Abstract A1: The physiological form of MetAP2 can be inhibited through binding to either of the two active-site metals

Abstract Methionine aminopeptidases (MetAP) are metalloenzymes that remove the N-terminal initiator methionine from newly synthesized polypeptides allowing essential post-translational modifications such as acetylation and myristoylation to take place. MetAP2, one of the two eukaryotic forms of the...

Full description

Saved in:
Bibliographic Details
Published inMolecular cancer therapeutics Vol. 8; no. 12_Supplement; p. A1
Main Authors Wallis, Nicola G., Berdini, Valerio, Besong, Gilbert, Chessari, Gianni, Coyle, Joe, Graham, Brent, Madin, Andrew, O'Brien, Alistair, Richardson, Caroline J., Smith, Kirsten, Thompson, Neil T., Vinkovic, Mladen, Williams, Pamela A.
Format Journal Article
LanguageEnglish
Published 10.12.2009
Online AccessGet full text

Cover

Loading…
Abstract Abstract Methionine aminopeptidases (MetAP) are metalloenzymes that remove the N-terminal initiator methionine from newly synthesized polypeptides allowing essential post-translational modifications such as acetylation and myristoylation to take place. MetAP2, one of the two eukaryotic forms of the enzyme, was identified as the target of fumagillin, a natural product with anti-angiogenic properties that inhibits the proliferation of endothelial cells. Clinical activity has been seen for a semi-synthetic analogue of fumagillin, TNP470, suggesting MetAP2 is a good target for inhibiting angiogenesis. In vitro, MetAP2 appears to have sites for two divalent metal ions within its active site but there has been much discussion around the identity and number of metal ions actually present in the physiological states of the various MetAPs. An understanding of the physiologically relevant metalloform of the enzyme is essential for designing inhibitors that are active in cells. We have used tool compounds that bind the active site metals in diverse ways to investigate the relevance of the two potential metal binding sites in MetAP2. Using our fragment-based screening approach, Pyramid™, we screened the manganese-form of the MetAP2 enzyme. We identified multiple low-molecular weight fragment hits and confirmed their modes of binding to the two metals in the active site of MetAP2 by X-ray crystallography. Three hit series, which bound metal 1 only, metal 2 only or both metals 1 and 2 were chosen for further optimization using structure-based drug design. Optimized lead compounds had potent inhibitory activity against the in vitro MetAP2 enzyme (∼100 nM) and in HUVEC proliferation assays, whilst also showing greater than 1000-fold selectivity for MetAP2 over MetAP1. Examples from each series, representing different active site metal binding modes, were used as tool compounds to investigate the mechanism of action in cells. The levels of the MetAP2 substrate, 14-3-3, were monitored by western blot in HUVECs treated with these compounds. Levels of methionylated 14-3-3 increased upon treatment with compounds from each of our series indicating the substrate was not being processed and that in each case the compound was inhibiting MetAP2 in these cells. These data indicate that the physiological form of MetAP2 can be inhibited by compounds which bind solely to either of the two active-site metals, suggesting that both metals must be present in the intra-cellular form of MetAP2 and allowing multiple approaches to inhibiting this key angiogenic target. The lead series identified here provide chemically diverse scaffolds for further optimization of drug like properties. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A1.
AbstractList Abstract Methionine aminopeptidases (MetAP) are metalloenzymes that remove the N-terminal initiator methionine from newly synthesized polypeptides allowing essential post-translational modifications such as acetylation and myristoylation to take place. MetAP2, one of the two eukaryotic forms of the enzyme, was identified as the target of fumagillin, a natural product with anti-angiogenic properties that inhibits the proliferation of endothelial cells. Clinical activity has been seen for a semi-synthetic analogue of fumagillin, TNP470, suggesting MetAP2 is a good target for inhibiting angiogenesis. In vitro, MetAP2 appears to have sites for two divalent metal ions within its active site but there has been much discussion around the identity and number of metal ions actually present in the physiological states of the various MetAPs. An understanding of the physiologically relevant metalloform of the enzyme is essential for designing inhibitors that are active in cells. We have used tool compounds that bind the active site metals in diverse ways to investigate the relevance of the two potential metal binding sites in MetAP2. Using our fragment-based screening approach, Pyramid™, we screened the manganese-form of the MetAP2 enzyme. We identified multiple low-molecular weight fragment hits and confirmed their modes of binding to the two metals in the active site of MetAP2 by X-ray crystallography. Three hit series, which bound metal 1 only, metal 2 only or both metals 1 and 2 were chosen for further optimization using structure-based drug design. Optimized lead compounds had potent inhibitory activity against the in vitro MetAP2 enzyme (∼100 nM) and in HUVEC proliferation assays, whilst also showing greater than 1000-fold selectivity for MetAP2 over MetAP1. Examples from each series, representing different active site metal binding modes, were used as tool compounds to investigate the mechanism of action in cells. The levels of the MetAP2 substrate, 14-3-3, were monitored by western blot in HUVECs treated with these compounds. Levels of methionylated 14-3-3 increased upon treatment with compounds from each of our series indicating the substrate was not being processed and that in each case the compound was inhibiting MetAP2 in these cells. These data indicate that the physiological form of MetAP2 can be inhibited by compounds which bind solely to either of the two active-site metals, suggesting that both metals must be present in the intra-cellular form of MetAP2 and allowing multiple approaches to inhibiting this key angiogenic target. The lead series identified here provide chemically diverse scaffolds for further optimization of drug like properties. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A1.
Author Vinkovic, Mladen
Graham, Brent
Williams, Pamela A.
Wallis, Nicola G.
Smith, Kirsten
O'Brien, Alistair
Madin, Andrew
Richardson, Caroline J.
Coyle, Joe
Thompson, Neil T.
Berdini, Valerio
Besong, Gilbert
Chessari, Gianni
Author_xml – sequence: 1
  givenname: Nicola G.
  surname: Wallis
  fullname: Wallis, Nicola G.
– sequence: 2
  givenname: Valerio
  surname: Berdini
  fullname: Berdini, Valerio
– sequence: 3
  givenname: Gilbert
  surname: Besong
  fullname: Besong, Gilbert
– sequence: 4
  givenname: Gianni
  surname: Chessari
  fullname: Chessari, Gianni
– sequence: 5
  givenname: Joe
  surname: Coyle
  fullname: Coyle, Joe
– sequence: 6
  givenname: Brent
  surname: Graham
  fullname: Graham, Brent
– sequence: 7
  givenname: Andrew
  surname: Madin
  fullname: Madin, Andrew
– sequence: 8
  givenname: Alistair
  surname: O'Brien
  fullname: O'Brien, Alistair
– sequence: 9
  givenname: Caroline J.
  surname: Richardson
  fullname: Richardson, Caroline J.
– sequence: 10
  givenname: Kirsten
  surname: Smith
  fullname: Smith, Kirsten
– sequence: 11
  givenname: Neil T.
  surname: Thompson
  fullname: Thompson, Neil T.
– sequence: 12
  givenname: Mladen
  surname: Vinkovic
  fullname: Vinkovic, Mladen
– sequence: 13
  givenname: Pamela A.
  surname: Williams
  fullname: Williams, Pamela A.
BookMark eNqdj0FOwzAQAC1UJFrgBxz2Ay7epoGEm4WAXpAQyt1y0k1slNiVbYr6exzgBZxmtdpZaVZs4bwjxm5QrBHL6hbLouT3eFesG_n-wkXNJZ6xZV5XvCpxu_iZf08u2CrGDyGwqje4ZEfZxhR0l0DiAzSG4GBO0frRD7bTI_Q-TOB7eKUk3zbQaQctgXXGtjbRHpIJ_nMw0Fq3t26A5IFsMhRmKRPSl4f83h6Jx2zAREmP8Yqd9xl0_cdLtn1-ah53vAs-xkC9OgQ76XBSKNTcqOYANQeouVGJWkks_ql9AwE_Xec
ContentType Journal Article
DBID AAYXX
CITATION
DOI 10.1158/1535-7163.TARG-09-A1
DatabaseName CrossRef
DatabaseTitle CrossRef
DatabaseTitleList CrossRef
DeliveryMethod fulltext_linktorsrc
Discipline Medicine
EISSN 1538-8514
EndPage A1
ExternalDocumentID 10_1158_1535_7163_TARG_09_A1
GroupedDBID ---
.55
123
18M
2FS
2WC
34G
39C
53G
5RE
5VS
AAYXX
ABOCM
ACGFO
ACIWK
ACPRK
ADBBV
ADCOW
AENEX
AFHIN
AFRAH
ALMA_UNASSIGNED_HOLDINGS
BAWUL
BR6
BTFSW
CITATION
CS3
DIK
DU5
E3Z
EBS
EJD
F5P
GX1
H13
H~9
IH2
KQ8
L7B
MVM
OK1
P2P
QTD
RCR
RHF
RHI
TR2
W8F
WOQ
X7M
YBU
ID FETCH-crossref_primary_10_1158_1535_7163_TARG_09_A13
ISSN 1535-7163
IngestDate Fri Aug 23 03:44:36 EDT 2024
IsPeerReviewed true
IsScholarly true
Issue 12_Supplement
Language English
LinkModel OpenURL
MergedId FETCHMERGED-crossref_primary_10_1158_1535_7163_TARG_09_A13
ParticipantIDs crossref_primary_10_1158_1535_7163_TARG_09_A1
PublicationCentury 2000
PublicationDate 2009-12-10
PublicationDateYYYYMMDD 2009-12-10
PublicationDate_xml – month: 12
  year: 2009
  text: 2009-12-10
  day: 10
PublicationDecade 2000
PublicationTitle Molecular cancer therapeutics
PublicationYear 2009
SSID ssj0018921
Score 3.943919
Snippet Abstract Methionine aminopeptidases (MetAP) are metalloenzymes that remove the N-terminal initiator methionine from newly synthesized polypeptides allowing...
SourceID crossref
SourceType Aggregation Database
StartPage A1
Title Abstract A1: The physiological form of MetAP2 can be inhibited through binding to either of the two active-site metals
Volume 8
hasFullText 1
inHoldings 1
isFullTextHit
isPrint
link http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwnV1LS8NAEF6KgngRn_hmDt5KapImaeItiFaUikoVb6Fbp7QgrbSpgr_emd1suj4Q6yVtk2YSMh_z2HwzI8SR5yLKTiKdIO65TuA9RU7iu-iEGMtQxuTxlKZb19HFfXD5GD5WKnaH4Gkua933H-tK_qNV2kd65SrZOTRbCqUd9J30S1vSMG3_pONU8kJFN6-mnmFPqJWK0qBxRKpILpinNz4zvKqSu4T0B1KFmmZKjxzo4hYKRJGLNMaGOpC_jVS7jVd0-DUzD5zuPE_siLZl5uuy9C4qzqKp6CrjdV6u180MFPI61WatXAdAgqiaK1V9IGc11rwwfWRS8IWbA-7EVRJ0Tvs8t0WXyDcHPHTp09qFGrxQsFiNuQ0dyti0icOZCaY4MLBtdGxD0c_UxNMZNUjb3tSzvLj-8d0_hFzzUF621k7vmsz-MOfa7bi_uMmSvKjSpjDOWErGUjKWkrlJllIavug3kpC5pVe3s9dZceIXjXv1dYsaTpJy_NO9WDGSFey0V8VKkaVAqiG3Jio4XBdLrYKHsSFeDfIg9U6AcAefcAeMOxj1QOMOCBkgEUrcQYE7KHAH-Qg07vgk-gTCHVi4A427TRGcn7VPLxxz39mLbpaS_fa06ltiYTga4rYAGUdIqXMkExmQW4llx_Uxqvs9F7sNyuJ3xHyid-f8_55YngF0Xyzk4ykeULiZy0Olyg-InYGf
link.rule.ids 315,786,790,27955,27956
linkProvider Colorado Alliance of Research Libraries
openUrl ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Abstract+A1%3A+The+physiological+form+of+MetAP2+can+be+inhibited+through+binding+to+either+of+the+two+active-site+metals&rft.jtitle=Molecular+cancer+therapeutics&rft.au=Wallis%2C+Nicola+G.&rft.au=Berdini%2C+Valerio&rft.au=Besong%2C+Gilbert&rft.au=Chessari%2C+Gianni&rft.date=2009-12-10&rft.issn=1535-7163&rft.eissn=1538-8514&rft.volume=8&rft.issue=12_Supplement&rft.spage=A1&rft.epage=A1&rft_id=info:doi/10.1158%2F1535-7163.TARG-09-A1&rft.externalDBID=n%2Fa&rft.externalDocID=10_1158_1535_7163_TARG_09_A1
thumbnail_l http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=1535-7163&client=summon
thumbnail_m http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=1535-7163&client=summon
thumbnail_s http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=1535-7163&client=summon