WO2003008439A1 - ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS - Google Patents

ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS Download PDF

Info

Publication number
WO2003008439A1
WO2003008439A1 PCT/US2001/022801 US0122801W WO03008439A1 WO 2003008439 A1 WO2003008439 A1 WO 2003008439A1 US 0122801 W US0122801 W US 0122801W WO 03008439 A1 WO03008439 A1 WO 03008439A1
Authority
WO
WIPO (PCT)
Prior art keywords
mono
alkyl
bicyclic
group
aryl
Prior art date
Application number
PCT/US2001/022801
Other languages
French (fr)
Inventor
Samuel H. Gellman
Bernard Weisblum
Emilie Ann Porter
Xifang Wang
Original Assignee
Wisconsin Alumni Research Foundation
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
Application filed by Wisconsin Alumni Research Foundation filed Critical Wisconsin Alumni Research Foundation
Priority to PCT/US2001/022801 priority Critical patent/WO2003008439A1/en
Publication of WO2003008439A1 publication Critical patent/WO2003008439A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention is directed to antimicrobial compositions containing active ingredients which are oligomers and/or polymers of ⁇ -amino acids.
  • Chemists have long sought to extrapolate the power of biological catalysis and recognition to synthetic systems. These efforts have focused largely on low molecular weight catalysts and receptors. Most biological systems, however, rely almost exclusively on large polymers such as proteins and RNA to perform complex chemical functions.
  • Proteins and RNA are unique in their ability to adopt compact, well-ordered conformations. These two biopolymers are unique also because they can perform complex chemical operations (e.g. , catalysis, highly selective recognition, etc.). Folding is linked to function in both proteins and RNA because the creation of an "active site" requires proper positioning of reactive groups. Consequently, there has been a long-felt need to identify synthetic polymer backbones which display discrete and predictable folding propensities (hereinafter referred to as "foldamers”) to mimic natural biological systems. Such backbones will provide molecular "tools" to probe the functionality of large-molecule interactions (e.g. protein-protein and protein-RNA interactions).
  • foldamers synthetic polymer backbones which display discrete and predictable folding propensities
  • Cationic peptides that adopt amphiphilic secondary structures constitute a widespread host defense against microbial invasions, and mimicry of these natural antibiotics led the present inventors on a search for new synthetic antimicrobial agents.
  • Many of the cationic antimicrobial peptides appear to act by disrupting bacterial membranes, a mechanism that may not be conducive to the development of resistance.
  • the subject invention is directed to antimicrobial compositions containing unnatural cationic oligomers that have no known natural counterpart and that display antibiotic activity comparable to that of a magainin derivative against at least four different bacterial species, including two pathogens that are resistant to common antibiotics.
  • the oligomers are constructed from ⁇ -amino acid building blocks rather than the ⁇ - amino acid building blocks of conventional peptides. These ⁇ -peptides exhibit lower hemolytic activity than does the magainin derivative, which indicates a favorable selectivity for bacterial cells relative to mammalian cells. Because ⁇ -peptides are not expected to be subject to protease degradation, these results point toward the rational design of a promising new antibiotic class.
  • the present invention is directed to compounds and to antimicrobial compositions containing the compounds, that is, compositions which inhibit the growth of microbes in general and bacteria and fungi in particular, the compositions comprising an antimicrobial-effective amount of a ⁇ -amino acid oligomer or polymer of the formula:
  • X and Y are preferably (and independently) selected from the group consisting of linear or branched C r C 6 -alkyl, hydroxy ⁇ C,-C 6 - alkyl, amino- - -alkyl, aryl-Cj-Cg-alkyl, heteroaryl- -Cg-alkyl, and carboxyl-C r C 6 - alkyl.
  • X and Y When X and Y are combined together into a cyclic substituent, together X and Y and the carbons to which they are bonded define a substituted or unsubstituted cyclopentyl, substituted or unsubstituted cyclohexyl, unsubstituted or N-substituted piperidinyl, or unsubstituted or N-substituted pyrrolidinyl.
  • the composition contains a ⁇ -amino acid oligomer or polymer of the above, wherein each residue (A, B, C, etc.) of the oligomer is a ⁇ - amino acid residue of formula:
  • X and Y combined, together with the carbon atoms to which they are bonded, independently from residue to residue define a substituted or unsubsituted C 3 - C 8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more nitrogen atoms as the sole heteroatom, the substituents being selected from the group consisting of hydroxy, linear or branched C,-C 6 -alkyl, alkenyl, alkynyl; hydroxy-C ⁇ -C 6 -alkyl, amino-C ⁇ -C 6 -alkyl, C,-C 6 -alkyloxy, C j - -alkyloxy-Cj-C f j-alkyl, amino, mono- or di- C j -C 6 -alkylamino, carboxamido, carboxamido-C,-C 6 -alkyl, sulfonamido, sulfonamido- C,
  • the invention is likewise directed to a method of inhibiting microbial growth in mammals, including humans, the method comprising administering an antimicrobial amount of a composition as recited above to a patient in need thereof.
  • Fig. 1 shows the structure of poly- ⁇ -alanine and further depicts the hydrogen bonds that define the six narrowest helices available to poly- ⁇ -alanine.
  • Poly- ⁇ -alanine is the simplest ⁇ -peptide polymer.
  • Fig. 2 A depicts the crystal structure of oligomer of trans-AC ⁇ Cl 14-helix.
  • the bottom depiction is the two-dimensional structure
  • the middle depiction is a view along the axis of the helix
  • the top depiction is a view perpendicular to the axis of the helix.
  • Fig. 2B depicts the crystal structure of oligomer of tra/w-ACPC/12-helix. The views shown are the same as in Fig. 2A.
  • Fig. 2C depicts the crystal structure of a standard ⁇ -helix. The views shown are the same as in Fig. 2 A.
  • Fig. 3 depicts 'H NMR spectra for a solution containing 2 mM trans-ACHC dimer and 2 mM trans-ACHC hexamer. The bottom spectrum was obtained in CD 3 OH with solvent suppression. The two NH resonances from the dimer are indicated with an asterisk (*). All other spectra were obtained in CD 3 OD at the times indicated after dissolution of the sample. Data obtained on a Bruker 300 MHz spectrometer at 20 ° C .
  • Fig. 4 is a circular dichroism (CD) plot for trans- ACPC hexamer in CH 3 OH.
  • Fig. 5 is a CD plot comparing trans- ACVC dimer, trimer, tetramer and hexamer.
  • Fig. 6 depicts X H NMR spectra for a solution containing 2 mM of a hexamer of alternating amino-substituted-tran_f-ACHA and trans-ACHA. All spectra were obtained in D 2 O, 100 mM deuteroacetate buffer, pD 3.9, at the times indicated after dissolution of the sample. Data obtained on a Bruker 300 MHz spectrometer at 20° C.
  • Fig. 7 is a comparison between the k ⁇ , for amide proton exchange in a hexamer of alternating amino-substituted-tr ⁇ /w-ACHA and trans-ACHA and a dimer of alternating amino-substituted-tra w-ACHA and trans-ACHA.
  • Fig. 8 is a comparison between the CD plot of a hexamer of alternating amino- substituted-tra/w-ACHA and trans-ACHA in water and the CD plot of a hexamer of trans-ACHC in methanol.
  • Fig. 9 is a comparison of CD data in methanol for a ⁇ -peptide tetramer, hexamer, and octamer containing alternating trans- ACPA residues and 4-pyrrolidinyl residues.
  • Fig. 10 is a comparison of CD data in water for a ⁇ -peptide tetramer, hexamer, and octamer containing alternating trans-ACPA residues and 4-pyrrolidinyl residues.
  • Fig. 11 is a superimposed plot of the CD data in water and the CD data in methanol for the octamer containing alternating trans-ACPA residues and 4- pyrrolidinyl residues.
  • Fig. 12 is a CD spectrum in water of an octamer containing alternating trans- ACPA residues and 3-pyrrolidinyl residues.
  • Fig. 13 is a comparison of CD spectra for a hexamer containing alternating residues of trans-ACHA and ammo-substituted-trans-ACHA and for a "mixed" ⁇ - peptide hexamer comprising alternating residues of trans-ACHA and an acyclic ⁇ - amino acid bearing an aminopropyl substituent on the ⁇ -carbon of the backbone.
  • Fig. 14A shows the infrared spectrum of two linked nipecotic acid residues wherein the two residues have the same absolute configuration.
  • Fig. 14B shows the infrared spectrum of two linked nipecotic acid residues wherein the two residues have the opposite absolute configuration. This diastereomer acts as a reverse turn in ⁇ -peptides which adopt a sheet structure.
  • Fig. 15 is a schematic representation of the "split and pool” method of generating combinatorial libraries.
  • Fig. 16 is a ball and stick representation of the solid state conformation of compound 1 described below. For clarity, all hydrogen atoms, except for those attached to nitogen, have been omitted. Hydrogen bonds are indicated with dotted lines.
  • Fig. 17 is a graph depicting the results of a direct hemolysis comparison between melittin (a positive control), a magainin derivative, and ⁇ -17 (see Example 11).
  • ⁇ -amino acid oligomers (“ ⁇ - peptides") are well suited for adoption of compact secondary structures stabilized by intramolecular hydrogen bonds.
  • Fig. 1 shows the hydrogen bonds that define the six narrowest helices available to poly- ⁇ -alanine, the simplest ⁇ -peptide polymer.
  • the 12-, 16-, and 20-helices (nomenclature derived from hydrogen-bonded ring size) contain hydrogen bonds from carbonyls toward NH groups in the N-terminal direction, as observed for 3 J0 - and ⁇ -helices in proteins, while the 10-, 14-, and 18-helices contain hydrogen bonds from carbonyls to NH groups in the N-terminal direction.
  • trans-ACHC trans-2-aminocyclohexanecarboxylic acid
  • trans-ACPC trans-2-aminocyclopentanecarboxylic acid
  • trans-ACHC optically active trans-ACHC was prepared by the reported route, Nohira et al. (1970) Bull. Chem. Soc. Jpn. 43:2230, and polypeptide oligomers synthesized via standard methods (see below).
  • the crystal structures of the trans-ACHC tetramer and the trans-ACHC hexamer reveal that these molecules adopt 14-helical conformations in the solid state.
  • the hexamer crystal contains three independent but very similar molecules, each of which forms the four possible 14-membered ring hydrogen bonds.
  • the regular helix revealed by the hexamer crystal structure matches the minimum energy conformation predicted for the decamer.
  • trans- ACPC trans-2-aminocyclopentanecarboxylic acid
  • Fig. 2B depicts the 12-helix rr ns-ACPC structure, as compared to a standard ⁇ -helix (Fig. 2C) and trans-ACHC/ 14-helix (Fig. 2A). This finding shows that ⁇ -peptides allow profound residue-based control of peptide conformation.
  • Routine 13 C-NMR spectra were obtained on a Bruker AC-300 and are referenced to the NMR solvent.
  • High resolution electron impact mass spectroscopy was performed on a Kratos MS-80RFA spectrometer with DS55/DS90.
  • Infrared Spectroscopy Spectra were obtained on a Nicolet Model 740 FT-IR spectrometer. IR samples were prepared under anhydrous conditions; CH 2 C1 2 was freshly distilled from CaH 2 , compounds and glassware were dried under vacuum for 1-2 days, and solutions were prepared under a nitrogen atmosphere. The pure solvent spectrum for a particular solution was subtracted from the sample spectrum prior to analysis. Peaks in the amide NH stretch region were baseline corrected, and analyzed without further manipulation. NMR Spectroscopy. 1. Aggregation Studies. One-dimensional spectra for aggregation studies were obtained on a Bruker AC-300 spectrometer.
  • Samples for aggregation studies were prepared by serial dilution from the most concentrated sample (50 mM or 27 mM). Dry compounds were dissolved in CD 2 C1 2 previously dried over 3 A molecular sieves, and samples were prepared with dry glassware under a nitrogen atmosphere.
  • NMR samples for conformational analysis were prepared by dissolving the dry compound in dry deuterated solvent under a nitrogen atmosphere.
  • CD 2 C1 2 samples were then degassed by the freeze-pump-thaw method, and the NMR tubes were sealed under vacuum.
  • Methanol samples were sealed with a close fitting cap and parafilm.
  • COSY spectra were obtained on a Bruker AC-300 spectrometer. TOCSY (Braunschweiler, L.; Ernst, R. R. (1983) J. Magn. Reson. 53:521), NOESY (Macura, S.; Ernst, R. R. (1980) Mol. Phys. 41:95), and ROESY (Bothner-By, A. A. ; Stephens, R.
  • NOESY and ROESY spectra were recorded with a similar number of t [ and tj points, and 32 and 40 scans per t ⁇ increment, depending on the sample concentration.
  • the width of the spectral window examined was between 2000 and 4000 Hz.
  • Sample concentrations for two-dimensional spectra were 2 mM in CD 2 C1 2 and 8 mM in CD 3 OD and CD 3 OH.
  • the ⁇ -amino acids used to assemble the peptides described herein can be manufactured using several different literature methods, as well as new methods described below.
  • the Arndt-Eisterdt homologation reaction can be used, see Reaction 1. See also Seebach et al. (1996) Helv. Chim. Acta 79:913. This route has advantages and disadvantages.
  • a distinct advantage is that the starting materials, ⁇ -amino acids, are readily available commercially in enantiomerically pure form.
  • R 1 and R 2 are aliphatic substituents.
  • ⁇ -Amino acids containing an unsubstituted cycloalkyl moiety involving the ⁇ and ⁇ carbons were synthesized using literature methods. See, for example, Nohira et al. (1970) Bull. Chem. Soc. Jpn. 43:2230; Herradon and Seebach (1989) Helv. Chim. Acta 72:690-714; and Tilley et al. (1992) J. Med. Chem. 35:3774-3783, all three of which are incorporated herein by reference.
  • cyclohexyl-containing ⁇ -amino acids can be synthesized via Reaction 2:
  • Methyl (15,6R)-6-benzyloxycarbonyIaminocyclohex-3-ene carboxylate (24) Ethylchloroforamate (4 mL, 0.042 mol) was added to a mixture of 23 (5.14 g, 0.028 mol) and triethylamine (6 mL, 0.043 mol) in acetone (100 mL) at 0° C and vigorously stirred for 10 min. An aqueous solution of NaN 3 (3.04 g, 0.047 mol, in 25 mL water) was added in one portion. The resulting mixture was stirred for 30 min at 0° C. The reaction mixture was diluted with water and extracted with diethyl ether.
  • Methyl (lS,6R)-6-te/f-butoxycarbonylaminocyclohexane carboxylate (25) The yellow oil from the previous reaction, which contains compound 24 (5.6 g, 0.020 mol) and benzyl alcohol, was dissolved in methanol. 0.525 g of 10% Pd on carbon was added to the methanol solution, and the heterogenous mixture was placed under 50 psi H 2 and shaken at rt for 24 h. The mixture was filtered through celite, and the filtrate was concentrated to yield 13.74 g of dark golden yellow liquid. 25 mLof IM HCl was added to the filtrate, and the benzyl alcohol was extracted with diethyl ether (3 x 25 mL).
  • the pH of the aqueous solution was adjusted to 9 using K 2 C0 3 .
  • 25 mL of dioxane and Boc 2 O (5 g, 0.023 mol) were added to the solution, and the reaction was stirred at rt for 20 h.
  • 15 mL of water was added and the solution was extracted with ethyl acetate (3 x 50 mL).
  • the combined organic extracts were dried over anhydrous magnesium sulfate and concentrated.
  • Residue was purified via column chromatography (SiO 2 , eluting with 6: 1 Hex:EtOAc), to yield 2.00 g viscous clear oil.
  • Methyl (IR,6R)-6-te/*-butoxycarbonylaminocyclohexane carboxylate (26) Sodium metal (0.14 g, 6.1 mmol) was placed into a flame dried flask under nitrogen atmosphere and cooled to 0° C. 10 mL of freshly distilled methanol was added and the mixture stirred until all the sodium dissolved. An amount of 25 (2.00 g, 7.7 mmol) was dissolved in 10 mL of freshly distilled methanol and transferred to NaOMe solution via cannula. The solution was refluxed under nitrogen for 5.5 h, cooled to rt and acidified with 0.5 M aqueous 0.5 M ammonium chloride (18 mL, 9 mmol).
  • the 4-position amino substituent is protected by a Boc group and the 1 -position amino substituent is protected by a Cbz group.
  • the starting material is available commercially (Aldrich Chemical Co. , Milwaukee, Wisconsin).
  • Reaction 4 details an illustrative synthesis of a ⁇ -proline wherein the exocyclic amino substituent is in the 3-position relative to the ring nitrogen.
  • Compound 42 Tap water (200 ml) and baker's yeast (25 g) were mixed, and were shaken on an orbital shaker for 1 hour. Compound 41 (1.0 g) was then added. The mixture was shaken at room temperature for 24 hours. The mixture was filtered through a bed of Celite. The Celite was washed with water (20 ml). The filtrate was extracted with diethyl ether (5 x 100 ml). The extracts were washed with water (2 x 50 ml), dried over MgS0 4 , and concentrated to yield a slightly yellow oil. The crude product was purified by column chromatography with ethyl acetate/hexane (1/1, v/v) as eluent to give a colorless oil (0.5 g) in 50% yield.
  • Compound 44 Compound 43 (1.1 g) was dissolved in methanol (50 ml) . SnCl 2 (2.2 g) was then added. The mixture was stirred at room temperature for 30 hours. The methanol was then removed under reduced pressure. The residue was dissolved in Baker's Yeast Tap Water oc
  • Fmoc-OSU 9-FIuorenylmethyIoxycarbonyl-N-hydroxysuccinimide
  • Compound 45 Compound 44 (1.35 g) was dissolved in methanol/ water (3/1, v/v, 80 ml), cooled to 0° C. LiOH.H20 (1.68 g) was added. The mixture was stirred at 0° C for 24 hours, by which time TLC indicated that the hydrolysis was complete. Saturated ammonium hydroxide (20 ml) was added. The methanol was removed under reduced pressure. The aqueous was washed with diethyl ether (50 ml), acidified with IN HCl to pH 3, extracted with methylene chloride (3 x 150 ml). The extracts were washed with dilute brine (50 ml), dried over MgSO 4 , concentrated to give a sticky colorless residue (1.25 g, 99%), which was used directly without further purification.
  • Compound 46 Compound 45 (1.25 g) was dissolved in methanol (50 ml) in a hydrogenation flask. 5 % Palladium on activated carbon (190 mg) was added. The flask was pressurized with hydrogen to 35 psi, rocked at room temperature for 7 hours, by which time TLC indicated that the hydrogenolysis was complete. The Pd/C was removed by filtration. The filtrate was concentrated to give a white solid. The white solid was dissolved in acetone/water (2/1, v/v, 70 ml), cooled to 0° C. NaHCO 3 (1.7 g) was added, followed by FMOC-OSU (1.39 g). The reaction mixture was stirred at room temperature for 16 hours.
  • Reaction 5 illustrates the synthesis of a ⁇ -amino acid wherein the exocyclic amino substituent the nitrogen heteroatom is in the 4-position relative to the ring nitrogen.
  • Compound 52 Compound 51 (2.0 g) and NaBH 3 CN (0.54 g) were dissolved in methanol (40 ml), IN HCl (aqueous) was added dropwise to maintain pH 3-4. After 15-20 minutes, pH change slowed. The mixture was stirred for an additional 1.0 hour, while IN HCl was added occasionally to keep pH 3-4. Water (100 ml) was added. The mixture was extracted diethyl ether (3 x 150 ml). The extracts were washedwith IN NaHC03 (100 ml) and dilute brine (100 ml), dried over MgS0 4 , and concentrated to give a colorless oil (1.9 g) in 95 % yield. The product was used directly without further purification.
  • Compound 53 Compound 52 (1.9 g) and Ph 3 P (2.8 g) were dissolved in toluene (anhydrous, 30 ml) under nitrogen. A solution of diethyl azodicarboxylate (1.5 ml) in toluene (10 ml) was subsequenely introduced via syringe over 15 minutes. The reaction mixture was stirred under nitrogen at room temperature for 12 hours. The toluene was removed under reduced pressure. The residue was purified by column chromatography with ethyl acetate/hexane (3/7, v/v) as eluent to afford a colorless oil (1.6 g) in 91 % yield.
  • Compound 54 Compound 53 (1.0 g) and R-(+)- ⁇ -methylbenzylamine (l. l ml) were mixed with water (15 ml). The mixture was stirred at 55° C for 67 hours. The mixture was taken up in diethyl ether (300 ml), and the aqueous layer was separated. The ether solution was washed with water (3 x 50 ml), dried over MgSO 4 , and concentrated to give a slight yellow oil. The diastereometic isomers were separated by column chromatography with ethyl acetate/hexane (2/8, v/v) as eluent to give RSS (0.2 g) and RRR (0.34 g) in 51 % overall yield.
  • Compound 56 Compound 55 (4.6 g) was dissolved in 95 % ethanol (150 ml) in a hydrogenation flask. 10% Palladium on activated carbon (0.5 g) was added. The flask was pressurized with hydrogen to 50 psi, rocked at room temperature for 22 hours, by which time NMR spectroscopy indicated that the hydrogenolysis was complete. The Pd/C was removed by filtration. The filtrate was concentrated to give a white solid. The white solid was dissolved in acetone/water (2/1 , v/v, 150 ml). NaHCOj (9.7 g) was added, followed by Cbz-OSU (3.4 g). The reaction mixture was stirred at room temperature for 14 hours.
  • Compound 57 Compound 56 (2.0 g) was dissolved in methanol/water (3/1 , v/v, 115 ml), cooled to 0° C, LiOH.H20 (2.4 g) was added. The mixture was stirred at O° C for 15 hours, by which time TLC indicated that the hydrolysis was complete. Saturated ammonium hydroxide (aqueous, 100 ml) was added. The methanol was removed under reduced pressure. The aqueous was acidified with IN HCl to pH 3, extracted with ethyl acetate (3 x 200 ml). The extracts were washed with dilute brine (100 ml), dried over MgSO 4 , concentrated to give a foamy solid (1.63 g, 88%), which was used directly without further purification).
  • Compound 58 Compound 57 (1.63 g) was dissolved in methanol (70 ml) in a hydrogenation flask. 5 % Palladium on activated carbon (250 mg) was added. The flask was pressurized with hydrogen to 35 psi, rocked at room temperature for 15 hours, by which time NMR spectroscopy indicated that the hydrogenolysis was complete. The Pd/C was removed by filtration. The filtrate was concentrated to ive a white solid. The white solid was dissolved in acetone/water (2/1 , v/v, 90 ml), cooled to O° C. NaHCO 3 (2.27 g) was added, followed by FMOC-OSU (1.83 g).
  • the reaction mixture was stirred at O° C for 2 hours, then at room temperature for 28 hours. Water (50 ml) was added. The acetone was removed under reduced pressure. The aqueous was acidified with IN HCl to pH 3, extracted with ethyl acetate (3 x 200 ml). The extracts were washed with dilute brine (100 ml), dried over MgSO 4 , concentrated to give a foamy white solid. The crude white solid was purified by column chromatography with methanolfethyl acetate (3/7, v/v) as eluent to give the clean product as a white solid (1.68 g) in 84% yield.
  • (2S,3R)-3-Amino-2-methylpentanoic acid was prepared according to the procedures given by Jefford and McNulty (1994), J. Helv. Chim. Acta 77:2142.
  • the synthesized (2S,3S)-2-methyl-3- (tosylamino)butano-4-lactone contained up to 8 % (2R,3S)-2-methyl-3- (tosylamino)butano-4-lactone as a byproduct, which could be removed by recrystallization from toluene.
  • (2S,3S)-3-Amino-2-benzyl-4-phenylthiobutanoic acid was prepared in a synthetic sequence derived from the one by Jefford and McNulty. This synthesis is described below. Homo- ⁇ -amino acids were prepared according to the procedures by Podlech and Seebach (1995), Liebigs Ann. 1217.
  • Depsi- ⁇ -peptides were synthesized by conventional dicyclohexylcarbodiimide / N-hydroxysuccinimide (DCC/HOSu) or l-ethyl-3-(3'dimethylaminopropyl)carbodiimidde hydrochloride / N,N-dimethyl-4-aminopyridine (EDCI/DMAP) solution-phase coupling procedures (see, for example, Bodanszky, M. ; Bodanszky, A. The Practice of Peptide Synthesis; Springer Verlag: New York, 1984). Illustrative procedures are given below.
  • a solution of lithium diisopropylamine (LDA) in THF was generated by adding 1.5 M methyllithium in diethyl ether (30 mL, 45.0 mmol) to a solution of diisopropylamine (6.4 mL, 45.7 mmol) in 100 mL THF at O°C under nitrogen and stirring for 10 min. The solution was then cooled to -78°C, and a solution of (3S)-3-(tosylamino)butano-4-lactone (5.36 g, 21.1 mmol) in 30 mL THF was added dropwise.
  • LDA lithium diisopropylamine
  • (2S,3S)-2-Benzyl-4-phenylthio-3-(tosylamino)butanoic acid (7) (2S,3S)-2-Benzyl-3-(tosylamino)butano-4-lactone (4) (0.91 g, 2.64 mmol) was dissolved in 10 mL methylene chloride. At O°C trimethylsilyliodide (1 mL, 7.03 mmol) and anhydrous ethanol (0.72 mL, 12.2 mmol) were added under nitrogen. The solution was stirred 30 min. at 0°C, allowed to warm to room temperature and stirred for 1 day.
  • thiophenol (0.73 ml, 7.11 mmol) was added to a suspension of NaH (289.7 mg, 7.24 mmol) in 6 mL DMF under nitrogen, warmed to room temperature and stirred for 15 min.
  • a solution of crude 5 (1.78 g) in 10 mL DMF was added to the thiophenolate solution at O°C. After warming to room temperature the solution was stirred for 1 hour.
  • the reaction was quenched with 50 ml water and extracted three times with methylene chloride. The combined organic extracts were washed with brine, dried over Na 2 SO 4 and concentrated in vacuo to give 2.43 g of 6 as a colorless oil, which was used in the next step without further purification.
  • (2S,3S)-3-Amino-2-Benzyl-4-phenylthiobutanoic acid (8) Compound 7 and phenol (0.77g) were dissolved in 50 mL 48% HBr and heated to reflux for 1.5 hours under nitrogen. After cooling to room temperature 150 mL water was added and the solution extracted two times with diethyl ether. The yellow aqueous layer was evaporated to give 0.58 g of (2S,3S)-3-amino-2-benzyl-4-phenylthiobutanoic acid hydrobromide as an orange solid.
  • the deprotected amide and triethylamine (85 ⁇ l, 0.610 mmol) were dissolved in 1 mL DMF and transferred into the activated ester solution. After stirring the resulting solution for 2 days at room temperature, the solvent was removed in a stream of nitrogen and the residue dried in vacuo. The residue was separated by chromatography (silica gel, CHCl 3 /MeOH 4: 1) to yield impure 11 (192.7 mg), which was used in the next step without further purification.
  • Methyl-3-(t-butoxycarbonylamino)propionic amide 14
  • BOC- ⁇ -alanine (0.50 g, 2.64 mmol) was dissolved in 4 mL DMF.
  • Methylamine hydrochloride (198 mg, 2.93 mmol) and DMAP (427.2 mg, 3.50 mmol) were added, followed by EDCI (1.06 g, 5.53 mmol).
  • EDCI (1.06 g, 5.53 mmol
  • Compound 16 Compound 14 (0.33 g, 1.63 mmol) was dissolved in 5 mL of 4 M HCl/dioxane, and the solution was stirred at 12° C for 1 hour. The HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo.
  • An activated ester solution was prepared by adding DCC (509.9 mg, 2.47 mmol) to a solution of glycolic acid (145.7 mg, 1.92 mmol) and HOSu (326.4 mg, 2.84 mmol) in 10 mL methylene chloride. A white precipitate formed after a few minutes. The suspension was stirred at 12°C for 6 hours.
  • Compound 21 Compound 21.
  • Compound 20 (52.1 mg, 0. 140 mmol) was dissolved in 1 mL 4 M HCl/dioxane and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo.
  • the deprotected depsipeptide and BOC-homophenylalanine (42.5 mg, 0. 152 mmol) were dissolved in 5 mL methylene chloride.
  • DMAP (32.4 mg, 0.265 mmol) was added, followed by EDCI (59.4 mg, 0.310 mmol). After stirring at room temperature for 2 days the solvent was removed in a stream of nitrogen.
  • polypeptides using any type of ⁇ -amino acid can be accomplished using conventional and widely recognized solid-phase or solution- phase synthesis.
  • solid-phase synthesis the desired C-terminal amino acid residue is linked to a polystyrene support as a benzyl ester.
  • the amino group of each subsequent amino acid to be added to the N-terminus of the growing peptide chain is protected with Boc, Fmoc, or another suitable protecting group.
  • the carboxylic acid group of each subsequent amino acid to be added to the chain is activated with DCC and reacted so that the N-terminus of the growing chain always bears a removable protecting group.
  • the process is repeated (with much rinsing of the beads between each step) until the desired polypeptide is completed.
  • the N-terminus of the growing chain is protected with a Boc group, which is removed using trifluoracetic acid, leaving behind a protonated amino group.
  • Triethylamine is used to remove the proton from the N- terminus of the chain, leaving a the free amino group, which is then reacted with the activated carboxylic acid group from a new protected amino acid.
  • a strong acid such as hydrogen bromide in trifluoracetic acid, is used to both cleave the C-terminus from the polystyrene support and to remove the N-terminus protecting group.
  • AA n incoming amino acid to be added to chain
  • Fmoc the protecting group 9-fluorenylmethyloxycarbonyl
  • NMP N-methyl pyrrolidone
  • HOBt N-hydroxy-benzotriazole
  • DIEA diisopropylethyl amine
  • Solution phase synthesis can also be used with equal success.
  • solution-phase synthesis of a ⁇ -peptide chain containing alternating residues of unsubstituted cyclohexane rings and amino-substituted cyclohexane rings proceeds in conventional fashion as outlined in Reaction 8:
  • REACTION 8 Reaction 8 works with equal success to build peptides wherein the residues are the same or different.
  • Reaction 9 is an illustration of a homologation reaction combined with conventional solution-phase peptide synthesis which yields a ⁇ -peptide having acyclic-substituted residues alternating with ring-constrained residues:
  • the ⁇ -peptides of the present invention can be substituted with any number of substituents, including hydroxy, linear or branched C r C 6 -alkyl, alkenyl, alkynyl; hydroxy-C,-C 6 -alkyl, amino-C ⁇ -alkyl, C r C 6 -alkyloxy, C,-C 6 - alkyloxy-C j -C 6 -alkyl, amino, mono- or di-C,-C 6 -alkylamino, carboxamido, carboxamido-C,-C 6 -alkyl, sulfonamido, sulfonamido-C,-C 6 -alkyl, urea, cyano, fluoro, thio, - -alkyithio, mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and
  • appending a sulfonamido moiety to the cylic backbone substituent can be accomplished in conventional fashion using Reaction 10.
  • Methanesulfonylchloride (71 ⁇ l) was added dropwise. After the addition, the reaction mixture was stirred at room temperature for 12 hours. The pyridine was then removed in vacuo. The residue was taken up in ethyl acetate (50 ml). The mixture was washed with dilute brine (2 x 10 ml), dried over MgSO 4 , and concentrated to give the clean product as a colorless oil (70 mg) in 82% yield.
  • Compound 64 Compound 62 (30 mg) was dissolved in 4 N HCl in dioxane (2.0 ml). The reaction mixture was stirred for 1.5 hours. The dioxane was then removed in vacuo. The residue was dissolved in pyridine (1.0 ml), then cooled to O° C in an ice-bath. Toluenesulfonylchloride (63 mg) was added in portions. After the addition, the reaction mixture was stirred at room temperature for 12 tours. The pyridine was then removed in vacuo. The residue was taken up in methylene chloride/dithyl ether (1/1, v/v, 100 ml).
  • compositions containing the ⁇ -peptide compounds described herein are selectively antimicrobial.
  • the compounds exhibit good antimicrobial activity, both MIC and MBC, while simultaneously exhibiting very low hemolytic activity. Consequently, the subject compounds are useful in the formulation of pharmaceutical compositions for the treatment microbial infections because they inhibit microbial growth at low concentrations without lysing human cells.
  • the compounds described herein being effective to inhibit the growth of microbial cells, the compounds are suitable for the therapeutic treatment of microbial infestation (infection, etc.) in mammals, including humans.
  • Microbial cell growth inhibition at pharmacologically-acceptable concentrations has been shown in four different types of bacteria (see Examples 10 and 11).
  • Administration of the ⁇ -peptides to a human or non-human patient can be accomplished by any means known.
  • the preferred administration route is parenteral, including intravenous administration, intraarterial administration, intratumor administration, intramuscular administration, intraperitoneal administration, and subcutaneous administration in combination with a pharmaceutical carrier suitable for the chosen administration route.
  • the treatment method is also amenable to oral administration.
  • the concentration or amount of the ⁇ -peptide administered will vary depending upon the severity of the ailment being treated, the mode of administration, the condition and age of the subject being treated, and the particular ⁇ -peptide or combination of ⁇ -peptides being used.
  • the compounds can be administered in the form of tablets, pills, powder mixtures, capsules, injectables, solutions, suppositories, emulsions, dispersions, food premixes, and in other suitable forms.
  • the pharmaceutical dosage form which contains the compounds described herein is conveniently admixed with a non-toxic pharmaceutical organic carrier or a non-toxic pharmaceutical inorganic carrier.
  • Typical pharmaceutically-acceptable carriers include, for example, mannitol, urea, dextrans, lactose, potato and maize starches, magnesium stearate, talc, vegetable oils, polyalkylene glycols, ethyl cellulose, poly(vinylpyrrolidone), calcium carbonate, ethyl oleate, isopropyl myristate, benzyl benzoate, sodium carbonate, gelatin, potassium carbonate, silicic acid, and other conventionally employed acceptable carriers.
  • the pharmaceutical dosage form may also contain non-toxic auxiliary substances such as emulsifying, preserving, or wetting agents, and the like.
  • Solid forms such as tablets, capsules and powders, can be fabricated using conventional tabletting and capsule-filling machinery, which is well known in the art.
  • Solid dosage forms may contain any number of additional non-active ingredients known to the art, including excipients, lubricants, dessicants, binders, colorants, disintegrating agents, dry flow modifiers, preservatives, and the like.
  • Liquid forms for ingestion can be formulated using known liquid carriers, including aqueous and non-aqueous carriers, suspensions, oil-in-water and/or water- in-oil emulsions, and the like.
  • Liquid formulation may also contain any number of additional non-active ingredients, including colorants, fragrance, flavorings, viscosity modifiers, preservatives, stabilizers, and the like.
  • the subject compounds may be administered as injectable dosages of a solution or suspension of the compound in a physiologically- acceptable diluent or sterile liquid carrier such as water or oil, with or without additional surfactants or adjuvants.
  • a physiologically- acceptable diluent or sterile liquid carrier such as water or oil
  • carrier oils would include animal and vegetable oils (peanut oil, soy bean oil), petroleum-derived oils (mineral oil), and synthetic oils.
  • water, saline, aqueous dextrose and related sugar solutions, and ethanol and glycol solutions such as propylene glycol or polyethylene glycol are preferred liquid carriers.
  • the pharmaceutical unit dosage chosen is preferably fabricated and administered to provide a concentration of drug at the point of contact with the microbial cell of from 1 ⁇ M to 10 mM. More preferred is a concentration of from 1 to 100 ⁇ M. This concentration will, of course, depend on the chosen route of administration and the mass of the subject being treated.
  • Amide proton exchange is one of the most powerful methods for assessing conformational stability of peptides and proteins; adoption of a stable intramolecularly hydrogen-bonded conformation leads to diminution of the rate of exchange.
  • NH/ND exchange behavior of the trans-ACHC hexamer relative to the corresponding dimer shows that the hexamer adopts a very stable intramolecularly hydrogen- bonded folding pattern in methanol solution.
  • this Example was conducted with solutions containing 2 mM of the trans-ACHC dimer and 2 mM of the trans-ACHC hexamer.
  • the 1H NMR results are shown in Fig. 3.
  • the amide proton and the urethane proton of the dimer are completely exchanged within 6 min, according to 'H NMR (Fig. 3).
  • three of the six amide protons of the trans-ACHC hexamer show strong resonances at this point.
  • trans-ACHC/ 14-helix combination would generate a stable ⁇ -peptide secondary structure.
  • the trans- ACPCl 12-helix combination was predicted to be almost as favorable. This latter prediction is interesting because there is no precedent for the 12-helix in the contradictory literature on polymers constructed from optically active ⁇ -amino acids. Among these polymers, poly( ⁇ -isobutyl-L-aspartate) has been particularly intensively studied, and proposed secondary structures include 14-, 16-, 18- and 20-helix, as well as sheet. Since the computational predictions regarding the trans-ACHC/ 14-helix relationship described above proved to be correct, the trans- ACVCI 12-helix prediction was then explored.
  • Optically active trans- ACPC was prepared using the protocols described above, and ⁇ -peptides were generated via standard coupling methods.
  • a trans - ACPC octamer displays the predicted 12-helical conformation in the solid state; all six of the possible 12-membered ring hydrogen bonds are formed.
  • a tran - ACPC hexamer also displays the predicted 12-helical conformation in the solid state, with all four of the possible 12-membered ring hydrogen bonds formed. In both cases, the regular helix frays at the C-terminus, perhaps because the C-terminal ester group cannot serve as a hydrogen bond donor.
  • Circular dichroism data for trans- ACPC hexamer in CH 3 OH indicates the adoption of a distinctive secondary structure.
  • the main graph in Fig. 4 depicts two virtually superimposable CD plots: one at a hexamer concentration of 2.0 mM, the other at hexamer concentration of 0.1 mM. Data were obtained on a Jasco J-715 instrument at 20°C using a 1 mm pathlength.
  • the inset graph shows the CD data at 0.1 mM and 0.02 mM using a 5 mm pathlength.
  • the 14-helical conformation in ⁇ -peptides composed of acyclic residues has a far-UV CD signature comprising a maximum at ca.
  • the CD signature for the trans-ACPC hexamer is clearly different: maximum at ca. 204 nm, zero crossing at ca. 214 nm, and minimum at ca. 221 nm. Since the CD signature of trans- ACPC hexamer in CH 3 OH does not vary significantly between 2.0 mM and 0.02 mM, it is unlikely that aggregation occurs under these conditions.
  • the C-terminal residue could be identified because it had the only C ⁇ H that did not show an NOE to an NH of another residue. Assignment of these terminal resonances allowed us to "walk through" the remaining backbone resonances by virtue of short-range C ⁇ H, - NH, +7 NOEs.
  • the secondary structure of the trans-ACPC hexamer in pyridine-d s is defined by the long-range NOEs summarized in Table 1, below.
  • C p H,- ⁇ NH, +2 and C p H, - C ⁇ H /+2 NOEs are expected for the 12-helix. All four possible C p H, - NH /+2 NOEs were observed for the ACPC hexamer, as were two of the four C ⁇ H/ - C ⁇ H /+2 NOEs; NOEs consistent with the other two C p H, - C ⁇ H, +2 interactions were observed but could not be unambiguously assigned because of overlap of the C ⁇ H resonances of residues 3 and 4.
  • Fig. 5 shows CD data for the dimer, trimer, tetramer, and hexamer of trans- ACPC in methanol.
  • a comparison between the dimer and the hexamer indicates the profound change in secondary structure which takes place as the peptide chain increases in length.
  • the dimer is essentially unstructured.
  • the tetramer clearly displays a CD curve indicative of 12-helical secondary structure.
  • This Example shows that short ⁇ -peptides of trans-ACPC have a high propensity to adopt the 12-helical folding pattern.
  • amino-substituted-trans-ACHA i.e. , ACHA containing an exocyclic amino substituent
  • the amino-substituted-trans-ACHA was then coupled with unsubstituted trans-ACHA as shown in Reaction 7 to yield ⁇ -peptides wherein the residues alternate between unsubstituted-trans-ACHA and amino-substituted-trans-ACHA.
  • These molecules were synthesized because it was anticipated that the amino group would be protonated in water and that the resulting positive charge would render these ⁇ - peptides water-soluble. They are indeed water-soluble.
  • Fig. 6 depicts the two-dimensional *H NMR data obtained for the alternating unsubstituted-rrans-ACHA/amino-substituted-trans-ACHA hexamer in D 2 0, 100 mM deuteroacetate buffer, pD 3.9.
  • Hydrogen/ deuterium exchange can be examined at five of the six backbone NH groups in this acidic D 2 0 solution. The spectra were taken at room temperature. Five NH peaks are observed (marked with asterisks) in the 8 minute plot. Each peak disappears at a different rate over the course of two days as the NH groups become ND groups.
  • the protons slowest to exchange are those hydrogen bonded and buried in a folded secondary conformation.
  • the CD data from the alternating hexamer described in Example 3 was compared with a hexamer of unsubstituted trans-ACHC.
  • the trans- ACHC hexamer adopts a 14-helical conformation in methanol solution. Therefore, the CD plot for trans-ACHC should be representational of the 14-helix structure and can serve as a means of comparison for other peptides.
  • Fig. 8 compares the CD data of trans-ACHC hexamer in methanol with the CD data of the alternating unsubstituted-trans- ACHA/amino-substituted-trans- ACHA described in Example 3.
  • Examples 3 and 4 in combination show that in addition to the substantial peptide folding indicated by the slow amide proton exchange rate of the alternating hexamer, the CD data strongly indicates that the alternating hexamer adopts a 14- helix secondary structure in aqueous solution.
  • Example 5 CD Data of ⁇ -Peptides Containing Alternating fr ⁇ ns-ACPA Residues and 3-Amino-4-Carboxy-PyrrolidinyI Residues
  • Fig. 10 shows another plot of CD data, in this instance using water as the solvent. Again, the maximum and minimum characteristic peaks increase in intensity with increasing chain length.
  • Fig. 11 is a direct comparison of the CD data in water versus the CD data in methanol for the alternating 4-pyrrolidinyl/trans-ACPA octamer. These data indicate that the 12-helix formation is only slightly less stable in water than it is in methanol.
  • Example 6 CD Data of Octamer Containing Alternating frans-ACPA Residues and 3-Amino-2-Carboxy-Pyrrolidinyl residues.
  • This Example shows that the nitrogen heteroatom introduced in the pyrrolidinyl moiety can be located in different postions along the heterocyclic ring without adversely affecting the formation of the 12-helical structure in solution.
  • Example 7 Comparison of Secondary Structure in Amino-Subsituted-trans- ACHC and ⁇ -Peptides Containing Aliphatic-Substituted Residues
  • the CD spectrum of a ⁇ -peptide containing alternating residues of trans-ACHA and amino-substituted-trans-ACHA was compared to the CD spectrum of a "mixed" ⁇ -peptide comprising alternating residues of trans- ACHA and an acyclic ⁇ -amino acid bearing an aminopropyl substituent on the ⁇ - carbon of the backbone.
  • the acyclic ⁇ -amino acid was synthesized and coupled to the trans-ACHA residue as detailed in Reaction 9, described above.
  • Example 8 Infrared Analysis of Reverse Turn Using Linked Nipecotic Acid Moieties
  • Fig. 14A shows the IR spectrum for the diastereomer in which the two nipecotic acid residues have the same absolute configuration
  • Fig. 14B shows the IR spectrum for the diastereomer in which the two nipecotic acid residues have opposite absolute configuration. Both samples were taken in dilute solution (to minimize intermolecular hydrogen bonding) with solvent subtraction. As is clear from the diastereomer shown in Fig. 14A, there is little intramolecular hydrogen bonding as evidenced by the large peak at 3454 cm '1 which is due to N-H units not involved in hydrogen bonding. This was as predicted from computer modeling studies.
  • Example 9 X-ray Crystallography of Synthetic Reverse Turn
  • Compound 1 was synthesized as described above. Crystals of compound 1 suitable for X-ray analysis were obtained by vapor diffusion (over 2 weeks) of n- heptane into a solution of the sample in ethyl acetate. The data were collected on a Siemens P4/CCD diffractometer running software provided by the manufacturer. The ball and stick schematic of the crystal stracture in the solid state is presented in Fig. 16. All hydrogens except those bonded to nitrogen have been removed for clarity. Hydrogen bonds are shown in dotted lines.
  • ⁇ -polypeptides described herein make these polyamide compounds highly useful for constructing large libraries of potentially useful compounds via combinatorial chemistry.
  • equatorial positions of the cyclohexyl ⁇ - peptides can be substituted with virtually any substituent, including very large substituents, without disrupting the helical secondary structure. At least in helical structures, this is because any equatorial substituent extends essentially perpendicular from the axis of rotation of the helix, thereby leaving the hydrogen bonds of the helix undisturbed.
  • amino acids which comprise the finished peptides can be functionalized prior to being incorporated into a polypeptide, or an unfunctionalized polypeptie can be constructed and then the entire oligomer functionalized. Neither method is preferred over the other as they are complementary depending upon the types of compounds which are desired.
  • Combinatorial libraries utilizing the present compounds may be constructed using any means now known to the art or developed in the future.
  • the preferred methods are the "split and pool” method using solid-phase polypeptide synthesis on inert solid substrates and parallel synthesis, also referred to as multipin synthesis.
  • the substrates are generally small diameter beads, e.g. about 100 ⁇ m, formed from inert polymeric materials such as polyoxyethylene-grafted polystyrene or polydimethylacrylamide.
  • An illustrative substrate, marketed under the trademark "ARGOGEL” is available from Argonaut Technologies, Washington, D.C.
  • a plurality of inert substrates are divided into two or more groups and then a first set of subunits is covalently linked to the inert support.
  • the initial plurality of substrates is divided into three subgroups.
  • the appearance of the three groups of beads after the first round of coupling is shown at I of Fig. 15.
  • the three groups of beads are then pooled together to randomize the beads.
  • the beads are then again split into a number of subgroups.
  • Another round of coupling then takes place wherein a second subunit is bonded to the first subunit already present on each bead.
  • the process is then repeated (theoretically ad infinitum) until the desired chain length is attained.
  • the split and pool process is highly flexible and has the capability of generating literally millions of different compounds which, in certain applications, can be assayed for activity while still attached to the inert substrate.
  • a critical aspect of the split and pool methodology is that each reaction be driven to completion to prior to initiating a subsequent round of coupling. So long as each coupling reaction is driven to completion, each substrate bead will only display a single compound. Because the rate of reaction will differ from bead to bead as the library construction progresses, the beads can be monitored using conventional dyes to ensure that coupling is completed prior to initiating another round of synthesis. The presence of only a single compound per bead comes about because each individual bead encounters only one amino acid at each coupling cycle. So long as the coupling cycle is driven to completion, all available coupling sites on each bead will be reacted during each cycle and therefore only one type of peptide will be displayed on each bead.
  • the resulting combinatorial library is comprised of a plurality of inert , substrates, each having covalently linked thereto a different ⁇ -polypeptide.
  • the polypeptides can be screened for activity while still attached to the inert support, if so desired and feasible for the activity being investigated. Beads which display the desired activity are then isolated and the polypeptide contained thereon characterized via conventional peptide chemistry, such as the Edman degradation. Where a solution-phase assay is to be used to screen the library, the polypeptides are cleaved from the solid substrate and tested in solution.
  • one or more of the subunits coupled to the inert substrate are selected from the ⁇ -amino acids described herein. In this fashion, large libraries of ⁇ -polypeptides can be assembled, all of compounds contained therein which display predictable secondary structure.
  • a known set of first subunits is covalently linked to a known location on a inert substrate, one subunit type to each location.
  • the substrate may be a series of spots on a suitable divisible substrate such as filter paper or cotton.
  • a substrate commonly used is an array of pins, each pin being manufactured from a suitable resin, described above. After the initial round of coupling, each pin of the array bears a first subunit covalently linked thereto. The array is then reacted with a known set of second subunits, generally different from the first, followed by reactions with a third set of subunits, and so on.
  • each individual pin (or location) is coupled with a incoming subunit selected from a distinct set of subunits, with the order of the subunits being recorded at each step.
  • the final result is an array of polypeptides, with a different polypeptide bonded to each solid substrate. Because the ordering of the subunits is recorded, the identity of the primary sequence of the polypeptide at any given location on the substrate (i.e. , any given pin) is known.
  • each coupling reaction must be driven to completion in order to ensure that each location on the substrate contains only a single type of polypeptide.
  • Another use for the present compounds is as molecular probes to investigate the interactions between biological macromolecules to identify antagonists, agonists, and inhibitors of selected biological reactions.
  • many biological reactions take place between very large macromolecules.
  • the surface areas in which these reactions take place are thought by many to be far too large to be disrupted, altered, or mimicked by a small molecule.
  • it has been difficult, if not impossible, to manufacture molecular probes of modest size that display a well-defined conformation. Because the compounds described herein assume a highly predictable helical or sheet conformation, even when functionalized, they find use as reagents to probe the interaction between large biomolecules.
  • This secondary structure is referred to herein as the " 12-helix.
  • the ⁇ -peptide 12-helix has approximately 2.5 residues per turn.
  • APC (R,R)-rr ⁇ ns-4-aminopyrrolidine-3-carboxylic acid
  • Each ⁇ -peptide bears an acetyl capping group at the N-terminus and a primary amide at the C-terminus.
  • These ⁇ -peptides were designed with the intent that they would form amphiphilic 12-helices in aqueous solution, with all cationic APC residues on one side of the helix and all hydrophobic ACPC residues on the other side.
  • Staphylococcus aureus 1206 (Weisblum et al , Erythromycin-inducible resistance in Staphylococcus aurens: survey of antibiotic cases involved, /. BacterioL , 98:447-52 (1969)) are clinical isolates, while Bacillus subtilis BR151 (Young et al , Chromosomal location of genes regulating resistance to bacteriophage in Bacillus subtilis, J. BacterioL, 98:2087-97 (1969)) and Esche ⁇ chia coli JM109 (Yanisch-Perron et al.
  • M13 phage cloning vectors and host strains nucleotide sequences of the M13mpl8 and pUC19 vectors, Gene 33: 103-19 (1985)
  • M13mpl8 and pUC19 vectors are non-pathogenic strains commonly used in the laboratory for genetic constructions.
  • B. subtilis itself shows little tendency to colonize humans, a related species, B. anthracis, causes anthrax.
  • ⁇ -17 is potent against all four bacteria, and this ⁇ - peptide matches or exceeds the magainin in all four cases.
  • the sizes of ⁇ -17 and the magainin are similar: there are 64 carbon and nitrogen atoms in the backbone of ⁇ -17 and 72 carbon and nitrogen atoms in the backbone of the magainin. If the 23- residue magainin were fully folded into an ⁇ -helix (3.6 residues per turn), this molecule would form roughly 6.4 helical turns and have a length of roughly 34 A. If the 17-residue ⁇ -peptide were fully folded into a 12-helix, the molecule would form roughly 6.8 helical turns and have a length of roughly 37 A.
  • Example 11 Therapeutic utility requires that a cationic peptide act selectively against bacterial cells in the presence of human cells. This issue is typically probed by comparing red blood cell lytic activity ("hemolysis") with antimicrobial activity. Magainins display low hemolytic activity, but other natural cationic helix-forming peptides are highly hemolytic. For example, the 26-residue peptide melittin, isolated from honey bee venom, readily lyses both bacterial and human red blood cells. Fig. 17 shows the results of a direct hemolysis comparison between melittin (a positive control), the magainin derivative, and ⁇ -17.
  • hemolysis red blood cell lytic activity
  • Fig. 17 shows the results of a direct hemolysis comparison between melittin (a positive control), the magainin derivative, and ⁇ -17.
  • human red blood cells (1 % suspension in PBS) were incubated at room temperature for 1 h with a two-fold serial dilution of peptide in PBS . Release of hemoglobin was measured by determining the OD 415 of the supernatant after centrifugation. Controls consisted of human red blood cells suspended in PBS (zero hemolysis) and suspended in 1 % SDS (100% hemolysis).
  • Minimal inhibitory concentration is defined here as the lowest concentration of peptide required for complete inhibition of growth as determined by optical density at 590 nm.
  • Bacterium in BHI medium ( ⁇ 10 6 CFU/ml) were incubated for 6 h at 37 °C with a two-fold dilution series of peptide in medium in a sterile 96-well plate. Microbial growth was determined by the increase in OD 590 over the 6 h period.
  • ⁇ Minimal bactericidal concentration refers to the lowest concentration of peptide required for the absence of viable colonies.
  • the bactericidal activities of the MIC, MICx2, and MICx4 were examined.
  • the well solution was diluted and plated in order to allow a maximum of ⁇ 10 3 colony-forming units to grow on the agar plate. Plates for t 0 contained approximately 1000 colonies. The plates were incubated overnight at 37° C, and colonies were counted for determination of bactericidal activity.
  • Teptide concentration of 100 ⁇ g/ml revealed 10 colonies ( " 99% killing).

Abstract

Disclosed are antimicrobial compositions containing β-peptides and methods of inhibiting microbial growth in mammals using the compositions. The β-peptides present in the compositions contain ring structures in the peptide backbone which limit the conformational flexibility of the peptide backbone.

Description

ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS
FIELD OF THE INVENTION
The present invention is directed to antimicrobial compositions containing active ingredients which are oligomers and/or polymers of β-amino acids.
DESCRIPTION OF THE PRIOR ART
Chemists have long sought to extrapolate the power of biological catalysis and recognition to synthetic systems. These efforts have focused largely on low molecular weight catalysts and receptors. Most biological systems, however, rely almost exclusively on large polymers such as proteins and RNA to perform complex chemical functions.
Proteins and RNA are unique in their ability to adopt compact, well-ordered conformations. These two biopolymers are unique also because they can perform complex chemical operations (e.g. , catalysis, highly selective recognition, etc.). Folding is linked to function in both proteins and RNA because the creation of an "active site" requires proper positioning of reactive groups. Consequently, there has been a long-felt need to identify synthetic polymer backbones which display discrete and predictable folding propensities (hereinafter referred to as "foldamers") to mimic natural biological systems. Such backbones will provide molecular "tools" to probe the functionality of large-molecule interactions (e.g. protein-protein and protein-RNA interactions).
Much work on β-amino acids and peptides synthesized therefrom has been performed by a group led by Dieter Seebach in Zurich, Switzerland. See, for example, Seebach et al. (1996) Helv. Chim. Acta. 79:913-941 ; and Seebach et al. (1996) Helv. Chim. Acta. 79:2043-2066. In the first of these two papers Seebach et al. describe the synthesis and characterization of a β-hexapeptide, namely (H-β-HVal-β-HAla-β- HLeu)2-OH. Interestingly, this paper specifically notes that prior art reports on the structure of β-peptides have been contradictory and "partially controversial. " In the second paper, Seebach et al. explore the secondary structure of the above-noted β- hexapeptide and the effects of residue variation on the secondary structure.
Dado and Gellman (1994) J. Am. Chem. Soc. 116: 1054-1062 describe intramolecular hydrogen bonding in derivatives of β-alanine and γ-amino butyric acid. This paper postulates that β-peptides will fold in manners similar to α-amino acid polymers if intramolecular hydrogen bonding between nearest neighbor amide groups on the polymer backbone is not favored.
Suhara et al. (1996) Tetrahedron Lett. 37(10): 1575-1578 report a polysaccharide analog of a β-peptide in which D-glycocylamine derivatives are linked to each other via a C-1 β-carboxylate and a C-2 α-amino group. This class of compounds has been given the trivial name "carbopeptoids. "
Hamuro et al. (1999) J. Am. Chem. Soc. 121 : 12200-12201 , describe antibacterial compositions containing β-peptides having a repeating 3-peptide residue motif. The compounds described are: Fmoc-(β3-HVal-β3-HLys-β3-HLeu)n-OH (n =2- 4); H-(β3-HVal-β3-HLys-β3-HLeu)n-OH (n=2-4); and H-(β3-HLeu-β3-HLys-β3- HLeu)n-OH (n =2-6). While these β-peptides are described as being antibacterial, they are also hemolytic at concentrations near the effective antibacterial concentrations, thus limiting their utility as medicaments. SUMMARY OF THE INVENTION
The increasing prevalence of pathogenic bacteria that are resistant to common chemotherapies has prompted an intensive search for new antibiotics. Cationic peptides that adopt amphiphilic secondary structures (e.g. , magainins) constitute a widespread host defense against microbial invasions, and mimicry of these natural antibiotics led the present inventors on a search for new synthetic antimicrobial agents. Many of the cationic antimicrobial peptides appear to act by disrupting bacterial membranes, a mechanism that may not be conducive to the development of resistance. The subject invention is directed to antimicrobial compositions containing unnatural cationic oligomers that have no known natural counterpart and that display antibiotic activity comparable to that of a magainin derivative against at least four different bacterial species, including two pathogens that are resistant to common antibiotics. The oligomers are constructed from β-amino acid building blocks rather than the α- amino acid building blocks of conventional peptides. These β-peptides exhibit lower hemolytic activity than does the magainin derivative, which indicates a favorable selectivity for bacterial cells relative to mammalian cells. Because β-peptides are not expected to be subject to protease degradation, these results point toward the rational design of a promising new antibiotic class.
Specifically , the present invention is directed to compounds and to antimicrobial compositions containing the compounds, that is, compositions which inhibit the growth of microbes in general and bacteria and fungi in particular, the compositions comprising an antimicrobial-effective amount of a β-amino acid oligomer or polymer of the formula:
W- (« -A-B-C-« » »)-Z
wherein " • • • -A-B-C- • • • " represents a β-amino acid oligomer or polymer chain, each of A, B, C, etc. representing individual residues of the oligomer or polymer chain and the ellipses representing adjacent N-terminal and C-terminal β-amino acid residues of the same genus, wherein each residue of the oligomer or polymer chain (that is, each A, B, C, etc.) is the same or different and is independently selected from the group consisting of:
Figure imgf000005_0001
wherein X is selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl--C1-C6- alkyl, mono- or bicyclic heteroaryl-Ci-Ce-alkyl, -(CΑ a+l-OR,
Figure imgf000005_0002
-(CH2)n+1-S(=O)-CH2-R, -(CH2)n+1-S(=O)2-CH2- R, -(CH2)n+1-NRR, -(CH2)n+1-NHC(=0)R,-(CH2)n+1-NHS(=O)2-CH2-R, -(CH^-O- CH^-R1, -(CH2)0+1-S-(CH2)m-R1, -(CH2)n+1-S(=O)-(CH2)m-R1, -(CH2)n+1-S(=O)2-(CH2)m-R1, -(CH^.-NH^CH^-R1, -(CH^.-N-UCH^-R1},, -(CH2)n+1-NHC(=O)-(CH2)n+1-R1, and -(CH2)n+1-NHS(=O)2-(CH2)m-R1; wherein R is independently selected from the group consisting of hydrogen, C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl- - -alkyl, mono- or bicyclic heteroaryl-CrC6-alkyl; and wherein R1 is selected from the group consisting of hydroxy, C C6- alkyloxy, aryloxy, heteroaryloxy, thio, C,-C6-alkylthio, Cι-C6-alkylsulfinyl, Cr C6-alkylsulfonyl, arylthio, arylsulfinyl, arylsulfonyl, heteroarylthio, heteroarylsulfinyl, heteroarylsulfonyl, amino, mono- or di-C,-C6-alkylamino, mono- or diarylamino, mono- or diheteroarylamino, N-alkyl-N-arylamino, N- alkyl-N-heteroarylamino, N-aryl-N-heteroarylamino, aryl-C,-C6-alkylamino, carboxylic acid, carboxamide, mono- or di-Cj-Cg-alkylcarboxamide, mono- or diarylcarboxamide, mono- or diheteroarylcarboxamide, N-alkyl-N- arylcarboxamide, N-alkyl-N-heteroarylcarboxamide, N-aryl-N- heteroarylcarboxamide, sulfonic acid, sulfonamide, mono- or di-C,-C6- alkylsulfonamide, mono- or diarylsulfonamide, mono- or diheteroarylsulfonamide, N-alkyl-N-arylsulfonamide, N-alkyl-N- heteroarylsulfonamide, N-aryl-N-heteroarylsulfonamide, urea; mono- di- ortri- substituted urea, wherein the subsitutent(s) is selected from the group consisting of Cj-C6-alkyl, aryl, heteroaryl; O-alkylurethane, O-arylurethane, and O- heteroarylurethane; wherein Y is selected from the group consisting of hydrogen, linear or branched Cι-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic arylτC,-C6- alkyl, mono- or bicyclic heteroaryl-CrC6-alkyl, -(CH^^OR, -(CH^-SR, -(CH2)n-S(=O)-CH2-R, -(CH2)n-S(=O)2-CH2-R, -(CH2)n-NRR, -(CH2)n-NHC(=O)R, -(CH2)n-NHS(=O)2-CH2-R, -(CH2)n-0-(CH2)1I1-R1, -(CH2)n-S-(CH2)m-R1, -(CH2)n-S(=O)-(CH2)m-R', -(CH2)n-S(=0)2-(CH2)m-R1, -(CH2)B-NH-(CH2)m-Rl, -(CH2)n-N-{(CH2)m-R1}2 -(CH2)n-NHC(=O)-(CH2)m-R1, and -(CH2)n-NHS(=O)2-(CH2)m-R1; wherein R and R1 are as defined above; or X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted or unsubsituted C3-C8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more N, O or S atom(s) as the heteroatom (s); the substituents on carbon atoms of these rings being independently selected from the group consisting of linear or branched CrC6-alkyl, alkenyl, alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-CrC6-alkyl, mono- or bicyclic heteroaryl-Cj- -alkyl, and the substituents listed above for X and Y when X and Y are not combined; the substituents on nitrogen heteroatoms being independently selected from the group consisting of -S(=O)2-CH2-R, -C(=O)-R, -S(=O)2-(CH2)m-Rl, -C(=O)-(CH2)0+1-R1; wherein R and R1 are as defined above; m is an integer of from 2-6 and n is an integer of from 0-6; W is hydrogen or an a ino-terminal capping group (such as formyl, acetyl, tBoc, Fmoc, etc.); Z is hydroxy or a carboxy-terminal capping group (such as NH2, NH(alkyl), N(alkyl)2, etc.); and wherein the total number of residues is greater than 6; and further wherein in at least one residue, X and Y are combined in a cyclic moiety; pharmaceutically-suitable salts thereof, and combinations thereof; optionally in combination with a pharmaceutically-suitable caπier.
When X is not combined with Y, X and Y are preferably (and independently) selected from the group consisting of linear or branched CrC6-alkyl, hydroxyτC,-C6- alkyl, amino- - -alkyl, aryl-Cj-Cg-alkyl, heteroaryl- -Cg-alkyl, and carboxyl-CrC6- alkyl. When X and Y are combined together into a cyclic substituent, together X and Y and the carbons to which they are bonded define a substituted or unsubstituted cyclopentyl, substituted or unsubstituted cyclohexyl, unsubstituted or N-substituted piperidinyl, or unsubstituted or N-substituted pyrrolidinyl.
In a another embodiment, the composition contains a β-amino acid oligomer or polymer of the above, wherein each residue (A, B, C, etc.) of the oligomer is a β- amino acid residue of formula:
Figure imgf000007_0001
wherein X and Y combined, together with the carbon atoms to which they are bonded, independently from residue to residue define a substituted or unsubsituted C3- C8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more nitrogen atoms as the sole heteroatom, the substituents being selected from the group consisting of hydroxy, linear or branched C,-C6-alkyl, alkenyl, alkynyl; hydroxy-Cι-C6-alkyl, amino-Cι-C6-alkyl, C,-C6-alkyloxy, Cj- -alkyloxy-Cj-Cfj-alkyl, amino, mono- or di- Cj-C6-alkylamino, carboxamido, carboxamido-C,-C6-alkyl, sulfonamido, sulfonamido- C,-C6-alkyl, urea, cyano, fluoro, thio, CrC6-alkylthio, mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, heteroaryl-Cj- -alkyl, and combinations thereof; where n is a positive integer greater than 6; and pharmaceutically-acceptable salts thereof, optionally in combination with a pharmaceutically-suitable carrier.
The invention is likewise directed to a method of inhibiting microbial growth in mammals, including humans, the method comprising administering an antimicrobial amount of a composition as recited above to a patient in need thereof.
Other aims, objects, and advantages of the invention will appear more fully from a complete reading of the following Detailed Description of the Invention and the attached claims.
BRIEF DESCRIPTION OF THE DRAWINGS
Fig. 1 shows the structure of poly-β-alanine and further depicts the hydrogen bonds that define the six narrowest helices available to poly-β-alanine. Poly-β-alanine is the simplest β-peptide polymer.
Fig. 2 A depicts the crystal structure of oligomer of trans-ACΑCl 14-helix. The bottom depiction is the two-dimensional structure, the middle depiction is a view along the axis of the helix, and the top depiction is a view perpendicular to the axis of the helix.
Fig. 2B depicts the crystal structure of oligomer of tra/w-ACPC/12-helix. The views shown are the same as in Fig. 2A.
Fig. 2C depicts the crystal structure of a standard α-helix. The views shown are the same as in Fig. 2 A. Fig. 3 depicts 'H NMR spectra for a solution containing 2 mM trans-ACHC dimer and 2 mM trans-ACHC hexamer. The bottom spectrum was obtained in CD3OH with solvent suppression. The two NH resonances from the dimer are indicated with an asterisk (*). All other spectra were obtained in CD3OD at the times indicated after dissolution of the sample. Data obtained on a Bruker 300 MHz spectrometer at 20 ° C .
Fig. 4 is a circular dichroism (CD) plot for trans- ACPC hexamer in CH3OH.
Fig. 5 is a CD plot comparing trans- ACVC dimer, trimer, tetramer and hexamer.
Fig. 6 depicts XH NMR spectra for a solution containing 2 mM of a hexamer of alternating amino-substituted-tran_f-ACHA and trans-ACHA. All spectra were obtained in D2O, 100 mM deuteroacetate buffer, pD 3.9, at the times indicated after dissolution of the sample. Data obtained on a Bruker 300 MHz spectrometer at 20° C.
Fig. 7 is a comparison between the k^, for amide proton exchange in a hexamer of alternating amino-substituted-trα/w-ACHA and trans-ACHA and a dimer of alternating amino-substituted-tra w-ACHA and trans-ACHA.
Fig. 8 is a comparison between the CD plot of a hexamer of alternating amino- substituted-tra/w-ACHA and trans-ACHA in water and the CD plot of a hexamer of trans-ACHC in methanol.
Fig. 9 is a comparison of CD data in methanol for a β-peptide tetramer, hexamer, and octamer containing alternating trans- ACPA residues and 4-pyrrolidinyl residues.
Fig. 10 is a comparison of CD data in water for a β-peptide tetramer, hexamer, and octamer containing alternating trans-ACPA residues and 4-pyrrolidinyl residues.
Fig. 11 is a superimposed plot of the CD data in water and the CD data in methanol for the octamer containing alternating trans-ACPA residues and 4- pyrrolidinyl residues. Fig. 12 is a CD spectrum in water of an octamer containing alternating trans- ACPA residues and 3-pyrrolidinyl residues.
Fig. 13 is a comparison of CD spectra for a hexamer containing alternating residues of trans-ACHA and ammo-substituted-trans-ACHA and for a "mixed" β- peptide hexamer comprising alternating residues of trans-ACHA and an acyclic β- amino acid bearing an aminopropyl substituent on the β-carbon of the backbone.
Fig. 14A shows the infrared spectrum of two linked nipecotic acid residues wherein the two residues have the same absolute configuration.
Fig. 14B shows the infrared spectrum of two linked nipecotic acid residues wherein the two residues have the opposite absolute configuration. This diastereomer acts as a reverse turn in β-peptides which adopt a sheet structure.
Fig. 15 is a schematic representation of the "split and pool" method of generating combinatorial libraries.
Fig. 16 is a ball and stick representation of the solid state conformation of compound 1 described below. For clarity, all hydrogen atoms, except for those attached to nitogen, have been omitted. Hydrogen bonds are indicated with dotted lines.
Fig. 17 is a graph depicting the results of a direct hemolysis comparison between melittin (a positive control), a magainin derivative, and β-17 (see Example 11)..
DETAILED DESCRIPTION OF THE INVENTION
Overview
Helices in β-Peptides:
Initial molecular modeling studies indicated that β-amino acid oligomers ("β- peptides") are well suited for adoption of compact secondary structures stabilized by intramolecular hydrogen bonds. Fig. 1 shows the hydrogen bonds that define the six narrowest helices available to poly-β-alanine, the simplest β-peptide polymer. The 12-, 16-, and 20-helices (nomenclature derived from hydrogen-bonded ring size) contain hydrogen bonds from carbonyls toward NH groups in the N-terminal direction, as observed for 3J0- and α-helices in proteins, while the 10-, 14-, and 18-helices contain hydrogen bonds from carbonyls to NH groups in the N-terminal direction. Molecular mechanics studies of a β-alanine decamer suggested that all six of these helices constitute local minima on the conformational energy surface. β-Alanine oligomers, however, have been shown experimentally to be unordered in solution and to adopt sheetlike packing patterns in the solid state.
Incorporation of the two backbone carbons of a β-amino acid into a small carbocycle or heterocycle provides substantial rigidity to the backbone. Computational methods were used to evaluate whether any particular helix/small ring combination would lead to enhanced conformational stability. For each of the six minimized deca- β-alanine helices shown in Fig. 1, each residue was modified by incorporation of the backbone carbons into a three-, four-, five-, and six-membered cycloalkyl ring. For each ring size, both cis and trans relationships between the amino and carboxyl substituents were examined, and for the cis rings, both of the possible ring orientations relative to the helix were examined. This process yields 72 helical starting structures (6 helices x 4 cycloalkyl ring sizes x (1 trans + 2 cis forms)). A combination of minimization and dynamics studies predicted that the 14-helical form of the decamer of trans-2-aminocyclohexanecarboxylic acid (trans-ACHC) (the corresponding monomer is refered to herein as trans-ACHA) and the 12-helical form of the decamer of trans-2-aminocyclopentanecarboxylic acid (trans- ACPC) (corresponding monomer referred to as trans- ACPA) would be the most stable among these hypothetical helices. These two structures are shown below:
Figure imgf000012_0001
trans-ACHC
Figure imgf000012_0002
trans-ACPC n > 3
In order to test this computational prediction, optically active trans-ACHC was prepared by the reported route, Nohira et al. (1970) Bull. Chem. Soc. Jpn. 43:2230, and polypeptide oligomers synthesized via standard methods (see below). The crystal structures of the trans-ACHC tetramer and the trans-ACHC hexamer reveal that these molecules adopt 14-helical conformations in the solid state. The hexamer crystal contains three independent but very similar molecules, each of which forms the four possible 14-membered ring hydrogen bonds. The regular helix revealed by the hexamer crystal structure matches the minimum energy conformation predicted for the decamer.
An alternatively rigidified β-amino acid, trans-2-aminocyclopentanecarboxylic acid (trans- ACPC), provides β-peptides with a dramatically altered secondary structure, the 12-helix. Fig. 2B depicts the 12-helix rr ns-ACPC structure, as compared to a standard α-helix (Fig. 2C) and trans-ACHC/ 14-helix (Fig. 2A). This finding shows that β-peptides allow profound residue-based control of peptide conformation.
Chemistry:
General. Melting points are uncorrected. CH2C12 was freshly distilled from CaH2 under N2. DMF was distilled under reduced pressure from ninhydrin and stored over 4 A molecular sieves. Triethylamine was distilled from CaH2 before use. Other solvents and reagents were used as obtained from commercial suppliers. For BOC removal, 4 M HCl in dioxane from was used. Column chromatography was carried out by using low air pressure (typically 6 psi) with 230-400 mesh silica gel 60. Routine 'H-NMR spectra were obtained on a Bruker AC-300 and are referenced to residual protonated NMR solvent. Routine 13C-NMR spectra were obtained on a Bruker AC-300 and are referenced to the NMR solvent. High resolution electron impact mass spectroscopy was performed on a Kratos MS-80RFA spectrometer with DS55/DS90.
Infrared Spectroscopy. Spectra were obtained on a Nicolet Model 740 FT-IR spectrometer. IR samples were prepared under anhydrous conditions; CH2C12 was freshly distilled from CaH2, compounds and glassware were dried under vacuum for 1-2 days, and solutions were prepared under a nitrogen atmosphere. The pure solvent spectrum for a particular solution was subtracted from the sample spectrum prior to analysis. Peaks in the amide NH stretch region were baseline corrected, and analyzed without further manipulation. NMR Spectroscopy. 1. Aggregation Studies. One-dimensional spectra for aggregation studies were obtained on a Bruker AC-300 spectrometer. Samples for aggregation studies were prepared by serial dilution from the most concentrated sample (50 mM or 27 mM). Dry compounds were dissolved in CD2C12 previously dried over 3 A molecular sieves, and samples were prepared with dry glassware under a nitrogen atmosphere.
2. Conformational Analysis. NMR samples for conformational analysis were prepared by dissolving the dry compound in dry deuterated solvent under a nitrogen atmosphere. CD2C12 samples were then degassed by the freeze-pump-thaw method, and the NMR tubes were sealed under vacuum. Methanol samples were sealed with a close fitting cap and parafilm. COSY spectra were obtained on a Bruker AC-300 spectrometer. TOCSY (Braunschweiler, L.; Ernst, R. R. (1983) J. Magn. Reson. 53:521), NOESY (Macura, S.; Ernst, R. R. (1980) Mol. Phys. 41:95), and ROESY (Bothner-By, A. A. ; Stephens, R. L.; Lee, J.; Warren, C. D. ; Jeanloz R. W. (1984) J. Am. Chem. Soc. (1984) 106:811) spectra were squired on a Varian Unity-500 spectrometer using standard Varian pulse sequences and hypercomplex phase cycling (States-Haberkorn method), and the data were processed with Varian " VNMR" version 5.1 software. Proton signals were assigned via COSY and TOCSY spectra, and NOESY and ROESY spectra provided the data used in the conformational analyses. TOCSY spectra were recorded with 2048 points in t^ 320 or 350 points in ij, and 8 or 40 scans per t^ increment. NOESY and ROESY spectra were recorded with a similar number of t[ and tj points, and 32 and 40 scans per t^ increment, depending on the sample concentration. The width of the spectral window examined was between 2000 and 4000 Hz. Sample concentrations for two-dimensional spectra were 2 mM in CD2C12 and 8 mM in CD3OD and CD3OH.
Far UV Circular Dichroism (CD). Data were obtained on a Jasco J-715 instrument at 20° C. In all CD plots contained herein, the mean residue ellipticity is presented on the vertical axis. Presenting the mean residue ellipticity is a standard practice in peptide chemistry wherein the intensity of each CD spectrum is normalized for the number of amide chromophores in the peptide backbone. Consequently, when the intensities of the maximum (ca. 205 nm) and minimum (ca. 220 nm) peaks characteristic of helix formation increase with increasing chain length, this change represents an increase in the population of the helix structure, rather than simply an increase in the number of chromophores present in each molecule.
Synthesis. The β-amino acids used to assemble the peptides described herein can be manufactured using several different literature methods, as well as new methods described below. For unsubstituted β-amino acids and β-amino acids containing one or two acyclic substituents on the carbon adjacent to the amino group in the product β- peptide, the Arndt-Eisterdt homologation reaction can be used, see Reaction 1. See also Seebach et al. (1996) Helv. Chim. Acta 79:913. This route has advantages and disadvantages. A distinct advantage is that the starting materials, α-amino acids, are readily available commercially in enantiomerically pure form. The Arndt-Eisterdt homologation also results in the simultaneous coupling of two β-amino residues. A distinct disadvantage is that the reaction cannot be used to synthesize β-amino acids having rings in the backbone or α-carbon substituents. The reaction proceeds via a Wolff rearrangement of a diazoketone with subequent trapping of the reactive intermediate with an amino moiety, as shown in Reaction 1:
Figure imgf000015_0001
Figure imgf000015_0002
(Pg designates a protecting group such as (t-butoxy)carbonyl (Boc) or an adjacent β- a ino residue, R1 and R2 are aliphatic substituents. β-Amino acids containing an unsubstituted cycloalkyl moiety involving the α and β carbons were synthesized using literature methods. See, for example, Nohira et al. (1970) Bull. Chem. Soc. Jpn. 43:2230; Herradon and Seebach (1989) Helv. Chim. Acta 72:690-714; and Tilley et al. (1992) J. Med. Chem. 35:3774-3783, all three of which are incorporated herein by reference.
In particular, the cyclohexyl-containing β-amino acids can be synthesized via Reaction 2:
Figure imgf000016_0001
NaOMe, MeOH, Reflux
Figure imgf000016_0002
Figure imgf000016_0003
REACTION 2 (lR,6S)-6-Methoxycarbonyl-3-cyclohexene-l-carboxylic acid (23): 4600 u of PLE was suspended in pH 8.01 aqueous buffer solution (0.17 M KH2PO4). The diester 22 (10.1 g, 0.05 mol) was dissolved in 30 mL of acetone and added to the buffer solution. Reaction was allowed to stir at rt overnight. The enzyme was filtered off through a well-packed celite pad, the solution was then acidified to pH 1 with IM HCl and the product was extracted with ethyl acetate (5 x 400 mL). The combined organic extracts were dried over anhydrous magnesium sulfate and concentrated to yield 9.00 g yellow oil. Product taken on without further purification.
Methyl (15,6R)-6-benzyloxycarbonyIaminocyclohex-3-ene carboxylate (24): Ethylchloroforamate (4 mL, 0.042 mol) was added to a mixture of 23 (5.14 g, 0.028 mol) and triethylamine (6 mL, 0.043 mol) in acetone (100 mL) at 0° C and vigorously stirred for 10 min. An aqueous solution of NaN3 (3.04 g, 0.047 mol, in 25 mL water) was added in one portion. The resulting mixture was stirred for 30 min at 0° C. The reaction mixture was diluted with water and extracted with diethyl ether. The organic extracts were dried over anhydrous magnesium sulfate and concentrated without heat to yield a viscous yellow liquid. The liquid was dissolved in 100 mL of benzene and refluxed under nitrogen atmosphere for 30 min. Benzyl alcohol (12 mL, 0.116 mol) was added and solution was refluxed for an additional 16 h. The reaction was cooled to rt and concentrated to yield 17.12 g of a yellow liquid (mixture of benzyl alcohol and desired product in a 5.4: 1 ratio, respectively by lH NMR, -5.67 g product). Mixture taken on without further purification.
Methyl (lS,6R)-6-te/f-butoxycarbonylaminocyclohexane carboxylate (25): The yellow oil from the previous reaction, which contains compound 24 (5.6 g, 0.020 mol) and benzyl alcohol, was dissolved in methanol. 0.525 g of 10% Pd on carbon was added to the methanol solution, and the heterogenous mixture was placed under 50 psi H2 and shaken at rt for 24 h. The mixture was filtered through celite, and the filtrate was concentrated to yield 13.74 g of dark golden yellow liquid. 25 mLof IM HCl was added to the filtrate, and the benzyl alcohol was extracted with diethyl ether (3 x 25 mL). The pH of the aqueous solution was adjusted to 9 using K2C03. 25 mL of dioxane and Boc2O (5 g, 0.023 mol) were added to the solution, and the reaction was stirred at rt for 20 h. 15 mL of water was added and the solution was extracted with ethyl acetate (3 x 50 mL). The combined organic extracts were dried over anhydrous magnesium sulfate and concentrated. Residue was purified via column chromatography (SiO2, eluting with 6: 1 Hex:EtOAc), to yield 2.00 g viscous clear oil.
Methyl (IR,6R)-6-te/*-butoxycarbonylaminocyclohexane carboxylate (26): Sodium metal (0.14 g, 6.1 mmol) was placed into a flame dried flask under nitrogen atmosphere and cooled to 0° C. 10 mL of freshly distilled methanol was added and the mixture stirred until all the sodium dissolved. An amount of 25 (2.00 g, 7.7 mmol) was dissolved in 10 mL of freshly distilled methanol and transferred to NaOMe solution via cannula. The solution was refluxed under nitrogen for 5.5 h, cooled to rt and acidified with 0.5 M aqueous 0.5 M ammonium chloride (18 mL, 9 mmol). The methanol was removed under reduced pressure, and the resulting solid collected by filtration to yield 1.27 g of desired product. β-Amino acids containing a substituted cycloalkyl moiety were synthesized using the following illustrative protocol, the first four steps of which are described in Kobayashi et al. (1990) Chem. Pharm. Bull. (1990) 38:350. The remaining steps to yield a cyclohexyl ring having two differentially protected amino substituents were developed in furtherance of the present invention and have not heretofore been described in the literature and are shown in Reaction 3 :
Figure imgf000019_0001
REACTION 3
As depicted in Reaction 3, the 4-position amino substituent is protected by a Boc group and the 1 -position amino substituent is protected by a Cbz group. The starting material is available commercially (Aldrich Chemical Co. , Milwaukee, Wisconsin).
Synthesis of β-amino acids containing a heterocylic ring moiety encompassing the α and β carbons were synthesized using Reactions 4 and 5, below. Reaction 4 details an illustrative synthesis of a β-proline wherein the exocyclic amino substituent is in the 3-position relative to the ring nitrogen.
Compound 42: Tap water (200 ml) and baker's yeast (25 g) were mixed, and were shaken on an orbital shaker for 1 hour. Compound 41 (1.0 g) was then added. The mixture was shaken at room temperature for 24 hours. The mixture was filtered through a bed of Celite. The Celite was washed with water (20 ml). The filtrate was extracted with diethyl ether (5 x 100 ml). The extracts were washed with water (2 x 50 ml), dried over MgS04, and concentrated to yield a slightly yellow oil. The crude product was purified by column chromatography with ethyl acetate/hexane (1/1, v/v) as eluent to give a colorless oil (0.5 g) in 50% yield.
Compound 43: Compound 42 (228 mg) and Ph3P (346 mg) were dissolved in benzene (anhydrous, 4 ml) under nitrogen. HN3 (1.64 M in benzene, 0.8 ml) was then added. A solution of diethyl azodicarboxylate (0.18 ml) in benzene (1.0 ml) was subsequently introduced via syringe over 5 minutes. The reaction mixture turned cloudy towards the end of the addition. The reaction mixture was stirred under nitrogen at room temperature for 3.0 hours. The reaction mixture was then taken up in ethyl acetate (50 ml), washed with IN NaOH (10 ml), saturated NaHCO3 (10 ml), and finally dilute brine (5 ml). The organic was dried over MgS04, and concentrated to give a slightly yellow oil. The crude oil was purified by column chromatography with ethyl acetate/hexane (1/1 , v/v) as eluent to afford a colorless oil (190 mg) in 76 % yield.
Compound 44: Compound 43 (1.1 g) was dissolved in methanol (50 ml) . SnCl2 (2.2 g) was then added. The mixture was stirred at room temperature for 30 hours. The methanol was then removed under reduced pressure. The residue was dissolved in Baker's Yeast Tap Water
Figure imgf000021_0001
Figure imgf000021_0002
oc
4-1 63%,99% ee 42
PhjP/DEAD/HNj 1) SnCi MeOH
Benzene 2) Cbz-OSU/ aHCOj Acetone/Water
Figure imgf000021_0003
76%
43
Figure imgf000021_0004
86% 46
DEAD = Diethyl azodicarboxylate
Cbz-OSU = N-φenzyloxycarbonyloxy)succinimide
Fmoc-OSU = 9-FIuorenylmethyIoxycarbonyl-N-hydroxysuccinimide
Starting material: Blake, J. et alj. Am. Chem. Soc. 1964, 86, 5293.
Yeast reduction: Cooper, J. et alj. Chem. Soc. Perkin Trans. 1 1993, 1313.
REACTION 4 methylene chloride (50 ml). The resulting cloudy solution was filtered through Celite. The methylene chloride was then removed under reduced pressure. The residual white solid was dissolved in acetone/water (2/1, v/v, 50 ml). NaHCO3 (3.3 g) was added, followed by Cbz-OSU ( 1.16 g) . The reaction mixture was stirred at room temperature for 24 hours. Water (50 ml) was added. The acetone was removed under reduced pressure. The aqueous mixture was extracted with ethyl acetate (3 x 100 ml). The extracts were washed with dilute brine (30 ml), dried over MgS04, and concentrated to give a colorless oil. The crude product was purified by column chromatography with ethyl acetate/hexane (3/7, v/v) as eluent to give the clean product as a colorless oil (1.35 g) in 89% yield.
Compound 45: Compound 44 (1.35 g) was dissolved in methanol/ water (3/1, v/v, 80 ml), cooled to 0° C. LiOH.H20 (1.68 g) was added. The mixture was stirred at 0° C for 24 hours, by which time TLC indicated that the hydrolysis was complete. Saturated ammonium hydroxide (20 ml) was added. The methanol was removed under reduced pressure. The aqueous was washed with diethyl ether (50 ml), acidified with IN HCl to pH 3, extracted with methylene chloride (3 x 150 ml). The extracts were washed with dilute brine (50 ml), dried over MgSO4, concentrated to give a sticky colorless residue (1.25 g, 99%), which was used directly without further purification.
Compound 46: Compound 45 (1.25 g) was dissolved in methanol (50 ml) in a hydrogenation flask. 5 % Palladium on activated carbon (190 mg) was added. The flask was pressurized with hydrogen to 35 psi, rocked at room temperature for 7 hours, by which time TLC indicated that the hydrogenolysis was complete. The Pd/C was removed by filtration. The filtrate was concentrated to give a white solid. The white solid was dissolved in acetone/water (2/1, v/v, 70 ml), cooled to 0° C. NaHCO3 (1.7 g) was added, followed by FMOC-OSU (1.39 g). The reaction mixture was stirred at room temperature for 16 hours. Water (50 ml) was added. The acetone was removed under reduced pressure. The aqueous was washed with diethyl ether (50 ml), acidified with IN HCl to pH 3, extracted with methylene chloride (3 x 150 ml). The extracts were washed with dilute brine (50 ml), dried over MgS04, concentrated to give a foamy white solid. The crude white solid was purified by column chromatography with methanol/ethyl acetate (3/7, v/v) as eluent to give the clean product as a white solid (1.3 g) in 86% yield.
Reaction 5 illustrates the synthesis of a β-amino acid wherein the exocyclic amino substituent the nitrogen heteroatom is in the 4-position relative to the ring nitrogen.
Compound 52: Compound 51 (2.0 g) and NaBH3CN (0.54 g) were dissolved in methanol (40 ml), IN HCl (aqueous) was added dropwise to maintain pH 3-4. After 15-20 minutes, pH change slowed. The mixture was stirred for an additional 1.0 hour, while IN HCl was added occasionally to keep pH 3-4. Water (100 ml) was added. The mixture was extracted diethyl ether (3 x 150 ml). The extracts were washedwith IN NaHC03 (100 ml) and dilute brine (100 ml), dried over MgS04, and concentrated to give a colorless oil (1.9 g) in 95 % yield. The product was used directly without further purification.
Compound 53: Compound 52 (1.9 g) and Ph3P (2.8 g) were dissolved in toluene (anhydrous, 30 ml) under nitrogen. A solution of diethyl azodicarboxylate (1.5 ml) in toluene (10 ml) was subsequenely introduced via syringe over 15 minutes. The reaction mixture was stirred under nitrogen at room temperature for 12 hours. The toluene was removed under reduced pressure. The residue was purified by column chromatography with ethyl acetate/hexane (3/7, v/v) as eluent to afford a colorless oil (1.6 g) in 91 % yield.
Compound 54: Compound 53 (1.0 g) and R-(+)-α-methylbenzylamine (l. l ml) were mixed with water (15 ml). The mixture was stirred at 55° C for 67 hours. The mixture was taken up in diethyl ether (300 ml), and the aqueous layer was separated. The ether solution was washed with water (3 x 50 ml), dried over MgSO4, and concentrated to give a slight yellow oil. The diastereometic isomers were separated by column chromatography with ethyl acetate/hexane (2/8, v/v) as eluent to give RSS (0.2 g) and RRR (0.34 g) in 51 % overall yield.
Compound 55: Compound 54 (4.2 g) was dissolved in ethyl acetate (200 ml). 4N HCl in dioxane (4.35 ml) was added dropwise while stirring. A white precipitate
Figure imgf000024_0001
63 95%
Figure imgf000024_0002
RRR 51% RSS RSS 100%
RRR/RSS: L7/1
Trans/Or. 2&1
Figure imgf000024_0003
90% 88%
bonyl-
Figure imgf000024_0004
Starting material: Blake, J. ύtάlJ. Am. Chem. Soc. 1961, 86, 5293. REACTION 5 resulted. The ethyl acetate was removed under reduced pressure, and the resulting white solid (4.6 g, 100%) was dried in vacuo.
Compound 56: Compound 55 (4.6 g) was dissolved in 95 % ethanol (150 ml) in a hydrogenation flask. 10% Palladium on activated carbon (0.5 g) was added. The flask was pressurized with hydrogen to 50 psi, rocked at room temperature for 22 hours, by which time NMR spectroscopy indicated that the hydrogenolysis was complete. The Pd/C was removed by filtration. The filtrate was concentrated to give a white solid. The white solid was dissolved in acetone/water (2/1 , v/v, 150 ml). NaHCOj (9.7 g) was added, followed by Cbz-OSU (3.4 g). The reaction mixture was stirred at room temperature for 14 hours. Water (100 ml) was added. The acetone was removed under reduced pressure. The aqueous mixture was extracted with ethyl acetate (3 x 200 ml). The extracts were washed with IN HCl (3 x 100 ml) and saturated NaHCO3 (aqueous), dried over MgS04, and concentrated to give a colorless oil. The crude product was purified by column chromatography with ethyl acetate/hexane (3/7, v/v) as eluent lo give the clean product as a colorless sticky oil (4.0 g) in 90% yield.
Compound 57: Compound 56 (2.0 g) was dissolved in methanol/water (3/1 , v/v, 115 ml), cooled to 0° C, LiOH.H20 (2.4 g) was added. The mixture was stirred at O° C for 15 hours, by which time TLC indicated that the hydrolysis was complete. Saturated ammonium hydroxide (aqueous, 100 ml) was added. The methanol was removed under reduced pressure. The aqueous was acidified with IN HCl to pH 3, extracted with ethyl acetate (3 x 200 ml). The extracts were washed with dilute brine (100 ml), dried over MgSO4, concentrated to give a foamy solid (1.63 g, 88%), which was used directly without further purification).
Compound 58: Compound 57 (1.63 g) was dissolved in methanol (70 ml) in a hydrogenation flask. 5 % Palladium on activated carbon (250 mg) was added. The flask was pressurized with hydrogen to 35 psi, rocked at room temperature for 15 hours, by which time NMR spectroscopy indicated that the hydrogenolysis was complete. The Pd/C was removed by filtration. The filtrate was concentrated to ive a white solid. The white solid was dissolved in acetone/water (2/1 , v/v, 90 ml), cooled to O° C. NaHCO3 (2.27 g) was added, followed by FMOC-OSU (1.83 g). The reaction mixture was stirred at O° C for 2 hours, then at room temperature for 28 hours. Water (50 ml) was added. The acetone was removed under reduced pressure. The aqueous was acidified with IN HCl to pH 3, extracted with ethyl acetate (3 x 200 ml). The extracts were washed with dilute brine (100 ml), dried over MgSO4, concentrated to give a foamy white solid. The crude white solid was purified by column chromatography with methanolfethyl acetate (3/7, v/v) as eluent to give the clean product as a white solid (1.68 g) in 84% yield.
To synthesize the nipecotic reverse turn moiety, Reaction 6 was used.
Figure imgf000027_0001
AciO (quantitative yield)
Figure imgf000027_0002
(98%)
Figure imgf000027_0003
Figure imgf000027_0004
REACTION 6 To synthesize β -peptides having reverse turn moiety which is a prolyl-glycolic acid residue, the following protocols are preferred:
(2S,3R)-3-Amino-2-methylpentanoic acid was prepared according to the procedures given by Jefford and McNulty (1994), J. Helv. Chim. Acta 77:2142. However, unlike the description in this paper, the synthesized (2S,3S)-2-methyl-3- (tosylamino)butano-4-lactone contained up to 8 % (2R,3S)-2-methyl-3- (tosylamino)butano-4-lactone as a byproduct, which could be removed by recrystallization from toluene. (2S,3S)-3-Amino-2-benzyl-4-phenylthiobutanoic acid was prepared in a synthetic sequence derived from the one by Jefford and McNulty. This synthesis is described below. Homo-α-amino acids were prepared according to the procedures by Podlech and Seebach (1995), Liebigs Ann. 1217. Depsi- β-peptides were synthesized by conventional dicyclohexylcarbodiimide / N-hydroxysuccinimide (DCC/HOSu) or l-ethyl-3-(3'dimethylaminopropyl)carbodiimidde hydrochloride / N,N-dimethyl-4-aminopyridine (EDCI/DMAP) solution-phase coupling procedures (see, for example, Bodanszky, M. ; Bodanszky, A. The Practice of Peptide Synthesis; Springer Verlag: New York, 1984). Illustrative procedures are given below.
Figure imgf000028_0001
(2S,3S)-2-Benzyl-3-(tosylamino)butano-4-lactone (4). A solution of lithium diisopropylamine (LDA) in THF was generated by adding 1.5 M methyllithium in diethyl ether (30 mL, 45.0 mmol) to a solution of diisopropylamine (6.4 mL, 45.7 mmol) in 100 mL THF at O°C under nitrogen and stirring for 10 min. The solution was then cooled to -78°C, and a solution of (3S)-3-(tosylamino)butano-4-lactone (5.36 g, 21.1 mmol) in 30 mL THF was added dropwise. The resulting yellow solution was stirred for 1 hour at -78°C, and then benzyl bromide (10 mL, 84.1 mmol) was added rapidly. Stirring at -78°C was continued for 2 hours, and the reaction was quenched with 20 mL sat. aq. NH4C1 solution and allowed to warm to room temperature. The mixture was acidified with 1 M HCl and extracted three times with methylene chloride. The combined organic extracts were dried over NajSO,, and evaporated to give an orange semisolid that was purified by chromatography (silica gel, hexane/ethyl acetate 3:2) to yield 2.22 g (8.70 mmol, 41 %) recovered starting material and 3.37 g (9.76 mmol; 46 %) of 4. No diastereomeric addition product could be detected. For further purification 4 can be recrystallized from toluene to give colorless needles, mp. 108.5- 109°C, 'H-NMR (300 MHz, CDC13) δ 7.58 (d, J = 8.5 Hz, 2 H), 7.31 (d, J = 8.2 Hz, 2 H), 7.20 (m, 3 H), 6.92 (dd, J = 7.7, 1.7 Hz, 2H), 4.97 (d, J = 5.5 Hz, 1 H), 4.27 (dd, J = 7.2, 9.8 Hz, 1 H), 3.98 (dd, J = 7.2, 9.8 Hz, 1 H), 3.65 (m, 1 H), 3.00 (m, 1 H), 2.77 (m, 2 H), 2.46 (s, 3 H), 13C-NMR (75.5 MHz, CDC13) δ 144.27 (C), 138.00 (C), 135.83 (C), 129.89 (CH), 128.97 (CH), 128.88 (CH), 127.10 (CH) 71.25 (CH2) 53.21 (CH), 46.54 (CH), 33.54 (CHj) 21.50 (CH3), FJ MS m/e 345.1027 calc. for C18H19NO4S 345.1035.
Figure imgf000029_0001
(2S,3S)-2-Benzyl-4-phenylthio-3-(tosylamino)butanoic acid (7). (2S,3S)-2-Benzyl-3-(tosylamino)butano-4-lactone (4) (0.91 g, 2.64 mmol) was dissolved in 10 mL methylene chloride. At O°C trimethylsilyliodide (1 mL, 7.03 mmol) and anhydrous ethanol (0.72 mL, 12.2 mmol) were added under nitrogen. The solution was stirred 30 min. at 0°C, allowed to warm to room temperature and stirred for 1 day. Then the addition of trimethylsilyliodide and ethanol was repeated and stirring at room temperature was continued for 12 hours. The reaction was quenched by the addition of 3 mL ethanol and stirring for 30 min. To the solution 20 mL of water were added, the layers were separated, and the aqueous layer was extracted five times with methylene chloride. The combined organic extracts were washed with 5 % aq. Na2S2O3 solution, dried over Na2SO4 and concentrated in vacuo to give 1.78 g of crude 5 as an orange solid, which was used in the next step without further purification.
At O°C, thiophenol (0.73 ml, 7.11 mmol) was added to a suspension of NaH (289.7 mg, 7.24 mmol) in 6 mL DMF under nitrogen, warmed to room temperature and stirred for 15 min. A solution of crude 5 (1.78 g) in 10 mL DMF was added to the thiophenolate solution at O°C. After warming to room temperature the solution was stirred for 1 hour. The reaction was quenched with 50 ml water and extracted three times with methylene chloride. The combined organic extracts were washed with brine, dried over Na2SO4 and concentrated in vacuo to give 2.43 g of 6 as a colorless oil, which was used in the next step without further purification.
To a solution of 6 (2.43 g) in 18 mL methanol a 1.5 M aq. NaOH solution was added and the mixture heated to 60 °C for 2 hours. After evaporation of methanol in vacuo, 20 mL water was added and the mixture extracted two times with diethyl ether. The aqueous layer was acidified with cone. HCl and extracted four times with diethyl ether. The organic extracts were dried over Na2SO4 and evaporated to yield 1.04 g (2.28 mmol, 86%) of 7. Η-NMR (300 MHz, CDC13) δ 7.45 (d, J = 8.3 Hz, 2 H), 7.24-7.17 (m, 6 H), 7.09-7.00 (m, 6 H), 5.54 (d, J = 8.3 Hz, NH), 3.46 (m, 1 H), 3.28 (m, 1 H), 3.00 (m, 3 H), 2.67 (dd, J = 7.1, 14.0 Hz, IH), 2.34 (s, 3H).
Figure imgf000030_0001
(2S,3S)-3-Amino-2-Benzyl-4-phenylthiobutanoic acid (8). Compound 7 and phenol (0.77g) were dissolved in 50 mL 48% HBr and heated to reflux for 1.5 hours under nitrogen. After cooling to room temperature 150 mL water was added and the solution extracted two times with diethyl ether. The yellow aqueous layer was evaporated to give 0.58 g of (2S,3S)-3-amino-2-benzyl-4-phenylthiobutanoic acid hydrobromide as an orange solid. Η-NMR (300 MHz, CDC13) δ 7.69 (b, 3 NH), 7.43 (m, 2 H), 7.34-7.01 (m 8 H), 3.60 (m, IH), 3.35 (m, 3 H), 3.08 (dd, J = 8.2, 14.2 Hz, 1 H), 2.87 (dd, J = 7.5, 14.2 Hz, 1 H).
The hydrobromide was dissolved in 140 mL anhydrous ethanol, and 28 mL methyloxirane was added. The solution was heated to reflux for 1 hour under nitrogen. The solvent was evaporated to yield 0.45 g (1.45 mmol, 65%) of 8.
Figure imgf000031_0001
(2S,3S)-3-(t-Butoxycarbonylamino)-2-benzyI-4-phenylthiobutanoic acid.
To a solution of 8 (0.18 g, 0.597 mmol) in 1 mL water and 2 mL dioxane was added K2CO3 (167.9 mg, 1.21 mmol). After cooling to O°C, di-t-butyl-dicarbonate (153.2 mg, 0.681 mmol) was added, the solution warmed to room temperature and stirred for 1 day. The solution was concentrated in vacuo, and the residue dissolved in 20 mL water. The solution was acidified to pH 2-3 (congo red) with 1 M HCl and extracted five times with ethyl acetate. The combined organic extracts were dried over MgSO4 and evaporated to give an orange oil that was purified by chromatography (silica gel, hexane/ethyl acetate 1:2) to yield 63.4 mg (0.159 mmol, 27%) of 9. 1H-NMR (300 MHz, CDC13) δ 7.37-7.13 ( , 10 H), 5.47 (d, J = 8.5 Hz, NH), 3.88 (m, 1 H), 3.20 (m, 1 H), 3.00 (m, 1 H), 2.84 (m, 3 H), 1.39 (s, 9 H), l3C-NMR (75.5 MHz, CDC13) δ 174.82 (C), 156.49 (C), 140.14 (C), 136.80 (C), 130.44 (C), 130.02 (C), 129.81 (C), 129.36 (C), 127.33 (C), 127.27 (C), 79.68 (C), 52.46 (CH), 52.33 (CH), 37.37 (CHj), 35.25 (CHj), 28.55 (3 CH3).
ONHMe BocHN γ
10
MethyI-(2S,3R)-3-(t-butoxycarbonylamino)-2-methylpentanoic amide (10) . (2S,3R)-3-(t-Butoxycarbonylamino)-2-methylpentanoic acid (149.1 mg, 0.645 mmol) was dissolved in 1 mL DMF. At O°C methylamine hydrochloride (88.6 mg, 1.31 mmol) and DMAP (195.7 mg, 1.60 mmol) were added, followed by EDCI (l-ethyl-3- (3' dim ethylaminopropyl)carbodiimidde hydrochloride) (376.9 mg, 1.97 mmol). After stirring at room temperature for 2 days, the solvent was removed in a stream of nitrogen and the residue dried in vacuo. The residue was titurated with 1 mL 1 M HCl and 4 mL water, and the white precipitate was collected by suction filtration to yield 121.0 mg (0.495 mmol, 66%) of the amide 10 mp. 206-207°C, 1H-NMR (300 MHz, CDC13) δ 5.92 (b, NH), 4.72 (b, NH), 3.58 (m, 1 H), 2.77 (d, J = 4.8 Hz, 3 H), 2.45 (m, 1 H), 1.45 (m, 1 H), 1.41 (s, 9 H), 1.40 (m, 1 H), 1.13 (d, J = 7.2 Hz, 3 H), 0.90 (t, J = 7.4 Hz, 3 H), 13C-NMR (75.5 MHz, CDC13) δ 174.83 (C), 156.15 (C), 79.35 (C), 54.62 (CH), 45.02 (CH), 28.35 (3 CH3), 26.24 (CH3), 25.18 (CH^, 13.71 (CH3), 10.85 (CH3), El MS m/e 244.1789 calc. for CI2H24N203 244.1787.
Figure imgf000032_0001
11 12 Compound 12. Compound 10 (121.0 mg, 0.495 mmol) was dissolved in 2 mL of 4 M HCl/dioxane, and the resulting solution was stirred 1 hour at room temperature. HCl/dioxane was then removed in a stream of nitrogen and the deprotected amide dried in vacuo. The activated glycolic ester was prepared by adding EDCI (188.7 mg, 0.635 mmol) to a solution of glycolic acid (45.5 mg, 0.598 mmol) and HOSu (N- hydroxysuccinimide) (72.7 mg, 0.632 mmol) in 1 mLDMF and stirring of the solution at room temperature for 2 hours. The deprotected amide and triethylamine (85 μl, 0.610 mmol) were dissolved in 1 mL DMF and transferred into the activated ester solution. After stirring the resulting solution for 2 days at room temperature, the solvent was removed in a stream of nitrogen and the residue dried in vacuo. The residue was separated by chromatography (silica gel, CHCl3/MeOH 4: 1) to yield impure 11 (192.7 mg), which was used in the next step without further purification.
Compound 11 (192.7 mg) and BOC-L-proline (213.3 mg, 0.991 mmol) were dissolved in 3 mL DMF. DMAP (15.6 mg, 0.128 mmol) was added, followed by DCC (dicyclohexylcarbodiimide) (248.3 mg, 1.20 mmol), and the resulting solution was stirred overnight at room temperature. The white precipitate was filtered off by suction filtration, and the filtrate was concentrated in vacuo. The residue was separated by chromatography (silica gel, CHCl3/MeOH 19: 1) to yield 145.7 mg (0.365 mmol, 74% based on 10) of 12. 1H-NMR (300 MHz, CDC13) δ 7.02 (d, J = 8.6 Hz, NH major rotamer 89%), 6.91 (d, J = 9.0 Hz, NH minor rotamer 11 %), 6.10 (m, NH), 4.78 (AB, A part, J = 15.3 Hz, 1 H), 4.49 (AB, B part, J = 15.3 Hz, 1 H) 4.26 (m, I H), 3.90 (m, 1 H), 3.44 (m, 2 H), 2.74 (d,J = 4.6 Hz, 3 H), 2.45 (quint. , J = 7.0 Hz, 1 H), 2.22 (m, 1 H), 1.98 (m, 2 H), 1.88 (m, 1 H), 1.56 (m, 1 H), 1.43 (s, 9 H), 1.43 (m, 1 H), 1.08 (d, J = 7.0 Hz, 3 H), 0.86 (t, J = 7.4 Hz), 13C-NMR (75.5 MHz, CDC13) δ 174.80 (C), 172.24 (C), 167.47 (C), 154.78 (C), 80.33 (C), 62.79 (CHj), 58.77 (CH), 53.74 (CH), 46.75 (CH2), 45.58 (CH), 29.91 (CH2), 28.26 (3 CH3), 26.05 (CH3), 24.97 (CH2), 24.49 (CH,), 14.43 (CH3), 10.62 (CH3).
Figure imgf000034_0001
13
Compound 13. Compound 12 (12.3 mg, 30.8 μmol) was dissolved in 1 mmol 4 M HCl/dioxane and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. The deprotected depsipeptide and (2S,3S)-2-benzyl-3-(t-butoxycarbonylamino)-4- phenylthiobutanoic acid (9) (14.3 mg, 35.6 μmol) were dissolved in 0.5 mL methylene chloride. DMAP (5.0 mg, 40.9 μmol) was added, followed by EDCI (13.7 mg, 71.5 μmol). After stirring at room temperature for 2 days, the solvent was removed in a stream of nitrogen and the residue dried in vacuo. The residue was titurated with 1 mL water, which was acidified to pH 2. The resulting solid was collected and purified by chromatography (silica gel, CHCl3/MeOH 19: 1) to yield 14.1 mg (20.6 μmol, 67%) of 13. 'H-NMR (300 MHz, CDC13) δ 7.41 (d, J = 10.1 Hz, NH), 7.38-7.13 (m, 10 H + NH), 5.06 (AB, A part, J = 15.3 Hz, 1 H), 5.03 (d, J = 10.5 Hz, NH), 4.45 (m, 1 H), 4.32 (AB, B part, J = 15.5 Hz, 1 H), 4.26 (m, 1 H), 4.02 (t, J = 7.6 Hz, 1 H), 3.31 (m, 1 H), 3.08 (m, 3 H), 2.97 (m, 1 H), 2.83 (m, 1 H), 2.78 (d, J = 4.6 Hz, 3 H), 2.53 (m, 1 H), 2.39 (dq, J = 10.1 Hz, 6.9 Hz, 1 H), 1.98 (m, 1 H), 1.73 (m, 2H), 1.50 (m, 1 H), 1.44 (m, 1 H), 1.41 (s, 9 H), 1.30 (m, 1 H), 1.07 (d, J = 6.9 Hz, 3 H), 0.98 (t, J = 7.4 Hz, 3 H), 13C-NMR (75.5 MHz, CDC13) δ 175.44 (C), 171.63 (C), 167.03 (C), 155.54 (C), 138.29 (C), 135,46 (C), 129.43 (CH), 129.06 (CH), 128.68 (CH), 128.30 (CH), 126.60 (CH), 126.47 (CH), 80.24 (C), 62.69 (CH2), 59.27 (CH), 52.71 (CH), 52.46 (CH), 49.39 (CH), 46.75 (OE J, 46.66 (CH), 38.16 (CH2), 36.32 (CH2), 28.58 (CH2), 28.11 (3 CH3), 26.26 (CH2), 25.85 (CH3), 25.05 (CH2), 16.22 (CH3), 10.47 (CH3).
Figure imgf000035_0001
Compound 1. Compound 13 (14.1 mg, 20.6 μmol) was dissolved in 1 mL 4 M HCl/dioxane and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. The deprotected depsipeptide and triethylamine (5.8 μL, 41.6 μmol) were dissolved in 0.41 mL methylene chloride, and acetic anhydride (2.4 μL, 25.4 μmol) was added. After stirring the solution at room temperature overnight the solvent was removed in a stream of nitrogen and the residue dried in vacuo. The residue was purified by chromatography (silica gel, CHCl3/MeOH 19:1) to yield 9.2 mg (14.7 μmol, 71 %) of 1. mp. 196.5-197°C, Η-NMR (300 MHz, CDC13) δ 7.40 (d, J = 9.0 Hz, NH), 7.39- 7.11 (m, 10 H + NH), 5.99 (d, J = 10.1 Hz, NH), 5.03 (AB, A part, J = 15.3 Hz, 1 H), 4.78 (tt, J = 10.3 Hz, 3.6 Hz, 1 H), 4.34 (AB, B part, J = 15.3 Hz, 1 H), 4.25 (dq, J = 10.0 Hz, 1 H), 4.02 (t, J = 7.4 Hz, 1 H), 3.36 (m, 1 H), 3.20-3.00 (m, 3 H), 2.85-2.75 (m, 2 H), 2.79 (d, J = 4.6 Hz, 3 H), 2.60 (m, 1 H), 2.42 (dq, J = 10.1 Hz, 6.9 Hz, 1 H), 2.00 (m, 1 H), 1.86 (s, 3 H), 1.85-1.62 (m, 3 H), 1.52 (m, 1 H), 1.31 (m, 1 H), 1.07 (d, J = 6.8 Hz, 3 H), 0.97 (t, J = 7.4 Hz, 3 H), 13C-NMR (75.5 MHz, CDC13) δ 175.46 (C), 171.59 (C), 170.17 (C), 167.08 (C), 138.14 (C), 136.87 (C), 135,36 (C), 129.49 (CH), 129.15 (CH), 128.60 (CH), 128.34 (CH), 126.88 (CH), 126.64 (CH), 62.69 (CH2), 59.30 (CH), 52.80 (CH), 51.08 (CH), 48.69 (CH), 46.83 (CH2), 46.28 (CH), 37.37 (CH2), 36.30 (O , 34.45 (CH2), 28.59 (CH2), 26.03 (CH3), 25.07 (CH2), 23.00 (CH3), 16.09 (CH3), 10.46 (CH3), IR (1 mM in CH2C12) 3423, 3367, 1753, 1669, 1626 cm 1, El MS m/e 624.2989 calc. for Cj-H^ S 624.2981. BθCHN^^C0NHMe 14
Methyl-3-(t-butoxycarbonylamino)propionic amide (14). BOC-β-alanine (0.50 g, 2.64 mmol) was dissolved in 4 mL DMF. Methylamine hydrochloride (198 mg, 2.93 mmol) and DMAP (427.2 mg, 3.50 mmol) were added, followed by EDCI (1.06 g, 5.53 mmol). After stirring at room temperature for 2 days the solvent was removed in a stream of nitrogen and the residue dried in vacuo. It was dissolved in 5 mL 1 M HCl, and the solution was extracted five times with ethyl acetate. The combined organic extracts were dried over MgSO4 and concentrated to yield 0.43 g (2.13 mmol, 81 %) of BOC-β-alanine methylamide (14) as a white solid, mp. 117- 118°C, Η-NMR (300 MHz, CDC13) δ 5.78 (b, NH), 5.15 (b, NH), 3.38 (q, J = 6.1 Hz, 2 H), 2.78 (d, J = 4.8 Hz, 3 H), 2.36 (t, J = 6.1 Hz, 2 H), 1.40 (s, 9 H) 13C- NMR (75.5 MHz, CDC13) δ 171.74 (C), 79.15 (C), 36.41 (CHj), 36.03 (CHj), 28.17 (3 CH3), 26.04 (CH3).
Figure imgf000036_0001
15 16
Compound 16. Compound 14 (0.33 g, 1.63 mmol) was dissolved in 5 mL of 4 M HCl/dioxane, and the solution was stirred at 12° C for 1 hour. The HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. An activated ester solution was prepared by adding DCC (509.9 mg, 2.47 mmol) to a solution of glycolic acid (145.7 mg, 1.92 mmol) and HOSu (326.4 mg, 2.84 mmol) in 10 mL methylene chloride. A white precipitate formed after a few minutes. The suspension was stirred at 12°C for 6 hours. The deprotected amide and triethylamine (0.27 mL, 1.94 mmol) were dissolved in 10 mL methylene chloride and transferred into the activated ester solution. After stirring the resulting solution overnight at room temperature, the white precipitate was filtered off by suction filtration and the filtrate concentrated to give a white solid, which was purified by chromatography (silica gel, CHC /MEOH 19: 1) to yield 0.30 g of impure 15, which was used in the next step without further purification.
Compound 15 (0.30 g) and BOC-L-proline (371.5 mg, 1.73 mmol) were dissolved in 50 mL methylene chloride. At O°C DMAP (25.6 mg, 0.210 mmol) was added, followed by DCC (402.9 mg, 1.95 mmol). After stirring 1 hour at O°C the suspension was allowed to warm to room temperature and stirred overnight. The white precipitate was filtered off by suction filtration and the filtrate concentrated. The residue was subjected to chromatography (silica gel, CHCl3/MEOH 19: 1) to yield 0.23 g (0.644 mmol, 40% based on 14) of 15 as a colorless glass. 1H-NMR (300 MHz, CDC13) δ 7.55 (b, NH major rotamer 84%), 7.05 (b, NH minor rotamer 16%), 6.25 (b, NH major rotamer 83 %), 6.04 (b, NH, minor rotamer 17%), 4.59 (s, 2 H), 4.25 (m, 1 H), 3.59 (m, 1 H), 3.42 (m, 3 H), 2.73 (d, J = 4.8 Hz, 3 H), 2.41 (t, J = 6.5 Hz, 2 H), 2.22 (m, 1 H), 1.96 (m, 2 H), 1.88 (m, 1 H), 1.42 (s, 9 H).
Figure imgf000037_0001
17
Compound 17. Compound 16 (0.23 g, 0.644 mmol) was dissolved in 2 mL 4 M HCl/dioxane, and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. The deprotected depsipeptide and BOC-β-alanine (133.3 mg, 0.705 mmol) were dissolved in 5 mL methylene chloride. DMAP (96.9 mg, 0.793 mmol) was added, followed by EDCI (258.7 mg, 1.349 mmol). After stirring at room temperature for 2 days the solvent was removed in a stream of nitrogen. The residue was dissolved in 0.1 M HCl and the solution was extracted four times with methylene chloride. The combined organic extracts were dried over MgSO4 and concentrated to give a white solid that was purified by chromatography (silica gel, CHC13/ MeOH 19:1) to yield 0.18 g (0.420 mmol, 66%) of 17 as a white solid. Η-NMR (300 MHz, CDC13) δ 7.54 (b, NH), 6.30 (b, NH), 5.58 (b, NH), 4.66 (AB, A part, J = 15.4 Hz, 1 H), 4.47 (AB, B part, J = 15.5 Hz, 1 H), 4.35 (m, 1 H), 3.51 ( , 4 H), 3.35 (m, 2 H), 2.72 (d, J = 4.8 Hz, 3 H), 2.51 (m, 2 H), 2.41 (m, 2 H), 2.20 (m, 1 H), 2.10 (m, 1 H), 1.98 (m, 2 H), 1.37 (s, 9 H).
Figure imgf000038_0001
Compound 2. Compound 17 (0.18 g, 0.420 mmol) was dissolved in 2 mL 4 M HCl/dioxane and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. The deprotected depsipeptide and triethylamine (0.12 mL, 0.861 mmol) were dissolved in 5 mL methylene chloride. At O°C acetic anhydride (50 μL, 0.530 mmol) was added and the solution was stirred 1 hour at O°C and then allowed to warm to room temperature with stirring overnight. The solvent was removed in a stream of nitrogen and the residue dried in vacuo. The remaining white solid was purified by chromatography (silica gel, CHCl3/MeOH 19: 1) to yield 0.12 g (0.324 mmol, 77%) of 2 as a white solid, mp. 153.5-154°C, 1H-NMR (300 MHz, CDC13) 7.79 (d, J = 4.4 Hz, NH), 7.32 (d, J = 3.9 Hz, NH), 6.08 (b, NH), 4.75 (AB, A part, J 15.4 Hz, 1 H), 4.44 (AB, B part, J = 15.3 Hz, 1 H), 4.32 (m, 1 H), 3.62-3.40 (m, 5 H), 2.74 (d, J = 4.8 Hz, 3 H), 2.59-2.34 (m, 4 H), 2.25-1.91 (m, 3 H), 1.97 (s, 3 H), 13C- NMR (75.5 MHz, CDC13) δ 171.57 (C), 171.30 (C), 170.63 (C), 167.42 (C), 62.78 (CHj), 59.10 (CH), 47.02 (CH^, 35.83 (CH,), 35.54 (CHj), 34.69 (CH^, 33.75 (CH2), 29.00 (CHj), 26.17 (CH3), 25.05 (CHj), 22.83 (CH3), IR (1 mM in CH^lj) 3452, 3334, 1757, 1669, 1635, 1539 cm 1, El MS m/e 370.1868 calc. for C16H26N4O6 370.1852.
ocH ^°° Me
18 MethyI-(S)-3-(t-butoxycarbonyIamino)butanoic amide (18). BOC- homoalanine (Podlech, J.; Seebach, D. (1995) Liebigs Ann. 1217) (0.44 g, 2.17 mmol) was dissolved in 5 mL methylene chloride. HOSu (376.4 mg, 3.27 mmol) was added and the solution cooled to O°C. After addition of DCC (587.8 mg, 2.85 mmol) the solution was stirred 1 hour at O°C, warmed to room temperature and stirred for an additional 2 hours. A stream of methylamine was bubbled through the suspension for 10 minutes, and stirring was continued overnight. The white precipitate was filtered off by suction filtration and the filtrate concentrated to give a pale yellow solid that was purified by chromatography (silica gel, CHCl3/MeOH 19: 1) to yield 0.41 g (1.90 mmol, 88 %) Of BOC-homoalanine methylamide (18) as a white solid. 1H-NMR (300 MHz, CDC13) δ 6.11 (b, NH), 5.23 (b, NH), 3.92 (m, 1 H), 2.75 (d, J = 4.8 Hz, 3 H), 2.35 (m, 2 H), 1.39 (s, 9 H), 1.17 (d, J = 6.6 Hz, 3 H).
Figure imgf000039_0001
Compound 20. Compound 18 (0.41 g, 1.90 mmol) was dissolved in 2 mL of 4 M HCl/dioxane, and the solution was stirred at room temperature for 1 hour. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. An activated ester solution was prepared by adding DCC (0.59 g, 2.86 mmol) to a solution of glycolic acid (175.5 mg, 2.31 mmol) and HOSu (421.9 mg, 3.67 mmol) in 5 mL DMF at O°C. The suspension was stirred at O°C for 1 hour and then 2 hours at room temperature. The deprotected amide and triethylamine (0.32 mL,
2.30 mmol) were dissolved in 5 mL DMF and transferred into the activated ester solution. After stirring the resulting solution overnight at room temperature the white precipitate was filtered off by suction filtration and the filtrate concentrated to give a semisolid that was chromatographed (silica gel, CHCl3/MeOH 9:1) to yield 0.42 g of impure 19, which was used in the next step without further purification.
Compound 19 (55 mg, 0.317 mmol, impure) and BOC-D-proline (148 mg, 0.688 mmol) were dissolved in 2 mL DMF. DMAP (10.0 mg, 0.082 mmol) was added, followed by DCC (171.4 mg, 0.831 mmol). After stirring the resulting suspension for 1 day at room temperature the white precipitate was filtered off by suction filtration and the filtrate concentrated. The remaining semisolid was purified by chromatography (silica gel, CHCl3/MeOH 19: 1) to yield 52.1 mg (0.140 mmol, 44%) of 20. Η-NMR (300 MHz, CDC13) δ 7.53 (d, J = 6.3 Hz, NH minor rotamer 21 %), 7.30 (d, J = 7.4 Hz, NH major rotamer 79 %), 6.35 (b, NH major rotamer 84%), 6.13 (b, NH minor rotamer 16%), 4.74 (AB, A Part, J = 15.4 Hz, 1 H), 4.44 (AB, B part, J = 15.4 Hz, 1 H), 4.28 (m, 2 H), 3.45 (m, 2 H), 2.72 (d, J = 4.8 Hz, 3 H), 2.41 (dA-B, A part, J =7.4 Hz, 14.3 Hz, 1 H),
2.31 (dAB, B part, J = 5.3 Hz, 14.3 Hz, 1 H), 2.23 (m, 1 H), 1.98 (m, 2 H), 1.88 (m, 1 H), 1.43 (s, 9 H), 1.25 (d, J = 6.8 Hz, 3 H).
Figure imgf000040_0001
21 Compound 21. Compound 20 (52.1 mg, 0. 140 mmol) was dissolved in 1 mL 4 M HCl/dioxane and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. The deprotected depsipeptide and BOC-homophenylalanine (42.5 mg, 0. 152 mmol) were dissolved in 5 mL methylene chloride. DMAP (32.4 mg, 0.265 mmol) was added, followed by EDCI (59.4 mg, 0.310 mmol). After stirring at room temperature for 2 days the solvent was removed in a stream of nitrogen. The residue was dissolved in 0.1 M HCl, and the solution was extracted three times with methylene chloride. The combined organic extracts were dried over MgSO4 and concentrated to give a colorless glass that was purified by chromatography (silica gel, CHCL/MeOH 19: 1) to yield 62.8 mg (0.118 mmol, 84%) of 21. Η-NMR (300 MHz, CDC13) δ 7.36 (b, NH), 7.31-7.12 ( , 5 H), 6.43 (b, NH), 5.22 (b, NH), 4.82 (AB, A part, J = 14.9 Hz, IH), 4.49 (m, 2 H), 4.41 (AB, B part, J = 15.6 Hz, 1 H), 4.21 (m, IH), 3.52 (m, 1 H), 3.32 (m, 1 H), 2.89 (m, 1 H), 2.78 (m, 1 H), 2.71 (d, J = 4.8 Hz, 3 H), 2.46 (m, 3 H), 2.40 (m, 1 H), 2.24 (m, 1 H), 2.12-1.89 (m, 3 H), 1.38 (s, 9 H), 1.25 (d, J = 6.8 Hz, 3 H).
AcHNv/ 1 I-- v-O-N °' N Ϊs/CONH e
O O
Ph" 3
Compound 3. Compound 21 (62.8 mg, 0.118 mmol) was dissolved in 1 mL 4 M HCl/dioxane and the solution was stirred for 1 hour at room temperature. HCl/dioxane was removed in a stream of nitrogen and the residue dried in vacuo. The deprotected depsipeptide and triethylamine (90 μL, 0.646 mmol) were dissolved in 1 mL methylene chloride. At O°C acetic anhydride (35 μL, 0.371 mmol) was added and the solution was stirred 1 hour at O°C and then allowed to warm to room temperature with stirring overnight. The solvent was removed in a stream of nitrogen and the residue dried in vacuo. The residue was purified by chromatography (silica gel, CHCl3/MeOH 19:1) to yield 52.1 mg (0.110 mmol, 93 %) of 3. mp. 128-129°C, 1H-NMR (300 MHz, CDC13) δ 7.49 (d, J = 8.5 Hz, NH), 7.31-7.16 (m, 5 H), 6.80 (d, J = 8.5 Hz, NH), 6.35 (m, NH), 4.73 (AB, A part, J = 15.1 Hz, 1 H), 4.49 (m, 1 H), 4.45 (AB, B part, J = 15.4 Hz, 1 H), 4.38 (m, 2 H), 3.55 (m, 1 H), 3.28 (m, 1 H), 2.99 (dAB, A part, J = 6.2 Hz, 13.4 Hz, 1 H), 2.88 (dAB, B part, J = 8.5 Hz, 13.6 Hz, 1 H), 2.72 (d, J = 4.8 Hz, 3 H), 2.55 (dAB, A part, J = 5.2 Hz, 15.6 Hz, 1 H), 2.43 (dAB, B part, J = 5.9 Hz, 15.5 Hz, 1 H), 2.45 (d, J = 6.3 Hz, 2 H), 2.25 (m, 1 H), 2.21 - 1.89 (m, 3 H), 1.93 (s, 3 H), 1.26 (d, J = 6.6 Hz, 3 H), 13C-NMR (75.5 MHz CDC13) δ 170.90 (C), 170.31 (C), 169.75 (C), 128.93 (CH), 128.32 (CH), 126.40 (CH), 62.60 (CH2), 58.89 (CH), 47.31 (CH2), 42.44 (CH, CH2), 39.57 (OLJ, 36.22 (CH2),28.92 (CH2), 25.96 (CH3), 24.94 (CHj), 23.02 (CH3), 20.21 (CH3), IR (1 mM in CI^Clj) 3452, 3433, 3346 cm"1, El MS m/e 474.2474 calc. for C^H^ A 474.2478.
Construction of polypeptides using any type of β-amino acid can be accomplished using conventional and widely recognized solid-phase or solution- phase synthesis. Very briefly, in solid-phase synthesis, the desired C-terminal amino acid residue is linked to a polystyrene support as a benzyl ester. The amino group of each subsequent amino acid to be added to the N-terminus of the growing peptide chain is protected with Boc, Fmoc, or another suitable protecting group. Likewise, the carboxylic acid group of each subsequent amino acid to be added to the chain is activated with DCC and reacted so that the N-terminus of the growing chain always bears a removable protecting group. The process is repeated (with much rinsing of the beads between each step) until the desired polypeptide is completed. In the classic route, the N-terminus of the growing chain is protected with a Boc group, which is removed using trifluoracetic acid, leaving behind a protonated amino group. Triethylamine is used to remove the proton from the N- terminus of the chain, leaving a the free amino group, which is then reacted with the activated carboxylic acid group from a new protected amino acid. When the desired chain length is reached, a strong acid, such as hydrogen bromide in trifluoracetic acid, is used to both cleave the C-terminus from the polystyrene support and to remove the N-terminus protecting group.
The preferred solid-phase synthesis used herein is shown in Reaction 7:
1. Piperidine 20% in NMP 9 min.
NH, NH-AA,-Fmoc
Fmoc-AA;-OH (3.0 eq.) 2. Fmoc-AA OH (3.0 eq.) PyBOP (3.0 eq.) PyBOP (3.0 eq.) HOBt (3.0 eq.) HOBt (3.0 eq.) DEEA (3.0 eq.) DIEA (3.0 eq.) NMP/CH2Cl2 (3/l, v/v) NMP/CH2Cl, (3/l, v/v) 10 h 10 h
NH-AArAA2-Fmoc NH-(AArAA2-)n-Fmoc
1. Piperidine l. TFA/EDT/H2O 20% in NMP (95 2.5 2.5, v/v/v) 9 min. 2 h
NH-(AA1-AA2-)„-Ac
2. NMP/Ac20/Et3N 2. Precipitation (25/5/1, v/v/v) from ether 1.5 h
Ac-(AA2-AA1)n-NH2
70 - 75% overall yield
REACTION 7
AAn = incoming amino acid to be added to chain
Fmoc = the protecting group 9-fluorenylmethyloxycarbonyl
NMP = N-methyl pyrrolidone
EDT = Ethanedithiol
PyBOP = benzotriazol-1 -yloxytripyrrolidinophosphonium hexafluorophosphate
HOBt = N-hydroxy-benzotriazole
DIEA = diisopropylethyl amine Solid-phase peptide synthesis is widely employed and well know. Consequently, it will not be described in any further detail here.
Solution phase synthesis, noted above, can also be used with equal success. For example, solution-phase synthesis of a β-peptide chain containing alternating residues of unsubstituted cyclohexane rings and amino-substituted cyclohexane rings proceeds in conventional fashion as outlined in Reaction 8:
Figure imgf000044_0001
Water Soluble
REACTION 8 Reaction 8 works with equal success to build peptides wherein the residues are the same or different.
Reaction 9 is an illustration of a homologation reaction combined with conventional solution-phase peptide synthesis which yields a β-peptide having acyclic-substituted residues alternating with ring-constrained residues:
Figure imgf000045_0001
Tetramer Dimer to Tetramer Couplin 80%
TFA
Figure imgf000045_0002
REACTION 9 As noted above, the β-peptides of the present invention can be substituted with any number of substituents, including hydroxy, linear or branched CrC6-alkyl, alkenyl, alkynyl; hydroxy-C,-C6-alkyl, amino-C^ -alkyl, CrC6-alkyloxy, C,-C6- alkyloxy-Cj-C6-alkyl, amino, mono- or di-C,-C6-alkylamino, carboxamido, carboxamido-C,-C6-alkyl, sulfonamido, sulfonamido-C,-C6-alkyl, urea, cyano, fluoro, thio, - -alkyithio, mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-Cr C6-alkyl, heteroaryl- - -alkyl, and combinations thereof. Effecting such substitutions is well within the set of skills possessed by a synthetic peptide chemist.
For example, appending a sulfonamido moiety to the cylic backbone substituent can be accomplished in conventional fashion using Reaction 10.
Compound 63: Compound 61 (90 mg) was dissolved in 4 N HCl in dioxane (2.0 ml). The reaction mixture was stirred for 1.5 hours. The dioxane was then removed in vacuo. The residue was dissolved in pyridine (2.0 ml), then cooled to O° C in an ice-bath.
Methanesulfonylchloride (71 μl) was added dropwise. After the addition, the reaction mixture was stirred at room temperature for 12 hours. The pyridine was then removed in vacuo. The residue was taken up in ethyl acetate (50 ml). The mixture was washed with dilute brine (2 x 10 ml), dried over MgSO4, and concentrated to give the clean product as a colorless oil (70 mg) in 82% yield.
Compound 64: Compound 62 (30 mg) was dissolved in 4 N HCl in dioxane (2.0 ml). The reaction mixture was stirred for 1.5 hours. The dioxane was then removed in vacuo. The residue was dissolved in pyridine (1.0 ml), then cooled to O° C in an ice-bath. Toluenesulfonylchloride (63 mg) was added in portions. After the addition, the reaction mixture was stirred at room temperature for 12 tours. The pyridine was then removed in vacuo. The residue was taken up in methylene chloride/dithyl ether (1/1, v/v, 100 ml). The mixture was washed with dilute brine (3 x 20 ml), dried over MgSO4, and concentrated to give a liquid residue. The crude product was purified by column chromatography with ethyl acetate/hexane (4/6, v/v). Synthesis of Sulfonamides
Figure imgf000047_0001
1.4N HCl in dioxane
2. Tosyichloride Pyridine
Figure imgf000047_0002
Figure imgf000047_0003
74%
REACTION 10 as eluent to give the clean product as a colorless oil (25 g) in 74% yield.
Analogous reactions will append a carboxyamido group. β-peptide residues having a hydroxyalkyl substituent either trans or cis to the peptide backbone carbonyl group (and thereby providing a hydroxy group for further functionalization) can be synthesized as shown in Reactions 11 and 12, respectively:
NaBTt,
Figure imgf000049_0001
Figure imgf000049_0002
Figure imgf000049_0003
Key Intermediate
Figure imgf000049_0004
REACTION 11
Figure imgf000050_0001
Key Intermediate
Figure imgf000050_0002
REACTION 12 Antimicrobial Activity with Low Hemolytic Activity:
As demonstrated in the Examples, pharmaceutical compositions containing the β-peptide compounds described herein are selectively antimicrobial. In short, the compounds exhibit good antimicrobial activity, both MIC and MBC, while simultaneously exhibiting very low hemolytic activity. Consequently, the subject compounds are useful in the formulation of pharmaceutical compositions for the treatment microbial infections because they inhibit microbial growth at low concentrations without lysing human cells.
Pharmaceutical Compositions:
Administration and Pharmaceutical Unit Dosage Forms:
The compounds described herein being effective to inhibit the growth of microbial cells, the compounds are suitable for the therapeutic treatment of microbial infestation (infection, etc.) in mammals, including humans. Microbial cell growth inhibition at pharmacologically-acceptable concentrations has been shown in four different types of bacteria (see Examples 10 and 11).
Administration of the β-peptides to a human or non-human patient can be accomplished by any means known. The preferred administration route is parenteral, including intravenous administration, intraarterial administration, intratumor administration, intramuscular administration, intraperitoneal administration, and subcutaneous administration in combination with a pharmaceutical carrier suitable for the chosen administration route. The treatment method is also amenable to oral administration.
It must be noted, as with all pharmaceuticals, the concentration or amount of the β-peptide administered will vary depending upon the severity of the ailment being treated, the mode of administration, the condition and age of the subject being treated, and the particular β-peptide or combination of β-peptides being used.
The compounds can be administered in the form of tablets, pills, powder mixtures, capsules, injectables, solutions, suppositories, emulsions, dispersions, food premixes, and in other suitable forms. The pharmaceutical dosage form which contains the compounds described herein is conveniently admixed with a non-toxic pharmaceutical organic carrier or a non-toxic pharmaceutical inorganic carrier. Typical pharmaceutically-acceptable carriers include, for example, mannitol, urea, dextrans, lactose, potato and maize starches, magnesium stearate, talc, vegetable oils, polyalkylene glycols, ethyl cellulose, poly(vinylpyrrolidone), calcium carbonate, ethyl oleate, isopropyl myristate, benzyl benzoate, sodium carbonate, gelatin, potassium carbonate, silicic acid, and other conventionally employed acceptable carriers. The pharmaceutical dosage form may also contain non-toxic auxiliary substances such as emulsifying, preserving, or wetting agents, and the like.
Solid forms, such as tablets, capsules and powders, can be fabricated using conventional tabletting and capsule-filling machinery, which is well known in the art. Solid dosage forms may contain any number of additional non-active ingredients known to the art, including excipients, lubricants, dessicants, binders, colorants, disintegrating agents, dry flow modifiers, preservatives, and the like.
Liquid forms for ingestion can be formulated using known liquid carriers, including aqueous and non-aqueous carriers, suspensions, oil-in-water and/or water- in-oil emulsions, and the like. Liquid formulation may also contain any number of additional non-active ingredients, including colorants, fragrance, flavorings, viscosity modifiers, preservatives, stabilizers, and the like.
For parenteral administration, the subject compounds may be administered as injectable dosages of a solution or suspension of the compound in a physiologically- acceptable diluent or sterile liquid carrier such as water or oil, with or without additional surfactants or adjuvants. An illustrative list of carrier oils would include animal and vegetable oils (peanut oil, soy bean oil), petroleum-derived oils (mineral oil), and synthetic oils. In general, for injectable unit doses, water, saline, aqueous dextrose and related sugar solutions, and ethanol and glycol solutions such as propylene glycol or polyethylene glycol are preferred liquid carriers.
The pharmaceutical unit dosage chosen is preferably fabricated and administered to provide a concentration of drug at the point of contact with the microbial cell of from 1 μM to 10 mM. More preferred is a concentration of from 1 to 100 μM. This concentration will, of course, depend on the chosen route of administration and the mass of the subject being treated.
Conformational Analysis - EXAMPLES:
The following Examples are included herein solely to provide a more complete understanding of the invention. The Examples do not limit the scope of the invention disclosed and claimed herein in any fashion.
Example 1: Amide Proton Exchange in trans-ACHC Dimer and Hexamer:
Amide proton exchange is one of the most powerful methods for assessing conformational stability of peptides and proteins; adoption of a stable intramolecularly hydrogen-bonded conformation leads to diminution of the rate of exchange. NH/ND exchange behavior of the trans-ACHC hexamer relative to the corresponding dimer (which is too small to form a favorable internal hydrogen bond) shows that the hexamer adopts a very stable intramolecularly hydrogen- bonded folding pattern in methanol solution.
To ensure a direct comparison, this Example was conducted with solutions containing 2 mM of the trans-ACHC dimer and 2 mM of the trans-ACHC hexamer. The 1H NMR results are shown in Fig. 3. Upon dissolution of the 1: 1 dimer: hexamer mixture in CD3OD, the amide proton and the urethane proton of the dimer (marked with asterisks) are completely exchanged within 6 min, according to 'H NMR (Fig. 3). In contrast, three of the six amide protons of the trans-ACHC hexamer show strong resonances at this point.
One of these protected protons exchanges within a ca. 20 h, but the other two require > 2 days for complete exchange. Thus, two of the amide protons of hexamer display > 100-fold protection from NH/ND exchange with CD3OD, which indicates remarkable conformational stability for this six-residue foldamer in a hydrogen-bonding solvent. The two most protected protons of the hexamer are believed to be the amide groups of residues 2 and 3 (numbering from the N- terminus), although Applicants do not wish to be limited to this interpretation. The amide protons of residues 5 and 6 should exchange rapidly because they cannot be involved in 14-helical hydrogen bonds. The protons of residues 1 and 4 occur at the ends of the 14-helix in the hexamer crystal. The ends of cc-helices in short α- peptides are "frayed" in solution, and similar fraying in the trans-ACHC hexamer would presumably enhance the NH/ND exchange rate. This conclusion is supported by the observation that only one of the NH resonances of the conesponding tetramer can be detected by lH NMR within a few minutes of dissolution in CD3OD, and this proton exchanges completely in less than an hour. Adoption of a stable folding pattern in solution requires the intrinsic rigidity of the trans-ACHC residue; dissolution of an analogous β-alanine hexamer in CD3OD causes all NH groups to exchange within 6 min. This Example shows that trans β-peptide oligomers constructed from an appropriately rigidified residue are highly predisposed to form a specific helix.
Example 2: Circular Dichroism and NOE's in trans-ACPC
As noted above, computational comparisons involving alternative cycloalkane-based β-amino acids and alternative β-peptide helices suggested that the trans-ACHC/ 14-helix combination would generate a stable β-peptide secondary structure. Using the same technique, the trans- ACPCl 12-helix combination was predicted to be almost as favorable. This latter prediction is intriguing because there is no precedent for the 12-helix in the contradictory literature on polymers constructed from optically active β-amino acids. Among these polymers, poly(α-isobutyl-L-aspartate) has been particularly intensively studied, and proposed secondary structures include 14-, 16-, 18- and 20-helix, as well as sheet. Since the computational predictions regarding the trans-ACHC/ 14-helix relationship described above proved to be correct, the trans- ACVCI 12-helix prediction was then explored.
Optically active trans- ACPC was prepared using the protocols described above, and β-peptides were generated via standard coupling methods. A trans - ACPC octamer displays the predicted 12-helical conformation in the solid state; all six of the possible 12-membered ring hydrogen bonds are formed. A tran - ACPC hexamer also displays the predicted 12-helical conformation in the solid state, with all four of the possible 12-membered ring hydrogen bonds formed. In both cases, the regular helix frays at the C-terminus, perhaps because the C-terminal ester group cannot serve as a hydrogen bond donor.
Circular dichroism data for trans- ACPC hexamer in CH3OH (Fig. 4) indicates the adoption of a distinctive secondary structure. The main graph in Fig. 4 depicts two virtually superimposable CD plots: one at a hexamer concentration of 2.0 mM, the other at hexamer concentration of 0.1 mM. Data were obtained on a Jasco J-715 instrument at 20°C using a 1 mm pathlength. The inset graph shows the CD data at 0.1 mM and 0.02 mM using a 5 mm pathlength. The 14-helical conformation in β-peptides composed of acyclic residues has a far-UV CD signature comprising a maximum at ca. 197 nm, a zero crossing at ca. 207 nm, and minimum at ca. 215 nm. In contrast, the CD signature for the trans-ACPC hexamer is clearly different: maximum at ca. 204 nm, zero crossing at ca. 214 nm, and minimum at ca. 221 nm. Since the CD signature of trans- ACPC hexamer in CH3OH does not vary significantly between 2.0 mM and 0.02 mM, it is unlikely that aggregation occurs under these conditions.
Two-dimensional 'H NMR data obtained for the trans-ACPC hexamer in pyridine-d5 and in CD3OH indicate that the 12-helix is highly populated in these solvents. Resonances of all NH groups, all protons at Cp of each trans-ACPC residue, and nearly all protons at Cα of each residue were resolved in pyridine-d5, and could be assigned via a combination of TOCSY and ROESY data. The N- terminal NH (residue 1) could be identified because it was the only one of the six amidic protons that did not show an NOE to CαH of another residue (this NH also showed an NOE to the Boc methyl groups). The C-terminal residue could be identified because it had the only CαH that did not show an NOE to an NH of another residue. Assignment of these terminal resonances allowed us to "walk through" the remaining backbone resonances by virtue of short-range CαH, - NH,+7 NOEs.
The secondary structure of the trans-ACPC hexamer in pyridine-ds is defined by the long-range NOEs summarized in Table 1, below. CpH,- → NH,+2 and CpH, - CαH/+2 NOEs are expected for the 12-helix. All four possible CpH, - NH/+2 NOEs were observed for the ACPC hexamer, as were two of the four CαH/ - CαH/+2 NOEs; NOEs consistent with the other two CpH, - CαH,+2 interactions were observed but could not be unambiguously assigned because of overlap of the CαH resonances of residues 3 and 4. An NOE between NH of residue 3 and the Boc methyl groups provided further evidence for 12-helical folding. In CD3OH, NOEs consistent with all of the long range NOEs detected in pyridine-d5 were observed, and there did not appear to be any additional NOEs in CD3OH. Interpretation of the CD3OH data was less definitive, however, because of greater overlap among proton resonances (especially for CpH of the trans-ACPC residues). The pattern of NOEs in CD3OH was identical for 25 mM and 2.5 mM samples of the trans-ACPC hexamer; chemical shifts and line widths were also invariant between these two concentrations, suggesting that aggregation does not occur under these conditions.
Table 1 Inter-residue NOEs of hexamer 2 in pyridine-d5
Residue H-atom Residue H-atom NOE*
CαH 2 NH Strong
CpH 2 NH Weak
CpH 3 NH Medium
CPH 3 CβH Mediumt
CHafBoc) 3 NH Strong
CpH 4 NH Weak
2 CβH 3 NH Strong
2 CPH 4 NH Medium
2 CpH 4 CβH Mediumt
3 CβH 4 NH Strong
3 CpH 5 NH Medium
3 CpH 5 C«H Me ium
4 CttH 5 NH Strong
4 CpH 6 NH Medium
4 CβH 6 C„H Weak
5 C„H 6 NH Strong
5 CpH 6 NH Weak
" Strong, 2.3 A; medium.3.0 A; weak, 4.0 A. t NOE present, but overlap of C„H of residues 3 and 4 prevents definite assignment.
NOE-restrained molecular dynamics simulations of the trans-ACPC hexamer generated 10 low-energy conformations. Superposition of these conformers shows a high degree of order (RMSD of backbone atoms < 0.2 A), with fraying at the C- terminus). The conformer from this set that is closest to the average agrees quite well with the crystal structure of the hexamer, although there is variation at the C- terminus and in individual cyclopentane ring conformations.
Fig. 5 shows CD data for the dimer, trimer, tetramer, and hexamer of trans- ACPC in methanol. A comparison between the dimer and the hexamer indicates the profound change in secondary structure which takes place as the peptide chain increases in length. The dimer is essentially unstructured. However, the tetramer clearly displays a CD curve indicative of 12-helical secondary structure. This Example shows that short β-peptides of trans-ACPC have a high propensity to adopt the 12-helical folding pattern. In combination with the 14-helix formation by short oligomers of trans-ACHC, the present results indicate that β- peptide secondary structure can be profoundly and rationally altered by manipulating torsional preferences about the Cα-Cp bonds of individual residues.
Example 3: Amide Proton Exchange in Amino-Substituted irøws-ACHA/trans ACHA Dimers and Hexamers:
In this Example, amino-substituted-trans-ACHA (i.e. , ACHA containing an exocyclic amino substituent) was synthesized according to Reaction 3, above. The amino-substituted-trans-ACHA was then coupled with unsubstituted trans-ACHA as shown in Reaction 7 to yield β-peptides wherein the residues alternate between unsubstituted-trans-ACHA and amino-substituted-trans-ACHA. These molecules were synthesized because it was anticipated that the amino group would be protonated in water and that the resulting positive charge would render these β- peptides water-soluble. They are indeed water-soluble.
The amide exchange of the dimer and the hexamer were then compared in the same fashion as described in Example 1 in order to probe for 14-helix formation in water. Fig. 6 depicts the two-dimensional *H NMR data obtained for the alternating unsubstituted-rrans-ACHA/amino-substituted-trans-ACHA hexamer in D20, 100 mM deuteroacetate buffer, pD 3.9. Hydrogen/ deuterium exchange can be examined at five of the six backbone NH groups in this acidic D20 solution. The spectra were taken at room temperature. Five NH peaks are observed (marked with asterisks) in the 8 minute plot. Each peak disappears at a different rate over the course of two days as the NH groups become ND groups. The protons slowest to exchange are those hydrogen bonded and buried in a folded secondary conformation.
As shown in Fig. 7, the rate of amide proton exchange in the dimer had a k^ of 6.6 x 10^/sec. In contrast, the slowest amide proton exchange in the hexamer had a kobs of 1 x lO^/sec, more than 6-fold slower (protection factor = 6.6). In other words, the slowest exchanging NH of the hexamer takes 6.6 times longer to exchange than the NH of the dimer (which is too small to adopt a folded conformation). This Example shows that there is substantial peptide folding of the alternating unsubstituted-trans-ACHA/amino-substituted-trans-ACHA hexamer in aqueous solution. The data strongly suggest that the hexamer is forming a 14-helix.
Example 4: Comparison of CD Data from Amino-Substituted trans- ACHAl trans-ACHA Hexamers and trans-ACHC:
In this Example, the CD data from the alternating hexamer described in Example 3 was compared with a hexamer of unsubstituted trans-ACHC. The trans- ACHC hexamer adopts a 14-helical conformation in methanol solution. Therefore, the CD plot for trans-ACHC should be representational of the 14-helix structure and can serve as a means of comparison for other peptides. Fig. 8 compares the CD data of trans-ACHC hexamer in methanol with the CD data of the alternating unsubstituted-trans- ACHA/amino-substituted-trans- ACHA described in Example 3. The similarity of the two plots provides strong evidence that the alternating hexamer adopts a 14-helical conformation in aqueous solution. In contrast, conventional peptides made from α-amino acids never show evidence of α-helicity where there are fewer than 10 residues.
Examples 3 and 4 in combination show that in addition to the substantial peptide folding indicated by the slow amide proton exchange rate of the alternating hexamer, the CD data strongly indicates that the alternating hexamer adopts a 14- helix secondary structure in aqueous solution.
Example 5: CD Data of β-Peptides Containing Alternating frøns-ACPA Residues and 3-Amino-4-Carboxy-PyrrolidinyI Residues
In this Example, a β-amino acid containing a 3-amino-4-carboxy- pyrrolidinyl residue was synthesized according to Reaction 5 , and coupled into a series of peptides with alternating trans- ACPA residues using conventional coupling methods (Reaction 7). CD data for the tetramer, hexamer, and octamer in this series were then gathered. The results, in methanol, are shown in Fig. 9. Note the distinct and increasing intensity of the characteristic maximum at about 205 nm and minimum at about 221 nm as the chain length increases.
Fig. 10 shows another plot of CD data, in this instance using water as the solvent. Again, the maximum and minimum characteristic peaks increase in intensity with increasing chain length.
Fig. 11 is a direct comparison of the CD data in water versus the CD data in methanol for the alternating 4-pyrrolidinyl/trans-ACPA octamer. These data indicate that the 12-helix formation is only slightly less stable in water than it is in methanol.
Taken together, the CD data presented in this Example strongly indicate that the pyrrolidinyl β-peptides assume a 12-helix conformation in both methanol and aqueous solutions.
Example 6: CD Data of Octamer Containing Alternating frans-ACPA Residues and 3-Amino-2-Carboxy-Pyrrolidinyl residues.
In this Example, a β-amino acid containing a 3-amino-2-carboxy- pyrrolidinyl residue was synthesized according to Reaction 4, and coupled into a peptide with alternating trans- ACPA residues using conventional coupling methods (Reaction 7). CD data for the octamer of this peptide was then gathered. The results, in aqueous solution, are shown in Fig. 12. This β-peptide clearly adopts the 12-helix conformation in water as evidenced by the characteristic maximum at about 205 nm and the characteristic minimum at about 221 nm.
This Example shows that the nitrogen heteroatom introduced in the pyrrolidinyl moiety can be located in different postions along the heterocyclic ring without adversely affecting the formation of the 12-helical structure in solution.
Example 7: Comparison of Secondary Structure in Amino-Subsituted-trans- ACHC and β-Peptides Containing Aliphatic-Substituted Residues In this Example, the CD spectrum of a β-peptide containing alternating residues of trans-ACHA and amino-substituted-trans-ACHA was compared to the CD spectrum of a "mixed" β-peptide comprising alternating residues of trans- ACHA and an acyclic β-amino acid bearing an aminopropyl substituent on the β- carbon of the backbone. The acyclic β-amino acid was synthesized and coupled to the trans-ACHA residue as detailed in Reaction 9, described above.
The CD data are presented in Fig. 13. While the removal of 3 cyclohexyl units clearly diminishes the extent of helix formation, the extent of helix formation in the mixed β-peptide is still significant. The data presented here indicate that β- peptides which include acyclic residues along with ring-constrained residues will adopt moderately stable secondary structures in solution.
Example 8: Infrared Analysis of Reverse Turn Using Linked Nipecotic Acid Moieties
In this Example, two nipecotic acid residues were linked to each other to act as a reverse tum moiety. This was done using Reaction 6. Fig. 14A shows the IR spectrum for the diastereomer in which the two nipecotic acid residues have the same absolute configuration and Fig. 14B shows the IR spectrum for the diastereomer in which the two nipecotic acid residues have opposite absolute configuration. Both samples were taken in dilute solution (to minimize intermolecular hydrogen bonding) with solvent subtraction. As is clear from the diastereomer shown in Fig. 14A, there is little intramolecular hydrogen bonding as evidenced by the large peak at 3454 cm'1 which is due to N-H units not involved in hydrogen bonding. This was as predicted from computer modeling studies.
In contrast, the spectrum shown in Fig. 14B for the heterochiral diastereomer exhibits a very large peak at 3350 cm"1, indicative of hydrogen-bonded N-H groups.
This Example shows that synthetic reverse turn moieties can be constructed from two nipecotic β-amino acid residues having opposite configuration. Example 9: X-ray Crystallography of Synthetic Reverse Turn
Compound 1 was synthesized as described above. Crystals of compound 1 suitable for X-ray analysis were obtained by vapor diffusion (over 2 weeks) of n- heptane into a solution of the sample in ethyl acetate. The data were collected on a Siemens P4/CCD diffractometer running software provided by the manufacturer. The ball and stick schematic of the crystal stracture in the solid state is presented in Fig. 16. All hydrogens except those bonded to nitrogen have been removed for clarity. Hydrogen bonds are shown in dotted lines.
Combinatorial Chemistry:
The defined conformation conferred by the β -polypeptides described herein makes these polyamide compounds highly useful for constructing large libraries of potentially useful compounds via combinatorial chemistry. Combinatorial exploration of functionalized oligomers of, for instance, trans-ACHC, trans-ACPC, or β-peptides wherein the α and β carbons are part of a cycloalkenyl ring or a heterocyclic ring such a pyrrolidinyl or piperidinyl moiety, or "mixed" β-peptides containing acyclic residues in addition to cyclic residues, has a potential yield of literally millions of novel polypeptide molecules, all of which display a well- defined helical or sheet secondary structure.
Of particular note here is that the equatorial positions of the cyclohexyl β- peptides can be substituted with virtually any substituent, including very large substituents, without disrupting the helical secondary structure. At least in helical structures, this is because any equatorial substituent extends essentially perpendicular from the axis of rotation of the helix, thereby leaving the hydrogen bonds of the helix undisturbed.
The amino acids which comprise the finished peptides can be functionalized prior to being incorporated into a polypeptide, or an unfunctionalized polypeptie can be constructed and then the entire oligomer functionalized. Neither method is preferred over the other as they are complementary depending upon the types of compounds which are desired.
Combinatorial libraries utilizing the present compounds may be constructed using any means now known to the art or developed in the future. The preferred methods, however, are the "split and pool" method using solid-phase polypeptide synthesis on inert solid substrates and parallel synthesis, also referred to as multipin synthesis.
The "split and pool" concept is based on the fact that combinatorial bead libraries contain single beads which display only one type of compound, although there may be up to 1013 copies of the same compound on a single 100 μm diameter bead. The process proceeds as follows, utilizing standard solid-phase peptide synthesis as described above:
Several suitable solid substrates are available commercially. The substrates are generally small diameter beads, e.g. about 100 μm, formed from inert polymeric materials such as polyoxyethylene-grafted polystyrene or polydimethylacrylamide. An illustrative substrate, marketed under the trademark "ARGOGEL" is available from Argonaut Technologies, Washington, D.C.
Referring now to Fig. 15, which is a schematic depicting the split and pool method, a plurality of inert substrates are divided into two or more groups and then a first set of subunits is covalently linked to the inert support. As depicted in Fig. 15, the initial plurality of substrates is divided into three subgroups. The appearance of the three groups of beads after the first round of coupling is shown at I of Fig. 15. The three groups of beads are then pooled together to randomize the beads. The beads are then again split into a number of subgroups. Another round of coupling then takes place wherein a second subunit is bonded to the first subunit already present on each bead. The process is then repeated (theoretically ad infinitum) until the desired chain length is attained.
The split and pool process is highly flexible and has the capability of generating literally millions of different compounds which, in certain applications, can be assayed for activity while still attached to the inert substrate. A critical aspect of the split and pool methodology is that each reaction be driven to completion to prior to initiating a subsequent round of coupling. So long as each coupling reaction is driven to completion, each substrate bead will only display a single compound. Because the rate of reaction will differ from bead to bead as the library construction progresses, the beads can be monitored using conventional dyes to ensure that coupling is completed prior to initiating another round of synthesis. The presence of only a single compound per bead comes about because each individual bead encounters only one amino acid at each coupling cycle. So long as the coupling cycle is driven to completion, all available coupling sites on each bead will be reacted during each cycle and therefore only one type of peptide will be displayed on each bead.
The resulting combinatorial library is comprised of a plurality of inert , substrates, each having covalently linked thereto a different β -polypeptide. The polypeptides can be screened for activity while still attached to the inert support, if so desired and feasible for the activity being investigated. Beads which display the desired activity are then isolated and the polypeptide contained thereon characterized via conventional peptide chemistry, such as the Edman degradation. Where a solution-phase assay is to be used to screen the library, the polypeptides are cleaved from the solid substrate and tested in solution.
As applied in the present invention, one or more of the subunits coupled to the inert substrate are selected from the β-amino acids described herein. In this fashion, large libraries of β -polypeptides can be assembled, all of compounds contained therein which display predictable secondary structure.
An alternative approach to generating combinatorial libraries uses parallel synthesis. In this approach, a known set of first subunits is covalently linked to a known location on a inert substrate, one subunit type to each location. The substrate may be a series of spots on a suitable divisible substrate such as filter paper or cotton. A substrate commonly used is an array of pins, each pin being manufactured from a suitable resin, described above. After the initial round of coupling, each pin of the array bears a first subunit covalently linked thereto. The array is then reacted with a known set of second subunits, generally different from the first, followed by reactions with a third set of subunits, and so on. During each reiteration, each individual pin (or location) is coupled with a incoming subunit selected from a distinct set of subunits, with the order of the subunits being recorded at each step. The final result is an array of polypeptides, with a different polypeptide bonded to each solid substrate. Because the ordering of the subunits is recorded, the identity of the primary sequence of the polypeptide at any given location on the substrate (i.e. , any given pin) is known. As in the split and pool method, each coupling reaction must be driven to completion in order to ensure that each location on the substrate contains only a single type of polypeptide.
Large Molecule Interactions:
Another use for the present compounds is as molecular probes to investigate the interactions between biological macromolecules to identify antagonists, agonists, and inhibitors of selected biological reactions. As noted above, many biological reactions take place between very large macromolecules. The surface areas in which these reactions take place are thought by many to be far too large to be disrupted, altered, or mimicked by a small molecule. Until the present invention, it has been difficult, if not impossible, to manufacture molecular probes of modest size that display a well-defined conformation. Because the compounds described herein assume a highly predictable helical or sheet conformation, even when functionalized, they find use as reagents to probe the interaction between large biomolecules.
Employing the combinatorial methods described herein greatly expands the medicinal application of the compounds as vast libraries of compounds can be screened for specific activities, such as inhibitory and antagonist activity in a selected biological reaction. Antimicrobial Activity/Non-Hemolytic Activity - EXAMPLES 10 and 11:
Example 10: Oligomers of (R,R)-trαns-2-aminocyclopentanecarboxylic acid (ACPC) adopt a helix defined by 12-membered ring having "C=0 — H-N" hydrogen bonds between each backbone carbonyl and the amide proton of the third residue in the C-terminal direction. This secondary structure is referred to herein as the " 12-helix. " (For comparison, the familiar α-helix in conventional peptides is defined by 13-membered ring with C=0— H-N hydrogen bonds between each backbone carbonyl and the amide proton of the fourth residue in the C-terminal direction.) The β-peptide 12-helix has approximately 2.5 residues per turn.
A protected version of (R,R)-rrαns-4-aminopyrrolidine-3-carboxylic acid (APC) was prepared from the known β-ketoester as shown in Reaction 5 (compound 58). After incoφoration into a β-peptide and removal of the Boc protecting group from the ring nitrogen atom, the APC residue should be cationic at pH ≤ 8. The APC derivative shown in Reaction 5 and Fmoc-protected ACPC were used for solid-phase synthesis of oligomers β-7, β-12 and β-17:
Figure imgf000066_0001
Each β-peptide bears an acetyl capping group at the N-terminus and a primary amide at the C-terminus. These β-peptides were designed with the intent that they would form amphiphilic 12-helices in aqueous solution, with all cationic APC residues on one side of the helix and all hydrophobic ACPC residues on the other side.
The activities of β-7, β-12 and β-17 against four bacteria were assessed in liquid medium. Also examined, as a positive control, was a synthetic magainin derivative (GIGKFLHAAKKFAKAFVAETMNS-NH2) (SEQ. ID. NO. 1) that displays enhanced antimicrobial activity relative to the natural magainins. Enterococcus faecium A436 (Nicus et al. , Characterization of vancomucin resistance Enterococcus faecium and Enterococcus faecalis , Antimicrob. Agents Chemother. 33:11121-24 (1989)) and Staphylococcus aureus 1206 (Weisblum et al , Erythromycin-inducible resistance in Staphylococcus aurens: survey of antibiotic cases involved, /. BacterioL , 98:447-52 (1969)) are clinical isolates, while Bacillus subtilis BR151 (Young et al , Chromosomal location of genes regulating resistance to bacteriophage in Bacillus subtilis, J. BacterioL, 98:2087-97 (1969)) and Escheήchia coli JM109 (Yanisch-Perron et al. , Improved M13 phage cloning vectors and host strains: nucleotide sequences of the M13mpl8 and pUC19 vectors, Gene 33: 103-19 (1985)) are non-pathogenic strains commonly used in the laboratory for genetic constructions. (Pathogenic strains of E. coli are well known; while B. subtilis itself shows little tendency to colonize humans, a related species, B. anthracis, causes anthrax.)
Preliminary studies showed that the antibacterial activity of the β-peptides against all four bacteria improves steadily as the length increases, with β-17 comparable to the magainin. β-7 showed some, albeit very small, activity against all of the four bacteria, but β-12 was moderately active against B. subtilis and E. faecium, and showed detectable activity against E. coli and S. aureus. Table 2 provides minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) data for β-17 and the magainin.
As shown in Table 2, β-17 is potent against all four bacteria, and this β- peptide matches or exceeds the magainin in all four cases. The sizes of β-17 and the magainin are similar: there are 64 carbon and nitrogen atoms in the backbone of β-17 and 72 carbon and nitrogen atoms in the backbone of the magainin. If the 23- residue magainin were fully folded into an α-helix (3.6 residues per turn), this molecule would form roughly 6.4 helical turns and have a length of roughly 34 A. If the 17-residue β-peptide were fully folded into a 12-helix, the molecule would form roughly 6.8 helical turns and have a length of roughly 37 A. In this example, the β-7, β-12 and β-17 were used in the trifluoroacetate salt forms (trifluoroacetic acid was a component of the eluent used for HPLC purification). Therefore, control experiments were run to determine whether trifluoroacetate exerts any antibacterial effect. Addition of up to 50 mM sodium trifluoroacetate to the growth media had no effect on the growth of any of the four bacteria used in this Example. The conclusion, therefore, is that the activities summarized in Table 2 result from the β-peptides themselves rather than their counterions.
Example 11 : Therapeutic utility requires that a cationic peptide act selectively against bacterial cells in the presence of human cells. This issue is typically probed by comparing red blood cell lytic activity ("hemolysis") with antimicrobial activity. Magainins display low hemolytic activity, but other natural cationic helix-forming peptides are highly hemolytic. For example, the 26-residue peptide melittin, isolated from honey bee venom, readily lyses both bacterial and human red blood cells. Fig. 17 shows the results of a direct hemolysis comparison between melittin (a positive control), the magainin derivative, and β-17. Significant hemolysis occurs at 50 μg/ml melittin, and hemolysis is complete by 100 μg/ml. In contrast, the magainin derivative displays hemolytic activity only above 100 μg/ml, and β-17 is even less hemolytic than the magainin.
To generate the data shown in Fig. 17, human red blood cells (1 % suspension in PBS) were incubated at room temperature for 1 h with a two-fold serial dilution of peptide in PBS . Release of hemoglobin was measured by determining the OD415 of the supernatant after centrifugation. Controls consisted of human red blood cells suspended in PBS (zero hemolysis) and suspended in 1 % SDS (100% hemolysis).
These results clearly show that the subject compounds with a completely unnatural backbone have a specific and very useful antimicrobial activity. Table 2. Bacteriostatic" and Bactericidal* Activities of β-17 and (Ala* 13 18)-
Magainin II amide (μg/ml)
Figure imgf000069_0001
"Minimal inhibitory concentration (MIC) is defined here as the lowest concentration of peptide required for complete inhibition of growth as determined by optical density at 590 nm. Bacterium in BHI medium (~106 CFU/ml) were incubated for 6 h at 37 °C with a two-fold dilution series of peptide in medium in a sterile 96-well plate. Microbial growth was determined by the increase in OD590 over the 6 h period.
^Minimal bactericidal concentration (MBC) refers to the lowest concentration of peptide required for the absence of viable colonies. The bactericidal activities of the MIC, MICx2, and MICx4 were examined. The well solution was diluted and plated in order to allow a maximum of ~103 colony-forming units to grow on the agar plate. Plates for t0 contained approximately 1000 colonies. The plates were incubated overnight at 37° C, and colonies were counted for determination of bactericidal activity.
Teptide concentration of 100 μg/ml revealed 10 colonies ("99% killing).
It is understood that the invention is not confined to the particular reagents, reactions, and methodologies described above, but embraces all modified and equivalent forms thereof as come within the scope of the following claims.

Claims

CLAIMSWhat is claimed is:
1. A pharmaceutical composition characterized by an antimicrobial-effective amount of a β-amino acid oligomer or polymer wherein each residue of the oligomer or polymer is a β-amino acid residue comprising formula:
Figure imgf000070_0001
wherein p is an integer greater than 6; wherein X is independently selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, mono- or bicyclic heteroaryl-C ,-C6- alkyl, -(CH2)n+1- OR, -(CH2)n+1-SR, -(CH2)n+rS(=0)-CH2-R, -(CH2)n+1-S(=0)2-CH2- R, -(CH2)n+1-NRR, -(CH2)n+1-NHC(=0)R,-(CH2)n+1-NHS(=0)2-CH2-R, -(CH2)n+1-0-(CH2)n-R1, -(CH^.-S-fCH^-R1, -(CH2)n+1-S(=0)-(CH2)m-R\ -(CH2)n+1-S( = 0)2-(CH2)m-R1, -(CH2)n+1-NH-(CH2)m-R1, -(CH2)n+1-N-{(CH2)m-R1}2, -(CH2)n+rNHC(=0)-(CH2)n+1-R\ and -(CH2)n+1-NHS(=0)2-(CH2)m-R1; wherein R is independently selected from the group consisting of hydrogen, C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl- -Ce-alkyl, mono- or bicyclic heteroaryl-C ,-C6- alkyl; and wherein R1 is independently selected from the group consisting of hydroxy, C,-C6-alkyloxy, aryloxy, heteroaryloxy, thio, C,-C6-alkylthio, C,- C6-alkylsulfιnyl, C,-C6-alkylsulfonyl, arylthio, arylsulfinyl, arylsulfonyl, heteroarylthio, heteroarylsulfinyl, heteroarylsulfonyl, amino, mono- or di- C,-C6-alkylamino, mono- or diarylamino, mono- or diheteroarylamino, N- alkyl-N-arylamino, N-alkyl-N-heteroarylamino, N-aryl-N-heteroarylamino, aryl-C,-C6-alkylamino, carboxylic acid, carboxamide, mono- or di-C,-C6- alkylcarboxamide, mono- or diarylcarboxamide, mono- or diheteroarylcarboxamide, N-alkyl-N-arylcarboxamide, N-alkyl-N- heteroarylcarboxamide, N-aryl-N-heteroarylcarboxamide, sulfonic acid, sulfonamide, mono- or
Figure imgf000071_0001
mono- or diarylsulfonamide, mono- or diheteroarylsulfonamide, N-alkyl-N- arylsulfonamide, N-alkyl-N-heteroarylsulfonamide, N-aryl-N- heteroarylsulfonamide, urea; mono- di- or tri-substituted urea, wherein the subsitutent(s) is selected from the group consisting of C,-C6-alkyl, aryl, heteroaryl; O-alkylurethane, O-arylurethane, and O-heteroarylurethane; wherein Y is independently selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-CrC6-alkyl, mono- or bicyclic heteroaryl-Cj- -alkyl, -(CH^-OR, -(CH2)n-SR, -(CH2)n-S( = 0)-CH2-R, -(CH2)„-S(=0)2-CH2-R, -(CH2)n-NRR, -(CH2)n-NHC( = 0)R, -(CH2)n-NHS( = 0)2-CH2-R, -(CH2)n-0-(CH2)m-R1, -(CH2)n-S-(CH2)m-R', -(CH2)n-S(=0)-(CH2)m-R', -(CH2)n-S(=0)2-(CH2)m-R', -(CH2)n-NH-(CH2)m-R1, -(CH2)n-N-{(CH2)m-R1}2-(CH2)n-NHC(=0)-(CH2)m-R,, and -(CH2)n-NHS(=0)2-(CH2)m-R, ; wherein R and R1 are as defined hereinabove; or X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted or unsubsituted C3-C8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more N, O or S atom(s) as the heteroatom(s); the substituents on carbon atoms of the rings being independently selected from the group consisting of linear or branched C,-C6-alkyl, alkenyl, alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,- C6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, and the substituents listed above for X and Y when X and Y are not combined; the substituents on nitrogen heteroatoms of the rings being independently selected from the group consisting of -S(=0)2-CH2-R,
-C(=0)-R, -S(=0)2-(CH2)m-R1, -C(=0)-(CH2)n+1-R1; wherein R and R* are as defined hereinabove; m is an integer of from 2-6 and n is an integer of from 0-6;
W is hydrogen or an amino-terminal capping group;
Z is hydroxy or a carboxy-terminal capping group; and further wherein in at least one residue, X and Y are combined in a cyclic moiety; and pharmaceutically-suitable salts thereof; in combination with a pharmaceutically-suitable carrier.
2. The composition according to claim 1, wherein X is selected from the group consisting of hydrogen, linear or branched Cι-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, -(CH2)n+1- OR, -(CH2)n+1-SR, -(CH2)n+1-S(=0)-CH2-R, -(CH2)n+1-S(=0)2-CH2- R, -(CH2)n+1-NRR, -(CH2)n+1-NHC(=0)R,-(CH2)n+1-NHS(=0)2-CH2-R, -(CH2)n+rO-(CH2)m-R\ -(CH2)n+ 1-S-(CH2)m-R1, -(CH2)n+^S( = 0)-(CH2)m-R,, -(CH2)n+1-S(=0)2-(CH2)m-R1, -(CH2)n+1-NH-(CH2)m-R,, -(CH2)n+I-N-{(CH2)m-R1}2, -(CH2)n+rNHC(=0)-(CH2)n+1-R\ and -(CH2)n+1-NHS(=0)2-(CH2)m-R1; and wherein Y is selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, -(CH2)n-OR, -(CH2)n-SR, -(CH2)n-S(=0)-CH2-R, -(CH2)n-S(=0)2-CH2-R, -(CH2)n-NRR, -(CH2)n-NHC(=0)R, -(CH2)n-NHS(=0)2-CH2-R, -(CH2)n-0-(CH2)m-R\ -(CH2)n-S-(CH2)m-R\ -(CH2)n-S(=0)-(CH2)m-R\ -(CH2)n-S( = 0)2-(CH2)m-R\
Figure imgf000072_0001
-(CH2)n-NHS( = 0)2-(CH2)m-R1.
3. The composition according to claim 1 or 2, wherein X and Y are independently selected from the group consisting of linear or branched C,-C6-alkyl, hydroxy- -Cg-alkyl, amino-C C6-alkyl, aryl-C^ -alkyl, heteroaryl-C, -C6-alkyl, and carboxyl- - -alkyl.
4. The composition according to any one of the preceding claims, wherein X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted or unsubsituted C3-C8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more N, O or S atom(s) as the heteroatom (s).
5. The composition according to any one of the preceding claims, wherein X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted or unsubstituted C5-C6 cycloalkyl, cycloalkenyl, or heterocyclic ring having one nitrogen atom as the sole heteroatom.
6. The composition according to any one of the preceding claims, wherein X and Y, together with the carbons to which they are bonded, independently define a substituted or unsubstituted cyclopentyl, substituted or unsubstituted cyclohexyl, unsubstituted or N-substituted piperidinyl, or unsubstituted or N-substituted pyrrolidinyl.
7. The composition according to any one of the preceding claims, wherein X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted cyclopentyl, cyclohexyl, pyrrolidinyl, or piperidinyl ring, wherein the subsituent is selected from the group consisting of amino, mono- or di-C,-C6-alkylamino, carboxamido, sulfonamido, urea, thio, and C,-C6-alkylthio.
8. The composition according to any one of the preceding claims, wherein X and Y combined, together with the carbon atoms to which they are bonded, independently define an amino-substituted cyclopentyl, amino-substituted cyclohexyl, N-substituted pyrrolidinyl, or N-substituted piperidinyl ring.
9. The composition according to any one of the preceding claims, wherein W is selected from the group consisting of hydrogen, formyl, acetyl, tBoc, and Fmoc; and Z is selected from the group consisting of hydroxy, NH2, NH(alkyl), and N(alkyl)2.
10. A β-amino acid oligomer or polymer characterized by formula:
Figure imgf000074_0001
wherein p is an integer greater than 6; wherein X is independently selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-CrC6-alkyl, mono- or bicyclic heteroaryl-CrC6-alkyl, -(CH2)n+I- OR, -(CH2)n+1-SR, -(CH2)n+ rS(=0)-CH2-R, -(CH2)n+1-S(=0)2-CH2- R, -(CH2)n+1-NRR, -(CH2)n+ 1-NHC( = 0)R,-(CH2)n+I-NHS(=0)2-CH2-R, -(CH2)n+rO-(CH2)m-R\ -(CH2)n+1-S-(CH2)m-R\ -(CH2)n+rS( = 0)-(CH2)m-R\ -(CH2)0+1-S(=0)2-(CH2)m-Rl, -(CH2)n+1-NH-(CH2)m-R1, -(CH2)rl+ I-N-{(CH2)m-R1}2, -(CH2)n+1-NHC( = 0)-(CH2)α+1-R1, and -(CH2)n+1-NHS(=0)2-(CH2)m-R1; wherein R is independently selected from the group consisting of hydrogen, C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, mono- or bicyclic heteroaryl-C,-C6- alkyl; and wherein R1 is independently selected from the group consisting of hydroxy, CrC6-alkyloxy, aryloxy, heteroaryloxy, thio, C,-C6-alkylthio, C,- C6-alkylsulfιnyl, C,-C6-alkylsulfonyl, arylthio, arylsulfinyl, arylsulfonyl, heteroarylthio, heteroarylsulfinyl, heteroarylsulfonyl, amino, mono- or di- C,-C6-alkylamino, mono- or diarylamino, mono- or diheteroarylamino, N- alkyl-N-arylamino, N-alkyl-N-heteroarylamino, N-aryl-N-heteroarylamino, aryl-C,-C6-alkylamino, carboxylic acid, carboxamide, mono- or di-C,-C6- alkylcarboxamide, mono- or diarylcarboxamide, mono- or diheteroarylcarboxamide, N-alkyl-N-arylcarboxamide, N-alkyl-N- heteroarylcarboxamide, N-aryl-N-heteroarylcarboxamide, sulfonic acid, sulfonamide, mono- or di-C,-C6-alkylsulfonamide, mono- or diarylsulfonamide, mono- or diheteroarylsulfonamide, N-alkyl-N- arylsulfonamide, N-alkyl-N-heteroarylsulfonamide, N-aryl-N- heteroarylsulfonamide, urea; mono- di- or tri-substituted urea, wherein the subsitutent(s) is selected from the group consisting of C,-C6-alkyl, aryl, heteroaryl; O-alkylurethane, O-arylurethane, and O-heteroarylurethane; wherein Y is independently selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, -(CH2)n-OR, -(CH2)n-SR, -(CH2)n-S(=0)-CH2-R, -(CH2)n-S(=0)2-CH2-R, -(CH^-NRR, -(CH2)n-NHC(=0)R, -(CH2)n-NHS(=0)2-CH2-R, -(CH2)n-0-(CH2)m-R1, -(CH^-S-^H,)^1, -(CH2)n-S(=0)-(CH2)m-R1, -(CH2)n-S( = 0)2-(CH2)m-R1, -(CH2)n-NH-(CH2)m-R1, -(CH2)n-N-{(CH2)m-R1}2 -(CH2)n-NHC(=0)-(CH2)m-R1, and -(CH2)n-NHS(=0)2-(CH2)m-R1; wherein R and R1 are as defined hereinabove; or X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted or unsubsituted C3-C8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more N, O or S atom(s) as the heteroatom(s); the substituents on carbon atoms of the rings being independently selected from the group consisting of linear or branched C,-C6-alkyl, alkenyl, alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteraryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,- C6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, and the substituents listed above for X and Y when X and Y are not combined; the substituents on nitrogen heteroatoms of the rings being independently selected from the group consisting of -S(=0)2-CH2-R, -C(=0)-R, -S(=0)2-(CH2)m-R1, -C(=0)-(CH2)n+1-R1; wherein R and R1 are as defined hereinabove; m is an integer of from 2-6 and n is an integer of from 0-6; W is hydrogen or an amino-terminal capping group; Z is hydroxy or a carboxy-terminal capping group; and further wherein in at least one residue, X and Y are combined in a cyclic moiety, and salts thereof.
11. The β-amino acid oligomer or polymer according to claim 10, wherein X is selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-CrC6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, -(CH2)n+1- OR, -(CH2)n+rSR, -(CH2)n+1-S(=0)-CH2-R, -(CH2)n+rS(=0)2-CH2- R, -(CH2)n+1-NRR, -(CH2)n+rNHC( = 0)R,-(CH2)n+1-NHS(=0)2-CH2-R, -(CH^^-O-^H^^R -^H^^-S-^H,)^1, -(CH2)n+1-S( = 0)-(CH2)m-R\ -(CH2)n+1-S(=0)2-(CH2)m-R1, -(CH2)n+rNH-(CH2)m-R1, -(CH2)n+ 1-N-{(CH2)m-R,}2, -(CH2)n+1-NHC(=0)-(CH2)n+1-R\ and -(CH2)n+1-NHS( = 0)2-(CH2)m-R1; and wherein Y is selected from the group consisting of hydrogen, linear or branched C,-C6-alkyl, alkenyl, or alkynyl; mono- or bicyclic aryl, mono- or bicyclic heteroaryl having up to 5 heteroatoms selected from N, O, and S; mono- or bicyclic aryl-C,-C6-alkyl, mono- or bicyclic heteroaryl-C,-C6-alkyl, -(CH2)n-OR, -(CH2)n-SR, -(CH2)n-S(=0)-CH2-R, -(CH2)n-S(=0)2-CH2-R, -(CH2)n-NRR, -(CH2)n-NHC( = 0)R, -(CH2)n-NHS(=0)2-CH2-R, -(CH2)B-0-(CH2)n-R1, -(CH2)n-S-(CH2)m-R' , -(CH2)n-S( = 0)-(CH2)„-R1 , -(CH2)n-S ( = 0)2-(CH2)m-R1 , -(CH2)n-NH-(CH2)m-R,, -(CH2)n-N-{(CH2)m-R,}2 -(CH2)n-NHC(=0)-(CH2)m-R1, and -(CH2)n-NHS(=0)2-(CH2)m-R1.
12. The β-amino acid oligomer or polymer according to any one of claims 10 or 11 , wherein X and Y are independently selected from the group consisting of linear or branched - -alkyl, hydroxy-Cj-C6-alkyl, amino-C,-C6- alkyl, aryl-C,-C6-alkyl, heteroaryl-C,-C6-alkyl, and carboxyl-C,-C6-alkyl.
13. The β-amino acid oligomer or polymer according to any one of claims 10, 11, or 12, wherein X and Y combined, together with the carbon atoms to which they are bonded, independently define a substituted or unsubsituted C3-C8 cycloalkyl, cycloalkenyl or heterocyclic ring having one or more N, O or S atom(s) as the heteroatom (s).
PCT/US2001/022801 2001-07-18 2001-07-18 ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS WO2003008439A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2001/022801 WO2003008439A1 (en) 2001-07-18 2001-07-18 ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2001/022801 WO2003008439A1 (en) 2001-07-18 2001-07-18 ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS

Publications (1)

Publication Number Publication Date
WO2003008439A1 true WO2003008439A1 (en) 2003-01-30

Family

ID=21742713

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/022801 WO2003008439A1 (en) 2001-07-18 2001-07-18 ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS

Country Status (1)

Country Link
WO (1) WO2003008439A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006099170A2 (en) * 2005-03-10 2006-09-21 Wisconsin Alumni Research Foundation Beta-polypeptides that inhibit cytomegalovirus infection
WO2007025141A3 (en) * 2005-08-26 2007-09-13 Wisconsin Alumni Res Found Poly-beta-peptides from functionalized beta-lactam monomers and antibacterial compositions containing same

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997047593A2 (en) * 1996-06-13 1997-12-18 Novartis Ag Peptides
US5739160A (en) * 1992-05-29 1998-04-14 Bayer Aktiengesellschaft Cyclopentane- and -pentene-β-amino acids
WO2000076974A2 (en) * 1999-06-14 2000-12-21 Wisconsin Alumni Research Foundation Oligomers and polymers of cyclic imino carboxylic acids

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5739160A (en) * 1992-05-29 1998-04-14 Bayer Aktiengesellschaft Cyclopentane- and -pentene-β-amino acids
WO1997047593A2 (en) * 1996-06-13 1997-12-18 Novartis Ag Peptides
WO2000076974A2 (en) * 1999-06-14 2000-12-21 Wisconsin Alumni Research Foundation Oligomers and polymers of cyclic imino carboxylic acids

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
KRICHELDORF H R ET AL: "13C-NMR SEQUENCE ANALYSIS XVIII. TACTICITY OF POLY(D,L-BETA-AMINO ACIDS)", JOURNAL OF MACROMOLECULAR SCIENCE: PART A - CHEMISTRY, MARCEL DEKKER, NEW YORK, NY, US, vol. A14, no. 7, 1980, pages 977 - 990, XP000918572, ISSN: 0022-233X *
MCREYNOLDS K D ET AL: "Examining the secondary structures of unnatural peptides and carbohydrate-based compounds utilizing circular dichroism", TETRAHEDRON: ASYMMETRY, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 11, no. 2, February 2000 (2000-02-01), pages 337 - 362, XP004202389, ISSN: 0957-4166 *
PORTER E ET AL: "Non-haemolytic beta-amino acid oligomers", NATURE, vol. 404, 2000, pages 565, XP002194514 *
ZANUY D ET AL: "On the structure of the phase A of comb-like poly(alpha-alkyl-beta,l-aspartate)s: a molecular modelling study", POLYMER, ELSEVIER SCIENCE PUBLISHERS B.V, GB, vol. 42, no. 1, January 2001 (2001-01-01), pages 281 - 287, XP004208418, ISSN: 0032-3861 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006099170A2 (en) * 2005-03-10 2006-09-21 Wisconsin Alumni Research Foundation Beta-polypeptides that inhibit cytomegalovirus infection
WO2006099170A3 (en) * 2005-03-10 2007-03-29 Wisconsin Alumni Res Found Beta-polypeptides that inhibit cytomegalovirus infection
WO2007025141A3 (en) * 2005-08-26 2007-09-13 Wisconsin Alumni Res Found Poly-beta-peptides from functionalized beta-lactam monomers and antibacterial compositions containing same
US7951912B2 (en) 2005-08-26 2011-05-31 Wisconsin Alumni Research Foundation Poly-β-peptides from functionalized β-lactam monomers and antibacterial compositions containing same
AU2006282942B2 (en) * 2005-08-26 2012-07-26 Wisconsin Alumni Research Foundation Poly-beta-peptides from functionalized beta-lactam monomers and antibacterial compositions containing same
US8519095B2 (en) 2005-08-26 2013-08-27 Wisconsin Alumni Research Foundation Poly-beta-peptides from functionalized beta-lactam monomers and antibacterial compositions containing same
US9120892B2 (en) 2005-08-26 2015-09-01 Wisconsin Alumni Research Foundation Poly-β-peptides from functionalized β-lactam monomers and antibacterial compositions containing same
US9683081B2 (en) 2005-08-26 2017-06-20 Wisconsin Alumni Research Foundation Poly-beta-peptides from functionalized beta-lactam monomers and antibacterial compositions containing same
US10323121B2 (en) 2005-08-26 2019-06-18 Wisconsin Alumni Research Foundation Poly-β-peptides from functionalized β-lactam monomers and antibacterial compositions containing same

Similar Documents

Publication Publication Date Title
US6683154B1 (en) Antimicrobial compositions containing β-amino acid oligomers
US7253146B2 (en) Template-fixed peptidomimetics with antimicrobial activity
EP1979374B1 (en) Template-fixed peptidomimetics
US7582604B2 (en) Template-fixed peptidomimetics with antibacterial activity
US10322998B2 (en) Heterogeneous foldamers containing α, β, and/or γ-amino acids
US20200140489A1 (en) Peptide-Based Quorum Sensing Inhibitors for the Attenuation of Virulence in Staphylococcus Aureus
JPH05500515A (en) Chimeric amino acid analogs
KR20020029908A (en) Mutilin Derivatives and Their Use as Antibacterials
Sarciaux et al. Total synthesis and structure–activity relationships study of odilorhabdins, a new class of peptides showing potent antibacterial activity
EA034926B1 (en) Beta-hairpin peptidomimetics
KR20190093600A (en) Antibacterial peptide
US8772450B2 (en) Beta-amino acids
US6958384B2 (en) Polypeptides containing γ-amino acids
Jeremic et al. Solution‐phase synthesis of Aib‐containing cyclic hexapeptides
WO2003008439A1 (en) ANTIMICROBIAL COMPOSITIONS CONTAINING β-AMINO ACID OLIGOMERS
Guichard β‐Peptides, γ‐Peptides and Isosteric Backbones: New Scaffolds with Controlled Shapes for Mimicking Protein Secondary Structure Elements
KR100298136B1 (en) Synthetic Aglucodalbaheptide Antibiotics
WO1998004584A1 (en) Cyclic decapeptide antibiotics
US6790829B1 (en) Cyclic decapeptide antibiotics
Richaud et al. Structure–Property Relationship Study of N-(Hydroxy) Peptides for the Design of Self-Assembled Parallel β-Sheets
Shyam Cationic amphipathic peptoid oligomers as antimicrobial peptide mimics
D’Amato Chiral Bioactive Cyclopeptoids: Concepts and Purposes
JP3232729B2 (en) New peptides and antibacterial agents
Jia et al. Importance of the structure of vancomycin binding pocket in designing compounds active against vancomycin-resistant enterococci (VRE)
Schmitt c12) United States Patent

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP