structure–activity relationships of ustiloxin analogues

4
Structure–activity relationships of ustiloxin analogues Madeleine M. Joullié a,, Simon Berritt a , Ernest Hamel b a Department of Chemistry, University of Pennsylvania, 231 S. 34th Street, Philadelphia, PA 19104, USA b Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute at Frederick, National Institutes of Health, Frederick, MD 21702, USA article info Article history: Available online 13 January 2011 Dedicated to Professor Harry Wasserman on the occasion of his 90th birthday. Keywords: Ustiloxin Tubulin polymerization Aziridine ring opening Structure–activity relationships abstract Four novel ustiloxin D analogues were synthesized focusing on the size of the macrocyclic core, the ste- reochemistry at the bridgehead ether, and the enantiomer of ustiloxin D. All four were subjected to bio- logical evaluation testing the inhibition of tubulin polymerization. Only 2,2-dimethyl-ustiloxin D retained any activity. Ó 2011 Elsevier Ltd. All rights reserved. 1. Introduction The ustiloxins A–F (Fig. 1) are a class of antimitotic natural products isolated from the water extracts of false smut balls, caused by the parasitic fungus Ustilaginoidea virens. 1 These com- pounds universally inhibit the assembly of the a,b-tubulin dimer into microtubules, thus resulting in mitotic arrest of eukaryotic cells. Many antimitotic agents are known, ranging from the vinca alkaloids and taxoids to chalcones, 2 but the mechanism of action of the naturally occurring peptides that inhibit tubulin polymeriza- tion is still not fully elucidated. 3 Recent syntheses of ustiloxin D 4 and F 5 have been published, and the syntheses of eight analogues focusing on structural varia- 0040-4039/$ - see front matter Ó 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.tetlet.2010.11.165 Corresponding author. Tel.: +1 215 898 3158; fax: +1 215 573 9711. E-mail address: [email protected] (M.M. Joullié). OH O HO HN HN O O N H O R 2 N H O OH R 1 HO 2 C NH 2 OH S O R 2 CH(CH 3 ) 2 R 1 HO 2 C NH 2 OH S O R 1 R 2 CH 3 HO S O R 1 R 2 CH 3 R 2 CH 2 (CH 3 ) 2 R 1 H R 1 H R 2 CH 3 Ustiloxin A Ustiloxin B Ustiloxin C Ustiloxin D Ustiloxin F 6 9 10 IC 50 /μM 1.0 1.8 4.4 2.5 10.3 Figure 1. Ustiloxins A–F with IC 50 values based on inhibition of porcine brain microtubule assembly. (2S)-epi-ustiloxin D 3 OH O HO HN HN O O N H O N H O OH OH O HO HN HN O O N H O N H O OH OH O HO HN HN O O N H O N H O OH OH O HN O NH OH O HO HN O O NH N H O ent-ustiloxin D 1 2,2-dimethyl-ustiloxin D 2 7-N-Gly-ustiloxin D 4 Figure 2. Four ustiloxin D analogues. Tetrahedron Letters 52 (2011) 2136–2139 Contents lists available at ScienceDirect Tetrahedron Letters journal homepage: www.elsevier.com/locate/tetlet

Upload: madeleine-m-joullie

Post on 02-Jul-2016

221 views

Category:

Documents


6 download

TRANSCRIPT

Page 1: Structure–activity relationships of ustiloxin analogues

Tetrahedron Letters 52 (2011) 2136–2139

Contents lists available at ScienceDirect

Tetrahedron Letters

journal homepage: www.elsevier .com/ locate / tet let

Structure–activity relationships of ustiloxin analogues

Madeleine M. Joullié a,⇑, Simon Berritt a, Ernest Hamel b

a Department of Chemistry, University of Pennsylvania, 231 S. 34th Street, Philadelphia, PA 19104, USAb Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute at Frederick,National Institutes of Health, Frederick, MD 21702, USA

a r t i c l e i n f o a b s t r a c t

Article history:Available online 13 January 2011

Dedicated to Professor Harry Wasserman onthe occasion of his 90th birthday.

Keywords:UstiloxinTubulin polymerizationAziridine ring openingStructure–activity relationships

0040-4039/$ - see front matter � 2011 Elsevier Ltd. Adoi:10.1016/j.tetlet.2010.11.165

⇑ Corresponding author. Tel.: +1 215 898 3158; faxE-mail address: [email protected] (M.M. Jou

OH

O

HO

HN

HN

O

O

NH

O

R2

NH

R1

HO2C

NH2 OH

S

O

R2 CR1

HO2C

NH2 OH

S

O

R1 R2 C

HOS

O

R1 R2 C

R2 CR1H

R1H R2 C

Ustiloxin A

Ustiloxin B

Ustiloxin C

Ustiloxin D

Ustiloxin F

6910

Figure 1. Ustiloxins A–F with IC50 values based onmicrotubule assembly.

Four novel ustiloxin D analogues were synthesized focusing on the size of the macrocyclic core, the ste-reochemistry at the bridgehead ether, and the enantiomer of ustiloxin D. All four were subjected to bio-logical evaluation testing the inhibition of tubulin polymerization. Only 2,2-dimethyl-ustiloxin Dretained any activity.

� 2011 Elsevier Ltd. All rights reserved.

1. Introduction

The ustiloxins A–F (Fig. 1) are a class of antimitotic naturalproducts isolated from the water extracts of false smut balls,caused by the parasitic fungus Ustilaginoidea virens.1 These com-

ll rights reserved.

: +1 215 573 9711.llié).

O

OH

H(CH3)2

H3

H3

H2(CH3)2

H3

IC50/µM

1.0

1.8

4.4

2.5

10.3

inhibition of porcine brain

pounds universally inhibit the assembly of the a,b-tubulin dimerinto microtubules, thus resulting in mitotic arrest of eukaryoticcells. Many antimitotic agents are known, ranging from the vincaalkaloids and taxoids to chalcones,2 but the mechanism of actionof the naturally occurring peptides that inhibit tubulin polymeriza-tion is still not fully elucidated.3

Recent syntheses of ustiloxin D4 and F5 have been published,and the syntheses of eight analogues focusing on structural varia-

(2S)-epi-ustiloxin D 3

OH

O

HO

HN

HN

O

O

NH

O

NH

O

OHOH

O

HO

HN

HN

O

O

NH

O

NH

O

OH

OH

O

HO

HN

HN

O

O

NH

O

NH

O

OH OH

O HN

O

NH

OH

O

HO

HN

O

O

NH

NH O

ent-ustiloxin D 1 2,2-dimethyl-ustiloxin D 2

7-N-Gly-ustiloxin D 4

Figure 2. Four ustiloxin D analogues.

Page 2: Structure–activity relationships of ustiloxin analogues

OH

O

HO

HN

HN

OR3R2

O

NH

O

R4

NH O

OH

HO

NH

O

OH

OH

OH

H2N

O

OH

R4

NH

R3R2 O

NH O

OH

5

6

7

Figure 3. Retrosynthetic analysis of the ustiloxins.

M. M. Joullié et al. / Tetrahedron Letters 52 (2011) 2136–2139 2137

tions at R2, stereochemistry at C9 and C10, and methylation of thephenol were also reported by our group.6 To complete the SARanalysis we wished to probe the stereochemistry and the substitu-ents at the tertiary aryl-ether bridge, the size of the macrocycliccore, and the enantiomer of ustiloxin D. The four analogues areshown in Figure 2.

2. Synthesis of ustiloxin D analogues

The convergent synthesis of the ustiloxins developed in our lab-oratory allowed for expeditious synthesis of the four ustiloxin ana-logues. In a retrosynthetic manner, the ustiloxins can be dividedinto three constituent parts: b-hydroxytyrosine 5, aziridine 6,and commercially available amino acid 7 (Fig. 3).

The aziridine 6 and b-hydroxytyrosine moiety 5 can be quicklyaccessed from D-valine,4e and a (S)-salen-aluminium catalyzed al-

NNs

O

NH

O

OBn

HOBocN

O a HO

NHNs

OH b,c HO

NHNs

NH

O

OBn

O

d

BocNO

O

HOBocN

O

HO OH

NHNsNNs

O

NH

O

OBn

e

a b,c

8 9 10

11

12 13

14 15

HOBocN

O

13a

+

Scheme 1. Synthesis of aziridines 11 and 15. Reagents and conditions: (a) HCl, THF,0 �C, 1 h, then NsCl, Na2CO3, THF/H2O, rt, 16 h, 58% for 9, 90% for 14; (b) TEMPO,NaClO2, NaClO, Na2HPO4, MeCN, 40 �C, 18 h; (c) H-GlyOBn�HCl, EDCI, HOBt,NaHCO3, DMF, 12 h, rt, 95% for 10, 90% for alkynyl precursor; (d) DIAD, PPh3,CH2Cl2, 18 h, rt, 77% for 11, 68% for 15; (e) ethynylMgBr, THF, rt, 80% ds = 11:1(13a:13).

TBSONCbz

O

OPMB

OBnOH

NNs

R2

R1 O

NH O

OB

11 R1= R2= Me15 R1= Ethyne; R2= M

+

16

Scheme 2. Ring opening of aziridines 11 and 15. Reage

dol7 reaction, respectively. This convergent route allows for rapidderivation of any of the three components.

To access 2,2-dimethyl ustiloxin D, oxazolidine 8 was utilized toform the requisite dimethylaziridine 11 (Scheme 1).

Thus, the 2,2-dimethyl aziridine was synthesized by treatmentof the tertiary alcohol 8 with aqueous HCl and subsequentortho-nitrobenzenesulfonamide protection of the amine affordeddiol 9. TEMPO oxidation, followed by EDCI-mediated coupling ofglycine benzyl ester gave the precursor to aziridine 10, and a Mits-unobu reaction afforded the dimethyl aziridine 11 in good yield.The (2S,3R)-ethynyl aziridine 15 was synthesized by Grignardaddition to ketone 12 which resulted in a 11:1 diastereomeric ra-tio; the minor diastereomer (13) being utilized within this route.The synthesis of 15 was continued supra vida to afford the secondaziridine.

The requisite b-hydroxytyrosine moiety (16) was synthesized aspreviously reported6 and coupled to aziridines 11 and 15 via a reg-ioselective 1,5,7-triazabicyclo[4.4.0]dec-5-ene (TBD) mediated ringopening reaction developed in our laboratory,8 to afford the nosylprotected amines 17 and 18 (Scheme 2).

The synthesis of ent-ustiloxin D followed the previous6 synthe-sis of ustiloxin D and will not be reported in detail here. An (R)-salen-aluminium catalyzed aldol reaction afforded the b-hydrox-ytyrosine, L-serine provided the aziridine and D-valine was usedto complete the protected linear precursor. The synthesis of 7-N-Gly-ustiloxin D (4) also followed the previous synthesis to givethe nosyl protected amine 21 (Scheme 3).

Removal of the nosyl protecting group on each of the three ana-logues with benzene thiol proceeded without incident to providethe free amine. EDCI-mediated coupling of tripeptides 17 and 18with N-Boc-D-valine gave compounds 19 and 20, respectively. Cou-pling of amine 21 with N-Boc-glycine-valine afforded 22 (Scheme3).

With all three analogues and the ent-ustiloxin D protectedlinear precursor in hand (not shown), the synthesis was concludedwith a deprotection/macrocyclization/deprotection sequence(Scheme 4).

Treatment of the linear precursors with TFA/Et3SiH afforded theTFA salts, which were subsequently converted into the hydrochlo-ride salts. Subjection of each amino acid salt to EDCI-mediatedmacrocyclization and concomitant Pd/C catalyzed hydrogenolysisgave ent-ustiloxin D (1), 2,2-dimethyl ustiloxin D (2), (2S)-epi-usti-loxin D (3), and 7-N-Gly-ustiloxin D (4).

OBnO

TBSOCbzN

NHNs

OR2R1

O

OPMB

NH O

OBnn

e

a

17 R1= R2= Me (72%)18 R1= Ethyne; R2= Me (78%)

nts and conditions: (a) TBD, toluene, 0 �C to rt, 3 h.

Page 3: Structure–activity relationships of ustiloxin analogues

OBnO

TBSOCbzN

NHNs

OR2R1

O

OPMB

NH O

OBn

17 R1= R2= Me18 R1= Ethyne; R2= Me

OBnO

TBSOCbzN

HN

OR2R1

O

OPMB

O

NH O

OBn

BocHN

a,b

19 R1= R2= Me (96%)20 R1= Ethyne; R2= Me (76%)

OBnO

TBSOCbzN

NHNs

O

O

OPMB

NH

O

OBn OBnO

TBSOCbzN

HN

O

O

OPMB

O

NH O

OBn

HN

NHBocO

a,c

21 22

Scheme 3. Deprotection and coupling of amino acid residues. Reagents andconditions: (a) PhSH, Cs2CO3, DMF, 0 �C, 4 h; (b) N-Boc-Gly, EDCI, HOBt, NaHCO3,DMF, 12 h, rt; (c) N-Boc-Val-Gly, EDCI, HOBt, NaHCO3, DMF, 12 h, 0 �C to rt, 79%.

OBnO

TBSOCbzN

HN

OR2

R1

O

OPMB

O

NH O

OBn

BocHN

19 R1= R2= Me20 R1= Ethyne; R2= Me

OBnO

TBSOCbzN

HN

O

O

OPMB

O

NH O

OBn

HN

NHBocO

OHO

HOHN

HN

OR2R1

O

NH

O

NH O

OH

2 R1= R2= Me3 R1= Ethyne; R2= Me

OHO HN

ONH

OH

O

HOHN

O

O

NHNH O

a,b,c

a,b,c

22 4

Scheme 4. Reagents and conditions: (a) TFA, Et3SiH, CH2Cl2, 0 �C, 4 h; (b) EDCI,HOBt, NaHCO3, DMF, 0 �C to rt, 24 h, 46% for R1 = R2 = Me, 30% for R1 = MeR2 = ethyne, 34% for N-Gly macrocycle; (c) Pd/C, H2, THF/H2O, 24 h, 80% for 2, 60%for 3, and 55% for 4.

Figure 4. Spartan�’03 energy minimized molecular models at HF/AM1 level oftheory. Ustiloxin D (A), 2,2-dimethyl-ustiloxin D (B), and (2S)-epi-ustiloxin D (C).

2138 M. M. Joullié et al. / Tetrahedron Letters 52 (2011) 2136–2139

3. Bioactivity

The effect of the changes present in the four analogues on thepolymerization of purified tubulin was investigated using theIC50 value of ustiloxin D (2.5 lM) as the benchmark. Biologicalevaluation of ent-ustiloxin D (1) showed no inhibition with anIC50 value >40 lM. Similarly, 7-N-Gly-ustiloxin D (4) showed noinhibition (IC50 >40 lM), suggesting that the naturally occurringustiloxin D macrocycle size is optimum. (2S)-epi-Ustiloxin D (3)exhibited a very slight inhibitory effect, but overall had an IC50

>40 lM. Interestingly, removal of the stereogenic carbon at theether bridge afforded 2,2-dimethyl ustiloxin D (2) with anIC50 = 9.2 ± 2 lM.

The difference in inhibitory activity of ustiloxin D, (2S)-epi, andthe dimethyl species was investigated using molecular modeling(Fig. 4). Comparison of ustiloxin D (A) to 2,2-dimethyl ustiloxin(B) and (2S)-epi-ustiloxin (C) quite clearly shows distortion of theglycine sidechain, progressively pushing it toward the valine resi-due. The out-of-plane glycine sidechain may lessen the ability ofthe molecule to bind at this position, but it may also block thebinding ability of the valine residue, which was shown to play animportant role in the ustiloxins inhibitory effect.6,9

4. Conclusions

Novel ustiloxin analogues were synthesized to investigate theeffect of changes at the C2 center, the macrocycle size, and theenantiomeric series on tubulin inhibition. The structural modifica-tions produced significant changes in the biological activity sug-gesting the importance of the valine residue in the binding process.

Acknowledgments

We thank NIH (CA-40081) and NSF (CHE-0951394) for supportof this work. Financial support for the departmental instrumenta-tion was provided by NIH (1S10RR23444-1). We also thank Dr.George Furst and Dr. Rakesh Kohli for NMR and MS assistance,respectively.

References and notes

1. (a) Koiso, Y.; Li, Y.; Kobayashi, H.; Natori, M.; Hashimoto, Y.; Iwasaki, S.; Fujita,Y.; Sonoda, R.; Yaegashi, H., et al Tennen Yuki Kagobutsu Toronkai Koen Yoshishu1992, 34th, 566; (b) Koiso, Y.; Natori, M.; Iwasaki, S.; Sato, S.; Sonoda, R.; Fujita,Y.; Yaegashi, H.; Sato, Z. Tetrahedron Lett. 1992, 33, 4157; (c) Koiso, Y.; Li, Y.;Iwasaki, S.; Hanaoka, K.; Kobayashi, T.; Sonoda, R.; Fujita, Y.; Yaegashi, H.; Sato,Z. J. Antibiot. 1994, 47, 765.

2. Hamel, E. Med. Res. Rev. 1996, 16, 207.3. Hamel, E.; Covell, D. G. Curr. Med. Chem.: Anti-Cancer Agents 2002, 2, 19.4. (a) Cao, B. University of Pennsylvania, 2002; (b) Cao, B.; Park, H.; Joullié, M. M. J.

Am. Chem. Soc. 2002, 124, 520; (c) Tanaka, H.; Sawayama, A. M.; Wandless, T. J. J.Am. Chem. Soc. 2003, 125, 6864; (d) Sawayama, A. M.; Tanaka, H.; Wandless, T. J.J. Org. Chem. 2004, 69, 8810; (e) Li, P.; Evans, C. D.; Joullié, M. M. Org. Lett. 2005, 7,5325.

5. Li, P.; Evans, C. D.; Forbeck, E. M.; Park, H.; Bai, R.; Hamel, E.; Joullié, M. M. Bioorg.Med. Chem. Lett. 2006, 16, 4804.

Page 4: Structure–activity relationships of ustiloxin analogues

M. M. Joullié et al. / Tetrahedron Letters 52 (2011) 2136–2139 2139

6. Li, P.; Evans, C. D.; Wu, Y.; Cao, C.; Hamel, E.; Joullié, M. M. J. Am. Chem. Soc. 2008,130, 2351.

7. Evans, D. A.; Janey, J. M.; Magomedov, N.; Tedrow, J. S. Angew. Chem., Int. Ed.2001, 40, 1884.

8. Forbeck, E. M.; Evans, C. D.; Gilleran, J. A.; Li, P.; Joullié, M. M. J. Am. Chem. Soc.2007, 129, 14463.

9. Cormier, A.; Marchand, M.; Ravelli, R. B. G.; Knossow, M.; Gigant, B. EMBO Rep.2008, 9, 1101.