throbber
WO2010/065751
`
`PCT/US2009/066600
`
`are inhibitors of neprilysin are useful analgesic agents which can be administered with
`
`the GCC agonists described herein or covalently linked to a GCC agonist to form a
`
`therapeutic conjugate. Sialorphin and related polypeptides are described in U.S. Patent
`
`6,589,750; U.S. 20030078200 Al; and WO 02/051435 A2.
`
`[197]
`In another embodiment, a GCC agonist formulation of the invention is
`administered as part of a regimen of combination therapy with an opioid receptor
`antagonist or agonist. In one embodiment, the GCC agonist and the opioid receptor
`antagonist or agonist are linked via a covalent bond. Non-limiting examplesof opioid
`
`receptor antagonists include naloxone, naltrexone, methyl nalozone, nalmefene,
`cypridime, beta funaltrexamine, naloxonazine,naltrindole, nor-binaltorphimine,
`enkephalin pentapeptide (HOE825; Tyr-D-Lys-Gly-Phe-L-homoserine; SEQ ID
`NO:258), trimebutine, vasoactive intestinal polypeptide, gastrin, glucagons. Non-
`limiting examples of opioid receptor agonists include fedotozine, asimadoline, and
`ketocyclazocine, the compounds described in WO03/09705 1! and WO05/007626,
`morphine, diphenyloxylate, frakefamide (H-Tyr-D-Ala-Phe(F)-Phe-NH 23 SEQ ID
`NO:259; WO 01/019849 Al), and loperamide.
`
`Further non-limiting examples of analgesic agents that can be used ina
`[198]
`regimen of combination therapy along with the GCCagonist formulations ofthe
`invention include the dipeptide Tyr-Arg (kyotorphin); the chromogranin-derived
`‘polypeptide (CgA 47-66; See, e.2., Ghia et al. 2004 Regulatory polypeptides 119:199);
`CCKreceptor agonists such as caerulein; conotoxin polypeptides; peptide analogs of
`thymulin (FR Application 2830451); CCK (CCKa or CCKb) receptor antagonists,
`including loxiglumide and dexloxiglumide (the R- isomer of loxighumide) (WO
`
`88/05774); 5-HT4 agonists such as tegaserod (Zelnorm®), mosapride, metoclopramide,
`
`zacopride, cisapride, renzapride, benzimidazolone derivatives such as BIMU 1 and
`
`BIMU8,andlirexapride; calcium channel blockers such as ziconotide and related
`
`compoundsdescribedin, for example, EP625162B1, US 5,364,842, US 5,587,454, US
`5,824,645, US 5,859,186, US 5,994,305, US 6087,091, US 6,136,786, WO 93/13128 Al,
`
`88
`
`RECTIFIED SHEET (RULE 91) ISA/EP
`MYLAN - EXHIBIT 1022 Part 12 of 16
`3776
`
`3776
`
`MYLAN - EXHIBIT 1022 Part 12 of 16
`
`

`

`WO2010/065751
`
`PCT/US2009/066600
`
`EP 1336409 Al, EP 835126 Al, EP 835126 Bl, US 5,795,864, US 5,891,849, US
`6,054,429, WO 97/01351 Al; NK-I, receptor antagonists such as aprepitant (Merck & Co
`Inc), vofopitant, ezlopitant (Pfizer, Inc.), R-673 (Hoffmann-La Roche Ltd), SR-48968
`(Sanofi Synthelabo), CP-122,721 (Pfizer, Inc.), GW679769 (Glaxo Smith Kline), TAK-
`637 (Takeda/Abbot), SR-14033,
`
`88a
`
`RECTIFIED SHEET (RULE 91) ISA/EP
`
`3777
`
`3777
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`and related compounds describedin, for example, EP 873753 Al, US 20010006972 Al, US
`
`20030109417 Al, WO 01/52844 Al (for a review see Giardinaet al. 2003.Drugs 6:758); NK-2
`
`receptor antagonists such as nepadutant (Menarini Ricerche SpA), saredutant (Sanofi-
`
`Synthelabo), GW597599 (Glaxo Smith Kline), SR-144190 (Sanofi-Synthelabo) and UK-290795
`
`(Pfizer Inc); NK3 receptor antagonists such as osanetant (SR-142801; Sanofi-Synthelabo), SSR-
`
`241586, talnetant and related compounds described in, for example, WO 02/094187 A2, EP
`
`876347 Al, WO 97/21680 Al, US 6,277,862, WO 98/1 1090, WO 95/28418, WO 97/19927, and
`
`Bodenet al. (J Med Chem. 39:1664-75, 1996); norepinephrine-serotonin reuptake inhibitors
`
`C(NSRI) such as milnacipran and related compounds described in WO 03/077897; and vanilloid
`
`receptor antagonists such as arvanil and related compouds described in WO 01/64212 AL.
`
`[199]
`
`In addition to sialorphin-related polypeptides, analgesic polypeptides include:
`
`AspPhe, endomorphin-1, endomorphin-2, nocistatin, dalargin, lupron, ziconotide, and substance
`
`P.
`
`EXAMPLES
`
`Example 1: Synthesis and Purification of GCC Agonist Peptides
`
`[200]
`
`The GCC agonist peptides were synthesized using standard methods for solid-
`
`phase peptide synthesis. Either a Boc/Bzl or Fmoc/tBu protecting group strategy was seleceted
`
`depending upon the scale of the peptide to be produced. In the case of smaller quantities, it is
`
`possible to get the desired product using an Fmoc/tBu protocol, but for larger quantitics (1 g or
`
`more), Boc/Bzl is superior.
`
`[201]
`
`In each case the GCC agonist peptide was started by either using a pre-loaded
`
`Wang (Fmoc) or Merrifield (Boc) or Pam (Boc)resin. For products with C-terminal Leu, Fmoc-
`
`Leu-Wang (D-1115) or Boc-Leu-Pam resin (D-1230) or Boc-Leu-Merrifield (D-1030) Thus, for
`
`peptides containing the C-terminal d-Leu, the resin was Fmoc-dLeu-Wang Resin (D-2535) and
`
`Boc-dLeu-Merrifield, Boc-dLeu-Pam-Resin (Bachem Product D-1230 and D-1590, respectively)
`
`(SP-332 and rclated analogs). For peptides produced as C-terminal amides, a resin with Ramage
`
`89
`
`3778
`
`3778
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`linker (Bachem Product D-2200) (Fmoc) or mBHA (Boc) (Bachem Product D-1210 was used
`
`and loaded with the C-terminal residue as the first synthetic step.
`
`Fmoc-tBu Overview
`
`[202]
`
`Each synthetic cycle consisted deprotection with 20% piperidine in DMF. Resin
`
`washes were accomplished with alternating DMF and IpOHto swell and shrink the resin,
`
`respectively. Peptide synthesis elongated the chain from the C-terminus to the N-terminus.
`
`Activation chemistry for cach amino acid was with HBTU/DIEAin a 4 fold cxcess for 45
`
`minutes. In automated chemistries, each amino acid was double coupled to maximize the
`
`coupling efficiency. To insure the correct position of disulfide bonds, the Cys residues were
`
`introduced as Cys (Acm)at positions 15 and 7. Cys (Trt) was positioned at Cys4 and Cys12.
`
`This protecting group strategy yields the correct topoisomer as the dominant product (75:25).
`
`(For enterotoxin analogs, a third disulfide bond protecting group (Mob) wasutilized).
`
`[203]
`
`For peptides containing C-terminal Acea (aminoethyloxyethyloxyacetyl) groups,
`
`these were coupled to a Ramage amide linker using the same activation chemistry above by
`
`using an Fmoc-protected Acea derivative. The Cys numbering in these cases remains the same
`
`and the positioning of the protecting groups as well. For the peptides containing the N-terminal
`
`extension of Acea, the Cys residue numbering will be increased by three Cys4 becomes Cys7,
`
`Cys12 becomes Cys15; Cys7 becomes Cys10 and Cys 15 becomes Cys18. The latter pair is
`
`protected with Acm and the former pair keeps the Trt groups.
`
`[204]
`
`For analogs containing D-aminoacid substitutions, these were introduced directly
`
`by incorporating the correctly protected derivative at the desired position using the same
`
`activation chemistry described in this document. For Fmocstrategics, Fmoc-dAsn(Trt)-OH,
`
`Fmoc-dAsn(Xan)-OH, Fmoc-dAsp(tBu)-OH, Fmoc-dGlu(tBu)-OH and for Boc strategies, Boc-
`
`dAsn(Xan)-OH, Boc-dAsn(Trt)-OH, Boc-dAsp(Chx), Boc-dAsp(Bzl)-OH, Boc-dGlu(Chx)-OH
`
`and Boc-dGlu(Bzl)-OH would be utilized.
`
`[205]
`
`Each peptide is cleaved from the solid-phase support using a cleavage cocktail of
`
`TFA:H20:Trisisopropylsilane (8.5:0.75:0.75) ml/g of resin for 2 hr at RT. The crude deprotected
`
`peptideis filtered to remove the spent resin beads and precipitated into ice-cold diethylether.
`
`90
`
`3779
`
`3779
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`[206]
`
`Each disulfide bonds was introduced orthogonally. Briefly, the crude synthetic
`
`product wasdissolved in water containing NH4OHto increase the pH to 9. Following complete
`
`solubilization of the product, the disulfide bond was made between the Trt deprotected Cys
`
`residues bytitration with H2O2. The monocyclic product was purified by RP-HPLC. The purified
`
`mono-cyclic product was subsequently treated with a solution of iodine to simultaneously
`
`remove the Acm protecting groups and introduce the second disulfide bond.
`
`[207]
`
`For cntcrotoxin analogs, the Mob group was removed via treatment of the dicyclic
`
`product with TFA 85% containing 10% DMSO and 5% thicanisole for 2 hr at RT.
`
`[208]
`
`Each product was then purified by RP-HPLC using a combination buffer system
`
`of TEAP in H20 versus MeCN,followed by TFA in H20 versus MeCN. Highly purefractions
`
`were combined and lyophilized. The final product was converted to an Acetate salt using either
`
`ion exchange with Acetate loaded Dow-Exresin or using RP-HPLC using a base-wash step with
`
`NH,4OAc followed by 1% AcOH in water versus MeCN.
`
`[209]
`
`It is also possible to prepare enterotoxin analogs using a random oxidation
`
`methodology using Cys(Trt) in Fmoc or Cys(MeB)in Boc. Following cleavage, the disulfide
`
`bonds can be formed using disulfide interchange redox pairs such as glutathione (red/ox) and/or
`
`cysteine/cystine. This process will yield a folded product that the disulfide pairs must be
`
`determined as there would be no way of knowing their position directly.
`
`Boc-Bil Process
`
`[210]
`
`Peptide synthesis is initiated on a Merrifield or Pam pre-loaded resin or with
`
`mBHAfor peptides produced as C-terminal amides. Each synthetic cycle consists of a
`
`deprotection step with 50% TFA in MeCL2. The resin is washed repetitively with MeCl2 and
`
`MeOH. The TFAsalt formed is neutralized with a base wash of 10% TEA in MeCl2. Theresin is
`
`washed with MeCl2 and MeOHandlastly with DMFprior to coupling steps. A colorimetric test
`
`is conducted to ensure deprotection. Each coupling is mediated with diisopropyl carbodiimide
`
`with HOBTto form the active ester. Each coupling is allowed to continue for 2 hr at RT or
`
`overnight on difficult couplings. Recouplings are conducted with either Uronium or
`
`Phosphoniumreagents until a negative colorimetric test is obtained for free primary amines. The
`
`91
`
`3780
`
`3780
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`resin is then washed with DMF, MeCl2 and MeOHandprepared for the next solid-phase step.
`
`Cysprotection utilizes Cys(Acm) at positions 7 and 15, and Cys(MeB)at Cys 4 and Cys12.
`
`[211]
`
`Cleavage and simultaneous deprotection is accomplished by treatment with HF
`
`using anisole as a scavenger (9:1:1) ml:ml:g (resin) at 0°C for 60 min. The peptide is
`
`subsequently extracted from the resin and precipitated in ice cold ether. The introduction of
`
`disulfide bonds and purification follows the exact same protocol described above for the Fmoc-
`
`produced product.
`
`Example 2: Jn vitro Biological and Chemical Stability of SP-304 after Incubation in
`
`Simulated Gastric Fluid (SGF)
`
`[212]
`
`Thestability of SP-304 in the presence of simulated gastric fluid (SGF) was
`
`determined by biological activity measurements and HPLC analyses (Figs. 1A & 1B). SP-304
`
`(final concentration of 8.5 mg/ml) was incubated in SGF (Proteose peptone (8.3 g/liter; Difco),
`
`D-Glucose (3.5 g/liter; Sigma), NaCl (2.05 g/liter; Sigma), KH 2PO, (0.6 g/liter; Sigma), CaCl,
`
`(0.11 g/liter), KCI (0.37 g/liter; Sigma), Porcine bile (final 1 X concentration 0.05 g/liter; Sigma)
`
`in PBS, Lysozyme(final 1 X concentration 0.10 g/liter; Sigma) in PBS, Pepsin (final 1 X
`
`concentration 0.0133 g/liter; Sigma) in PBS). SGF was made on the day of the experiment and
`
`the pH wasadjusted to 2.0 + 0.1 using HCl or NaOHas necessary. After the pH adjustment,
`
`SGFis filter sterilized with 0.22 um membranefilters. SP-304 (final concentration of 8.5
`
`mg/ml) was incubated in SGF at 37°C for 0, 15, 30, 45, 60 and 120 min, respectively, in
`
`triplicate aliquots. Following incubations, samples were snap frozen in dry ice then stored in a -
`
`80°C freezer until assayed in duplicate.
`
`[213]
`
`Figure 1A showsa bar chart showing the biological activity of SP-304 after
`
`incubation with SGF for times as indicated. The activity at 0 min was taken as 100%. The data
`
`are an average oftriplicates + SD for each data point. The data demonstrate that SP-304 is
`
`resistant to breakdown in SGFfor incubationslasting as long as two hours. In addition, the data
`
`also suggest that the activity of SP-304 is unaltcred by exposure to the acidic pH of the SGF.
`
`[214]
`
`The HPLC chromatograms of samples of SP-304 incubated in SGF for 0 and 120
`
`min are shown in Fig. 1B. Here, aliquots of the two samples were analyzed by HPLC using a
`
`92
`
`3781
`
`3781
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`previously developed method for analyzing SP-304 peptide. Samples from the SGF incubations
`
`were diluted to give a final concentration 0.17 mg/mL of SP-304. The major peak of SP-304 did
`
`not change following incubation with SGF, indicating that the peptide was resistant to SGF
`
`treatment.
`
`Example 3: én vitro Biological and Chemical Stability of SP-304 after Incubation in
`
`Simulated Intestinal Fluid (SLF)
`
`[215]
`
`The stability of SP-304 was also evaluated after incubation with simulated
`
`intestinal fluid (SIF) by measuring its biological activity and by HPLC analyses (Figs. 2A & 2B).
`
`SIF solution was prepared by the method as described in the United States Pharmacopoeia, 24th
`
`edition, p2236. The recipe to prepare SIF solution was as described below. The SIF solution
`
`contained NaCl (2.05 g/liter; Sigma), KH 2PO4 (0.6 g/liter; Sigma), CaCl, (0.11 g/liter), KCl
`
`(0.37 g/liter; Sigma), and Pacreatin 10 mg/ml. The pH wasadjusted to 6 and the solution was
`
`filter sterilized. A solution of SP-304 (8.5 mg/ml) was incubated in SGF at 37°C for 0, 30, 60,
`
`90, 120, 150 and 300 min respectively, in triplicate aliquots. Following incubations, samples
`
`were removed and snap frozen with dry ice and stored in a -80°C freezer until assayed in
`
`duplicate. Figure 2A is a bar chart showing the ability of SP-304, after incubation in SIF for
`
`times as indicated, to stimulate cGMP synthesis in T84 cells. The cGMPstimulation activity at 0
`
`min was taken as 100%. The data are an average of 3 triplicates + SD. The data indicated that
`
`the biological activity of SP-304 is reduced by about 30% following incubation in SIFfor 300
`
`min.
`
`[216]
`
`The physical stability of SP-304 peptide exposed to SIF was evaluated by HPLC
`
`using the method described for SGF digestion. Figure 2B shows HPLC chromatograms for SP-
`
`304 after incubation with heat-inactivated SIF for 300 min, and SIF for 120 min, respectively.
`
`SP-304 treated with heat-inactivated SIF remained intact (Note: the major peak of SP-304
`
`eluting at 16.2 min), whereas SP-304 treated with SIF for 120 min was completely converted
`
`into another peak eluting at 9.4 min plus a few minor additional peaks.
`
`[217]
`
`Figure 3 is a schematic representation of the possible metabolites of SP-304. The
`
`major degradation products involve Asn and Aspclipped from the N-terminus and Leu from the
`
`93
`
`3782
`
`3782
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`C-terminus of SP304. The fact that only 30% reduction in biological activity was observed even
`
`after 2 hours incubation in SIF implies that one or more of the degradation products observed in
`
`Fig. 2B are also biologically active. To address this possibility, several truncated peptides were
`
`synthesized and evaluated for their abilities to stimulate cGMPsynthesis in T84 cells (Figure 4).
`
`[218]
`
`Figure 4 showsdata from the analyses of various peptides in the T&84 cell cGMP
`
`stimulation assay (essentially as described in Shailubhai, et a/., Cancer Research 60, 5151-5157
`
`(2000) . Bricfly, conflucnt monolayers of T-84 cells in 24-well plates were washed twice with
`
`250 wl of DMEM containing 50 mM HEPES (pH 7.4) and pre-incubated at 37°C for 10 minutes
`
`with 250 ul of DMEM containing 50 mM HEPES (pH 7.4) and 1 mM isobutyl methylxanthine
`
`(BMX). Monolayers of T&4 cells were then incubated with 250 ul of DMEM containing 50 mM
`
`HEPES(pH 7.4) containing one of the peptides shown in the Figure 4 at a concentration of 1.0
`
`uM for 30 min. After the 30 min incubation, the medium was aspirated and the reaction was
`
`terminated by the addition of 3% perchloric acid. Following centrifugation and the addition of
`
`NaOH (0.1 N) to neutralize the pH,intracellular cGMP levels were determined in lysates using a
`
`cGMP ELISA kit (Cat. No. 581021 ; Cayman Chemical, Ann Arbor, MI). Peptide incubations
`
`were run in duplicate, and samples taken from each incubation were run as duplicates in the
`
`ELISA test.
`
`[219]
`
`The data indicate that SP-338, the 15-mer peptide missing the leucine (L) residue
`
`at the C-terminus of SP-304, retains about 80% ofthe biological activity of the full length 16-
`
`mer SP-304 peptide. Thus, the C-terminal Leu clearly does make some contribution to the
`
`biological potency of the peptide. Similarly, peptides SP-327, SP-329 and SP-331, whichareall
`
`missing their C-terminal Leu, also showed a 20-25% reduction in biological potency relative to
`
`their counterpart parent peptides SP-326, SP-328 and SP-330, respectively. In addition, the data
`
`also suggest that amino acid residues at the N-terminus may also contribute to the stability and/or
`
`potency of the peptides. Several additional peptides were synthesized with D-forms of amino
`
`acids replacing the corresponding L-forms at the C- and N-termini of the peptides. These
`
`peptides were evaluated for their abilities to stimulate cGMP synthesis in T84 cells as shown in
`
`Figure 5.
`
`94
`
`3783
`
`3783
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`[220]
`
`The results presented in Figure 5 indicate that substitution of L-aminoacids with
`
`D-aminoacids at the C- and N-termini did not significantly alter their potency relative to SP-304.
`
`Peptides SP-332, SP-333 and SP-335 all showed comparable ability to stimulate cGMP synthesis
`
`in T84 cells. These results suggest that the amino acid residues Asn, Asp and Glu at the N-
`
`terminus and Leuat the C-terminus can be replaced with their respective D-aminoacid forms.
`Onthe other hand, substitution of L-leucine with D-leucineat the 6"position (SP-337) resulted
`
`in virtually complete loss of biological activity.
`
`[221]
`
`Figure 7 (A-F) showsthe stabilities of peptides SP-332, SP-333 and SP-304 when
`
`incubated in SIF for two hours. The results demonstrate that SP-333, which has D-Asnat the N-
`
`terminus and D-Leu at the C-terminus, remained virtually 100% biologically active after a two
`
`hour incubation in SIF (Figure 7A), and remained virtually intact to digestion with SIF after two
`
`hours (Figs. 7F-1 & 7F-2). Subsequent incubation studies with SP-333 performed in SIF for up
`
`to 24 hours indicate that there is very little degradation even after 24 hours in SIF (Fig. 7G). The
`
`data indicated that SP-333 is stable against digestion with SIF for up to 24 hours. Peptide SP-
`
`332 with D-Leu at the C-terminus showed a minor reduction in potency following the 120 min
`
`incubation with SIF (Figure 7B). Interestingly, the HPLC analyses of SP-332 did not reveal any
`
`clear-cut degradation of the peptide (Figure 7E-1 & 7E2), also suggesting that this peptide is also
`
`almost completely resistant to proteoysis by SIF during the 2-hr incubation. On the other hand,
`
`peptide SP-304 lost about 30% of its potency following digestion with SIF for just one hour
`
`(Figure 7C). The HPLC analysis of SP-304 following SIF incubation confirmedits degradation
`
`(Figure 7D-1 & 7D-2). These results suggest that SP-304 undergoes substantial proteolysis
`
`following incubation with SIF within one hour.
`
`Example 4: Cyclic GMP Stimulation Assays
`
`[222]
`
`Theability of the GCC agonist peptide to bind to and activate the intestinal GC-C
`
`receptor wastested usingT 84 human colon carcinomacell line. Human T84 colon carcinoma
`
`cells were obtained from the American Type Culture Collection. Cells were grown in a 1:1
`
`mixture of Ham's F-12 medium and Dulbecco's modified Eagle's medium (DMEM)
`
`supplemented with 10% fetal bovine serum, 100 U penicillin/ml, and 100 Lg/ml streptomycin.
`
`The cells were fed fresh medium every third day andsplit at a confluence of approximately 80%.
`
`95
`
`3784
`
`3784
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`[223]
`
`Biological activity of the GCC agonist peptides was assayed as previously
`
`reported (Shailubhai, et a/., Cancer Research 60, 5151-5157 (2000)). Briefly, the confluent
`
`monolayers of T-84 cells in 24-well plates were washed twice with 250 ul of DMEMcontaining
`
`50 mM HEPES(pH 7.4), pre-incubated at 37°C for 10 min with 250 wl of DMEM containing 50
`
`mM HEPES(pH 7.4) and 1 mM isobutylmcthylxanthinc (BMX), followed by incubation with
`
`GCC agonist peptides (0.1 nM to 10 .mu.M) for 30 min. The medium wasaspirated, and the
`
`reaction was terminated by the addition of 3% perchloric acid. Following centrifugation, and
`
`neutralization with 0.1 N NaOH, the supernatant was used directly for measurements of cGMP
`
`using an ELISA kit (Caymen Chemical, Ann Arbor, Mich.).
`
`[224]
`
`Figure 6 showsresults from experiments evaluating the potency of peptides (via
`
`cGMPstimulation assay) having structures similar to the 14-mer peptide SP-339, also referred to
`
`as linaclotide. SP-339 is a truncated analog of the E. coli enterotoxin ST peptide. SP-354 was
`
`foundto be virtually identical to SP-339 in biological activity. Notably, peptide SP-353, which
`has a Serresidue at the 6" position, was found to be more potent than SP-339, and was the most
`
`potent of all the peptides tested. Peptide SP-355 which has a D-Tyrat the C-terminus showed
`
`considerably less potency than the other peptidestested.
`
`Example 5: Peggylated Peptides
`
`[225]
`
`An additional strategy to render peptides moreresistant towards digestion by
`
`digestive proteases is to peggylate them at the N- and C-terminus. The peptide SP-333 was
`
`peggylated with the aminoethyloxy-ethyloxy-acetic acid (Acea) group at the C-terminus (SP-
`
`347) or at the N-terminus (SP-350) or at both termini (SP-343). Cyclic GMP synthesis in T84
`
`cells was measured by the method as described above.
`
`[226]
`
`The peptides SP-347 and SP-350 showed potencies comparable to SP-333 in their
`
`abilities to stimulate cGMPsynthesis in T84 cells. However, peptide SP-343 was considerably
`
`less potent as compared to the other peptides tested. The poor activity of SP-343 might be due to
`
`the considerable steric hindrance afforded by the large Aeea groups at both termini.
`
`96
`
`3785
`
`3785
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`Example 6: Combination of Guanylate Cyclase Receptor Agonists with
`Phosphodiesterase Inhibitors
`
`[227]
`
`Regulation of intracellular concentrations of cyclic nucleotides (7.e., cAMP and
`
`cGMP)and thus, signaling via these second messengers, has been generally considered to be
`
`governedbytheir rates of production versus their rates of destruction within cells. Thus, levels of
`
`cGMPintissues and organs can also be regulated by the levels of expression of cGMP-specific
`
`phosphodiesterases (CGMP-PDE), which are generally overexpressed in cancer and
`
`inflammatory diseases. Therefore, a combination consisting of an agonist of GC-C with an
`
`inhibitor of cGMP-PDE might produce synergistic effect on levels of cGMPin the target tissues
`
`and organs.
`
`[228]
`
`Sulindac Sulfone (SS) and Zaprinast (ZAP) are two of the knowninhibitors of
`
`cGMP-PDEand have been shownto induce apoptosis in cancer cells via a cGMP-dependent
`
`mechanism. SS and ZAP in combination with SP-304 or SP-333 were evaluated to see if these
`
`PDEinhibitors had any synergistic effects on intracellular accumulation of cGMP(Fig. 9-12).
`
`As the data show, SS at a concentration of 100 uM did not enhance intracellular accumulation of
`
`cGMP. However, the combination of SS with SP-304 stimulated cGMP production several-fold
`
`more then stimulation by SP-304 alone. This synergistic effect on cGMP levels was more
`
`pronounced when SP-304 were used at a 0.1 uM concentration (Fig 10). Similar observations
`
`were made when SP-304 or SP-333 were used in combination with ZAP (Fig 10, Fig 11 and Fig
`
`12). These results suggest that the intracellular levels of cGMPare stabilized because SS inhibits
`
`cGMP-PDEthat might be responsible for depletion of intracellular cGMP. Thus, the approach to
`
`use a combination of GCC agonist with a cGMP-PDEinhibitoris attractive.
`
`[229]
`
`For the results shown in Figure 9, cyclic GMP synthesis in T84 cclls was assessed
`
`essentially as described in Shailubhai et al., Cancer Research 60, 5151-5157 (2000). Briefly,
`
`confluent monolayers of T-84 cells in 24-well plates were washed twice with 250 ul of DMEM
`
`containing 50 mM HEPES(pH 7.4) and pre-incubated at 37°C for 10 minutes with 250 ul of
`
`DMEMcontaining 50 mM HEPES (pH 7.4) and 1 mM isobutyl methylxanthine (BMX).
`
`Monolayers of T84 cells were then incubated with 250 ul of DMEM containing 50 mM HEPES
`
`(pH 7.4) containing SP-304 or PDE inhibitors either alone or in combinations, as indicated below
`
`in the following experimental sets: 1) Control; 2) SP-304 (0.1 uM); 3) Sulindac Sulfone (100
`
`97
`
`3786
`
`3786
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`uM); 4) Zaprinast (100 uM); 5) SP-304 (0.1 uM) + Sulindac Sulfone (100 uM); and 6) SP-304
`
`(0.1 uM) + Zaprinast (100 uM). After the 30 min incubation, the medium wasaspirated and the
`
`reaction was terminated by the addition of 3% perchloric acid. Following centrifugation and the
`
`addition of NaOH (0.1 N) to neutralize the pH,intracellular cGMP levels were determined in
`
`lysates using a cGMPELISAkit (Cat. No. 581021 ; Cayman Chemical, Ann Arbor, MI).
`
`Incubations were performed in duplicate, and cach sample was run in duplicate using the ELISA
`
`test.
`
`[230]
`
`For the results shown in Figure 10, the method used was same as the one used for
`
`Fig. 9 except that the monolayers of T84 cells were incubated with 500 wl of DMEM containing
`
`50 mM HEPES(pH 7.4) containing SP-304 (0.1 or 1.0 uM)or increasing concentrations of PDE
`
`inhibitors (0 to 750 uM) either alone or in combination with SP-304. After the 30 min
`
`incubation, the medium wasaspirated and the reaction was terminated by the addition of 3%
`
`perchloric acid. Following centrifugation and the addition of NaOH (0.1 N) to neutralize the pH,
`
`intracellular cGMPlevels were determined in lysates using a cGMP ELISAkit (Cat. No.
`
`581021; Cayman Chemical, Ann Arbor, MI). Samples were runintriplicate using the ELISA
`
`test.
`
`[231]
`
`For the results shown in Figure 11,
`
`the method used was sameas the one used for
`
`Fig. 10 except that the monolayers of T84 cells were incubated with 500 ul of DMEM containing
`
`50 mM HEPES(pH 7.4) containing SP-3333 (0.1 or 1.0 uM) or increasing concentrations of
`
`ZAP (0 to 500 uM) either alone or in combination with SP-333. After the 30 min incubation, the
`
`medium wasaspirated and the reaction was terminated by the addition of 3% perchloric acid.
`
`Following centrifugation and the addition of NaOH (0.1 N) to neutralize the pH, intracellular
`
`cGMPlevels were determined in lysates using a cGMP ELISAkit (Cat. No. 581021 ; Cayman
`
`Chemical, Ann Arbor, MI). Samples were run in triplicate using the ELISAtest.
`
`[232]
`
`For the results shown in Figure 12, the method used was sameas the one usedfor
`
`Fig. 10 except that the monolayers of T84 cells were incubated with 500 Ul of DMEM containing
`
`50 mM HEPES(pH 7.4) containing SP-333 (0.1 LM) or increasing concentrations of Sulindac
`
`Sulfone (0 to 500 UM) either alone or in combination with SP-333. After the 30 min incubation,
`
`the medium was aspirated and the reaction was terminated by the addition of 3% perchloric acid.
`
`98
`
`3787
`
`3787
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`Following centrifugation and the addition of NaOH (0.1 N) to neutralize the pH, intracellular
`
`cGMPlevels were determined in lysates using a cGMP ELISA kit (Cat. No. 581021 ; Cayman
`
`Chemical, Ann Arbor, MI). Samples were run in triplicate using the ELISAtest.
`
`Example 7: A Repeated Oral Dose Toxicity Study of SP-304 in Cynomolgus Monkeys.
`
`[233]
`
`The primary purpose ofthis experiment was to evaluate the toxicity and
`
`pharmacokinetics of a repeated oral dose of SP-304 in cynomolgus monkeys. Treatment with a
`
`daily dose of 250 mg of SP-304 for 14 consecutive days was well tolerated by all of the
`
`monkeys, however the treatment consistently produced liquid feces and watery diarrhea (Figure
`
`14). Monkeys returned to normal stool consistency within 24-48 hours following the last dose of
`
`SP-304.
`
`Example 8:
`
`SP-304 Treatment Improves Stool Consistency and Clears TNBS-induced
`Intestinal Blockage in a TNBS-induced Murine Modelof Colitis.
`
`[234]
`
`SP-304 is a superior analog of uroguanylin and an agonist of GC-C. The anal
`
`administration of trinitrobenzene sulphonic acid (TNBS)is typically used to produce intestinal
`
`blockage, resulting in poor stool consistency. As shown in Figure 13, oral administration of SP-
`
`304 considcrably improved stool consistency in mice treated with TNBS. Treatment with SP-304
`
`at a dose between 0.05 to 0.5 mg/kg body weight was sufficient to completely restore the
`
`consistency score to the level observed in mice treated with phosphate buffer instead of TNBS
`
`(minus TNBS control). Sulfasalazine, a FDA approved drug used as a positive control, also
`
`restored normal stool consistency.
`
`Example 9: A Randomized, Double-blind, Placebo-Controlled, Single-, Ascending-, Oral-
`Dose Safety, Tolerability, and Pharmacokinetic Study of SP-304 in Healthy
`Adult Human Male and Female Volunteers
`
`[235]
`
`The objectives of this study were to assess the safety and pharmacokinetics of a
`
`single oral dose of SP-304 in healthy subjects. This was a phase 1, single-site, randomized,
`
`double-blind, placebo-controlled, single-, ascending-, oral-dose, sequential dose escalation study
`
`of SP-304 in fasted, healthy male and female subjects. A total of 9 cohorts utilizing 8 subjects
`
`per cohort (6 SP-304; 2 placebo) were utilized, totaling 71 volunteers administered drug (one
`
`99
`
`3788
`
`3788
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`volunteer dropping out). Each cohort was administered a single, oral dose or matching placebo
`
`administered in 10-fold diluted phosphate buffered saline (PBS) (240 mL). Subjects were only
`
`administered one dose of study treatment and could not be enrolled in subsequent cohorts. The
`
`nine cohort doses included 0.1, 0.3, 0.9, 2.7, 5.4, 8.1, 16.2, 24.3 mg and 48.6 mg SP-304.
`
`Doses of SP-304
`
`0.1 mg (6 active, 2 placebo)
`0.3 mg (6 active, 2 placebo)
`0.9 mg (6 active, 2 placcbo)
`2.7 mg (6 active, 2 placebo)
`5.4 mg (6 active, 2 placebo)
`8.1 mg (6 active, 2 placebo)
`16.2 mg (5 active, 2 placcbo)
`24.3 mg(6 active, 2 placebo)
`48.6 mg(6 active, 2 placebo)
`
`[236]
`
`The decision to proceed to the next cohort was madeby the study sponsor and
`
`principal investigator after reviewing the preliminary blinded, safety information from the
`
`cohort. All safety data collected through the 48 hours after dosing were reviewedto assess
`
`tolerability of the dose level. A minimum of 3 evaluable subjects were required for the
`
`determination of safety and tolerability at each dose level.
`
`[237]
`
`The stopping criteria were: 1) clinically significant adverse events [including
`
`clinically significant changes in laboratory or electrocardiogram (ECG) parameters] in >4
`
`subjects (collectively within a cohort), or 2) 1 drug related, serious adverse event (SAE). No
`
`higher doses were to be administered if one of these criteria was met. Otherwise, the study could
`
`proceed to the next higher dose cohort.
`
`[238]
`
`Safety was monitored by physical examinations, vital signs, clinical laboratory
`
`tests (hematology, chemistry, urinalysis, fecal occult blood), ECG, and adverse experience
`
`assessments). Serial blood samples were collected 0, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 16, 24, 36, and
`
`48 hours after dosing. Plasma samples were assayed by a validated method for SP-304, and
`
`pharmacokinetic parameters calculated. Pharmacodynamic endpoints that were evaluated
`
`includedtimeto first stool, stool frequency (48-hour period), and stool consistency (48-hour
`
`period) using the Bristol Stool Form Scale (BSFS).
`
`100
`
`3789
`
`3789
`
`

`

`WO 2010/065751
`
`PCT/US2009/066600
`
`[239]
`
`The phase | study (Protocol No. SP-SP304101-08) used an oral solution prepared
`
`by a registered licensed pharmacist at the investigation site not more than one hour before
`
`administration of dose.
`
`[240]
`
`The primary objectives of this clinical evaluation were to determine safety,
`
`toxicity and systemic absorption of a single oral dose of SP-304. The data indicated that SP-304
`
`was well-tolerated at all dosage levels and there were no severe adverse events (SAEs). The
`
`most prevalent adverse event (AE) observed during this study was grade | diarrhea (12.7%), as
`
`defined using the Common Terminology Criteria for Adverse Events (CTCAE), which is an
`
`increase in the number of bowel movements from 1 and <4 in a 24-hour period. Notably, SP-
`
`304 was expected to promote bowel movement, thus the increase in number of bowel
`
`movements was considered to be related to the pharmacodynamic (PD) action of SP-304.
`
`[241]
`
`The effect of a single oral dose of SP-304 on stool consistency, as judged by the
`
`Bristol form stool scale (BSFS), was also examined in volunteers. The BSFS sco

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket