mixed hybrid
- 24 2017200078 06 Jan 2017 senetic background composed of C57BL/DBA2/SW, as published in ref. 59. These mice develop NFTs in the basal telencephalon, diencephalon, brainstem, and spinal cord, with severe patholosy accompanied by degeneration in the spinal cord leading to dystonia, paralysis, and death in mice >12 months in age. Nine month-old male JNPL3 mice (n = 2) were treated intraperitoneally with PU-DZ8 or vehicle for 5 days. Mice were sacrificed 12 h after last treatment by cervical dislocation under anesthesia.
To further examine the effect of Hsp90 on tau phosphorylation, we used the JNPL3 line of mice expressing mutant (P301L) tau protein (59). Genetic analyses have linked mutations in the tau gene to FTDP-17 (60, 61). Over 20 distinct pathogenic mutations have been identified, with P301L as the most common mutation in tauophaties (33). JNPL3 mice exhibit an age and gene-dose-dependent increase in tau phosphorylation and development of NFTs (59, 62 ). The tau protein in JNPL3 is predominantly human and is phosphorylatcd at multiple sites: T181 (AT270). S202/T205 (AT8), T2I2 (AT100),T231 (AT 180), S262. S396/S404, S409 and S422 (59, 62). In concordance with the experiments in the juvenile nude athymic mice, a five day treatment of nine-month old male JNPL3 mice with PU-DZ8, a water soluble PU24FC1 derivative (2), reduced p35 levels in whole brains and led to a significant amelioration of tau phosphorylation at the putative cdk5 sites, S2O2/T2O5 and T212. The degree of p35 expression translated well into alleviation of phosphorylation. A 50% reduction in p35 levels translated in approximately similar effect on S202/T205 (Ab AT-8). while reducing phosphorylation on T212/S214 (Ab AT-100) almost completely. No significant effect on tau phosphorylated at T231 (Ab AT-180), associated with tau in PHF and tangles (63, 64) was seen at a reduction by 50% in p35 expression. However, in mice where effects were more prominent and p35 expression declined to approximately 2()7( as compared to control, a significant effect on tau phosphorylation at S202/T205 and T2I2/S2I4 and a 50% reduction on T231 was observed. We could not detect a significant amount of tau phosphorylation at T181, site found to be hyperphosphorylatcd in PHF, tangles and neurofilaments (65). Again, whole brain expression of cdk5 was not affected (Fig. 3). Pharmacologically relevant levels of PU-DZ8 were recorded in these brains.
- 25 2017200078 06 Jan 2017
Example 3
JNPL3 female mice 6.5 months of age were treated for 30 days. 5day/wcek, with the Hsp90 inhibitor PU-DZ8 (Fig. 5) or vehicle, or sacrificed for time zero. n=4/group. Brains were divided in subcortical and cortical regions and processed using the Greenberg and Davies extraction protocol. (77) Sarkosyl soluble fractions (SI) were analyzed by WB for p35 and Hsp70. and for tau epitopes found abnormally hyperphosphorylated in AD brains such as: S202 and T205 recognized by ATS, T181 by AT270, T231 by AT 180. These are putative cdk5/p35 sites. Protein bands were normalized to Hsp90 and plotted as relative units The results are shown in Fig. 8A and B. Since tauopathy, characterized by pathogenic phosphorylation of tau can be due to aberrant kinase activity, the hsp90 inhibitor is effective because it affects the expression of the p35 protein, an activator of cdk5 known to phosphorylate tau at pathogenic sites, and thus alleviates tau phosphorylation at these sites.
Example 4
JNPL3 female mice 6months of age were treated IP with the Hsp90 inhibitor PU-DZ8 (75mg/kg) and sacrificed various times as indicated in Fig. 9. Brains were divided in subcortical and cortical regions and processed using the Greenberg and Davies extraction protocol (77). Sarkosyl soluble fractions (S1) extracted from the subcortical region were analyzed by WB for p35, cdk5, mutant tau (HT7), Hsp90 and Hsp70. Protein bands were normalized to actin and plotted as relative change from untreated mice. Fig. 9 shows degradation of the mutant protein, mtau (HT7) after one dose administration of DZ8. It also shows the change in chaperone levels (hsp70 increase) and kinase expression (p35 levels).
Example 5
COS-7 cells were transfected with cDNAs corresponding to WT and mTau and cells were further treated with PU24FCI for 24h. Cells were lysed and protein content analyzed by Western blot. The results are shown in Fig. 10. As shown, the mutant Tau (P301L) is very sensitive to the Hsp90 inhibitor PU24FCI, while the WT tau is unaffected by similar doses ol dru a.
-262017200078 06 Jan 2017
Example 6
The ability of composition according to the inventions Hsp90 inhibitors to bind Hsp90 was tested using a fluorescence polarization assay developed by Chiosis et al (W020050124S2.66, 67, 68). SK-N-SH neuroblastoma cells were treated with Hsp90 inhibitors for 24 h and Hsp70 levels were detected by a phenotypic cell-based assay developed by Chiosis et al ( W02005012482,69). The results are summarized in Figs. 11 A and B. As shown, the inhibitors induce a stress response in the SK-N-SH neuroblastoma cells and Hsp70 induction by Hsp90 inhibitors correlates with their potency in binding to the ATP-regulatorv pocket of the Hsp90 chaperone.
Example 7
Embryonic primary rat cortical neurons and COS-7 cells transfected with cDNAs corresponding to either p35 alone (COS-7/ p35) or both p35 and Tau (COS-7/p35/Tau) arc relevant experimental systems to study aberrant neuronal kinase activity because phosphorylation of Tau at putative cdk5 sites is both enhanced in these cells and in embryonic and juvenile brains (50, 52) and is similar to that in AD-afflicted brains (50). COS-7 cells transfected with cDNAs corresponding to either human WT Tau (COS-7/Tau) or Tau harboring the P301L mutation characteristic of frontotemporal dementia and parkinsonism linked to chromosome 7 (COS-7/TauP301L) are cellular models that may be used to differentiate the effect of Hsp90 inhibition on a mutant protein compared with its normal counterpart.
To further examine the roles played by Hsp90 in tauopathy, we made use of both PU24FC1 and 17-(allyllamino)-17-demethoxygeldanamycin (17AAC) and investigated their effects on both cdk5/p35 and TauP301L in primary neuronal and COS-7 cell cultures.
Primary neuronal cultures were derived from the cerebral cortices of embryonic day 17 rat embryos and maintained as described previously (105). To determine the effects of PU24FCI on protein steady-states and on Tau phosphorylation, PU24FCI was added at day 6 of culture, and cells were incubated at 37°C as indicated. COS-7 cells grown inDMEM with 10% FBS and penicillin/ streptomycin (50 units and 50 gg/ml. respectively) were transiently transfected by using FuGENE 6 reagent (Roche Molecular Biochemicals, Indianapolis, IN) to overexpress p35 and either WT Tau or Tau harboring a P301L mutation. At 12 h al ter
- 27 2017200078 06 Jan 2017 transfection, cells were incubated for 24 h with the indicated concentration of PU24FCI. After incubation, cells were harvested and lysed in 2% SDS, and the resulting samples were analyzed by Western blotting.
Phosphorylation of Tau by cdk5 is initiated through activation by complex formation with one of the neuron-specific proteins p35 or p39. However, only suppression of p35 by antisense oligonucleotide treatment but not of the highly related isoform p39 selectively reduces cdk5 activity. In addition, levels of p35 but not of cdk5 protein are rate-limiting for cdk5 activity. In concordance, we assessed the influence of Hsp90 inhibition on p35 cellular expression. A dose- and time-dependent degradation of p35 by PU24FCI was detected in primary neurons by immunoblot and by immunofluorescence techniques, as well as in COS-7/p35 and COS-7/p35/Tau cells. Effects were seen at ~l-5 μΜ PU24FCI and were maximal at 10 μΜ Hsp90 inhibitor, in agreement with the affinity of this compound for Hsp90. Exogenously introduced p35 was more sensitive to Hsp90 inhibition than the endogenous protein, suggesting that by analogy to Hsp90 oncoproteins, buffering and stabilization of aberrant proteins in tauopathy may be accomplished by co-opting Hsp90. Reduction of p35 levels by Hsp90 inhibition affected the activity of the cdk5/p35 complex, as measured by using a substrate of cdk5, the histone-Hl, and lessened Tau phosphorylation at putative cdk5 shown to be phosphorylated in AD brains without affecting normal Tau protein expression. mTau however, was sensitive to concentrations of PU24FCI that did not interfere with WT Tau expression. The higher sensitivity to Hsp90 inhibition of mTau compared with WT Tau is in agreement with the observed lability of the mutant oncoprotein clients of Hsp90. Analogous effects on p35 and mTau were observed with I7AAG. The effect of PU24FCI on neuronal proteins was well-defined and selective, us the expression of several kinases and phosphatases that regulate normal Tau activity (PKA, CK.-1, CK-2, PP-1-alpha, PP-1-gamma, and PP2A) was not affected by the Hsp90 inhibitor.
Induction of Hsp70 by Hsp90 inhibitors is documented in several neurodegenerative disease models (12, 16, 19). Expression of Hsp70 is indirectly regulated by Hsp90 (7). Accordingly, treatment of either primary neurons or transfected COS-7 cells with PU24FCI led to a dose-dependent increase in Hsp70. Induction of Hsp70 occurred at doses of PU24FCI that also modulated both p35 and mTau, suggesting that degradation of aberrant proteins and
-282017200078 06 Jan 2017 induction of a heat-shock response are both direct consequences of Hsp90 inhibition by
PU24FCI.
Example 8
To examine whether Hsp90 plays a direct role in maintaining the stability of these p35 and mTau, we tested whether inhibition of Hsp90 function by PU24FCI affected their half-life. Primary neuronal cultures were treated with inhibitor or vehicle in the presence of cycloheximide. Quantification of protein levels demonstrated that the half-life of endogenous p35 was 120 min in the presence of vehicle and decreased to 60 min when PU24FCI was added to the system. The exogenous p35 was more labile and had a significantly shorter half-life than the endogenous protein (t1/2 = 60 min in the presence of vehicle and 30 min in the presence of PU24FCI) for both COS-7/p35/ Tau cells and primary neurons. Similar results were observed for mTau: whereas 50% of the protein was degraded at 2-4 h in the presence of the Hsp90 inhibitor, the half-life of mTau in vehicle treated cells exceeded 10 h. The inhibitor had no effect on the level of WT Tau. Moreover, mTau and p35 were degraded upon PU24FCI treatment even when induction of Hsp70 was blocked by cycloheximide. These findings strongly position Hsp90 as a direct and important regulator of both p35 and mutant Tau stability.
Example 9
To examine whether Hsp90 regulates the stability of these proteins through protein complex formation, we made use of several chemical and immunological tools that selectively bind either Hsp90 or its putative client proteins. Association of Hsp90 with p35 as well as with mTau, was observed. No significant association was observed when cells were immunopurified with a control IgG. Cdc37, a cochaperone of H$p90 found associated with several chaperone-kinase assemblies, was absent in the p35-immunopurificd complexes, in concordance with previous observations of Lamphcre et al. (106). Pretreatment of cells with PU24FC1 altered the interaction of Hsp90 with p35.
The cellular models presented above demonstrate that an interaction between Hsp90 and aberrant neuronal proteins is possible at a molecular level. However, exogenous introduction of proteins by transfection, may destabilize the cell’s protein content and impose
2017200078 06 Jan 2017
- 29 a regulation of the alien protein’s stability by Hsp90. Therefore, to evaluate the interaction of Hsp90 with TauP301E and p35 in an endogenous environment, wc made use of brain homogenates obtained from animal models of tauopathy. The JNPL3 line of mice expressing mutant (P301L) human Tau (hTau) protein exhibit an age, gender and gene dosc-depcndcnt increase in Tau phosphorylation and insoluble Tau deposits. To isolate proteins associated with Hsp90 in these brains, we made use of brain homogenates obtained from female JNPL3 mice (n = 4) 10 months of age and used either a biotinylated PU derivative immobilized on streptavidin beads ora specific anti-Hsp90 antibody. Hsp90 isolated by PU beads bound mTau specifically. The presence of the C terminus of heat-shock cognate 70-intcracting protein, an ubiquitin E3 ligase found to collaborate with molecular chaperones in facilitating protein folding, was also identified in the Hsp90 complex, in agreement with findings of Sahara et al. (62). An Hsp90 antibody specifically identified the chaperone in complex with p35 and its kinase partner cdk5. Collectively, these data position Hsp90 as a regulator of p35 and mTau stability through direct protein complex formation.
Example 10
Binding to JNPL3 brain Hsp90. The assay buffer (HFB) contained 20 mM HEPES (K) pH 7.3, 50 mM KCI, 5 mM MgCE, 20 mM Na2MoO4, 0.019?' NP40. Before each use. 0.1 mg/mL bovine gamma globulin (BGG) (Panvera Corporation, Madison, WI) and 2 mM DTT (Fisher Biotech, Fair Lawn, NJ) were freshly added. GM-cy3B, a specific Hsp90 ligand, was synthesized as previously reported (10) and was dissolved in DMSO to form 10 μΜ solutions. Brains were homogenized in HFB with added protease and phosphatase inhibitors. Saturation curves were recorded in which GM-cy3B (3 nM) was treated with increasing amounts of brain homogenates. The Hill and Scatchard plot analyses of the experiment were constructed to show that at the low amounts of brain homogenates required to reach saturation, interaction from other cellular material was precluded. The amount of brain homogenate for which over 90% of GM-cy3B was Hsp90 bound at equilibrium (24 h) was chosen for the competition study. For the competition experiments, each 96-wcll contained 3 nM GM-cy3B. brain homogenate and tested inhibitor (initial stock in DMSO) in a final volume of 100 uL. The plate was left on a shaker at 4 °C for 24 h and the fluorescence polarization values in mP were recorded. ECS0 values were determined as the competitor
-302017200078 06 Jan 2017 concentrations at which 50% of GM-cv3B was displaced. Fluorescence polarization measurements were performed on an Analyst GT instrument (Molecular Devices, Sunnyvale, CA). For GM-cy3B, an excitation filter at 545 nm and an emission filter at 610 to 675 nm were used with a dichroic mirror of 565 nm. Measurements were taken in black 96-wcll microtiter plates.
Figs. 12 shows the binding affinity of PU-DZ8, PU24FCI and 17AAG to hsp9O in JNPL3 brain extracts determined using this procedure. As shown, the EC5() for PU-DZ8 is lower than that of the other compounds. (46.71 nM, as opposed to 822.6 nM for PU24FCI and 98.40 nM for 17AAG).
The same procedure was repeated using the compounds of Fig. 5. The EC5ll values determined for these compounds are set forth in Table 1. Hsp70 induction in neuroblastoma cells by the various purine scaffold compounds was determined. The potency for hsp70 induction corresponds to the hsp90 binding affinity.
ΓΟ
CM
-31 Tabic 1
Η-S
Ό
Ο
Compound EC50 JNPL3 brain Hsp90 binding (nM)
PU-H71 (multiple measurements) 15.2, 30.8
PU-DZ8 (multiple measurements) 85.3, 40.1
PU-HZ150 5.7
PU-HZ151 6.9
PU-DZ13 8.1
PU-DZ14 8.4
PU-HT65 212.9
PU-HT64 122.4
PU-DZ10 192.8
PU-HT70 146.6
PU-HT78 9561
PU-HT133 812.9
PU-BSI12 39.3
PU-BSI8 30.2
PU-BSI6 60.8
PU-BSI11 29.9
PU-BSI7 43.5
PU-BSI13 44.5
PU-BSI14 42.8
PU-BSI5 26.8
PU-BSI10 105.4
PU-BSI3 199.4
PU-BSI15 122.2
PU-BSI16 202.4
PU-BSI4 155.2
PU-DZ12 219.0
PU-DZ16 35.6
PU-DZ15 165.6
PU-DZ17 92.3
| PU-DZ18 107.1
2017200078 06 Jan 2017
- 32 Example I 1
Assessment of PU-DZ8 brain levels. Concentrations of compound were determined and quantitated by a MRM mode using a tandem high-performance liquid chromatographymass/muss spectrometry (HPLC/MS/MS). A weighed piece of brain was rinsed with saline isotonic solution, dried with gauze and then homogenized in mobile phase (acetonitrile (ACN)/0.l% formic acid = 1.2/2.8. v/v). Haloperidol was added as internal standard. PU-DZ8 was extracted in methylene chloride, the organic layer was separated, speedily dried under vacuum and reconstituted in the mobile phase. Compound analysis was performed in the API 4000™ LC/MS/MS (Applied Biosystems) which was coupled with a Shimadzu LC system and a 96-well plate autosampler. A Gemini Cl8 column (5μ particle size. 50 x 4.6 mm I.D.) was used for the LC separation. The analyte was eluted under an isocratic condition for 4 min at a flow rate of 0.4 mL/min.
One dose of PU-DZ8 (75 mg/kg) was administered intraperitoncally (i.p.) to female mice of 2.5-4 months in age (n = 32) and animals were sacrificed in the interval of 0 to 36 h. Both aggregate-free Tau (SI) and insoluble Tau (P3) fractions were prepared from the subcortical and cortical regions of these mice. PU-DZ8 levels in the brain reached 0.35 ,ug/g (-700 nM) at 4 h, and the pharmacologically relevant dose was retained for at least 12 h post-administration (0.2 ug/g, -390 nM). The results are shown in Fig. 13.
Fig. 13 shows that PU-DZ8 reaches pharmacologically relevant concentrations in JNPL3 transgenic mouse brain following administration of one dose of 75 mg/kg PU-DZ8 administered i.p . This shows that PU-DZ8 arrives in the bruin tissue much sooner than PU24FC1 (Fig. IB).
Example 12
In a cluster of tauopathies termed frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17), transformation is caused by several mutations in human Tau isoforms on chromosome 17, that result in and are characterized by the accumulation of aggregated Tau similar to that in AD (10, 11). Over 20 distinct pathogenic mutations have been identified, with P301L as the most common mutation in tauopathies.
To investigate whether release of mTau and p35 from Hsp90 regulation restores normal neuronal activity and results in elimination of toxic Tau aggregates, wc made use of
-332017200078 06 Jan 2017 the JNPL3 mouse model of tauopathy. Brain tissues of JNPL3 mice contain Tau proteins with different solubilities and these can be separated into buffcr-extructabic (S1). high-salt extractable (S2) and sarkosyl-insoluble (P3) fractions. The S 1 fractions contain a 50-60 kDa human Tau protein, whereas sarkosyl-insoluble Tau proteins of 64 kDa and higher molecular weights are detected in the subcortical bruin regions of JNPL3 mice as early as 3 months in hemizygous females. These contain insoluble toxic Tau phosphorylated at multiple sites such as T181, S202/T205, T212 andT231 (37,38).
To investigate whether the human TauP301L present in the JNPL3 line of mice is a sensitive target for Hsp90 inhibition, animals were treated with the brain-banicr permeable Hsp90 inhibitor PU-DZ8. This agent is a higher-potency water soluble derivative of PU24FCI (EC5OJNPL3 brain Hsp90 = 70 nM). One dose of PU-DZ8 (75 mg/kg) was administered intraperitoneally (i.p.) to female mice of 2.5-4 months in age (n = 32) and animals were sacrificed in the interval of 0 to 36 h. Both aggregate-free Tau (SI) and insoluble Tau (P3) fractions were prepared from the subcortical and cortical regions of these mice. Human Tau levels were assessed by immunobloting w'ith a human specific anti-Tau antibody (HT-7). At 4 h post-administration, the Hsp90 inhibitor induced a significant decrease in the soluble precursor pool mTau as present in the subcortical brain regions (P = 0.0031 at 4 h). with these effects maintained up to 36 h (P = 0.0066 at 8 h, 0.0030 at 12 h, 0.0111 at 24 h and 0.042 at 36 h) (Fig. 14A). We next examined in a 4- to 6-month old mouse group (n = 15) w'hcthcr the stability of mTau as present in tau aggregates (P3 fraction) was additionally regulated by Hsp90. As demonstrated in Fig. 14B, a significant reduction of insoluble (P < 0.0001) and hyperphosphorylated (P = 0.001) Tau was observed in treated mice (n = 8), as compared to those mice receiving no Hsp90 inhibitor (n = 7).
No significant changes in cdk5 expression were detected, indicating that management of cdk5 by Hsp90 in the brain may be limited to regulating the activity of the p35/cdk5 complex. The expressions of Akt and Raf-l, nodal proteins in cell survival and growth pathways, respectively, tightly regulated by Hsp90 in malignant cells were not altered by PU-DZS.
For experiments designed to test the kinetics of mTau and p35 modulation by Hsp90 inhibitors, animals were administered intraperitoneally (i.p.) 75 mg/kg PU-DZ8 in PBS (6% DMSO). Mice were sacrificed by CO2 euthanasia at the indicated times following PU-DZ8
-342017200078 06 Jan 2017 administration. Hemibrains were separated into cortico-limbic (cortex, amygdale and hippocampus) and subcortical (basal ganglia, diencephalon, brain stem and cerebellum) resions, quickly frozen on dry ice and stored at -80°C and processed. In summary, each brain piece was weighed and homogenized in three volumes of Tris-buffered saline (TBS) containing protease and phosphatase inhibitors (25 mM Tris-HCI, pH 7.4, 150 mM NaCl, 1 mM EDTA, I mM EGTA, 5 mM sodium pyrophosphate, 30 mM β-glyccrophosphatc. 30 mM sodium fluoride. 1 mM phenylmethylsulfonyl fluoride (PMSF)). The homogenates were centrifuged at 27,000 g for 15 min at 4°C. Supernatants were collected as SI fractions, and the pellets (Pl) were re-homogenized in three volumes of salt/sucrose buffer (0.8 M NaCl. 10% sucrose, 10 mM Tris/HCl, pH 7.4. 1 mM EGTA, 1 mM PMSF) and centrifuged us above. The resulting pellets were discarded and the supernatants were incubated with sarkosyl (Sigma, St Louis, MO, USA; 1% final concentration) for 1 h at 37°C. The sarkosyl mixtures were then centrifuged at 150,000 g for 30 min at 4°C. The supernatants (S2 fraction) were collected, and the pellets (P3) were resuspended in 50 ,uL 2% SDS in TBS and stored at -80°C for western blotting. Proteins were analyzed by Western blot.
Fig. 14A. shows the effects of one dose, short term administration of PU-DZ8 on 1 he levels of soluble mutant tau in the JNPL3 mouse brain The subcortical brain region of 2.5 to 4-month old mice is presented. Human Tau levels were normalized to those of Hsp90. Fig. 14B shows the effect of one dose, short-term administration of PU-DZ8 on the levels of insoluble mutant tau in the JNPL3 mouse brain.Analysis of the insoluble tau (P3) fractions extracted from the subcortical brain region of 6-month old mice treated with PU-DZ8 (75 mg/kg) for 4, 8. 12 and 24h is presented
-352017200078 06 Jan 2017
Example 13
To investigate whether modulation of mTau could be sustained over a longer Hsp90 inhibitor-treatment period, without being toxic to mice, JNPL3 mice were subjected for 30 days to these agents. Female JNPL3 mice 6.5 months of age (n = 10) were administered i.p. vehicle (n = 5) or one of the Hsp90 inhibitors, PU24FC1 (200 mg/kg) or PU-DZ8 (75 mg/kg) (n = 5). on a daily, five-times per week schedule and animals were sacrificed at 8 h following the last administered dose of inhibitor. No toxicity was observed as evidenced by lack of significant change in animal weight, fur appearance, appetite and posture. Furthermore, no visible internal organ damage was detected at sacrifice upon gross inspection. Both SI and P3 fractions extracted from the subcortical brain region of these mice were analyzed for mTau expression and phosphorylation. A significant reduction in Tau expression and phosphorylation in both the precursor protein pool (SI fraction) (hTau, P < ().()001) and the toxic aggregate (P3 fraction) (phosphorylated Tau at T231, P = 0.0034) (Fig. 15). as well as p35 reduction in SI fraction was observed in mice treated with the Hsp90 inhibitor.
Collectively, the rapid kinetics of Tau degradation in both the soluble pool and the aggregated form by the Hsp90 inhibitors suggests that Hsp90 regulates the toxic Tau aggregate and facilitates its formation and accumulation. These data also suggest that an Hsp90 inhibitor may be used in the treatment of tauopathies both to prevent the formation of toxic aggregates and to solubilize the already aggregated toxic tau.
Fi?ure 15 shows the effect of long term PU-DZ8 administration on hyperphosphorylated tau in toxic tau aggregates.
Example 14
In tauopathies transformation is characterized by abnormalities in the Tau protein leading to an accumulation of hyperphosphorylated and aggregated Tau (5-7). In Alzheimer's disease (AD), Tau hyperphosphorylation is suggested to be a pathogenic process caused by aberrant activation of several kinases, in particular cyclin-dependent protein kinase 5 (cdk5) and glycogen synthase kinase-3 beta (gsk3 β. leading to phosphorylation of Tau on pathogenic sites. Hyperphosphorylated Tau in AD is believed to misfold, undergo net dissociation from microtubules and form toxic Tau aggregates (9, 10). Phosphorylation of Tau by cdk5 is initiated through activation by complex formation with one of the
-362017200078 06 Jan 2017 neuron-specific proteins p35 or p39 (.22. 23). However, only suppression of p35 by antisense oligonucleotide treatment, and not of the highly related isoform p39, selectively reduces cdk5 activity (24). In addition, levels of p35 but not cdk5 protein are rate-limiting for cdk5 activity (25). In concordance, we assessed the influence of Hsp90 inhibition on p35 expression.
We detected a dose- and time-dependent degradation of p35 by PU24FCI in primary neurons, as well as in COS-7/p35 and COS-7/p35/Tau cells. Embryonic primary rat conical neurons and COS-7 cells transfected with cDNAs corresponding to cither p35 alone (COS-7/p35) or both p35 and Tau (COS-7/p35/Tau) are cellular systems that enable the evaluation of these inhibitors on cdk5/p35 activity and stability and also on Tau phosphorylation at putative cdk5 sites. These are relevant experimental systems to study aberrant neuronal kinase activity because phosphorylation of Tau at these sites is enhanced in embryonic and juvenile brains (20) and is similar to AD afflicted brains (21). In addition. COS-7 cells transfected with the cdk5 activator p35 express Tau phosphorylated al pathogenic sites (21). Effects were seen at approximately 1-5 μΜ PU24FCI and were maximal at 10 uM Hsp90 inhibitor, in agreement with the affinity of this compound for Hsp90. Exogenously introduced p35 was more sensitive to Hsp90 inhibition than the endogenous protein, suggesting that by analogy to Hsp90 oncoproteins, buffering and slabilization of aberrant proteins in tauopathy may be accomplished by co-opting Hsp90. Reduction of p35 levels by Hsp90 inhibition affected the activity of the cdk5/p35 complex, as measured using a substrate of cdk5. the histone-Hl.
To investigate whether decreased p35 expression resulted in reduced phosphorylation of Tau, we measured Tau phosphorylation on three putative cdk5 sites, namely S2O2/T2O5. T231 andTl81 (26, 27). These sites have been shown to be abnormally phosphorylated in AD brains (28). PU24FCI lessened phosphorylation on these sites in a dosc-depcndcnt manner without affecting normal Tau protein expression. As observed for p35 levels and activity, effects were evident at 5 μΜ and maximal at 10 μΜ inhibitor. In addition, the kinetics of p35 degradation were similar to those observed for reduction in Tau phosphorylation.
To investigate the in vivo effect of Hsp90 inhibition on p35 in a WT Tau environment, we made use of hTau mice (41). hTau mice develop Tau pathology with a distribution that is comparable to that occurring in the early stages of AD. The majority of
-372017200078 06 Jan 2017
Tau pathology in hTau mice is located in the cortical brain region. These mice express six isoforms of non-mutant human Tau, but develop AD-like Tau-pathology. Heat-stable fractions (SI) prepared from cortical homogenates of these mice indicate an age-related accumulation of Tau phosphorylated at putative cdk5 sites. Wc examined whether inhibition of Hsp90 in these brains may lead to a reduction in p35 expression and a consequent alleviation of Tau phosphorylation. hTau female mice (n = 10) 4 and 8-10 months of age were administered either vehicle or one dose of PU-DZ8 (75 mg/kg) i.p. and animals were sacrificed at 4 h or 8 h post-administration. Aggregate-free Tau (SI) fractions were prepared from the cortical region of these mice and human Tau levels assessed by immunobloling with an antibody specific for 3-repeat domain Tau (RD3). By analogy to experiments on primary neuronal cultures and WT Tau transfected cells, the Hsp90 inhibitor had no effect on soluble WT Tau expression. However, both a significant time-dependent reduction in p35 levels (P = 0.0019) (Figure 16A) and alleviation of Tau phosphorylation on Ser202, as detected byantibody CP13 (P = 0.0078), were evident at 8 h post-administration of the Hsp90 inhibitor (figure 16B). The monoclonal antibody CP13 is commonly used to detect Tau pathology in both early and more advanced stages of Tau aggregate accumulation (41). Collectively, these data position p35/cdk5 as a kinase complex prone to aberrantly phosphorylate WT and mutant Tau, and suggest Hsp90 as a regulator of its activity in both Tau environments.
Fig.l6A shows the effect of PU-DZ8 on p35 in the htau mice that express pathogenically hyperphosphorylated WT tau similarly to Alzheimer's patients. Fig. 16B shows the effect of PU-DZ8 tau phosphorylation in the htau mice that express pathogenically hyperphosphorylated WT tau similarly to Alzheimer's patients.
References
The following references are cited herein, and are incorporated herein by reference in their entirety.
1. He H. Llauger L, Rosen N, Chiosis G. General Method for the Synthesis of 8-Arylsulfanyl Adenine Derivatives, J. Org. Chem. 2004. 69, 3230-3232.
2017200078 06 Jan 2017
- 38 2. He H, Zatorska D, Kim J, Aguirre J, Llauger L, She Y, Wu N, Immormino RM. Gewirth DT, Chiosis G. Identification of Potent Water-Soluble Purinc-Seaffold Inhibitors of the Heat Shock Protein 90. J. Med. Chem. 2006. 49( 1 ):381 -90.
3. Soti C, Csermely P. Chaperones and aging: role in neurodegeneration and in other civilizational diseases. Neurochem Int. 2002 Dec;41(6):383-9.
4. Poletti A, Negri-Cesi P, Martini L. Endocrine. 2005 Dec;28(3):243-62.
5. Beglopoulos V, Shen J. Trends Pharmacol Sci. 2006 Jan;27( 1 ):33-40.
6. Sorger PK. Heat shock factor and the heat shock response. Cell. 1991 May 3:65(3):363-6.
7. Zou J. Guo Y, Guettouche T, Smith DF, Voellmy R. Repression of heat shock transcription factor HSF1 activation by HSP90 (HSP90 complex) that forms a stress-sensitive complex with HSF1. Cell. 1998 Aug 21:94(4):471-80.
8. Morimoto RI, Santoro MG. Stress-inducible responses and heat shock proteins: new pharmacologic targets for cytoprotection. Nat Biotechnol. 1998 Sep: 16(9):833-8.,
9. Gardian G. Vecsei L. Huntington’s disease: pathomechanism and therapeutic perspectives. J Neural Transm. 2004 Oct;l 11 (10-11): 1485-94.
10. Rajgopal Y. Vemuri MC.Ethanol induced changes in eye I in-dependent kinasc-5 activity and its activators, P35, P67 (Munc-18) in rat brain.Neurosci Lett. 2001 Aug 10:308(3):173-6.
11. Patrick GN, Zukerberg L, Nikolic M, de la Monte S, Dikkes P, Tsai LH. Conversion of p35 to p25 deregulates Cdk5 activity and promotes neurodcgeneration. Nature. 1999 Dec 9:402(6762):615-22.
12. Dou F. Netzer WJ, Tanemura K, Li F, Hartl FU, Takashima A, Gouras GK, Greengard P, Xu H. Chaperones increase association of tau protein with microtubules Proc Natl Acad Sci USA. 2003 Jan 21:100(2):721-6.
13. Nakamura S. Kawamoto Y, Nakano S. Ikemoto A. Akiguchi I, Kimura J.Cyclin-dependent kinase 5 in Lewy body-like inclusions in anterior horn cells of a patient with sporadic amyotrophic lateral sclerosis. Neurology. 1997
Jari;48( 1 ):267-70.
-392017200078 06 Jan 2017
14. Bajaj NP. Al-Sarraj ST, Anderson V, Kibble M. Leigh N, Miller CC.Cyclin-dependent kinase-5 is associated with lipofuscin in motor neurones in amyotrophic lateral sclerosis. Neurosci Lett. 1998 Mar 27:245(1):45-8.
15. Wang J, Liu S, Fu Y, Wang JH, Lu Y. Cdk5 activation induces hippocampal CA I cell death by directly phosphorylating NMDA receptors. Nat Neurosci. 2003 Oct;6(10): 1039-47.
16. Sittler A, Lurz R, Lueder G, Priller J, Lehrach H, Hayer-Hartl MK, Haiti FU, Wanker EE. Geldanamycin activates a heat shock response and inhibits huntingtin aggregation in a cell culture model of Huntington's disease. Hum Mol Genet. 2001 Jun 1-,10(12):1307-15.
17. Winklhofer KF, Reintjes A, Hoener MC, Voellmy R, Tatzelt J. Geldanamycin restores a defective heat shock response in vivo. J Biol Chem. 2001 Nov 30:276(48):45160-7.
18. Tatzelt J, Zuo J, Voellmy R. Scott M, Hart! U, Prusiner SB, Welch WJ. Scrapie prions selectively modify the stress response in neuroblastoma cells. Proc Natl Acad Sci U S A. 1995 Mar 28:92(7):2944-8.
19. Auluck PK, Bonini NM. Pharmacological prevention of Parkinson disease in Drosophila. Nat Med. 2002 Nov;8(l 1):1185-6.
20. Weishaupt JH, Kussmaul L, Grotsch P, Heckel A, Rohde G, Romig H, Bahr M, Gillardon F.Inhibition of CDK5 is protective in necrotic and apoptotic paradigms of neuronal cell death and prevents mitochondrial dysfunction. Mol Cell Neurosci. 2003 Oct;24(2):489-502.
21. Bibb JA, Chen J, Taylor JR, Svenningsson P, Nishi A, Snyder GL, Yan Z, Sagawa ZK, Ouimet CC, Naim AC, Nestler EJ, Greengard P.Effects of chronic exposure to cocaine are regulated by the neuronal protein Cdk5.Nature. 2001 Mar 15:410(6826):376-80.
22. Brion JP, Couck AM.Cortical and brainstem-type Lewy bodies are immunorcactivc for the cyclin-dependent kinase 5.Am J Pathol. 1995 Nov; 147(5): 1465-76.
23. Chen J, Zhang Y. Kelz MB, Steffen C, Ang ES, Zeng L, Nestler EJ. Induction of cyclin-dependent kinase 5 in the hippocampus by chronic electroconvulsive seizures: role of [DeltajFosB.J Neurosci. 2000 Dec 15:20(24):8965-71.
-402017200078 06 Jan 2017
24. Sisodiya SM, Thom M, Lin WR, Bajaj NP, Cross JH. Harding BN. Abnormal expression of cdk5 in focal cortical dysplasia in humans.Neurosci Lett. 2002 Aug 16;328(3):217-20.
25. Green SL. Vulliet PR, Pinter MJ. Cork LC.Alterations in cyclin-dependcnt protein kinase 5 (CDK5) protein levels, activity and immunocytochemistry in canine motor neuron disease.J Neuropathol Exp Neurol. 1998 Nov;57(l 1): 1070-7.
26. Nakano S. Akiguchi 1, Nakamura S, Satoi H, Kawushima S, Kimura Aberrant expression of cyclin-dependent kinase 5 in inclusion body myositis. Neurology. 1999 Nov 10:53(8): 1671-6.
27. Nakamura S, Kawamoto Y, Nakano S, Akiguchi 1, Kimura J.Cyclin-dependcnt kinase 5 and mitogen-activated protein kinase in glial cytoplasmic inclusions in multiple system atrophy. J Neuropathol Exp Neurol. 1998 Jul;57(7):690-8.
28. Honjyo Y. Kawamoto Y, Nakamura S, Nakano S, Akiguchi 1.P39 immunorcaelivity in glial cytoplasmic inclusions in brains with multiple system atrophy.Acta Neuropathol (Berl). 2001 Mar: 101(3): 190-4.
30. E. Mandelkow and E.-M. Mandelkow, Kinesin motors and disease. Trends Cell Biol. 12(2002), pp. 585-591.
31. Hong M. Trojanowski JQ, Lee VMY. Tau-based neurofibrillary lesions. In: Clark CM. Trojanowski JQ. eds. Neurodegenerative dementia: clinical features and pathological mechanisms. New York: McGraw-Hill, 2000; 161-175.
32. von Bergen, M. , Barghom, S. , Li, L. , Marx, A. , Biemat, J. , Mandelkow. E. M. & Mandelkow, E. Mutations of tau protein in frontotemporal dementia promote aggregation of paired helical filaments by enhancing local beta-structure. (2001) J. Biol. Chem. 276, 48165-48174
33. Lee, V. M.. Goedert, M. & Trojanowski. J. Q. Neurodegenerative tauopalhics (2001) Annu. Rev. Neurosci. 24, 1121-1159
34. Matsumura, N., Yamazaki, T. & lhara, Y. Stable expression in Chinese hamster ovary cells of mutated tau genes causing frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). (1999) Am. J. Pathol. 154(6):1649-56.
-41 2017200078 06 Jan 2017
35. Fath. T., Eidenmuller, J., and Brandt, R. Tau-mediated cytotoxicity in a pseudohyperphosphorylation model of Alzheimer's disease (2002) J Neurosci 22, 9733-9741.
36. Ahlijanian, Μ. K., Barrezueta, N. X., Williams, R. D., Jakowski, A., Kowsz, K. P.. McCarthy, S., Coskran, T., Carlo, A., Seymour, P. A., Burkhardt, J. E., Nelson, R. B.,and McNeish, J. D. Hyperphosphorylated tau and neurolilament and cyloskeletal disruptions in mice overexpressing human p25, an activator of cdk5. (2000) Proc Natl Acad Sci U S A 97, 2910-2915.
37. Noble. W„ Olm, V., Takata, K., Casey, E., Mary, 0., Meyerson, J., Gaynor, K... LaFrancois, J.. Wang, L., Kondo, T., Davies, P., Bums, M., Veeranna, Nixon, R., Dickson. D.. Matsuoka, Y„ Ahlijanian, M., Lau, L. F., and Duff, K. Cdk5 is a key factor in tau aggregation and tangle formation in vivo. (2003) Neuron 38, 555-565.
38. Lucas, J. J., Hernandez, F., Gomez-Ramos, P., Moran, M. A., Hen, R., and Avila, J. Decreased nuclear beta-catenin, tau hyperphosphorylation and ncurodegencration in GSK-3beta conditional transgenic mice. (2001) Embo J 20, 27-39.
39. Tsai, L.H., Takahashi, T., Caviness, V. & Harlow, E. (1993) Activity and expression patterns of cyclin-dependent kinase 5 in the embryonic mouse nervous system. Development 119, 1029-1040.
40. Paudel, H.K., Lew, J., Ali, Z. & Wang, J. (1993) Brain proline-diiccted protein kinase phosphorylates tau on sites that are abnormally phosphorylated in tau associated with Alzheimer's paired helical filaments. J. Biol. Chem. 268, 23512-23518.
41. Lazaro, J.B., Kitzman. M., Poul, M.A., Vandromme, M., Lamb. N.Y. & Fernandez,
A. (1997) Cdk5, is a positive regulator of myogenesis in mouse C2 cells. J. Cell Sci. 110, 1251-1260.
42. Alvarez, A., Toro, R., Caceres, A. & Maccioni, R.B. (1999) Inhibition of tau phosphorylating protein kinase Cdk5 prevents beta-amyloid induced neuronal death. FEBS Lett. 459, 421-426.
43. Maccioni RB, Otth C, Concha II, Munoz JP. The protein kinase Cdk5. Structural aspects, roles in neurogenesis and involvement in Alzheimer's pathology. Fur 1 Biochem. 2001 Mar;268(6): 1518-27
-422017200078 06 Jan 2017
44. Walling AD.Amyotrophic lateral sclerosis: Lou Gehrig's disease.Am Fam Physician. 1999 Mar 15,59(6): 1489-96.
45. Julien JP, Beaulieu JM.Cytoskeletal abnormalities in amyotrophic lateral sclerosis: beneficial or detrimental effects, J Neurol Sci. 2000 Nov 1:180(1-2):7-14.
47. Julien JP, Couillard-Despres S, Meier J.Transgenic mice in the study of ALS: the role of neurofilaments.Brain Pathol. 1998 Oct;8(4):759-69.
48. Fahn S.Description of Parkinson's disease as a clinical syndrome.Ann N Y Acad Sci. 2003 Jun;991:1-14.
49. Neystat M, Rzhetskaya M. Oo TF. Kholodilov N, Yarygina O, Wilson A. El-Khodor BF, Burke RE.Expression of cyclin-dependent kinase 5 and its activator p35 in models of induced apoptotic death in neurons of the substantia nigra in vivo.J Neurochem. 2001 Jun;77(6):1611-25.
50. Kanemaru K, Takio K, Miura R, Titani K, Ihara Y. Fetal-type phosphorylation of the tau in paired helical filaments. J Neurochem. 1992 May:58(5): 1667-75.
51. Stoothoff WH, Johnson GV. Tau phosphorylation: physiological and pathological consequences. Biochim Biophys Acta. 2005 Jan 3:1739(2-3):280-97.
52. Michel G, Mercken M, Murayama M, Noguchi K, Ishiguro K, Imahori K, Takashima A. Characterization of tau phosphorylation in glycogen synthase kinasc-3bcta and cyclin dependent kinase-5 activator (p23) transfected cells. Biochim Biophys Acta. 1998 Apr 10:1380(2):177-82.
53. Stancato LF. Silverstein AM, Owens-Grillo JK, Chow YH, Jove R, Pratt WB. The hsp90-binding antibiotic geldanamycin decreases Raf levels and epidermal growth factor signaling without disrupting formation of signaling complexes or reducing the specific enzymatic activity of Raf kinase. J Biol Chem 1997; 272:4013-20.
54. Donze O, Abbas-Terki T, Picard D. The Hsp90 chaperone complex is both a facilitator and a repressor of the dsRNA-dependent kinase PKR. Embo J 2001: 20:3771-80.
55. Cadepond F, Schweizer-Groyer G, Segard-Maurel I, Jibard N, Hollenberg SM, Giguere V,e al. Heat shock protein 90 as a critical factor in maintaining glucocorticosteroid receptor in a nonfunctional state. J Biol Chem 1991; 266:5834-41
2017200078 06 Jan 2017
-43 56. Oshima, T., Ward. J.M.. Huh, C.G., Longenecker, G., Veeranna, C.. Pant, H.C.,
Brady. R.O.. Martin, L.J. & Kulkami, A.B. (1996) Targeted disruption of the cyclin-dependent kinase 5 gene results in abnormal corticogenesis, neuronal pathology and perinathan death. Proc. Natl Acad. Sci. USA 93, 11173 11178.
57. Chae, T., Kwon, Y.T., Bronson, R., Dikkes, P., Li. E. & Tsai, L.H. (1997) Mice lacking p35. a neuronal specific activator of Cdk5 display cortical lamination defects, seizures, and adult lethality. Neuron 18. 29-42.
58. Lee KY, Clark AW, Rosales JL, Chapman K, Fung T, Johnston RN. Elevated neuronal Cdc2-like kinase activity in the Alzheimer disease brain. Ncurosci Res. 1999 May;34(l):21-9.
59. Lewis J. McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtcnhorst M. Gwinn-Hardy K, Paul Murphy M, Baker M, Yu X, Duff K, Hardy J, Conal A, Lin WL, Yen SH, Dickson DW, Davies P, Hutton M. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet. 2000 Aug;25(4):402-5.
60. Hutton M, Lendon CL, Rizzu P, Baker M. Froelich S, Houlden H. Pickcring-Brown S. Chakraverty S, Isaacs A, Grover A, Hackett J. Adamson J, Lincoln S. Dickson D, Davies P, Petersen RC, Stevens M. de Graaff E, Wauters E, van Baren J, Hillcbrand M, Joosse M, Kwon JM, Nowotny P, Che LK, Norton J, Morris JC, Reed LA, Trojanowski J, Basun H, Lannfelt L, Neystat M, Fahn S, Dark F, Tanncnberg T, Dodd PR, Hayward N. Kwok JB, Schofield PR, Andreadis A, Snowden J, Craufurd D,
Neary D, Owen F, Oostra BA, Hardy J, Goate A, van Swieten J, Mann D, Lynch T, Heutink P. Association of missense and 5'-splice-site mutations in tau with the inherited dementia FTDP-17. Nature. 1998 Jun 18;393(6686):702-5.
61. Poorkaj P, Bird TD, Wijsman E, Nemens E, Garruto RM, Anderson L, Andreadis A, Wiederholt WC, Raskind M, Schellenbers GD. Tau is a candidate gene for chromosome 17 frontotemporal dementia.Ann Neurol. 1998 Jun;43(6):815-25.
62. Sahara N, Lewis J, DeTure M, McGowan E, Dickson DW. Hutton M, Yen SH. Assembly of tau in transgenic animals expressing P301L tau: alteration of phosphorylation and solubility. J Neurochem. 2002 Dec;83(6): 1498-1508.
2017200078 06 Jan 2017
-44 63. Lewis J., Dickson D. W., Lin W.-L., Chisholm L., Corral A., Jones G., Yen S.-H., Sahara N., Skipper L.. Yager D., Eckman C., Hardy J., Hutton M. and McGrowan E. (2001) Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 293. 1487-1491.
64. Tatebayashi Y, Miyasaka T. Chui DH, Akagi T, Mishima K, Iwasaki K, Fujiwara M. Tanemura K, Murayama M. Ishiguro K, Planel E, Sato S, Hashikawa T, Takashima A. Tau filament formation and associative memory deficit in aged mice expressing mutant (R406W) human tau. Proc Natl Acad Sci U S A. 2002 Oct 15 ;
99(21):13896-901.
65. Gotz J. Chen F. van Dorpe J, Nitsch RM. Formation of neurofibrillary tangles in P301I tau transgenic mice induced by Abeta 42 fibrils. Science. 2001 Aug 24:293 (5534): 1491-5.
66. Kim J. Felts S, He H, Llauger L. Huezo H, Rosen N, Chiosis G. Development of a fluorescence polarization assay for the molecular chaperone Hsp90. J. Biomolccular Screening 2004, 9(5):375-81.
67. Llauger L. Felts S, Huezo H. Rosen N, Chiosis G. Synthesis of novel fluorescent probes for the molecular chaperone Hsp90. Bioorg. Med. Chem. Lett. 2003, 13. 3975-3978.
68. Moulick K, Clement CC, Aguirre J, Kim J, Kang Y, Felts S, Chiosis G. Synthesis of a red-shifted fluorescence polarization probe for Hsp90. Bioorg. Med. Chem. Lett. Available online 22 June 2006.
69. Huezo H. Vilenchik M, Rosen N, Chiosis G. Microtiter cell-based assay for the detection of agents that alter cellular levels of Her2 and EGFR. Chem Biol. 2003, 10(7),629-634.
70. Ferrer I, Gomez-IslaT. Puig B, Freixes M, Ribe E, Dalfo E, Avila J. Current advances on different kinases involved in tau phosphorylation, and implications in Alzheimer’s disease and tauopathies. Cun' Alzheimer Res. 2005 Jan;2( I ):3-18.
71. Fu WY, Fu AK, Lok KC, Ip FC, Ip NY.Induction of Cdk5 activity in rat skeletal muscle after nerve injury.Neuroreport. 2002 Feb 11:13(2):243-7.
77. Greenberg, S. G., and Davies, P. (1990) Proc. Natl. Acad. Sci.USA 87, 5827-5831.
2017200078 06 Jan 2017
-45 89. Hansch C. Leo AJ. Substituent constant for correlation analysis in chemistry and biology. New York: Wiley, 1979.
90. Hansch C, Bjorkroth J. Leo AJ. Hydrophobicity and central nervous system agents: on the principle of minimal hydrophobicity in drug design. Pharm Sci 76: 663. 1987.
91. Osterberg T, Norinder U. Prediction of polar surface area and drug transport processes using simple parameters and PLS statistics. J Chem Inf Comput Sci 40: 1408-1411. 2000.
92. Feng RM. Assessment of blood-brain barrier penetration: in silico, in vitro and in vivo. Cuit Drug Metab 3: 647-657, 2002.
93. Skaaeda T, Okamura N, Nagata S, Yagami T, Horinouchi M, Okumura K, Yamahila F, Hashida M. Molecular and pharmacokinetic properties of 222 commercially available oral drugs in humans. Biol Pharm Bull 24: 935-940, 2001.
94. Abraham MH, Chadha HS, Martins F, Mitchell RC, Bradbury MW, Gratton J A. Hydrogen bonding part 46: a review of the correlation and prediction of transport properties by an LFER method: physicochemical properties brain penetration and skin permeability. Pestic Sci 55: 78-88, 1999.
95. Kelder J, Grootenhuis PDJ, Bayada DM, Delbressine LPC, Ploemcn J-P. Polar molecular surface as a dominating determinant for oral absorption and brain penetration of drugs. Pharm Res 16: 1514-1519, 1999.
96. van de Waterbeemd H, Camenish G, Folkers G, Chretien JR, Raevsky OA. Estimation of blood-brain barrier crossing of drugs using molecular size and shape, and H-bonding characteristics. J Drug Target 6: 151-165, 1998.
97. P. Ertl, B. Rohde, P. Selzer, Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. J.Med.Chem. 43, 3714-3717 (2000).
98. C.A. Lipinski, F. Lombardo, B.W. Dominy, PJ. Feeney, Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv.Drug.Delivery Rev. 23. 4-25 (1997).
99. D.F. Veber. S.R. Johnson, H.-Y. Cheng. B.R. Smith, K.W. Ward, K.D. Kopplc. Molecular properties that influence the oral bioavailability of drug candidates. J.Med.Chem. 45, 2615-2623 (2002).
-462017200078 06 Jan 2017
100. Clark DE. In silico prediction of blood-brain barrier permeation, Drug Discovery Today 8: 927-933, 2003.
101. Lin JH, Rodrigues AD. In vitro model for early studies of drug metabolism. In: Pharmacokinetics optimization in drug research: biological, physicochemical and computational strategies (Testa, Van de Wuterbeemed H, Folkcr G, Guy R, eds.), pp 217-243. New York: Wiley-VCH, 2001.
102. Raub JT. Early preclinical evaluation in support of hit identification and lead optimization for brain exposure. AAPS Workshop on Optimization of Drug-Like Properties During Lead Optimization. Parsippany, NJ, 19-22 September. 2004.
103. Klettner A. The induction of heat shock proteins as a potential strategy to treat neurodegenerative disorders. Drug News Perspect. 2004 Jun; 17(5):299-306.
104. Barral JM, Broadley SA, Schaffar G, Hartl FU. Roles of molecular chaperones in protein misfolding diseases. Semin Cell Dev Biol. 2004 Feb; 15( 1): 17-29.
105. Xu H., Gouras GK, Greenfield JP, Vincent B. Naslund J, Mazzarcllu L. Fired G, Jovanovic JN. Seeger M, Relkin NR et al. (1998) Nat. Ned. 4: 447-451.
106. Lamphere L. Fiore F, Xu X, Brizuela L, Keezcr S, Sardet C, Draeltu GF, Gyuris J (1997) Oncogene 14:1999-2004.
1002340256
2017200078 17 Oct 2018

Claims (27)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
    1. Use of a compound in the preparation of a medicament for treatment of neurodegenerative disease, wherein the compound is of the formula:
    or a salt thereof, wherein
    R is hydrogen, or a Ci to Cl0 alkyl, C2 to Cio alkenyl, C2 to Cio alkynyl, or C2 to Cio alkoxyalkyl group, optionally including heteroatoms;
    Yi and Y2 are independently C orN;
    X4 is hydrogen or halogen;
    X3 is CH2, CF2 S, SO, SO2, Ο, NH, or NR2, wherein R2 is alkyl; and
    X2 is halogen, alkyl, halogenated alkyl, alkoxy, halogenated alkoxy, hydroxyalkyl, pyrollyl, optionally substituted aryloxy, alkylamino, dialkylamino, carbamyl, amido, alkylamido dialkylamido, acylamino, alkylsulfonylamido, trihalomethoxy, trihalomethyl, thioalkyl, SO2alkyl, COO-alkyl, NH2, OH, or CN; and
    Xi has the formula -X-Y-Z-, wherein X, Y and Z are independently C, N, S or O, connected by single or double bonds and with appropriate hydrogen substitution to satisfy valence, or Y may be (CH2)2; and wherein one of X and Z is bonded at the 5'-position of the aryl ring and the other is bonded to the 4' position, with the proviso that the compound is not PU-H71 or PU-DZ8.
  2. 2. Use of a compound in the preparation of a medicament for treatment of neurodegenerative disease, wherein the compound is of the formula:
    1002340256
    2017200078 17 Oct 2018 or a salt thereof, wherein:
    n is an integer no greater than 2;
    R is a Cj-Cio alkyl moiety which includes a nitrogen heteroatom;
    X4 is hydrogen or halogen;
    X3 is CH2, CF2, S, SO, SO2, Ο, NH or NR2, wherein R2 is alkyl; and X2 is halogen.
  3. 3. A method of treatment of neurodegenerative disease comprising administering to an individual in need of such treatment a therapeutically effective amount of a compound according to the formula:
    or a salt thereof, wherein
    R is hydrogen, or a Ci to C10 alkyl, C2 to C10 alkenyl, C2 to C10 alkynyl, or C2 to C10 alkoxyalkyl group, optionally including heteroatoms;
    Yi and Y2 are independently C or N;
    X4 is hydrogen or halogen;
    X3 is CH2, CF2 S, SO, SO2, Ο, NH, or NR2, wherein R2 is alkyl; and
    X2 is halogen, alkyl, halogenated alkyl, alkoxy, halogenated alkoxy, hydroxyalkyl, pyrollyl, optionally substituted aryloxy, alkylamino, dialkylamino, carbamyl, amido, alkylamido dialkylamido, acylamino, alkylsulfonylamido, trihalomethoxy, trihalomethyl, thioalkyl, SO2alkyl, COO-alkyl, NH2, OH, or CN; and
    1002340256
    2017200078 17 Oct 2018
    Xi has the formula -X-Y-Z-, wherein X, Y and Z are independently C, N, S or O, connected by single or double bonds and with appropriate hydrogen substitution to satisfy valence, or Y may be (CKfyh; and wherein one of X and Z is bonded at the 5'-position of the aryl ring and the other is bonded to the 4' position;
    with the proviso that the compound is not PU-H71 or PU-DZ8.
  4. 4. A method of treatment of neurodegenerative disease comprising administering to an individual in need of such treatment a therapeutically effective amount of a compound according to the formula:
    R or a salt thereof, wherein:
    n is an integer no greater than 2;
    R is a Ci-Cio alkyl moiety which includes a nitrogen heteroatom;
    X4 is hydrogen or halogen;
    X3 is CH2, CF2, S, SO, SO2, Ο, NH or NR2, wherein R2 is alkyl; and X2 is halogen.
    wherein one of X and Z is bonded at the 5'-position of the aryl ring and the other is bonded to the 4' position.
  5. 5. The use of claim 1 or 2 or method of claim 3 or 4, wherein at least one of X, Y and Z is a carbon atom.
  6. 6. The use or method of claim 5, wherein Xi is -O-(CH2)n-O-, wherein n is 1 or 2.
  7. 7. The use or method of claim 5 or 6, wherein X2 is halogen.
    1002340256
    2017200078 17 Oct 2018
  8. 8. The use or method of any one of claims 5-7, wherein R is an alkyl group containing a nitrogen heteroatom.
  9. 9. The use or method of claim 8, wherein R is 3-isopropylaminopropyl,
    3 -(isoprop yl(methyl)amino)propyl, 3 -(isopropyl(ethyl)amino)propyl,
    3 -((2-hydroxyethy l)(isopropyl)amino)propyl, 3 -(methyl(prop-2-ynyl)amino)propyl, 3-(allyl(methyl)amino)propyl, 3-(ethyl(methyl)amino)propyl,
    3-(cyclopropyl (propyl)amino)propyl, 3-(cyclohexyl(2-hydroxyethyl)amino)propyl, 3-(2-methylazirdin-l-yl)propyl, 3-(piperidin- l-yl)propyl,
    3-(4-(2-hydroxyethyl )piperazin-l-yl)propyl, 3-morpholinopropyl,
    3-(trimethylammonio)propyl, 2-(isopropylamino)ethyl, 2-(isobutylamino)ethyl, 3-(neopentylamino)ethyl, 2-(cyclopropylmethylamino)ethyl, 2-(ethyl(methyl)amino)ethyl,
    2- (isobutyl(methyl)amino)ethyl, or 2-(methyl(prop-2-ynyl)amino)ethyl.
  10. 10. The use or method of claim 9, wherein R is 3-(methyl(prop-2-ynyl)amino)propyl, 2(isobutylamino)ethyl, or 2-(neopentylamino)ethyl.
  11. 11. The use or method of any one of claims 1-7, wherein R is a terminal alkyne.
  12. 12. The use or method of claim 11, wherein R is propynyl.
  13. 13. The use or method of any one of claims 5-12, wherein X3 is CH2 or S.
  14. 14. The use or method of any one of claims 1-7 and 13, wherein R is 3(isopropyl(methyl)amino)propyl, 3 -(isopropyl(ethyl)amino)propyl, 3 -((2hydroxyethyl)(isopropyl)amino)propyl, 3-(methyl(prop-2-ynyl )amino)propyl, 3(allyl(methyl)amino)propyl, 3-(ethyl(methyl)amino)propyl,
    3- (cyclopropyl(propyl)amino)propyl, 3-(cyclohexyl(2-hydroxyethyl)amino)propyl, 3-(2-methylaziridin-l-yl)propyl, 3-(piperidin-l-yl)propyl,
    3-(4-(2-hydroxyethyl)piperazin-l -yl)propyl, 3-morpholinopropyl,
    3-(trimethylammonio)propyl, 2-(isopropylamino)ethyl, 2-(isobutylamino)ethyl, 2-(neopentylamino)ethyl, 2-(cyclopropylmethylamino)ethyl, 2-(ethyl(methyl)amino)ethyl, 2-(isobutyl(methyl)amino)ethyl, or 2-(methyl(prop-2-ynyl)amino)ethyl.
    1002340256
    2017200078 17 Oct 2018
  15. 15. The use or method of claim 14, wherein R is 3-(methyl(prop-2-ynyl)amino)propyl, 2(isobutylamino)ethyl, or 2-(neopentylamino)ethyl.
  16. 16. The use or method of claim 14 or 15, wherein X2 is I.
  17. 17. The use or method of any one of claims 1-16, wherein the neurodegenerative disease is one in which aggregate, plaque or tangle formation occurs, or is a tauopathy.
  18. 18. The use or method of any one of claims 1-16, wherein the neurodegenerative disease is characterized by abnormalities in a signalling pathway and/or by aberrant neuronal protein folding.
  19. 19. The use or method of any one of claims 1-16 wherein the neurodegenerative disease is complete androgen insensitivity syndrome (CAIS), spinal and bulbar muscular atrophy (SBMA or Kennedy's disease), Alzheimer's Disease (AD), Gerstmann Straussler syndrome, hereditary Creutzfeldt-Jakob disease, autosomal dominant Parkinson's disease, or Huntington disease.
  20. 20. The use or method of any one of claims 1-16, wherein the neurodegenerative disease is Alzheimer's Disease.
  21. 21. The use or method of claim 17, wherein the neurodegenerative disease is a tauopathy.
  22. 22. The use or method of claim 21, wherein the compound is PU-HZ150:
    , or a salt thereof.
  23. 23. The use or method of claim 21, wherein the compound is PU-HZ151:
    1002340256
    2017200078 17 Oct 2018 , or a salt thereof.
  24. 24. The use or method of claim 21, wherein the compound is PU-BSI5:
    , or a salt thereof.
  25. 25. The use or method of claim 21, wherein the compound is PU-DZ13:
    , or a salt thereof.
  26. 26. The use or method of claim 21, wherein the compound is PU-DZ14:
    , or a salt thereof.
  27. 27. The use or method of claim 21, wherein the compound is PU-BSI11
    1002340256
    2017200078 17 Oct 2018 or a salt thereof.
    2017200078 06 Jan 2017
    1/16
    Fig. 1A
    Fig. 1B
    2017200078 06 Jan 2017
    2/16 mg/kg t=ZI p35 cdk5
    B p-tau (AT-8) B§ Hsp70 1^· Hsp90
    Fig . 2
    Fig. 3
    3/16
    2017200078 06 Jan 2017
    X. Υ, Z = 0. C, N, S
    NH, I z benzoturans, 2.3-dihydiobenzolurans, indoles, beinlsoxazoles, benoxazoles. benzimidazoles 2.3-Cyctopcnlenopyhdines, benzothiophen-3(2H)ones. benzothiophene
    Fig. 4
    2017200078 06 Jan 2017
    4/16
    Fig. 5, Part 1
    2017200078 06 Jan 2017
    5/16
    Fig. 5, part 2
    2017200078 06 Jan 2017
    6/16
    Fig. 5, Part 3
    2017200078 06 Jan 2017
    7/16
    Reagents and conditions: (a) K2CO3, DMAP, DMF, 120°C; (b) NaOMe, MeOH, i-BuOH, 1O5°C; (c) HF-pyridine, NaNO2, rt; (d) MBS, DMF, rt or NIS, CF3COOH, DMF, rt; (e) Cs2CO3,DMF, 50°C.
    Fig. 6
    8/16
    2017200078 06 Jan 2017
    Reagents and conditions: (a) NaOt-Bu. neocuproine, Cul, DMF, 110°C; (b) NIS, CF3COOH, acetonitrile, rt.
    Fig· 7
    9/16
    2017200078 06 Jan 2017
    4000
    Untreated Tx 75mg/kg PUDZ8
    Fig. 8A
    Untreated Tx75mg/kg PUDZ8
    Fig. 8B
    2017200078 06 Jan 2017
    10/16
    JNPL3 mice Tx IP with 75mg/kg PUDZ8
    Fig. 9
    Hsp70 Htau (HT7) p35
    Hsp90
    11/16
    2017200078 06 Jan 2017
    Fig. 10
    Hsp90 binding
    0 T-1-1' ' ·' T
    0 12 3 log 10 [PU] (nM) PU-DZ2 A PU-DZ3 ▼ PU-DZ7 ♦ PU-DZB • PUH58 □ PUH84 Δ PU-H85 v PU-H71 o PU24FCI
    Fig. 11 A
    12/16
    2017200078 06 Jan 2017
    Hsp70 induction in SK-N-SH
    430300
    150000' [PU] (ulvl) —B-PU-DZ2 -A- PU-DZ3 -T-PU-DZ7
    PU-DS -·- PU-H58 -a- PU-HS4 -A-PU-H65 -V- PU-H71
    PU24FCI
    Fig. 11 B
    13/16
    2017200078 06 Jan 2017
    Log10 [Hsp90 inhibitor] (nM)
    Fig. 12
    Fig. 13
    2017200078 06 Jan 2017
    14/16
    Fig. 14A
    Fig. 14B
    15/16
    2017200078 06 Jan 2017 <Λ c
    ω >
    J5 φ
    &
    12.510.07.55.02.50.0
    AT180 (P-T231)(P3)
    Fig. 15 °-cro^Untreated
    Treated p35
    Fig. 16A
    16/16
    2017200078 06 Jan 2017
    CP13(P-S202)
    30to c
    Φ >
    a
    Φ a:
    2010** □
    0J--ΓΟΗ
    Fig. 16B
    8h
AU2017200078A 2006-06-30 2017-01-06 Treatment of neurodegenerative diseases through inhibition of Hsp90 Ceased AU2017200078C1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2017200078A AU2017200078C1 (en) 2006-06-30 2017-01-06 Treatment of neurodegenerative diseases through inhibition of Hsp90

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60/806,427 2006-06-30
AU2007269144A AU2007269144B2 (en) 2006-06-30 2007-07-02 Treatment of neurodegenerative diseases through inhibition of HSP90
AU2013205338A AU2013205338B2 (en) 2006-06-30 2013-04-11 Treatment of neurodegenerative diseases through inhibition of Hsp90
AU2017200078A AU2017200078C1 (en) 2006-06-30 2017-01-06 Treatment of neurodegenerative diseases through inhibition of Hsp90

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2013205338A Division AU2013205338B2 (en) 2006-06-30 2013-04-11 Treatment of neurodegenerative diseases through inhibition of Hsp90

Publications (3)

Publication Number Publication Date
AU2017200078A1 AU2017200078A1 (en) 2017-02-02
AU2017200078B2 true AU2017200078B2 (en) 2018-11-08
AU2017200078C1 AU2017200078C1 (en) 2019-03-07

Family

ID=48446965

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2013205338A Ceased AU2013205338B2 (en) 2006-06-30 2013-04-11 Treatment of neurodegenerative diseases through inhibition of Hsp90
AU2017200078A Ceased AU2017200078C1 (en) 2006-06-30 2017-01-06 Treatment of neurodegenerative diseases through inhibition of Hsp90

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU2013205338A Ceased AU2013205338B2 (en) 2006-06-30 2013-04-11 Treatment of neurodegenerative diseases through inhibition of Hsp90

Country Status (1)

Country Link
AU (2) AU2013205338B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006130469A1 (en) * 2005-05-27 2006-12-07 Oregon Health & Science University Stimulation of neurite outgrowth by small molecules

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006130469A1 (en) * 2005-05-27 2006-12-07 Oregon Health & Science University Stimulation of neurite outgrowth by small molecules

Also Published As

Publication number Publication date
AU2013205338B2 (en) 2016-10-06
AU2017200078A1 (en) 2017-02-02
AU2017200078C1 (en) 2019-03-07
AU2013205338A1 (en) 2013-05-02

Similar Documents

Publication Publication Date Title
US20230060990A1 (en) Treatment of neurodegenerative diseases through inhibiton of hsp90
US8357673B2 (en) 4-arylazo-3,5-diamino-pyrazole compounds and use thereof
Šála et al. Novel human neutral sphingomyelinase 2 inhibitors as potential therapeutics for Alzheimer’s disease
WO2007015632A1 (en) Atm and atr inhibitor
JP2011504474A (en) Use of Mnk inhibitors to treat Alzheimer&#39;s disease
Dou et al. Rational modification, synthesis and biological evaluation of 3, 4-dihydroquinoxalin-2 (1H)-one derivatives as potent and selective c-Jun N-terminal kinase 3 (JNK3) inhibitors
AU2017200078B2 (en) Treatment of neurodegenerative diseases through inhibition of Hsp90
US20240158397A1 (en) Treatment of neurodegenerative diseases through inhibiton of hsp90
ES2897712T3 (en) Procedures for Screening Selective and Nonselective Phosphatase Inhibitors
Liu et al. Development of bisindole-substituted aminopyrazoles as novel GSK-3β inhibitors with suppressive effects against microglial inflammation and oxidative neurotoxicity
WO2013149976A1 (en) Indole-pyrimidine derivatives and their therapeutic uses
KR102352624B1 (en) Pyridopyrimidine derivatives as lrrk2 (leucine rich repeat kinase 2) inhinitor
EP2662081A1 (en) Therapeutic use of indole-dihydro-imidazole derivatives
EP2970118A1 (en) Compounds for the treatment of neurological disorders

Legal Events

Date Code Title Description
DA2 Applications for amendment section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 22 NOV 2018

DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS AS SHOWN IN THE STATEMENT(S) FILED 22 NOV 2018

FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired