WO2000015797A2 - Compositions and methods for the treatment of immune related diseases - Google Patents

Compositions and methods for the treatment of immune related diseases Download PDF

Info

Publication number
WO2000015797A2
WO2000015797A2 PCT/US1999/021547 US9921547W WO0015797A2 WO 2000015797 A2 WO2000015797 A2 WO 2000015797A2 US 9921547 W US9921547 W US 9921547W WO 0015797 A2 WO0015797 A2 WO 0015797A2
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
antibody
cells
pro301
mammal
Prior art date
Application number
PCT/US1999/021547
Other languages
French (fr)
Other versions
WO2000015797A3 (en
Inventor
Sherman Fong
Audrey Goddard
Austin L. Gurney
Daniel Tumas
William I. Wood
Original Assignee
Genentech, Inc.
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
Priority claimed from PCT/US1998/019437 external-priority patent/WO1999014241A2/en
Application filed by Genentech, Inc. filed Critical Genentech, Inc.
Priority to CA002343006A priority Critical patent/CA2343006A1/en
Priority to AU64984/99A priority patent/AU6498499A/en
Priority to KR1020017003430A priority patent/KR20010085816A/en
Priority to IL14155199A priority patent/IL141551A0/en
Priority to EP99952930A priority patent/EP1114154A2/en
Priority to JP2000570324A priority patent/JP2003524600A/en
Priority to CA002503475A priority patent/CA2503475A1/en
Priority to PCT/US1999/028313 priority patent/WO2000032221A2/en
Priority to EP06014175A priority patent/EP1734051A3/en
Priority to AU17482/00A priority patent/AU771751C/en
Priority to CA002347835A priority patent/CA2347835A1/en
Priority to NZ535590A priority patent/NZ535590A/en
Priority to AT99960624T priority patent/ATE458050T1/en
Priority to JP2000584912A priority patent/JP3695642B2/en
Priority to MXPA01005169A priority patent/MXPA01005169A/en
Priority to KR10-2001-7006818A priority patent/KR100529270B1/en
Priority to IL14274299A priority patent/IL142742A0/en
Priority to EP99960624A priority patent/EP1135485B1/en
Priority to AT07001711T priority patent/ATE432987T1/en
Priority to IL14303199A priority patent/IL143031A0/en
Priority to EP99960644A priority patent/EP1141284A2/en
Priority to ES07001711T priority patent/ES2327785T3/en
Priority to EP07001711A priority patent/EP1820859B9/en
Priority to DE69935085T priority patent/DE69935085T2/en
Priority to EP04007617A priority patent/EP1484338B1/en
Priority to DK04007617T priority patent/DK1484338T3/en
Priority to MXPA01006330A priority patent/MXPA01006330A/en
Priority to DE69940964T priority patent/DE69940964D1/en
Priority to PT04007617T priority patent/PT1484338E/en
Priority to KR10-2001-7007850A priority patent/KR100499600B1/en
Priority to JP2000589692A priority patent/JP3993746B2/en
Priority to CA002353799A priority patent/CA2353799A1/en
Priority to ES04007617T priority patent/ES2281704T3/en
Priority to PCT/US1999/028565 priority patent/WO2000037638A2/en
Priority to EP09004092A priority patent/EP2075335A3/en
Priority to CA002450402A priority patent/CA2450402A1/en
Priority to AU17499/00A priority patent/AU768230B2/en
Priority to AT04007617T priority patent/ATE353339T1/en
Priority to PCT/US1999/030911 priority patent/WO2000075316A1/en
Priority to AU22153/00A priority patent/AU2215300A/en
Priority to AU26008/00A priority patent/AU2600800A/en
Priority to PCT/US2000/000219 priority patent/WO2000053753A2/en
Priority to PCT/US2000/004414 priority patent/WO2001004311A1/en
Priority to AU28839/00A priority patent/AU2883900A/en
Priority to PCT/US2000/005841 priority patent/WO2000053758A2/en
Priority to CA002362427A priority patent/CA2362427A1/en
Priority to KR1020017011406A priority patent/KR20010103046A/en
Priority to AU35144/00A priority patent/AU3514400A/en
Priority to EP00913764A priority patent/EP1220905A2/en
Priority to JP2000603379A priority patent/JP2004516227A/en
Priority to PCT/US2000/007377 priority patent/WO2001019991A1/en
Priority to AU39026/00A priority patent/AU3902600A/en
Publication of WO2000015797A2 publication Critical patent/WO2000015797A2/en
Priority to AT00916675T priority patent/ATE478145T1/en
Priority to CA002383254A priority patent/CA2383254A1/en
Priority to AU37743/00A priority patent/AU3774300A/en
Priority to EP00916675A priority patent/EP1210418B1/en
Priority to DE60044849T priority patent/DE60044849D1/en
Priority to JP2001500766A priority patent/JP2004522402A/en
Priority to PCT/US2000/008439 priority patent/WO2000073454A1/en
Priority to AT07025118T priority patent/ATE448246T1/en
Priority to DE60043322T priority patent/DE60043322D1/en
Priority to EP00936172A priority patent/EP1208195A2/en
Priority to AT07025116T priority patent/ATE449109T1/en
Priority to JP2001503894A priority patent/JP2003529324A/en
Priority to EP07025117A priority patent/EP1978029A3/en
Priority to PCT/US2000/014042 priority patent/WO2000077037A2/en
Priority to CA2372511A priority patent/CA2372511C/en
Priority to EP07025116A priority patent/EP1953173B1/en
Priority to EP07025118A priority patent/EP1956030B1/en
Priority to DE60043367T priority patent/DE60043367D1/en
Priority to AU51527/00A priority patent/AU5152700A/en
Priority to AU54601/00A priority patent/AU5460100A/en
Priority to PCT/US2000/015264 priority patent/WO2000073452A2/en
Publication of WO2000015797A3 publication Critical patent/WO2000015797A3/en
Priority to US09/902,759 priority patent/US20030077654A1/en
Priority to US09/902,979 priority patent/US20030113718A1/en
Priority to US09/902,736 priority patent/US20030049676A1/en
Priority to US09/902,853 priority patent/US20020192659A1/en
Priority to US09/902,572 priority patent/US20030108983A1/en
Priority to US09/902,903 priority patent/US20030044839A1/en
Priority to US09/902,615 priority patent/US20030092002A1/en
Priority to US09/902,634 priority patent/US20030082540A1/en
Priority to US09/902,692 priority patent/US20030054400A1/en
Priority to US09/902,713 priority patent/US20030082541A1/en
Priority to US09/903,823 priority patent/US20030104381A1/en
Priority to US09/903,806 priority patent/US20030130489A1/en
Priority to US09/904,011 priority patent/US20030003530A1/en
Priority to US09/904,119 priority patent/US20030049621A1/en
Priority to US09/903,603 priority patent/US20030148419A1/en
Priority to US09/903,786 priority patent/US20030044793A1/en
Priority to US09/903,925 priority patent/US20030096233A1/en
Priority to US09/903,562 priority patent/US6965015B2/en
Priority to US09/903,749 priority patent/US7147853B2/en
Priority to US09/903,943 priority patent/US20030054349A1/en
Priority to US09/903,640 priority patent/US7208308B2/en
Priority to US09/903,520 priority patent/US20030054401A1/en
Priority to US09/904,786 priority patent/US7557192B2/en
Priority to US09/904,938 priority patent/US20030211569A1/en
Priority to US09/904,992 priority patent/US20030135025A1/en
Priority to US09/905,088 priority patent/US20030073077A1/en
Priority to US09/904,766 priority patent/US20030152999A1/en
Priority to US09/905,291 priority patent/US20020160374A1/en
Priority to US09/905,056 priority patent/US20030054441A1/en
Priority to US09/904,859 priority patent/US20030036060A1/en
Priority to US09/905,125 priority patent/US6664376B2/en
Priority to US09/904,877 priority patent/US20030186358A1/en
Priority to US09/905,449 priority patent/US6965011B2/en
Priority to US09/904,532 priority patent/US7151160B2/en
Priority to US09/905,381 priority patent/US6818746B2/en
Priority to US09/904,462 priority patent/US6878807B2/en
Priority to US09/904,485 priority patent/US20030064367A1/en
Priority to US09/905,075 priority patent/US7169906B2/en
Priority to US09/905,348 priority patent/US20030064923A1/en
Priority to US09/904,920 priority patent/US6806352B2/en
Priority to US09/904,820 priority patent/US20030036094A1/en
Priority to US09/904,838 priority patent/US20030148370A1/en
Priority to US09/904,553 priority patent/US20030059828A1/en
Priority to US09/904,956 priority patent/US20030049622A1/en
Priority to US09/906,618 priority patent/US6828146B2/en
Priority to US09/906,722 priority patent/US6946262B2/en
Priority to US09/906,646 priority patent/US6852848B2/en
Priority to US09/906,700 priority patent/US6723535B2/en
Priority to US09/906,742 priority patent/US20030023054A1/en
Priority to US09/906,760 priority patent/US20030096340A1/en
Priority to US09/906,777 priority patent/US20030148371A1/en
Priority to US09/906,838 priority patent/US7070979B2/en
Priority to US09/906,815 priority patent/US7094567B2/en
Priority to US09/907,652 priority patent/US20030104469A1/en
Priority to US09/907,613 priority patent/US20030027145A1/en
Priority to US09/907,841 priority patent/US7033825B2/en
Priority to US09/907,824 priority patent/US20020197671A1/en
Priority to US09/907,979 priority patent/US20030082542A1/en
Priority to US09/908,093 priority patent/US20030017498A1/en
Priority to US09/907,794 priority patent/US6635468B2/en
Priority to US09/907,575 priority patent/US20030073079A1/en
Priority to US09/907,942 priority patent/US7087738B2/en
Priority to US09/907,925 priority patent/US20030054352A1/en
Priority to US09/907,728 priority patent/US20030190611A1/en
Priority to US09/909,320 priority patent/US7074592B2/en
Priority to US09/909,064 priority patent/US6818449B2/en
Priority to US09/909,088 priority patent/US20020146709A1/en
Priority to US09/908,576 priority patent/US20040005553A1/en
Priority to US09/909,204 priority patent/US20030036061A1/en
Priority to US09/941,992 priority patent/US20030082546A1/en
Priority to US09/990,726 priority patent/US20030054359A1/en
Priority to US09/990,562 priority patent/US20030027985A1/en
Priority to US09/990,438 priority patent/US20030027754A1/en
Priority to US09/990,443 priority patent/US20030054987A1/en
Priority to US09/990,711 priority patent/US20030032023A1/en
Priority to US09/990,456 priority patent/US20020137890A1/en
Priority to US09/990,427 priority patent/US20030073809A1/en
Priority to US09/991,854 priority patent/US20030059780A1/en
Priority to US09/990,444 priority patent/US6930170B2/en
Priority to US09/993,469 priority patent/US20030068623A1/en
Priority to US09/991,163 priority patent/US20020132253A1/en
Priority to US09/992,521 priority patent/US20030083461A1/en
Priority to US09/993,748 priority patent/US20030069403A1/en
Priority to US09/992,598 priority patent/US6956108B2/en
Priority to US09/993,583 priority patent/US7074897B2/en
Priority to US09/993,667 priority patent/US20030022187A1/en
Priority to US09/990,436 priority patent/US20020198148A1/en
Priority to US09/990,442 priority patent/US20020132252A1/en
Priority to US09/990,440 priority patent/US20030060407A1/en
Priority to US09/993,687 priority patent/US20020198149A1/en
Priority to US09/991,073 priority patent/US20020127576A1/en
Priority to US09/997,440 priority patent/US20030059833A1/en
Priority to US09/998,041 priority patent/US20030119001A1/en
Priority to US09/997,573 priority patent/US20030049682A1/en
Priority to US09/997,585 priority patent/US7166282B2/en
Priority to US09/998,041 priority patent/US7309775B2/en
Priority to US09/997,542 priority patent/US20030068647A1/en
Priority to US09/997,514 priority patent/US7019116B2/en
Priority to US09/997,529 priority patent/US7309761B2/en
Priority to US09/997,614 priority patent/US20030124531A1/en
Priority to US09/997,641 priority patent/US7112656B2/en
Priority to US09/997,384 priority patent/US7119177B2/en
Priority to US09/997,653 priority patent/US7034122B2/en
Priority to US09/997,349 priority patent/US7034106B2/en
Priority to US09/997,683 priority patent/US20030059783A1/en
Priority to US10/002,796 priority patent/US20030032057A1/en
Priority to US09/997,666 priority patent/US7244816B2/en
Priority to US09/997,585 priority patent/US20030119055A1/en
Priority to US09/997,428 priority patent/US20030027162A1/en
Priority to US09/997,628 priority patent/US20030059782A1/en
Priority to US09/997,559 priority patent/US20030054403A1/en
Priority to US09/997,529 priority patent/US20030134284A1/en
Priority to US09/997,641 priority patent/US20030224358A1/en
Priority to US09/997,666 priority patent/US20030027163A1/en
Priority to US09/997,601 priority patent/US7189814B2/en
Priority to US09/998,156 priority patent/US20030044806A1/en
Priority to US09/997,384 priority patent/US20030087305A1/en
Priority to US09/997,601 priority patent/US20030054404A1/en
Priority to US09/997,333 priority patent/US6953836B2/en
Priority to US09/997,857 priority patent/US20030064375A1/en
Priority to US09/991,181 priority patent/US6913919B2/en
Priority to US09/991,150 priority patent/US20030194760A1/en
Priority to US09/991,172 priority patent/US20030050457A1/en
Priority to US09/990,441 priority patent/US7041804B2/en
Priority to US09/990,437 priority patent/US20030045463A1/en
Priority to US09/991,157 priority patent/US7101687B2/en
Priority to US09/989,723 priority patent/US20020072092A1/en
Priority to US09/989,722 priority patent/US20020072067A1/en
Priority to US09/989,726 priority patent/US7018811B2/en
Priority to US09/989,721 priority patent/US20020142961A1/en
Priority to US09/989,735 priority patent/US6972185B2/en
Priority to US09/989,279 priority patent/US7083978B2/en
Priority to US09/989,732 priority patent/US7037679B2/en
Priority to US09/989,328 priority patent/US7056736B2/en
Priority to US09/989,727 priority patent/US20020072497A1/en
Priority to US09/989,734 priority patent/US7491529B2/en
Priority to US09/989,729 priority patent/US20030059831A1/en
Priority to US09/989,862 priority patent/US20030130182A1/en
Priority to US09/989,730 priority patent/US7157247B2/en
Priority to US09/989,293 priority patent/US7034136B2/en
Priority to US09/989,731 priority patent/US20020103125A1/en
Priority to US09/989,728 priority patent/US7029873B2/en
Priority to US09/989,725 priority patent/US20030139329A1/en
Priority to US10/028,072 priority patent/US20030004311A1/en
Priority to US10/066,269 priority patent/US20030040014A1/en
Priority to US10/066,193 priority patent/US20030044902A1/en
Priority to US10/066,198 priority patent/US20030170721A1/en
Priority to US10/066,203 priority patent/US20030180796A1/en
Priority to US10/066,273 priority patent/US7317092B2/en
Priority to US10/066,211 priority patent/US20030044844A1/en
Priority to US10/066,500 priority patent/US20020177165A1/en
Priority to US10/066,494 priority patent/US20030032063A1/en
Priority to US10/081,056 priority patent/US20040043927A1/en
Priority to US10/121,046 priority patent/US20030194791A1/en
Priority to US10/121,040 priority patent/US20030082759A1/en
Priority to US10/121,042 priority patent/US20030096386A1/en
Priority to US10/121,059 priority patent/US20030190721A1/en
Priority to US10/121,051 priority patent/US20030092147A1/en
Priority to US10/121,047 priority patent/US20030077778A1/en
Priority to US10/121,044 priority patent/US20030190717A1/en
Priority to US10/121,041 priority patent/US20030077776A1/en
Priority to US10/121,045 priority patent/US20030073210A1/en
Priority to US10/121,048 priority patent/US20030199051A1/en
Priority to US10/121,043 priority patent/US7220831B2/en
Priority to US10/121,061 priority patent/US20030082761A1/en
Priority to US10/121,060 priority patent/US20030190722A1/en
Priority to US10/121,055 priority patent/US20030190718A1/en
Priority to US10/121,049 priority patent/US20030022239A1/en
Priority to US10/121,062 priority patent/US20030077779A1/en
Priority to US10/121,056 priority patent/US20030082760A1/en
Priority to US10/121,050 priority patent/US20030054516A1/en
Priority to US10/121,052 priority patent/US20030199052A1/en
Priority to US10/121,058 priority patent/US20030190720A1/en
Priority to US10/121,054 priority patent/US20030199054A1/en
Priority to US10/121,057 priority patent/US20030190719A1/en
Priority to US10/121,053 priority patent/US20030199053A1/en
Priority to US10/121,063 priority patent/US20030199055A1/en
Priority to US10/123,213 priority patent/US20030199057A1/en
Priority to US10/123,213 priority patent/US7193048B2/en
Priority to US10/123,155 priority patent/US20030068794A1/en
Priority to US10/123,212 priority patent/US7276577B2/en
Priority to US10/123,291 priority patent/US20030199058A1/en
Priority to US10/123,215 priority patent/US7291329B2/en
Priority to US10/123,108 priority patent/US7635478B2/en
Priority to US10/123,214 priority patent/US7343721B2/en
Priority to US10/123,261 priority patent/US20030068796A1/en
Priority to US10/123,109 priority patent/US20030190723A1/en
Priority to US10/123,235 priority patent/US20030082762A1/en
Priority to US10/123,771 priority patent/US20030199060A1/en
Priority to US10/123,292 priority patent/US20030073211A1/en
Priority to US10/123,154 priority patent/US20030190724A1/en
Priority to US10/123,157 priority patent/US20030190725A1/en
Priority to US10/123,322 priority patent/US20030199059A1/en
Priority to US10/123,156 priority patent/US20030194792A1/en
Priority to US10/123,236 priority patent/US20030068795A1/en
Priority to US10/123,262 priority patent/US20030049816A1/en
Priority to US10/123,904 priority patent/US20030022328A1/en
Priority to US10/123,906 priority patent/US20030190726A1/en
Priority to US10/123,905 priority patent/US20030087344A1/en
Priority to US10/123,907 priority patent/US7084258B2/en
Priority to US10/123,913 priority patent/US20030203462A1/en
Priority to US10/123,908 priority patent/US7335728B2/en
Priority to US10/123,902 priority patent/US20030077781A1/en
Priority to US10/123,905 priority patent/US7285625B2/en
Priority to US10/123,909 priority patent/US7193049B2/en
Priority to US10/123,912 priority patent/US20030100087A1/en
Priority to US10/123,910 priority patent/US7329404B2/en
Priority to US10/123,911 priority patent/US7408032B2/en
Priority to US10/123,903 priority patent/US20030073212A1/en
Priority to US10/124,816 priority patent/US20030190728A1/en
Priority to US10/125,805 priority patent/US20030194794A1/en
Priority to US10/124,817 priority patent/US20030077786A1/en
Priority to US10/124,822 priority patent/US7109305B2/en
Priority to US10/125,704 priority patent/US7357926B2/en
Priority to US10/124,818 priority patent/US20030082763A1/en
Priority to US10/124,824 priority patent/US20030077659A1/en
Priority to US10/125,795 priority patent/US7304131B2/en
Priority to US10/124,823 priority patent/US20030199062A1/en
Priority to US10/124,814 priority patent/US7105335B2/en
Priority to US10/124,813 priority patent/US7312307B2/en
Priority to US10/124,819 priority patent/US7285626B2/en
Priority to US10/124,821 priority patent/US20030199023A1/en
Priority to US10/124,820 priority patent/US20030190729A1/en
Priority to US10/125,927 priority patent/US20030190731A1/en
Priority to US10/125,931 priority patent/US20030199063A1/en
Priority to US10/125,932 priority patent/US7317079B2/en
Priority to US10/125,922 priority patent/US7309762B2/en
Priority to US10/125,924 priority patent/US7342097B2/en
Priority to US10/127,831 priority patent/US20030082689A1/en
Priority to US10/128,689 priority patent/US20030087365A1/en
Priority to US10/131,825 priority patent/US7282566B2/en
Priority to US10/131,817 priority patent/US7291701B2/en
Priority to US10/131,823 priority patent/US7304132B2/en
Priority to US10/137,865 priority patent/US20030032155A1/en
Priority to US10/137,867 priority patent/US20030207349A1/en
Priority to US10/137,868 priority patent/US20030082764A1/en
Priority to US10/139,963 priority patent/US7288625B2/en
Priority to US10/140,020 priority patent/US20030207415A1/en
Priority to US10/140,470 priority patent/US20030022331A1/en
Priority to US10/140,024 priority patent/US20040058424A1/en
Priority to US10/140,023 priority patent/US20030207416A1/en
Priority to US10/140,474 priority patent/US20030032156A1/en
Priority to US10/139,980 priority patent/US7247710B2/en
Priority to US10/140,805 priority patent/US20030207417A1/en
Priority to US10/140,921 priority patent/US7317080B2/en
Priority to US10/140,865 priority patent/US20030207420A1/en
Priority to US10/140,928 priority patent/US20030068798A1/en
Priority to US10/140,860 priority patent/US7307151B2/en
Priority to US10/140,808 priority patent/US7425621B2/en
Priority to US10/140,925 priority patent/US20030073215A1/en
Priority to US10/140,864 priority patent/US20030207419A1/en
Priority to US10/140,809 priority patent/US20030207418A1/en
Priority to US10/141,755 priority patent/US7297764B2/en
Priority to US10/141,760 priority patent/US7342104B2/en
Priority to US10/141,756 priority patent/US7488586B2/en
Priority to US10/141,701 priority patent/US20030207421A1/en
Priority to US10/141,754 priority patent/US7361732B2/en
Priority to US10/142,425 priority patent/US20030207424A1/en
Priority to US10/143,114 priority patent/US20030036180A1/en
Priority to US10/142,417 priority patent/US7304133B2/en
Priority to US10/142,430 priority patent/US7309766B2/en
Priority to US10/143,113 priority patent/US7329730B2/en
Priority to US10/142,431 priority patent/US7285629B2/en
Priority to US10/142,419 priority patent/US7153941B2/en
Priority to US10/143,032 priority patent/US7408033B2/en
Priority to US10/142,423 priority patent/US20030049817A1/en
Priority to US10/146,730 priority patent/US20030207427A1/en
Priority to US10/146,792 priority patent/US20030207428A1/en
Priority to US10/147,528 priority patent/US20030219885A1/en
Priority to US10/147,536 priority patent/US20040077064A1/en
Priority to US10/147,519 priority patent/US20030077791A1/en
Priority to US10/147,492 priority patent/US20030082765A1/en
Priority to US10/152,395 priority patent/US7189534B2/en
Priority to US10/153,934 priority patent/US20030129695A1/en
Priority to US10/156,843 priority patent/US20030207805A1/en
Priority to US10/157,786 priority patent/US20030208055A1/en
Priority to US10/157,782 priority patent/US20030077792A1/en
Priority to US10/160,498 priority patent/US20030073216A1/en
Priority to US10/158,791 priority patent/US20030207429A1/en
Priority to US10/158,782 priority patent/US20030082766A1/en
Priority to US10/176,913 priority patent/US20030022298A1/en
Priority to US10/187,885 priority patent/US20030032138A1/en
Priority to US10/226,739 priority patent/US7390879B2/en
Priority to JP2002341509A priority patent/JP4017507B2/en
Priority to US10/771,187 priority patent/US7355002B2/en
Priority to JP2004212340A priority patent/JP3803681B2/en
Priority to US10/970,823 priority patent/US7307152B2/en
Priority to JP2005066570A priority patent/JP2005237383A/en
Priority to JP2005066569A priority patent/JP4037876B2/en
Priority to US11/100,159 priority patent/US7425613B2/en
Priority to JP2005180188A priority patent/JP4358159B2/en
Priority to JP2005235120A priority patent/JP2006068006A/en
Priority to JP2005238274A priority patent/JP2006051032A/en
Priority to JP2005238266A priority patent/JP2006025795A/en
Priority to JP2005238244A priority patent/JP2006061156A/en
Priority to JP2005238217A priority patent/JP2006051031A/en
Priority to US11/240,891 priority patent/US20060246540A1/en
Priority to US11/341,175 priority patent/US7468427B2/en
Priority to US11/518,609 priority patent/US20070077623A1/en
Priority to JP2007138469A priority patent/JP2007291116A/en
Priority to JP2007138468A priority patent/JP2007222180A/en
Priority to JP2007172879A priority patent/JP5512073B2/en
Priority to IL186154A priority patent/IL186154A0/en
Priority to IL186153A priority patent/IL186153A0/en
Priority to IL202176A priority patent/IL202176A0/en
Priority to JP2011138276A priority patent/JP2011236224A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/18Drugs for disorders of the endocrine system of the parathyroid hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

The present invention relates to a composition containing novel proteins and methods for the diagnosis and treatment of immune related diseases.

Description

COMPOSITIONS AND METHODS FOR THE TREATMENT OF IMMUNE RELATED
DISEASES
Field of the Invention The present invention relates to compositions and methods for the diagnosis and treatment of immune related diseases.
Background of the Invention
Immune related and inflammatory diseases are the manifestation or consequence of fairly complex, often multiple interconnected biological pathways which in normal physiology are critical to respond to insult or injury, initiate repair from insult or injury, and mount innate and acquired defense against foreign organisms. Disease or pathology occurs when these normal physiological pathways cause additional insult or injury either as directly related to the intensity of the response, as a consequence of abnormal regulation or excessive stimulation, as a reaction to self, or as a combination of these.
Though the genesis of these diseases often involves multistep pathways and often multiple different biological systems/pathways, intervention at critical points in one or more of these pathways can have an ameliorative or therapeutic effect. Therapeutic intervention can occur by either antagonism of a detrimental process/pathway or stimulation of a beneficial process/pathway.
Many immune related diseases are known and have been extensively studied. Such diseases include immune-mediated inflammatory diseases, non-immune-mediated inflammatory diseases, infectious diseases, immunodeficiency diseases, neoplasia, etc.
T lymphocytes (T cells) are an important component of a mammalian immune response. T cells recognise antigens which are associated with a self-molecule encoded by genes within the major histocompatibility complex (MHC). The antigen may be displayed together with MHC molecules on the surface of antigen presenting cells, virus infected cells, cancer cells, grafts, etc. The T cell system eliminates these altered cells which pose a health threat to the host mammal. T cells include helper T cells and cytotoxic T cells. Helper T cells proliferate extensively following recognition of an antigen -MHC complex on an antigen presenting cell. Helper T cells also secrete a variety of cytokines, i.e. lymphokines, which play a central role in the activation of B cells, cytotoxic T cells and a variety of other cells which participate in the immune response.
A central event in both humoral and cell mediated immune responses is the activation and clonal expansion of helper T cells. Helper T cell activation is initiate by the interaction of the T cell receptor (TCR) - CD3 complex with an antigen-MHC on the surface of an antigen presenting cell. This interaction mediates a cascade of biochemical events that induce the resting helper T cell to enter a cell cycle (the Go to G 1 transition) and results in the expression of a high affinity receptor for IL-2 and sometimes I -4 The activated T cell progresses through the cycle proliferating and differentiating into memory cells or effector cells
In addition to the signals mediated through the TCR, activation of T cells involves additional costimulation induced by cytokines released by the antigen presenting cell or through interactions with membrane bound molecules on the antigen presenting cell and the T cell The cytokines IL- 1 and IL-6 have been shown to provide a costimulatory signal Also, the interaction between the B7 molecule expressed on the surface of an antigen presenting cell and CD28 and CTLA-4 molecules expressed on the T cell surface effect T cell activation Activated T cells express an increased number of cellular adhesion molecules, such as ICAM-1, mtegnns, VLA-4, LFA-1, CD56, etc
T-cell proliferation in a mixed lymphocyte culture or mixed lymphocyte reaction (MLR) is an established indication of the ability of a compound to stimulate the immune system In many immune responses, inflammatory cells infiltrate the site of injury or infection The migrating cells may be neutrophihc, eosinophilic, monocytic or lymphocytic Histologic examination of the affected tissues provides evidence of an immune stimulating or inhibiting responseCurrent Protocols in Immunology, ed John E Coligan, 1994, John Wiley & Sons, Ine
Immune related diseases can be treated by suppressing the immune response Using neutralizing antibodies that inhibit molecules having immune stimulatory activity would be beneficial in the treatment of immune-mediated and inflammatory diseases Molecules which inhibit the immune response can be utilized (proteins directly or via the use of antibody agonists) to inhibit the immune response and thus ameliorate immune related disease
Summary of the Invention The present invention concerns compositions and methods for the diagnosis and treatment of immune related disease in mammals, including humans The present invention is based on the identification of proteins (including agonist and antagonist antibodies) which either stimulate or inhibit the immune response in mammals Immune related diseases can be treated by suppressing or enhancing the immune response Molecules that enhance the immune response stimulate or potentiate the immune response to an antigen Molecules which stimulate the immune response can be used therapeutically where enhancement of the immune response would be beneficial Such stimulatory- molecules can also be inhibited where suppression of the immune response would be of value Neutralizing antibodies are examples of molecules that inhibit molecules having immune stimulatory activity and which would be beneficial in the treatment of immune related and inflammatory diseases Molecules which inhibit the immune response can also be utilized (proteins directly or via the use of antibody agonists) to inhibit the immune response and thus ameliorate immune related disease Accordingly, the proteins of the invention encoded by the genes of the invention are useful for the diagnosis and/or treatment (including prevention) of immune related diseases. Antibodies which bind to stimulatory proteins are useful to suppress the immune system and the immune response. Antibodies which bind to inhibitory proteins are useful to stimulate the immune system and the immune response. The proteins and antibodies of the invention are also useful to prepare medicines and medicaments for the treatment of immune related and inflammatory diseases.
In one embodiment, the present invention concerns an isolated antibody which binds a PR0245, PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 polypeptide. In one aspect, the antibody mimics the activity of a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide (an agonist antibody) or conversely the antibody inhibits or neutralizes the activity of a PR0245, PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide (an antagonist antibody). In another aspect, the antibody is a monoclonal antibody, which preferably has nonhuman complementarity determining region (CDR) residues and human framework region (FR) residues. The antibody may be labeled and may be immobilized on a solid support. In a further aspect, the antibody is an antibody fragment, a single-chain antibody, or an anti-idiotypic antibody.
In another embodiment, the invention concerns a composition containing a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide or an agonist or antagonist antibody which binds the polypeptide in admixture with a carrier or excipient. In one aspect, the composition contains a therapeutically effective amount of the peptide or antibody. In another aspect, when the composition contains an immune stimulating molecule, the composition is useful for: (a) increasing infiltration of inflammatory cells into a tissue of a mammal in need thereof, (b) stimulating or enhancing an immune response in a mammal in need thereof, or (c) increasing the proliferation of T-lymphocytes in a mammal in need thereof in response to an antigen. In a further aspect, when the composition contains an immune inhibiting molecule, the composition is useful for: (a) decreasing infiltration of inflammatory cells into a tissue of a mammal in need thereof, (b) inhibiting or reducing an immune response in a mammal in need thereof, or (c) decreasing the proliferation of T- lymphocytes in a mammal in need thereof in response to an antigen. In another aspect, the composition contains a further active ingredient, which may, for example, be a further antibody or a cytotoxic or chemotherapeutic agent. Preferably, the composition is sterile.
In another embodiment, the invention concerns the use of the polypeptides and antibodies of the invention to prepare a composition or medicament which has the uses described above.
In a further embodiment, the invention concerns nucleic acid encoding an anti-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 antibody, and vectors and recombinant host cells comprising such nucleic acid. In a still further embodiment, the invention concerns a method for producing such an antibody by culturing a host cell transformed with nucleic acid encoding the antibody under conditions such that the antibody is expressed, and recovering the
* antibody from the cell culture.
The invention further concerns antagonists and agonists of a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide that inhibit one or more of the functions or activities of the PR0245, PR0217, PRO301. PR0266, PR0335, PR0331 or PR0326 polypeptide.
In a further embodiment, the invention concerns isolated nucleic acid molecules that hybridize to the complement of the nucleic acid molecules encoding the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptides. The nucleic acid preferably is DNA, and hybridization preferably occurs under stringent conditions. Such nucleic acid molecules can act as antisense molecules of the amplified genes identified herein, which, in turn, can find use in the modulation of the respective amplified genes, or as antisense primers in amplification reactions. Furthermore, such sequences can be used as part of ribozyme and/or triple helix sequence which, in turn, may be used in regulation of the amplified genes.
In another embodiment, the invention concerns a method for determining the presence of a PR0245, PR0217, PRO301, PR0266. PR0335, PR0331 or PR0326 polypeptide comprising exposing a cell suspected of containing the polypeptide to an anti-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 antibody and determining binding of the antibody to the cell.
In yet another embodiment, the present invention concerns a method of diagnosing an immune related disease in a mammal, comprising detecting the level of expression of a gene encoding a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide (a) in a test sample of tissue cells obtained from the mammal, and (b) in a control sample of known normal tissue cells of the same cell type, wherein a higher expression level in the test sample indicates the presence of immune related disease in the mammal from which the test tissue cells were obtained.
In another embodiment, the present invention concerns a method of diagnosing an immune disease in a mammal, comprising (a) contacting an anti-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 antibody with a test sample of tissue cells obtained from the mammal , and (b) detecting the formation of a complex between the antibody and the PR0245, PR0217, PRO301, PR0266. PR0335, PR0331 or PR0326 polypeptide in the test sample. The detection may be qualitative or quantitative, and may be performed in comparison with monitoring the complex formation in a control sample of known normal tissue cells of the same cell type. A larger quantity of complexes formed in the test sample indicates the presence of tumor in the mammal from which the test tissue cells were obtained. The antibody preferably carries a detectable label. Complex formation can be monitored, for example, by light microscopy, flow cytometry, fluorimetry, or other techniques known in the art. The test sample is usually obtained from an individual suspected of having a deficiency or abnormality of the immune system.
In another embodiment, the present invention concerns a diagnostic kit, containing an anti-
PR0245, PR0217, PRO301, PR0266. PR0335. PR0331 or PR0326 antibody and a carrier (e.g. a buffer) in suitable packaging. The kit preferably contains instructions for using the antibody to detect the PR0245, PR0217, PRO301, PR0266. PR0335. PR0331 or PR0326 polypeptide.
In a further embodiment, the invention concerns an article of manufacture, comprising: a container; a label on the container; and a composition comprising an active agent contained within the container; wherein the composition is effective for stimulating or inhibiting an immune response in a mammal, the label on the container indicates that the composition can be used to treat an immune related disease, and the active agent in the composition is an agent stimulating or inhibiting the expression and/or activity of the PR0245, PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 polypeptide. In a preferred aspect, the active agent is a PR0245, PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide or an anti-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 antibody.
A further embodiment is a method for identifying a compound capable of inhibiting the expression and/or activity of a PR0245. PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide by contacting a candidate compound with a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide under conditions and for a time sufficient to allow these two components to interact. In a specific aspect, either the candidate compound or the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide is immobilized on a solid support. In another aspect, the non-immobilized component carries a detectable label.
Brief Description of the Drawings
Figures 1 A-D and Table 4 show hypothetical exemplifications for using the below described method to determine % amino acid sequence identity (Figures 1 A-B) and % nucleic acid sequence identity (Figures 1C-D) using the ALIGN-2 sequence comparison computer program, wherein "PRO" represents the amino acid sequence of a hypothetical polypeptide of the invention of interest, "Comparison Protein" represents the amino acid sequence of a polypeptide against which the "PRO" polypeptide of interest is being compared, "PRO-DNA" represents a hypothetical "PRO"-encoding nucleic acid sequence of interest, "Comparison DNA" represents the nucleotide sequence of a nucleic acid molecule against which the "PRO-DNA" nucleic acid molecule of interest is being compared, "X", "Y" and "Z" each represent different hypothetical amino acid residues and "N", "L" and "V" each represent different hypothetical nucleotides.
Figures 2A-P and Table 5 provide the complete source code for the ALIGN-2 sequence comparison computer program. This source code may be routinely compiled for use on a UNIX operating system to provide the ALIGN-2 sequence comparison computer program.
Figure 3 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PR0245(UNQ219), wherein the nucleotide sequence (SEQ ID NO: 1) is a clone designated herein as "DNA35638" Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 4 and Table 6 show the ammo acid sequence (SEQ ID NO 2) of a native sequence PR0245 polypeptide as derived from the coding sequence of Figure 3 Also shown are the approximate locations of vanous other important polypeptide domains if known
Figure 5 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PR0217(UNQ 191 ), wherein the nucleotide sequence (SEQ ID NO 3) is a clone designated herein as "DNA33094" Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 6 and Table 7 show the amino acid sequence (SEQ ID NO 4) of a native sequence PR0217 polypeptide as deπved from the coding sequence of Figure 5 Also shown are the approximate locations of vanous other important polypeptide domains if known
Figure 7 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PRO301(UNQ264), wherein the nucleotide sequence (SEQ ID NO 5) is a clone designated herein as "DNA40628" Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 8 and Table 8 show the amino acid sequence (SEQ ID NO 6) of a native sequence PRO301 polypeptide as deπved from the coding sequence of Figure 7 Also shown are the approximate locations of vanous other important polypeptide domams if known
Figure 9 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PR0266 (UNQ233), wherein the nucleotide sequence (SEQ ID NO 7) is a clone designated herein as "DNA37150" Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 10 and Table 9 show the amino acid sequence (SEQ ID NO 8) of a native sequence PR0266 polypeptide as deπved from the coding sequence of Figure 9 Also shown are the approximate locations of vanous other important polypeptide domains if known
Figure 11 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PR0335 (UNQ287V), wherein the nucleotide sequence (SEQ ID NO 9) is a clone designated herein as "DNA41388" Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 12 and Table 10 show the ammo acid sequence (SEQ ID NO 10) of a native sequence PR0335 polypeptide as deπved from the coding sequence of Figure 1 1 Also shown are the approximate locations of vanous other important polypeptide domains if known
Figure 13 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PR0331 (UNQ292), wherein the nucleotide sequence (SEQ ID NO 1 1) is a clone designated herein as "DNA40981 " Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 14 and Table 1 1 show the ammo acid sequence (SEQ ID NO 12) of a native sequence PR0331 polypeptide as deπved from the coding sequence of Figure 13 Also shown are the approximate locations of vanous other important polypeptide domains if known
Figure 15 shows the nucleotide sequence of a cDNA containing a nucleotide sequence encoding native sequence PR0326 (UNQ287), wherein the nucleotide sequence (SEQ ID NO 13) is a clone designated herein as "DNA37140" Also presented in bold font and underlined are the positions of the respective start and stop codons
Figure 16 and Table 12 show the ammo acid sequence (SEQ ID NO 14) of a native sequence PR0331 polypeptide as deπved from the coding sequence of Figure 15 Also shown are the approximate locations of vanous other important polypeptide domains if known
Detailed Descπption of the Preferred Embodiments I Definitions
The term "immune related disease" means a disease in which a component of the immune system of a mammal causes, mediates or otherwise contnbutes to a morbidity in the mammal Also included are diseases in which stimulation or intervention of the immune response has an ameliorative effect on progression of the disease Included within this term are immune-mediated inflammatory diseases, non-immune-mediated inflammatory diseases, infectious diseases, immunodeficiency diseases, neoplasia, etc
The term "T cell mediated" disease means a disease in which T cells directly or indirectly mediate or otherwise contribute to a morbidity in a mammal The T cell mediated disease may be associated with cell mediated effects, lymphokine mediated effects, etc , and even effects associated with B cells if the B cells are stimulated, for example, by the lymphokmes secreted by T cells
Examples of immune-related and inflammatory diseases, some of which are lmune or T cell mediated, which can be treated according to the invention include systemic lupus erythematosis, rheumatoid arthntis, juvenile chronic arthπtis, spondyloarthropathies, systemic sclerosis (scleroderma). idiopathic inflammatory myopathies (dermatomyositis, polymyositis), Sjsgren's syndrome, systemic vascuhtis, sarcoidosis, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuria), autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura. immune-mediated thrombocytopenia), thyroiditis (Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic thyroiditis), diabetes mellitus. immune-mediated renal disease (glomerulonephntis, tubulointerstitial nephπtis), demvehnating diseases of the central and peπpheral nervous systems such as multiple sclerosis, idiopathic demyehnatmg polyneuropathy or Guillam-Barre syndrome, and chronic inflammatory demyehnatmg polyneuropathy, hepatobi ary diseases such as infectious hepatitis (hepatitis A, B, C, D, E and other non-hepatotropic viruses), autoimmune chronic active hepatitis, pnmary biliary cirrhosis, granulomatous hepatitis, and sclerosmg cholangitis, inflammatory and fibrotic lung diseases such as inflammatory bowel disease (ulcerative colitis Crohn's disease), gluten-sensitive enteropathy, and Whipple's disease, autoimmune or immune-mediated skin diseases including bullous skin diseases, erythema multiforme and contact dermatitis, psonasis, allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and urticana, lmmunologic diseases of the lung such as eosmophilic pneumomas, idiopathic pulmonary fibrosis and hypersensitivity pneumonitis, transplantation associated diseases including graft rejection and graft -versus-host-disease Infectious diseases include AIDS (HIV infection), hepatitis A, B, C, D, and E, bacteπal infections, fungal infections, protozoal infections and parasitic infections
"Treatment" is an intervention performed with the intention of preventing the development or alteπng the pathology of a disorder Accordingly, "treatment" refers to both therapeutic treatment and prophylactic or preventative measures Those in need of treatment include those already with the disorder as well as those m which the disorder is to be prevented In treatment of an immune related disease, a therapeutic agent may directly decrease or increase the magnitude of response of a component of the immune response, or render the disease more susceptible to treatment by other therapeutic agents, e g antibiotics, antifungals, anti-mflammatory agents, chemotherapeutics, etc
The "pathology" of an immune related disease includes all phenomena that compromise the well-being of the patient This includes, without limitation, abnormal or uncontrollable cell growth (neutrophihc, eosmophilic, monocytic, lymphocytic cells), antibody production, auto-antibody production, complement production, interference with the normal functioning of neighbonng cells, release of cytokines or other secretory products at abnormal levels, suppression or aggravation of any inflammatory or immunological response, infiltration of inflammatory cells (neutrophihc. eosmophilic, monocytic, lymphocytic) into cellular spaces, etc
"Mammal" for purposes of treatment refers to any animal classified as a mammal, including humans, domestic and farm animals, and zoo, sports, or pet animals, such as dogs, horses, cats, cows, etc Preferably, the mammal is human
Administration "in combination with" one or more further therapeutic agents includes simultaneous (concurrent) and consecutive administration m any order
"Chronic" administration refers to administration of the agent(s) in a continuous mode as opposed to an acute mode, so as to maintain the initial therapeutic effect (activity) for an extended peπod of time "Intermittent" administration is treatment that is not consecutively done v\ ithout interruption, but rather is cyclic in nature "Carriers" as used herein include pharmaceutically acceptable carriers, excipients. or stabilizers which are nontoxic to the cell or mammal being exposed thereto at the dosages and concentrations employed. Often the physiologically acceptable earner is an aqueous pH buffered solution. Examples of physiologically acceptable earners include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid, low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrohdone; amino acids such as glycme, glutamme, asparagine, arginme or lysine; monosacchaπdes, disacchandes, and other carbohydrates including glucose, mannose, or dextπns; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol, salt-forming countenons such as sodium; and/or nomonic surfactants such as TWEEN™, polyethylene glycol (PEG), and PLURONICS™
The term "cytotoxic agent" as used herein refers to a substance that inhibits or prevents the function of cells and/or causes destruction of cells The term is intended to include radioactive isotopes (e g I131, 1125, Y90 and Re186), chemotherapeutic agents, and toxins such as enzymatically active toxins of bacteπal, fungal, plant or animal ongin, or fragments thereof.
A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include adπamycm, doxorubicm, epirubicin, 5-fluorouracιl, cytosine arabmoside ("Ara-C"), cyclophosphamide, thiotepa, busulfan, cytoxm, taxoids, e.g. paclitaxel (Taxol, Bπstol-Myers Squibb Oncology, Pπnceton, NJ), and doxetaxel (Taxotere, Rhone- Poulenc Rorer, Antony, Rnace), toxotere, methotrexate, cisplatin, melphalan, vinblastme, bleomycm, etoposide, lfosfamide, mitomycin C. mitoxantrone, vmcπstine, vinorelbme, carboplatin, temposide, daunomycm, carmmomycm, ammoptenn. dactinomycin, mitomycins, esperamicins (see U.S. Pat. No. 4,675, 187), melphalan and other related nitrogen mustards. Also included in this definition are hormonal agents that act to regulate or inhibit hormone action on tumors such as tamoxifen and onapπstone.
A "growth inhibitory agent" when used herein refers to a compound or composition which inhibits growth of a cell, especially cancer cell overexpressmg any of the genes identified herein, either in vitro or in vivo Thus, the growth inhibitory agent is one which significantly reduces the percentage of cells overexpressmg such genes in S phase. Examples of growth inhibitory agents include agents that block cell cycle progression (at a place other than S phase), such as agents that induce Gl arrest and M-phase arrest Classical M-phase blockers include the vincas (vincnstine and vinblastme), taxol, and topo II inhibitors such as doxorubicm, epirubicin, daunorubicm, etoposide, and bleomycm. Those agents that arrest G 1 also spill over into S-phase arrest, for example, DNA alkylating agents such as tamoxifen. pred sone, dacarbazine, mechlorethamme, cisplatin, methotrexate, 5-fluorouracil, and ara-C Further information can be found in The Molecular Basis of
Cancer, Mendelsohn and Israel, eds . Chapter 1, entitled "Cell cycle regulation, oncogens, and antineoplastic drugs" by Murakami et al. (WB Saunders: Philadelphia, 1995), especially p. 13.
The term "cytokine" is a generic term for proteins released by one cell population which act on another cell as intercellular mediators. Examples of such cytokines are lymphokines, monokines, and traditional polypeptide hormones. Included among the cytokines are growth hormone such as human growth hormone, N-methionyl human growth hormone, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin; prorelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and luteinizing hormone (LH); hepatic growth factor; fibroblast growth factor; prolactin; placental lactogen; tumor necrosis factor-alpha and -beta; mullerian-inhibiting substance; mouse gonadotropin-associated peptide; inhibin; activin; vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF-beta; platelet-growth factor; transforming growth factors (TGFs) such as TGF- alpha and TGF-beta; insulin-like growth factor-I and -II; erythropoietin (EPO); osteoinductive factors; interferons such as interferon-alpha, -beta, and -gamma; colony stimulating factors (CSFs) such as macrophage-CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G- CSF); interleukins (ILs) such as IL-1, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-11, IL- 12; a tumor necrosis factor such as TNF-alpha or TNF-beta; and other polypeptide factors including LIF and kit ligand (KL). As used herein, the term cytokine includes proteins from natural sources or from recombinant cell culture and biologically active equivalents of the native sequence cytokines.
As used herein, a "PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide" refers to a native sequence PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 having the same amino acid sequence as a PR0245, PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326 derived from nature. Such native sequence PR0245, PR0217, PRO301, PR0266. PR0335, PR0331 or PR0326 can be isolated from nature or can be produced by recombinant and/or synthetic means. The term specifically encompasses naturally-occurring truncated or secreted forms (e.g., an extracellular domain sequence), naturally-occurring variant forms (e.g., alternatively spliced forms) and naturally-occurring allelic variants of the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326. In one embodiment of the invention, the native sequence PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 is a mature or full- length native sequence PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 comprising the amino acid sequences shown in Figures 4, 6, 8, 10, 12, 14 and 16.
The term "polypeptide of the invention" refers to each individual PR0245, PR0217, PRO301. PR0266, PR0335, PR0331 or PR0326 polypeptide. All disclosures in this specification which refer to the "polypeptide of the invention" or to "the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide" refer to each of the polypeptides individually as well as jointly. For example, descriptions of the preparation of, purification of. derivation of, formation of antibodies to or against, administration of, compositions containing, treatment of a disease with, etc., pertain to each polypeptide of the invention individually The term "compound of the invention" includes the polypeptide of the invention, as well as agonist antibodies for and antagonist antibodies to these polypeptide, peptides or small molecules having agonist or antagonist activity developed from the polypeptide, etc
The term "polypeptide of the invention" also includes variants of the PR0245 PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptides A "vaπant" polypeptide means an active polypeptide as defined below having at least about 80% ammo acid sequence identity with the amino acid sequence of the PR0245, PR0217. PRO301, PR0266. PR0335, PR0331 or PR0326 polypeptides Such vaπant polypeptides include, for instance, polypeptides wherein one or more amino acid residues are added, or deleted, at the N- and/or C-terminus, as well as within one or more internal domains Ordinaπly, a vaπant polypeptide will have at least about 80% ammo acid sequence identity, more preferably at least about 81 % amino acid sequence identity, more preferably at least about 82% ammo acid sequence identity, more preferably at least about 83% ammo acid sequence identity, more preferably at least about 84% amino acid sequence identity, more preferably at least about 85% amino acid sequence identity, more preferably at least about 86% amino acid sequence identity, more preferably at least about 87% ammo acid sequence identity, more preferably at least about 88% ammo acid sequence identity, more preferably at least about 89% ammo acid sequence identity, more preferably at least about 90% amino acid sequence identity, more preferably at least about 91% amino acid sequence identity, more preferably at least about 92% ammo acid sequence identity, more preferably at least about 93% amino acid sequence identity, more preferably at least about 94% amino acid sequence identity, more preferably at least about 95% ammo acid sequence identity, more preferably at least about 96% amino acid sequence identity, more preferably at least about 97% ammo acid sequence identity, more preferably at least about 98% ammo acid sequence identity and yet more preferably at least about 99% amino acid sequence identity with the amino acid sequence of the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptides Vanants do not encompass the native polypeptide sequence
Ordmanly, vaπant polypeptides of the invention are at least about 10 amino acids in length, often at least about 20 amino acids in length, more often at least about 30 ammo acids in length, more often at least about 40 amino acids in length, more often at least about 50 ammo acids in length, more often at least about 60 amino acids in length, more often at least about 70 amino acids in length, more often at least about 80 amino acids m length, more often at least about 90 amino acids in length, more often at least about 100 amino acids in length, more often at least about 150 amino acids in length, more often at least about 200 amino acids m length, more often at least about 300 ammo acids in length, or more
"Percent (%) amino acid sequence identity" with respect to the polypeptide sequences identified herein is defined as the percentage of ammo acid residues in a candidate sequence that are identical with the ammo acid residues in a sequence of the PR0245, PR0217, PRO301. PR0266, PR0335, PR0331 or PR0326 polypeptides, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not consideπng any conservative substitutions as part of the sequence identity Alignment for purposes of determining percent amino acid sequence identity can be achieved in vanous ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megahgn (DNASTAR) software Those skilled in the art can determine appropπate parameters for measuπng alignment, including any algorithms needed to achieve maximal alignment over the full- length of the sequences being compared For purposes herein, however, % ammo acid sequence identity values are obtained as descπbed below by using the sequence compaπson computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Figures 2A-P The ALIGN-2 sequence compaπson computer program was authored by Genentech, Ine and the source code shown in Figures 2A-P has been filed with user documentation in the U S Copyπght Office. Washington D C . 20559, where it is registered under U S Copynght Registration No TXU510087 The ALIGN-2 program is publicly available through Genentech, Ine , South San Francisco, California or may be compiled from the source code provided in Figures 2A-P The ALIGN-2 program should be compiled for use on a UNIX operating system, preferably digital UNIX V4.0D All sequence compaπson parameters are set by the ALIGN-2 program and do not vary
For purposes herein, the % ammo acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or compπses a certain % amino acid sequence identity to, with, or agamst a given ammo acid sequence B) is calculated as follows
100 times the fraction X/Y
where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of ammo acid residues in B It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % ammo acid sequence identity of A to B will not equal the % ammo acid sequence identity of B to A As examples of % amino acid sequence identity calculations, Figures 1 A-B demonstrate how to calculate the % amino acid sequence identity of the amino acid sequence designated "Compaπson Protein" to the amino acid sequence designated "PRO"
Unless specifically stated otherwise, all % amino acid sequence identity values used herein are obtained as descπbed above using the ALIGN-2 sequence companson computer program However, % ammo acid sequence identity may also be determined using the sequence companson program NCBI-BLAST2 (Altschul et al , Nucleic Acids Res 25 3389-3402 (1997)) The NCBI- BLAST2 sequence compaπson program may be downloaded from http //www ncbi nlm mh gov NCBI-BLAST2 uses several search parameters, wherein all of those search parameters are set to default values including, for example, unmask = yes, strand = all, expected occurrences = 10, minimum low complexity length = 15/5, multi-pass e-value = 0 01, constant for multi-pass = 25, dropoff for final gapped alignment = 25 and scoπng matπx = BLOSUM62
In situations where NCBI-BLAST2 is employed for ammo acid sequence compansons. the % ammo acid sequence identity of a given ammo acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given ammo acid sequence A that has or compπses a certain % amino acid sequence identity to, with, or against a given ammo acid sequence B) is calculated as follows
100 times the fraction X/Y
where X is the number of ammo acid residues scored as identical matches by the sequence alignment program NCBI-BLAST2 m that program's alignment of A and B, and where Y is the total number of amino acid residues m B It will be appreciated that where the length of ammo acid sequence A is not equal to the length of ammo acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A
Also included within the term "polypeptides of the invention" are polypeptides which in the context of the amino acid sequence identity compansons performed as descπbed above, include ammo acid residues in the sequences compared that are not only identical, but also those that have similar properties These polypeptides are termed "positives" Amino acid residues that score a positive value to an ammo acid residue of interest are those that are either identical to the ammo acid residue of interest or are a preferred substitution (as defined in Table 1 below) of the ammo acid residue of interest For purposes herein, the % value of positives of a given amino acid sequence A to, with, or against a given ammo acid sequence B (which can alternatively be phrased as a given ammo acid sequence A that has or compπses a certain % positives to, with, or against a given ammo acid sequence B) is calculated as follows
100 times the fraction X/Y
where X is the number of ammo acid residues scoπng a positive value as defined above by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of ammo acid residues in B It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % positives of A to B will not equal the % positives of B to A An " invention vanant polynucleotide" or " invention vanant nucleic acid sequence" means a nucleic acid molecule which encodes an active polypeptide of the invention as defined herein and which has at least about 80% nucleic acid sequence identity with the nucleotide acid sequence of DNA35638. DNA33094. DNA40628. DNA37150, DNA41388, DNA40981, or DNA37140 or a specifically deπved fragment thereof Ordinaπly, an invention vaπant polynucleotide will have at least about 80% nucleic acid sequence identity, more preferably at least about 81% nucleic acid sequence identity, more preferably at least about 82% nucleic acid sequence identity, more preferably at least about 83% nucleic acid sequence identity, more preferably at least about 84% nucleic acid sequence identity, more preferably at least about 85% nucleic acid sequence identity, more preferably at least about 86% nucleic acid sequence identity, more preferably at least about 87% nucleic acid sequence identity, more preferably at least about 88% nucleic acid sequence identity, more preferably at least about 89% nucleic acid sequence identity, more preferably at least about 90% nucleic acid sequence identity, more preferably at least about 91% nucleic acid sequence identity, more preferably at least about 92% nucleic acid sequence identity, more preferably at least about 93% nucleic acid sequence identity, more preferably at least about 94% nucleic acid sequence identity, more preferably at least about 95% nucleic acid sequence identity, more preferably at least about 96% nucleic acid sequence identity, more preferably at least about 97% nucleic acid sequence identity, more preferably at least about 98% nucleic acid sequence identity and yet more preferably at least about 99% nucleic acid sequence identity with the nucleic acid sequence of DNA35638, DNA33094, DNA40628, DNA37150, DNA41388, DNA40981 , or DNA37140 or a denved fragment thereof Vaπants do not encompass the native nucleotide sequence In this regard, due to the degeneracy of the genetic code, one of ordinary skill m the art will immediately recognize that a large number of invention vaπant polynucleotides having at least about 80% nucleic acid sequence identity to nucleotides DNA35638, DNA33094. DNA40628, DNA37150, DNA41388, DNA40981. or DNA37140 will encode a polypeptide having an amino acid sequence which is identical to the ammo acid sequence of a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 or the amino acid sequence encoded by the clones deposited with the ATCC descπbed below
Ordinarily, invention vaπant polynucleotides are at least about 30 nucleotides m length, often at least about 60 nucleotides in length, more often at least about 90 nucleotides in length, more often at least about 120 nucleotides in length, more often at least about 150 nucleotides m length, more often at least about 180 nucleotides in length, more often at least about 210 nucleotides m length, more often at least about 240 nucleotides in length, more often at least about 270 nucleotides m length, more often at least about 300 nucleotides m length, more often at least about 450 nucleotides in length, more often at least about 600 nucleotides m length, more often at least about 900 nucleotides in length, or more "Percent (%) nucleic acid sequence identity" with respect to the invention polypeptide- encoding nucleic acid sequences identified herein is defined as the percentage of nucleotides in a candidate sequence that are identical with the nucleotides in an invention polypeptide-encoding sequence of interest, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity. Alignment for purposes of determining percent nucleic acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full- length of the sequences being compared. For purposes herein, however, % nucleic acid sequence identity values are obtained as described below by using the sequence comparison computer program ALIGN-2, wherein the complete source code for the ALIGN-2 program is provided in Figures 2A-P. The ALIGN-2 sequence comparison computer program was authored by Genentech, Inc. and the source code shown in Figures 2A-P has been filed with user documentation in the U.S. Copyright Office, Washington D.C, 20559, where it is registered under U.S. Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available through Genentech, Inc., South San Francisco, California or may be compiled from the source code provided in Figures 2A-P. The ALIGN-2 program should be compiled for use on a UNIX operating system, preferably digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary.
For purposes herein, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or comprises a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows:
100 times the fraction W/Z
where W is the number of nucleotides scored as identical matches by the sequence alignment program
ALIGN-2 in that program's alignment of C and D, and where Z is the total number of nucleotides in
D. It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C. As examples of % nucleic acid sequence identity calculations.
Figures 1C-D demonstrate how to calculate the % nucleic acid sequence identity of the nucleic acid sequence designated "Comparison DNA" to the nucleic acid sequence designated "PRO-DNA".
Unless specifically stated otherwise, all % nucleic acid sequence identity values used herein are obtained as described above using the ALIGN-2 sequence comparison computer program.
However, % nucleic acid sequence identity may also be determined using the sequence comparison program NCBI-BLAST2 (Altschul et al , Nucleic Acids Res 25 3389-3402 (1997)) The NCBI- BLAST2 sequence compaπson program may be downloaded from http //www ncbr nlm nih gov NCBI-BLAST2 uses several search parameters, wherein all of those search parameters are set to default values including, for example, unmask = yes, strand = all, expected occurrences = 10, minimum low complexity length = 15/5, multi-pass e-value = 0 01, constant for multi-pass = 25, dropoff for final gapped alignment = 25 and scoπng matπx = BLOSUM62
In situations where NCBI-BLAST2 is employed for sequence compansons, the % nucleic acid sequence identity of a given nucleic acid sequence C to, with, or against a given nucleic acid sequence D (which can alternatively be phrased as a given nucleic acid sequence C that has or compπses a certain % nucleic acid sequence identity to, with, or against a given nucleic acid sequence D) is calculated as follows
100 times the fraction W/Z
where W is the number of nucleotides scored as identical matches by the sequence alignment program NCBI-BLAST2 in that program's alignment of C and D, and where Z is the total number of nucleotides m D It will be appreciated that where the length of nucleic acid sequence C is not equal to the length of nucleic acid sequence D, the % nucleic acid sequence identity of C to D will not equal the % nucleic acid sequence identity of D to C
In other embodiments, invention vaπant polynucleotides are nucleic acid molecules that encode an active polypeptide of the invention and which are capable of hybπdizmg, preferably under stnngent hybπdization and wash conditions, to nucleotide sequences encoding the full-length invention polypeptide Invention vaπant polypeptides include those that are encoded by an invention vaπant polynucleotide
An "isolated" nucleic acid molecule encoding a polypeptide of the invention is a nucleic acid molecule that is identified and separated from at least one contaminant nucleic acid molecule with which it is ordinanly associated in the natural source of the polypeptide-encodmg nucleic acid An isolated polypeptide-encoding nucleic acid molecule is other than in the form or setting in which it is found in nature Isolated nucleic acid molecules therefore are distinguished from the polypeptide- encodmg nucleic acid molecule as it exists in natural cells However, an isolated nucleic acid molecule encoding a polypeptide of the invention includes polypeptide-encodmg nucleic acid molecules contained m cells that ordinarily express a polypeptide of the invention where, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells
The term "control sequences" refers to DNA sequences necessary for the expression ot an operably linked coding sequence m a particular host organism The control sequences that are suitable for prokaryotes, for example, include a promoter, optionally an operator sequence and a πbosome binding site Eukaryotic cells are known to utilize promoters, polyadenylation signals, and enhancers.
Nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence For example, DNA for a presequence or secretory leader is operably linked to DNA for a polypeptide if it is expressed as a preprotem that participates m the secretion of the polypeptide, a promoter or enhancer is operably linked to a coding sequence if it affects the transcπption of the sequence, or a πbosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation Generally, "operably linked" means that the DNA sequences being linked are contiguous, and, in the case of a secretory leader, contiguous and in reading phase. However, enhancers do not have to be contiguous. Linking is accomplished by ligation at convenient restπction sites If such sites do not exist, the synthetic oligonucleotide adaptors or linkers are used in accordance with conventional practice
"Stnngency" of hybπdization reactions is readily determmable by one of ordinary skill in the art. and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybndization generally depends on the abrhty of denatured DNA to reanneal when complementary strands are present rn an environment below their melting temperature. The higher the degree of desired homology between the probe and hybndizable sequence, the higher the relative temperature which can be used. As a result, it follows that higher relatrve temperatures would tend to make the reaction conditions more stnngent, while lower temperatures less so. For additional details and explanation of stnngency of hybπdization reactions, see Ausubel et al , Cmrent Protocols in Molecular Biology, Wiley Interscience Publishers, (1995)
"Stnngent conditions" or "high stnngency conditions", as defined herein, may be identified by those that. (1) employ low ionic strength and high temperature for washing, for example 0 015 M sodium chloπde/0.0015 M sodium cιtrate/0 1% sodium dodecyl sulfate at 50C, (2) employ duπng hybridization a denatuπng agent, such as formamide, for example, 50% (v/v) formamide with 0 1% bovine serum albumιn 0.1% Fιcoll/0 1% polyvιnylpyrrolιdone/50mM sodium phosphate buffer at pH 6.5 wrth 750 mM sodium chlonde, 75 mM sodium citrate at 42C; or (3) employ 50% formamide, 5 x SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6 8), 0.1% sodium pyrophosphate, 5 x Denhardt's solution, sonicated salmon sperm DNA (50 ug/ml), 0.1% SDS, and 10% dextran sulfate at 42C, with washes at 42C in 0 2 x SSC (sodium chloπde/sodium citrate) and 50% formamide at 55C, followed by a high-stnngency wash consisting of 0 1 x SSC containing EDTA at 55C.
"Moderately stnngent conditions" may be identified as descπbed by Sambrook et al , Molecular Cloning. A Laboratory Manual, New York Cold Spnng Harbor Press, 1989, and include the use of washing solution and hybridization conditions (e g , temperature, ionic strength and %SDS) less stnngent that those described above An example of moderately stringent conditions is overnight incubation at 37C m a solution compnsmg 20% formamide, 5 x SSC (150 mM NaCl, 15 mM tnsodium citrate), 50 mM sodium phosphate (pH 7 6), 5 x Denhardt's solution, 10% dextran sulfate, and 20 mg/mL denatured sheared salmon sperm DNA. followed by washing the filters in 1 x SSC at about 37-50C The skilled artrsan wrll recognize how to adjust the temperature, ionic strength, etc as necessary to accommodate factors such as probe length and the like
The term "epitope tagged" when used herein refers to a chimeπc polypeptide compnsmg a polypeptide of the invention fused to a "tag polypeptide" The tag polypeptide has enough residues to provide an epitope against which an antibody can be made, yet is short enough such that it does not interfere with activity of the polypeptide to which it is fused The tag polypeptide preferably also is fairly unique so that the antibody does not substantially cross-react with other epitopes Suitable tag polypeptides generally have at least six amino acid residues and usually between about 8 and 50 ammo acid residues (preferably, between about 10 and 20 ammo acid residues)
"Active" or "activity" m the context of vaπants of the nolypeptide of the invention refers to form(s) of proteins of the invention which retain the biologic and or mimunologic activities of a native or naturally-occurπng polypeptide of the invention
"Biological activity" in the context of an antibody or another molecule that can be identified by the screening assays disclosed herein (e g an organic or inorganic small molecule, peptide, etc ) is used to refer to the abrlrty of such molecules to duce or rnhrbrt infiltration of inflammatory cells into a tissue, to stimulate or inhibit T-cell proliferation and to stimulate or inhibit lymphokine release by cells Another preferred activity is increased vascular permeability or the inhibition thereof
The term "antagonist" is used in the broadest sense, and includes any molecule that partrally or fully blocks, inhibits, or neutralizes a biological activity of a native polypeptide of the invention disclosed herein In a similar manner, the term "agonrst" is used rn the broadest sense and includes any molecule that mimics a biological activity of a native polypeptide of the invention disclosed herein Suitable agonist or antagonist molecules specifically include agonist or antagonist antibodies or antibody fragments, fragments or ammo acid sequence vaπants of native polypeptides of the invention , peptides, small organic molecules, etc
A "small molecule" is defined herein to have a molecular weight below about 600 daltons
"Antibodies" (Abs) and "immunoglobulins" (Igs) are glycoprotems having the same structural characteπstics While antibodies exhibit binding specificity to a specific antigen, immunoglobulins include both antibodies and other antibody-like molecules whrch lack antigen specificity
Polypeptides of the latter kind are. for example, produced at low levels by the lymph system and at increased levels by myelomas The term "antibody" is used m the broadest sense and specifically covers, without limitation, intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e g bispecific antibodres) formed from at least two tact antibodies, and antibody fragments so long as they exhibit the desired biological activity. An anti- PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 antibody is an antibody which immunologically binds to a PR0245, PR0217, PRO301 , PR0266, PR0335. PR0331 or PR0326 polypeptide. The antibody may bind to any domain of the polypeptide of the invention which may be contacted by the antibody. For example, the antibody may bind to any extracellular domain of the polypeptide and when the entire polypeptide is secreted, to any domain on the polypepetide which is available to the antibody for binding.
"Native antibodies" and "native immunoglobulins" are usually heterotetrameric glycoproteins of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light- chain variable domain is aligned with the variable domain of the heavy chain. Particular amino acid residues are believed to form an interface between the light- and heavy-chain variable domains.
The term "variable" refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called complementarity-determining regions (CDRs) or hypervariable regions both in the light-chain and the heavy-chain variable domains. The more highly conserved portions of variable domains are called the framework (FR). The variable domains of native heavy and light chains each comprise four FR regions, largely adopting a beta-sheet configuration, connected by three CDRs, which form loops connecting, and in some cases forming part of, the beta-sheet structure. The CDRs in each chain are held together in close proximity by the FR regions and, with the CDRs from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al, NIH Publ. No.91 -3242, Vol. I, pages 647-669 (1991)). The constant domains are not involved directly in binding an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity.
The term "hypervariable region" or "complementarity-determining regions" (CDRs) as used herein define a subregion within the variable region of extreme sequence variability of the antibody, which form the antigen-binding site and are the main determinants of antigen specificity. According to one definition, they can be, for example, residues (Kabat nomenclature) 24-34 (LI), 50-56 (L2) and
89-97 (L3) in the light chain variable region and residues (Kabat nomenclature) 31-35 (HI), 50-65 (H2), 95-102 (H3) in the heavy chain variable region. Kabat et al, Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institute of Health, Bethesda, MD. [1991]. Alternatively, or in combination with the region defined by Kabat, the hypervariable region can be the "hypervariable loop", comprising, for example, residues (Chothia nomenclature) 26-32 (LI), 50-53 (L2), 91-96 (L3) in the light chain variable region and residue (Chothia nomenclature) 26- 32 (HI), 53-55 (L2) and 96-101 (L3); Chothia and Lesk, J. Mol. Biol. 196: 901-917 [1987]. "Framework" or "FR" residues are those variable domain residues of relatively low sequence variability which lie in between the CDR regions.
"Antibody fragments" comprise a portion of an intact antibody, preferably the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (Zapata et al, Protein Eng. 8(10): 1057-1062 [1995]); single-chain antibody molecules; and multispecifc antibodies formed from antibody fragments.
Papain digestion of antibodies produces two identical antigen- binding fragments, called "Fab" fragments, each with a single antigen-binding site, and a residual "Fc" fragment, whose PR0245, PR0217, PRO301, Pro266, pro335, pro331 or pro326 reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-combining sites and is still capable of cross-linking antigen.
"Fv" is the minimum antibody fragment which contains a complete antigen-recognition and - binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VJJ-VL dimer. Collectively, the six
CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
The Fab fragment also contains the constant domain of the light chain and the first constant domain (CHI) of the heavy chain. Fab' fragments differ from Fab fragments by the addition of a few residues at the carboxy terminus of the heavy chain CHI domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
The "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (K) and lambda (λ), based on the amino acid sequences of their constant domains.
Depending on the amino acid sequence of the constant domain of their heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called called , δ, ε, γ, and μ, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. In addition to their specificity, the monoclonal antibodies are advantageous in that they are synthesized by the hybridoma culture, uncontaminated by other immunoglobulins. The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present invention may be made by the hybridoma method first described by Kohler et al., Nature, 256:495 [1975], or may be made by recombinant DNA methods (see, e.g., U.S. Patent No. 4,816,567). The "monoclonal antibodies" may also be isolated from phage antibody libraries using the techniques described in Clackson et al., Nature. 352:624-628 [1991] and Marks et al, J. Mol. Biol.. 222:581-597 (1991), for example. See also U.S Patent Nos. 5,750,373, 5,571,698, 5,403,484 and 5,223,409 which describe the preparation of antibodies using phagemid and phage vectors.
The monoclonal antibodies herein specifically include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (U.S. Patent No. 4,816,567; Morrison et al, Proc. Natl. Acad. Sci. USA. 81:6851- 6855 [1984]).
"Humanized" forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementanty-determinmg region (CDR) of the recipient are replaced by residues from a CDR of a non-human specres (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity, and capacity. In some mstances, Fv framework regron (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues Furthermore, humamzed antibodies may compnse residues which are found neither m the recipient antibody nor in the imported CDR or framework sequences. These modrfications are made to further refine and maxrmize antibody performance. In general, the humanized antibody will compnse substantially all of at least one, and typically two, vanable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence. The humanized antibody optimally also will compnse at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al , Nature, 321 :522-525 (1986); Reichmarm et al , Nature, 332.323-329 [1988], and Presta, Curr. Op Struct. Biol.. 2:593-596 (1992) The term "humanized antibody" includes a "pnmatized" antibody where the antigen-bmding region of the antibody is deπved from an antibody produced by immunizing macaque monkeys with the antigen of interest. Antibodies containing residues from Old World monkeys are also possible within the invention. See, for example, U.S. Patent Nos. 5,658,570; 5,693,780; 5,681,722; 5,750,105; and 5,756,096.
Antibodies and fragments thereof in this mvention also include "affinity matured" antibodies in which an antibody is altered to change the ammo acid sequence of one or more of the CDR regions and/or the framework regions to alter the affinity of the antibody or fragment thereof for the antigen to which it binds. Affinity maturation may result in an increase or in a decrease in the affinity of the matured antibody for the antrgen relative to the starting antibody. Typically, the starting antibody will be a humamzed, human, chimenc or murine antibody and the affinity matured antibody will have a higher affinity than the starting antibody Dunng the maturation process, one or more of the amino acid residues in the CDRs or in the framework regrons are changed to a different resrdue using any standard method. Suitable methods include point mutations using well known cassette mutagenesis methods (Wells et al., 1985, Gene, 34 315) or oligonucleotide mediated mutagenesis methods (Zoller et al., 1987, Nucleic Acids Res., 10 6487-6504). Affinity maturation may also be performed using known selection methods in which many mutations are produced and mutants having the desired affinity are selected from a pool or library of mutants based on improved affinity for the antigen or ligand Known phage display techniques can be conveniently used m this approach See, for example, U.S. 5,750,373, U.S. 5,223.409, etc.
Human antibodies are also with in the scope of the antibodies of the invention. Human antibodies can be produced using \ anous techniques known in the art. including phage display hbranes [Hoogenboom and Wrnter, J. Mol. Biol.. 227.381 (1991), Marks et al., J Mol. Biol.. 222:581 (1991)]. The techniques of Cole et al. and Boerner et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985); Boerner et al., J Immunol . 147(0.86-95 (1991), U. S. 5,750, 373] Similarly, human antibodies can be made by introducing of human immunoglobulin loci mto transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, whrch closely resembles that seen rn humans rn all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is descπbed, for example, in U.S. Patent Nos. 5,545.807; 5,545,806; 5,569,825; 5,625,126, 5,633,425; 5,661,016, and in the followmg screntrfrc publications. Marks et al, Bio/Technology 10, 779-783 (1992), Lonberg et al . Nature 368 856-859 (1994), Morπson, Nature 368. 812-13 (1994); Frshwrld et al , Nature Brotechnologv 14, 845-51 (1996); Neuberger, Nature Brotechnology \A, 826 (1996); Lonberg and Huszar, Intern Rev. Immunol L3 65-93 (1995).
"Single-chain Fv" or "sFv" antibody fragments compnse the VH and VL domarns of antrbody, wherein these domams are present in a single polypeptide chain. Preferably, the Fv polypeptrde further compπses a polypeptide linker between the VJ and VL domarns which enables the sFv to form the desrred structure for antrgen binding. For a review of sFv see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer- Verlag, New York, pp. 269-315 (1994).
The term "diabodies" refers to small antrbody fragments with two antigen-bmding srtes, whrch fragments compnse a heavy-chain vanable domain (Vjj) connected to a light-chain vanable domain (VjJ in the same polypeptide chain (Vjj - VjJ. By using a linker that is too short to allow paiπng between the two domains on the same chain, the domains are forced to pair with the complementary domarns of another chain and create two antigen-bmding sites. Diabodies are descπbed more fully in, for example, EP 404,097; WO 93/11161; and Holhnger et al , Proc Natl. Acad. Sci. USA. 90:6444-6448 (1993).
An "isolated" antibody is one which has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are matenals which would interfere with diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or nonprotemaceous solutes. In preferred embodrments, the compound of the invention will be puπfied (1) to greater than 95% by weight of the compound as determined by the Lowry method, and most preferably more than 99% by weight, (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under reducing or nonreducrng condrtions using Coomassie blue or, preferably, silver stain. Isolated compound, e.g. antibody or polypeptide. includes the compound in situ within recombinant cells since at least one component of the compound's natural environment will not be present Ordinarily, however, isolated compound will be prepared by at least one purification step
The word "label" when used herein refers to a detectable compound or composition which is conjugated drrectly or indirectly to the compound, e g antibody or polypeptide, so as to generate a "labelled" compound The label may be detectable by itself (e g radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition which is detectable
By "solid phase" is meant a non-aqueous matnx to which the compound of the present invention can adhere Examples of solid phases encompassed herein include those formed partially or entirely of glass (e g , controlled pore glass), polysacchaπdes (e g , agarose), polyacrylamrdes, polystyrene, polyvinyl alcohol and sihcones In certarn embodiments, depending on the context, the solid phase can comprise the well of an assay plate, in others it is a punfication column (e g , an affinity chromatography column) Thrs term also mcludes a drscontmuous solrd phase of discrete partrcles, such as those descnbed m U S Patent No 4,275,149
A "hposome" is a small vesicle composed of vanous types of hpids, phosphohpids and/or surfactant which is useful for delivery of a drug (such as the antι-ErbB2 antibodies disclosed herein and, optionally, a chemotherapeutic agent) to a mammal The components of the hposome are commonly arranged rn a brlayer formatron, similar to the lipid arrangement of biological membranes
As used herein, the term "lmmunoadhesm" designates antibody-like molecules which combine the binding specificity of a heterologous protem (an "adhesin") with the effector functrons of immunoglobulin constant domains Structurally, the immunoadhesins compnse a fusron of an ammo acid sequence with the desired binding specificity which is other than the antigen recognition and binding site of an antibody (1 e , is "heterologous"), and an immunoglobulin constant domain sequence The adhesm part of an rmmunoadhesm molecule typically is a contiguous ammo acid sequence comprising at least the binding site of a receptor or a ligand The immunoglobulin constant domain sequence in the lmmunoadhesm may be obtained from any rmmunoglobuhn, such as IgG- 1 , IgG-2, IgG-3, or IgG-4 subtypes, IgA (including IgA-1 and IgA-2), IgE, IgD or IgM
II Compositions and Methods of the Invention
1 Preparation of the polypeptrdes of the invention
The present invention provides newly identified and isolated nucleotide sequences encoding polypeptides refeπed to rn the present applrcation as PR0245, PR0217. PRO301, PR0266 PR0335, PR0331 or PR0326 (UNQ219, UNQ191, UNQ264, UNQ233, UNQ287V, UNQ292 or UNQ287 respectively) In particular, cDNA encoding a PR0245, PR0217. PRO301 , PR0266, PR0335,
PR03 1 oi PR0326 polypeptide has been identified and isolated, as disclosed m further detail in the Examples below It is noted that proteins produced in separate expression rounds may be given different PRO numbers but the UNQ number is unique for any given DNA and the encoded protein, and will not be changed However, for sake of simplicity, in the present specification the protein encoded by DNA35638, DNA33094. DNA40628, DNA37150, DNA41388, DNA40981 and DN A37140 as well as all further native homologues and variants included in the foregoing definition of PR0245. PR0217. PRO301. PR0266, PR0335, PR0331 or PR0326, will be referred to as PR0245, PR0217, PRO301 , PR0266. PR0335, PR0331 or PR0326 or simply as "the polypeptide of the invention", regardless of their ongm or mode of preparation
The descπption below relates pnmaπly to production of the polypeptide of the invention by cultunng cells transformed or transfected with a vector containing nucleic acid which encodes of the polypeptide of the invention It is, of course, contemplated that alternative methods, which are well known in the art, may be employed to prepare of the polypeptrde of the mvention For instance, the polypeptide sequence, or portions thereof, may be produced by drrect peptrde synthesrs usmg solid- phase technrques [see, e g , Stewart et al , Solrd-Phase Peptide Synthesis. W H Freeman Co , San Francisco, CA (1969), Mernfield, J Am Chem Soc . 85 2149-2154 (1963)] In vitro protein synthesis may be performed usmg manual techniques or by automation Automated synthesis may be accomplished, for mstance, usmg an Applred Brosystems Peptide Synthesizer (Foster City, CA) usmg manufacturer's mstructrons Vanous portrons of the polypeptrde of the invention may be chemically synthesized separately and combined using chemical or enzymatic methods to produce the full-length polypeptide
In addition to the full-length native sequence polypeptides descπbed herein, it is contemplated that variants can be prepared Vaπants can be prepared by introducing appropriate nucleotide changes into the DNA, and/or by synthesis of the desired polypeptide Those skilled in the art will appreciate that amino acid changes may alter post-translational processes of the polypeptide of the mvention, such as changing the number or position of glycosylation sites or altenng the membrane anchoring characteπstics
Vanations in the native full-length sequence or in vanous domains of the polypeptide of the invention descnbed herem, can be made, for example, using any of the techniques and guidelines for conservatrve and non-conservatrve mutatrons set forth, for mstance, rn U S Patent No 5,364,934 Vaπatrons may be a substitution, deletion or insertion of one or more codons encoding the polypeptide that results in a change in the ammo acid sequence of the polypeptide as compared with the native sequence polypeptide sequence Optionally the variation is by substitution of at least one amino acid with any other amino acid in one or more of the domains of the polypeptide of the invention Guidance in determining which ammo acid residue may be inserted, substituted or deleted without adversely affecting the desired activrty may be found by compaπng the sequence of the polypeptide of the invention with that of homologous known protein molecules and minimizing the number of amino acid sequence changes made in regions of high homology Amino acid substitutions can be the result of replacing one amino acid with another amino acid having similar structural and/or chemrcal properties, such as the replacement of a leucme with a seπne, 1 e , conservatrve ammo acid replacements Insertions or deletions may optionally be in the range of about 1 to 5 ammo acids The vaπation allowed may be determined by systematically making insertions, deletions or substitutions of amino acids in the sequence and testing the resulting vaπants for actrvrty exhrbrted by the full- length or mature native sequence
Polypeptide fragments of the polypeptides of the invention are also withm the scope of the mventron Such fragments may be truncated at the N-termmus or C-terminus, or may lack internal residues, for example, when compared with a full length native protein Certain fragments lack ammo acid residues that are not essential for a desrred brologrcal actrvrty of the invention polypeptide
Polypeptide fragments may be prepared by any of a number of convent onal technrques Desrred peptide fragments may be chemrcally synthesized An alternative approach involves generating fragments by enzymatrc digestion, e g , by treating the protein with an enzyme known to cleave proteins at sites defined by particular ammo acid residues, or by digesting the DNA with suitable restnction enzymes and isolating the desired fragment Yet another surtable technrque involves isolating and amplifying a DNA fragment encodmg a desrred polypeptrde fragment, by polymerase chain reaction (PCR) Oligonucleotides that define the desired termrnr of the DNA fragment are employed at the 5' and 3' pπmers rn the PCR Preferably, polypeptrde fragments share at least one biological and/or immunological activity with the native invention polypeptide
In particular embodiments, conservative substitutions of interest are shown in Table 1 under the heading of prefeπed substrtutrons If such substitutions result m a change in biological activity, then more substantial changes, denominated exemplary substrtutrons rn Table 1 , or as further descπbed below in reference to amino acid classes, are introduced and the products screened
Table 1
Original Exemplary Preferred
Residue Substitutions Substitutions
Ala (A) val; leu; ile val
Arg (R) lys; gin; asn lys
Asn (N) gin; his; lys; arg gin
Asp (D) glu glu
Cys (C) ser ser
Gln (Q) asn asn
Glu (E) asp asp
Gly (G) pro; ala ala
His (H) asn; gin; lys; arg arg
He (I) leu; val; met; ala; phe; norleucine leu
Leu (L) norleucine; ile; val; met; ala; phe ile
Lys (K) arg; gin; asn arg
Met (M) leu; phe; ile leu
Phe (F) leu; val; ile; ala; tyr leu
Pro (P) ala ala
Ser (S) thr thr
Thr (T) ser ser
Trp (W) tyr; phe tyr
Tyr (Y) trp; phe; thr; ser phe
Val (V) ile; leu; met; phe; ala; norleucine leu
Substantial modifications in function or immunological identity of the invention polypeptide are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are divided into groups based on common side- chain properties:
(1) hydrophobic: norleucine, met, ala, val, leu, ile; (2) neutral hydrophilic cys, ser, thr;
(3) acidic: asp, glu;
(4) basic asn, gin, his, lys. arg,
(5) residues that influence chain onentation: gly, pro; and
(6) aromatic: tip, tyr, phe
Non-conservatrve substitutions will entail exchanging a member of one of these classes for another class Such substituted residues also may be introduced mto the conservatrve substitution sites or, more preferably, mto the remarnmg (non-conserved) srtes.
The vaπatrons can be made using methods known in the art such as olrgonucleotrde-medrated (srte-drrected) mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis [Carter et al., Nucl Acids Res.. 13.4331 (1986), Zoller et al , Nucl. Acids Res . JO'6487 (1987)], cassette mutagenesis [Wells et al., Gene. 34:315 (1985)], restnction selection mutagenesis [Wells et al., Philos Trans. R Soc. London SerA, 317:415 (1986)] or other known techniques can be performed on the cloned DNA to produce the vaπant DNA.
Scannmg amino acid analysis can also be employed to identify one or more ammo acrds along a contrguous sequence. Among the preferred scanning amino acids are relatively small, neutral amino acids Such amino acids include alanme, glycine, seπne, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta- carbon and is less likely to alter the main-chain conformatron of the vaπant [Cunnmgham and Wells, Science. 244- 1081-1085 (1989)]. Alamne is also typically preferred because rt rs the most common ammo acid Further, it is frequently found m both buπed and exposed positions [Creighton, The Proteins. (W H Freeman & Co , N.Y.); Chothia, J Mol Biol . 150: 1 (1976)]. If alanine substitution does not yield adequate amounts of vaπant, an isoteπc amino acid can be used.
Covalent modifications of polypeptides of the invention are included withm the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of an invention polypeptide with an organic deπvatizing agent that is capable of reacting with selected side chains or the N- or C- terminal residues of the polypeptide. Denvatization with bifunctional agents is useful, for mstance, for crosslmking the invention polypeptide to a water-insoluble support matnx or surface for use rn the method for punfymg anti-polypeptide antibodies, and vice-versa Commonly used crosslmking agents include, e.g., 1 , 1 -bιs(dιazoacetyl)-2-phenylethane, glutaraldehyde, N- hydroxysuccmimide esters, for example, esters with 4-azιdosalιcylιc acid, homobifunctional rmidoesters, including disuccimmidyl esters such as 3,3'-dιthιobιs(succιnιmιdylpropιonate), bifunctronal maleimides such as bιs-N-maleιmιdo-1.8-octane and agents such as methyl-3-[(p- azιdophenyl)dιthιo]propιoιmιdate. Other modifications include deamidation of glutaminyl and asparaginyl residues to the corresponding glutamyl and aspartyl residues, respectively, hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the -amino groups of lysine, arginine, and histidine side chains [T.E. Creighton, Proteins: Structure and Molecular Properties. W.H. Freeman & Co., San Francisco, pp. 79-86 (1983)], acetylation of the N-terminal amine, and amidation of any C-terminal carboxyl group.
Another type of covalent modification of the invention polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of the polypeptide. "Altering the native glycosylation pattern" is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence polypeptide (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence. In addition, the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.
Addition of glycosylation sites to the polypeptide may be accomplished by altering the amino acid sequence. The alteration may be made, for example, by the addition of, or substitution by, one or more serine or threonine residues to the native sequence polypeptide (for O-linked glycosylation sites). The amino acid sequence may optionally be altered through changes at the DNA level, particularly by mutating the DNA encoding the polypeptide at preselected bases such that codons are generated that will translate into the desired amino acids.
Another means of increasing the number of carbohydrate moieties on the polypeptide of the invention is by chemical or enzymatic coupling of glycosides to the polypeptide. Such methods are described in the art, e.g., in WO 87/05330 published 11 September 1987, and in Aplin and Wriston, CRC Crit. Rev. Biochem.. pp. 259-306 (1981).
Removal of carbohydrate moieties present on the polypeptide of the invention may be accomplished chemically or enzymatically or by mutational substitution of codons encoding for amino acid residues that serve as targets for glycosylation. Chemical deglycosylation techniques are known in the art and described, for instance, by Hakimuddin, et al., Arch. Biochem. Biophys., 259:52 (1987) and by Edge et al., Anal. Biochem., U_8:131 (1981). Enzymatic cleavage of carbohydrate moieties on polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al., Meth. EnzvmoL 138:350 (1987).
Another type of covalent modification comprises linking the invention polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337. The polypeptide of the present invention may also be modified in a way to form a chimenc molecule comprising the invention polypeptide fused to another, heterologous polypeptide or amino acid sequence.
In one embodiment, such a chimenc molecule compnses a fusion of the invention polypeptide with a tag polypeptide which provides an epitope to which an anti-tag antibody can selectively bind. The epitope tag is generally placed at the ammo- or carboxyl- terminus of the polypeptide of the invention. The presence of such epitope-tagged forms of the polypeptide of the invention can be detected using an antibody against the tag polypeptide Also, provision of the epitope tag enables the polypeptide of the invention to be readily punfied by affinity punfication using an anti-tag antibody or another type of affinity matnx that binds to the epitope tag. Vanous tag polypeptides and their respective antibodies are well known in the art. Examples mclude poly-histidine (poly-his) or poly- histidme-glycine (poly-his-gly) tags; the flu HA tag polypeptide and its antibody 12CA5 [Field et al., Mol Cell. Biol.. 8:2159-2165 (1988)]; the c-myc tag and the 8F9, 3C7, 6E10, G4, B7 and 9E10 antibodies thereto [Evan et al., Molecular and Cellular Biology. 5:3610-3616 (1985)], and the Herpes Simplex virus glycoprotem D (gD) tag and its antibody [Paborsky et al., Protein Engineenng. 3(6):547-553 (1990)]. Other tag polypeptides include the Flag-peptide [Hopp et al., BioTechnology. 6: 1204-1210 (1988)]; the KT3 eprtope peptrde [Martin et al, Scrence. 255: 192-194 (1992)]; an - tubulrn epitope peptide [Skmner et al., J. Biol. Chem.. 266: 15163-15166 (1991)]; and the T7 gene 10 protein peptide tag [Lutz-Freyermuth et al, Proc Natl. Acad. Ser. USA. 87:6393-6397 (1990)].
In an alternative embodiment, the chimenc molecule may compnse a fusron of the polypeptide of the invention with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimenc molecule (also referred to as an "immunoadhesin"). such a fusron could be to the Fc regron of an IgG molecule. The Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of an invention polypeptide in place of at least one vanable region withm an Ig molecule. In a particularly prefeπed embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CHI, CH2 and CH3 regions of an IgGl molecule. For the production of immunoglobulin fusrons see also US Patent No 5,428,130 issued June 27, 1995.
l. Isolation of DNA Encoding the Polypeptide of the Invention
DNA encoding the polypeptide of the invention may be obtained from a cDNA library prepared from trssue belreved to possess the polypeptide mRNA and to express it at a detectable level
Accordingly, human DNA can be conveniently obtained from a cDNA hbrary prepared from human tissue, such as descπbed in the Examples. The gene encoding the polypeptrde of the invention may also be obtained from a genomic library or by oligonucleotide synthesis
Libraπes can be screened with probes (such as antibodies to the polypeptide of the invention or oligonucleotides of at least about 20-80 bases) designed to identify the gene of interest or the protein encoded by it. Screening the cDNA or genomic library with the selected probe may be conducted using standard procedures, such as described in Sambrook et al., Molecular Cloning: A Laboratory Manual (New York: Cold Spring Harbor Laboratory Press, 1989). An alternative means to isolate the gene encoding the polypeptide of the invention is to use PCR methodology [Sambrook et al., supra: Dieffenbach et al., PCR Primer: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1995)].
The Examples below describe techniques for screening a cDNA library. The oligonucleotide sequences selected as probes should be of sufficient length and sufficiently unambiguous that false positives are minimized. The oligonucleotide is preferably labeled such that it can be detected upon hybridization to DNA in the library being screened. Methods of labeling are well known in the art, and include the use of radiolabels like 32P-labeled ATP, biotinylation or enzyme labeling. Hybridization conditions, including moderate stringency and high stringency, are provided in Sambrook et al., supra.
Sequences identified in such library screening methods can be compared and aligned to other known sequences deposited and available in public databases such as GenBank or other private sequence databases. Sequence identity (at either the amino acid or nucleotide level) within defined regions of the molecule or across the full-length sequence can be determined through sequence alignment using computer software programs such as ALIGN, DNAstar, and INHERIT which employ various algorithms to measure homology.
Nucleic acid having protein coding sequence may be obtained by screening selected cDNA or genomic libraries using the deduced amino acid sequence disclosed herein for the first time, and, if necessary, using conventional primer extension procedures as described in Sambrook et al., supra, to detect precursors and processing intermediates of mRNA that may not have been reverse-transcribed into cDNA.
ii. Selection and Transformation of Host Cells
Host cells are transfected or transformed with expression or cloning vectors described herein for production of the polypeptides of the invention and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences. The culture conditions, such as media, temperature, pH and the like, can be selected by the skilled artisan without undue experimentation. In general, principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: a Practical Approach, M. Butler, ed. (IRL Press, 1991) and Sambrook et al., supra. Methods of transfection are known to the ordinarily skilled artisan, for example, CaPθ4 and electroporation. Depending on the host cell used, transformation is performed using standard techniques appropriate to such cells. The calcium treatment employing calcium chloride, as described in Sambrook et al., supra, or electroporation is generally used for prokaryotes or other cells that contain substantial cell-wall barriers. Infection with Agrobacterium tumefaciens is used for transformation of certain plant cells, as described by Shaw et al., Gene, 23:315 (1983) and WO 89/05859 published 29 June 1989. For mammalian cells without such cell walls, the calcium phosphate precipitation method of Graham and van der Eb, Virology, 52:456-457 (1978) can be employed. General aspects of mammalian cell host system transformations have been described in U.S. Patent No. 4,399,216. Transformations into yeast are typically carried out according to the method of Van Solingen et al, J. Bact.. 130:946 (1977) and Hsiao et al., Proc. Natl. Acad. Sci. (USA). 76:3829 (1979). However, other methods for introducing DNA into cells, such as by nuclear microinjection, electroporation, bacterial protoplast fusion with intact cells, or polycations, e.g., polybrene, polyornithine, may also be used. For various techniques for transforming mammalian cells, see Keown et al., Methods in Enzymology, 185:527-537 (1990) and Mansour et al., Nature. 336:348-352 (1988).
Suitable host cells for cloning or expressing the DNA in the vectors herein include prokaryote, yeast, or higher eukaryote cells. Suitable prokaryotes include but are not limited to eubacteria, such as Gram-negative or Gram-positive organisms, for example, Enterobacteriaceae such as E. coli. Various E. coli strains are publicly available, such as E. coli K12 strain MM294 (ATCC 31,446); E. coli X1776 (ATCC 31,537); E. coli strain W3110 (ATCC 27,325) and K5 772 (ATCC 53,635).
In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for vectors encoding the polypeptides of the invention. Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism.
Suitable host cells for the expression of glycosylated polypeptides of the invention are derived from multicellular organisms. Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sf9, as well as plant cells. Examples of useful mammalian host cell lines include Chinese hamster ovary (CHO) and COS cells. More specific examples include monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol., 36:59 (1977)); Chinese hamster ovary cells/-DHFR (CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA, 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod.. 23:243-251 (1980)); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); and mouse mammary tumor (MMT 060562, ATCC CCL51). The selection of the appropriate host cell is deemed to be within the skill in the art. iii. Selection and Use of a Replicable Vector The nucleic acid (e.g., cDNA or genomic DNA) encoding the polypeptides of the invention may be inserted into a replicable vector for cloning (amplification of the DNA) or for expression. Various vectors are publicly available. The vector may, for example, be in the form of a plasmid, cosmid, viral particle, phagemid or phage. The appropriate nucleic acid sequence may be inserted into the vector by a variety of procedures. In general, DNA is inserted into an appropriate restriction endonuclease site(s) using techniques known in the art. Vector components generally include, but are not limited to, one or more of a signal sequence, an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence. Construction of suitable vectors containing one or more of these components employs standard ligation techniques which are known to the skilled artisan.
The polypeptide of the invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which may be a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide. In general, the signal sequence may be a component of the vector, or it may be a part of the DNA encoding the polypeptide of the invention that is inserted into the vector. The signal sequence may be a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin II leaders. For yeast secretion the signal sequence may be, e.g., the yeast invertase leader, alpha factor leader (including Saccharomyces and Kluyveromyces alpha-factor leaders, the latter described in U.S. Patent No. 5,010,182), or acid phosphatase leader, the C. albicans glucoamylase leader (EP 362,179 published 4 April 1990), or the signal described in WO 90/13646 published 15 November 1990. In mammalian cell expression, mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species, as well as viral secretory leaders.
Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram- negative bacteria, the 2u plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g. , ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the nucleic acid encoding the polypeptide of the invention, such as DHFR or thymidine kinase. An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described by Urlaub et al, Proc. Natl. Acad. Sci. USA, 77:4216 (1980). A suitable selection gene for use in yeast is the trjσl gene present in the yeast plasmid YRp7 [Stinchcomb et al., Nature. 282:39 (1979); Kingsman et al, Gene. 7: 141 (1979); Tschemper et al, Gene. JO: 157 (1980)]. The trp I gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEP4-1 [Jones, Genetics. 85:12 (1977)].
Expression and cloning vectors usually contain a promoter operably linked to the nucleic acid sequence encoding the polypeptide of the invention to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Promoters suitable for use with prokaryotic hosts include the beta-lactamase and lactose promoter systems [Chang et al., Nature. 275:615 (1978); Goeddel et al., Nature. 281:544 (1979)], alkaline phosphatase, a tryptophan (trp) promoter system [Goeddel, Nucleic Acids Res.. 8:4057 (1980); EP 36,776], and hybrid promoters such as the tac promoter [deBoer et al., Proc. Natl. Acad. Sci. USA, 80:21-25 (1983)]. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the polypeptide of the invention.
Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase [Hitzeman et al., J. Biol. Chem.. 255:2073 (1980)] or other glycolytic enzymes [Hess et al., J. Adv. Enzyme Reg.. 7: 149 (1968); Holland, Biochemistry. J7:4900 (1978)], such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.
Other yeast promoters, which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, glyceraldehyde-3-phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
Transcription of the polypeptide of the invention from vectors in mammalian host cells is controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus (UK 2,21 1,504 published 5 July 1989), adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems.
Transcription of a DNA encoding the polypeptide of the invention by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Many enhancer sequences are now known from mammalran genes (globm, elastase, albumin, alpha- fetoprotem, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus Examples include the SV40 enhancer on the late side of the replication ongm (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication ongm, and adenovirus enhancers The enhancer may be spliced mto the vector at a position 5' or 3' to the coding sequence of the polypeptide of the invention, but is preferably located at a site 5' from the promoter
Expression vectors used m eukaryotic host cells (yeast, fungr, msect, plant, anrmal, human, or nucleated cells from other multrcellular organisms) will also contain sequences necessary for the termination of transcnptron and for stabilizing the mRNA Such sequences are commonly available from the 5' and, occasronally 3', untranslated regrons of eukaryotic or viral DNAs or cDNAs These regions contain nucleotide segments transcnbed as polyadenylated fragments rn the untranslated portion of the mRNA encoding the polypeptide of the invention
Still other methods, vectors, and host cells suitable for adaptatron to the synthesis of the polypeptide of the invention in recombinant vertebrate cell culture are descπbed rn Gethmg et al., Nature. 293:620-625 (1981); Mantel et al., Nature. 281 :40-46 (1979); EP 1 17,060; and EP 1 17,058. in Detecting Gene Expression
Gene expression may be measured in a sample directly, for example, by conventronal Southern blotting, Northern blotting to quantitate the transcnption of mRNA [Thomas, Proc Natl Acad Sci USA, 77"5201-5205 (1980)], dot blotting (DNA analysis), or in situ hybπdization, using an appropπately labeled probe, based on the sequences provided herein. Alternatrvely, antrbodies may be employed that can recognize specific duplexes, including DNA duplexes. RNA duplexes, and DNA-RNA hybnd duplexes or DNA-protein duplexes The antibodies in turn may be labeled and the assay may be earned out where the duplex is bound to a surface, so that upon the formatron of duplex on the surface, the presence of antibody bound to the duplex can be detected
Gene expressron, alternatively, may be measured by immunological methods, such as immunohistochemical staining of cells or tissue sections and assay of cell culture or body fluids, to quantitate drrectly the expression of gene product Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal. and may be prepared in any mammal Conveniently, the antibodies may be prepared against a native sequence of the inventive polypeptide or against a synthetic peptide based on the DNA sequences provided herein or against exogenous sequence fused to DNA encoding the polypeptide of the invention and encoding a specific antibody epitope lv Punfication of Polypeptide Forms of the polypeptide of the invention may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g. Triton-X 100) or by enzymatic cleavage. Cells employed in expression of the polypeptide of the invention can be disrupted by various physical or chemical means, such as freeze- thaw cycling, sonication, mechanical disruption, or cell lysing agents.
It may be desired to purify the polypeptide of the invention from recombinant cell proteins or polypeptides. The following procedures are exemplary of suitable purification procedures: by fractionation on an ion-exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS- PAGE; ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and metal chelating columns to bind epitope-tagged forms of the polypeptide of the invention . Various methods of protein purification may be employed and such methods are known in the art and described for example in Deutscher, Methods in Enzvmology. 182 (1990); Scopes, Protein Purification: Principles and Practice, Springer- Verlag, New York (1982). The purification step(s) selected will depend, for example, on the nature of the production process used and the particular polypeptide of the invention produced. 2. Tissue Distribution
The location of tissues expressing the polypeptides of the invention can be identified by determining mRNA expression in various human tissues. The location of such genes provides information about which tissues are most likely to be affected by the stimulating and inhibiting activities of the polypeptides of the invention. The location of a gene in a specific tissue also provides sample tissue for the activity blocking assays discussed below.
As noted before, gene expression in various tissues may be measured by conventional Southern blotting, Northern blotting to quantitate the transcription of mRNA (Thomas, Proc. Natl. Acad. Sci. USA. 77:5201-5205 [1980]), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies may be employed that can recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes.
Gene expression in various tissues, alternatively, may be measured by immunological methods, such as immunohistochemical staining of tissue sections and assay of cell culture or body fluids, to quantitate directly the expression of gene product. Antibodies useful for immunohistochemical staining and/or assay of sample fluids may be either monoclonal or polyclonal, and may be prepared in any mammal. Conveniently, the antibodies may be prepared against a native sequence of a polypeptide of the invention or against a synthetic peptide based on the DNA sequences encoding the polypeptide of the invention or against an exogenous sequence fused to a DNA encoding a polypeptide of the invention and encoding a specific antibody epitope. General techniques for generating antibodies, and special protocols for Northern blotting and in situ hybridization are provided below.
3. Antibody Binding Studies
The activity of the polypeptides of the invention can be further verified by antibody binding studies, in which the ability of anti-PR0245. PR0217, PRO301, PR0266. PR0335, PR0331 or PR0326 antibodies to inhibit the effect of the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptides on tissue cells is tested. Exemplary antibodies include polyclonal, monoclonal, humanized, bispecific, and heteroconjugate antibodies, the preparation of which will be described hereinbelow.
Antibody binding studies may be carried out in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies: A Manual of Techniques, pp.147-158 (CRC Press, Inc., 1987).
Competitive binding assays rely on the ability of a labeled standard to compete with the test sample analyte for binding with a limited amount of antibody. The amount of target protein in the test sample is inversely proportional to the amount of standard that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bound, the antibodies preferably are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte which remain unbound.
Sandwich assays involve the use of two antibodies, each capable of binding to a different immunogenic portion, or epitope, of the protein to be detected. In a sandwich assay, the test sample analyte is bound by a first antibody which is immobilized on a solid support, and thereafter a second antibody binds to the analyte, thus forming an insoluble three-part complex. See, e.g., U.S. Pat No. 4,376,110. The second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.
For immunohistochemistry, the tissue sample may be fresh or frozen or may be embedded in paraffin and fixed with a preservative such as formalin, for example. 4. Cell-Based Assays
Cell-based assays and animal models for immune related diseases can be used to further understand the relationship between the genes and polypeptides identified herein and the development and pathogenesis of immune related disease.
In a different approach, cells of a cell type known to be involved in a particular immune related disease are transfected with the cDNAs described herein, and the ability of these cDNAs to stimulate or inhibit immune function is analyzed. Suitable cells can be transfected with the desired gene, and monitored for immune function activity. Such transfected cell lines can then be used to test the ability of poly- or monoclonal antibodies or antibody compositions to inhibit or stimulate rmmune functron, for example to modulate T-cell prohferatron or rnflammatory cell infiltration Cells transfected with the coding sequences of the genes identified herein can further be used to identify drug candidates for the treatment of immune related diseases.
In addition, pπmary cultures denved from transgenic animals (as descπbed below) can be used in the cell-based assays herein, although stable cell lines are preferred Technrques to deπve continuous cell lines from transgenrc anrmals are well known in the art (see, e.g. Small et al. Mol. Cell. Biol 5. 642-648 [1985]).
One suitable cell based assay is the mixed lymphocyte reaction (MLR). Current Protocols in Immunology, unit 3.12; edited by J. E. Cohgan, A. M. Krursbeek, D. H. Marghes, E. M. Shevach, W. Strober, Natronal Insrtutes of Health, Published by John Wiley & Sons, Inc. In this assay, the ability of a test compound to stimulate the proliferation of activated T cells is assayed. A suspension of responder T cells is cultured with allogeneic stimulator cells and the prohferatron of T cells is measured by uptake of tπtiated thymidme. This assay is a general measure of T cell reactivity. Since the majonty of T cells respond to and produce IL-2 upon activation, differences n responsrveness rn thrs assay rn part reflect drfferences rn IL-2 productron by the responding cells. The MLR results can be veπfied by a standard lymphokine (IL-2) detection assay. Current Protocols in Immunology, above, 3.15, 6.3.
A prohferatrve T cell response rn an MLR assay may be due to drrect mitogemc propertres of an assayed molecule or to external antigen induced activation. Additional venficatron of the T cell stimulatory activity of the polypeptides of the invention can be obtained by a costimulation assay. T cell activation requires an antigen specific signal mediated through the T-cell receptor (TCR) and a costimulatory signal mediated through a second ligand binding interaction, for example, the B7(CD80, CD86)/CD28 binding interaction. CD28 crosslmking increases lymphokine secretion by activated T cells. T cell activation has both negative and positive controls through the binding of ligands which have a negative or positive effect CD28 and CTLA-4 are related glycoprote s in the Ig superfamrly which bind to B7. CD28 binding to B7 has a positive costimulation effect of T cell activation; conversely, CTLA-4 binding to B7 has a negative T cell deactivating effect. Chambers, C. A. and Allison, J P., Curr. Opin. Immunol. (1997) 9.396. Schwartz, R. H , Cell (1992) 71.1065; Linsey, P. S. and Ledbetter, J. A., Annu. Rev. Immunol. (1993) 1 1:191; June, C. H. et al., Immunol. Today (1994) 15:321; Jenkins, M K., Immunity (1994) L405. In a costimulation assay, the polypeptides of the invention are assayed for T cell costimulatory or rnhibitory activity
Polypeptrdes of the mventron, as well as other compounds of the mvention, which are stimulators (costimulators) of T cell prohferatron and agonists, e.g. agonist antibodies, thereto as determined by MLR and costimulation assays,for example, are useful in treating immune related diseases characterized by poor, suboptimal or inadequate immune function These diseases are treated by stimulatmg the proliferation and activation of T cells (and T cell mediated immunity) and enhancing the immune response in a mammal through administration of a stimulatory compound, such as the stimulating polypeptides of the invention The stimulatmg polypeptide may, for example, be a PR0245 PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide or an agonist antibody therefor
Direct use of a stimulating compound as in the invention has been validated in expenments with 4- IBB glycoprotem, a member of the tumor necrosis factor receptor family, which binds to a ligand (4-1BBL) expressed on pnmed T cells and signals T cell activation and growth Alderson, M E et al . J Immunol (1994) 24 2219
The use of an agonist stimulating compound has also been validated experimentally Activation of 4- IBB by treatment with an agomst antι-4-lBB antibody enhances eradication of tumors Hellstrom, I and Hellstrom, K E , Cnt Rev Immunol (1998) 18 1 Immunoadjuvant therapy for treatment of tumors, descnbed n more detail below, is another example of the use of the stimulating compounds of the invention
An immune stimulating or enhancing effect can also be achreved by antagomzrng or blocking the activity of a protein which has been found to be nhrbrtmg in the MLR assay Negatmg the inhibitory actrvrty of the compound produces a net strmulatory effect Suitable antagonists/blocking compounds are antibodies or fragments thereof whrch recognize and bind to the inhibitory protein, thereby blocking the effectrve mteractron of the protem with its receptor and inhibiting signaling through the receptor This effect has been validated in expenments usmg antι-CTLA-4 antibodies which enhance T cell proliferation, presumably by removal of the inhibitory signal caused by CTLA-4 binding Walunas, T L et al , Immunrty ( 1994) 1 405
On the other hand, polypeptides of the invention, as well as other compounds of the invention, which are direct inhibitors of T cell proliferation/activation and/or lymphokine secretion, can be directly used to suppress the immune response These compounds are useful to reduce the degree of the immune response and to treat immune related diseases charactenzed by a hyperactive, superoptrmal, or autormmune response Thrs use of the compounds of the mventron has been valrdated by the expenments descnbed above in which CTLA-4 binding to receptor B7 deactivates T cells The direct inhibitory compounds of the invention functron in an analogous manner
Alternatrvely, compounds, e g antibodies, which bind to stimulating polypeptides of the invention and block the stimulating effect of these molecules produce a net rnhrbrtory effect and can be used to suppress the T cell medrated rmmune response by rnhrbrtmg T cell prolrferatron/actrvation and or lymphokme secretion Blocking the stimulating effect of the polypeptides suppresses the immune response of the mammal This use has been validated in expenments usmg an antr-IL2 antrbody In these expenments, the antrbody bmds to IL2 and blocks brnding of IL2 to its receptor thereby achieving a T cell lnhibitorv effect 5. Animal Models
The results of the cell based in vitro assays can be further verified using in vivo animal models and assays for T-cell function. A variety of well known animal models can be used to further understand the role of the genes identified herein in the development and pathogenesis of immune related disease, and to test the efficacy of candidate therapeutic agents, including antibodies, and other antagonists of the native polypeptides, including small molecule antagonists. The in vivo nature of such models makes them predictive of responses in human patients. Animal models of immune related diseases include both non-recombinant and recombinant (transgenic) animals. Non- recombinant animal models include, for example, rodent, e.g., murine models. Such models can be generated by introducing cells into syngeneic mice using standard techniques, e.g. subcutaneous injection, tail vein injection, spleen implantation, intraperitoneal implantation, implantation under the renal capsule, etc.
Graft-versus-host disease occurs when immunocompetent cells are transplanted into immunosuppressed or tolerant patients. The donor cells recognize and respond to host antigens. The response can vary from life threatening severe inflammation to mild cases of diarrhea and weight loss. Graft-versus-host disease models provide a means of assessing T cell reactivity against MHC antigens and minor transplant antigens. A suitable procedure is described in detail in Current Protocols in Immunology, above, unit 4.3.
An animal model for skin allograft rejection is a means of testing the ability of T cells to mediate in vivo tissue destruction and a measure of their role in transplant rejection. The most common and accepted models use murine tail-skin grafts. Repeated experiments have shown that skin allograft rejection is mediated by T cells, helper T cells and killer-effector T cells, and not antibodies. Auchincloss, H. Jr. and Sachs, D. H., Fundamental Immunology, 2nd ed., W. E. Paul ed., Raven Press, NY, 1989, 889-992. A suitable procedure is described in detail in Current Protocols in Immunology, above, unit 4.4. Other transplant rejection models which can be used to test the compounds of the invention are the allogeneic heart transplant models described by Tanabe, M. et al., Transplantation (1994) 58:23 and Tinubu, S. A. et al., J. Immunol. (1994) 4330-4338.
Animal models for delayed type hypersensitivity provides an assay of cell mediated immune function as well. Delayed type hypersensitivity reactions are a T cell mediated in vivo immune response characterized by inflammation which does not reach a peak until after a period of time has elapsed after challenge with an antigen. These reactions also occur in tissue specific autoimmune diseases such as multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE, a model for MS). A suitable procedure is described in detail in Current Protocols in Immunology, above, unit 4.5.
EAE is a T cell mediated autoimmune disease characterized by T cell and mononuclear cell inflammation and subsequent demyelination of axons in the central nervous system. EAE is generally considered to be a relevant animal model for MS in humans Bolton. C , Multiple Sclerosis (1995) 1 143 Both acute and relapsmg-remittmg models have been developed The compounds of the invention can be tested for T cell stimulatory or inhibitory activity against immune mediated demyelinatmg disease usmg the protocol descπbed in Current Protocols in Immunology, above, units 15 1 and 15 2 See also the models for myelrn disease in which ohgodendrocytes or Schwann cells are grafted mto the central nervous system as descnbed in Duncan, I D et al, Molec Med Today (1997) 554-561
Contact hypersensitπ lty rs a srmple delayed type hypersensitivity in vivo assay of cell mediated immune function In this procedure, cutaneous exposure to exogenous haptens which gives nse to a delayed type hypersensitivity reaction which is measured and quantitated Contact sensitivity involves an initial sensitizing phase followed by an ehcitation phase The ehcrtatron phase occurs when the T lymphocytes encounter an antrgen to which they have had previous contact Swelling and inflammation occur, making this an excellent model of human allergic contact dermatrtis A surtable procedure is descnbed in detail in Current Protocols in Immunology, Eds J E Cologan, A M Krursbeek, D H Margulres, E M Shevach and W Strober, John Wiley & Sons, Ine , 1994, unit 4 2 See also Grabbe, S and Schwarz, T , Immun Today 19(1) 37-44 (1998)
An animal model for arthπtis is collagen-induced arthπtis Thrs model shares clinical, histological and immunological charactenstrcs of human autoimmune rheumatoid arthritis and is an acceptable model for human autormmune arthntrs Mouse and rat models are charactenzed by synovrtis, erosron of cartilage and subchondral bone The compounds of the invention can be tested for actrvity against autoimmune arthritis using the protocols descπbed in Current Protocols m Immunology, above, units 15 5 See also the model using a monoclonal antibody to CD 18 and VLA- 4 integnns descnbed rn Issekutz. A C et al . Immunology (1996) 88 569
A model of asthma has been descnbed in which antigen-induced airway hyper-reactivity, pulmonary eosmophiha and inflammation are induced bv sensitizing an animal with ovalbumm and then challenging the animal with the same protein delivered by aerosol Several animal models (guinea pig, rat, non-human pnmate) show symptoms similar to atopic asthma in humans upon challenge with aerosol antigens Munne models have many of the features of human asthma Suitable procedures to test the compounds of the mvention for actrvity and effectrveness n the treatment of asthma are descnbed by Wolynrec, W W et al , Am J Resprr Cell Mol Brol (1998) 18 777 and the references cited therein
Additionally, the compounds of the invention can be tested on animal models for psonasis like diseases Evidence suggests a T cell pathogenesis for psoriasis The compounds of the invention can be tested n the scrd/scrd mouse model descnbed Schon M P et al , Nat Med (1997) 3 183, in which the mice demonstrate histopathologic skm lesions resembling psonasis Another suitable model is the human skrn/scid mouse chrmera prepared as descnbed by Nickoloff. B J et al , Am J Path (1995) 146 580
Recombinant (transgenic) animal models can be engineered by introducing the coding portion of the genes identrfϊed herein mto the genome of animals of interest, using standard techniques for producing transgenic animals Animals that can serve as a target for transgenic manipulation include, without limitation, mice, rats, rabbits, guinea pigs, sheep, goats, pigs, and non-human pnmates, e g baboons chimpanzees and monkeys Techniques known m the art to introduce a transgene mto such animals include pronucleic mrcromjectron (Hoppe and Wanger, U S Patent No 4,873,191), retrovrrus-medrated gene transfer mto germ lrnes (e g , Van der Putten et al , Proc Natl Acad Sci USA 82, 6148-615 [1985]), gene targeting in embryonic stem cells (Thompson et al , Cell 56, 313- 321 [1989]), electroporation of embryos (Lo. Mol Cel Biol 3. 1803-1814 [1983]), sperm-mediated gene transfer (Lavitrano et al , Cell 57, 717-73 [ 1989]) For review, see, for example, U S Patent No 4,736,866
For the purpose of the present invention, transgenic animals include those that carry the transgene only rn part of therr cells ("mosaic anrmals") The transgene can be mtegrated either as a srngle transgene, or n concatamers, e g , head-to-head or head-to-tarl tandems Selectrve mtroduction of a transgene mto a particular cell type is also possible by followmg, for example, the technrque of Lasko et al , Proc Natl Acad Sci USA 89, 6232-636 (1992)
The expression of the transgene rn transgenic animals can be monitored by standard techniques For example, Southern blot analysis or PCR amplification can be used to venfy the mtegratron of the transgene The level of mRNA expressron can then be analyzed using techniques such as in situ hybndization, Northern blot analysrs, PCR, or rmmunocytochemrstry
The animals may be further examined for srgns of rmmune drsease pathology, tor example by histological exammation to determine infiltration of immune cells mto specific tissues Blocking expenments can also be performed in which the transgenic anrmals are treated with the compounds of the invention to determine the extent of the T cell proliferation strmulation or inhibition of the compounds In these expenments, blocking antibodies which bind to the polypeptide of the mventron, prepared as descπbed above, are admmrstered to the animal and the effect on immune functron is determmed
Alternatrvely, "knock out" animals can be constructed which have a defective or altered gene encoding a polypeptide identified herein, as a result of homologous recombination between the endogenous gene encoding the polypeptide and altered genomic DNA encoding the same polypeptide introduced into an embryonic cell of the animal For example, cDNA encoding a particular polypeptide can be used to clone genomic DNA encoding that polypeptide in accordance with established techniques A portion of the genomic DNA encoding a partrcular polypeptide can be deleted or replaced wrth another gene, such as a gene encoding a selectable marker which can be used to monitor integration Typically, several kilobases of unaltered flankrng DNA (both at the 5' and 3' ends) are included in the vector [see e.g., Thomas and Capecchi, Cell, 5J_:503 (1987) for a descnption of homologous recombination vectors] The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected [see e.g., Li et al , Cell, 69:915 (1992)]. The selected cells are then injected into a blastocyst of an animal (e g., a mouse or rat) to form aggregation chimeras [see e.g., Bradley, m Teratocarcinomas and Embryonic Stem Cells A Practical Approach, E. J. Robertson, ed (IRL, Oxford, 1987), pp 113-152], A chrmenc embryo can then be rmplanted mto a suitable pseudopregnant female foster ammal and the embryo brought to term to create a "knock out" ammal Progeny harboπng the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed ammals in which all cells of the ammal contarn the homologously recombmed DNA. Knockout animals can be characterized for mstance, for their ability to defend agamst certain pathological conditions and for therr development of pathologrcal condrtions due to absence of the polypeptide.
6 ImmunoAdiuvant Therapy
In one embodrment, the rmmunostrmulatmg compounds of the mventron can be used rn rmmunoadjuvant therapy for the treatment of tumors (cancer). It is now well established that T cells recognize human tumor specific antigens. One group of tumor antigens, encoded by the MAGE, BAGE and GAGE families of genes, are silent in all adult normal tissues , but are expressed in significant amounts m tumors, such as melanomas, lung tumors, head and neck tumors, and bladder carcinomas. DeSmet, C. et al, (1996) Proc. Natl. Acad. Sci. USA, 93:7149 It has been shown that costimulation of T cells induces tumor regression and an antitumor response both in vitro and in vivo. Melero, I. et al., Nature Medicine (1997) 3.682; Kwon, E. D. et al., Proc. Natl. Acad. Sci. USA (1997) 94:8099; Lynch, D. H. et al., Nature Medicine (1997) 3.625; Finn, O. J. and Lotze, M T., J. Immunol (1998) 21:114 The stimulatory compounds of the invention can be administered as adjuvants, alone or together with a growth regulating agent, cytotoxic agent or chemotherapeutic agent , to stimulate T cell prohferatron/actrvatron and an antrtumor response to tumor antrgens. The growth regulatmg, cytotoxrc, or chemotherapeutrc agent may be administered in conventional amounts using known administration regimes Immunostimulating activity by the compounds of the invention allows reduced amounts of the growth regulating, cytotoxic, or chemotherapeutic agents thereby potentially lowenng the toxicity to the patient
7. Screening Assays for Drug Candidates
Screening assays for drug candrdates are desrgned to rdentify compounds that bind or complex with the polypeptides encoded by the genes identified herein or a biologically active fragment thereof, or otherwrse interfere with the mteractron of the encoded polypeptrdes wrth other cellular proteins. Such screening assays will include assays amenable to high-throughput screening of chemical libraries, making them particularly suitable for identifying small molecule drug candrdates Small molecules contemplated mclude synthetrc organic or inorganic compounds, including peptides, preferably soluble peptides, (poly)peptιde-ιmmunoglobulm fusions, and. in particular, antrbodres including, without limitation, poly- and monoclonal antibodies and antibody fragments, smgle-cham antibodies, anti-idiotypic antibodies, and chimenc or humanized versions of such antibodies or fragments, as well as human antibodies and antibody fragments. The assays can be performed rn a varrety of formats, including protein-protein binding assays, biochemical screening assays, immunoassays and cell based assays, which are well characteπzed in the art.
All assays are common rn that they call for contacting the drug candidate with a polypeptide encoded by a nucleic acid identified herein under conditions and for a trme sufficient to allow these two components to interact.
In binding assays, the interaction is binding and the complex formed can be rsolated or detected rn the reaction mrxture. In a particular embodiment, the polypeptrde encoded by the gene identified herein or the drug candrdate rs immobilized on a solid phase, e.g. on a microtiter plate, by covalent or non-covalent attachments. Non-covalent attachment generally is accomplished by coating the solid surface wrth a solution of the polypeptide and drymg. Alternatively, an rmmobihzed antibody, e.g. a monoclonal antibody, specific for the polypeptrde to be rmmobrhzed can be used to anchor it to a solid surface. The assay rs performed by addmg the non-immobilized component, whrch may be labeled by a detectable label, to the rmmobrhzed component, e.g. the coated surface containing the anchored component. When the reaction is complete, the non-reacted components are removed, e.g. by washing, and complexes anchored on the solid surface are detected. When the ongmally non-immobrhzed component carries a detectable label, the detectron of label immobilized on the surface mdrcates that complexmg occurred. Where the ongmally non-immobilized component does not carry a label, complexmg can be detected, for example, by usmg a labelled antrbody specifically binding the immobilized complex
If the candidate compound interacts with but does not bind to a particular protem encoded by a gene rdentrfred herein, its interaction with that protein can be assayed by methods well known for detecting protem-protem interactions. Such assays include traditional approaches, such as, cross- lrnkrng, co-rmmunoprecrprtatron, and co-punfication through gradients or chromatographic columns
In additron, protem-protein interactions can be monitored by using a yeast-based genetic system descπbed by Fields and co-workers [Fields and Song, Nature (London) 340, 245-246 (1989); Chien et al , Proc Natl Acad Sci USA 88. 9578-9582 (1991)] as disclosed by Chevray and Nathans [Proc.
Natl. Acad. Ser. USA 89, 5789-5793 (1991)] Many transcπptronal activators, such as yeast GAL4, consrst of two physically discrete modular domains, one acting as the DNA-bmdmg domain, while the other one functioning as the transcnption activation domain The yeast expression system descnbed in the foregoing publications (generally referred to as the "two-hybnd system") takes advantage of this property, and employs two hybnd proteins, one m which the target protein is fused to the DNA- bindmg domain of GAL4, and another, in which candidate activating proteins are fused to the actrvation domain The expression of a GAL 1 -lacZ reporter gene under control of a G AL4-actιvated promoter depends on reconstitution of GAL4 activity via protem-protem rnteractron Colonres containing interacting polypeptides are detected with a chromogemc substrate for beta-galactosidase A complete kit (MATCHMAKER™) for identifyrng protem-protem interactions between two specific proteins using the two-hybnd technique is commercially available from Clontech This system can also be extended to map protein domains involved in specific protein interactions as well as to pinpoint amino acid residues that are crucral for these teractrons
In order to find compounds that mterfere wrth the rnteractron of a gene rdentified herein and other intra- or extracellular components can be tested, a reaction mixture is usually prepared containing the product of the gene and the intra- or extracellular component under conditions and for a time allowing for the rnteractron and binding of the two products To test the ability of a test compound to inhibit binding, the reaction is run rn the absence and in the presence of the test compound In addition, a placebo may be added to a third reaction mixture, to serve as posrtive control The binding (complex formation) between the test compound and the intra- or extracellular component present in the mixture is monitored as descπbed above The formation of a complex in the control reactιon(s) but not in the reaction mixture containing the test compound indicates that the test compound interferes with the interaction of the test compound and its reaction partner
8 Composrtions and Methods for the Treatment of Immune Related Diseases
The composrtrons useful m the treatment of immune related diseases include, without limitation, antibodies, small organic and inorganic molecules, peptides, phosphopeptides, antisense and nbozyme molecules, tπple helix molecules, etc that inhibit or stimulate immune function, for example, T cell proliferation/activation, lymphokine release, or immune cell infiltration
For example, antisense RNA and RNA molecule act to directly block the translation of mRNA by hybridizing to targeted mRNA and preventing protein translation When antisense DNA is used, ohgodeoxyπbonucleotides deπved from the translation initiation site, e g between about -10 and +10 positions of the target gene nucleotide sequence, are preferred
Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA Ribozymes act by sequence-specific hybndization to the complementary target RNA, followed by endonucleolytic cleavage Specific ribozyme cleavage sites within a potential RNA target can be identified by known techniques For further details see, e g Rossi, Current Brology 4, 469-471 (1994), and PCT publication No WO 97/33551 (published September 18, 1997)
Nucleic acid molecules in tnple helix formation used to inhibit transcnption should be single- stranded and composed of deoxynucleotides The base composition of these oligonucleotides is designed such that it promotes tπple helix formation via Hoogsteen base pairing rules, which generally require sizeable stretches of puπnes or pyπmidines on one strand of a duplex For further detarls see, e g PCT publication No WO 97/33551, supra
These molecules can be identified by any or any combination of the screening assays discussed above and/or by any other screening techniques well known for those skrlled in the art 9 Antibodies
Some of the most promrsmg drug candidates according to the present invention are antibodies and antibody fragments whrch may rnhrbit (antagonists) or strmulate (agonrsts) T cell prolrferation, eosmophil infiltration, etc l Polyclonal Antibodies
Methods of preparmg polyclonal antibodies are known to the skilled artisan Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or mtrapentoneal injections The immunizing agent may include the polypeptide of the invention or a fusion protein thereof It may be useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobuhn, and soybean trypsin inhibitor Examples of adjuvants which may be employed include Freund's complete adjuvant and MPL-TDM adjuvant (monophosphoryl Liprd A, synthetrc trehalose dicorynomycolate) The immunization protocol may be selected by one skilled the art without undue expenmentation ii Monoclonal Antibodies
Antibodies which recognize and bind to the polypeptides of the invention or which act as agonist therefor may, alternatively, be monoclonal antrbodres Monoclonal antibodies may be prepared using hybπdoma methods, such as those described by Kohler and Milstem, Nature. 256 495 ( 1975) In a hybndoma method, a mouse, hamster, or other appropπate host ammal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent Alternatively, the lymphocytes may be immunized in vitro
The immunizing agent will typically include the polypeptide of the invention or a fusion protein thereof Generally, either penpheral blood lymphocytes ("PBLs") are used if cells of human ongin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybndoma cell [Godmg, Monoclonal Antibodies Principles and Practice, Academic Press, (1986) pp 59-103] Immortalrzed cell lrnes are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human ongm Usually, rat or mouse myeloma cell lines are employed The hybridoma cells may be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused. immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanrne phosphonbosyl transferase (HGPRT or HPRT), the culture medrum for the hybπdomas typically will mclude hypoxanthine, ammopterm. and thymidme ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells
Preferred rmmortahzed cell lines are those that fuse efficrently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred rmmortahzed cell lrnes are munne myeloma lines, which can be obtained, for instance, from the Salk Instrtute Cell Distrrbutron Center. San Drego, California and the Ameπcan Type Culture Collection, Rockville, Maryland. Human myeloma and mouse-human heteromyeloma cell lrnes also have been descπbed for the productron of human monoclonal antrbodres [Kozbor, J Immunol.. 133.3001 (1984), Brodeur et al., Monoclonal Antibody Production Techniques and Applications, Marcel Dekker, Ine . New York, (1987) pp 51-63].
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antrbodies drrected against the polypeptrde of the mvention or having similar activity as the polypeptide of the invention. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determmed by lmmunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked lmmunoabsorbent assay (ELISA) Such techniques and assays are known in the art. The brndmg affinity of the monoclonal antibody can, for example, be determmed by the Scatchard analysis of Munson and Pollard, Anal. Biochem.. 107:220 (1980).
After the desrred hybndoma cells are rdentrfied, the clones may be subcloned by limiting dilution procedures and grown by standard methods [Godmg, supra] Suitable culture media for this purpose mclude, for example, Dulbecco's Modrfied Eagle's Medium and RPMI-1640 medium Alternatively, the hybπdoma cells may be grown in vivo as ascites in a mammal
The monoclonal antibodies secreted by the subclones may be isolated or punfied from the culture medium or ascites fluid by conventional immunoglobulin punfication procedures such as, for example, protein A-Sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography
The monoclonal antrbodres may also be made by recombinant DNA methods, such as those described in U.S. Patent No 4,816,567 DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains ot munne antibodies) The hybndoma cells of the invention serve as a prefeπed source of such DNA Once isolated, the DNA may be placed to expression vectors, which are then transfected mto host cells such as simian COS cells. Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwrse produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also may be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences [U.S. Patent No. 4,816,567; Morrison et al., supra] or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non- immunoglobulin polypeptide. Such a non- immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
The antibodies are preferably monovalent antibodies. Methods for preparing monovalent antibodies are well known in the art. For example, one method involves recombinant expression of immunoglobulin light chain and modified heavy chain. The heavy chain is truncated generally at any point in the Fc region so as to prevent heavy chain crosslinking. Alternatively, the relevant cysteine residues are substituted with another amino acid residue or are deleted so as to prevent crosslinking.
In vitro methods are also suitable for preparing monovalent antibodies. Digestion of antibodies to produce fragments thereof, particularly, Fab fragments, can be accomplished using routine techniques known in the art. iii. Human and Humanized Antibodies
The antibodies of the invention may further comprise humanized antibodies or human antibodies. Humanized forms of non-human (e.g., murine) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity. In some instances, Fv framework residues of the human immunoglobulin are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-329 (1988); and Presta, Curr. Op. Struct. Biol. 2:593-596 (1992)].
Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more ammo acid residues introduced into it from a source which is non-human. These non-human ammo acid residues are often referred to as "import" residues, which are typically taken from an "import" vanable domarn. Humamzation can be essentially performed followmg the method of Winter and coworkers [Jones et al., Nature, 321.522-525 ( 1986); Riechmann et al., Nature, 332.323-327 (1988); Verhoeyen et al, Science. 239: 1534-1536 (1988)], by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimenc antibodies (U.S. Patent No. 4,816,567), wherein substantially less than an intact human vanable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies m which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
Human antibodies can also be produced using various techniques known the art, including phage display libraries [Hoogenboom and Winter, J Mol. Biol.. 227.381 (1991); Marks et al., J. Mol. Brol, 222:581 (1991)]. The technrques of Cole et al. and Boerner et al. are also available for the preparation of human monoclonal antibodies (Cole et al., Monoclonal Antibodies and Cancer Therapy. Alan R. Liss, p. 77 (1985); Boerner et al, J. Immunol. 147(0:86-95 (1991); U. S. 5,750, 373]. Similarly, human antibodies can be made by introducing of human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, whrch closely resembles that seen rn humans in all respects, including gene rearrangement, assembly, and antrbody repertoire This approach is descnbed, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825, 5.625,126; 5,633,425; 5,661,016, and in the followmg screntrfic publications: Marks et al , Bio/Technology 10. 779-783 (1992), Lonberg et al . Nature 368 856-859 (1994); Mornson, Nature 368, 812-13 (1994); Fishwild et al . Nature Biotechnology 14. 845-51 (1996), Neuberger, Nature Biotechnology 14, 826 (1996), Lonberg and Huszar. Intern. Rev. Immunol. 13 65-93 (1995). lv. Brspecrfic Antibodies
Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two drfferent antrgens. In the present case, one of the brnding specificities may be for the polypeptide of the invention, the other one is for any other antigen, and preferably for a cell-surface protern or receptor or receptor subunrt.
Methods for making brspecrfic antrbodies are known in the art Tradrtronally, the recombinant production of bispecific antibodies is based on the coexpression of two immunoglobulin heavy-chain/hght-cham pairs, where the two heavy chains have different specificities (Milstem and
Cuello, Nature, 305:537-539 [1983]) Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of ten different antibody molecules, of which only one has the correct bispecific structure. The puπfication of the coπect molecule is usually accomplished by affinity chromatography steps Similar procedures are disclosed m WO 93/08829, published 13 May 1993. and in Traunecker et al , EMBO J , 10 3655-3659 (1991)
Antibody vanable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to rmmunoglobulm constant domain sequences The fusion preferably is with an immunoglobulin heavy-chain constant domain, compnsmg at least part of the hinge, CH2, and CH3 regions It is preferred to have the first heavy-chain constant region (CH 1 ) containing the site necessary for light-chain binding present in at least one of the fusions DNAs encoding the immunoglobulin heavy-chain fusrons and, rf desired, the immunoglobulin light chain, are inserted mto separate expressron vectors, and are cotransfected into a suitable host organism For further details of generating bispecific antibodies see, for example. Suresh et al , Methods in Enzvmology, 121 210 (1986) v Heterocomugate Antibodies
Heteroconjugate antibodres are composed of two covalently joined antibodies Such antibodies have, for example, been proposed to target immune system cells to unwanted cells [U S Patent No 4,676,980], and for treatment of HIV infection [WO 91/00360, WO 92/200373, EP 03089] It is contemplated that the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslmking agents For example, immunotoxins may be constructed using a disulfide exchange reaction or by forming a thioether bond Examples of suitable reagents for thrs purpose mclude iminothiolate and methyl-4- mercaptobutyπmidate and those disclosed, for example, n U S Patent No 4,676,980 vr Effector function engrneenng
It may be desrrable to modify the antibody of the mvention with respect to effector function, so as to enhance the effectiveness of the antibody in treating an immune related disease, for example For example cysteine resιdue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region The homodimenc antibody thus generated may have improved mternahzation capabrlrty and/or increased complement-mediated cell killing and antibody- dependent cellular cytotoxicity ( ADCC) See Caron et al , J Exp Med 176 1 191-1 195 (1992) and Shopes, B J Immunol 148 2918-2922 (1992) Homodimenc antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-lrnkers as descnbed rn Wolff et al Cancer Research 53 2560-2565 (1993) Alternatively, an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities See Stevenson et al , Anti-Cancer Drug Design 3 219-230 (1989) vii Immunocomugates
The invention also pertains to immunoconjugates compnsmg an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e g an enzymatically active toxin of bactenal, fungal, plant or animal ongm, or fragments thereof), or a radioactive isotope (/ e a radioconjugate)
Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above Enzymatically actrve toxrns and fragments thereof which can be used include diphthena A chain, nonbindmg active fragments of diphthena toxm, exotoxm A chain (from Pseudomonas aeruginosa), ncm A chain, abπn A chain, modeccm A chain, alpha-sarcm, Aleuntes fordn proteins, dianthm proteins. Phvtolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcm, crotin, sapaonaπa officinahs inhibitor, gelonm, mrtogellm, restnctocm, phenomycm, enomycm and the tncothecenes A variety of radionuchdes are available for the production of radioconjugated antibodies Examples include 212Bι, 131I, 131In, 90Y and 186Re
Conjugates of the antibody and cytotoxic agent are made using a vaπety of bifunctional protein coupling agents such as N-succmιmιdyl-3-(2-pyndyldrthιol) propionate (SPDP), lm othiolane (IT), bifunctional deπvatives of lmidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccmrmrdyl suberate). aldehvdes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamme), bis-diazomum deπvatives (such as bιs-(p- dιazomumbenzoyl)-ethylenedιamιne), dnsocyanates (such as tolyene 2,6-dnsocyanate), and bis-active fluorrne compounds (such as l,5-difluoro-2,4-dimtrobenzene) For example, a ncm immunotoxin can be prepared as descπbed in Vitetta et al , Science 238 1098 (1987) Carbon- 14-labeled 1- ιsothιocyanatobenzyl-3-methyldιethylene tπaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody See W094/11026
In another embodiment, the antibody may be conjugated to a "receptor" (such streptavidin) for utilization in tisue pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation usmg a clearing agent and then administration of a "ligand" (e g avidm) which is conjugated to a cytotoxic agent (e g a radionucleotide) viii Immunohposomes
The protems, antrbodres, etc disclosed herein may also be formulated as immunohposomes Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al . Proc Natl Acad Sci USA, 82 3688 (1985), Hwang et al Proc Natl Acad Sci USA. 77 4030 (1980), and U S Pat Nos 4,485,045 and 4 544.545 Liposomes with enhanced circulation time are disclosed in U S Patent No 5,013,556
Particularly useful hposomes can be generated by the reverse phase evaporation method with a lipid composrtion compnsmg phosphatidylcholme. cholesterol and PEG-denvatized phosphatidylethanolamme (PEG-PE) Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter Fab' fragments of the antibody of the present invention can be conjugated to the liposomes as described in Martin et al , J Biol Chem 257 286-288 (1982) via a disulfide interchange reaction A chemotherapeutic agent (such as doxorubicm) may be optionally contained within the hposome See Gabizon et al , J National Cancer Inst 81( 19) 1484 (1989) 10 Pharmaceutical Compositions
The active molecules of the invention, polypeptides and antibodies, as well as other molecules identified by the screening assays disclosed above, can be administered for the treatment of immune related diseases, in the form of pharmaceutical compositions
Therapeutic formulations of the active molecule, preferably a polypeptide or antrbody of the invention, are prepared for storage by mixing the active molecule having the desired degree of punty with optional pharmaceutically acceptable earners, excipients or stabilizers (Remington 's Pharmaceutical Sciences 16th edrtron, Osol, A Ed [1980]), in the form of lyophrhzed formulations or aqueous solutions Acceptable earners, excrprents, or stabrhzers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, crtrate, and other organrc acrds, antroxidants including ascorbic acid and methionine, preservatives (such as octadecyldimethylbenzyl ammonium chlonde, hexamethonium chloπde, benzalkonium chloπde, benzethomum chlonde, phenol, butyl or benzyl alcohol, alkyl parabens such as methyl or propyl paraben. catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol), low molecular weight (less than about 10 residues) polypeptides, proteins, such as serum albumin, gelatin, or immunoglobulins, hydrophilic polymers such as polyvmylpyrrolrdone, ammo acrds such as glycme, glutamine, asparagme, histrdme, arginine, or lysine, monosacchaπdes, disacchandes, and other carbohydrates including glucose, mannose, or dextπns, chelating agents such as EDTA, sugars such as sucrose, mannitol. trehalose or sorbitol, salt-formrng counter-ions such as sodium, metal complexes (e g , Zn- protein complexes), and/or non-iomc surfactants such as TWEEN™, PLURONICS™ or polyethylene glycol (PEG)
Compounds identified by the screening assays of the present invention can be formulated in an analogous manner, using standard techniques well known in the art
Lipofections or liposomes can also be used to deliver the polypeptide, antibody, or an antibody fragment, mto cells Where antibody fragments are used, the smallest inhibitory fragment which specifically binds to the binding domain of the target protein is preferred For example, based upon the vanable region sequences of an antibody, peptide molecules can be designed which retain the abrlity to bind the target protein sequence Such peptides can be synthesized chemically and/or produced by recombinant DNA technology (see, e g Marasco et al , Proc Natl Acad Sci USA 90,
7889-7893 [1993])
The formulation herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other Alternatively, or rn addition, the composition may compnse a cytotoxic agent, cytokine or growth inhibitory agent Such molecules are suitably present in combination in amounts that are effective for the purpose intended The active molecules may also be entrapped in microcapsules prepared, for example, by coacervation technrques or bv mterfacial polymenzation, for example, hydroxymethylcellulose or gelatm-mrcrocapsules and poly-(methylmethacylate) mrcrocapsules, respectrvely, rn colloidal drug delivery systems (for example, liposomes. albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions Such techniques are disclosed in Remingto 's Pharmaceutical Sciences 16th edition, Osol. A Ed (1980)
The formulations to be used for in vivo admrnrstration must be stenle Thrs is readily accomplished by filtration through stenle filtration membranes
Sustained-release preparations may be prepared Suitable examples of sustained-release preparations include semipermeable matnces of solrd hydrophobic polymers contarning the antibody, which matnces are in the form of shaped articles, e g films, or microcapsules Examples of sustained-release matnces include polyesters, hydrogels (for example, poly(2-hydroxyethyl- methacrylate), or poly(vmylalcohol)), polylactides (U S Pat No 3,773,919), copolymers of L- glutamrc acrd and ethyl-L-glutamate, non-degradable ethylene-vmyl acetate, degradable lactrc acid- glycohc acid copolymers such as the LUPRON DEPOT ™ (injectable microspheres composed of lactic acid-glycohc acid copolymer and leuprohde acetate), and poly-D-(-)-3-hydroxybutync acid While polymers such as ethylene-vmyl acetate and lactic acid-glycohc acid enable release of molecules for over 100 days, certarn hydrogels release proteins for shorter time peπods When encapsulated antibodies remain in the body for a long trme, they may denature or aggregate as a result of exposure to morsture at 37C, resulting in a loss of biological activity and possible changes in lmmunogenrcrty Rational strategies can be devised for stabilization depending on the mechamsm involved For example, if the aggregation mechanism is discovered to be mtermolecular S-S bond formation through thro-drsulfrde interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophihzing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matnx compositions 11 Methods of Treatment
It is contemplated that the polypeptides, antibodies and other active compounds of the present invention may be used to treat vanous immune related diseases and conditrons, such as T cell medrated drseases, including those charactenzed by infiltration of inflammatory cells mto a tissue, stimulation of T-cell prohferatron, inhibition of T-cell proliferation, mcreased or decreased vascular permeability or the inhibition thereof
Exemplary conditions or disorders to be treated with the polypeptides, antibodies and other compounds of the invention, include, but are not limited to systemic lupus erythematosis, rheumatoid arthntrs, juvenile chronic arthntis, osteoarthπtrs, spondyloarthropathies, systemic sclerosis (scleroderma), idiopathic inflammatory myopathies (dermatomyositis. polymyositis), Sjogren's syndrome systemic vascuhtis. sarcordosrs, autormmune hemolytic anemia (immune pancytopema, paroxysmal nocturnal hemoglobinuπa), autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), thyroiditis (Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic thyroiditis), diabetes mellitus, immune-mediated renal disease (glomerulonephntrs. tubulomterstrtial nephrrtis), demyehnatmg diseases of the central and penpheral nervous systems such as multiple sclerosis, idiopathic demyehnatmg polyneuropathy or Guillain-Barre syndrome, and chronic inflammatory demyehnatmg polyneuropathy, hepatobihary diseases such as infectious hepatitis (hepatitis A, B, C, D, E and other non-hepatotropic viruses), autoimmune chronic active hepatitis, pπmary biliary cirrhosis, granulomatous hepatitis, and sclerosmg cholangitis, mflammatory bowel disease (ulcerative colitis Crohn's disease), gluten- sensitive enteropathy, and Whipple's disease, autoimmune or immune-mediated skm diseases including bullous skm diseases, erythema multiforme and contact dermatitis, psonasis, allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and urticana, lmmunologic diseases of the lung such as eosmophilic pneumonias, idiopathic pulmonary fibrosis and hypersensrtrvity pneumomtis, transplantation associated diseases including graft rejection and graft - versus-host-drsease
In systemrc lupus erythematosus, the central medrator of drsease rs the productron of autoreactive antibodies to self proteins/tissues and the subsequent generation of immune-mediated inflammation Antibodies either directly or indirectly mediate tissue injury Though T lymphocytes have not been shown to be directly involved in tissue damage, T lymphocytes are required for the development of auto-reactive antibodies The genesis of the disease is thus T lymphocyte dependent Multiple organs and systems are affected clinically including kidney, lung, musculoskeletal system, mucocutaneous, eye, central nervous system, cardrovascular system, gastrointestinal tract, bone marrow and blood
Rheumatoid arthritis (RA) is a chronic systemic autoimmune mflammatory disease that mainly involves the synovial membrane of multiple joints with resultant injury to the articular cartilage The pathogenesis is T lymphocyte dependent and is associated with the production of rheumatoid factors, auto-antrbodres directed agamst self IgG. with the resultant formation of immune complexes that attain high levels in joint fluid and blood These complexes in the joint may induce the marked infiltrate of lymphocytes and monocytes mto the synovium and subsequent marked synovial changes, the joint space/fluid if infiltrated by similar cells with the addition of numerous neutrophils Tissues affected are pπmanly the joints, often m symmetncal pattern However, extra- articular drsease also occurs n two major forms One form is the development of extra-articular lesions with ongoing progressive joint disease and typical lesions of pulmonary fibrosis, vascuhtis, and cutaneous ulcers The second form of extra-articular drsease is the so called Felty's syndrome which occurs late in the RA disease course, sometimes after joint disease has become quiescent, and involves the presence of neutropenia, thrombocytopenia and splenomegaly This can be accompanied by vascuhtis in multiple organs with formations of nfarcts, skm ulcers and gangrene Patients often also develop rheumatoid nodules in the subcutis tissue overlying affected jomts; the nodules late stage have necrotic centers surrounded by a mrxed inflammatory cell infiltrate. Other mamfestatrons whrch can occur n RA include- pencarditis, pleuπtis, coronary arterrtis, mtestitial pneumonitis with pulmonary fbrosrs, keratoconjunctrvrtis srcca, and rhematoid nodules.
Juvenile chronic arthritis is a chronic idiopathic inflammatory disease which begins often at less than 16 years of age Its phenotype has some similaπties to RA; some patients which are rhematoid factor posrtrve are classified as juvenile rheumatoid arthntrs. The drsease rs sub-classrfred mto three major categoπes: paucrartrcular, polyarticular, and systemrc. The arthntrs can be severe and is typically destructive and leads to joint ankylosis and retarded growth. Other manifestations can include chronic antenor uveitis and systemic amylordosrs
Spondyloarthropathies are a group of drsorders wrth some common clrmcal features and the common association with the expression of HLA-B27 gene product. The disorders include ankylosmg sponyhtis, Rerter's syndrome (reactive arthntrs), arthntrs assocrated with inflammatory bowel drsease, spondyhtrs associated with psoriasis, juvenile onset spondyloarthropathy and undifferentiated spondyloarthropathy. Distinguishing features include sacroileitis with or without spondyhtis; inflammatory asymmetnc arthntis, assocratron with HLA-B27 (a serologically defined allele of the HLA-B locus of class I MHC); ocular inflammation, and absence of autoantibodies associated with other rheumatoid disease. The cell most implicated as key to induction of the disease is the CD8+ T lymphocyte, a cell which targets antigen presented by class I MHC molecules. CD8+ T cells may react against the class I MHC allele HLA-B27 as if it were a forergn peptrde expressed by MHC class I molecules It has been hypothesized that an epitope of HLA-B27 may mimic a bactenal or other microbial antigenic epitope and thus induce a CD8+ T cells response
Systemic sclerosis (scleroderma) has an unknown etiology A hallmark of the disease is induration of the skin, likely this is induced by an active inflammatory process. Scleroderma can be localized or systemic; vascular lesions are common and endothelial cell injury m the microvasculature is an early and important event in the development of systemic sclerosis, the vascular injury may be immune mediated. An immunologic basis is implied by the presence of mononuclear cell infiltrates in the cutaneous lesions and the presence of anti-nuclear antibodies in many patients. ICAM-1 is often upregulated on the cell surface of fibroblasts rn skm lesrons suggesting that T cell interaction with these cells may have a role m the pathogenesis of the disease. Other organs involved include, the gastrointestinal tract" smooth muscle atrophy and fibrosis resulting in abnormal peπstalsis/motihty, kidney concentnc subendothelial mtimal proliferation affecting small arcuate and interlobular arterres wrth resultant reduced renal cortical blood flow, results in proteinuna, azotemia and hypertension. skeletal muscle, atrophy, interstitial fibrosis, inflammation, lung: interstitial pneumonitis and interstitial fibrosis; and heart: contraction band necrosis, scarnng fibrosis.
Idiopathic mflammatory myopathies mcludrng dermatomyositis, polymyositis and others are disorders of chronic muscle inflammation of unknown etiology resulting m muscle weakness. Muscle injury/inflammation rs often symmetπc and progressive. Autoantibodies are associated with most forms These myositis-specific autoantibodies are directed against and inhibit the function of components, proteins and RNA's, mvolved in protein synthesis.
Sjogren's syndrome is due to immune-mediated inflammation and subsequent functional destruction of the tear glands and salivary glands. The disease can be associated with or accompanied by inflammatory connective tissue diseases The disease is associated with autoantibody production agamst Ro and La antigens, both of which are small RNA-protein complexes Lesions result in keratoconjunctivitis sicca, xerostomia, with other manifestations or associations including bilary cirrhosis, peπpheral or sensory neuropathy, and palpable purpura.
Systemrc vasculrtrs are diseases rn whrch the pnmary lesion is inflammation and subsequent damage to blood vessels which results m ischemia/necrosis/degeneration to tissues supphed by the affected vessels and eventual end-organ dysfunction in some cases. Vascuhtides can also occur as a secondary lesron or sequelae to other rmmune-rnflammatory mediated diseases such as rheumatoid arthntis, systemic sclerosis, etc., particularly in diseases also associated with the formation of rmmune complexes. Drseases rn the pnmary systemrc vasculitrs group mclude: systemrc necrotrzing vascuhtis: polyarteπtis nodosa, allergic angntis and granulomatosis, polyangntis; Wegener's granulomatosis; lymphomatord granulomatosrs; and grant cell artentrs. Mrscellaneous vasculrtrdes include: mucocutaneous lymph node syndrome (MLNS or Kawasaki's drsease), isolated CNS vascuhtis. Behet's disease, thromboangiitis oblrterans (Buerger's disease) and cutaneous necrotizing venuhtis The pathogenic mechanism of most of the types of vascuhtis listed is believed to be pπmaπly due to the deposition of immunoglobulin complexes in the vessel wall and subsequent induction of an inflammatory response erther via ADCC, complement activation, or both
Sarcoidosis is a condition of unknown etiology which is charactenzed by the presence of epithelioid granulomas in nearly any tissue in the body; involvement of the lung is most common The pathogenesis involves the persistence of activated macrophages and lymphoid cells at sites of the disease with subsequent chronic sequelae resultant from the release of locally and systemically active products released by these cell types
Autoimmune hemolytic anemia including autoimmune hemolytic anemia, immune pancytopenia, and paroxysmal noctural hemoglobinuria is a result of production of antibodies that react with antigens expressed on the surface of red blood cells (and rn some cases other blood cells including platelets as well) and is a reflection of the removal of those antibody coated cells via complement mediated lysis and/or ADCC/Fc-receptor-mediated mechanrsms In autoimmune thrombocytopenra including thrombocytopenic purpura, and immune- mediated thrombocytopenia in other clinical settings, platelet destruction/removal occurs as a result of either antibody or complement attaching to platelets and subsequent removal by complement lysis, ADCC or FC-receptor mediated mechanisms
Thyroiditis including Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, and atrophic thyroiditis, are the result of an autoimmune response against thyroid antigens with production of antibodies that react with proteins present in and often specific for the thyroid gland Experimental models exist including spontaneous models rats (BUF and BB rats) and chickens (obese chicken strain), rnducrble models immunization of animals with either thyroglobulm, thyroid microsomal antigen (thyroid peroxidase)
Type I diabetes mellitus or sulm-dependent diabetes is the autoimmune destruction of pancreatrc islet β cells, this destruction rs medrated by auto-antrbodies and auto-reactive T cells Antibodies to insulin or the insulin receptor can also produce the phenotype of lnsuhn-non- responsiveness
Immune mediated renal diseases, including glomerulonephntis and tubulomterstrtral nephπtis, are the result of antibody or T lymphocyte mediated injury to renal tissue either directly as a result of the production of autoreactive antibodies or T cells agamst renal antigens or indirectly as a result of the deposition of antibodies and/or immune complexes in the kidney that are reactive agamst other, non-renal antigens Thus other immune-mediated diseases that result in the formation of immune- complexes can also induce immune mediated renal disease as an indirect sequelae Both direct and indirect immune mechanisms result in inflammatory response that produces/ induces lesion development in renal tissues with resultant organ function impairment and in some cases progression to renal failure Both humoral and cellular immune mechanisms can be involved in the pathogenesis of lesions
Demyehnatmg diseases of the central and penpheral nervous systems, including Multiple Sclerosis, idiopathic demyehnatmg polyneuropathy or Guillain-Baπ syndrome, and Chronic Inflammatory Demyehnatmg Polyneuropathy are believed to have an autoimmune basis and result m nerve demyelination as a result of damage caused to ohgodendrocytes or to myelrn directly In MS there is evrdence to suggest that disease induction and progression is dependent on T lymphocytes Multiple Sclerosis is a demyehnatmg disease that is T lymphocyte-dependent and has either a relapsing-remitting course or a chronic progressive course The etiologj is unknown, however, viral infections, genetic predisposition, environment, and autoimmunity all contnbute Lesions contain infiltrates of predominantly T lymphocyte mediated, microghal cells and infiltrating macrophages, CD4+T lymphocytes are the predominant cell type at lesions The mechanism of ohgodendrocyte cell death and subsequent demyelination is not known but is likely T lymphocyte dnven Inflammatory and Fibrotic Lung Disease, including Eosmophilic Pneumonias, Idiopathic Pulmonary Fibrosis and Hypersensitivity Pneumonitis may involve a deregulated immune- inflammatory response Inhibition of that response would be of therapeutic benefit
Autoimmune or Immune-mediated Skm Disease including Bullous Skm Diseases, Erythema Multiforme, and Contact Dermatitis are mediated by auto-antibodies, the genesis of which is T lymphocyte-dependent
Psonasis is a T lymphocyte-mediated inflammatory disease Lesions contain infiltrates of T lymphocytes, macrophages and antigen processing cells, and some neutrophrls
Allergrc drseases, including asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity, and urtrcana are T lymphocyte dependent These diseases are predommantly mediated by T lymphocyte induced inflammation, IgE mediated-mflammation or a combination of both
Transplantation associated diseases, including Graft rejection and Graft- Versus-Host-Disease (GVHD) are T lymphocyte-dependent, mhrbition of T lymphocyte functron is amelroratrve
Other diseases in which intervention of the immune and/or mflammatory response have benefit are infectious disease including but not limited to viral infection (including but not limited to AIDS, hepatitis A, B, C, D, E and herpes) bacteπal infection, fungal infections, and protozoal and parasitic infections (molecules (or denvatives/agomsts) which stimulate the MLR can be utilized therapeutically to enhance the immune response to infectious agents), diseases of immunodeficiency (molecules/denvatives/agonists) which stimulate the MLR can be utihzed therapeutically to enhance the immune response for conditions of mhented, acquired, infectious induced (as in HIV infectron), or ratrogemc (i.e as from chemotherapy) immunodeficiency), and neoplasia
It has been demonstrated that some human cancer patients develop an antibody and/or T lymphocyte response to antigens on neoplastic cells It has also been shown in ammal models of neoplasia that enhancement of the immune response can result in rejection or regression of that particular neoplasm Molecules that enhance the T lymphocyte response in the MLR have utility in vivo in enhancing the immune response agamst neoplasia Molecules which enhance the T lymphocyte proliferative response in the MLR (or small molecule agonists or antibodies that affected the same receptor m an agonistic fashion) can be used therapeutically to treat cancer Molecules that inhibit the lymphocyte response in the MLR also functron rn vivo duπng neoplasia to suppress the immune response to a neoplasm; such molecules can either be expressed by the neoplastic cells themselves or their expression can be induced by the neoplasm m other cells Antagonism of such inhibitory molecules (either with antibody, small molecule antagonists or other means) enhances rmmune-mediated tumor rejection
Additionally, inhibition of molecules with proinflammatory properties may have therapeutic benefit in reperfusion rnjury, stroke, myocardial infarction, atherosclerosis, acute lung injury, hemorrhagic shock; burn, sepsis/ septic shock, acute tubular necrosis, endometπosis; degenerative joint disease and pancreatis
The compounds of the present mventron, e g polypeptrdes or antrbodres, are admrmstered to a mammal, preferably a human, in accord with known methods, such as intravenous administration as a bolus or by continuous infusion over a penod of time, by intramuscular, mtrapeπtoneal, mtracerobrospinal, subcutaneous, lntra-articular. mtrasynovial, intrathecal, oral, topical, or inhalation (mtranasal, mtrapulmonary) routes. Intravenous or inhaled administration of polypeptides and antibodies is preferred
In lmmunoadjuvant therapy, other therapeutic regrmens, such admrnrstration of an anti-cancer agent, may be combined wrth the admrnrstration of the proterns, antibodies or compounds of the instant mvention. For example, the patient to be treated with a the lmmunoadjuvant of the mventron may also recerve an anti-cancer agent (chemotherapeutic agent) or radiation therapy. Preparation and dosmg schedules for such chemotherapeutic agents may be used according to manufacturers' instructions or as determmed empiπcally by the skilled practitioner Preparation and dosmg schedules for such chemotherapy are also descnbed rn Chemotherapy Service Ed., M.C. Perry, Wrlhams & Wilkins, Baltimore, MD ( 1992) The chemotherapeutic agent may precede, or follow administration of the lmmunoadjuvant or may be given simultaneously therewith. Additionally, an anti-oestrogen compound such as tamoxifen or an anti-progesterone such as onapπstone (see, EP 616812) may be given in dosages known for such molecules.
It may be desirable to also administer antibodies against other immune disease associated or tumor associated antigens, such as antibodies which bind to CD20, CD1 la, CD 18, ErbB2, EGFR, ErbB3, ErbB4, or vascular endothelial factor (VEGF) Alternatively, or in addrtion, two or more antibodies binding the same or two or more different antigens disclosed herein may be coadmmistered to the patient. Sometimes, it may be beneficial to also administer one or more cytokines to the patient. In one embodiment, the polypeptides of the mvention are coadmmistered with a growth inhibitory agent. For example, the growth inhibitory agent may be administered first, followed by a polypeptide of the mventron. However, simultaneous administration or administration first is also contemplated. Suitable dosages for the growth inhibitory agent are those presently used and may be lowered due to the combined action (synergy) of the growth inhibitory agent and the polypeptide of the invention.
For the treatment or reduction in the severity of immune related disease, the appropnate dosage of an a compound of the invention will depend on the type of disease to be treated, as defined above, the severity and course of the disease, whether the agent is administered for preventive or therapeutic purposes, previous therapy, the patient's clinical history and response to the compound, and the drscretion of the attending physician The compound is suitably administered to the patient at one time or over a senes of treatments For example, depending on the type and severity of the disease, about 1 ug/kg to 15 mg/kg (e g 0 l-20mg/kg) of polypeptide or antibody is an initial candidate dosage for administration to the patient, whether, for example, by one or more separate administrations, or by continuous infusion A typical daily dosage might range from about 1 ug/kg to 100 mg/kg or more, depending on the factors mentioned above For repeated administrations over several days or longer, depending on the condition, the treatment is sustained until a desired suppression of disease symptoms occurs However, other dosage regimens mav be useful The progress of this therapy is easily monitored by conventional techniques and assays
12 Articles of Manufacture
In another embodrment of the mvention, an article of manufacture containing materials useful for the dragnosis or treatment of the disorders descnbed above is provided The article of manufacture compπses a contamer and a label Surtable containers include, for example, bottles, vials, synnges. and test tubes The containers may be formed from a vanety of matenals such as glass or plastic The container holds a composition which is effective for dragnosmg or treating the condition and may have a sterile access port (for example the contamer may be an mtravenous solutron bag or a vral havrng a stopper pierceable by a hypodermrc injection needle) The active agent m the composition is usually a polypeptide or an antibody of the mvention The label on. or associated with, the container indicates that the composition is used for diagnosing or treating the condition of choice The article of manufacture may further compnse a second container compnsmg a pharmaceutically-acceptable buffer, such as phosphate-buffered salrne, Rmger's solution and dextrose solution It may further mclude other matenals desirable from a commercral and user standpomt, including other buffers, diluents, filters, needles, synnges, and package inserts with instructions for use
13 Diagnosis and Prognosis of Immune Related Disease
Cell surface proteins, such as proteins which are overexpressed in certain immune related diseases, are excellent targets for drug candidates or disease treatment The same proteins along with secreted proteins encoded by the genes amplified in immune related disease states find additional use in the diagnosis and prognosis of these diseases For example, antibodies directed agamst the protein products of genes amplified in multiple sclerosis, rheumatoid arthntis, or another immune related disease, can be used as diagnostics or prognostics
For example, antibodies, including antibody fragments, can be used to qualitatively or quantitatively detect the expression of proteins encoded by amplified or overexpressed genes ("marker gene products") The antibody preferably is equipped with a detectable, e g fluorescent label, and binding can be monitored by light microscopy, flow cytometry, fluoπmetry, or other technrques known m the art These technrques are particularly suitable, if the overexpressed gene encodes a cell surface protern Such binding assavs are performed essentially as decπbed above In situ detection of antibodv binding to the marker gene products can be performed, for example, by immunofluorescence or immunoelectron microscopy For this purpose, a histological specimen is removed from the patient, and a labeled antibody is applied to it, preferably by overlaying the antibodv on a biological sample This procedure also allows for determining the distπbution of the marker gene product in the tissue examined It will be apparent for those skilled in the art that a wrde varrety of histological methods are readily available for in situ detectron
The followmg examples are offered for rllustratrve purposes only, and are not mtended to hmrt the scope of the present mvention in any way
All patent and literature references cited in the present specification are hereby incoφorated by reference in their entirety
EXAMPLES Commercially available reagents referred to in the examples were used according to manufacturer's instructions unless otherwise indicated The source of those cells identified in the followmg examples, and throughout the specification, by ATCC accession numbers is the Amencan Type Culture Collection, Manassas, VA Unless otherwrse noted, the present mvention uses standard procedures of recombinant DNA technology, such as those descnbed hereinabove and in the followmg textbooks Sambrook et al , Molecular Cloning A Laboratory Manual, Cold Spnng Harbor Press N Y , 1989, Ausubel et al , Current Protocols in Molecular Biology, Green Publishing Associates and Wiley Interscience, N Y , 1989, Innis et al , PCR Protocols A Guide to Methods and Applications, Academic Press, mc , N Y , 1990, Harlow et al , Antibodies A Laboratory Manual, Cold Spnng Harbor Press, Cold Spnng Harbor, 1988, Gait, M J , Oligonucleotide Synthesis, IRL Press. Oxford, 1984, R I Freshney. Animal Cell Cultur e, 1987 Cohgan t α/ Current Pr otocols in Immunology, 1991
EXAMPLE 1 Isolation of cDNA clones Encoding Human PRQ245 PRQ217 PRO301 PRQ266. PRQ335 PRQ331 or PRQ326
I Isolation of cDNA Clones Encoding Human PRQ245
The extracellular domain (ECD) sequences (including the secretion signal, if any) of from about 950 known secreted proterns from the Swrss-Prot publrc protern database were used to search expressed sequence tag (EST) databases The EST databases included publrc EST databases (e g ,
GenBank) and a proprietary EST DNA database (LIFESEQ™, Incyte Pharmaceuticals, Palo Alto,
CA) The search was performed usmg the computer program BLAST or BLAST2 (Altshul et al ,
Methods m Enzymology 266 460-480 ( 1996)) as a compaπson of the ECD protern sequences to a 6 frame translation of the EST sequence Those comparisons resulting in a BLAST score of 70 (or rn some cases 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green, University of Washington, Seattle, Washington)
A consensus DNA sequence encoding PR0245 was assembled relative to the other identified EST sequences, where the consensus sequence was designated herein as DNA30954, and the polypeptide showed some structural homology to transmembrane protein receptor tyrosine kinase proteins
Based on the DNA30954 consensus sequence, oligonucleotides were synthesized to identify by PCR a cDNA library that contamed the sequence of mterest and for use as probes to isolate a clone of the full-length coding sequence for PR0245
A parr of PCR primers (forward and reverse) were synthesized forward PCR pnmer 5'-ATCGTTGTGAAGTTAGTGCCCC-3' (SEQ ID NO 15) reverse PCR pπmer 5'-ACCTGCGATATCCAACAGAATTG-3' (SEQ ID NO 16)
Addrtronally, a synthetic oligonucleotide hybndization probe was constructed from the consensus DNA30954 sequence whrch had the followmg nucleotrde sequence hvbπdrzatron probe
5'-GGAAGAGGATACAGTCACTCTGGAAGTATTAGTGGCTCCAGCAGTTCC-3' (SEQ ID NO 17)
In order to screen several hbranes for a source of a full-length clone, DNA from the hbranes was screened by PCR amplification with the PCR pnmer pair identified above A positive library was then used to isolate clones encoding the PR0245 gene using the probe oligonucleotide and one of the PCR pnmers
RNA for construction of the cDNA libraries was isolated from human fetal liver tissue The cDNA hbranes used to isolate the cDNA clones were constructed by standard methods using commercially available reagents such as those from Invrtrogen, San Drego, CA The cDNA was pruned with oligo dT containing a Notl site, linked with blunt to Sail hemikmased adaptors, cleaved with Notl, sized appropnately by gel electrophoresis, and cloned in a defined onentation mto a suitable cloning vector (such as pRKB or pRKD, pRK5B is a precursor of pRK5D that does not contain the Sfil site, see, Holmes et al , Science. 253 1278- 1280 ( 1991)) in the unique Xliol and Notl sites
DNA sequencing of the clones isolated as descnbed above gave the full-length DNA sequence for PR0245 [herein designated as UNQ219 (DNA35638)] and the denved protein sequence for PR0245
The entire nucleotide sequence of UNQ219 (DNA35638) is shown in Figure 3 (SEQ ID NO
1 ) Clone UNQ219 (DNA35638) contains a single open reading frame wrth an apparent translatronal initiation site at nucleotide positions 89-91 [Kozak et al , supra] and ending at the stop codon at nucleotide positions 1025-1027 (Fig. 3). The predicted polypeptide precursor is 312 amino acids long (Figure. 4. PR0245; SEQ ID NO 2) Clone UNQ219 (DNA35638) has been deposited with ATCC on September 17, 1997 and is assigned ATCC Deposit No. 209265.
Analysis of the amino acid sequence of the full-length PR0245 suggests that a portion of it possesses 60% amino acid identity with the human c-myb protein and, therefore, may be a new member of the transmembrane protein receptor tyrosine kinase family
II. Isolation of cDNA clones Encoding PRQ217
The extracellular domain (ECD) sequences (including the secretion signal sequence, if any) from about 950 known secreted proteins from the Swiss-Prot public database were used to search EST databases The EST databases included public databases (e.g., Dayhof, GenBank), and propπetary databases (e g LIFESEQ™, Incyte Pharmaceuticals, Palo Alto, CA). The search was performed using the computer program BLAST or BLAST2 (Altschul, SF and Gish ( 1996), Methods in Enzx'mology 266" 460-80 (1996), http://blast.wustl/edu/blast/README.html) as a companson of the ECD protein sequences to a 6 frame translatron of the EST sequences. Those compansons wrth a Blast score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled mto consensus DNA sequences with the program "phrap" (Phrl Green, Unrversrty of Washrngton, Seattle, WA; (http://bozeman.mbt.washmgton.edu/phrap.docs/phrap.html).
Consensus DNA sequences encoding EGF-hke homologues were assembled (DNA28726, DNA28730 and DNA28760) using phrap. In some cases, the consensus DNA sequence was extended using repeated cycles of blast and phrap to extend the consensus sequence as far as possrble usmg the three sources of EST sequences listed above
Based on this consensus sequence, oligonucleotides were synthesized. 1 ) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to rsolate a clone of the full-length codmg sequence The parr of forward and reverse PCR pnmers (notated as * f and *.r, respectively) may range from 20 to 30 nucleotides (typically 24), and are designed to give a PCR product of 100-1000 bp m length. The probe sequences (notated as *.p) are typically 40-55 bp (typically 50) in length In some cases additional oligonucleotides are synthesrzed when the consensus sequence is greater than 1-1 5 kbp. In order to screen several hbranes for a source of a full-length clone. DNA from the hbranes was screened by PCR amphficatron, as per Ausubel et al , Current Protocols in Molecular Bwlog ', with the PCR pnmer pair. A positive library was then used to isolate clones encoding the gene of interest by the in vivo cloning procedure using the probe oligonucleotide and one of the PCR pnmers This library was used to isolate DNA32279, DNA32292 and DNA33094 was fetal krdney, fetal lung and fetal lung, respectively RNA for the construction of the cDNA libraries was isolated using standard isolation protocols, e.g., Ausubel et al, Current Protocols in Molecular Biolog}', from tissue or cell line sources or it was purchased from commercial sources (e.g., Clontech). The cDNA libraries used to isolate the cDNA clones were constructed by standard methods (e.g., Ausubel et al.) using commercially available reagents (e.g., Invitrogen). The cDNA was primed with oligo dT containing a Noti site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoesis, and cloned in a defined orientation in a suitable cloning vector (pRK5B or pRK5D) in the unique Xhol and Notl sites.
A cDNA clone was sequenced in its entirety. The entire nucleotide sequence of EGF-like homologue PR0217 is shown in Figure 5 (SEQ ID NO: 3). The predicted polypeptide is 379 (PR0217; Figure 6; SEQ ID NO: 4) amino acids in length with a molecule weight of approximately 41.52 kDa.
The oligonucleotide sequences used in the above procedure were the following: 28726.p (OLI500) (SEQ ID NO: 18) GGGTACACCTGCTCCTGCACCGACGGATATTGGCTTCTGGAAGGCC
28726.f (OLI 502) (SEQ ID NO: 19) ACAGATTCCCACCAGTGCAACC
28726.r (OLI 503) (SEQ ID NO: 20) CACACTCGTTCACATCTTGGC
28730.p (OLI 516) (SEQ ID NO: 21) AGGGAGCACGGACAGTGTGCAGATGTGGACGAGTGCTCACTAGCA
28730.f (OLI 517) (SEQ ID NO: 22) AGAGTGTATCTCTGGCTACGC
28730.r (OLI 518) (SEQ ID NO: 23) TAAGTCCGGCACATTACAGGTC
28760.p (OLI 617) (SEQ ID NO: 24) CCCACGATGTATGAATGGTGGACTTTGTGTGACTCCTGGTTTCTGCATC
28760.f (OLI 618) (SEQ ID NO: 25) AAAGACGCATCTGCGAGTGTCC
28760.r (OLI 619) (SEQ ID NO: 26) TGCTGATTTCACACTGCTCTCCC
III. Isolation of cDNA clones Encoding Human PRO301
The extracellular domain (ECD) sequences (including the secretion signal sequence, if any) from about 950 known secreted proteins from the Swiss-Prot public database were used to search EST databases. The EST databases included public EST databases (e.g., GenBank), a proprietary EST database (LIFESEQ™, Incyte Pharmaceuticals, Palo Alto, CA). The search was performed using the computer program BLAST or BLAST2 [Altschul et al, Methods in Enzymology, 266:460-480 (1996); http://blast.wustl/edu/blast/README.html] as a comparison of the ECD protein sequences to a 6 frame translation of the EST sequences. Those comparisons resulting in a BLAST score of 70 (or in some cases, 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green, University of Washington, Seattle, Washington; http://bozeman.mbt.washington.edu phrap.docs/phrap.html).
A consensus DNA sequence encoding DNA35936 was assembled using phrap. In some cases, the consensus DNA sequence was extended using repeated cycles of blast and phrap to extend the consensus sequence as far as possible using the three sources of EST sequences listed above.
Based on this consensus sequence, oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence. Forward and reverse PCR primers (notated as *.f and *.r, respectively) may range from 20 to 30 nucleotides (typically about 24), and are designed to give a PCR product of 100- 1000 bp in length. The probe sequences (notated as *.p) are typically 40-55 bp (typically about 50) in length. In some cases, additional oligonucleotides are synthesized when the consensus sequence is greater than 1-1.5 kbp. In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification, as per Ausubel et al, Current Protocols in Molecular Biology, with the PCR primer pair. A positive library was then used to isolate clones encoding the gene of interest by the in vivo cloning procedure suing the probe oligonucleotide and one of the PCR primers.
In order to screen several libraries for a source of a full-length clone, DNA from the libraries was screened by PCR amplification with the PCR primer pair identified above. A positive library was then used to isolate clones encoding the PRO301 gene using the probe oligonucleotide and one of the PCR primers.
RNA for construction of the cDNA libraries was isolated from human fetal kidney. The cDNA libraries used to isolated the cDNA clones were constructed by standard methods using commercially available reagents (e.g., Invitrogen, San Diego, CA; Clontech, etc.) The cDNA was primed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD; pRK5B is a precursor of pRK5D that does not contain the Sfil site; see, Holmes et al. Science, 253:1278-1280 (1991)) in the unique Xhol and Notl sites.
A cDNA clone was sequenced in its entirety. The full length nucleotide sequence of native sequence PRO301 is shown in Figure 7 (SEQ ID NO: 5). Clone DNA40628 contains a single open reading frame with an apparent translational initiation site at nucleotide positions 52-54 [Kozak et al, supra] (Fig. 7). The predicted polypeptide (PRO301; Figure 8; SEQ ID NO: 6) is 299 amino acids long with a predicted molecular weight of 32583 daltons and pi of 8.29. Clone DNA40628 has been deposited with ATCC and is assigned ATCC deposit No. 209432.
Based on a BLAST and FastA sequence alignment analysis of the full-length sequence, PRO301 shows amino acid sequence identity to A33 antigen precursor (30%) and coxsackie and adenovirus receptor protein (29%).
The oligonucleotide sequences used in the above procedure were the following:
OLI2162 (35936.fl) (SEQ ID NO: 27)
TCGCGGAGCTGTGTTCTGTTTCCC
OLI2163 (35936.pl) (SEQ ID NO: 28)
TGATCGCGATGGGGACAAAGGCGCAAGCTCGAGAGGAAACTGTTGTGCCT
OLI2164 (35936.f2) (SEQ ID NO: 29)
ACACCTGGTTCAAAGATGGG
OLI2165 (35936.rl) (SEQ ID NO: 30)
TAGGAAGAGTTGCTGAAGGCACGG
OLI2166 (35936.f3) (SEQ ID NO: 31)
TTGCCTTACTCAGGTGCTAC
OLI2167 (35936.r2) (SEQ ID NO: 32)
ACTCAGCAGTGGTAGGAAAG
IV. Isolation of cDNA Clones Encoding Human PRQ266
The extracellular domain (ECD) sequences (including the secretion signal, if any) of from about 950 known secreted proteins from the Swiss-Prot public protein database were used to search expressed sequence tag (EST) databases. The EST databases included public EST databases (e.g., GenBank) and a proprietary EST DNA database (LIFESEQ™, Incyte Pharmaceuticals, Palo Alto, CA). The search was performed using the computer program BLAST or BLAST2 (Altshul et al, Methods in Enzymology 266:460-480 (1996)) as a comparison of the ECD protein sequences to a 6 frame translation of the EST sequence. Those comparisons resulting in a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green, University of Washington, Seattle, Washington; http:/Λozeman.mbt.washington.edu phrap.docs/plιrap.html).
Based on an expression sequence tag oligonucleotides were synthesized: 1) to identify by PCR a cDNA library that contained the sequence of interest, and 2) for use as probes to isolate a clone of the full-length coding sequence for PR0266. Forward and reverse PCR primers generally range from 20 to 30 nucleotides and are often designed to give a PCR product of about 100-1000 bp in length. The probe sequences are typically 40-55 bp in length. In some cases, additional oligonucleotides are synthesized when the consensus sequence is greater than about l-1.5kbp. In order to screen several hbranes for a full-length clone. DNA from the hbranes was screened by PCR amplification, as ber Ausubel et al , Current Protocols in Molecular Biology, with the PCR pnmer pair A positive library was then used to isolate clones encodmg the gene of interest by the in vivo clongm procedure using the probe oligonucleotide and one of the pnmer pairs
A pair of PCR pπmers ( forward and reverse) were synthesized forward PCR pnmer 5'-GTTGGATCTGGGCAACAATAAC-3' (SEQ ID NO. 33) reverse PCR pnmer 5'-ATTGTTGTGCAGGCTGAGTTTAAG-3' (SEQ ID NO 34)
Addrtronally, a synthetic ohgonucleotide hybndization probe was constructed whrch had the followmg nucleotrde sequence hvbrrdrzation probe 5'-GGTGGCTATACATGGATAGCAATTACCTGGACACGCTGTCCCGGG-3' (SEQ ID NO 35)
In order to screen several hbranes for a source of a full-length clone, DNA from the hbranes was screened by PCR amplification with the PCR pnmer pair identified above A positive library was then used to isolate clones encoding the PR0266 gene using the probe oligonucleotide and one of the PCR pnmers
RNA for construction of the cDNA hbranes was isolated from human fetal bram tissue The cDNA hbranes used to isolate the cDNA clones were constructed by standard methods using commercially available reagents such as those from Invitrogen, San Diego, CA The cDNA was pruned with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropriately by gel electrophoresis, and cloned in a defined oπentation into a suitable clomng vector (such as pRKB or pRKD, pRK5B is a precursor of pRK5D that does not contain the Sfil site, see, Holmes et al , Science, 253 1278-1280 (1991)) in the unique Xhol and Notl sites
DNA sequencing of the clones isolated as descnbed above gave the full-length DNA sequence for PR0266 [herein designated as UNQ233 (DNA37150)] and the denved protein sequence for PR0266
The entire nucleotide sequence of UNQ233 (DNA37150) is shown in Figure 9 (SEQ ID NO 7) Clone UNQ233 (DNA37150) contains a single open reading frame wrth an apparent translational initiation site at nucleotide positions 1-3 [Kozak et al . supra] and ending at the stop codon after nucleotide position 2088 The predicted polypeptide precursor is 696 ammo acids long (Figure 10, PR0266. SEQ ID NO 8) Clone UNQ233 (DNA37150) has been deposited with ATCC and is assigned ATCC deposit no 209401
Analysis of the amino acid sequence of the full-length PR0266 polypeptide suggests that portions of it possess significant homology to a SLIT protein, thereby indicating that PR0266 may be a novel leucine rich repeat protein V Isolation of cDNA Clones Encoding Human PRQ335 PRQ331 or PRQ326
The extracellular domain (ECD) sequences (including the secretion signal, if any) of from about 950 known secreted proteins from the Swrss-Prot publrc protein database were used to search expressed sequence tag (EST) databases The EST databases included public EST databases (e g , GenBank) and a propπetary EST DNA database (LIFESEQ™, Incyte Pharmaceuticals, Palo Alto, CA) The search was performed using the computer program BLAST or BLAST2 (Altshul et al , Methods m Enzymology 266 460-480 (1996)) as a companson of the ECD protern sequences to a 6 frame translation of the EST sequence Those compansons resulting rn a BLAST score of 70 (or in some cases 90) or greater that did not encode known proteins were clustered and assembled into consensus DNA sequences with the program "phrap" (Phil Green, University of Washington, Seattle, Washington, http //bozeman mbt washmgton edu/phrap docs/phrap html)
A consensus DNA sequence was assembled relatrve to other EST sequences usmg phrap Based on the consensus sequence, oligonucleotides were synthesized 1 ) to identify by PCR a cDNA library that contamed the sequence of mterest, and 2) for use as probes to rsolate a clone of the full- length coding sequence for PR0335, PR0331 or PR0326 Forward and reverse PCR pnmers generally range from 20 to 30 nucleotides and are often designed to give a PCR product of about 100- 1000 bp in length The probe sequences are typically 40-55 bp in length In some cases, additional olrgonucleotides are synthesrzed when the consensus sequence rs greater than about 1-1 5kbp In order to screen several hbranes for a full-length clone, DNA from the hbranes was screened by PCR amplrficatron, as per Ausubel et al , Current Protocols in Molecular Biology, with the PCR pnmer pair A positive library was then used to isolate clones encoding the gene of interest by the in vivo clongin procedure using the probe oligonucleotide and one of the pnmer pairs
In order to screen several hbranes for a source of a full-length clone, DNA from the hbranes was screened by PCR amplification with the PCR pnmer pair identified above A positiv e library was then used to isolate clones encoding the PR0335, PR0331 or PR0326 gene usmg the probe oligonucleotide and one of the PCR pnmers
RNA for construction of the cDNA hbranes was isolated from human fetal krdney trssue (PR0335 and PR0326) and human fetal brain (PR0331) The cDNA hbranes used to isolate the cDNA clones were constructed by standard methods usmg commercially available reagents such as those from Invitrogen, San Diego, CA The cDNA was pnmed with oligo dT containing a Notl site, linked with blunt to Sail hemikinased adaptors, cleaved with Notl, sized appropπatelv by gel electrophoresis, and cloned in a defined orientation into a suitable cloning vector (such as pRKB or pRKD, pRK5B is a precursor of pRK5D that does not contain the Sfil site, see. Holmes et al , Science. 253 1278-1280 ( 1991)) in the unique Xhol and Notl sites
DNA sequencing of the clones isolated as descπbed above gave the full-length DNA sequence for PR0335 (Frgure 1 1 SEQ ID NO 9), PR0331 (Figure 13. SEQ ID NO 1 1 ) or PR0326 (Figure 15. SEQ ID NO 13) and the denved protein sequence for PR0335 (Figure 12. SEQ ID NO 10). PR0331 (Figure 14, SEQ ID NO. 12) or PR0326 (Figure 16, SEQ ID NO 14) The nucleic acid encoding PR0335 was deposited with the ATCC on 2 June 1998 and is assigned ATCC Accession No. 209927. the nucleic acid encoding PR0331 was deposited with the ATCC on 7 November 1997 and is assigned ATCC Accession No 209439, and the nucleic acrd encoding PR0326 was deposited with the ATCC on 21 November 1997 and is assigned ATCC Accession No. 209489
Analysis of the ammo acid sequence of the full-length PR0335, PR0331 or PR0326 polypeptide suggests that portions of it possess significant homology to the LIG- 1 protein, thereby indicating that PR0335, PR0331 and PR0326 may be a novel LIG-1-related protern
EXAMPLE 2 Stimulatory Activity in Mixed Lymphocyte Reaction (MLR) Assay (No 24)
This example shows that the polypeptides of the mvention are active as a stimulator of the proliferation of stimulated T-lymphocytes. Compounds which stimulate prohferatron of lymphocytes are useful therapeutically where enhancement of an immune response is beneficial Compounds which inhibit prohferatron of lymphocytes are useful therapeutically where suppression of an immune response is beneficial A therapeutic agent may take the form of antagonists of the polypeptide of the invention, for example, muπne-human chimenc, humanized or human antibodies agamst the polypeptide.
The basic protocol for this assay is descnbed in Current Protocols in Immunology, unit 3.12; edited by J. E. Cohgan, A. M. Kruisbeek, D. H. Marghes, E. M. Shevach, W Strober, National Insitutes of Health, Publrshed by John Wiley & Sons, Inc.
More specifically, in one assay vanant, penpheral blood mononuclear cells (PBMC) are isolated from mammalian individuals, for example a human volunteer, by leukopheresis (one donor will supply stimulator PBMCs, the other donor will supply responder PBMCs) If desired, the cells are frozen m fetal bovine serum and DMSO after isolation. Frozen cells may be thawed overnrght in assay media (37°C, 5% CO2 ) and then washed and resuspended to 3 x 10^ cells/ml of assay media
(RPMI. 10% fetal bovine serum, 1% penicillin/streptomycin, 1% glutamine, 1% HEPES. 1% non- essential amino acids, 1 % pyruvate)
The stimulator PBMCs are prepared by irradiating the cells (about 3000 Rads) The assay is prepared by plating in tnphcate wells a mixture of lOOμl of test sample diluted to 1% or to 0.1%
50 μl of lrradrated strmulator cells and
50 μl of responder PBMC cells 100 microliters of cell culture media or 100 microliter of CD4-IgG is used as the control. The wells are then incubated at 37°C, 5% CO2 for 4 days. On day 5 and each well is pulsed with tritiated thymidine (i.O mC/well; Amersham). After 6 hours the cells are washed 3 times and then the uptake of the label is evaluated.
In another variant of this assay, PBMCs are isolated from the spleens of Balb/c mice and C57B6 mice. The cells are teased from freshly harvested spleens in assay media (RPMI; 10% fetal bovine serum, 1 % penicillin/streptomycin. 1% glutamine, 1% HEPES, 1% non-essential amino acids, 1 % pyruvate) and the PBMCs are isolated by overlaying these cells over Lympholyte M (Organon Teknika), centrifuging at 2000 m for 20 minutes, collecting and washing the mononuclear cell layer in assay media and resuspending the cells to lx 107 cells/ml of assay media. The assay is then conducted as described above using a sample having a PRO concentration obtained by diluting a stock solution. The results of this assay for compounds of the invention are shown below. Positive increases over control are considered positive with increases of greater than or equal to 180% being preferred. However, any value greater than control indicates a stimulatory effect for the test protein.
Table 2
PRO PRO Concentration Percent Increase Over Control
PR0245 0.1% 189.7
0.1% 193.7
1.0% 212.5
1.0% 300.5
PR0217 0.1% 74.5 π 1.0% 89.5
0.99 nM 97.0
9.9 nM 122.3
0.25 nM 144.8
2.5 nM 126.9
PRO301 50.0% 109.4
70.0 nM 133.7
700.0 nM 83.6
0.1% 58.7
PRO301 1.0% 127.7 π 0.1% 181.7
1.0% 187.3
0.1% 127.5
1.0% 108.3 PR0266 0.1% 136.4
0.1% 139.2
1.0% 189.8
1.0% 245.1
PR0335 50.0% 91.0
50.0% 103.8
0.1% 130.0
1.0% 180.2
PR0331 50.0% 155.5
0.1% 169.3
1.0% 128.1
0.1% 129.3
1.0% 162.5
PR0326 50.0% 91.0
50.0% 103.8
0.1% 130.0
1.0% 180.2
EXAMPLE 3 Skin Vascular Permeabrlrty Assay (No. 64) This assay shows that certam polypeptides of the invention stimulate an immune response and induce inflammation by inducing mononuclear cell, eosinophil and PMN infiltration at the site of injection of the animal. This skm vascular permeabrlrty assay rs conducted as follows. Harrless guinea pigs weighing 350 grams or more are anesthetized with ketamme (75-80 mg/Kg) and 5 mg/Kg xylazme intramuscularly (IM). A sample of punfied polypeptide of the invention or a conditioned media test sample is injected intradermally onto the backs of the test ammals wrth 100 μL per injection site. It is possible to have about 10-30, preferably about 16-24, rnjection sites per animal. One L of Evans blue dye (1% in physiologic buffered saline) is injected mtracardially Blemishes at the injection sites are then measured (mm diameter) at lhr and 6 hr post injection. Anrmals were sacπficed at 6 hrs after injection. Each skm injection site was biopsied and fixed in formalm The skins were then prepared for hrstopathalogrc evaluatron Each site was evaluated for mflammatory cell infiltration mto the skin. Sites with visible inflammatory cell mflammatron were scored as positive. Inflammatory cells can be neutrophihc, eosinophilic, monocytic or lymphocytic. The results of this test for compounds of the invention is shown below.
In the Table below, at least a minimal perivascular infiltrate at the injection site is scored as positve, no infiltrate at the site of injection is scored as negative.
Table 3 PRO Hours Post Injection Infiltrate Designation
PR0245 24 hr positive
PR0217 24 hr positive
PRO301 24 hr positive
PR0266 24 hr positive
PR0335 24 hr positive
PR0331 24 hr positive
PR0326 24 hr positive
EXAMPLE 4 Human Co-Stimulation Assay In addition to the activation signal mediated by the T cell receptor, T cell activation requires a costimulatory signal. One costimulatory signal is generated by the interaction of B7 (CD3) with CD28. In this assay, 96 well plates are coated with CD3 with or without CD28 and then human peripheral blood lymphocytes followed by a test protein, are added. Proliferation of the lymphocytes is determined by tritiated thymidine uptake. A positive assay indicates that the test protein provided a stimulatory signal for lymphocyte proliferation.
Material:
1) Hy clone D-PBS without Calcium, Magnesium
2) Nunc 96 well certified plates #4-39454
3) Anti-human CD3 Amac 0178 200 μg/ml stock
4) Anti-human CD28 Biodesign P42235M
5) Media: Gibco RPMI 1640 + 10 % Intergen #1020-90 FBS, 1% Glu, 1% P/S, 50 μg/ml Gentamycin, 10 mM Hepes. Fresh for each assay.
6) Tritiated Thymidine
7) Frozen human peripheral blood lymphocytes (PBL) collected via a leukophoresis procedure Plates are prepared by coating 96 well flat bottom plates with anti-CD3 antibody (Amac) or anti-CD28 antibody (Biodesign) or both in Hyclone D-PBS without calcium and magnesium. Anti - CD3 antibody is used at 10 ng/well (50μl of 200 ng/ml) and anti -CD28 antibody at 25 ng/well (50 μl of 0.5 μg/ml) in 100 μl total volume.
PBLs are isolated from human donors using standard leukophoresis methods. The cell preparations are frozen in 50% fetal bovine serum and 50% DMSO until the assay is conducted. Cells are prepared by thawing and washing PBLs in media, resuspending PBLs in 25 mis of media and incubating at 37°C, 5% CO2 overnight.
In the assay procedure, the coated plate is washed twice with PBS and the PBLs are washed and resuspended to 1 x 10" cells/ml using 100 μL /well 100 μl of a test protein or control media are added to the plate making a total volume per well of 200 μL. The plate is incubated for 72 hours. The plate is then pulsed for 6 hours with tritiated thymidine ( 1 mC/well; Amersham) and the PBLs are harvested from the plates and evaluated for uptake of the tritiated thymidine.
EXAMPLE 5 In situ Hybridization In situ hybridization is a powerful and versatile technique for the detection and localization of nucleic acid sequences within cell or tissue preparations. It may be useful, for example, to identify sites of gene expression, analyze the tissue distribution of transcription, identify and localize viral infection, follow changes in specific mRNA synthesis and aid in chromosome mapping.
In situ hybridization was performed following an optimized version of the protocol by Lu and
Gillett, Cell Vision k 169- 176 ( 1994), using PCR-generated 33P-labeled riboprobes. Briefly, formalin- fixed, paraffin-embedded human tissues were sectioned, deparaffinized, deproteinated in proteinase K (20 g/ml) for 15 minutes at 37°C, and further processed for in situ hybridization as described by Lu and Gillett, supra. A [33P] UTP-labeled antisense riboprobe was generated from a PCR product and hybridized at 55C overnight. The slides were dipped in Kodak NTB2 nuclear track emulsion and exposed for 4 weeks.
33P-Riboprobe synthesis
6.0 μl (125 mCi) of 33P-UTP (Amersham BF 1002, SA<2000 Cr/mmol) were speed vac dried. To each tube containing dried 33P-UTP, the following ingredients were added: 2.0 μl 5x transcription buffer 1.0 μl DTT (100 mM) 2.0 μl NTP mix (2.5 mM : lOμ 1; each of 10 mM GTP, CTP & ATP + lOμ 1 H20)
1.0 μl UTP (50 μM) 1 0 μl Rnasm
1 0 μl DNA template (lμg)
1 0 μl H,0 The tubes were incubated at 37°C for one hour 1 0 μL RQ1 DNase were added, followed by incubation at 37°C for 15 minutes 90 μL TE (10 mM Tns pH 7 6/lmM EDTA pH 8 0) were added, and the mixture was pipetted onto DE81 paper The remaining solution was loaded in a Microcon-50 ultrafiltration unrt, and spun using program 10 (6 minutes) The filtration unit was inverted over a second tube and spun usmg program 2 (3 minutes) After the final recovery spin, 100 μL TE were added 1 μL of the final product was pipetted on DE81 paper and counted in 6 ml of Biofluor II
The probe was run on a TBE/urea gel 1-3 μL of the probe or 5 μL of RNA Mrk III were added to 3 μL of loading buffer After heating on a 95C heat block for three minutes, the gel was immediately placed on ice The wells of gel were flushed, the sample loaded, and run at 180-250 volts for 45 minutes The gel was wrapped in saran wrap and exposed to XAR film with an intensifying screen in -70C freezer one hour to overnrght
33P-Hybndιzatιon
Pretreatment of frozen sections The slides were removed from the freezer, placed on aluminium trays and thawed at room temperature for 5 minutes The trays were placed in 55C incubator for five minutes to reduce condensation The slides were fixed for 10 minutes 4% paraformaldehyde on ice m the fume hood, and washed in 0 5 x SSC for 5 minutes, at room temperature (25 ml 20 x SSC + 975 ml SQ H2O) After deproteination in 0 5 μg/ml proteinase K for
10 minutes at 37°C (12 5μL of 10 mg/ml stock in 250 ml prewarmed RNase-free RNAse buffer), the sections were washed in 0 5 x SSC for 10 minutes at room temperature The sections were dehydrated m 70%, 95%, 100%) ethanol, 2 minutes each
Pretreatment of paraffin-embedded sections The slides were deparaffinized, placed rn SQ H2O, and nnsed twice in 2 x SSC at room temperature, for 5 minutes each time The sections were deproteinated in 20 μg/ml proteinase K (500 μL of 10 mg/ml in 250 ml RNase-free RNase buffer, 37C, 15 minutes ) - human embryo, or 8 x prote nase K (100 μL in 250 ml Rnase buffer, 37C, 30 minutes) - formalin tissues Subsequent πnsing in 0 5 x SSC and dehydration were performed as descnbed above
Prehvbridization The slrdes were laid out in plastic box lined with Box buffer (4 x
SSC, 50% formamide) - saturated filter paper The tissue was covered with 50 μL of hybπdization buffer (3 75g Dextran Sulfate + 6 ml SQ H2O), vortexed and heated rn the mrcrowave for 2 minutes with the cap loosened After cooling on ice, 18 75 ml formamrde, 3 75 ml 20 x SSC and 9 ml SQ H2O were added, the trssue was vortexed well, and incubated at 42C for 1 -4 hours Hybridization 1.0 x 106 cpm probe and 1.0 μL tRNA (50 mg/ml stock) per slide were heated at 95C for 3 minutes. The slides were cooled on ice, and 48 μL hybridization buffer were added per slide. After vortexmg, 50 μL 33P mix were added to 50 μL prehybπdization on slide. The slides were incubated overnight at 55C
Washes Washing was done 2x10 minutes with 2xSSC, EDTA at room temperature
(400 ml 20 x SSC + 16 ml 0.25M EDTA, Vf=4L), followed by RNaseA treatment at 37C for 30 minutes (500 μL of 10 mg/ml in 250 ml Rnase buffer = 20 ug/ml), The slides were washed 2x10 minutes with 2 x SSC, EDTA at room temperature The stnngency wash conditions were as follows- 2 hours at 55C, 0.1 x SSC, EDTA (20 ml 20 x SSC + 16 ml EDTA, V =4L).
DNA 35638 (1 TM receptor)
Expression was observed in the endothelium lining of a subset of fetal and placental vessels Endothelial expression was confined to these tissue blocks. Expression was also observed over intermediate trophoblast cells of placenta.
Ohgo C-120N- (SEQ ID NO: 36)
GGA TTC TAA TAC GAC TCA CTA TAG GGC TGC GGC GGC TCA GGT CTT CAG TT
Ohgo c-120P (SEQ ID NO: 37)
CTA TGA AATTAA CCC TCA CTA AAG GGA GCATGG GAT GGG GAG GGA TAC GG
DNA 33094 (EGF Homolog)
A hrghly distinctive expression pattern was observed. In the human embryo expression was obseerved in outer smooth muscle layer of the Gl tract, respiratiry cartilage, branchmg respiratory epithelium, osteoblasts, tendons, gonad, in the optic nerve head and developing dermis. In the adult, expression was observed rn the eprdermal pegs of the chrmp tongue, the basal eprthelral / myoepithe al cells of the prostate and unnary bladder. Expression was also found rn the alveolar lrmng cells of the adult lung, mesenchymal cells juxtaposed to erectrle trssue in the penis and the cerebral cortex (probably ghal cells) In the kidney, expression was only seen in disease, m cells surrounding thyroidized renal tubules
Ohgo D-200V (SEQ ID NO. 38)
CTA TGA AATTAA CCC TCA CTA AAG GGA ATA GCA GGC CAT CCC AGG ACA
Olrgo D-200Z (SEQ ID NO 39) CTATGA AATTAA CCC TCA CTA AAG GGA TGTCTT CCA TGC CAA CCTTC
EXAMPLE 6 In situ Hybridization in Cells and Diseased Tissues The in situ hybridization method of Example 5 is used to determine gene expression, analyze the tissue distribution of transcription, and follow changes in specific mRNA synthesis for the genes DNAs and the proteins of the invention in diseased tissues isolated from human individuals suffering from a specific disease. These results show more specifically where in diseased tissues the genes of the invention are expressed and are more predictive of the particular localization of the therapeutic effect of the inhibitory or stimulatory compounds of the invention (and agonists or antagonists thereof) in a disease. Hybridization is performed according to the method of Example 5 using one or more of the following tissue and cell samples:
(a) lymphocytes and antigen presenting cells (dendritic cells, langherhans cells, macrophages and monocytes, NK cells);
(b) lymphoid tissues: normal and reactive lymph node, thymus, Bronchial Associated Lymphoid Tissues, (BALT), Mucosal Associated Lymphoid Tissues (MALT);
(c) human disease tissues
• Synovium and joint of patients with Arthritis and Degenerative Joint Disease
• Colon from patients with Inflammatory Bowel Disease including Ulcerative Colitis and Crohns' disease
• Skin lesions from Psoriasis and other forms of dermatitis
• Lung tissue including BALT and tissue lymph nodes from Chronic and acute bronchitis, pneumonia, pneumonitis, pleuritis
• Lung tissue including BALT and tissue lymph nodes from Asthma
• nasal and sinus tissue from patients with rhinitis or sinusitis
• Brain and Spinal cord from Multiple Sclerosis. Alzheimer's Disease and Stroke
• Kidney from Nephritis, Glomerulonephritis and Systemic Lupus Erythematosis
• Liver from Infectious and non-infectious Hepatitis
• Tissues from Neoplasms/Cancer.
Expression is observed in one or more cell or tissue samples indicating localization of the therapeutic effect of the compounds of the invention (and agonists or antagonists thereof) in the disease associated with the cell or tissue sample.
DNA 35638 (PR0245) was found to be expressed in inflamed human tissues (psoriasis, inflammatory bowel disease (IBD), inflamed kidney, inflamed lung, hepatitis (liver block), normal tonsil, adult and chimp (multiblocks). Expression was present in the endothelium/intima of large vessels in the lung afflicted with chronic inflammation, in the superficial dermal vessels of the psoriatic skin, in arterioles in a specimen of chronic sclerosing nephritis, and in capillaries including the perifollucular sinuses of the tonsil. These results indicate that PRO 245 is immunostimulatory (enhances T lymphocyte proliferation in the MLR and costimulation) and has proinflammatory properties (induces a neutrophjil infiltrate in vivo).
EXAMPLE 7 Use of PRQ245, PRQ217, PRO301. PRQ266. PRQ335. PRQ331 or PRQ326 as a hybridization probe
The following method describes use of a nucleotide sequence encoding PR0245, PR0217, PRO301 , PR0266, PR0335, PR0331 or PR0326 as a hybridization probe.
DNA comprising the coding sequence of full-length or mature PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 (as shown in Figures 4, 6, 8, 10, 12, 14 and 16) is employed as a probe to screen for homologous DNAs (such as those encoding naturally-occurring variants of PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326) in human tissue cDNA libraries or human tissue genomic libraries.
Hybridization and washing of filters containing either library DNAs is performed under the following high stringency conditions. Hybridization of radiolabeled PR0245. PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326-derived probe to the filters is performed in a solution of 50% formamide, 5x SSC, 0.1% SDS, 0.1% sodium pyrophosphate, 50 mM sodium phosphate, pH 6.8, 2x Denhardt's solution, and 10% dextran sulfate at 42°C for 20 hours. Washing of the filters is performed in an aqueous solution of 0. lx SSC and 0.1% SDS at 42°C.
DNAs having a desired sequence identity with the DNA encoding full-length native sequence PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 can then be identified using standard techniques known in the art.
EXAMPLE 8 Expression ofPRQ245. PRQ217. PRO301. PRQ266. PRQ335. PRQ331 or PRQ326 _ E. coli This example illustrates preparation of an unglycosylated form of PR0245, PR0217,
PRO301 , PR0266, PR0335, PR0331 or PR0326 by recombinant expression in E. coli.
11 The DNA sequence encoding PR0245, PR0217, PRO301 , PRO266, PR0335, PR0331 or PR0326 is initially amplified using selected PCR primers. The primers should contain restriction enzyme sites which correspond to the restriction enzyme sites on the selected expression vector. A variety of expression vectors may be employed. An example of a suitable vector is pBR322 (derived from E. coli; see Bolivar et al, Gene, 2:95 (1977)) which contains genes for ampicillin and tetracycline resistance. The vector is digested with restriction enzyme and dephosphorylated. The PCR amplified sequences are then ligated into the vector. The vector will preferably include sequences which encode for an antibiotic resistance gene, a tip promoter, a polyhis leader (including the first six STII codons, polyhis sequence, and enterokinase cleavage site), the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 coding region, lambda transcriptional terminator, and an argU gene.
The ligation mixture is then used to transform a selected E. coli strain using the methods described in Sambrook et al, supra. Transformants are identified by their ability to grow on LB plates and antibiotic resistant colonies are then selected. Plasmid DNA can be isolated and confirmed by restriction analysis and DNA sequencing.
Selected clones can be grown overnight in liquid culture medium such as LB broth supplemented with antibiotics. The overnight culture may subsequently be used to inoculate a larger scale culture. The cells are then grown to a desired optical density, during which the expression promoter is turned on.
After culturing the cells for several more hours, the cells can be harvested by centrifiigation. The cell pellet obtained by the centrifiigation can be solubilized using various agents known in the art, and the solubilized PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 protern can then be purified using a metal chelating column under conditions that allow tight binding of the protein.
PR0245, PR0217, PRO301 and PR0266 were expressed in E. coli in a poly-His tagged form, using the following procedure. The DNA encoding PR0245, PR0217, PRO301 and PR0266 was initially amplified using selected PCR primers. The primers contained restriction enzyme sites which correspond to the restriction enzyme sites on the selected expression vector, and other useful sequences providing for efficient and reliable translation initiation, rapid purification on a metal chelation column, and proteolytic removal with enterokinase. The PCR-amplified, poly-His tagged sequences were then ligated into an expression vector, which was used to transform an E. coli host based on strain 52 (W3110 fuhA(tonA) Ion galE φoHts(htpRts) clpP(lacIq). Transformants were first grown in LB containing 50 mg/ml carbenicillin at 30C with shaking until an O.D.600 of 3-5 was reached. Cultures were then diluted 50-100 fold into CRAP media (prepared by mixing 3.57 g (NH )2S0 , 0.71 g sodium citrate.2H20, 1.07 g KC1, 5.36 g Difco yeast extract, 5.36 g Sheffield hycase SF in 500 mL water, as well as 110 mM MPOS, pH 7.3, 0.55% (w/v) glucose and 7 mM MgS04) and grown for approximately 20-30 hours at 30C with shaking. Samples were removed to verify expression by SDS-PAGE analysis, and the bulk culture is centrifuged to pellet the cells. Cell pellets were frozen until purification and refolding.
E. coli paste from 0.5 to 1 L fermentations (6-10 g pellets) was resuspended in 10 volumes (w/v) in 7 M guanidine, 20 mM Tris, pH 8 buffer. Solid sodium sulfite and sodium tetrathionate is added to make final concentrations of 0.1M and 0.02 M, respectively, and the solution was stirred overnight at 4C. This step results in a denatured protein with all cysteine residues blocked by sulfitolization. The solution was centrifuged at 40,000 φm in a Beckman Ultracentifuge for 30 min. The supernatant was diluted with 3-5 volumes of metal chelate column buffer (6 M guanidine, 20 mM Tris, pH 7.4) and filtered through 0.22 micron filters to clarify. Depending the clarified extract was loaded onto a 5 ml Qiagen Ni-NTA metal chelate column equilibrated in the metal chelate column buffer. The column was washed with additional buffer containing 50 mM imidazole (Calbiochem, Utrol grade), pH 7.4. The protein was eluted with buffer containing 250 mM imidazole. Fractions containing the desired protein were pooled and stored at 4C. Protein concentration was estimated by its absorbance at 280 nm using the calculated extinction coefficient based on its amino acid sequence.
The proteins were refolded by diluting sample slowly into freshly prepared refolding buffer consisting of: 20 mM Tris, pH 8.6, 0.3 M NaCl, 2.5 M urea, 5 mM cysteine, 20 mM glycine and 1 mM EDTA. Refolding volumes were chosen so that the final protein concentration was between 50 to 100 micrograms/ml The refolding solution was stirred gently at 4C for 12-36 hours. The refolding reaction was quenched by the addition of TFA to a final concentration of 0.4% (pH of approximately 3). Before further purification of the protein, the solution was filtered through a 0.22 micron filter and acetonitrile was added to 2-10% final concentration. The refolded protein was chromatographed on a Poros Rl/H reversed phase column using a mobile buffer of 0.1% TFA with elution with a gradient of acetonitrile from 10 to 80%. Aliquots of fractions with A280 absorbance were analyzed on SDS polyacrylamide gels and fractions containing homogeneous refolded protein were pooled. Generally, the properly refolded species of most proteins are eluted at the lowest concentrations of acetonitrile since those species are the most compact with their hydrophobic interiors shielded from interaction with the reversed phase resin. Aggregated species are usually eluted at higher acetonitrile concentrations. In addition to resolving misfolded forms of proteins from the desired form, the reversed phase step also removes endotoxin from the samples.
Fractions containing the desired folded PR0245, PR0217, PRO301 and PR0266 proteins, respectively, were pooled and the acetonitrile removed using a gentle stream of nitrogen directed at the solution. Proteins were formulated into 20 mM Hepes, pH 6.8 with 0.14 M sodium chloride and 4% mannitol by dialysis or by gel filtration using G25 Superfine (Pharmacia) resins equilibrated in the formulation buffer and sterile filtered.
EXAMPLE 9 Expression of PR0245. PRQ217. PRO301. PRQ266. PRQ335. PRQ331 or PRQ326 in mammalian cells
This example illustrates preparation of a potentially glycosylated form of PR0245, PR0217, PRO301, PR0266. PR0335, PR0331 or PR0326 by recombinant expression in mammalian cells. The vector, pRK5 (see EP 307,247, published March 15, 1989), is employed as the expression vector. Optionally, the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 DNA is ligated into pRK5 with selected restriction enzymes to allow insertion of the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 DNA using ligation methods such as described in Sambrook et al, supra. The resulting vector is called pRK5-PR0245, PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326.
In one embodiment, the selected host cells may be 293 cells. Human 293 cells (ATCC CCL 1573) are grown to confluence in tissue culture plates in medium such as DMEM supplemented with fetal calf serum and optionally, nutrient components and/or antibiotics. About 10 ug pRK5-PR0245, PR0217, PRO301. PRO266, PR0335, PR0331 or PR0326 DNA is mixed with about 1 ug DNA encoding the VA RNA gene [Thimmappaya et al, Cell. 31:543 (1982)] and dissolved in 500 uL of 1 mM Tris-HCl, 0.1 mM EDTA, 0.227 M CaCl2. To this mixture is added, dropwise, 500 uL of 50 mM HEPES (pH 7.35), 280 mM NaCl, 1.5 mM NaP04, and a precipitate is allowed to form for 10 minutes at 25°C. The precipitate is suspended and added to the 293 cells and allowed to settle for about four hours at 37°C. The culture medium is aspirated off and 2 ml of 20% glycerol in PBS is added for 30 seconds. The 293 cells are then washed with serum free medium, fresh medium is added and the cells are incubated for about 5 days.
Approximately 24 hours after the transfections, the culture medium is removed and replaced with culture medium (alone) or culture medium containing 200 uCi/ml 35S-cysteine and 200 uCi/ml 35S-methionine. After a 12 hour incubation, the conditioned medium is collected, concentrated on a spin filter, and loaded onto a 15% SDS gel. The processed gel may be dried and exposed to film for a selected period of time to reveal the presence of PR0245, PR0217, PRO301, PR0266. PR0335, PR0331 or PR0326 polypeptide. The cultures containing transfected cells may undergo further incubation (in serum free medium) and the medium is tested in selected bioassays.
In an alternative technique, PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 may be introduced into 293 cells transiently using the dextran sulfate method described by Somparyrac et al, Proc. Natl. Acad. Sci. 12:7575 (1981). 293 cells are grown to maximal density in a spinner flask and 700 ug pRK5-PR0245. PR0217. PRO301, PR0266, PR0335. PR0331 or PR0326 DNA is added The cells are first concentrated from the spinner flask by centnfugation and washed with PBS The DNA-dextran precipitate is mcubated on the cell pellet for four hours The cells are treated with 20% glycerol for 90 seconds, washed with tissue culture medium, and re- mtroduced mto the spinner flask containing tissue culture medium. 5 μg/ml bovine insulin and 0.1 ug/ml bovine transfernn After about four days, the conditroned medra rs centrrfuged and filtered to remove cells and debπs The sample contarmng expressed PR0245, PR0217. PRO301. PR0266, PR0335. PR0331 or PR0326 can then be concentrated and purified by any selected method, such as dialysis and/or column chromatography
In another embodrment, PR0245. PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 can be expressed m CHO cells The pRK5-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 can be transfected into CHO cells using known reagents such as CaP04 or DEAE-dextran. As descπbed above, the cell cultures can be mcubated, and the medium replaced with culture medium (alone) or medium containing a radiolabel such as 3:>S-methιonme After determrnmg the presence of PR0245. PR0217. PRO301. PR0266, PR0335, PR0331 or PR0326 polypeptide, the culture medium may be replaced with serum free medrum. Preferably, the cultures are mcubated for about 6 days, and then the condrtioned medium is harvested. The medrum contarmng the expressed PR0245, PR0217, PRO301. PR0266, PR0335, PR0331 or PR0326 can then be concentrated and puπfred by any selected method.
Epitope-tagged PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 may also be expressed m host CHO cells The PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 may be subcloned out of the pRK5 vector The subclone msert can undergo PCR to fuse m frame with a selected epitope tag such as a poly-his tag into a Baculovirus expression vector. The poly-his tagged PR0245. PR0217. PRO301, PR0266, PR0335. PR0331 oi PR0326 insert can then be subcloned into a SV40 dnven vector containing a selection marker such as DHFR for selection of stable clones Finally, the CHO cells can be transfected (as descnbed above) with the SV40 driven vector. Labeling may be performed, as described above, to veπfy expression The culture medium containing the expressed poly-His tagged PR0245, PR0217. PRO301. PR0266. PR0335. PR0331 or PR0326 can then be concentrated and puπfied by any selected method, such as by Nι2+-chelate affinity chromatography
PR0245, PR0217 and PRO301 were expressed m CHO cells by both a transient and a stable expression procedure
Stable expression in CHO cells was performed usmg the followmg procedure The proterns were expressed as an IgG construct (lmmunoadhesm), in which the coding sequences for the soluble forms (e.g extracellular domains) of the respective proteins were fused to an IgGl constant region sequence containing the hinge, CH2 and CH2 domains and/or is a poly-His tagged form.
Followmg PCR amplification, the respective DNAs were subcloned m a CHO expression vector using standard techniques as descnbed in Ausubel et al , Current Protocols of Molecular Biology, Umt 3 16, John Wiley and Sons (1997) CHO expression vectors are constructed to have compatible restriction sites 5' and 3' of the DNA of mterest to allow the convement shuttling of cDNAs The vector used expression in CHO cells is as descπbed in Lucas et al , Nucl Acids Res 24 9 (1774-1779 (1996), and uses the SV40 early promoter/enhancer to dnve expression of the cDNA of interest and dihydrofolate reductase (DHFR) DHFR expressron permits selection for stable maintenance of the plasmid followmg transfectron
Twelve micrograms of the desired plasmid DNA were introduced into approximately 10 million CHO cells using commercially available transfectron reagents Superfect (Quragen), Dosper or Fugene (Boehπnger Mannherm) The cells were grown and descπbed in Lucas et al , supra Approximately 3 x 10 7 cells are frozen in an ampule for further growth and production as descnbed below
The ampules containing the plasmid DNA were thawed by placement mto water bath and mixed by vortexmg The contents were pipetted into a centrifuge tube containing 10 mLs of medra and centπfuged at 1000 φm for 5 mmutes The supernatant was asprrated and the cells were resuspended rn 10 mL of selectrve media (0 2 μm filtered PS20 with 5% 0 2 μm diafiltered fetal bovme serum) The cells were then alrquoted to a 100 mL sprnner containing 90 mL of selective media After 1-2 days, the cells were transferred into a 250 mL spmner filled with 150 mL selective growth medium and incubated at 37C After another 2-3 days, a 250 mL, 500 mL and 2000 mL spinners were seeded with 3 x 105 cells/mL The cell medra was exchanged wrth fresh media by centnfugation and resuspension m production medium Although any suitable CHO media may be employed, a production medium descnbed in U S Patent No 5,122,469, issued June 16, 1992 was actually used 3L production spmner is seeded at 1 2 x 106 cells/mL On day 0, the cell number pH were determmed On day 1 , the sprnner was sampled and sparging with filtered air was commenced On day 2, the spinner was sampled, the temperature shifted to 33C, and 30 mL of 500 g/L glucose and 0 6 mL of 10% antifoam (e g , 35% polydimethylsiloxane emulsion, Dow Cornrng 365 Medrcal Grade Emulsron) Throughout the production, pH was adjusted as necessary to keep at around 7 2 After 10 days, or until viability dropped below 70%, the cell culture was harvested by centnfugtion and filtering through a 0 22 μm filter The filtrate was erther stored at 4C or rmmedrately loaded onto columns for punfrcatron
For the poly-His tagged constructs, the proteins were punfied using a Ni-NTA column (Qiagen) Before puπfrcation, imidazole was added to the conditioned media to a concentration of 5 mM The conditioned media was pumped onto a 6 ml Ni-NTA column equilrbrated in 20 mM Hepes, pH 7 4, buffer containing 0 3 M NaCl and 5 mM imidazole at a flow rate of 4-5 ml/mm at 4C After loading, the column was washed with additional equilibration buffer and the protein eluted with equilibration buffer containing 0.25 M imidazole. The highly purified protein was subsequently desalted into a storage buffer containing 10 mM Hepes, 0.14 M NaCl and 4% mannitol, pH 6.8, with a 25 ml G25 Superfine (Pharmacia) column and stored at -80C.
Immunoadhesin (Fc containing) constructs of were purified from the conditioned media as follows. The conditioned medium was pumped onto a 5 ml Protein A column (Pharmacia) which had been equilibrated in 20 mM Na phosphate buffer, pH 6.8. After loading, the column was washed extensively with equilibration buffer before elution with 100 mM citric acid, pH 3.5. The eluted protein was immediately neutralized by collecting 1 ml fractions into tubes containing 275 μL of 1 M Tris buffer, pH 9. The highly purified protein was subsequently desalted into storage buffer as described above for the poly-His tagged proteins. The homogeneity was assessed by SDS polyacrylamide gels and by N-terminal amino acid sequencing by Edman degradation.
PR0326 was also produced by transient expression in COS cells.
EXAMPLE 10 Expression of PRQ245. PRQ217. PRO301. PRQ266. PRQ335. PRQ331 or PRQ326 in Yeast
The following method describes recombinant expression of PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 in yeast.
First, yeast expression vectors are constructed for intracellular production or secretion of PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 from the ADH2/GAPDH promoter. DNA encoding PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 and the promoter is inserted into suitable restriction enzyme sites in the selected plasmid to direct intracellular expression of PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326. For secretion, DNA encoding PR0245, PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 can be cloned into the selected plasmid, together with DNA encoding the ADH2/GAPDH promoter, a native PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 signal peptide or other mammalian signal peptide, or, for example, a yeast alpha-factor or invertase secretory signal/leader sequence, and linker sequences (if needed) for expression of PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326.
Yeast cells, such as yeast strain ABI 10, can then be transformed with the expression plasmids described above and cultured in selected fermentation media. The transformed yeast supernatants can be analyzed by precipitation with 10% trichloroacetic acid and separation by SDS- PAGE, followed by staining of the gels with Coomassie Blue stain.
Recombinant PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 can subsequently be isolated and purified by removing the yeast cells from the fermentation medium by centrifugation and then concentrating the medium using selected cartridge filters. The concentrate containing PR0245, PR0217, PRO301. PR0266. PR0335. PR0331 or PR0326 may further be purified using selected column chromatography resins.
EXAMPLE 1 1 Expression of PRQ245. PRQ217, PRO301. PRQ266. PRQ335. PRQ331 or PRQ326 in Baculovirus- Infected Insect Cells
The following method describes recombinant expression of PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 in Baculovirus-infected insect cells.
The sequence coding for PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 is fused upstream of an epitope tag contained within a baculovirus expression vector. Such epitope tags include poly-his tags and immunoglobulin tags (like Fc regions of IgG). A variety of plasmids may be employed, including plasmids derived from commercially available plasmids such as pVL1393 (Novagen). Briefly, the sequence encoding PR0245, PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 or the desired portion of the coding sequence of PR0245. PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 [such as the sequence encoding the extracellular domain of a transmembrane protein or the sequence encoding the mature protein if the protein is extracellular] is amplified by PCR with primers complementary to the 5' and 3' regions. The 5' primer may incoφorate flanking (selected) restriction enzyme sites. The product is then digested with those selected restriction enzymes and subcloned into the expression vector.
Recombinant baculovirus is generated by co-transfecting the above plasmid and BaculoGold™ virus DNA (Pharmingen) into Spodoptera frugiperda ("Sf9") cells (ATCC CRL 1711) using lipofectin (commercially available from GIBCO-BRL). After 4 - 5 days of incubation at 28°C, the released viruses are harvested and used for further amplifications. Viral infection and protein expression are performed as described by O'Reilley et al, Baculovirus expression vectors: A Laboratory Manual, Oxford: Oxford University Press (1994).
Expressed poly-his tagged PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or
PR0326 can then be purified, for example, by Ni2+-chelate affinity chromatography as follows.
Extracts are prepared from recombinant virus-infected Sf9 cells as described by Rupert et al, Nature.
362: 175-179 (1993). Briefly, Sf9 cells are washed, resuspended in sonication buffer (25 mL Hepes, pH 7.9: 12.5 mM MgCl2; 0.1 mM EDTA; 10% glycerol; 0.1% NP-40; 0.4 M KC1), and sonicated twice for 20 seconds on ice. The sonicates are cleared by centrifugation, and the supernatant is diluted 50-fold in loading buffer (50 mM phosphate, 300 mM NaCl, 10% glycerol, pH 7.8) and filtered through a 0.45 um filter. A Ni2+-NTA agarose column (commercially available from Qiagen) is prepared with a bed volume of 5 mL, washed with 25 mL of water and equilibrated with 25 mL of loading buffer. The filtered cell extract is loaded onto the column at 0.5 mL per minute. The column is washed to baseline A2so with loading buffer, at which point fraction collection is started. Next, the column is washed with a secondary wash buffer (50 mM phosphate; 300 mM NaCl, 10% glycerol, pH 6.0), which elutes nonspecifically bound protein. After reaching A28o baseline again, the column is developed with a 0 to 500 mM Imidazole gradient in the secondary wash buffer. One mL fractions are collected and analyzed by SDS-PAGE and silver staining or Western blot with Ni2+-NTA- conjugated to alkaline phosphatase (Qiagen). Fractions containing the eluted Hisι0-tagged PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 are pooled and dialyzed against loading buffer.
Alternatively, purification of the IgG tagged (or Fc tagged) PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 can be performed using known chromatography techniques, including for instance, Protein A or protein G column chromatography.
PR0245, PRO301 and PR0266 were expressed in baculovirus infected Sf9 insect cells. While the expression was actually performed in a 0.5-2 L scale, it can be readily scaled up for larger (e.g. 8 L) preparations. The proteins were expressed as an IgG construct (immunoadhesin), in which the protein extracellular region was fused to an IgG 1 constant region sequence containing the hinge, CH2 and CH3 domains and or in poly-His tagged forms.
Following PCR amplification, the respective coding sequences were subcloned into a baculovirus expression vector (pb.PH.IgG for IgG fusions and pb.PH.His.c for poly-His tagged proteins), and the vector and Baculogold baculovirus DNA (Pharmingen) were co-transfected into 105 Spodoptera frugiperda ("Sf9") cells (ATCC CRL 1711), using Lipofectin (Gibco BRL). pb.PH.IgG and pb.PH.His are modifications of the commercially available baculovirus expression vector pVL1393 (Pharmingen), with modified polylinker regions to include the His or Fc tag sequences. The cells were grown in Hink's TNM-FH medium supplemented with 10% FBS (Hyclone). Cells were incubated for 5 days at 28C. The supernatant was harvested and subsequently used for the first viral amplification by infecting Sf9 cells in Hink's TNM-FH medium supplemented with 10% FBS at an approximate multiplicity of infection (MOI) of 10. Cells were incubated for 3 days at 28C. The supernatant was harvested and the expression of the constructs in the baculovirus expression vector was determined by batch binding of 1 ml of supernatant to 25 mL of Ni-NTA beads (QIAGEN) for histidine tagged proteins or Protein-A Sepharose CL-4B beads (Pharmacia) for IgG tagged proteins followed by SDS-PAGE analysis comparing to a known concentration of protein standard by Coomassie blue staining.
The first viral amplification supernatant was used to infect a spinner culture (500 ml) of Sf9 cells grown in ESF-921 medium (Expression Systems LLC) at an approximate MOI of 0.1. Cells were incubated for 3 days at 28C. The supernatant was harvested and filtered. Batch binding and SDS-PAGE analysis was repeated, as necessary, until expression of the spinner culture was confirmed.
The conditioned medium from the transfected cells (0.5 to 3 L) was harvested by centrifiigation to remove the cells and filtered through 0.22 micron filters. For the poly-His tagged constructs, the protein construct were purified using a Ni-NTA column (Qiagen). Before purification, imidazole was added to the conditioned media to a concentration of 5 mM. The conditioned media were pumped onto a 6 ml Ni-NTA column equilibrated in 20 mM Hepes, pH 7.4, buffer containing 0.3 M NaCl and 5 mM imidazole at a flow rate of 4-5 ml/min. at 4C. After loading, the column was washed with additional equilibration buffer and the protein eluted with equilibration buffer containing 0.25 M imidazole. The highly purified protein was subsequently desalted into a storage buffer containing 10 mM Hepes, 0.14 M NaCl and 4% mannitol, pH 6.8, with a 25 ml G25 Superfine (Pharmacia) column and stored at -80C.
Irnmunoadhesin (Fc containing) constructs of proteins were purified from the conditioned media as follows. The conditioned media were pumped onto a 5 ml Protein A column (Pharmacia) which had been equilibrated in 20 mM Na phosphate buffer, pH 6.8. After loading, the column was washed extensively with equilibration buffer before elution with 100 mM citric acid, pH 3.5. The eluted protein was immediately neutralized by collecting 1 ml fractions into tubes containing 275 mL of 1 M Tris buffer, pH 9. The highly purified protein was subsequently desalted into storage buffer as described above for the poly-His tagged proteins. The homogeneity of the proteins was verified by SDS polyacrylamide gel (PEG) electrophoresis and N-terminal amino acid sequencing by Edman degradation.
PR0245, PR0217, PRO301, PR0266, PR0331 and PR0326 were also expressed in baculovirus infected High-5 cells using an analogous procedure.
EXAMPLE 12 Preparation of Antibodies that Bind PRQ245. PRQ217. PRO301. PRQ266. PRQ335. PRQ331 or PRQ326
This example illustrates preparation of monoclonal antibodies which can specifically bind PR0245. PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326.
Techniques for producing the monoclonal antibodies are known in the art and are described, for instance, in Goding, supra. Immunogens that may be employed include purified PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326, fusion proteins containing PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326. and cells expressing recombinant PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 on the cell surface. Selection of the immunogen can be made by the skilled artisan without undue experimentation.
Mice, such as Balb/c, are immunized with the PR0245. PR0217. PRO301, PR0266,
PR0335, PR0331 or PR0326 immunogen emulsified in complete Freund's adjuvant and injected subcutaneously or intraperitoneally in an amount from 1-100 micrograms. Alternatively, the immunogen is emulsified in MPL-TDM adjuvant (Ribi Immunochemical Research, Hamilton, MT) and injected into the animal's hind foot pads. The immunized mice are then boosted 10 to 12 days later with additional immunogen emulsified in the selected adjuvant Thereafter, for several weeks, the mice may also be boosted with additional immunization injections Serum samples may be penodrcally obtamed from the mrce by retro-orbrtal bleedmg for testing rn ELISA assays to detect antr-PR0245. PR0217, PRO301. PR0266, PR0335, PR0331 or PR0326 antibodies.
After a suitable antibody titer has been detected, the animals "positive" for antibodies can be injected with a final intravenous injection of PR0245, PR0217. PRO301, PR0266, PR0335, PR0331 or PR0326 Three to four days later, the mice are sacrificed and the spleen cells are harvested. The spleen cells are then fused (usmg 35% polyethylene glycol) to a selected munne myeloma cell line such as P3X63AgU.l, available from ATCC, No CRL 1597 The fusions generate hybπdoma cells which can then be plated in 96 well tissue culture plates containing HAT (hypoxanthine. ammopteπn. and thymidme) medium to inhibit proliferation of non- fused cells, myeloma hybπds, and spleen cell hybnds.
The hybndoma cells will be screened m an ELISA for reactivity agamst PR0245, PR0217, PRO301, PR0266. PR0335. PR0331 or PR0326 Determination of "positive" hybndoma cells secreting the desired monoclonal antibodies agamst PR0245, PR0217, PRO301. PR0266. PR0335. PR0331 oi PR0326 is within the skill in the art.
The posrtrve hybndoma cells can be mjected intraperitoneally into syngeneic Balb/c mice to produce ascites containing the antι-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 monoclonal antibodies. Alternatively, the hybπdoma cells can be grown in tissue culture flasks or roller bottles. Puπfication of the monoclonal antibodies produced m the ascites can be accomplished using ammonium sulfate precipitation, followed by gel exclusion chromatography Alternatively, affnrty chromatography based upon bindmg of antibody to protein A or protein G can be employed
Deposit of Material
The followmg matenals have been deposrted wrth the Amencan Type Culture Collection, 10801 University Blvd , Manassas, VA 201 10-2209, USA (ATCC)
Matenal ATCC Dep. No Deposit Date
DNA40981 209439 7 November 1997
DNA37140 209489 21 November 1997
DNA41388 209927 2 June 1998
DNA35638 209265 17 September 1997
DNA37150 209401 17 October 1997 DNA33094 209256 16 September 1997
DNA32292 209258 16 September 1997
DNA32279 209259 16 September 1997
DNA40628 209432 7 November 1997
This deposit was made under the provisions of the Budapest Treaty on the International Recognrtron of the Deposrt of Microorganisms for the Puφose of Patent Procedure and the Regulatrons thereunder (Budapest Treaty) This assures maintenance of a viable culture of the deposit for 30 years from the date of deposrt The deposrt wrll be made avarlable by ATCC under the terms of the Budapest Treaty, and subject to an agreement between Genentech, Ine and ATCC, which assures permanent and unrestiicted avarlabrhty of the progeny of the culture of the deposit to the public upon issuance of the pertinent U S patent or upon laying open to the public of any U S or foreign patent application, whichever comes first, and assures avarlabrhty of the progeny to one determmed by the U S Commissioner of Patents and Trademarks to be entitled thereto according to 35 USC 122 and the Commissioner's rules pursuant thereto (including 37 CFR 1 14 with particular reference to 886 OG 638)
The assignee of the present application has agreed that if a culture of the matenals on deposit should die or be lost or destroyed when cultivated under suitable conditions, the matenals will be promptly replaced on notification with another of the same Availability of the deposited matenal is not to be construed as a license to practice the invention in contravention of the nghts granted under the authonty of any government m accordance with its patent laws
The foregoing wntten specification is considered to be sufficient to enable one skilled in the art to practrce the mventron The present mventron rs not to be lrmrted rn scope by the construct deposrted, smce the deposited embodiment is intended as a single illustration of certain aspects of the invention and any constructs that are functionally equivalent are within the scope of this mventron The deposrt of materral herein does not constitute an admission that the written descnption herem contained is inadequate to enable the practice of any aspect of the invention, including the best mode thereof, nor is it to be construed as limiting the scope of the claims to the specific illustrations that it represents Indeed, vanous modifications of the invention in addition to those shown and descnbed herein will become apparent to those skilled m the art from the foregoing descnption and fall wrthm the scope of the appended claims TABLE 4
PRO XXXXXXXXXXXXXXX (Length = 15 amino acids)
Comparison Protein XXXXXYYYYYYY (Length = 12 amino acids)
% amino acid sequence identity =
(the number of identically matching amino acid residues between the two polypeptide sequences as determined by ALIGN-2) divided by (the total number of ammo acid residues of the PRO polypeptide) =
5 divided by 15 = 33.3%
PRO XXXXXXXXXX (Length = 10 amino acids)
Comparison Protein XXXXXYYYYYYZZYZ (Length = 15 ammo acids)
% ammo acid sequence identity =
(the number of identically matching amino acid residues between the two polypeptide sequences as determined by ALIGN-2) divided by (the total number of ammo acid residues of the PRO polypeptide) =
5 divided by 10 = 50%
PRO-DNA NNNNNNNNNNNNNN (Length = 14 nucleotides)
Comparison DNA NNNNNNLLLLLLLLLL (Length = 16 nucleotides)
% nucleic acid sequence identity =
(the number of identically matching nucleotides between the two nucleic acid sequences as determined by ALIGN-2) divided by (the total number of nucleotides of the PRO-DNA nucleic acid sequence) =
6 divided by 14 = 42 9%
PRO-DNA NNNNNNNNNNNN (Length = 12 nucleotides)
Comparison DNA NNNNLLLVV (Length = 9 nucleotides)
% nucleic acid sequence identity =
(the number of identically matching nucleotides between the two nucleic acid sequences as determined by ALIGN-2) divided by (the total number of nucleotides of the PRO-DNA nucleic acid sequence) =
4 divided by 12 = 33 3% TABLE 5
* C-C increased from 12 to 15
* Z is average of EQ
* B is average of ND
* match with stop is _M; stop-stop = 0, J (joker) match = 0
# efιne M /* value of a match with a stop */ lilt _day[26][26] = {
/* A B C D E F G H I J K L M N O P Q R S T U V W X Y Z*/ /* A*/ 2.0,-2,0,0,-4, 1,-1,-1, 0,-1,-2,-1, 0,_M, 1,0,-2, 1, 1,0,0,-6,0,-3,0}, /*B*/ 0,3,-4,3,2,-5,0, 1.-2,0, 0,-3,-2, 2,_M,-1, 1,0,0,0,0,-2,-5,0,-3, 1},
/* */ {-2.-4,15.-5,-5,-4,-3,-3,-2, 0,-5,-6,-5,-4,_M,-3,-5,-4, 0,-2, 0,-2,-8, 0, 0,-5},
/* D*/ 0, 3,-5, 4, 3.-6, 1, 1,-2, 0, 0,-4,-3, 2,_M,-1, 2,-1, 0, 0, 0,-2,-7, 0,-4, 2},
/* E */ 0, 2,-5, 3, 4.-5, 0, 1,-2, 0, 0,-3,-2, 1,_M,-1, 2,-1, 0, 0, 0,-2,-7, 0,-4, 3}, /*F*/ (-4,-5,-4.-6,-5, 9,-5,-2, 1, 0,-5, 2, 0,-4, vl,-5,-5,-4,-3,-3, 0,-1, 0, 0, 7,-5}, l*G*l 1, 0,-3.1, 0,-5, 5,-2,-3, 0,-2,-4,-3, 0,_M,-l,-l,-3, 1,0, 0,-1,-7, 0,-5, 0}, /*H*/ -1, 1,-3, 1, 1,-2.-2, 6,-2, 0, 0,-2,-2, 2,_M, 0, 3, 2,-1,-1, 0,-2,-3, 0, 0, 2}, /* I */ -1.-2,-2,-2,-2, 1,-3,-2, 5, 0,-2, 2, 2,-2, vl,-2,-2,-2,-l, 0, 0, 4,-5, 0,-1,-2}, /* j */ 0, 0, 0, 0, 0.0, 0, 0, 0, 0, 0, 0, 0, 0._M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0},
/*K*/ -1, 0,-5, 0, 0.-5.-2.0,-2, 0, 5,-3, 0, 1,_M,-1, 1, 3, 0, 0, 0,-2,-3, 0,-4.0}, /* L*7 -2,-3.-6.-4,-3.2.-4.-2, 2.0,-3, 6, 4,-3,_M.-3,-2.-3,-3,-l, 0, 2,-2.0,-1,-2}. /*M */ -1.-2,-5,-3,-2, 0.-3,-2, 2, 0, 0, 4, 6,-2,Jvl,-2,-l, 0,-2,-1, 0, 2,-4, 0,-2,-1}, /*N*/ 0,2,-4,2.1,-4,0,2,-2,0, 1,-3,-2, 2,_M,-1, 1,0, 1,0,0.-2,-4.0,-2, 1}. ι*o*ι _M,_M,_M,_M,_M,_M,_M,_M,_M._M,_M,_M,_M,_M,0,_M,_M._M,_M,_M,_M,_M,_M,_M,_M,_M},
/*P*/ 1,-1,-3,-1,-1,-5,-1, 0,-2, 0,-l,-3,-2,-l,_M, 6, 0, 0, 1, 0, 0,-1,-6, 0,-5, 0}, /*Q*/ 0, 1,-5,2,2,-5,-1,3,-2,0, 1,-2,-1, 1,_M, 0,4, 1,-1,-1,0,-2,-5,0,-4,3}, /*R*/ -2, 0,-4,-1,-1,-4,-3, 2,-2, 0, 3,-3, 0, 0,Jvl, 0, 1, 6, 0,-1, 0,-2, 2, 0,-4, 0}, /*S*/ 1, 0, 0, 0, 0,-3, 1,-1,-1, 0, 0,-3,-2, 1,_M, 1,-1,0, 2, 1, 0,-1,-2, 0,-3, 0}, 1*1*1 1, 0,-2, 0, 0,-3, 0,-1, 0, 0, 0,-1,-1, 0,_M, 0,-1,-1, 1, 3, 0, 0,-5, 0,-3, 0}, ι*υ*ι 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0,_M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0}, i* v*/ 0,-2,-2,-2,-2,-1,-1,-2, 4, 0,-2, 2, 2,-2,_M,-l,-2,-2,-l, 0, 0, 4,-6, 0,-2,-2}, /*w*/ -6,-5,-8,-7,-7, 0,-7,-3,-5, 0,-3,-2.-4,-4,_M,-6,-5, 2,-2,-5, 0,-6,17, 0, 0,-6}, /*x*/ 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0,_M, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0, 0},
/* Y*/ -3,-3, 0,-4,-4, 7,-5, 0,-1, 0,-4,-1, -2,-2,_M,-5,-4,-4,-3,-3, 0,-2, 0, 0,10,-4}, ι*z*ι 0, 1,-5, 2, 3,-5, 0, 2,-2, 0, 0,-2,-1, 1,_M, 0, 3, 0, 0, 0, 0,-2,-6, 0,-4, 4} },
Page 1 of day h
TABLE 5 (cont.)
/* */
#include <stdιo h> #include <ctype h>
#defιne MAXJMP 16 /* max lumps in a diag */
#define MAXGAP 24 '* don't continue to penalize gaps larger than this */
#defιne JMPS 1024 * max jmps in an path */
#defιne MX 4 /* save if there's at least MX-1 bases since last jmp */
#define DMAT 3 /* value of matching bases */
#defϊne DMIS 0 /* penalty for mismatched bases */
#defιne DΓNSO 8 /* penalty for a gap */
#defιne DINS1 1 /* penalty per base */
#define PINSO 8 /* penalty tor a gap */
#define PI Sl 4 /* penalty per residue */ struct jmp { short n[MAXJMP], * size of jmp (neg tor dely) */ unsigned short x[MAXJMP], '* base no ot jmp in seq x */
}, /* limits seq to 2A16 -1 */ struct diag { int score. /* score at last jmp */ long offset, /* offset ot prev block */ short ymp, /* current jmp index */ struct jmp jp, /* list of jmps */
}, struct path { int spc, /* number of leading spaces */ short n[JMPS], /* size of jmp (gap) */ int x[JMPS], /* loc of jmp (last elem before gap) */
}, char *ofile, /* output file name */ char *namex[2], /* seq names getseqs() */ char *prog, /* prog name tor err msgs */ char *seqx[2], /* seqs getseqs() */ int dmax, /* best diag nw() */ int dmaxO, /* final diag */ int dna. /* set if dna maιn() */ int endgaps, /* set if penalizing end gaps */ int gapx, gapy, /* total gaps in seqs */ int lenO, len 1 , /* seq lens */ int ngapx, ngapy, /* total size ot gaps */ int smax, /* max score n () */ int *xbm, /* bitmap tor matching *' long offset, /* current offset in jmp tile */ struct diag *dx, /* holds diagonals */ struct path PP[2], /* holds path for seqs */ char *calloc(), *malloc(). *mdex(), *strcpy(), char *getseq(), *g callocl :),
Page 1 of nw h TABLE 5 (cont.)
/* Needleman-Wunsch alignment program
*
* usage progs filel file2
* where filel and fϊle2 are two dna or two protein sequences
* The sequences can be in upper- or lower-case an may contain ambiguity
* Any lines beginning with ',', '>' or '<' are ignored
* Max file length is 65535 (limited bv unsigned short x in the jmp struct)
* A sequence with 1/3 or more ot its elements ACGTU is assumed to be DNA _ * Output is in the file "align out"
* The program mav create a tmp file in /tmp to hold info about traceback
* Oπginal version developed under BSD 4 3 on a vax 8650 */
#include "nw h" #include "day h" static _dbval[26] = {
1,14,2,13,0,0,4,1 1,0,0,12,0,3,15,0,0,0,5,6,8,8,7,9,0,10,0
static _pbval[26] = {
1, 2|(1«('D'-'A'))|(1«('N,-'A')), 4, 8 16, 32, 64, 128, 256 OxFFFFFFF 1«10 1«1 1 1«12 1«13. 1«14 1«15. 1«16, 1«17, 1 «18, 1«19, 1«20, 1«21, 1«22, 1«23, 1«24, 1«25|(1«(,E'-,A'))|(1«('Q'-,A'))
Figure imgf000094_0001
fpπntf(stderr,"usage %s filel file2\n", prog), fpπntf(stderr, "where filel and file2 are two dna or two protern sequences \n"), fpπntf(stderr,"The sequences can be in upper- or lower-case\n"), fprmtf(stderr,"Any lrnes beginning with ',' or '<' are ιgnored\n"), fpπntf(stderr, "Output is in the file V'align out\"\n"), exιt(l),
} namexfO] = av[l], namex[l ] = av[2], seqx[0] = getseq(namex[0] &len0), seqx[l] = getseq(namex[l], &lenl), xbm = (dna)9 dbval _pbval, endgaps = 0, /* 1 to penalize endgaps */ ofile = "align out" /* output file */ nw(), /* fill in the matnx get the possible jmps */ readjmpsQ, /* get the actual jmps */ pπnt(), /* pπnt stats alignment */ cleanup(O), /* unlink any tmp files */
Page 1 of nw c TABLE 5 (cont.)
'* do the alignment, leturn best score maιn()
* dna values in Fitch and Smith, PNAS, 80, 1382-1386. 1983
* pro PAM 250 values
* When scores are equal, we prefer mismatches to any gap, prefer
* a new gap to extending an ongoing gap. and prefer a gap in seqx
Figure imgf000095_0001
nw( ) nw char *px, *py, /* seqs and ptrs */ int *ndely, *dely, * keep track ot dely */ int ndelx, delx. * keep track ot delx */ int *tmp, /* for swappmg rowO, row 1 */ int mis. /* score for each type */ int insO, insi, /* i nsertion penalties */ register id, /* diagonal index */ register 'J. '*jmp index */ register *col0, *coll. * score for curr, last row */ register xx, yy> /* index into seqs */ dx = (struct diag *)g_calloc("to get diags", lenO+len 1 + 1 , sizeof(struct diag)), ndely = (int *)g_calloc("to get ndelv". ienl + 1. sizeoffmt)), dely = (int *)g_calloc("to get dely". lenl+1, sizeof(int)), col0 = (int *)g_calloc("to get colO", lenl + 1. sizeof(int)), col 1 = (int *)g_calloc("to get col 1 ". len 1 + 1 , sizeof(int)), tnsO = (dna) DINSO PINSO, ins 1 = (dna)9 DINS 1 PINS1, smax = -10000, if (endgaps) { for (col0[0] = dely[0] = -insO, yy = 1 , yy <= len 1 , yy++) { colOfyy] = dely[yy] = col0[yy-l] - msl, ndelyfyy] = yy,
} col0[0] = 0, /* Waterman Bull Math Biol 84 */
} else for (yy = 1 , yy <= lenl , yy++)
Figure imgf000095_0002
'* fill in match matnx
(px = = seqx[0], xx = 1 , xx <= lenO px++ xx++) { /* mitii alize first entry in col */ if (end) gaps) { if (xx = 1) col 1 [0] = delx = -( insO+ins 1 ), else col 1 [0] = delx = col0[0] - ins 1 ndelx = xx.
} else {
Figure imgf000095_0003
ndelx = 0,
Page 2 of nw c TABLE 5 (cont.) eqx[l], yy = 1, yy <= lenl , py++, yy++) { mis = col0[yy-l], if (dna) mis += (xbm[*px-'A']&xbm[*py-,A'])9 DMAT DMIS, else mis += _day[*px-'A'][*py-'A'],
/* update penalty for del in x seq,
* favor new del over ongong del
* ignore MAXGAP if weighting endgaps */ if (endgaps || ndely[yy] < MAXGAP) { if (col0[yy] - insO >= delyfyy]) { dely[yy] = col0[yy] - (msO+insl), ndely[yy] = 1 , } else { dely[yy] -= ins 1 , ndely[yy]++, }
} else { if (col0[yy] - (msO+insl) >= dely[yy]) { dely[yy] = col0[yy] - (msO+insl ), ndely[yy] = 1
} else ndely[yy]++,
}
/* update penalty for del in y seq, * favor new del over ongong del */ if (endgaps || ndelx < MAXGAP) { if (coll[yy-l] - msO >= delx) { delx = col 1 [yy- 1 ] - (insO+ins 1 ), ndelx = 1 , } else { delx — insi, ndelx++,
} else { if (coll[yy-l] - (msO+insl ) >= delx) { delx = col 1 [yy- 1 ] - (insO+ins 1 ), ndelx = 1 ,
} else ndelx++,
/* pick the maximum score, we're favoπng * mis over anv del and delx over dely
*/
Page 3 of nw c
TABLE 5 (cont.)
id = xx - yy + lenl - 1 , if (mis >= delx && mis >= dely[yy]) coll [yy] = mιs, else if (delx >= dely[yy]) { coll[yy] = delx,
IJ = dx[ιd] ljmp, if (dx[ιd] jp n[0] && ('dna || (ndelx >= MAXJMP && xx > dx[ιd] jp x[ιj]+MX) || mis > dx[ιd] score+DINSO)) { dx[ιd] ιjmρ++. if (++ιj >= MAXJMP) { wπtejmps(ιd), ij = dx[ιd] ijmp = 0, dx[ιd] offset = offset, offset += sizeof(struct jmp) + sizeof(offset), } } dx[ιd] jp n[ιj] = ndelx, dx[ιd] jp x[ιj] = xx, dx[ιd] score = delx,
} else { coll[yy] = dely[yy], ij = dx[ιd] ijmp, if (dx[ιd] jp n[0] && ('dna || (ndely[yy] >= MAXJMP
&& xx > dx[ιd] jp x[ιj]+MX) || mis > dx[ιd] score+DINSO)) { dx[ιd] ιjmp++, if (++ιj >= MAXJMP) { wπtejmps(ιd), ij = dx[ιd] ymp = 0, dx[ιd] offset = offset, offset += sizeof(struct jmp) + sizeof(offset), } } dx[ιd]jp n[y] = -ndely[yy], dx[ιd]jp x[ιj] = xx, dx[ιd] score = dely[yy],
} if (xx = lenO && yy < lenl) { /* last col */ if (endgaps) col 1 [yy] -= msO+ins 1 *(len 1 -yy ), if(coll[yy] > smax) { smax = coll [yy], dmax = id, } } } if (endgaps &<&. xx < lenO) coll [yy-l ] -= msO+ιnsl *(lenO-xx), if (col 1 [yy- 1 ] > smax) { smax = coll[yy-l ], dmax = id,
} tmp = colO, colO = col 1 , col 1 = tmp,
}
(void) free((char *)ndely),
(void) free((char *)dely),
(void) free((char *)col0),
(void) iτee((char *)col 1 ), } Page 4 ot nw c TABLE 5 (cont.)
' pπnt() - only routine visible outside this module
' static
* getmat( ) - trace back best path, count matches pnnt()
* pr_align() - pπnt alignment of descπbed in array p[] pπnt()
* dumpblockO ~ dump a block of lines with numbers, stars pr_alιgn()
* nums( ) — put out a number line dumpblock( )
* putlmeO - put out a line (name, [num], seq, [num]) dumpblockO
* stars() - -put a line of stars dumpblockO
* stnpnameO - stπp any path and prefix from a seqname */
#include ' 'nw h"
#defme SPC 3 #defιne P LINE 256 /* maximum output line */ #defιne P SPC 3 /* space between name or num and seq */ extern _day[26][26], int olen, /* set output line length */
FILE *fx, /* output file *' pπnt() print { int llxx,, 1l'y, firstgap, lastgap, /* overlap */ if ((fx = fopen(ofile, "w")) = 0) { fpπntf(stderr,"%s can't wπte %s\n", prog, ofile), cleanup(l),
} fprmtf(fx, "<first sequence %s (length = %d)\n", namex[0], lenO), fpπntf(fx, "<second sequence %s (length = %d)\n", namex[l], lenl), olen = 60, lx = lenO, ly = lenl, firstgap = lastgap = 0, if (dmax < len 1 - 1 ) { /* leading gap m x */ pp[0] spc = firstgap = lenl - dmax - 1 , ly -= pp[0] spc,
} else if (dmax > lenl - 1) { '* leading gap m y *' pp[l] spc = firstgap = dmax - (lenl - 1 ), lx -= pp[l ] spc, } if (dmaxO < lenO - 1 ) { /* trailing gap in x *' lastgap = lenO - dmaxO - 1. lx — lastgap,
} else if (dmaxO > lenO - 1 ) { /* trailing gap in v *' lastgap = dmaxO - (lenO - 1 ), ly — lastgap,
} getmat(lx. ly, firstgap, lastgap), pr_ahgn().
Page 1 of nwpπnt c TABLE 5 (cont.)
/*
* trace back the best path, count matches
*/ static getmat(lx, ly, firstgap, lastgap) gctmat int lx, ly, /* "core" (minus endgaps) */ int firstgap, lastgap. /* leadmg trailing overlap */ int nm, IO. ii, sizO, sizl . char outx[32], double pet, register nO. nl, register char *p0, *pl.
/* get total matches, score */
Figure imgf000099_0001
else { if(xbm[*pO-'A,]&xbm[*pl-'A']) nm++, if (nO++ = pp[0] x[ιO]) sιzO = ρp[0] n[ιO++],
Figure imgf000099_0002
}
/* pet homology
* if penalizing endgaps, base is the shorter seq
* else, knock off overhangs and take shorter core */ if (endgaps)
Ix = (len0 < lenl) len0 lenl, else lx = (lx < ly)9 lx ly, pct = 100 *(doubie)nm (doubϊe)lx, tpπntf(fx, "\n"), tpπntf(fx, "<%d match%s in an overlap ot %d % 2f percent snnιlaπty\n" nm, (nm == 1 )9 "" "es", lx, pet).
Page 2 ot nwpπnt c TABLE 5 (cont.) fpπntf(fx "<gaps in first sequence %d", gapx), ...getmat if (gapx) {
(void) spπntt(outx. " (°/od %s%s)", ngapx, (dna)9 "base" "residue", (ngapx == I ) ' "" "s"). tpπntf(tx,"%s", outx), fpnnttf fx, ", gaps in second sequence %d", gapy), if (gapy) {
(void) spπntr(outx, " (%d %s%s)", ngapy, (dna)9 "base" "residue", (ngapy == 1 )9 "" "s"), fpπntf(fx,"%s". outx), if (dna) fpπntf(fx,
"\n<scoιe %d (match = %d, mismatch = %d, gap penalty = %d + %d per base)\n" smax, DMAT, DMIS, DINSO, DINS1), else fpπntt(fx,
"\n<score %d (Dayholf PAM 250 matnx, gap penalty = %d + %d per resιdue)\n" smax, PINSO, PINS 1 ), if (endgaps) fpnntf(fx,
"<endgaps penalized left endgap %d %s%s, right endgap %d %s%s\n" firstgap, (dna)9 "base" "lesidue". (firstgap == 1 )9 "" "s", lastgap, (dna)9 "base" "residue", (lastgap = 1 )9 "" "s"), else fpnntf(fx, "<endgaps not penahzed\n"),
static nm, /* matches in core — for checking */ static lmax. /* lengths ot stnpped file names */ static ιj[2], /* jmp index for a path */ static nc[2], /* number at start ot current line */ static nι[2], /* current elem number — for gapping */ static sιz[2], static char *ps[2], /* ptr to current element */ static char *po[2], /* ptr to next output char slot */ static char out[2][P LINE] /* output line */ static char star[P LINE], /* set by stars() */
/*
* print alignment of descnbed in struct path pp[] */ static pι_alιgn() pr align { int nn, /* char count *' int more, register I, for (ι = 0 lmax = 0, i < 2, ι++) { nn = stnpname(namex[ι]), if (nn > lmax) lmax = nn, nc[ι] = 1 , n.[.] = l , sιz[ι] = ιj[ι] = 0, ps[ι] = seqx[ι], po[ι] = out[ι],
} Page 3 of nwpnnt c TABLE 5 (cont.)
for (nn = nm = 0, more = 1 more, ) { ...pr aiign for (I = more = 0 K 2, ι++) {
/* * do we have more of this sequence9
*/ if ('*ps[ι]) continue, more++. if (pp[ι] spc) { /* leading space */
*po[ι]++ = pp[ι] spc-,
} else if (sιz[ι]) { /* in a gap */
*po[ι]- sιz[ι]- else { /* we're putting a seq element
*/ *po[ι] = *ps[ι], if (ιslower(*ps[ι]))
*ps[ι] = toupper(*ps[ι]), po[ι]++, ps[ι]++
/*
* are we at next gap for this seq9
*/ if (nι[ι] = pp[ι] x[ij[ι]]) { /*
* we need to merge all gaps
* at this location */ sιz[ι] = pp[ι] n[ιj[ι]++], while (m[ι] = ppfi] x[ιj[ι]]) sιz[ι] += pp[ι] n[ιj[ι]+
} nι[ι]++,
}
} if (++nn = olen || 'more && nn) { dumpblockO, for (l = 0 ι < 2, ι++) po[ι] = out[ι], nn = 0,
}
/*
* dump a block of lines, including numbers, stars pr_alιgn()
*/ static dumpblockO dumpblock register l, for (l = 0, K 2. ι++)
*po[ι]- = W,
Page 4 ofnwpnnt c TABLE 5 (cont.)
...dumpblock
(void) putc('\n', fx), for (i = 0, K 2, ι++) { if (*out[ι] && (*out[ι] '= " || *(po[ι]) ι= ' ')) { if (. = 0) nums(ι), if (ι==0 &.&*out[l]) stars(), puthne(ι) if(ι==0&&*out[l]) fpπntf(fx, star), if (ι==l) nums(ι), } }
* put out a number line dumpblockO
*/ static nums(ιx) nums int tx, /* index in out[]
! char nlιne[P_LINE], register i.J, register char *pn, *px, *py, for (pn = nlme, l = 0, l < lmax+P_SPC, ι++, pn++)
*pn = ' ', for (I = nc[ιx], py = out[tx], *py, py++, pn++) { if (*py = " II *py =='-')
*pn = ' ', else { if(ι%10 = 0||(ι= 1 &&nc[ιx]'=l)){ j = (ι<0) -ι l, for(px = pn,j,j /= 10, px-)
*px=j%10 + '0', if 0 < 0)
*px = '-', else
*pn =
} }
*pn = \0', nc[ιx] = l, for (pn = nline, *pn, pn++)
(void)putc(*pn, fx), (void) putc('\n', fx),
/*
* put out a line (name, [num], seq, [num]) dumpblockO
*/ static puthne(ιx) putline
Page 5 ot nwpπnt c TABLE 5 (cont.)
...putline int i, register char *px, for (px = namex[ιx], i = 0, *px && *px '= ' '. px++, ι++)
(void) putc(*px, fx), for (, l < lmax+P SPC, ι++)
(void) putc(' ', fx),
/* these count from 1
* m[] is current element (from 1 )
* nc[] is number at start of current line */ for (px = out[ιx], *px, px++)
(void) putc(*px&0x7F, fx). (void) putc('\n', fx),
' put a line of stars (seqs always in out[0], out[ 1 ]) dumpblockO
*/ static starsO stars i int 1, register char *p0, *pl, cx, *p\. if C*out[0] || (*out[0] = " && *(po[0]) == ' ') || ι*out[l] || (*ouf[l] == " && *(po[ l]) = ' ')) return, px = star, for (I = lmax+P_SPC, t, ι-)
*px++ = ' ', for (pO = out[0], pi = out[l], *p0 && *pl , p0++, pl++) { if(ιsalpha(*pO) && ιsalpha(*pl)) { if (xbm[*pO-'A']&xbm[*pl-'A']) { ex = '*', nm++, } else if ('dna <£.& _dav[*pO-'A'][*pl-'A'] > 0) ex = ' ' else ex = ' '
} else ex = ' ', *px++ = ex,
}
*px++ = V, *px = \0',
Page 6 of nwpπnt c TABLE 5 (cont.)
/*
* stπp path or prefix from pn, return len pr_ahgn()
*/ static stnpname(pn) stripname char *pn, /* file name (may be path) */
{ register char *px, *py, py = 0, for (px = pn, *px, px++) if (*px = '/') py = px + 1, if (py)
(void) strcpy(pn, py), return(strlen(pn)),
Page 7 of nwpnnt c
TABLE 5 (cont.)
/*
* cleanup!) - cleanup any tmp tile
* getseqO — read in seq. set dna. len. maxlen
* g_calloc() — callocO with error checkin
* read|mps() - get the good jmps. tiom tmp file if necessary
* wntejmps() — write a filled array ot jmps to a tmp file nw() */
#include "nw h" #include <sys/file h> char *jname = "'tmp/homgXXXXXX" /* tmp file tor jmps */
FILE *fj. int cleanupO, /* cleanup tmp file */ long lseek(),
/*
* remove any tmp file if we blow */ cleanup(ι) cleanup int l, ϊ if (fj)
(void) unlιnk(jname). exιt(ι).
/*
* read, return ptr to seq. set dna, len. maxlen
* skip lines starting with ',', '<', or '>'
* seq in upper or lower case */ char * getseq(file, len) getseq char *file, /* file name */ int *len, /* seq len */ char hne[1024], *pseq. register char *px, *py, int natgc, tlen,
FILE *Φ. if ((fp = fopen(file,"r")) = 0) { tpnntf(stderr,"%s can't read %s\n", prog, tile). exιt( 1 ),
} tlen = natgc = 0, while (fgets(hne, 1024, fp)) { if (*hne == ',' || *hne = '<' || *lιne == '>') continue, for (px = line, *px '= '\n', px++) if (ιsupper(*px) || ιslower(*px)) tlen++,
} if ((pseq = malloc((unsigned)(tlen+6))) = 0) { fpnntf(stderr."%s malloc() failed to get %d bytes for %s\n", piog, tlen+6, file), exιt( 1 ),
} pseq[0] = pseq[ 1 ] = pseq[2] = pseq[3] = Λ0',
Pase 1 ot nwsubr c TABLE 5 (cont.)
..getseq py = pseq + 4, *len = tlen, rewιnd(fp), while (fgets(lιne, 1024, fp)) { if (*lιne == ',' || *hne = '<' || *lιne = '>') continue for (px = line, *px '= '\n', px++) { if (ιsupper(*px))
*py++ = *px, else if (ιslower(*px))
*py++ = toupper(*px), if (ιndex( "ATGCU",*(py- 1 ))) natgc++, } }
*py++ = '\0', *py = W, (void) fclose(fp), dna = natgc > (tlen/3), return(pseq+4).
} char * g_calloc(msg, nx, sz) g calloc char *msg, /* program, calling routine */ int nx, sz, /* number and size of elements */
{ char *px, *calloc(). if ((px = calloc((unsigned)nx, (unsigned)sz)) = 0) { if (*msg) { φnntf(stderr, "%s g_calloc() failed %s (n=%d, sz=%d)\n", prog, msg, nx, sz), exιt(l), } } return(px),
: get final jmps from dx[] or tmp file, set pp[], reset dmax mam() readjmps( ) readjmps { int fd = -l ,
Figure imgf000106_0001
register ι,j, xx, if (fj) {
(void) fclose(fj), if ((fd = open name, O RDONLY, 0)) < 0) { φnntf(stderr, "%s can't open() %s\n", prog, jname), cleanup( 1 ),
} for (I = iO = 11 = 0, dmaxO = dmax, xx = lenO, , ι++) { while (1) { for (j = dx[dmax] ιjmp,j >= 0 && dxfdmax] jp x[j] >= xx,j-)
Page 2 of nwsubr c TABLE 5 (cont.)
...readjmps if (j < 0 &.& dx[dmax] offset && fj) {
(void) lseekltd. dx[dmax] offset, 0),
(void) read(td (char *)< dx[dmax] jp, sizeof(struct jmp)),
(void) read(td. (char *)<idx[dmax] offset, sιzeof(dx[dmax] offset)), dx[dmax] ιjmp = MAXJMP- 1 ,
} else break. if(ι >= JMPS) { fpπntf(stderr, "%s too many gaps in alignments", prog), cleanup(l), } if
* gap m second seq */ = -sιz,
Figure imgf000107_0001
yy + len 1 - 1 */ pp[l] x[ιl] = xx - dmax + lenl - 1 , gapy++ ngapy -= siz, /* ignore MAXGAP when doing endgaps */ siz = (-siz < MAXGAP || endgaps)9 -siz MAXGAP, ιl++,
} else if (siz > 0) { /* gap in first seq */
Figure imgf000107_0002
ngapx += siz, /* ignore MAXGAP when doing endgaps */ siz = (siz < MAXGAP || endgaps)9 siz MAXGAP, ι0++, } } else break, }
/* reverse the order of jmps */ for (j = 0, ι0~, j < iO, j++, iO-) { i = pp[0] n[|], pp[0] n[)] = pp[0] n[ι0], pp[0] n[ι0] = i, i = PP[0] ϋl. PPfO] xLl] = PPtO] x[ι0], pp[0] x[ι0] = i,
} for (j = 0, ιl-,j < ιl, j++, ιl-) { i = pp[l] n[)], pp[l] n ] = pp[l] n[ιl], pp[l] n[ιl] = i, i = pp[l] x|j], pp[l] x[j] = pp[l] x[ιl], pp[l] x[ιl] = t,
} if (fd >= 0)
(void) close(fd), if (fj) {
(void) unlinktjname), fj = o, offset = 0, }
Page 3 ot nwsubr c TABLE 5 (cont.)
/*
* wπte a filled jmp struct offset of the prev one (if any) nw()
*/ wπtejmps(ιx) writejmps
Figure imgf000108_0001
char *mktemp(), if ('fj) { if (mktemp(jname) < 0) { φnntf(stderr, "%s can't mktempO %s\n". prog, jname), cleanup(l ), } if ((fj = fopenljname, "w")) == 0) { φπntf(stderr, "%s can't wπte %s\n", prog, jname), exιt(l), } }
(void) fwπte((char *)&dx[ιx] jp, sizeof(struct jmp), 1, fj), (void) fwπte((char *)&dx[ιx] offset. sizeof(dx[ιx] offset), 1, fj),
TABLE 6
MARRSRHR LLLLLRYLVVALGYHKAYGFSAPKDQQλΛ/TAVEYQEAIJACKTPKKTVSS R EWKKLGRSVSFVYYQQTLQGDFK RAEMIDFNIRIKNVTRSDAGKYRCEVSAPSEQG QNLEEDTVTLEVLVAPAVPSCEVPSSALSGTWELRCQDKEGNPAPEYT FKDGIRLLE
NPR GSQSTNSSYTMNTKTGTLQFNTVSKLDTGEYSCEARNSVGYRRCPGKRMQVDDLN ISGIIAAVVWALVISVCG GVCYAQRKGYFSKETΞFQKSNSSSKATTMSENVQWLTPV IPALWKAAAGGSRGQEF
N-glycosylation siteat residues:
98-102
187-191
236-240
277-281
Casein kinase II phosphorylation siteat residues: 39-43 59-63 100-104 149-153
205-209 284-288
N-myristoylation siteat residues: 182-188
239-245 255-261 257-263 305-311
Amidation site at residues: 226-230 TABLE 7
MARRSAFPAAAL SI LCLLALRAEAGPPQEESLYLWIDAHQARV IGFEEDILIVS EGK^ .PFTHDFRKAQQR PAIPVNIHSMNFTWQ AGQAEYFYEFLS RSLDKGIMADPT V7>JVPLLGTΛ7PHKASVVQVGFPC GKQDGVAAFEVOVI'τMNSEGNTILQTPQNAIFFKTC QQAECPGGCRNGGFCNERRICECPDGFHGPHCEKA CTPRCMNGGLCVTPGFCICPPGF YGVNCDKANCSTTCFNGGTCFYPGKCICPPG EGEQCEISKCPQPCRNGGKCIGKSKCK CSKGYQGD CSKPVCEPGCGAHGTCHEPNKCQCQEG HGRHCNKRYEASLIHALRPAGA QLRQHTPS KKAEERRDPPESNYIW
N-glycosylation site at residues: 88-92 245-249
Casein kinase II phosphorylation site at residues:
319-323
Tyrosine kinase phosphorylation site at residues: 370-378
N-myristoylation site at residues:
184-190
185-191
189-195 315-321
ATP/GTP-binding site motif A (P-loop) at residues: 285-293
EGF-like domain cysteine pattern signature at residues:
198-210
230-242
262-274
294-306 326-338 TABLE 8
MGTKAQVERK LCLFILAIL CSLALGSVTVHSSEPEVRIPENNPVK SCAYSGFSSPR VE KFDQGDTTRLVCYNNKITASYEDRVTF PTGITFKSVTREDTGTYTCMVSEEGGNS YGEVKVK IV VPPSKPTVNIPΞSATIGNRAVLTCSEQDGSPPSEYT FKDGIVMPTNP
KSTRAFSNSSYVLNPTTGELVFDPLSASDTGEYSCEARNGYGTPMTSNAVRMEAVERNV GVIVAAVLVTLILLGILVFGIWFAYSRGHFDRTKKGTSSKKVIYSQPSARSEGEFKQTS SF V
N-glycosylation site at residues:
185-189
cAMP- and cGMP-dependent protein kinase phosphorylation site at residues : 270-274
Casein kinase II phosphorylation site at residues: 34-38 82-86 100-108
118-122 152-156 154-158 193-197 203-207
287-291
N-myristoylation site at residues: 105-111 116-122
158-164 219-225 237-243 256-262 TABLE 9
M WILL ETSLCFAAGNVTGDVCKEKICSCNEIEGDLHVDCE KGFTSLQRFTAPTSQ
FYHLFLHGNS LTRLFPNE F ANFYNAVS LHMENNGLHE I VPGAFLG QLVKRLH INN KI KSFRKQTFLGLDDLEY QADFNLLRDIDPGAFQDLNKLEVLILNDNLISTLPANVFQYV
PITHLDLRGNRLKTLPYEEVLEQIPGIAEI LEDNP DCTCDL SLKE LENIPK ALI
GRWCEAPTRLQGKDLNETTEQDLCP KNRVDSSLPAPPAQEETFAPGPLPTPFKTNGQ
EDHATPGSAPNGGTKIPGNWQIKIRPTAAIATGSSRNKPLANSLPCPGGCSCDHIPGSG KV1NCNNRNVSSI-ADLKPKLSIWQELFLRDNKIHSIRKSHFVDYKNLILLD GNNNIAT VEIMNTFKNLLD RWLYMDSNYLDTLSREKFAGLQN EYLNVEYNAIQLILPGTFNAMPK
LRILIL NN LRSLPVTJVFAGVSLSKLSLHNNYFMYLPVAGVLDQLTSIIQIDLHGNP
ECSCTIVPFKQ AERLGSEVLMSDLKCETPVNFFRKDFMLLSNDEICPQLYARISPT T
SHSK ΞTGLAETGTHSNSYLDTSRVSISVLVPGLL VFVTSAFTWGMLVFILR RKRS
KRRDANSSASEINSLQTVCDSSYWHNGPYNADGAHRVYDCGSHS SD
N-glycosylation site at residues: 18-22 253-257 363-367 416-420
595-599 655-659
cAMP- and cGMP-dependent protein kinase phosphorylation site at residues:
122-126 646-650
Casein kinase II phosphorylation site at residues: 30-34
180-184
222-226
256-260
366-370 573-577
608-612
657-661
666-670 693 -697
N-myristoylation site at residues: 17-23 67-73
100-106 302-308 328-334 343-349 354-360
465-471 493-499 598-604 603-609
Prokaryotic membrane lipoprotein lipid attachment site at residues :
337-348
TABLE 10
MVDVLL FSLC LFHISRPDLSHNRLSFIKASSMSH QS REVKLNNNELETIPN GPV
SANITLLS AGNRIVEILPEHLKEFQSLET DLSSNNISELQTAFPA QLKYLYLNSNR
VTSMEPGYFDN ANTLLVLKLNRNRISAIPPKMFKLPQ QHLELNRNKIKNVDGLTFQG LGALKSLKMQRNGVTKLMDGAFWGLSNMEILQLDHNNLTEITKGWLYGLLMLQELHLSQ
NAINRISPDAWEFCQKLSELDLTFNHLSRLDDSSFLGLSLLNTLHIGNNRVSYIADCAF
RGLSSLKTLDLKNNEISWTIEDMNGAFSGLDKLRRLILQGNRIRSITKKAFTGLDALEH
LDLSDNAIMSLQGNAFSQMKKLQQLHLNTSSLLCDCQLKWLPQWVAENNFQSFVNASCA
HPQLLKGRSIFAVSPDGFVCDDFPKPQITVQPETQSAIKGSNLSFICSAASSSDSPMTF AWKKDNELLHDAEMENYAHLRAQGGEVMEYTTI RLREVEFASEGKYQCVISNHFGSSY
SVKAKLTVNMLPSFTKTPMDLTIRAGAMARLECAAVGHPAPQIAWQKDGGTDFPAARER
RMHVMPEDDVFFIVDVKIEDIGVYSCTAQNSAGSISANATLTVLETPSFLRPLLDRTVT
KGETAVLQCIAGGSPPPKLNWTKDDSPLWTERHFFAAGNQLLIIVDSDVSDAGKYTCE
MSNTLGTERGNVRLSVIPTPTCDSPQMTAPSLDDDGWATVGWIIAWCCWGTSLVWV VIIYHTRRRNEDCSITNTDETNLPADIPSYLSSQGTLADRQDGYVSSESGSHHQFVTSS
GAGFFLPQHDSSGTCHIDNSSEADVEAATDLFLCPFLGSTGPMYLKGNVYGSDPFETYH
TGCSPDPRTVLMDHYEPSYIKKKECYPCSHPSEESCERSFSNISWPSHVRKLLNTSYSH
NEGPGMKNLCLNKSSLDFSANPEPASVASSNSFMGTFGKALRRPHLDAYSSFGQPSDCQ
PRAFYLKAHSSPDLDSGSEEDGKERTDFQEENHICTFKQTLENYRTPNFQSYDLDT
N-glycosylation site at residues: 62-66 96-100 214-218 382-386
409-413 455-459 628-632 669-673 845-849
927-931 939-943 956-960
Glycosaminoglycan attachment site at residues:
826-830
Casein kinase II phosphorylation site at residues: 17-21 39-43
120-124
203-207
254-258 264-268
314-318 323-327 347-351 464-468 548-552
632-636 649-653 671-675 739-743 783-787
803-807 847-851 943-947 958-962 1013-1017
1019-1023 1021-1025
Tyrosine kinase phosphorylation site at residues: 607-615
N-myristoylation site at residues:
179-185
197-203 320-326
367-373
453-459
528-534
612-618 623-629
714-720
873-879 TABLE 11
MLNKMTLHPQQIMIGPRFNRALFDPLLWLLALQLLWAGLVRAQTCPSVCSCSNQFSK VICVRKNLREVPDGISTNTRLLNLHENQIQIIKVNSFKHLRHLEILQLSRNHIRTIEIG AFNGLANLNTLELFDNRLTTIPNGAFVYLSKLKELWLRNNPIESIPSYAFNRIPSLRRL
DLGELKRLSYISEGAFEGLSNLRYLNLAMCNLREIPNLTPLIKLDELDLSGNHLSAIRP GSFQGLMHLQKLWMIQSQIQVIERNAFDNLQSLVEINLAHNNLTLLPHDLFTPLHHLER IHLHHNPWNCNCDILWLSWWIKDMAPSNTACCARCNTPPNLKGRYIGELDQNYFTCYAP VIVEPPADLNVTEGMAAELKCRASTSLTSVSWITPNGTVMTHGAYKVRIAVLSDGTLNF TNVTVQDTGMYTCMVSNSVGNTTASATLNVTAATTTPFSYFSTVTVETMEPSQDEARTT
DNNVGPTPWDWETTNVTTSLTPQSTRSTEKTFTIPVTDINSGIPGIDEVMKTTKIIIG CFVAITLMAAVMLVIFYKMRKQHHRQNHHAPTRTVEIINVDDEITGDTPMESHLPMPAI EHEHLNHYNSYKSPFNHTTTVNTINSIHSSVHEPLLIRMNSKDNVQETQI
N-glycosylation site at residues:
278-282
364-368
390-394
412-416 415-419
434-438
442-446
488-492
606-610
cAMP- and cGMP-dependent protein kinase phosphorylation site at residues :
183-187
Casein kinase II phosphorylation site at residues:
268-272
417-421
465-469
579-583 620-624
N-myristoylation site at residues: 40-46 73-79 118-124 191-197 228-234 237-243 391-397 422-428 433-439 531-537
TABLE 12
MSAPSLRARAAGLGLLLCAVLGRAGRSDSGGRGELGQPSGVAAERPCPTTCRCLGDLLD CSRKRLARLPEPLPSWVARLDLSHNRLSFIKASSMSHLQSLREVKLNNNELETIPNLGP VSANITLLSLAGNRIVEILPEHLKEFQSLETLDLSSNNISELQTAFPALQLKYLYLNSN
RVTSMEPGYFDNLANTLLVLKLNRNRISAIPPKMFKLPQLQHLELNRNKIKNVDGLTFQ GLGALKSLKMQRNGVTKLMDGAFWGLSNMEILQLDHNNLTEITKGWLYGLLMLQELHLS QNAINRISPDAWEFCQKLSELDLTFNHLSRLDDSSFLGLSLLNTLHIGNNRVSYIADCA FRGLSSLKTLDLKNNEISWTIEDMNGAFSGLDKLRRLILQGNRIRSITKKAFTGLDALE HLDLSDNAIMSLQGNAFSQMKKLQQLHLNTSSLLCDCQLKWLPQWVAENNFQSFVNASC
AHPQLLKGRSIFAVSPDGFVCDDFPKPQITVQPETQSAIKGSNLSFICSAASSSDSPMT FAWKKDNELLHDAEMENYAHLRAQGGEVMEYTTILRLREVEFASEGKYQCVISNHFGSS YSVKAKLTVNMLPSFTKTPMDLTIRAGAMARLECAAVGHPAPQIAWQKDGGTDFPAARE RRMHVMPEDDVFFIVDVKIEDIGVYSCTAQNSAGSISANATLTVLETPSFLRPLLDRTV TKGETAVLQCIAGGSPPPKLNWTKDDSPLWTERHFFAAGNQLLIIVDSDVSDAGKYTC
EMSNTLGTERGNVRLSVIPTPTCDSPQMTAPSLDDDGWATVGWIIAWCCWGTSLVW WIIYHTRRRNEDCSITNTDETNLPADIPSYLSSQGTLADRQDGYVSSESGSHHQFVTS SGAGFFLPQHDSSGTCHIDNSSEADVEAATDLFLCPFLGSTGPMYLKGNVYGSDPFETY HTGCSPDPRTVLMDHYEPSYIKKKECYPCSHPSEESCERSFSNISWPSHVRKLLNTSYS HNEGPGMKNLCLNKSSLDFSANPEPASVASSNSFMGTFGKALRRPHLDAYSSFGQPSDC
QPRAFYLKAHSSPDLDSGSEEDGKERTDFQEENHICTFKQTLENYRTPNFQSYDLDT
N-glycosylation site at residues:
122-126 156-160
274-278
442-446
469-473
515-519 688-692
729-733
905-909
987-991
999-1003 1016-1020
Glycosaminoglycan attachment site at residues: 886-890 Casein kinase II phosphorylation site at residues: 99-103 180-184 263-267
314-318 324-328 374-378 383-387 407-411
524-528 608-612 692-696 709-713 731-735
799-803 843-847 863-867 907-911 1003-1007
1018-1022 1073-1077 1079-1083 1081-1085
Tyrosine kinase phosphorylation site at residue: 667-675
N-myristoylation site at resudues: 14-20
36-42
239-245
257-263
380-386 427-433
513-519 588-594 672-678 683-689 774-780 933-999
Leucine zipper pattern at residues: 58-80 65-87
.18

Claims

What is claimed:
1. A composition, comprising a PR0245, PR0217. PRO301, PR0266, PR0335. PR0331 or PR0326 polypeptide, agonist or fragment thereof and a carrier or excipient, useful for:
(a) increasing infiltration of inflammatory cells into a tissue of a mammal in need thereof,
(b) stimulating or enhancing an immune response in a mammal in need thereof, or
(c) increasing the proliferation of T-lymphocytes in a mammal in need thereof in response to an antigen.
2. Use of a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide, agonist or a fragment thereof to prepare a composition useful for:
(a) increasing infiltration of inflammatory cells into a tissue of a mammal in need thereof,
(b) stimulating or enhancing an immune response in a mammal in need thereof, or
(c) increasing the proliferation of T-lymphocytes in a mammal in need thereof in response to an antigen.
3. A composition, comprising a PR0245, PR0217, PRO301 , PR0266, PR0335, PR0331 or PR0326 polypeptide, antagonist or a fragment thereof and a carrier or excipient, useful for:
(a) decreasing infiltration of inflammatory cells into a tissue of a mammal in need thereof,
(b) inhibiting or reducing an immune response in a mammal in need thereof, or
(c) decreasing the proliferation of T-lymphocytes in a mammal in need thereof in response to an antigen.
4. Use of a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide, antagonist or a fragment thereof to prepare a composition useful for:
(a) decreasing infiltration of inflammatory cells into a tissue of a mammal in need thereof,
(b) inhibiting or reducing an immune response in a mammal in need thereof, or
(c) decreasing the proliferation of T-lymphocytes in a mammal in need thereof in response to an antigen.
5. A method of treating an immune related disorder, such as a T cell mediated disorder, in a mammal in need thereof, compnsmg admmistenng to the mammal an effective amount of a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide, an agonist antibody thereof, an antagonist antibody thereto, or a fragment thereof
6 The method of claim 5, wherein the disorder is selected from systemic lupus erythematosis, rheumatoid arthntis, juvenile chronic arthritis, spondyloarthropathies, systemic sclerosis (scleroderma), idiopathic inflammatory myopathies (dermatomyositis, polymyositis), Sjogren's syndrome, systemic vascuhtis, sarcoidosis, autoimmune hemolytic anemia (immune pancytopenia, paroxysmal nocturnal hemoglobinuna), autoimmune thrombocytopenia (idiopathic thrombocytopenic purpura, immune-mediated thrombocytopenia), thyroiditis (Grave's disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, atrophic thyroiditis), diabetes mellitus, immune-mediated renal disease (glomerulonephntis, tubulointerstitial nephntis), demyehnatmg diseases of the central and penpheral nervous systems such as multiple sclerosis, idiopathic demyehnatmg polyneuropathy or Guillain-Barre syndrome, and chrome inflammatory demyehnatmg polyneuropathy, hepatobiliary diseases such as infectious hepatitis (hepatitis A, B, C, D, E and other non-hepatotropic viruses), autoimmune chronic active hepatitis, pnmary biliary cirrhosis, granulomatous hepatitis, and scleros g cholangitis, inflammatory and fibrotic lung diseases such as inflammatory bowel disease (ulcerative colitis- Crohn's disease), gluten-sensitive enteropathy, and Whipple's disease, autoimmune or immune-mediated skm diseases including bullous skin diseases, erythema multiforme and contact dermatitis, psonasis, allergic diseases such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity and urticana, immunologic diseases of the lung such as eosinophilic pneumonias, idiopathic pulmonary fibrosis and hypersensitivity pneumomtis, transplantation associated diseases including graft rejection and graft -versus-host-disease
7 The composition or use of any of the preceding claims, wherein the antibody is a monoclonal antibody
8 The composition or use of any of the preceding claims, wherein the antibody is an antibody fragment or a single-chain antibody
9 The composition or use of any of the preceding claims, wherein the antibody has nonhuman complementanty determining region (CDR) residues and human framework region (FR) residues
10 A method for determining the presence of a PR0245, PR0217, PRO301 , PR0266, PR0335, PR0331 or PR0326 polypeptide, comprising exposing a cell suspected of containing the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide to an anti-PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 antibody and determining binding of the antibody to the cell.
11. A method of diagnosing an immune related disease in a mammal, comprising detecting the level of expression of a gene encoding a PR0245, PR0217, PRO301 , PR0266, PR0335, PR0331 or PR0326 polypeptide (a) in a test sample of tissue cells obtained from the mammal, and (b) in a control sample of known normal tissue cells of the same cell type, wherein a higher expression level in the test sample indicates the presence of immune related disease in the mammal from which the test tissue cells were obtained.
12. A method of diagnosing an immune related disease in a mammal, comprising (a) contacting an anti-PR0245, PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 antibody with a test sample of tissue cells obtained from the mammal , and (b) detecting the formation of a complex between the antibody and the polypeptide in the test sample.
13. An immune related disease diagnostic kit, comprising an anti-PR0245, PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 antibody or fragment thereof and a carrier in suitable packaging.
14. The kit of claim 13, further comprising instructions for using the antibody to detect a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide.
15. An article of manufacture, comprising: a container; a label on the container; and a composition comprising an active agent contained within the container; wherein the composition is effective for stimulating or enhancing an immune response in a mammal, the label on the container indicates that the composition can be used for treating an immune related disease, and the active agent in the composition is an agent inhibiting the expression and/or activity of a PR0245. PR0217, PRO301, PR0266, PR0335. PR0331 or PR0326 polypeptide.
16. The article of manufacture of claim 21 wherein said active agent is an anti- PR0245, PR0217, PRO301. PR0266, PR0335, PR0331 or PR0326 antibody.
17. A method for identifying a compound capable of inhibiting the expression or activity of a PR0245 polypeptide, comprising contacting a candidate compound with a PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide under conditions and for a time sufficient to allow these two components to interact.
18. The method of claim 17, wherein the candidate compound or the PR0245, PR0217, PRO301, PR0266, PR0335, PR0331 or PR0326 polypeptide is immobilized on a solid support.
19. The method of claim 18, wherein the non-immobilized component carries a detectable label.
PCT/US1999/021547 1994-09-08 1999-09-15 Compositions and methods for the treatment of immune related diseases WO2000015797A2 (en)

Priority Applications (379)

Application Number Priority Date Filing Date Title
CA002343006A CA2343006A1 (en) 1998-09-17 1999-09-15 Compositions and methods for the treatment of immune related diseases
AU64984/99A AU6498499A (en) 1998-09-17 1999-09-15 Compositions and methods for the treatment of immune related diseases
KR1020017003430A KR20010085816A (en) 1998-09-17 1999-09-15 Compositions and Methods for the Treatment of Immune Related Diseases
IL14155199A IL141551A0 (en) 1998-09-17 1999-09-15 Compositions and methods for the treatment of immune related diseases
EP99952930A EP1114154A2 (en) 1998-09-17 1999-09-15 Compositions and methods for the treatment of immune related diseases
JP2000570324A JP2003524600A (en) 1998-09-17 1999-09-15 Compositions and methods for treating immune-related diseases
CA002503475A CA2503475A1 (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
PCT/US1999/028313 WO2000032221A2 (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
EP06014175A EP1734051A3 (en) 1998-12-01 1999-11-30 Composition and methods for the diagnosis of tumours
AU17482/00A AU771751C (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
CA002347835A CA2347835A1 (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
NZ535590A NZ535590A (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
AT99960624T ATE458050T1 (en) 1998-12-01 1999-11-30 PROMOTION OR INHIBITION OF ANGIOGENESIS AND CARDIOVASCULARIZATION
JP2000584912A JP3695642B2 (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
MXPA01005169A MXPA01005169A (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization.
KR10-2001-7006818A KR100529270B1 (en) 1998-12-01 1999-11-30 Promotion or Inhibition of Angiogenesis and Cardiovascularization
IL14274299A IL142742A0 (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
EP99960624A EP1135485B1 (en) 1998-12-01 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
AT07001711T ATE432987T1 (en) 1998-12-22 1999-12-02 METHODS AND COMPOSITIONS FOR INHIBITING NEOPLASTIC CELL GROWTH
IL14303199A IL143031A0 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
EP99960644A EP1141284A2 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
ES07001711T ES2327785T3 (en) 1998-12-22 1999-12-02 PROCEDURES AND COMPOUNDS TO INHIBIT THE GROWTH OF NEOPLASTIC CELLS.
EP07001711A EP1820859B9 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
DE69935085T DE69935085T2 (en) 1998-12-22 1999-12-02 Method and composition for inhibiting neoplastic cell growth
EP04007617A EP1484338B1 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
DK04007617T DK1484338T3 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
MXPA01006330A MXPA01006330A (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth.
DE69940964T DE69940964D1 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
PT04007617T PT1484338E (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
KR10-2001-7007850A KR100499600B1 (en) 1998-12-22 1999-12-02 Methods and Compositions for Inhibiting Neoplastic Cell Growth
JP2000589692A JP3993746B2 (en) 1998-12-22 1999-12-02 Compositions and methods for inhibiting neoplastic cell growth
CA002353799A CA2353799A1 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
ES04007617T ES2281704T3 (en) 1998-12-22 1999-12-02 PROCEDURES AND COMPOUNDS TO INHIBIT THE GROWTH OF NEOPLASSIC CELLS.
PCT/US1999/028565 WO2000037638A2 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
EP09004092A EP2075335A3 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
CA002450402A CA2450402A1 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting cancer cell growth comprising pro224
AU17499/00A AU768230B2 (en) 1998-12-22 1999-12-02 Methods and compositions for inhibiting neoplastic cell growth
AT04007617T ATE353339T1 (en) 1998-12-22 1999-12-02 METHOD AND COMPOSITION FOR INHIBITING NEOPLASTIC CELL GROWTH
PCT/US1999/030911 WO2000075316A1 (en) 1999-06-02 1999-12-20 Methods and compositions for inhibiting neoplastic cell growth
AU22153/00A AU2215300A (en) 1999-06-02 1999-12-20 Methods and compositions for inhibiting neoplastic cell growth
AU26008/00A AU2600800A (en) 1999-03-08 2000-01-05 Promotion or inhibition of angiogenesis and cardiovascularization
PCT/US2000/000219 WO2000053753A2 (en) 1999-03-08 2000-01-05 Promotion or inhibition of angiogenesis and cardiovascularization
PCT/US2000/004414 WO2001004311A1 (en) 1999-07-07 2000-02-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU28839/00A AU2883900A (en) 1999-07-07 2000-02-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
PCT/US2000/005841 WO2000053758A2 (en) 1999-03-08 2000-03-02 Compositions and methods for the treatment of immune related diseases
CA002362427A CA2362427A1 (en) 1999-03-08 2000-03-02 Compositions and methods for the treatment of immune related diseases
KR1020017011406A KR20010103046A (en) 1999-03-08 2000-03-02 Compositions and Methods for the Treatment of Immune Related Diseases
AU35144/00A AU3514400A (en) 1999-03-08 2000-03-02 Compositions and methods for the treatment of immune related diseases
EP00913764A EP1220905A2 (en) 1999-03-08 2000-03-02 Composition and methods for the treatment of immune related diseases
JP2000603379A JP2004516227A (en) 1999-03-08 2000-03-02 Compositions and methods for treating immune-related diseases
PCT/US2000/007377 WO2001019991A1 (en) 1999-09-15 2000-03-20 Compositions and methods for the treatment of immune related diseases
AU39026/00A AU3902600A (en) 1999-09-15 2000-03-20 Compositions and methods for the treatment of immune related diseases
AT00916675T ATE478145T1 (en) 1999-06-02 2000-03-30 SECRETED AND TRANSMEMBRANE POLYPEPTIDES AND NUCLEIC ACIDS CODING THEREFOR
CA002383254A CA2383254A1 (en) 1999-06-02 2000-03-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU37743/00A AU3774300A (en) 1999-06-02 2000-03-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP00916675A EP1210418B1 (en) 1999-06-02 2000-03-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
DE60044849T DE60044849D1 (en) 1999-06-02 2000-03-30 SECRETED AND TRANSMEMBRANE POLYPEPTIDES AND NUCLEIC ACIDS THEREFOR CODED
JP2001500766A JP2004522402A (en) 1999-06-02 2000-03-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
PCT/US2000/008439 WO2000073454A1 (en) 1999-06-02 2000-03-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
AT07025118T ATE448246T1 (en) 1999-06-15 2000-05-22 SECRETED AND TRANSMEMBRANE POLYPEPTIDES AND NUCLEIC ACIDS FOR THE ENCODING THEM
DE60043322T DE60043322D1 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids for their coding
EP00936172A EP1208195A2 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
AT07025116T ATE449109T1 (en) 1999-06-15 2000-05-22 SECRETED AND TRANSMEMBRANE POLYPEPTIDES AND NUCLEIC ACIDS FOR THE ENCODING THEM
JP2001503894A JP2003529324A (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids encoding them
EP07025117A EP1978029A3 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids endoding the same
PCT/US2000/014042 WO2000077037A2 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
CA2372511A CA2372511C (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP07025116A EP1953173B1 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids endoding the same
EP07025118A EP1956030B1 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids endoding the same
DE60043367T DE60043367D1 (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids for their coding
AU51527/00A AU5152700A (en) 1999-06-15 2000-05-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
AU54601/00A AU5460100A (en) 1999-06-02 2000-06-02 Compositions and methods for the treatment of immune related diseases
PCT/US2000/015264 WO2000073452A2 (en) 1999-06-02 2000-06-02 Compositions and methods for the treatment of immune related diseases
US09/902,759 US20030077654A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,979 US20030113718A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,736 US20030049676A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,853 US20020192659A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,572 US20030108983A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,903 US20030044839A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,615 US20030092002A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,634 US20030082540A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,692 US20030054400A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/902,713 US20030082541A1 (en) 1997-09-17 2001-07-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,823 US20030104381A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,806 US20030130489A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,011 US20030003530A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,119 US20030049621A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids enconding the same
US09/903,603 US20030148419A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,786 US20030044793A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,925 US20030096233A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,562 US6965015B2 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,749 US7147853B2 (en) 1997-09-17 2001-07-11 Anti-pro211 polypeptide antibodies
US09/903,943 US20030054349A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,640 US7208308B2 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/903,520 US20030054401A1 (en) 1997-09-17 2001-07-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,786 US7557192B2 (en) 1997-09-17 2001-07-12 Anti-PRO335 antibodies
US09/904,938 US20030211569A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,992 US20030135025A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,088 US20030073077A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,766 US20030152999A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,291 US20020160374A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,056 US20030054441A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,859 US20030036060A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,125 US6664376B2 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,877 US20030186358A1 (en) 1997-09-17 2001-07-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,449 US6965011B2 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,532 US7151160B2 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,381 US6818746B2 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,462 US6878807B2 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,485 US20030064367A1 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/905,075 US7169906B2 (en) 1997-09-17 2001-07-13 PRO211 polypeptides
US09/905,348 US20030064923A1 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,920 US6806352B2 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,820 US20030036094A1 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,838 US20030148370A1 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,553 US20030059828A1 (en) 1997-09-17 2001-07-13 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/904,956 US20030049622A1 (en) 1995-12-01 2001-07-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,618 US6828146B2 (en) 1997-09-17 2001-07-16 Nucleic acid encoding PRO229 polypeptides
US09/906,722 US6946262B2 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,646 US6852848B2 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,700 US6723535B2 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,742 US20030023054A1 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,760 US20030096340A1 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,777 US20030148371A1 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,838 US7070979B2 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/906,815 US7094567B2 (en) 1997-09-17 2001-07-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,652 US20030104469A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,613 US20030027145A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,841 US7033825B2 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,824 US20020197671A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,979 US20030082542A1 (en) 1994-09-08 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/908,093 US20030017498A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,794 US6635468B2 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,575 US20030073079A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,942 US7087738B2 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,925 US20030054352A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/907,728 US20030190611A1 (en) 1997-09-17 2001-07-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/909,320 US7074592B2 (en) 1997-09-17 2001-07-18 Secreted and transmembrane polypeptides nucleic acid encoding
US09/909,064 US6818449B2 (en) 1997-09-17 2001-07-18 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/909,088 US20020146709A1 (en) 1997-09-17 2001-07-18 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/908,576 US20040005553A1 (en) 1997-09-17 2001-07-18 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/909,204 US20030036061A1 (en) 1997-09-17 2001-07-18 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/941,992 US20030082546A1 (en) 1996-11-06 2001-08-28 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,726 US20030054359A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,562 US20030027985A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,438 US20030027754A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,443 US20030054987A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,711 US20030032023A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,456 US20020137890A1 (en) 1997-03-31 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,427 US20030073809A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,854 US20030059780A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,444 US6930170B2 (en) 1997-06-16 2001-11-14 PRO1184 polypeptides
US09/993,469 US20030068623A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,163 US20020132253A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/992,521 US20030083461A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/993,748 US20030069403A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/992,598 US6956108B2 (en) 1997-06-16 2001-11-14 PRO1184 antibodies
US09/993,583 US7074897B2 (en) 1997-06-16 2001-11-14 Pro943 polypeptides
US09/993,667 US20030022187A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,436 US20020198148A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,442 US20020132252A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,440 US20030060407A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/993,687 US20020198149A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,073 US20020127576A1 (en) 1997-06-16 2001-11-14 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,440 US20030059833A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/998,041 US20030119001A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,573 US20030049682A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,585 US7166282B2 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/998,041 US7309775B2 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,542 US20030068647A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,514 US7019116B2 (en) 1997-06-16 2001-11-15 PRO 1387 polypeptides
US09/997,529 US7309761B2 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,614 US20030124531A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,641 US7112656B2 (en) 1997-06-16 2001-11-15 PRO1312 polypeptides
US09/997,384 US7119177B2 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,653 US7034122B2 (en) 1997-06-16 2001-11-15 Antibodies to PRO1159 polypeptides
US09/997,349 US7034106B2 (en) 1997-06-16 2001-11-15 Pro1159 polypeptides
US09/997,683 US20030059783A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/002,796 US20030032057A1 (en) 1997-08-26 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,666 US7244816B2 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,585 US20030119055A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,428 US20030027162A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,628 US20030059782A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,559 US20030054403A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,529 US20030134284A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,641 US20030224358A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,666 US20030027163A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,601 US7189814B2 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/998,156 US20030044806A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,384 US20030087305A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,601 US20030054404A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/997,333 US6953836B2 (en) 1997-06-16 2001-11-15 PRO844 polypeptides
US09/997,857 US20030064375A1 (en) 1997-06-16 2001-11-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,181 US6913919B2 (en) 1997-06-16 2001-11-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,150 US20030194760A1 (en) 1997-06-16 2001-11-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,172 US20030050457A1 (en) 1997-06-16 2001-11-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/990,441 US7041804B2 (en) 1997-06-16 2001-11-16 Antibodies to PRO1387 polypeptides
US09/990,437 US20030045463A1 (en) 1997-06-16 2001-11-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/991,157 US7101687B2 (en) 1997-06-16 2001-11-16 Nucleic acids encoding PRO943
US09/989,723 US20020072092A1 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,722 US20020072067A1 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,726 US7018811B2 (en) 1997-06-16 2001-11-19 Nucleic acids encoding PRO189 polypeptides
US09/989,721 US20020142961A1 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,735 US6972185B2 (en) 1997-06-16 2001-11-19 Nucleic acids encoding PRO844 polypeptides
US09/989,279 US7083978B2 (en) 1997-06-16 2001-11-19 Nucleic acid encoding PRO1111 polypeptides
US09/989,732 US7037679B2 (en) 1997-06-16 2001-11-19 Nucleic acids encoding PRO1184 polypeptides
US09/989,328 US7056736B2 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,727 US20020072497A1 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,734 US7491529B2 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,729 US20030059831A1 (en) 1997-06-16 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,862 US20030130182A1 (en) 1997-11-05 2001-11-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,730 US7157247B2 (en) 1997-06-16 2001-11-20 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,293 US7034136B2 (en) 1997-06-16 2001-11-20 Nucleic acids encoding PRO1159 polypeptides
US09/989,731 US20020103125A1 (en) 1997-06-16 2001-11-20 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US09/989,728 US7029873B2 (en) 1997-06-16 2001-11-20 Nucleic acids to PRO1387 polypeptides
US09/989,725 US20030139329A1 (en) 1997-06-16 2001-11-20 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/028,072 US20030004311A1 (en) 1997-06-18 2001-12-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,269 US20030040014A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,193 US20030044902A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,198 US20030170721A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,203 US20030180796A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,273 US7317092B2 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,211 US20030044844A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/066,500 US20020177165A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding
US10/066,494 US20030032063A1 (en) 1997-08-26 2002-02-01 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/081,056 US20040043927A1 (en) 1997-09-19 2002-02-20 Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
US10/121,046 US20030194791A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,040 US20030082759A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,042 US20030096386A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,059 US20030190721A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,051 US20030092147A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,047 US20030077778A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,044 US20030190717A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,041 US20030077776A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,045 US20030073210A1 (en) 1997-03-31 2002-04-11 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,048 US20030199051A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,043 US7220831B2 (en) 1997-03-31 2002-04-12 PRO235 polypeptides
US10/121,061 US20030082761A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,060 US20030190722A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,055 US20030190718A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,049 US20030022239A1 (en) 1997-06-18 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,062 US20030077779A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,056 US20030082760A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,050 US20030054516A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,052 US20030199052A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,058 US20030190720A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,054 US20030199054A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,057 US20030190719A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,053 US20030199053A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/121,063 US20030199055A1 (en) 1997-03-31 2002-04-12 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,213 US20030199057A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,213 US7193048B2 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,155 US20030068794A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,212 US7276577B2 (en) 1997-03-31 2002-04-15 PRO1866 polypeptides
US10/123,291 US20030199058A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,215 US7291329B2 (en) 1997-03-31 2002-04-15 Antibodies against PRO4406
US10/123,108 US7635478B2 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,214 US7343721B2 (en) 1997-03-31 2002-04-15 PRO4406 polypeptide
US10/123,261 US20030068796A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,109 US20030190723A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,235 US20030082762A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,771 US20030199060A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,292 US20030073211A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,154 US20030190724A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,157 US20030190725A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,322 US20030199059A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,156 US20030194792A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,236 US20030068795A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,262 US20030049816A1 (en) 1997-03-31 2002-04-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,904 US20030022328A1 (en) 1997-03-31 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,906 US20030190726A1 (en) 1997-03-31 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,905 US20030087344A1 (en) 1997-06-18 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,907 US7084258B2 (en) 1997-03-31 2002-04-16 Antibodies against the PRO862 polypeptides
US10/123,913 US20030203462A1 (en) 1997-03-31 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,908 US7335728B2 (en) 1997-03-31 2002-04-16 PRO1310 polypeptides
US10/123,902 US20030077781A1 (en) 1997-03-31 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,905 US7285625B2 (en) 1997-06-18 2002-04-16 PRO536 polypeptides
US10/123,909 US7193049B2 (en) 1997-03-31 2002-04-16 PRO862 polypeptides
US10/123,912 US20030100087A1 (en) 1997-03-31 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/123,910 US7329404B2 (en) 1997-03-31 2002-04-16 Antibodies against PRO1310
US10/123,911 US7408032B2 (en) 1997-03-31 2002-04-16 PRO1188 polypeptides
US10/123,903 US20030073212A1 (en) 1997-03-31 2002-04-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,816 US20030190728A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/125,805 US20030194794A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,817 US20030077786A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,822 US7109305B2 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/125,704 US7357926B2 (en) 1997-03-31 2002-04-17 Antibodies against PRO1879 and the use thereof
US10/124,818 US20030082763A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,824 US20030077659A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/125,795 US7304131B2 (en) 1997-03-31 2002-04-17 PRO1483 polypeptides
US10/124,823 US20030199062A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,814 US7105335B2 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,813 US7312307B2 (en) 1997-03-31 2002-04-17 PRO1056 polypeptides
US10/124,819 US7285626B2 (en) 1997-03-31 2002-04-17 PRO1076 polypeptides
US10/124,821 US20030199023A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/124,820 US20030190729A1 (en) 1997-03-31 2002-04-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/125,927 US20030190731A1 (en) 1997-03-31 2002-04-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/125,931 US20030199063A1 (en) 1997-03-31 2002-04-19 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/125,932 US7317079B2 (en) 1997-03-31 2002-04-19 PRO812 polypeptides
US10/125,922 US7309762B2 (en) 1997-03-31 2002-04-19 PRO1360 polypeptides
US10/125,924 US7342097B2 (en) 1997-03-31 2002-04-19 PRO1309 polypeptides
US10/127,831 US20030082689A1 (en) 1997-03-31 2002-04-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/128,689 US20030087365A1 (en) 1997-03-31 2002-04-23 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/131,825 US7282566B2 (en) 1997-03-31 2002-04-24 PRO1779 polypeptide
US10/131,817 US7291701B2 (en) 1997-03-31 2002-04-24 PRO1777 polypeptides
US10/131,823 US7304132B2 (en) 1997-03-31 2002-04-24 PRO1693 polypeptides
US10/137,865 US20030032155A1 (en) 1997-03-31 2002-05-03 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/137,867 US20030207349A1 (en) 1997-03-31 2002-05-03 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/137,868 US20030082764A1 (en) 1997-03-31 2002-05-03 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/139,963 US7288625B2 (en) 1997-03-31 2002-05-06 PRO4395 polypeptides
US10/140,020 US20030207415A1 (en) 1997-03-31 2002-05-06 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,470 US20030022331A1 (en) 1997-03-31 2002-05-06 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,024 US20040058424A1 (en) 1997-03-31 2002-05-06 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,023 US20030207416A1 (en) 1997-03-31 2002-05-06 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,474 US20030032156A1 (en) 1997-03-31 2002-05-06 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/139,980 US7247710B2 (en) 1997-03-31 2002-05-06 PRO4395 antibodies
US10/140,805 US20030207417A1 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,921 US7317080B2 (en) 1997-03-31 2002-05-07 PRO4303 polypeptides
US10/140,865 US20030207420A1 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,928 US20030068798A1 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,860 US7307151B2 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,808 US7425621B2 (en) 1997-03-31 2002-05-07 Antibodies against the PRO4401 polypeptide
US10/140,925 US20030073215A1 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,864 US20030207419A1 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/140,809 US20030207418A1 (en) 1997-03-31 2002-05-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/141,755 US7297764B2 (en) 1997-03-31 2002-05-08 PRO4318 polypeptides
US10/141,760 US7342104B2 (en) 1997-03-31 2002-05-08 Antibodies against the PRO4320 polypeptide
US10/141,756 US7488586B2 (en) 1997-03-31 2002-05-08 PRO4409 polypeptides
US10/141,701 US20030207421A1 (en) 1997-03-31 2002-05-08 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/141,754 US7361732B2 (en) 1997-03-31 2002-05-08 PRO4400 polypeptides
US10/142,425 US20030207424A1 (en) 1997-03-31 2002-05-09 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/143,114 US20030036180A1 (en) 1997-03-31 2002-05-09 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/142,417 US7304133B2 (en) 1997-03-31 2002-05-09 PRO4389 polypeptides
US10/142,430 US7309766B2 (en) 1997-03-31 2002-05-09 PRO5774 polypeptides
US10/143,113 US7329730B2 (en) 1997-03-31 2002-05-09 PRO4348 polypeptides
US10/142,431 US7285629B2 (en) 1997-03-31 2002-05-10 Pro5005 polypeptides
US10/142,419 US7153941B2 (en) 1997-03-31 2002-05-10 Antibodies that bind PRO4994 polypeptides
US10/143,032 US7408033B2 (en) 1997-03-31 2002-05-10 PRO5995 polypeptides
US10/142,423 US20030049817A1 (en) 1997-03-31 2002-05-10 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/146,730 US20030207427A1 (en) 1997-03-31 2002-05-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/146,792 US20030207428A1 (en) 1997-03-31 2002-05-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/147,528 US20030219885A1 (en) 1997-03-31 2002-05-16 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/147,536 US20040077064A1 (en) 1997-03-31 2002-05-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/147,519 US20030077791A1 (en) 1997-03-31 2002-05-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/147,492 US20030082765A1 (en) 1997-03-31 2002-05-17 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/152,395 US7189534B2 (en) 1997-03-31 2002-05-21 PRO4320 polynucleotide
US10/153,934 US20030129695A1 (en) 1997-03-31 2002-05-22 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/156,843 US20030207805A1 (en) 1997-06-18 2002-05-28 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/157,786 US20030208055A1 (en) 1997-03-31 2002-05-29 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/157,782 US20030077792A1 (en) 1997-03-31 2002-05-29 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/160,498 US20030073216A1 (en) 1997-03-31 2002-05-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/158,791 US20030207429A1 (en) 1997-03-31 2002-05-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/158,782 US20030082766A1 (en) 1997-03-31 2002-05-30 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/176,913 US20030022298A1 (en) 1997-09-15 2002-06-20 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/187,885 US20030032138A1 (en) 1998-06-24 2002-07-02 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US10/226,739 US7390879B2 (en) 1999-06-15 2002-08-23 Secreted and transmembrane polypeptides and nucleic acids encoding the same
JP2002341509A JP4017507B2 (en) 1999-06-02 2002-09-25 Medicament containing polypeptide
US10/771,187 US7355002B2 (en) 1997-11-12 2004-02-02 Secreted and transmembrane polypeptides and nucleic acids encoding the same
JP2004212340A JP3803681B2 (en) 1998-12-01 2004-07-20 Promotion or inhibition of angiogenesis and cardiovascularization
US10/970,823 US7307152B2 (en) 1997-10-17 2004-10-20 Secreted and transmembrane polypeptides and nucleic acids encoding the same
JP2005066570A JP2005237383A (en) 1998-12-22 2005-03-10 Composition and method for inhibiting neoplastic cell growth
JP2005066569A JP4037876B2 (en) 1998-12-22 2005-03-10 Compositions and methods for inhibiting neoplastic cell growth
US11/100,159 US7425613B2 (en) 1997-11-05 2005-04-05 PRO1375 polypeptides
JP2005180188A JP4358159B2 (en) 1998-12-01 2005-06-21 Promotion or inhibition of angiogenesis and cardiovascularization
JP2005235120A JP2006068006A (en) 1999-06-15 2005-08-15 Secreted and transmembrane polypeptide and nucleic acid encoding the same
JP2005238274A JP2006051032A (en) 1999-06-15 2005-08-19 Secreted and transmembrane polypeptide and nucleic acid encoding the same
JP2005238266A JP2006025795A (en) 1999-06-15 2005-08-19 Secreted and transmembrane polypeptide and nucleic acid encoding the same
JP2005238244A JP2006061156A (en) 1999-06-15 2005-08-19 Secreted and transmembrane polypeptide and nucleic acid encoding the same
JP2005238217A JP2006051031A (en) 1999-06-15 2005-08-19 Secreted and transmembrane polypeptide and nucleic acid encoding the same
US11/240,891 US20060246540A1 (en) 1997-08-26 2005-09-29 Secreted and transmembrane polypeptides and nucleic acids encoding the same
US11/341,175 US7468427B2 (en) 1997-03-31 2006-01-27 Antibodies to PRO1275 polypeptide
US11/518,609 US20070077623A1 (en) 1997-09-17 2006-09-07 Secreted and transmembrane polypeptides and nucleic acids encoding the same
JP2007138469A JP2007291116A (en) 1998-12-22 2007-05-24 Composition and method for neoplastic cell growth inhibition
JP2007138468A JP2007222180A (en) 1998-12-22 2007-05-24 Composition and method for inhibiting neoplastic cell growth
JP2007172879A JP5512073B2 (en) 1999-06-02 2007-06-29 Medicament containing polypeptide
IL186154A IL186154A0 (en) 1998-12-22 2007-09-20 Methods and compositions for inhibiting neoplastic cell growth
IL186153A IL186153A0 (en) 1998-12-22 2007-09-20 Methods and compositions for inhibiting neoplastic cell growth
IL202176A IL202176A0 (en) 1998-12-01 2009-11-17 Promotion or inhibition of angiogenesis and cardiovascularization
JP2011138276A JP2011236224A (en) 1999-06-02 2011-06-22 Medicine containing polypeptide

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US10085898P 1998-09-17 1998-09-17
PCT/US1998/019437 WO1999014241A2 (en) 1997-09-17 1998-09-17 Compositions and methods for the treatment of immune related diseases
USPCT/US98/19437 1998-09-17
US60/100,858 1998-09-17

Related Parent Applications (6)

Application Number Title Priority Date Filing Date
US40329799A Continuation-In-Part 1997-08-26 1999-09-01
PCT/US1999/020111 Continuation-In-Part WO2000012708A2 (en) 1997-03-31 1999-09-01 Further pro polypeptides and sequences thereof
PCT/US1999/020594 Continuation-In-Part WO2000015666A2 (en) 1994-09-08 1999-09-08 Compositions and methods for the treatment of tumors
PCT/US1999/020944 Continuation-In-Part WO2000015792A2 (en) 1994-09-08 1999-09-13 Promotion or inhibition of angiogenesis and cardiovascularization
PCT/US1999/021547 Continuation-In-Part WO2000015797A2 (en) 1994-09-08 1999-09-15 Compositions and methods for the treatment of immune related diseases
PCT/US1999/021090 Continuation-In-Part WO2000015796A2 (en) 1994-09-08 1999-09-15 Secreted and transmembrane polypeptides and nucleic acids encoding the same

Related Child Applications (8)

Application Number Title Priority Date Filing Date
PCT/US1999/021547 Continuation-In-Part WO2000015797A2 (en) 1994-09-08 1999-09-15 Compositions and methods for the treatment of immune related diseases
US40329699A Continuation-In-Part 1996-11-06 1999-10-18
US40329699A A-371-Of-International 1996-11-06 1999-10-18
PCT/US1999/028214 Continuation-In-Part WO2001019987A1 (en) 1994-09-08 1999-11-29 Promotion or inhibition of angiogenesis and cardiovascularization
PCT/US1999/028313 Continuation-In-Part WO2000032221A2 (en) 1994-09-08 1999-11-30 Promotion or inhibition of angiogenesis and cardiovascularization
US80868901A Continuation 1996-11-06 2001-03-14
US09/903,749 Continuation US7147853B2 (en) 1997-09-17 2001-07-11 Anti-pro211 polypeptide antibodies
US10/081,056 Continuation US20040043927A1 (en) 1997-09-19 2002-02-20 Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis

Publications (2)

Publication Number Publication Date
WO2000015797A2 true WO2000015797A2 (en) 2000-03-23
WO2000015797A3 WO2000015797A3 (en) 2000-08-24

Family

ID=22281903

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/021547 WO2000015797A2 (en) 1994-09-08 1999-09-15 Compositions and methods for the treatment of immune related diseases

Country Status (6)

Country Link
JP (1) JP2003524600A (en)
KR (1) KR20010085816A (en)
AU (1) AU6498499A (en)
CA (1) CA2343006A1 (en)
IL (1) IL141551A0 (en)
WO (1) WO2000015797A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001019991A1 (en) * 1999-09-15 2001-03-22 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US6878806B2 (en) 1997-03-14 2005-04-12 Human Genome Sciences, Inc. Human secreted protein HTEEB42
EP1778273A2 (en) * 2004-08-13 2007-05-02 Genentech Inc. Promotion of axonal regeneration
JP2009159949A (en) * 2001-10-19 2009-07-23 Genentech Inc Composition and method for diagnosis and treatment of inflammatory bowel disorder
US7781176B2 (en) 1997-03-14 2010-08-24 Human Genome Sciences, Inc. Antibodies to HTEEB42 protein
JP2010207229A (en) * 2003-03-03 2010-09-24 Genentech Inc Composition and method for treating systemic lupus erythematosus

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013033271A2 (en) * 2011-08-29 2013-03-07 Derren Barken Method to augment immune system in response to disease or injury

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996027603A1 (en) * 1995-03-03 1996-09-12 Millennium Pharmaceuticals, Inc. Compositions and methods for the treatment and diagnosis of immune disorders
WO1998001541A1 (en) * 1996-07-10 1998-01-15 Tularik, Inc. Traf2-associated protein kinase and assays
WO1998011138A1 (en) * 1996-09-12 1998-03-19 Human Genome Sciences, Inc. Chemokine alpha-4
WO1998024908A1 (en) * 1996-12-05 1998-06-11 Human Genome Sciences, Inc. Human chemokine beta-13
WO1998040483A2 (en) * 1997-03-14 1998-09-17 Human Genome Sciences, Inc. 28 human secreted proteins
WO1999014241A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO1999014328A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO1999027098A2 (en) * 1997-11-21 1999-06-03 Genentech, Inc. A-33 related antigens and their pharmacological uses

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996027603A1 (en) * 1995-03-03 1996-09-12 Millennium Pharmaceuticals, Inc. Compositions and methods for the treatment and diagnosis of immune disorders
WO1998001541A1 (en) * 1996-07-10 1998-01-15 Tularik, Inc. Traf2-associated protein kinase and assays
WO1998011138A1 (en) * 1996-09-12 1998-03-19 Human Genome Sciences, Inc. Chemokine alpha-4
WO1998024908A1 (en) * 1996-12-05 1998-06-11 Human Genome Sciences, Inc. Human chemokine beta-13
WO1998040483A2 (en) * 1997-03-14 1998-09-17 Human Genome Sciences, Inc. 28 human secreted proteins
WO1999014241A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
WO1999014328A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
WO1999027098A2 (en) * 1997-11-21 1999-06-03 Genentech, Inc. A-33 related antigens and their pharmacological uses

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL - EMEST2 [Online] Entry/Acc.no. AA725566, 8 January 1998 (1998-01-08) STRAUSBERG, R.: "ai24d02.s1 Soares_testis_NHT Homo sapiens cDNA clone 1343715 3' similar to TR:Q99795 Q99795 A33 ANTIGEN PRECURSOR. ;, mRNA sequence." XP002132547 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878806B2 (en) 1997-03-14 2005-04-12 Human Genome Sciences, Inc. Human secreted protein HTEEB42
US7781176B2 (en) 1997-03-14 2010-08-24 Human Genome Sciences, Inc. Antibodies to HTEEB42 protein
WO2001019991A1 (en) * 1999-09-15 2001-03-22 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
JP2009159949A (en) * 2001-10-19 2009-07-23 Genentech Inc Composition and method for diagnosis and treatment of inflammatory bowel disorder
JP2013140167A (en) * 2001-10-19 2013-07-18 Genentech Inc Compositions and methods for diagnosis and treatment of inflammatory bowel disease
JP2013172712A (en) * 2001-10-19 2013-09-05 Genentech Inc Composition and method for diagnosis and treatment of inflammatory bowel disorder
JP2010207229A (en) * 2003-03-03 2010-09-24 Genentech Inc Composition and method for treating systemic lupus erythematosus
EP1778273A2 (en) * 2004-08-13 2007-05-02 Genentech Inc. Promotion of axonal regeneration
EP1778273A4 (en) * 2004-08-13 2010-08-25 Genentech Inc Promotion of axonal regeneration

Also Published As

Publication number Publication date
CA2343006A1 (en) 2000-03-23
WO2000015797A3 (en) 2000-08-24
IL141551A0 (en) 2002-03-10
KR20010085816A (en) 2001-09-07
JP2003524600A (en) 2003-08-19
AU6498499A (en) 2000-04-03

Similar Documents

Publication Publication Date Title
CA2586615C (en) Pro87299 (btla), and variants thereof, for treating immune related disorders
EP1220905A2 (en) Composition and methods for the treatment of immune related diseases
WO2000037638A2 (en) Methods and compositions for inhibiting neoplastic cell growth
WO2000073452A2 (en) Compositions and methods for the treatment of immune related diseases
WO2004039394A1 (en) Novel composition and methods for the treatment of immune related diseases
WO2000070050A1 (en) Compositions and methods for the treatment of immune related diseases
WO2001005972A1 (en) Compositions and methods for the treatment of immune related diseases
WO2000075327A1 (en) Methods and compositions for inhibiting neoplastic cell growth
EP1135491A2 (en) Methods and compositions for inhibiting neoplastic cell growth
WO2000015797A2 (en) Compositions and methods for the treatment of immune related diseases
WO2001040465A2 (en) Compositions and methods for the treatment of immune related diseases
NZ531311A (en) Poro20080 polypeptide and nucleotides encoding them, antibodies and the treatment of immune related disorders
WO2004024076A9 (en) Novel compositions and methods for the treatment of immune related diseases
WO2001029070A2 (en) Modulation of t cell differentiation for the treatment of t helper cell mediated diseases
EP1578368A2 (en) Compositions and methods for the treatment of rheumatoid arthritis
WO2000075316A1 (en) Methods and compositions for inhibiting neoplastic cell growth
WO2004096848A2 (en) Novel composition and methods for the treatment of immune disorders
WO2000021996A2 (en) Methods and compositions for inhibiting neoplastic cell growth
WO2001016319A2 (en) Compositions and methods for the treatment of immune related diseases
EP1114154A2 (en) Compositions and methods for the treatment of immune related diseases
AU2003215368B2 (en) Novel composition and methods for the treatment of immune related diseases
WO2001019991A1 (en) Compositions and methods for the treatment of immune related diseases
WO2000055319A1 (en) Methods and compositions for inhibiting neoplastic cell growth

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 09403296

Country of ref document: US

ENP Entry into the national phase

Ref document number: 1999 64984

Country of ref document: AU

Kind code of ref document: A

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM 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 MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW 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 BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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

Kind code of ref document: A3

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK DM 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 MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW 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 BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 141551

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 510212

Country of ref document: NZ

Ref document number: 64984/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200101628

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/002820

Country of ref document: MX

Ref document number: 1020017003430

Country of ref document: KR

ENP Entry into the national phase

Kind code of ref document: A

Ref document number: 2343006

Country of ref document: CA

Ref document number: 2000 570324

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 1999952930

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999952930

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 1020017003430

Country of ref document: KR

WWW Wipo information: withdrawn in national office

Ref document number: 1999952930

Country of ref document: EP

WWR Wipo information: refused in national office

Ref document number: 1020017003430

Country of ref document: KR