Access to Enantiopure α-Hydrazino Acids for N-Amino Peptide

Department of Chemistry, University of South Florida, Tampa, Florida 33620, United States. J. Org. Chem. , 2017, 82 (3), pp 1833–1841. DOI: 10.1021/...
0 downloads 0 Views 513KB Size
Subscriber access provided by Fudan University

Note

Access to Enantiopure #-Hydrazino Acids for N-Amino Peptide Synthesis Chang Won Kang, Matthew P Sarnowski, Yassin M Elbatrawi, and Juan R Del Valle J. Org. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.joc.6b02718 • Publication Date (Web): 11 Jan 2017 Downloaded from http://pubs.acs.org on January 13, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

The Journal of Organic Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 27

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The Journal of Organic Chemistry

Access to Enantiopure α-Hydrazino Acids for N-Amino Peptide Synthesis Chang Won Kang, Matthew P. Sarnowski, Yassin M. Elbatrawi, and Juan R. Del Valle* Department of Chemistry, University of South Florida, Tampa, FL 33620 [email protected]

TOC Graphic

1) EtO 2C EtO 2C

R H 2N

O N

Boc

sat. aq. NaHCO 3, THF

CO 2Me

R BocHN

2) aq. NaOH, THF

N H

CO 2H

chiral α-hydrazino acids 19 examples

O H 2N

N NH 2 O

H N

O N NH 2 O

H N

O N NH 2 O

NH 2

SPPS

N-amino peptide (NAP)

ABSTRACT Backbone N-Methylation of α-peptides has been widely employed to enhance the bioavailability and bioactivity of parent sequences. Heteroatomic peptide amide substituents have received less attention due, in part, to the lack of practical synthetic strategies. Here, we report the synthesis of α-hydrazino acids derived from 19 out of the 20 canonical proteinogenic amino acids and demonstrate their use in the solid-phase synthesis of N-amino peptide (NAP) derivatives.

KEYWORDS Peptides, Peptidomimetics, Electrophilic amination, Amino acids, Hydrazines ACS Paragon Plus Environment

1

The Journal of Organic Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 27

The introduction of backbone amide substituents has a profound impact on the conformation and proteolytic stability of peptides. Backbone N-methylation, for example, has been utilized extensively in peptide structure-activity-relationship (SAR) campaigns to probe for bioactive conformations and to improve the bioavailability of lead compounds.1 From a synthetic standpoint, substitution of a single amide within a sequence represents a subtle modification. However, the resulting increase in cis amide rotamer population and removal of a main chain hydrogen-bond donor can give rise to significant conformational heterogeneity. This is particularly evident in peptoids owing to the presence of multiple tertiary amides as well as to the absence of Cα substituents that would otherwise restrict backbone dihedral angles.2 The development of amide substitution strategies that maintain side-chain functionality while constraining main-chain torsions would thus be of considerable utility in peptidomimetic drug design.

R1

R1

O N peptoid

R1 NH N

O

N-azapeptoid

O

R1

N

NH N

O

R2

R2

peptide tertiary amide

N-amino peptide (NAP)

Figure 1. Representative backbone amide modified peptidomimetics

In an effort toward conformationally defined and proteolytically stable peptidomimetics, our group recently developed a class of N-amino peptide (NAP) derivatives that are constrained by both covalent and non-covalent interactions.3 As shown in Figure 1, backbone aminated peptides retain native Cα substituents, which reduces accessible φ and ψ torsions.4 This feature sets NAPs apart from the related but inherently more flexible glycine-derived N-azapeptoids.5 In contrast to oligomeric N-alkylated peptides,6 the N-amino substituent in NAPs also offers a handle for hydrogen bonding or subsequent chemical diversification. Given that ease-of-synthesis is a major consideration for peptidomimetic SAR studies, we sought a convenient route to diversely substituted α-hydrazino acid building blocks7 for the ACS Paragon Plus Environment

2

Page 3 of 27

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The Journal of Organic Chemistry

assembly of NAPs on solid support. Here, we describe a practical electrophilic amination approach to enantiopure α-hydrazino acids corresponding to 19 canonical proteinogenic amino acids, as well as an optimized protocol for their introduction into host peptides.

Peptoids and N-azapeptoids are typically prepared via a submonomer approach that relies on displacement of α-halo amides with amine or hydrazine nucleophiles.5,8 Unfortunately, chiral secondary α-bromo acids and amides are not readily available and may epimerize or undergo elimination during reaction with with basic nucleophiles.3b,9 As an alterantive to on-resin submonomer synthesis, we explored a conventional building block strategy toward NAPs that relied on SN2 displacement of chiral triflates with t-butylcarbazate (Scheme 1).3,10 Unlike reactions with α-bromoamides or esters, these reactions proceed rapidly at low temperature with little to no racemization. The triflate intermediates are, in turn, derived from D-amino acids via a diazotization reaction that proceeds with overall retention of stereochemistry.11 Although convenient for the synthesis of aliphatic α-hydrazino acids, a number of side chains bearing nucleophilic heteroatoms are incompatible with the triflation step. Some acid-labile protecting groups are also unstable under acidic diazotization conditions. Notably, we found that one-pot triflation and substitution reaction of tyrosine derivative 1 afforded Wagner-Meerwein rearrangement product 2, whose structure was confirmed by X-ray diffraction (Scheme 1).12 Although this reaction pathway was unique to 1, the Cα-N bond formation approach toward α-hydrazino acids appeared to impose inherent limitations on substrate scope.

ACS Paragon Plus Environment

3

The Journal of Organic Chemistry

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

R H 2N

CO 2H

1) NaNO 2, AcOH/H2O 2) MeI, K 2CO3, DMF

R HO

CO 2Me

Page 4 of 27

Tf2O, lutidine, DCM, -78oC, then BocNHNH 2, rt

aliphatic D-amino acids

R BocHN

N H

CO 2Me

Ot-Bu

aq. LIOH, THF

R BocHN

N H

Ot-Bu

Ot-Bu

Tf2O, lutidine, DCM

BocNHNH 2

H CO 2Me HO

O MeO

1

CO 2H

CO 2Me HN

NHBoc

X-ray

2

Scheme 1. Cα-Nα bond formation approach to α-hydrazino acids.

Electrophilic amination represents an attractive method to prepare substituted hydrazines starting from primary and secondary amines. A number of oxaziridine derivatives can rapidly transfer amine or carbamate groups onto nucleophilic heteroatoms under mild conditions, thus allowing for the use of Lamino acids as readily available substrates.13 Seminal contributions from the labortaories of Collet and Vidal have described N-acyloxy oxazirdines effective in aminating zwitterionic amino acid residues.13c,14 However, the poor solubility of these substrates in organic solvents neccesitates the use of phase transfer reagents to promote reaction, and often affords products in moderate overall yields. The reaction of primary amines with N-acyl oxaziridines is further complicated by competing diamination as well as the formation of undesired imine adducts. As a result, a protocol involving temporary N-benzyl protection of α-amino acids has also been reported.15 Given the existing methods, we sought to develop a direct route toward α-hydrazino acid building blocks based on each of the canonical proteinogenic amino acids in a form suitable for SPPS.

We first examined the electrophilic amination of H-Tyr(OtBu)-OMe using an N-Boc oxaziridine derivative (3, Table 1) recently reported by Armstrong and co-workers.16 Reagent 3 was previously shown to minimize the formation of imine adducts resulting from reaction of amine substrates with the diethylketomalonate byproduct. We employed a streamlined synthesis of 3 that requires only one ACS Paragon Plus Environment

4

Page 5 of 27

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The Journal of Organic Chemistry

chromatographic purification, yielding multi-gram quantities of the shelf-stable oxaziridine in under 48 hours starting from t-butyl carbazate.16-17 The reaction of 3 with the free base of H-Tyr(OtBu)-OMe was equally tolerant of different aprotic solvents as long as excess α-amino ester was employed (entries 1-3). Yield of the desired product decreased when the equivalency of H-Tyr(OtBu)-OMe was reduced (entries 4-6). In these cases, significant formation of 5 was observed, along with the undesired imine adduct. Low yields of 4a were obtained when H-Tyr(OtBu)-OMe.HCl was used as the substrate, even in the presence of auxiliary bases (entries 7-10). We were pleased to find that a moderate yield of 4a was acheived using 1:1 THF:H2O as solvent, indicating the suitability of an aqueous reaction medium. By adding excess NaHCO3 to the solvent mixture, the yield of 4a increased dramatically and diamination was suppressed, even when H-Tyr(OtBu)-OMe.HCl was the limiting reagent (entries 12-16). These aqueous conditions thus obviate the need for excess amino ester or prior neutralization of the substrate hydrochloride salts.

Table 1. Screening of α-amino ester amination conditions. t-BuO

t-BuO CO 2Et EtO 2C O N Boc

AA

see table BocHN

3

N H 4a

BocHN N CO 2Me CO 2Me NHBoc 5

entry 1

AA H-Tyr(OtBu)-OMe

equiv. AA 2.0

conditions DCM

yield 4aa 98

2 3

H-Tyr(OtBu)-OMe H-Tyr(OtBu)-OMe

2.0 2.0

THF PhMe

96 99

4 5

H-Tyr(OtBu)-OMe H-Tyr(OtBu)-OMe

1.2 1.0

PhMe PhMe

79 57

6

H-Tyr(OtBu)-OMe

0.5

PhMe

34

7 8

H-Tyr(OtBu)-OMe . HCl H-Tyr(OtBu)-OMe . HCl

2.0 2.0

PhMe THF

14 2

9 10

H-Tyr(OtBu)-OMe . HCl H-Tyr(OtBu)-OMe . HCl

2.0 2.0

PhMe, NEt3 PhMe, pyridine