NZ719369B2 - Conjugates of Cell Binding Molecules with Cytotoxic Agents - Google Patents

Conjugates of Cell Binding Molecules with Cytotoxic Agents Download PDF

Info

Publication number
NZ719369B2
NZ719369B2 NZ719369A NZ71936912A NZ719369B2 NZ 719369 B2 NZ719369 B2 NZ 719369B2 NZ 719369 A NZ719369 A NZ 719369A NZ 71936912 A NZ71936912 A NZ 71936912A NZ 719369 B2 NZ719369 B2 NZ 719369B2
Authority
NZ
New Zealand
Prior art keywords
alkyl
mab
independently
aryl
mmol
Prior art date
Application number
NZ719369A
Other versions
NZ719369A (en
Inventor
Yourang Ma
Yue Zhang
R Yongxin Zhao
Original Assignee
Hangzhou Dac Biotech Co Ltd
Filing date
Publication date
Application filed by Hangzhou Dac Biotech Co Ltd filed Critical Hangzhou Dac Biotech Co Ltd
Priority to NZ719369A priority Critical patent/NZ719369B2/en
Publication of NZ719369A publication Critical patent/NZ719369A/en
Publication of NZ719369B2 publication Critical patent/NZ719369B2/en

Links

Abstract

conjugate of a cytotoxic agent with a cell-surface receptor binding molecule having a structure of Formula (I) as defined in claim 1. The conjugate is used for targeted treatment of cancer, autoimmune disease, and infectious disease.

Description

CONJUGATES OF CELL BINDING MOLECULES WITH CYTOTOXIC AGENTS 1. FIELD OF THE INVENTION This invention relates to conjugates of potent antimitotic agents with a cell-surface receptor binding molecules for targeted therapy. The invention also relates to use of the compositions comprising binding molecule-antimitotic agent conjugates for treating cancer, autoimmune disease, and infectious disease. 2. BACKGROUND OF THE INVENTION There are many articles appeared on the attempted specific targeting of pathogenic cells, in particular, cancer cells utilizing cytotoxic drug conjugated to cell-surface receptor binding agents, such as antibodies (Sela et al, in Immunoconjugates 189-216 (C. Vogel, ed. 1987); Ghose et al, in Targeted Drugs 1-22 (E. Goldberg, ed. 1983); Diener et al, in Antibody mediated delivery systems 1-23 (J. Rodwell, ed. 1988); Silverstein, Nat. Immunol. 2004, 5, 1211–7; Fanning et al, Clin. Immunol. Immunopathol. 1996, 79, 1–14; Ricart A.D., et al., Nature Clinical Practice Oncology 2007, 4, 245-255 ; Singh R. et al Rickson H.K., Therapeutic Antibodies: Methods and Protocols, 2009, 525, 445-467), folic acids (Sudimack, J. et al, Adv. Drug Delivery Rev. 2000, 41, 147-162; Reddy, et al, Mol. Pharm. 2009, 6, 1518-25); PMSA (prostate specific membrane antigen) binding ligands (Low, et al, A1); albumin with peptides (Temming, et al, Bioconjugate Chem. 2006, 17, 1385-1394); Cobalamin and proteins (Gupta, et al, Crit. Rev.
Therap. Drug Carrier Syst. 2008, 25, 347-79; Petrus, et al, Angew. Chem. Int. Ed. 2009, 48, 1022-8); carbohydrate (Darbre, et al, Curr. Top. Med. Chem.. 2008, 8, 1286-93); bioactive polymers (Dhar, et al, Proc. Natl. Acad. Sci. 2008, 105, 17356-61); dendrimers (Lee, et al, Nat.
Biotechnol. 2005, 23, 1517-26; Almutairi, et al; Proc. Natl. Acad. Sci. 2009, 106, 685-90); nanoparticles (Liong, et al, ACS Nano, 2008, 19, 1309-12; Medarova, et al, Nat. Med. 2007, 13, 372-7; Javier, et al, Bioconjugate Chem. 2008, 19, 1309-12); liposomes (Medinai, et al, Curr.
Phar. Des. 2004, 10, 2981-9); viral capsides (Flenniken, et al, Viruses Nanotechnol. 2009, 327, 71-93), etc.
Different families of cytotoxic agents like calicheamicin derivative (Giles, et al Cancer 2003, 98, 2095-104; Hamann, et al, Bioconjug Chem 2002, 13, 47-58), maytansin derivative (Widdison, et al, J Med Chem 2006, 49, 4392-408; Ikeda, et al, Clin Cancer Res 2009, 15, 4028 - 37; Xie, et al, Expert Opin Biol Ther 2006, 6, 281-91), auristatins (Sutherland, et al, J Biol Chem 2006, 281, 10540-7; Doronina, et al, Bioconjug Chem 2006, 17, 114-24), taxane derivatives (Miller, et al, J Med Chem 2004, 47, 4802-5; WO 06061258), leptomycine derivatives (WO 07144709), CC-1065 and analogues (Suzawa, et al, J Control Release 2002, 79, 229-42; Suzawa, et al, Bioorg Med Chem 2000, 8, 2175-84; WO 2007102069), doxorubicin (Trail, et al, Science 1993, 261, 212-5; Saleh et al, J Clin Oncol 2000, 18, 2282-92), or like methotrexate, daunorubicin, vincristine, vinblastine, melphalan, mitomycin C, chlorambucil have been used for the conjugation with a cell-surface receptor binding agent, in particular with antibodies (Wu, et al, Nat. Biotechnol. 2005, 23, 1137–1146. Ricart, et al, Nat. Clin. Pract. Oncol. 2007, 4, 245–255).
The use of a cell-surface receptor binding agent, particularly a targeting antibody having an affinity for the pathogenic cells makes it possible to deliver the cytotoxic agent directly in the vicinity or directly in the pathogenic cell, thus increasing the efficiency of the cytotoxic agent while minimizing the side-effects commonly associated with the cytotoxic agents.
Several short peptidic compounds that found to have biological activity have been isolated from natural sources. One of them, Tubulysins (structures shown below), which were the first time isolated by Hofle and Reichenbach et al. (GBF Braunschweig) from a culture browth Tup, R = H Mep Ile Tuv iii Tut, R = OH H R O R COOH i ii iii Tubulysin R R R A CH OCOCH CH(CH ) OCOCH OH 2 2 3 2 3 B CH OCOCH CH CH OCOCH OH 2 2 2 3 3 C CH OCOCH CH OCOCH OH 2 2 3 3 D CH OCOCH CH(CH ) OCOCH H 2 2 3 2 3 E CH OCOCH CH CH OCOCH H 2 2 2 3 3 F CH OCOCH CH OCOCH H 2 2 3 3 G CH OCOCH=CH OCOCH OH 2 2 3 H CH OCOCH OCOCH H 2 3 3 I CH OCOCH OCOCH OH 2 3 3 U H OCOCH H V H OH H Z H OH OH Pretubulysin CH H H (The structures of existing tubulysin compounds) of the myxobacterial strains of Archangium gephyra (F. Sasse et al. J. Antibiot. 2000, 53, 879-885; WO9813375), are members of group of antimitotic peptides that inhibit tubulin polymerization in dividing cells, and thus inducing apoptosis. With the exceptional potency exceeding that of vinblastine, taxol and epothilones (Wipf, et al, Org. Lett. 2004, 6, 4057–60; Peltier, et al, J. Am.
Chem. Soc. 2006, 128, 16018-9; Wipf, et al, Org. Lett., 2007, 9, 1605–1607; Wang, et al, Chem.
Biol. Drug Des. 2007, 70, 75-86; Pando, et al, Org. Lett. 2009, 11, 5567-9), these antimitotic peptides are exciting leads for targeted therapies. Structurally, the tetrapeptide tubulysins are comprising of N-methylpipecolinic acid (Mep) at the N-terminus, isoleucine (Ile) as the second residue, the unique thiazole-containing tubuvaline (Tuv) as the third residue, and two possible γ- amino acids at the C-terminus (tubutyrosine (Tut) or tubuphenylalanine (Tup)). Despite several tubulysins have recently been synthesized, significant general toxicities (>20% animal body weight loss) of the existing tubulysins at doses required for achieving a therapeutic effect compromise their efficacy (US Patent appl. 2010/0048490). We have been interested in the art of a conjugate of a cell surface binding ligand, particularly using an antibody to conjugate with tubulysin derivaties for having significantly lower general toxicity, yet useful therapeutic efficiency. However, the tubulysins are hardly soluble in a buffer solution, resulting in significant amount of antibody aggregation when conjugated with the tubulysins. A simpler analog, such as using phenyl analine (Phe) and tyrosine (Tyr) to replace Tup and Tut components respectively for the antibody conjugation leads to the hydrolysis of Phe and Tyr in the animal blood circulation and generates the much less potent (over 300 fold less) Mep-Ile-Tuv moiety. Here this patent discloses conjugates of a cell surface binding ligand with soluble and stable tubulysin derivaties and using these conjugates for treating cancer and immune disorders. 3. SUMMARY OF THE INVENTION In one illustrative embodiment of the invention provides a conjugate of formula (I): R R O and pharmaceutical acceptable salts and solvates thereof Wherein T is a targeting or binding ligand; L is a releasable linker; is a linkage bond that L connects to an atom inside the bracket independently; n is 1-20 and m is 1-10; 1 2 3 4 Inside the bracket is a potent antimitotic agent wherein R , R R and R are independently C -C of alkyl; C -C of heteroalkyl, , alkylcycloalkyl, heterocycloalkyl; C -C of aryl, Ar-alkyl, 1 8 2 8 3 8 heterocyclic, carbocyclic, cycloalkyl, heteroalkylcycloalkyl, alkylcarbonyl; or two R's, such as 1 2 1 3 2 3 3 4 5 6 12 13 R R R R R R R R , R R , and R R can be 3-7 members of a carbocyclic, cycloalkyl, or , , , 1 3 4 heterocyclic, heterocycloalkyl ring system; Y is N or CH; In addition, R , R , and R can be H, and R can be absent; 6 8 10 Wherein R R , R and R are independently selected from H and C -C of alkyl or C -C , 1 4 2 4 of heteroalkyl. 7 14 14 1 15 14 1 15 1 Wherein R is selected from H, R or -R C(=O)X R ; -R X R X is selected from O, S, S-S, NH, or NR 9 14 14 14 14 15 Wherein R is H, -O-, -OR , -OC(=O)R -, -OC(=O)NHR -, -OC(=O) R SSR -, 14 14 15 OP(=O)(OR )-, or OR OP(=O)(OR ); 11 14 14 16 14 2 16 14 2 2 Wherein R is H, R , -R C(=O)R , -R X R , -R C(=O)X , wherein X is -O-, -S-, - 14 14 14 14 14 NH-, -N(R )-, -O-R -, -S-R -, -S(=O)-R -, or -NHR -; 12 14 14 Wherein R is R , -O-, -S-, -N-, =N-, =NNH-, -OH, -SH, -NH , =NH, =NNH , -NH(R ), 14 16 16 14 14 14 -OR , -C(O)O-, -C(O)OR -,-COR , -COOR -, C(O)NH-, C(O)NH , C(O)NHR , -SR , - 14 16 16 16 16 S(=O)R , -P(=O)(OR ) , -OP(=O)(OR ) , -CH OP(=O)(OR ) , -SO R ; 2 2 2 Wherein R is C -C of alkyl, alkyl acid, alkyl amide, alkyl amine; or C -C of 1 10 2 10 heteroalkyl; or C -C of Ar; Ar refers to an aromatic or hetero aromatic group, composed of 3 10 one or several rings, comprising four to ten carbon atoms, preferentially four to six carbon atoms. The term of hetero aromatic group refers to an aromatic group that has one or several carbon atoms replaced by hetero atoms, preferentially one, two or three carbon atoms replaced by O, N, Si, Se, P or S, more preferentially O, S, N. The term aryl or Ar also refers to an aromatic group, wherein one or several H atoms can be replaced independently by R , F, Cl, Br, 16 16 16 17 16 16 16 17 16 17 16 16 16 I, OR , SR , NR R , N=NR , N= R , NR R , NO SOR R , SO R , SO R OSO R , 2, , 2 3 3 16 17 16 17 16 17 17 17 17 PR R , POR R , PO R R , OP(O)(OR ) , OCH OP(O)(OR ) , OC(O)OP(O)(OR ) , 2 2 2 2 2 16 17 17 17 17 17 PO(OR )(OR ), OP(O)(OR )OP(O)(OR ) , OC(O)R or OC(O)NHR ; 14 15 Wherein R and R are independently H; C -C of alkyl; C -C of alkenyl, alkynyl, 1 8 2 8 heteroalkyl, heterocyclic, carbocyclic; C -C of aryl, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl; 14 14 Wherein when R is bivalent, it is a R that is further connected to an additional functional group of one to four amino acid units, or (CH CH O) r is an integer ranging from 0 to 12, or 2 2 r, C -C glycosides, or C -C of carboxylic acid. 4 12 1 8 16 14 Wherein R is H, OH, R or one to four amino acid units; Wherein R is H, C -C of alkyl; C -C of alkenyl, alkynyl, heteroalkyl, heterocyclic; C - 1 8 2 8 3 C of aryl, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl or C -C glycosides, or pharmaceutical salts; 4 12 12 13 In addition, R can be H when R is: k=0~4 , , wherein Z 18 18 18 18 18 18 is H, CH OP(O)(OR ) , C(O)OP(O)(OR ) , PO(OR ) , P(O)(OR )OP(O)(OR ) , C(O)R , 2 2 2 2 2 18 18 C(O)NHR , SO (OR ), C -C glycosides, C -C of alkyl, carboxyalkyl, or C -C of 2 4 12 1 8 3 8 heterocyclic; Wherein R is H, C -C of alkyl; C -C of carboxyalkyl, heteroalkyl, alkenyl, 1 8 2 8 19 18 alkynyl; C -C of aryl, alkylcarbonyl, heterocyclic; Wherein R is H, OH, NH , OSO (OR ), 3 8 2 2 18 18 18 18 18 XCH OP(O)(OR ) , XPO(OR ) XC(O)OP(O)(OR ) , XC(O) R , XC(O)NHR C -C of 2 2 2 2 1 8 alkyl, carboxylic acid derivative; C -C of alkenyl, alkynyl, carboxyalkyl C -C of aryl, 2 8 3 8 heterocyclic, alkylcarbonyl; or pharmaceutical salts; X is O, S, NH; Y and Y are N or CH, independently; 12 11 Or R can be H when R is: 2 8 8 , , wherein X is O, S, N-R ; R is H, C -C of alkyl or C -C of heteroalkyl; In another embodiment, the linker L of the potent antimitotic agent- binding molecule conjugates has the formula: --Ww—(Aa)r--Vv—; wherein: --W-- is a Stretcher unit; w is 0 or 1; each --Aa-- is independently an Amino Acid unit; r is independently an integer ranging from 0 to 12; --V-- is a Spacer unit; and v is 0, 1 or 2. The Stretcher unit W may indepen-dently contain a self-immolative spacer, peptidyl units, a hydrazone bond, disulfide or thiolether bonds.
In another embodiment, the cell-surface binding molecule T may be of any kind presently known, or which become known cell binding ligands, such as peptides and non-peptides.
Generally the binding molecule T is an antibody; a single chain antibody; an antibody fragment that binds to the target cell; a monoclonal antibody; a single chain monoclonal antibody; or a monoclonal antibody fragment that binds the target cell; a chimeric antibody; a chimeric antibody fragment that binds to the target cell; a domain antibody; a domain antibody fragment that binds to the target cell; adnectins that mimic antibodies; DARPins; a lymphokine; a hormone; a vitamin; a growth factor; a colony stimulating factor; or a nutrient-transport molecule (a transferrin); a binding peptide, or protein, or antibody, or small molecule attached on albumin, polymers, dendrimers, liposomes, nanoparticles, vesicles, (viral) capsids. Preferably the binding molecule T is a monoclonal antibody.
In yet another aspect, a compound of formula I~VII or a pharmaceutically acceptable salt or solvate thereof is used for treating cancer, an autoimmune disease or an infectious disease in a human or an animal. 4. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows the synthesis of branched linkers for conjugation of drugs to cell binding agents.
Figure 2 shows the synthesis of maleimido linkers and their use for the drug-binding molecule conjugation.
Figure 3 shows the synthesis of bromomaleimido and dibromomaleimido linkers and their uses for the drug-binding molecule conjugation.
Figure 4 shows the synthesis of amino acid (Val-Cit) linkers for the conjugation antimitotic agents with a cell surface binding ligand.
Figure 8 shows the synthesis of Tuv, Ile-Tuv and Mep-Ile-Tuv moieties of the antimitotic agents.
Figure 9 shows the synthesis of Ile-Tuv, Mep-Leu-Tuv, Val-Ile-Tuv and Val-Ile-Tuv (O- alkyl) moieties of the antimitotic agents.
Figure 10 shows the synthesis of a conjugatable antimotic agent and its conjugation to an antibody.
Figure 11 shows the synthesis of amino acid (Phe-(D)Lys) linkers for conjugation of the antimitotic agents.
Figure 12 shows the synthesis of an antibody-antimitotic agent conjugate.
Figure 13 shows the synthesis of antibody-antimitotic agent conjugates.
Figure 14 shows the synthesis of the binding molecule-antimitotic agent conjugates.
Figure 15 shows the synthesis of a binding molecule-antimitotic agent conjugate.
Figure 16 shows the synthesis of an antibody-antimitotic agent conjugate.
Figure 17 shows the synthesis of an antibody-antimitotic agent conjugate.
Figure 18 shows the synthesis of Tut analogs for the synthesis of the the hydrophilic (phosphate prodrug of) antimitotic agents.
Figure 19 shows the synthesis of the conjugates of the antimitotic agents with an antibody.
Figure 20 shows the Boc solid-phase synthesis of the conjugatable antimitotic agents Figure 21 shows the Fmoc solid-phase synthesis of the conjugatable antimitotic agents.
Figure 22 shows the synthesis of the hydrophilic antimitotic drugs and their conjugates with an antibody.
Figure 23 shows the synthesis of Tuv derivatives and the solid phase synthesis of components of Mep-Ile-Tuv and NMe -Val-Ile-Tuv.
Figure 24 shows the synthesis of the antimitotic agents and their conjugation with an antibody.
Figure 25 shows the solid phase synthesis of an antimitotic agent and its conjugate with an antibody.
Figure 26 shows the solid-phase synthesis of antimitotic drugs and their conjugates with an antibody.
Figure 27 shows the synthesis of transarylcyclopropylamines, transarylcyclopropyl- carboxyl acids and transarylethylepoxidyl carboxyl acids.
Figure 28 shows the synthesis of alkene amino acids and alkyl epoxidyl amino acids Figure 29 shows the synthesis of hydrophilic Tut analogs for the synthesis of the hydrophilic prodrugs of antimitotic agents Figure 30 shows the synthesis of the hydrophilic prodrugs of antimitotic agents for the conjugation with a cell binding agent.
Figure 31 shows the synthesis of the hydrophilic prodrugs of antimitotic agents for the conjugation with an antibody.
Figure 32 shows the synthesis of the hydrophilic prodrugs of antimitotic agents for the conjugation with an antibody.
Figure 33 shows the in vitro cytotoxic effects of antiCD22 antibody-antimitotic agent (TZ01~TZ09) conjugates with drug /antibody ratio (D/A) 3.0 ~ 4.3 on Ramos (Burkitt lymphoma cell line). The cells were incubated with the conjugates for 5 days and the IC50 values are: TZ07, D/A = 3.8, IC =24.0 pM; TZ02, D/A = 4.3, IC =30.8 pM; TZ03, D/A = 4.2, IC =36.7 pM; 50 50 50 TZ04, D/A = 4.0, IC =71.9 pM; TZ09, D/A = 3.5, IC =531 pM; TZ01, D/A = 3.3, IC =660 50 50 50 pM; TZ05, D/A = 3.2, IC =3.41 nM; TZ08, D/A = 3.0, IC =2.79 nM; 50 50 Figure 34 shows the cytotoxic effects of Trastuzumab-antimitotic agent (TZ03, TZ04, and TZ07) conjugates with drug /antibody ratio (DAR) 3.5 ~ 4.0 on KPL-4 (breast cancer cell line). huHerTZ03, DAR=4.0, IC = 0.09 nM; huHerTZ04, DAR=3.5, IC = 0.11 nM; huHer 50 50 TZ07, DAR=3.5, IC = 0.12 nM; huHerTZ03 plus huHer2 (1uM), IC > 20 nM; It shows that 50 50 Trastuzumab-TZ03 conjugate induces a specifically potent antipro-liferative effect with IC = 90 pM in the absence of unconjugated Trastuzumab and IC > 20 nM in the presence of 1 micromole concentration of Trastuzumab (to saturate the antigen binding) respectively. The specificity window is > 222 (IC = 20 nM/ IC =0.09 nM) 50 50 Figure 35 shows the cytotoxic effects of antiCD22 antibody-antimitotic agent (TZ03, TZ04, and TZ07) conjugates with drug /antibody ratio (D/A) 3.8 - 4.2 and of unconjugated CD22 antibody as well as of CD20 antibody (Rituximab) on BJAB (Burkitt lymphoma cell line). huCD22-TZ03, D/A =4.2, IC =0.005 nM; huCD22-TZ04, D/A =4.0, IC =0.017 nM; huCD22- 50 50 TZ07, D/A =3.8, IC =0.019 nM; huCD22-TZ03 plus huCD22 (1 uM), IC =3.3 nM; huCD22- 50 50 TZ07 plus huCD22 (1 uM), IC =15 nM; It shows that the conjugate induces much more potent antiproliferative effect –with IC =5 - 19 pM than the unconjugated antibodies with IC > 20 nM. 50 50 The specificity windows when used 1 micromole concentration of unconjugated CD22 antibody to saturate the antigen binding are > 660 (IC = 3.3 nM/ IC =0.005 nM) for huCD22-TZ03 50 50 conjugate and > 790 (IC = 15 nM/ IC =0.019 nM) for huCD-TZ07 conjugate. 50 50 Table 1 displays the structure of the antimitotic drugs made through solid phase synthesis and their molecular ion of mass spectra and in vitro cytotoxicity of these drugs against Ramos cell (ATCC, a human Burkitt’s lymphoma cell).
Table 2 displays the structures of some experimental antibody-antimitotic agent conjugates.
. DETAILED DESCRIPTION OF THE INVENTION .1 DEFINITIONS "Alkyl" means an aliphatic hydrocarbon group which may be straight or branched having 1 to 8 carbon atoms in the chain or branched. "Branched" means that one or more lower alkyl groups such as methyl, ethyl or propyl are attached to a linear alkyl chain. Exemplary alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, t-butyl, n-pentyl, 3-pentyl, octyl, nonyl, decyl, cyclopentyl, cyclohexyl, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 2,2-dimethylpentyl, 2,3- dimethylpentyl, 3,3-dimethylpentyl, 2,3,4-trimethylpentyl, 3-methylhexyl, 2,2-dimethylhexyl, 2,4-dimethylhexyl, 2,5-dimethylhexyl, 3,5-dimethylhexyl, 2,4-dimethylpentyl, 2-methylheptyl, 3- methylheptyl, n-heptyl, isoheptyl, n-octyl, and isooctyl. A C -C alkyl group can be unsubstituted or substituted with one or more groups including, but not limited to, -C ~C alkyl, -O-(C ~C 1 8 1 8 alkyl), -aryl, -C(O)R', -OC(O)R', -C(O)OR', -C(O)NH , -C(O)NHR', -C(O)N(R') -NHC(O)R', - S(O) R', -S(O)R', -OH, -halogen (F, Cl, Br or I), -N , -NH , -NH(R'), -N(R') and -CN; where 2 3 2 2 each R' is independently selected from -C ~C alkyl and aryl.
A "C ~C carbocycle" means a 3-, 4-, 5-, 6-, 7- or 8-membered saturated or unsaturated non-aromatic carbocyclic ring. Representative C ~C carbocycles include, but are not limited to, - cyclopropyl, -cyclobutyl, -cyclopentyl, -cyclopentadienyl, -cyclohexyl, -cyclohexenyl, -1,3- cyclohexadienyl, -1,4-cyclohexadienyl, -cycloheptyl, -1,3-cycloheptadienyl, -1,3,5- cycloheptatrienyl, -cyclooctyl, and -cyclooctadienyl. A C ~C carbocycle group can be unsubstituted or substituted with one or more groups including, but not limited to, --C ~C alkyl, - O-( C ~C alkyl), -aryl, -C(O)R', -OC(O)R', -C(O)OR', -C(O)NH , -C(O)NHR', -C(O)N(R') - 1 8 2 2 NHC(O)R', -S(O) R', -S(O)R', -OH, -halogen, -N , -NH , -NH(R'), -N(R') and -CN; where each 2 3 2 2 R' is independently selected from -C ~C alkyl and aryl.
A "C ~C carbocyclo" refers to a C ~C carbocycle group defined above wherein one of 3 8 3 8 hydrogen atoms on the carbocycle is replaced with a bond.
"Alkenyl” means an aliphatic hydrocarbon group containing a carbon-carbon double bond and which may be straight or branched having 2 to 8 carbon atoms in the chain. Exemplary alkenyl groups include ethenyl, propenyl, n-butenyl, i-butenyl, 3-methylbutenyl, n- pentenyl, hexylenyl, heptenyl, octenyl.
"Alkynyl" means an aliphatic hydrocarbon group containing a carbon-carbon triple bond and which may be straight or branched having 2 to 8 carbon atoms in the chain. Exemplary alkynyl groups include ethynyl, propynyl, n-butynyl, 2-butynyl, 3-methylbutynyl, pentynyl, n- pentynyl, hexylynyl, heptynyl, and octynyl.
“Heteroalkyl” is C ~C alkyl in which one to four carbon atoms are independently replaced with a heteroatom from the group consisting of O, S and N.
"Aryl" or Ar refers to an aromatic or hetero aromatic group, composed of one or several rings, comprising three to fourteen carbon atoms, preferentially six to ten carbon atoms. The term of hetero aromatic group refers one or several carbon on aromatic group, preferentially one, two, three or four carbon atoms are replaced by O, N, Si, Se, P or S, preferentially by O, S, N. The term aryl or Ar also refers to a aromatic group, wherein one or several H atoms are replaced 13 13 13 13 14 13 13 13 14 independently by R , F, Cl, Br, I, OR , or SR , NR R , N=NR , N=R NR R NO , , 2, 13 14 13 13 13 13 14 13 14 13 14 13 14 13 14 SOR R , SO R , SO R OSO R , PR R , POR R , PO R R , OPO R R , or PO R R 2 3 , 3 2 3 3 13 14 wherein R R are independently H, alkyl, alkenyl, alkinyl, heteroalkyl, aryl, arylalkyl, carbonyl, or pharmaceutical salts.
“Halogen atom” refers to fluorine, chlorine, bromine or iodine atom; preferably fluorine and chlorine atom.
“Heterocycle" refers to an aromatic or non-aromatic C ~C carbocycle in which one to four 3 14 of the ring carbon atoms are independently replaced with a heteroatom from the group of O, N, S Se, and P. Preferable heteroatoms are oxygen, nitrogen and sulfur. Suitable heterocycles are also disclosed in The Handbook of Chemistry and Physics, 76 Edition, CRC Press, Inc., 1995-1996, p. 2- to 2-26, the disclosure of which is hereby incorporated by reference. Preferred non aromatic heterocyclic include, but are not limited to pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxiranyl, tetrahydrofuranyl, dioxolanyl, tetrahydro-pyranyl, dioxanyl, dioxolanyl, piperidyl, piperazinyl, morpholinyl, pyranyl, imidazolinyl, pyrrolinyl, pyrazolinyl, thiazolidinyl, tetrahydrothiopyranyl, dithianyl, thiomorpholinyl, dihydro-pyranyl, tetrahydropyranyl, dihydropyranyl, tetrahydro-pyridyl, dihydropyridyl, tetrahydropyrinidinyl, dihydrothiopyranyl, azepanyl, as well as the fused systems resulting from the condensation with a phenyl group.
The term “heteroaryl” or aromatic heterocycles refers to a 5 to 14, preferably 5 to 10 membered aromatic hetero, mono-, bi- or multicyclic ring. Examples include pyrrolyl, pyridyl, pyrazolyl, thienyl, pyrimidinyl, pyrazinyl, tetrazolyl, indolyl, quinolinyl, purinyl, imidazolyl, thienyl, thiazolyl, benzothiazolyl, furanyl, benzofuranyl, 1,2,4-thiadiazolyl, isothiazolyl, triazoyl, tetrazolyl, isoquinolyl, benzothienyl, isobenzofuryl, pyrazolyl, carbazolyl, benzimidazolyl, isoxazolyl, pyridyl-N-oxide, as well as the fused systems resulting from the condensation with a phenyl group.
"Alkyl", "cycloalkyl", “alkenyl”, "alkynyl", "aryl", "heteroaryl", "heterocycle" and the like refer also to the corresponding "alkylene", "cycloalkylene", "alkenylene", "alkynylene", "arylene", "heteroarylene", "heterocyclene" and the likes which are formed by the removal of two hydrogen atoms.
“Pharmaceutically” or “pharmaceutically acceptable” refer to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to an animal, or a human, as appropriate.
“Pharmaceutically acceptable excipient” includes any carriers, diluents, adjuvants, or vehicles, such as preserving or antioxidant agents, fillers, disintegrating agents, wetting agents, emulsifying agents, suspending agents, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions as suitable therapeutic combinations.
As used herein, "pharmaceutical salts" refer to derivatives of the disclosed compounds wherein the parent compound is modified by making acid or base salts thereof. The pharmaceutically acceptable salts include the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, tartaric, citric, methanesulfonic, benzenesulfonic, glucoronic, glutamic, benzoic, salicylic, toluenesulfonic, oxalic, fumaric, maleic, lactic and the like. Further addition salts include ammonium salts such as tromethamine, meglumine, epolamine, etc., metal salts such as sodium, potassium, calcium, zinc or magnesium.
The pharmaceutical salts of the present invention can be synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17 ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the disclosure of which is hereby incorporated by reference. .2 DRUG-LINKER- BINDING LIGAND CONJUGATES As stated above, this invention provides a cell surface binding molecule - antimitotic agent conjugate of formula (I): 4 9 12 13 N 11 10 and pharmaceutical acceptable salts and solvates thereof, Wherein T is a targeting or binding ligand; L is a releasable linker; is a linkage bond that L connects to an atom inside the bracket independently; n is 1~20 and m is 1~10. 1 2 3 4 Inside the bracket is a potent antimitotic agent wherein R , R R and R are independently C ~C of alkyl, heteroalkyl; C ~C of heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, 1 8 2 8 heterocycloalkyl; C ~C of aryl, Ar-alkyl, heteroalkylcycloalkyl, alkylcarbonyl; or two R's, such 1 2 1 3 2 3 3 4 5 6 12 13 as R R R R R R R R , R R , and R R can be 3~7 members of a carbocyclic, cycloalkyl, or , , , 1 3 4 heterocyclic, heterocycloalkyl ring system; Y is N or CH; In addition, R , R , and R can be H, and R can be absent. 6 8 10 Wherein R R , R and R are independently selected from H and C ~C of alkyl or , 1 4 heteroalkyl. 7 14 14 1 15 14 1 15 Wherein R is independently selected from H, R or -R C(=O)X R ; -R X R wherein 14 15 R and R are independently selected from C ~C of alkyl, heteroalkyl; C ~C of alkenyl, 1 8 2 8 alkynyl, heterocyclic, carbocyclic, cycloalkyl,; C ~C of aryl, heterocycloalkyl, 1 14 heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl; X is selected from O, S, S-S, NH, or NR 9 14 14 14 Wherein R is independently H, -O-, -OR , -OC(=O)R -, -OC(=O)NHR -, -OC(=O) 14 15 14 14 15 14 15 R SSR -, OP(=O)(OR )-, or OR OP(=O)(OR ), wherein R R are independently C ~C of alkyl, heteroalkyl; C ~C of alkenyl, alkynyl, heterocyclic, carbocyclic; C ~C of aryl, 1 8 2 8 3 8 cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl. 11 14 14 16 14 2 16 14 2 2 Wherein R is independently H, R , -R C(=O)R , -R X R , -R C(=O)X , wherein X 14 14 14 14 14 14 is -O-, -S-, -NH-, -N(R )-, -O-R -, -S-R -, -S(=O)-R -, or -NHR -; R is C ~C of alkyl, heteroalkyl; C ~C of alkenyl, alkynyl, heterocyclic, carbocyclic; C ~C of aryl, cycloalkyl, 2 8 3 8 alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl; R is H, OH, R , or one to four amino acid units; 12 14 Wherein R is independently R , -O-, -OH, -S-, -SH, -NH , -NH-, =NH, =N-, =NNH , 14 14 16 14 14 14 14 =NNH-, -NH(R ), -OR , -COR , -COOR -, -C(O)NH , -C(O)NHR , -SR , -S(=O)R , - 16 16 16 16 14 P(=O)(OR ) , -OP(=O)(OR ) , -CH OP(=O)(OR ) , -SO R . R is independently C ~C of 2 2 2 2 2 1 8 alkyl, heteroalkyl; C ~C of alkenyl, alkynyl, hetero-cyclic, carbocyclic; C ~C of aryl, 2 8 3 8 cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl. 16 14 R is H, OH, R or one to four amino acid units; Wherein R is C ~C of alkyl, heteroalkyl, alkyl acid, alkyl amide, alkyl amine, or Ar; 1 10 Ar refers to an aromatic or hetero aromatic group, composed of one or several rings, comprising four to ten carbon atoms, preferentially four to six carbon atoms. The term of hetero aromatic group refers to an aromatic group that has one or several carbon atoms replaced by hetero atoms, preferentially one, two or three carbon atoms replaced by O, N, Si, Se, P or S, more preferentially O, S, N. The term aryl or Ar also refers to an aromatic group, wherein one or 17 16 16 16 17 several H atoms can be replaced independently by R , F, Cl, Br, I, OR , SR , NR R , 16 16 16 17 16 17 16 16 16 16 17 16 17 N=NR , N= R , NR R , NO SOR R , SO R , SO R OSO R , PR R , POR R , 2, , 2 3 3 16 17 17 17 17 16 17 PO R R , OP(O)(OR ) , OCH OP(O)(OR ) , OC(O)OP(O)(OR ) , PO(OR )(OR ), 2 2 2 2 2 17 17 17 17 16 17 OP(O)(OR )OP(O)(OR ) , OC(O)R or OC(O)NHR , wherein R R are independently H, C ~C of alkyl, heteroalkyl; C ~C of alkenyl, alkynyl, heterocyclic, carbocyclic; C ~C of aryl, 1 8 2 8 3 8 cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl or C ~C glycosides, or pharmaceutical salts. 4 12 12 10 13 In addition, R can be H when R is not H, or when R is: k=0~4 , , 1 18 18 18 wherein Z is H, CH OP(O)(OR ) , C(O)OP(O)(OR ) , PO(OR ) , 2 2 2 2 18 18 18 18 18 P(O)(OR )OP(O)(OR ) , C(O)R , C(O)NHR , SO (OR ), C ~C glycosides, or C ~C of 2 2 4 12 1 8 alkyl, carboxyalkyl, heterocyclic; R is H, C ~C of alkyl, carboxyalkyl, heteroalkyl; C ~C of 1 8 2 8 19 18 alkenyl, alkynyl, heterocyclic; C ~C of aryl, alkylcarbonyl; R is H, OH, NH , OSO (OR ), 3 8 2 2 18 18 18 18 18 XCH OP(O)(OR ) , XPO(OR ) XC(O)OP(O)(OR , XC(O) R , XC(O)NHR C ~C of , ) , 2 2 2 2 1 8 alkyl, carboxyalkyl, carboxylic acid derivative; C ~C of alkenyl, alkynyl, heterocyclic; C ~C 2 8 3 8 of aryl, alkylcarbonyl; or pharmaceutical salts; X is O, S, NH; Y and Y are N or CH respectively. 12 11 Or R can be H when R is: 2 8 6 8 , , wherein X is O, S, N-R ; R and R are H, C ~C of alkyl or heteroalkyl respectively.
Wherein the said conjugate of the invention specifically excludes the following structures: 24 25 26 Wherein R is H or CH ; R is alkyl or C(O)alkyl or C(O)aryl; R is H, C ~C alkyl, 3 1 5 28 27 C ~C alkenyl, C(=O)C ~C alkyl, or C(=O)C ~C alkenyl; R is H or C ~C alkyl; R is H, 1 to 2 5 1 5 2 5 1 5 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, 29 29 haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, or OR , where R is 30 30 30 substituted aryl, C(O)R , P(O)(OR )2, or SO R , where R is selected from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl or a metal cation; T is a targeting or binding ligand; L is a releasable linker.
In another embodiment, conjugates of antimitotic agents have the formula (II) R 13 T 11 Y N R R 10 (II) and pharmaceutical acceptable salts and solvates thereof; 1 2 3 4 5 6 7 8 10 11 13 Wherein T L, n, m, Y, R , R , R , R , R , R , R , R , R , R and R are defined the same as in the claim 1; 9 14 14 14 Wherein R is independently H, -OH, -OR , -OC(=O)R -, -OC(=O)NHR -, -OC(=O) 14 15 14 14 15 R SSR , OP(=O)(OR ) , or OR OP(=O)(OR ) 2 2; 12 14 14 14 Wherein R is independently R , -OH, -SH, -NH , =NH, =NNH , -NH(R ), -OR , - 16 14 14 14 14 16 16 COR , -COOR -, C(O)NH , C(O)NHR , -SR , -S(=O)R , -P(=O)(OR ) , -OP(=O)(OR ) , - 2 2 2 16 16 CH OP(=O)(OR ) , -SO R ; 14 15 16 17 Wherein R , R , R and R are defined the same as in the claim 1; 12 10 13 In addition, R can be H when R is not H, or when R is: k=0~4 k=0~4 , , 1 18 18 18 wherein Z is H, CH OP(O)(OR ) , C(O)OP(O)(OR ) , PO(OR ) , 2 2 2 2 18 18 18 18 18 P(O)(OR )OP(O)(OR ) , C(O)R , C(O)NHR , SO (OR ), C -C glycosides, C -C of alkyl, 2 2 4 12 1 8 carboxyalkyl, or C -C of heterocyclic; R is H, C -C of alkyl, carboxyalkyl; C -C of 3 8 1 8 2 8 heteroalkyl; C -C of alkenyl, alkynyl; C -C of aryl, heterocyclic, alkylcarbonyl; R is H, OH, 2 8 3 8 18 18 18 18 18 NH , OSO (OR ), XCH OP(O)(OR ) , XPO(OR ) XC(O)OP(O)(OR , XC(O) R , 2 2 2 2 2 2 XC(O)NHR C -C of alkyl, carboxyalkyl, carboxylic acid derivative; C -C of alkenyl, 1 8 2 8 alkynyl; C -C of aryl, heterocyclic, alkylcarbonyl; or pharmaceutical salts; X is O, S, NH; Y and Y are N or CH; 12 11 Or R can be H when R is: 2 8 8 wherein X is O, S, N-R ; R is H, C -C of alkyl or C -C of heteroalkyl; In addition, R can be absent.
Illustrative classes of compounds of formula II have the structures: IIa H NH O Aa T S Y' OPO(OH) H X' O HN H O N O Y' S IIh H OAc HN N O X' O NH O N S Z' 23 N T NH N O S H CO H 22 H 23 H T NH R' O S H IIo wherein Aa is a natural or an unnatural amino acid; n is 1 ~ 20; q = 1~5; X , Y and ’ 22 22 23 Z are independently CH, O, S, NH, or NR ; R and R are independently C ~C of alkyl; C ~C of alkenyl, alkynyl, heteroalkyl; C ~C of aryl, heterocyclic, carbocyclic, cycloalkyl, 2 8 3 8 alkylcycloalkyl, heterocycloalkyl, Ar-alkyl, heteroalkylcycloalkyl, heteroaralkyl, or - (OCH CH ) -; R’ and R’’ are independently H or CH . . Inside the round brackets are the 2 2 n 3 antimitotic drugs and the inside square brackets are the antimitotic drug conjugated to a cell binding agent T via a linker.
In another embodiment, a conjugate of a cell binding molecule-antimitotic agent has the formula (III): 8 13 R 10 O 5 6 10 R R R R (III) 1 2 3 4 5 6 8 9 10 11 12 13 wherein T, L, m, Y, R , R R R , R R R R R R R R and n are defined the , , , , , , , , , same as in formular (I) and (II). 7 14 14 1 15 14 1 15 Wherein R is independently selected from H, -R - or -R C(=O)X R - or -R X R - 14 15 wherein R and R are independently C ~C of alkyl, heteroalkyl; C ~C of alkenyl, alkynyl; 1 8 2 8 C ~C of aryl, heterocyclic, carbocyclic, cycloalkyl, heterocycloalkyl, heteroaralkyl 1 14 heteroalkylcycloalkyl, alkylcarbonyl; X is selected from O, S, S-S, NH, or NR Illustrative examples of compounds of formula (III) have the structures: IIIa IIIb IIIc IIId IIIe IIIf IIIg IIIh IIIi IIIj ’ ’ ’ 22 23 Wherein Ar, n, q, X , Y , Z , R , R , R’ and R’’ are defined the same as for formula IIa ~ IIr.
In another embodiment, a binding molecule-antimitotic agent conjugate has the formula (IV) 8 13 3 4 R Y N 11 O 5 6 (IV) 1 2 3 4 5 6 7 8 10 11 12 13 wherein T, L, m, R , R R R , R R R R R R R R and n are defined the same , , , , , , , , , as in formular (II). 9 14 14 14 Wherein R is independently H, -O-, -OR -, -OC(=O)R -, -OC(=O)NHR -, - 14 15 14 14 15 OC(=O)R SSR -, -OP(=O)(OR )O-, wherein R R are independently C ~C of alkyl, , 1 8 heteroalkyl; C ~C of aryl, heteroaryl, heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl or pharmaceutical salts.
Illustrative examples of compounds of formula (IV) have the structures: O 22 S NH R'' O R' q N COOH IV-1 HN q H 22 O NH O 2 IV-2 HN HO R'' R' O 23 N IV-2 IVc IV-2 q H O R'' R' T R' IV-3 q H O T NH O N O O O O H NH IV-4 ’ 22 23 wherein Ar, Aa, n, q, X , Y’, R , R , R’ and R’’ are defined the same as for formula IIa ~ IIr.
In another embodiment, a binding molecule -antimitotic agent conjugate has the formula (V) 8 13 3 4 R R R 6 10 R R R 1 2 3 4 5 6 7 8 9 10 12 13 wherein T, L, m, Y, R , R R R R R R R R R R R and n are defined the same , , , , , , , , , , as in formular (II). 11 14 14 17 14 2 17 14 2 Wherein R is independently H, -R -, -R C(=O)R -, - R X R -, - R C(=O)X -, wherein R is independently H, OH, C ~C of alkyl; C ~C of alkenyl, alkynyl, heteroalkyl; C ~C of aryl, 1 8 2 8 3 8 arylene, heterocyclic, carbocyclic, heterocycloalkyl; or an amino acid, or two amino acid units; 2 14 14 14 14 14 14 X is -O-, -S-, -NH-, -N(R )-, -O-R -, -S-R -, -S(=O)-R -, or -NHR -; R is C ~C of alkyl, heteroalkyl; C ~C of alkenyl, alkynyl; C ~C of aryl, heterocyclic, carbocyclic, cycloalkyl, 2 8 3 8 alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl.
Illustrative examples of compounds of formula (V) have the structures: N 22 S 23 H R' (HO) OP N R' N N S R R' R'' H V-2 H OAc O OPO(OH) OPO(OH) OPO(OH) N Aa 22 S H OAc O H 22 V-6 O O N N V-7 O Vg O N N H H n R' R'' ’ 22 23 Wherein Ar, Aa, n, q, X , R , R , R’ and R’’ are defined the same as for formula IIa ~ IIr.
In another embodiment, a conjugates of a cell binding-antimitotic agent have the formula (VI) 3 13 R O R R 10 (VI) 1 2 3 4 5 6 7 8 9 11 13 wherein T, L, m, Y, R , R R R R R R R R R R and n are defined the same as , , , , , , , , , in formular (II). 12 14 14 14 Wherein R is independently R , -O-, -S-, -NH-, =N-, =NNH-, -N(R )-, -OR -,C(O)O-, 14 14 14 14 15 15 C(O)NH-, C(O)NR -, -SR -, -S(=O)R - -NHR -, -CH OP(=O)(OR )- , -P(=O)(OR )-,. - 14 14 15 OP(=O)(OR )O-, -SO R , R R are independently C ~C of alkyl, heteroalkyl; C ~C of 2 , 1 8 2 8 alkenyl, alkynyl; C ~C of aryl, heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl.
Illustrative examples of compounds of formula VI have the following structures: 22 S R' R'' VI-1 R' R'' VI-2 NH T VI-3 N NH N R'' N R' T O HN 22 N VI-3 H O O COOH VI-3' O N R' R'' N NH O R' R'' VI-4 Ar R'' OAc N Ar 23 N R N Ar R' H O O N 22 H Ar VI-5 R' R'' N S N R N 22 T VI-6 VI-7 ’ 22 23 Wherein Ar, Aa, n, q, X , R , R , R’ and R’’ are defined the same as for formula IIa ~ IIr.
In another embodiment, the conjugates of the cell-surface binding molecule-antimitotic agents have the formula (VII) R 13 O S 10 (VII) wherein T, L, m, Y, R , R R R R R R R R R R and n are defined the same as 1 2, 3, 4, 5, 6, 7, 8, 9, 11, 12 in formular (II).
Wherein R is C ~C of alkyl, heteroalkyl, alkyl acid, alkyl amide, alkyl amine, or Ar; Ar 1 10 refers to a aromatic or hetero aromatic group, composed of one or several rings, comprising four to ten carbon, preferentially four to six carbon atoms. The term of hetero aromatic group refers one or several carbon on aromatic group, preferentially one, two or three carbon atoms are replaced by O, N, Si, Se, P or S, preferentially O, S, N. The term aryl or Ar also refers to a aromatic group, wherein one or several H atoms are replaced independently by R , F, Cl, Br, I, 16 16 16 18 16 16 16 18 16 18 16 16 16 OR , SR , NR R , N=NR , N=R NR R NO SOR R , SO R , SO R OSO R , , , 2, 2 3 , 3 16 18 16 18 16 18 16 18 16 18 16 18 PR R , POR R , PO R R , OPO R R , or PO R R wherein R R are independently H, 2 3 3 , C ~C of alkyl; C ~C of alkenyl, alkynyl, heteroalkyl; C ~C of aryl, heterocyclic, carbocyclic, 1 8 2 8 3 8 cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl; or C ~ C glycosides; or pharmaceutical salts. 4 12 12 13 In addition, R can be H when R is: Y OZ Y 18 k = 0 ~4 k=0~4 k = 0~4 , , . 1 18 18 18 wherein Z is H, CH OP(O)(OR )2, C(O)OP(O)(OR ) , PO(OR ) , 2 2 2 18 18 18 18 18 PO(OR )OPO(OR ) , C(O)R , C(O)NHR , SO (OR ), C ~C glycosides, or C ~C of alkyl, 2 2 4 12 1 8 carboxyalkyl, heterocyclic; R is H, C ~C of alkyl, carboxyalkyl, heteroalkyl; C ~C of 1 8 2 8 19 18 alkenyl, alkynyl, heterocyclic; C ~C of aryl, alkylcarbonyl; R is H, OH, NH , OSO (OR ), 3 8 2 2 18 18 18 18 18 XCH OP(O)(OR ) , XC(O)OP(O)(OR ) , XPO(OR ) , XC(O)R , XC(O)NHR , C ~C of 2 2 2 2 1 8 alkyl, carboxyalkyl, carboxylic acid derivative; C ~C of alkenyl, alkynyl, heterocyclic; C ~C 2 8 3 8 of aryl, alkylcarbonyl; or pharmaceutical salts; X is O, S, NH; Y and Y are N or CH respectively. 12 11 Or R can be H when R is: 2 8 8 wherein X is O, S, N-R ; R is H, C -C of alkyl or C -C of heteroalkyl; Illustrative examples of compounds of formula VII have the following structures: (HO) OPO H NH q H N O OAc N 22 O NH 2 H n VII-1 VIIa R' R'' R' R'' VII-2 VIIb OPO(OH) OAc 2 N S H S Aa O R' 24 O R'' VII-3 VIIc OPO(OH) OAc R' R'' R'' O n VII-4 OH VIId OPO(OH) S H O VII-4 VIIe OPO(OH) N O T 24 H N VII-4 OH 2 H VIIf OPO(OH) O OAc N O T OH n VII-4 O VIIg OSO H N O T VII-5 OH n VIIh O OH H O OAc X' 22 N O T VII-6 OH n VIIi OSO H R' T O H H R' R'' n 24 O VII-5 OH VIIj O OH H OAc OH O N 23 R' T O H R' H VII-6 VIIk O OH H OAc q OH 24 O VII-6 R OH VIIl OSO H H OAc VII-5 OH VIIm OPO(OH) H OAc HN 22 VII-7 VIIn NH H O OAc N O T O HN O NH VII-8 O VIIo VIIP H OAc X' O O S O O S n VII-9 VIIq H OAc O O N H H N VII-9 R VIIr H OAc HN R 24 OH VII-10 VIIs H OAc q O N 24 O VII-11 R H N VIIt ’ 22 23 Wherein Ar, Aa, n, q, X , R , R , R’ and R’’ are defined the same as for formula IIa ~ IIf; R is H or CH .
In another embodiment, the synthetic routes to produce the antimitotic agents and their conjugation to a cell-surface receptor binding molecules of the present invention are exampled, but not limited to, shown in figures 1-32.
In another embodiment, the releasable linker (L) is a chain of atoms selected from C, N, O, S, Si, and P that covalently connects the cell-surface binding ligand (T) to the potent antimitotic agents. The linker may have a wide variety of lengths, such as in the range from about 2 to about 100 atoms. The atoms used in forming the linker may be combined in all chemically relevant ways, such as forming alkylene, alkenylene, and alkynylene, ethers, polyoxyalkylene, esters, amines, imines, polyamines, hydrazines, hydrazones, amides, ureas, semicarbazides, carbazides, alkoxyamines, alkoxylamines, urethanes, amino acids, acyloxylamines, hydroxamic acids, and many others. In addition, it is to be understood that the atoms forming the releasable linker (L) may be either saturated or unsaturated, or may be radicals, or may be cyclized upon each other to form divalent cyclic structures, including cyclo alkanes, cyclic ethers, cyclic amines, arylenes, heteroarylenes, and the like in the linker.
The term releasable linker refers to a linker that includes at least one bond that can be broken under physiological conditions, such as a pH-labile, acid-labile, base-labile, oxidatively labile, metabolically labile, biochemically labile, or enzyme-labile bond. It is appreciated that such physiological conditions resulting in bond breaking do not necessarily include a biological or metabolic process, and instead may include a standard chemical reaction, such as a hydrolysis or substitution reaction, for example, an endosome having a lower pH than cytosolic pH, and/or disulfide bond exchange reaction with a intracellular thiol, such as the amillimolar range of abundant of glutathione inside the malignant cells.
The releasable linker L of conjugates may have the formula: --Ww—(Aa)r--Vv-- wherein: - -W-- is a Stretcher unit; w is 0 or 1; each --Aa-- is independently an Amino Acid unit; r is independently an integer ranging from 0 to 12; --V-- is a Spacer unit; and v is 0, 1 or 2.
The Stretcher unit (--W--), when present, links a targeted binding molecular unit (T) to an amino acid unit (--Aa--), or links V when an Aa is not present. The Stretcher unit W may independently contain a self-immolative spacer, peptidyl units, a hydrazone bond, disulfide or thiolether bonds. In this regard a binding molecular (T) has a functional group that can form a bond with a functional group of a Stretcher. Useful functional groups that can be present on a binding molecular, either naturally or via chemical manipulation include, but are not limited to, sulfhydryl (--SH), amino, hydroxyl, carbonyl, the anomeric hydroxyl group of a carbohydrate, and carboxyl. Preferred functional groups are sulfhydryl, carboxy and amino. Sulfhydryl groups can be generated by reduction of an intramolecular disulfide bond of a Ligand. Alternatively, sulfhydryl groups can be generated by reaction of an amino group of a lysine moiety of a binding molecular using 2-iminothiolane (Traut's reagent) or thiolactone or another sulfhydryl generating reagent, such as modifies T with a disulfide bond linker, or a thiol ester following by reduction or hydrolysis respectively.
Illustrative examples of W linked to T have the structures: T N T 20 S R N T NHNH O R' R'' H ; ; ; R N T NHNH HN O R N T ; ; ; S O NH+ O 20 R N T R N T H ; H ; H ; R'' R' 20 S R N N N O N C T S S T H H H ; ; ; O O 21 N T R N T S H H O ; ; ; 20 R S R S O H H 20 N R N R N T N T H O O Ar O O ; ; NH+ O NHNH R N T H ; ; ; 20 NHNH , , , , , , , , 21 , wherein R and R are selected from –C ~C alkylene-, -C ~C carbocyclo-, -O-( C ~C alkyl)-, -arylene-, -C ~C alkylene- 1 9 1 7 1 8 1 9 arylene-, -arylene, -C ~C alkylene-, -C ~C alkylene-(C ~C carbocyclo)-, -(C ~C carbocyclo)- 1 9 1 9 1 8 3 7 C ~C alkylene-, -C ~C heterocyclo-, -C ~C alkylene-(C ~C heterocyclo)-, -(C ~C 1 9 3 8 1 10 3 8 3 8 heterocyclo)- C ~C alkylene-, -(CH CH O) -, -(CH(CH )CH O) -, and -(CH CH O) -CH -; k is 1 9 2 2 k 3 2 k 2 2 k 2 an integer ranging from 1-20.; R’ and R’’ are independently H or CH .
In another embodiment, conjugation of W to T covalently as illustrated above can be via various chemical reactions.
Examples of the formation of amide linkages: + H N T N T R' R'' O O O O H N T + N T 2 NNH R Wherein the Stretcher unit contains a reactive site of E, which can form an amide bond with a primary or secondary amino group of a Ligand. Example of the reactive E, includes, but is not limited to, such as hydroxysuccinimidyl esters (NHS, Sulfo-NHS, etc), 4-nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl (includes sulfo-tetrafluorophenyl) esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates and isothiocyanates.
Examples of thiol ether or disulfide bond linkages: HS T 2 20 S HS H N T N T O R'' R' R'' R' S R'' R' R'' R' + HS T S S T Wherein the Stretcher unit contains a sulfhydryl reactive site, which can form a thiol ether or disulfide bond with a thiol group which is generated by reduction of an intramolecular disulfide bond of the binding ligand T, or generated by a chemical modification on the binding ligand T.
In yet another aspect of the invention, the reactive group of the Stretcher contains a reactive site that is reactive to an aldehyde (--CHO) or a ketone (-C(=O)R) group that can be chemically modified on a binding molecular T. For example, a carbohydrate on a binding molecular T can be mildly oxidized using a reagent such as sodium periodate to generate an aldehyde or a ketone (- C(=O)R) group; or an amine on an amino acid at the N-termini of antibodies (or proteins or peptides ) can react with pyridoxal 5’-phosphate (PLP) in a buffer solution to introduce ketone groups (Scheck & Francis, ACS Chem. Biol. 2007, 2, 247-251). The resulting (--C=O) unit can be condensed with a Stretcher that contains a functionality such as a hydrazide, an oxime, a primary or secondary amine, a hydrazine, a thiosemicarbazone, a hydrazine carboxylate, and an arylhydrazide.
Examples of the conjugation of the hydrazone, or the oxime or imine linkages: T + PLP (or NaIO ) 21 20 21 S R N R NH S R N R N 25 T + PLP (or NaIO ) S O C N N N NH S 20 R 25 R 25 R H R 21 25 wherein R and R are described above, R is an organic substituent of an amino acid.
In another aspect of the invention, the Stretchers (which may contain a spacer V and/or an amino acid) can be linked to the binding molecules (T), followed by conjugation of a potent antimitotic agent to the binding molecule-stretcher moiety in an aqueous buffered solution.
Examples of these kinds of two-step conjugations (a drug linked to R is omitted here): 26 26 O O 20 H N T 16 S S R 2 20 T S SH E N 16 R' R'' H R' R'' R'' H N T R E N R N N T O O O Ar NHNH H N T 20 Ar E Ar N T 16 20 N 20 H H N T R 16 N N T N N T O Br O O 16 S 20 H H N T R N R R N N T N N T Br 16 H N T T 2 J R T wherein E includes, but is not limited to, such as hydroxysuccinimidyl esters (NHS, Sulfo- NHS, etc), 4-nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl (includes sulfo- tetrafluorophenyl) esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates and 16 isothiocyanates. R’ and R’’ are independently H or CH ; R , R and Ar are defined in various embodiment through out this inventions; R is H, or F, or NO independently; J is F, Cl, Br, I, tosylate (TsO) or mesylate (MsO) independently and wherein bears at least one antimitotic agent/drug .
In another aspect of the invention, the Stretchers can be linked to a potent antimitotic agent first, followed by conjugation of the binding molecules (T) in an aqueous pH 3 ~ 10 (preferably pH 5 ~ 8.5) buffered solution containing up to 50% of organic cosolvents. Examples of these kinds of two-step conjugations: 20 H N T S SH 16 R 2 16 20 R R S R S R S R R' R'' R' R' R'' R 16 20 R R E N N H N T 16 N Ar O O O Ar H N T 16 NHNH R 20 16 N NHN 16 20 Ar E E R NHN R R R 20 SH R 16 16 E H N T H N E R 2 O 16 O R O S S 16 R H N T H R E 2 R O 16 S O 16 S T O O O O R SH 16 H N T J R S wherein E includes, but is not limited to, such as hydroxysuccinimidyl esters (NHS, Sulfo- NHS, etc), 4-nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl (includes sulfo- tetrafluorophenyl) esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates and 16 20 isothiocyanates. R’ and R’’ are independently H or CH ; R , R and Ar are defined in various embodiment through out this inventions; R is H, or F, or NO independently; J is F, Cl, Br, I, tosylate (TsO) or mesylate (MsO) independently and wherein 16 bears at least one antimitotic agent/drug.
The Amino Acid unit (--Aa--), when present, links the Stretcher unit to the Spacer unit if the Spacer unit is present, links the Stretcher unit to the antimitotic agent unit if the Spacer unit is absent, and links the binding molecule (T) unit to the antimitotic agent unit if the Stretcher unit and Spacer unit are absent. --(Aa)r-- is a natural or unnatural amino acid, dipeptide, tripeptide, tetrapeptide, pentapeptide, hexapeptide, heptapeptide, octapeptide, nonapeptide, decapeptide, undecapeptide or dodecapeptide unit, and r is an integer ranging from 0 to 12. The term amino acid as used herein refers generally to aminoalkylcarboxylate, where the alkyl radical is optionally substituted, such as with alkyl, acyl, hydroxy alkyl, sulfhydrylalkyl, aminoalkyl, carboxyalkyl, and the like, The structures of the natural and unnatural amino acids and peptides are described in the book: G. C. Barrett and D. T. Elmore, “Amino Acid and Peptide”, Cambridge University Press, 2004. In addition, amino acid refers to beta, gamma, and longer amino acids with intra chain containing methyl, benzyl, hydroxymethyl, thiomethyl, carboxyl, carboxylmethyl, guanidinopropyl, and the like. More preferably the amino acid is selected from asparagine, aspartic acid, cysteine, glycine, glutamic acid, lysine, glutamine, arginine, serine, ornithine, threonine, and the like.
The Amino Acid unit of the Invention can be enzymatically cleaved by one or more enzymes, including a tumor-associated protease, to liberate the antimitotic agent, which in one embodiment is protonated in vivo upon release to provide an antimitotic agent.
The Spacer unit (--V--), when present, links an Amino Acid unit to the antimitotic agent when an Amino Acid unit is present. Alternately, the Spacer unit links the Stretcher unit to antimitotic agent when the Amino Acid unit is absent. The Spacer unit also links antimitotic agent to the binding molecule (T) when both the Amino Acid unit and Stretcher unit are absent. The spacer linkers may contain function groups that substantially increase the water solubility, biological transport, preferential renal clearance, uptake, absorption, biodistribution, and/or bioavailability of the conjugate are described herein. Spacer units are of two general types: self- immolative and non self-immolative. A non self-immolative Spacer unit is one in which part or all of the Spacer unit remains bound to antimitotic agent after cleavage, particularly enzymatic, of an Amino Acid unit from the antimitotic agent-Linker- binding molecule conjugate or the antimitotic agent-Linker Compound.
Examples of the self-immolative spacer linkers: Z * Z * Z * v Y Z * Y Z * Y Z * ; ;.
; X Y*; wherein the (*) atom is the point of attachment of additional spacer or releasable linker units, the antimitotic agent, and/or the binding molecule (T); X, Y and Z are independently NH, O, or S; Z is H, NH, O or S independently. v is 0 or 1; Q is 17 17 17 18 17 independently H, OH, C ~C alkyl, (OCH CH ) F, Cl, Br, I, OR , or SR , NR R , N=NR , 1 6 2 2 n 17 17 18 17 18 17 17 17 17 18 17 18 17 18 N=R NR R NO SOR R , SO R , SO R OSO R , PR R , POR R , PO R R , , , 2, 2 3 , 3 2 17 18) 17 18 17 18 OPO(OR )(OR , or OCH PO(OR (OR ) wherein R R are independently H, C ~C of alkyl; 2 , 1 8 C ~C of alkenyl, alkynyl, heteroalkyl; C ~C of aryl, heterocyclic, carbocyclic, cycloalkyl, 2 8 3 8 heterocycloalkyl, heteroaralkyl, alkylcarbonyl; or pharmaceutical cation salts Examples of the non-self-immolative spacer linkers: (CH ) CO(OCH CH ) OCH (CH ) CON(CH CH O) COCH 2 m 2 2 n 3 2 m 2 2 n 3 *(CH CH O) * 2 2 n *CH* *CH* ; ; ; (CH ) (OCH CH ) OCOCH (CH ) CO(OCH CH ) OCOCH 2 m 2 2 n 3 2 m 2 2 n 3 *CH* H O ; *CH* ; ; ; H N HS HO HO H N HS 2 m m m P * * * *N *N * * * * * * OH O O O O O ; ; ; ; ; ; ; 17 17 COOH COOH COOH COOH * *N m m m O O ; ; ; ; ; ; * N* * *X Y* * * * N* N* O m m m m m *N ; ; ; ; ; ; ; COOH O Ar COOH *X Y * *X Y S* m O m H ; ; ; ; ; R' H Q Q OH R' R'' N *X Y* *X Y S* ; ; ; ; ; H O O O HOOC R' R'' COOH COOH ; ; ; ; COOH COOH O COOH O N N OH N OH COOH COOH COOH *N * *N O O O ; ; ; ; ; COOH (OCH CH ) OCH N (CH CH O) CH 2 2 n 3 2 2 n 3 COOH *N * * ; O ; ; H OH N(CH CH O) CH 2 2 n 3 N H N * *N * *N * *N * HO * O O HO O ; ; ; ; HN HN HN O O S HO m O OH O *N * *N * *N O O O ; ; ; ; SO H HO OH HO OH O HN N COOH O NHAc m m m *N * *N * *N * O O O ; ; ; SO H HN HN HN m m P O OH *N * * *N * *N H OH O O O ; ; ; Where the (*) atom is the point of attachment of additional spacer or releaseable linkers, the antimitotic agents, and/or the binding molecules; m is 1~10; n is 1~20.
The binding molecule (T) may be of any kind presently known, or that become known, molecule that binds to, complexes with or reacts with a moiety of a cell population sought to be therapeutically or otherwise biologically modified. The binding molecule unit acts to deliver the antimitotic agents to the particular target cell population with which the binding molecule (T) reacts.
The cell binding agents include, but are not limited to, large molecular weight proteins such as, for example, full-length antibodies (polyconal and monoclonal antibodies); single chain antibodies; fragments of antibodies such as Fab, Fab', F(ab') , F [Parham, J. Immunol. 131, 2 v, 2895-2902 (1983)], fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, CDR's, and epitope-binding fragments of any of the above which immuno- specifically bind to cancer cell antigens, viral antigens or microbial antigens; interferons (such as type I, II, III); peptides; lymphokines such as IL-2, IL-3, IL-4, IL-6, GM-CSF, interferon- gamma (IFN-γ); hormones such as insulin, TRH (thyrotropin releasing hormones), MSH (melanocyte-stimulating hormone), steroid hormones, such as androgens and estrogens, melanocyte-stimulating hormone (MSH); growth factors and colony-stimulating factors such as epidermal growth factors (EGF), granulocyte-macrophage colony-stimulating factor (GM-CSF), transforming growth factors (TGF), such as TGFα, TGFβ, insulin and insulin like growth factors (IGF-I, IGF-II) G-CSF, M-CSF and GM-CSF [Burgess, Immunology Today, 5, 155-158 (1984)]; vaccinia growth factors (VGF); fibroblast growth factors (FGFs); smaller molecular weight proteins, poly-peptide, peptides and peptide hormones, such as bombesin, gastrin, gastrin-releasing peptide; platelet-derived growth factors; interleukin and cytokines, such as interleukin-2 (IL-2), interleukin-6 (IL-6), leukemia inhibitory factors, granulocyte-macrophage colony-stimulating factor (GM-CSF); vitamins, such as folate; apoproteins and glycoproteins, such as transferrin {O'Keefe et al, 260 J. Biol. Chem. 932-937 (1985)}; sugar-binding proteins or lipoproteins, such as lectins; cell nutrient-transport molecules; and small molecular inhibitors, such as prostate-specific membrane antigen (PSMA) inhibitors and small molecular tyrosine kinase inhibitors (TKI), non-peptides or any other cell binding molecule or substance, such as bioactive polymers (Dhar, et al, Proc. Natl. Acad. Sci. 2008, 105, 17356-61); dendrimers (Lee, et al, Nat. Biotechnol. 2005, 23, 1517-26; Almutairi, et al; Proc. Natl. Acad. Sci. 2009, 106, 685-90); nanoparticles (Liong, et al, ACS Nano, 2008, 19, 1309-12; Medarova, et al, Nat. Med. 2007, 13, 372-7; Javier, et al, Bioconjugate Chem. 2008, 19, 1309-12); liposomes (Medinai, et al, Curr. Phar. Des. 2004, 10, 2981-9); viral capsides (Flenniken, et al, Viruses Nanotechnol. 2009, 327, 71-93). In general monoclonal antibodies are preferred as a cell-surface binding agent if an appropriate one is available.
Prior to conjugating with the antimitotic agents of this invention, the binding molecules can be modified through attachment of a more specific peptide, a protein, or a drug, or the other functional molecules with a heterobifunctional cross linker such as with linkers of Amine-to- Nonselective (succinimidyl (NHS)-diazirine (SDA), NHS ester /Azide), Amine-to-Sulfhydryl (NHS ester/maleimide, NHS ester/ pyridyldithiol, NHS esters/ haloacetyl), Sulfhydryl-to- Carbohydrate (Maleimide/Hydrazide, Pyridyldithiol /Hydrazide), Hydroxyl-to-Sulfhydryl (Isocyanate / Maleimide), Amine-to-DNA (NHS ester/ Psoralen), Amine-to-Carboxyl (Carbodiimide).
In the SDA linkage modification, the NHS ester of a SDA linker reacts with primary an amine group of a binding molecule backbone in pH 6~9 buffer to form a stable amide bond upon release of NHS. Then photoactivation of the diarzirine with long-wave UV light (330- 370nm) creates a reactive carbene intermediate that can react with an amine group of a more specific peptide or a protein or the other functional molecule. The order of these two steps can be different as this: an amine group of a functional molecule reacts with a SDA linker first following by photoactive reaction of a binding molecule with long-wave UV light (330-370nm).
The SDA crosslinkers can be cleavable (with a disulfide bond inside such as SDAD linker).
In the NHS ester /Azide linkage modification, the NHS ester of the linker reacts with primary an amine group of a binding molecule backbone in pH 6~9 buffer to form a stable amide. Then an alkynyl group on a more specific peptide or a protein or the other functional molecule reacts to the azide on the other side of the linker via Azide-Alkyne Huisgen Cycloaddition to form a 1,2,3-triazole linkage (click chemistry). Also, the NHS ester of the linker reacts with primary an amine group of a functional molecule in pH 6~9 buffer to form a stable amide. Then an alkynyl group being linked on a binding molecule reacts to the azide on the other side of the linker via 5 Azide-Alkyne Huisgen Cycloaddition to form a 1,2,3-triazole linkage.
In the Amine-to-Sulfhydryl linkage modification, the NHS ester of the linker reacts with a primary amine group of a binding molecule backbone in pH 6~9 buffer to form a stable amide bond. Then a sulfhydryl on a more specific peptide or a protein or the other functional molecule reacts to the maleimide, or pyridyldithiol, or haloacetyl on the other side of the Amine- tosulfhydryl linker at pH 4.5 ~ 8.5 to form a thioether or a disulfide bond. The conjugation with the Amine-to-Sulfhydryl linker can be in different orders. For instance, an amine group of a functional molecule can be reacted with the linker to form an amide bond first, following by reaction with a sulfhydryl on a binding molecule. Also a sulfhydryl group of a functional molecule can be reacted with the linker to form a thioether or a disulfide bond at pH 4.5 ~ 7 first, following by reaction with an amine group on a binding molecule at pH 6 ~ 9 to form an amide bond.
In the Sulfhydryl-to-Carbohydrate linkage modification, the sulfhydryl group of a binding molecule can be reacted with the maleimide or the pyridyldithiol on the linker to form a thioether or a disulfide bond at pH 4.5 ~ 8 first, Then a carbonyl (aldehyde/ketone) group on a functional molecule reacts with the hydrazide to form an hydrazone bond. Also the sulfhydryl group on a functional molecule can react with the linker to form a thioether or a disulfide bond at pH 4.5 ~ 8 first, following by reaction with a carbohydrate, or an oxidized carbohydrate, or an carbonyl (aldehyde/ketone) group on a binding molecule form an hydrazone bond.
In the Hydroxyl-to-Sulfhydryl linkage modification, the sulfhydryl group of a binding molecule can be reacted with the maleimide or the pyridyldithiol on the linker to form a thioether or a disulfide bond at pH 6 ~ 8 first, Then a hydroxy group on a functional molecule reacts with the isocyanate on the linker to form a carbamate bond at pH 8 ~9. Also the sulfhydryl group on a functional molecule can react with the linker to form a thioether or a disulfide bond at pH 6~ 8 first, following by reaction with a hydroxy on a binding molecule form a carbamate bond at pH 8~9.
In yet another aspect of the invention, the production of antibodies used in the present invention involves in vivo or in vitro procedures or combinations thereof. Methods for producing polyclonal anti-receptor peptide antibodies are well-known in the art, such as in U.S.
Pat. No. 4,493,795 (to Nestor et al). A monoclonal antibody is typically made by fusing myeloma cells with the spleen cells from a mouse that has been immunized with the desired antigen (Köhler, G.; Milstein, C. (1975). Nature 256: 495-497). The detailed procedures are described in “Antibodies--A Laboratory Manual”, Harlow and Lane, eds., Cold Spring Harbor Laboratory Press, New York (1988), which is incorporated herein by reference. Particularly monoclonal antibodies are produced by immunizing mice, rats, hamsters or any other mammal with the antigen of interest such as the intact target cell, antigens isolated from the target cell, whole virus, attenuated whole virus, and viral proteins. Splenocytes are typically fused with myeloma cells using polyethylene glycol (PEG) 6000. Fused hybrids are selected by their sensitivity to HAT (hypoxanthine-aminopterin-thymine). Hybridomas producing a monoclonal antibody useful in practicing this invention are identified by their ability to immunoreact specified receptors or inhibit receptor activity on target cells.
A monoclonal antibody used in the present invention can be produced by initiating a monoclonal hybridoma culture comprising a nutrient medium containing a hybridoma that secretes antibody molecules of the appropriate antigen specificity. The culture is maintained under conditions and for a time period sufficient for the hybridoma to secrete the antibody molecules into the medium. The antibody-containing medium is then collected. The antibody molecules can then be further isolated by well-known techniques, such as using protein-A affinity chromatography; anion, cation, hydrophobic, or size exclusive chromatographies (particularly by affinity for the specific antigen after Protein A, and sizing column chromatography); centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
Media useful for the preparation of these compositions are both well-known in the art and commercially available and include synthetic culture media. An exemplary synthetic medium is Dulbecco's minimal essential medium (DMEM; Dulbecco et al., Virol. 8:396 (1959)) supplemented with 4.5 gm/l glucose, 20 mm glutamine, 20% fetal calf serum and with an anti- foaming agent, such as polyoxyethylene-polyoxypropylene block copolymer.
In addition, antibody-producing cell lines can also be created by techniques other than fusion, such as direct transformation of B lymphocytes with oncogenic DNA, or transfection with an oncovirus, such as Epstein-Barr virus (EBV, also called human herpesvirus 4 (HHV-4)) or Kaposi's sarcoma-associated herpesvirus (KSHV). See, U.S. Pat. Nos. 4,341,761; 4,399,121; 4,427,783; 4,444,887; 4,451,570; 4,466,917; 4,472,500; 4,491,632; 4,493,890. A monoclonal antibody may also be produced via an anti-receptor peptide or peptides containing the carboxyl terminal as described well-known in the art. See Niman et al., Proc. Natl. Acad. Sci. USA, 80: 4949-4953 (1983); Geysen et al., Proc. Natl. Acad. Sci. USA, 82: 178-182 (1985); Lei et al.
Biochemistry 34(20): 6675-6688, (1995). Typically, the anti-receptor peptide or a peptide analog is used either alone or conjugated to an immunogenic carrier, as the immunogen for producing anti-receptor peptide monoclonal antibodies.
There are also a number of other well-known techniques for making monoclonal antibodies as binding molecules in this invention. Particularly useful are methods of making fully human antibodies. One method is phage display technology which can be used to select a range of human antibodies binding specifically to the antigen using methods of affinity enrichment. Phage display has been thoroughly described in the literature and the construction and screening of phage display libraries are well known in the art, see, e.g., Dente et al, Gene. 148(1):7-13 (1994); Little et al, Biotechnol Adv. 12(3):539-55 (1994); Clackson et al., Nature 352:264-628 (1991); Huse et al., Science 246:1275-1281 (1989).
Moncolonal antibodies derived by hybridoma technique from another species than human, such as mouse, can be humanized to avoid human anti-mouse antibodies when infused into humans. Among the more common methods of humanization of antibodies are complementarity- determining region grafting and resurfacing. These methods have been extensively described, see e.g. U.S. Pat. Nos. 5,859,205 and 6,797,492; Liu et al, Immunol Rev. 222:9-27 (2008); Almagro et al, Front Biosci. 1;13:1619-33 (2008); Lazar et al, Mol Immunol. 44(8):1986-98 (2007); Li et al, Proc. Natl. Acad. Sci. U S A. 103(10):3557-62 (2006) each incorporated herein by reference.
Fully human antibodies can also be prepared by immunizing transgenic mice, rabbits, monkeys, or other mammals, carrying large portions of the human immunoglobulin heavy and light chains, with an immunogen. Examples of such mice are: the Xenomouse. (Abgenix, Inc.), the HuMAb- Mouse (Medarex/BMS), the VelociMouse (Regeneron), see also U.S. Pat. No. 6,596,541 , 6,207,418, No. 6,150,584, No. 6,111,166, No. 6,075,181, No. 5,922,545, Nos. 5,661,016, ,545,806, 5,436,149 and 5,569,825. In human therapy, murine variable regions and human constant regions can also be fused to construct called “chimeric antibodies” that are considerably less immunogenic in man than murine mAbs (Kipriyanov et al, Mol Biotechnol. 26:39-60 (2004); Houdebine, Curr Opin Biotechnol. 13:625-9 (2002) each incorporated herein by reference). In addition, site-directed mutagenesis in the variable region of an antibody can result in an antibody with higher affinity and specificity for its antigen (Brannigan et al, Nat Rev Mol Cell Biol. 3:964- 70, (2002)); Adams et al, J Immunol Methods. 231:249-60 (1999)) and exchanging constant regions of a mAb can improve its ability to mediate effector functions of binding and cytotoxicity.
Antibodies immunospecific for a malignant cell antigen can also be obtained commercially or produced by any method known to one of skill in the art such as, e.g., chemical synthesis or recombinant expression techniques. The nucleotide sequence encoding antibodies immunospecific for a malignant cell antigen can be obtained commercially, e.g., from the GenBank database or a database like it, the literature publications, or by routine cloning and sequencing.
Apart from an antibody, a peptide or protein that bind/block/target or in some other way interact with the epitopes or corresponding receptors on a targeted cell can be used as a binding molecule. These peptides or proteins could be any random peptide or proteins that have an affinity for the epitopes or corresponding receptors and they don't necessarily have to be of the immunoglobulin family. These peptides can be isolated by similar techniques as for phage display antibodies (Szardenings, J Recept Signal Transduct Res. 2003; 23(4):307-49). The use of peptides from such random peptide libraries can be similar to antibodies and antibody fragments. The binding molecules of peptides or proteins may be conjugated on or linked to a large molecules or materials, such as, but is not limited, an albumin, a polymer, a liposome, a nano particle, as long as such attachment permits the peptide or protein to retain its antigen binding specificity.
Examples of antibodies used for conjugation of antimitotic agents in this prevention for treating cancer, autoimmune disease, and infectious disease include, but are not limited to, 3F8 (anti-GD2), Abagovomab (anti CA-125), Abciximab (anti CD41 (integrin alpha-IIb), Adalimumab (anti-TNF-α), Adecatumumab (anti-EpCAM, CD326), Afelimomab (anti-TNF-α); Afutuzumab (anti-CD20), Alacizumab pegol (anti-VEGFR2), ALD518 (anti-IL-6), Alemtuzumab (Campath, MabCampath, anti- CD52), Altumomab (anti-CEA), Anatumomab ( anti-TAG-72), Anrukinzumab (IMA-638, anti-IL-13), Apolizumab (anti-HLA-DR), Arcitumomab (anti-CEA), Aselizumab (anti-L-selectin (CD62L), Atlizumab (tocilizumab, Actemra, RoActemra, anti-IL-6 receptor), Atorolimumab (anti-Rhesus factor), Bapineuzumab (anti-beta amyloid), Basiliximab (Simulect, antiCD25 (α chain of IL-2 receptor), Bavituximab (anti-phosphatidylserine), Bectumomab (LymphoScan, anti-CD22), Belimumab (Benlysta, LymphoStat-B, anti-BAFF), Benralizumab (anti-CD125), Bertilimumab (anti-CCL11 (eotaxin-1)), Besilesomab (Scintimun, anti-CEA-related antigen), Bevacizumab (Avastin, anti-VEGF-A), Biciromab (FibriScint, anti- fibrin II beta chain), Bivatuzumab (anti-CD44 v6), Blinatumomab (BiTE, anti-CD19), Brentuximab (cAC10, anti-CD30 TNFRSF8), Briakinumab (anti-IL-12, IL-23) Canakinumab (Ilaris, anti-IL-1), Cantuzumab (C242, anti-CanAg), Capromab, Catumaxomab (Removab, anti- EpCAM, anti-CD3), CC49 (anti-TAG-72), Cedelizumab (anti-CD4), Certolizumab pegol (Cimzia anti-TNF-α), Cetuximab (Erbitux, IMC-C225, anti-EGFR), Citatuzumab bogatox (anti-EpCAM), Cixutumumab (anti-IGF-1), Clenoliximab (anti-CD4), Clivatuzumab (anti-MUC1), Conatumumab (anti-TRAIL-R2), CR6261 (anti-Influenza A hemagglutinin), Dacetuzumab (anti- CD40), Daclizumab (Zenapax, anti-CD25 (α chain of IL-2 receptor)), Daratumumab (anti-CD38 (cyclic ADP ribose hydrolase), Denosumab (Prolia, anti-RANKL), Detumomab (anti-B- lymphoma cell), Dorlimomab, Dorlixizumab, Ecromeximab (anti-GD3 ganglioside), Eculizumab (Soliris, anti-C5), Edobacomab (anti-endotoxin), Edrecolomab (Panorex, MAb17-1A, anti- EpCAM), Efalizumab (Raptiva, anti-LFA-1 (CD11a), Efungumab (Mycograb, anti-Hsp90), Elotuzumab (anti-SLAMF7), Elsilimomab (anti-IL-6), Enlimomab pegol (anti-ICAM-1 (CD54)), Epitumomab (anti-episialin), Epratuzumab (anti-CD22), Erlizumab (anti-ITGB2 (CD18)), Ertumaxomab (Rexomun, anti-HER2/neu, CD3), Etaracizumab (Abegrin, anti-integrin α β ), Exbivirumab ( anti-hepatitis B surface antigen), Fanolesomab (NeutroSpec, anti-CD15), Faralimomab (anti-interferon receptor), Farletuzumab (anti-folate receptor 1), Felvizumab (anti- respiratory syncytial virus), Fezakinumab (anti-IL-22), Figitumumab (anti-IGF-1 receptor), Fontolizumab (anti-IFN-γ), Foravirumab (anti-rabies virus glycoprotein), Fresolimumab (anti- TGF-β), Galiximab (anti-CD80), Gantenerumab (anti- beta amyloid), Gavilimomab (anti-CD147 (basigin)), Gemtuzumab (anti-CD33), Girentuximab (anti-carbonic anhydrase 9), Glembatumumab (CR011, anti-GPNMB), Golimumab (Simponi, anti-TNF-α), Gomiliximab (anti-CD23 (IgE receptor)), Ibalizumab (anti-CD4), Ibritumomab (anti-CD20), Igovomab (Indimacis-125, anti-CA-125), Imciromab (Myoscint, anti-cardiac myosin), Infliximab (Remicade, anti-TNF-α), Intetumumab (anti-CD51), Inolimomab (anti-CD25 (α chain of IL-2 receptor)), Inotuzumab (anti-CD22), Ipilimumab (anti-CD152), Iratumumab (anti- CD30 (TNFRSF8)), Keliximab (anti-CD4), Labetuzumab (CEA-Cide, anti-CEA), Lebrikizumab (anti- IL-13), Lemalesomab (anti-NCA-90 (granulocyte antigen)), Lerdelimumab (anti-TGF beta 2), Lexatumumab (anti-TRAIL-R2), Libivirumab (anti-hepatitis B surface antigen), Lintuzumab (anti-CD33), Lucatumumab (anti-CD40), Lumiliximab (anti- CD23 (IgE receptor), Mapatumumab (anti-TRAIL-R1), Maslimomab (anti- T-cell receptor), Matuzumab (anti-EGFR), Mepolizumab (Bosatria, anti-IL-5), Metelimumab (anti-TGF beta 1), Milatuzumab (anti-CD74), Minretumomab (anti-TAG-72), Mitumomab (BEC-2, anti-GD3 ganglioside), Morolimumab (anti-Rhesus factor), Motavizumab (Numax, anti-respiratory syncytial virus), Muromonab-CD3 (Orthoclone OKT3, anti-CD3), Nacolomab (anti-C242), Naptumomab (anti-5T4), Natalizumab (Tysabri, anti-integrin α ) Nebacumab (anti-endotoxin), Necitumumab (anti-EGFR), Nerelimomab (anti-TNF-α), Nimotuzumab (Theracim, Theraloc, anti-EGFR), Nofetumomab, Ocrelizumab (anti-CD20), Odulimomab (Afolimomab, anti-LFA-1 (CD11a)), Ofatumumab (Arzerra, anti-CD20), Olaratumab (anti-PDGF-R α), Omalizumab (Xolair, anti-IgE Fc region), Oportuzumab (anti-EpCAM), Oregovomab (OvaRex, anti-CA-125), Otelixizumab (anti-CD3), Pagibaximab (anti-lipoteichoic acid), Palivizumab (Synagis, Abbosynagis, anti-respiratory syncytial virus), Panitumumab (Vectibix, ABX-EGF, anti-EGFR), Panobacumab (anti- Pseudomonas aeruginosa), Pascolizumab (anti-IL-4), Pemtumomab (Theragyn, anti-MUC1), Pertuzumab (Omnitarg, 2C4, anti-HER2/neu), Pexelizumab (anti-C5), Pintumomab (anti- adenocarcinoma antigen), Priliximab (anti-CD4), Pritumumab (anti-vimentin), PRO 140 (anti- CCR5), Racotumomab (1E10, anti-(N-glycolylneuraminic acid (NeuGc, NGNA)-gangliosides GM3)), Rafivirumab (anti-rabies virus glycoprotein), Ramucirumab (anti-VEGFR2), Ranibizumab (Lucentis, anti-VEGF-A), Raxibacumab (anti-anthrax toxin, protective antigen), Regavirumab (anti-cytomegalovirus glycoprotein B), Reslizumab (anti-IL-5), Rilotumumab (anti-HGF), Rituximab (MabThera, Rituxanmab, anti-CD20), Robatumumab (anti-IGF-1 receptor), Rontalizumab (anti-IFN-α), Rovelizumab (LeukArrest, anti-CD11, CD18), Ruplizumab (Antova, anti-CD154 (CD40L)), Satumomab (anti-TAG-72), Sevirumab (anti-cytomegalovirus), Sibrotuzumab (anti-FAP), Sifalimumab (anti-IFN-α), Siltuximab (anti-IL-6), Siplizumab (anti- CD2), (Smart) MI95 (anti-CD33), Solanezumab (anti-beta amyloid), Sonepcizumab (anti- sphingosinephosphate), Sontuzumab (anti-episialin), Stamulumab (anti-myostatin), Sulesomab (LeukoScan, (anti-NCA-90 (granulocyte antigen), Tacatuzumab (anti-alpha-fetoprotein), Tadocizumab (anti-integrin α β ), Talizumab (anti-IgE), Tanezumab (anti-NGF), Taplitumomab IIb 3 (anti-CD19), Tefibazumab (Aurexis, (anti-clumping factor A), Telimomab, Tenatumomab (anti- tenascin C), Teneliximab (anti-CD40), Teplizumab (anti-CD3), TGN1412 (anti-CD28), Ticilimumab (Tremelimumab, (anti-CTLA-4), Tigatuzumab (anti-TRAIL-R2), TNX-650 (anti- IL-13), Tocilizumab (Atlizumab, Actemra, RoActemra, (anti-IL-6 receptor), Toralizumab (anti- CD154 (CD40L)), Tositumomab (anti-CD20), Trastuzumab (Herceptin, (anti-HER2/neu), Tremelimumab (anti-CTLA-4), Tucotuzumab celmoleukin (anti-EpCAM), Tuvirumab (anti- hepatitis B virus), Urtoxazumab (anti- Escherichia coli), Ustekinumab (Stelara, anti-IL-12, IL-23), Vapaliximab (anti-AOC3 (VAP-1)), Vedolizumab, (anti-integrin α β ), Veltuzumab (anti-CD20), Vepalimomab (anti-AOC3 (VAP-1), Visilizumab (Nuvion, anti-CD3), Vitaxin (anti-vascular integrin avb3), Volociximab (anti-integrin α β ), Votumumab (HumaSPECT, anti-tumor antigen CTAA16.88), Zalutumumab (HuMax-EGFr, (anti-EGFR), Zanolimumab (HuMax-CD4, anti- CD4), Ziralimumab (anti-CD147 (basigin)), Zolimomab (anti-CD5), Etanercept (Enbrel®), Alefacept (Amevive®), Abatacept (Orencia®), Rilonacept (Arcalyst), 14F7 [anti-IRP-2 (Iron Regulatory Protein 2)], 14G2a (anti-GD2 ganglioside, from Nat. Cancer Inst. for melanoma and solid tumors), J591 (anti-PSMA, Weill Cornell Medical School for prostate cancers), 225.28S [anti-HMW-MAA (High molecular weight-melanoma-associated antigen), Sorin Radiofarmaci S.R.L. (Milan, Italy) for melanoma], COL-1 (anti-CEACAM3, CGM1, from Nat. Cancer Inst.
USA for colorectal and gastric cancers), CYT-356 (Oncoltad®, for prostate cancers), HNK20 (OraVax Inc. for respiratory syncytial virus), ImmuRAIT (from Immunomedics for NHL), Lym-1 (anti-HLA-DR10, Peregrine Pharm. for Cancers), MAK-195F [anti-TNF (tumor necrosis factor; TNFA, TNF-alpha; TNFSF2), from Abbott / Knoll for Sepsis toxic shock], MEDI-500 [T10B9, anti-CD3, TRαβ (T cell receptor alpha/beta), complex, from MedImmune Inc for Graft-versus- host disease], RING SCAN [ anti-TAG 72 (tumour associated glycoprotein 72), from Neoprobe Corp. for Breast, Colon and Rectal cancers], Avicidin (anti-EPCAM (epithelial cell adhesion molecule), anti-TACSTD1 (Tumor-associated calcium signal transducer 1), anti-GA733-2 (gastrointestinal tumor-associated protein 2), anti-EGP-2 (epithelial glycoprotein 2); anti-KSA; KS1/4 antigen; M4S; tumor antigen 17-1A; CD326, from NeoRx Corp. for Colon, Ovarian, Prostate cancers and NHL]; LymphoCide (Immunomedics, NJ), Smart ID10 (Protein Design Labs), Oncolym (Techniclone Inc, CA), Allomune (BioTransplant, CA), anti-VEGF (Genentech, CA); CEAcide (Immunomedics, NJ), IMC-1C11 (ImClone Systems, NJ) and Cetuximab (ImClone, NJ) .
Other antibodies as binding ligands include, but are not limited to, are antibodies against the following antigens: Aminopeptidase N (CD13), Annexin A1, B7-H3 (CD276, various cancers), CA125 (ovarian), CA15-3 (carcinomas), CA19-9 (carcinomas), L6 (carcinomas), Lewis Y (carcinomas), Lewis X (carcinomas), alpha fetoprotein (carcinomas), CA242 (colorectal), placental alkaline phosphatase (carcinomas), prostate specific antigen (prostate), prostatic acid phosphatase (prostate), epidermal growth factor (carcinomas), CD2 (Hodgkin’s disease, NHL lymphoma, multiple myeloma), CD3 epsilon (T cell lymphoma, lung, breast, gastric, ovarian cancers, autoimmune diseases, malignant ascites), CD19 (B cell malignancies), CD20 (non- Hodgkin's lymphoma), CD22 (leukemia, lymphoma, multiple myeloma, SLE), CD30 (Hodgkin’s lymphoma), CD33 (leukemia, autoimmune diseases), CD38 (multiple myeloma), CD40 (lymphoma, multiple myeloma, leukemia (CLL)), CD51 (Metastatic melanoma, sarcoma), CD52 (leukemia), CD56 (small cell lung cancers, ovarian cancer, Merkel cell carcinoma, and the liquid tumor, multiple myeloma), CD66e (cancers), CD70 (metastatic renal cell carcinoma and non- Hodgkin lymphoma), CD74 (multiple myeloma), CD80 (lymphoma), CD98 (cancers), mucin (carcinomas), CD221 (solid tumors), CD227 (breast, ovarian cancers), CD262 (NSCLC and other cancers), CD309 (ovarian cancers), CD326 (solid tumors), CEACAM3 (colorectal, gastric cancers), CEACAM5 (carcinoembryonic antigen; CEA, CD66e) (breast, colorectal and lung cancers), DLL4 (Δ-like-4), EGFR (Epidermal Growth Factor Receptor, various cancers), CTLA4 (melanoma), CXCR4 (CD184, Heme-oncology, solid tumors), Endoglin (CD105, solid tumors), EPCAM (epithelial cell adhesion molecule, bladder, head, neck, colon, NHL prostate, and ovarian cancers), ERBB2 (Epidermal Growth Factor Receptor 2; lung, breast, prostate cancers), FCGR1 (autoimmune diseases), FOLR (folate receptor, ovarian cancers), GD2 ganglioside (cancers), G- 28 (a cell surface antigen glyvolipid, melanoma), GD3 idiotype (cancers), Heat shock proteins (cancers), HER1 (lung, stomach cancers), HER2 (breast, lung and ovarian cancers), HLA-DR10 (NHL), HLA-DRB (NHL, B cell leukemia), human chorionic gonadotropin (carcinoma), IGF1R (insulin-like growth factor 1 receptor, solid tumors, blood cancers), IL-2 receptor (interleukin 2 receptor,T-cell leukemia and lymphomas), IL-6R (interleukin 6 receptor, multiple myeloma, RA, Castleman’s disease, IL6 dependent tumors), Integrins (αvβ3, α5β1, α6β4, αllβ3, α5β5, αvβ5, for various cancers), MAGE-1 (carcinomas), MAGE-2 (carcinomas), MAGE-3 (carcinomas), MAGE 4 (carcinomas), anti-transferrin receptor (carcinomas), p97 (melanoma), MS4A1 (membrane- spanning 4-domains subfamily A member 1, Non-Hodgkin's B cell lymphoma, leukemia), MUC1 or MUC1-KLH (breast, ovarian, cervix, bronchus and gastrointestinal cancer), MUC16 (CA125) (Ovarian cancers), CEA (colorectal), gp100 (melanoma), MART1 (melanoma), MPG (melanoma), MS4A1 (membrane-spanning 4-domains subfamily A, small cell lung cancers, NHL), Nucleolin, Neu oncogene product (carcinomas), P21 (carcinomas), Paratope of anti-(N-glycolylneuraminic acid, Breast, Melanoma cancers), PLAP-like testicular alkaline phosphatase (ovarian, testicular cancers), PSMA (prostate tumors), PSA (prostate), ROBO4, TAG 72 (tumour associated glycoprotein 72, AML, gastric, colorectal, ovarian cancers), T cell transmembrane protein (cancers), Tie (CD202b), TNFRSF10B (tumor necrosis factor receptor superfamily member 10B, cancers), TNFRSF13B (tumor necrosis factor receptor superfamily member 13B, multiple myeloma, NHL, other cancers, RA and SLE), TPBG (trophoblast glycoprotein, Renal cell carcinoma), TRAIL-R1 (Tumor necrosis apoprosis Inducing ligand Receptor 1,lymphoma, NHL, colorectal, lung cancers), VCAM-1 (CD106, Melanoma), VEGF, VEGF-A, VEGF-2 (CD309) (various cancers). Some other tumor associated antigens recognized by antibodies have been reviewed (Gerber, et al, mAbs 1:3, 247-253 (2009); Novellino et al, Cancer Immunol Immunother. 54(3),187-207 (2005). Franke, et al, Cancer Biother Radiopharm. 2000, 15, 459-76). Examples of these antigens that antibodies against are: Many other Cluster of Differentiations (CD4, CD5, CD6, CD7, CD8, CD9, CD10, CD11a, CD11b, CD11c, CD12w, CD14, CD15, CD16, CDw17, CD18, CD21, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD31, CD32, CD34, CD35, CD36, CD37, CD41, CD42, CD43, CD44, CD45, CD46, CD47, CD48, CD49b, CD49c, CD53, CD54, CD55, CD58, CD59, CD61, CD62E, CD62L, CD62P, CD63, CD68, CD69, CD71, CD72, CD79, CD81, CD82, CD83, CD86, CD87, CD88, CD89, CD90, CD91, CD95, CD96, CD100, CD103, CD105, CD106, CD109, CD117, CD120, CD127, CD133, CD134, CD135, CD138, CD141, CD142, CD143, CD144, CD147, CD151, CD152, CD154, CD156, CD158, CD163, CD166, .CD168, CD184, CDw186, CD195, CD202 (a, b), CD209, CD235a, CD271, CD303, CD304), Annexin A1, Nucleolin, Endoglin (CD105), ROBO4, Amino-peptidase N, Δ-like-4 (DLL4), VEGFR-2 (CD309), CXCR4 9CD184), Tie2, B7-H3, WT1, MUC1, LMP2, HPV E6 E7, EGFRvIII, HER-2/neu, Idiotype, MAGE A3, p53 nonmutant, NY-ESO-1, GD2, CEA, MelanA/MART1, Ras mutant, gp100, p53 mutant, Proteinase3 (PR1), bcr-abl, Tyrosinase, Survivin, hTERT, Sarcoma translocation breakpoints, EphA2, PAP, ML-IAP, AFP, EpCAM, ERG (TMPRSS2 ETS fusion gene), NA17, PAX3, ALK, Androgen receptor, Cyclin B1, Polysialic acid, MYCN, RhoC, TRP-2, GD3, Fucosyl GM1, Mesothelin, PSCA, MAGE A1, sLe(a), CYP1B1, PLAC1, GM3, BORIS, Tn, GloboH, ETV6-AML, NY-BR-1, RGS5, SART3, STn, Carbonic anhydrase IX, PAX5, OY-TES1, Sperm protein 17, LCK, HMWMAA, AKAP-4, SSX2, XAGE 1, B7H3, Legumain, Tie 2, Page4, VEGFR2, MAD-CT-1, FAP, PDGFR-β, MAD- CT-2, Fos-related antigen 1.
In another specific embodiment, the antimitotic agent- binding molecule conjugates of the invention are used in accordance with the compositions and methods of the invention for the treatment of cancers. The cancers include, but are not limited, Adrenocortical Carcinoma, Anal Cancer, Bladder Cancer, Brain Tumor (Adult, Brain Stem Glioma, Childhood, Cerebellar Astrocytoma, Cerebral Astrocytoma, Ependymoma, Medulloblastoma, Supratentorial Primitive Neuroectodermal and Pineal Tumors, Visual Pathway and Hypothalamic Glioma), Breast Cancer, Carcinoid Tumor, Gastrointestinal, Carcinoma of Unknown Primary, Cervical Cancer, Colon Cancer, Endometrial Cancer, Esophageal Cancer, Extrahepatic Bile Duct Cancer, Ewings Family of Tumors (PNET), Extracranial Germ Cell Tumor, Eye Cancer, Intraocular Melanoma, Gallbladder Cancer, Gastric Cancer (Stomach), Germ Cell Tumor, Extragonadal, Gestational Trophoblastic Tumor, Head and Neck Cancer, Hypopharyngeal Cancer, Islet Cell Carcinoma, Kidney Cancer (renal cell cancer), Laryngeal Cancer, Leukemia (Acute Lymphoblastic, Acute Myeloid, Chronic Lymphocytic, Chronic Myelogenous, Hairy Cell), Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer (Non-Small Cell, Small Cell, Lymphoma (AIDS-Related, Central Nervous System, Cutaneous T-Cell, Hodgkin's Disease, Non-Hodgkin's Disease, Malignant Mesothelioma, Melanoma, Merkel Cell Carcinoma, Metasatic Squamous Neck Cancer with Occult Primary, Multiple Myeloma, and Other Plasma Cell Neoplasms, Mycosis Fungoides, Myelodysplastic Syndrome, Myeloproliferative Disorders, Nasopharyngeal Cancer, Neuroblastoma, Oral Cancer, Oropharyngeal Cancer, Osteosarcoma, Ovarian Cancer (Epithelial, Germ Cell Tumor, Low Malignant Potential Tumor), Pancreatic Cancer (Exocrine, Islet Cell Carcinoma), Paranasal Sinus and Nasal Cavity Cancer, Parathyroid Cancer, Penile Cancer, Pheochromocytoma Cancer, Pituitary Cancer, Plasma Cell Neoplasm, Prostate Cancer Rhabdomyosarcoma, Rectal Cancer, Renal Cell Cancer (kidney cancer), Renal Pelvis and Ureter (Transitional Cell), Salivary Gland Cancer, Sezary Syndrome, Skin Cancer, Skin Cancer (Cutaneous T-Cell Lymphoma, Kaposi's Sarcoma, Melanoma), Small Intestine Cancer, Soft Tissue Sarcoma, Stomach Cancer, Testicular Cancer, Thymoma (Malignant), Thyroid Cancer, Urethral Cancer, Uterine Cancer (Sarcoma), Unusual Cancer of Childhood, Vaginal Cancer, Vulvar Cancer, Wilms' Tumor In another specific embodiment, the antimitotic agent- binding molecule conjugates of the invention are used in accordance with the compositions and methods of the invention for the treatment or prevention of an autoimmune disease. The autoimmune diseases include, but are not limited, Achlorhydra Autoimmune Active Chronic Hepatitis, Acute Disseminated Encephalomyelitis, Acute hemorrhagic leukoencephalitis, Addison's Disease, Agammaglobulinemia, Alopecia areata, Amyotrophic Lateral Sclerosis, Ankylosing Spondylitis, Anti-GBM/TBM Nephritis, Antiphospholipid syndrome, Antisynthetase syndrome, Arthritis, Atopic allergy, Atopic Dermatitis, Autoimmune Aplastic Anemia, Autoimmune cardiomyopathy, Autoimmune hemolytic anemia, Autoimmune hepatitis, Autoimmune inner ear disease, Autoimmune lymphoproliferative syndrome, Autoimmune peripheral neuropathy, Autoimmune pancreatitis, Autoimmune polyendocrine syndrome Types I, II, & III, Autoimmune progesterone dermatitis, Autoimmune thrombocytopenic purpura, Autoimmune uveitis, Balo disease/Balo concentric sclerosis, Bechets Syndrome, Berger's disease, Bickerstaff's encephalitis, Blau syndrome, Bullous Pemphigoid, Castleman's disease, Chagas disease, Chronic Fatigue Immune Dysfunction Syndrome, Chronic inflammatory demyelinating polyneuropathy, Chronic recurrent multifocal ostomyelitis, Chronic lyme disease, Chronic obstructive pulmonary disease, Churg- Strauss syndrome, Cicatricial Pemphigoid, Coeliac Disease, Cogan syndrome, Cold agglutinin disease, Complement component 2 deficiency, Cranial arteritis, CREST syndrome, Crohns Disease (a type of idiopathic inflammatory bowel diseases), Cushing's Syndrome, Cutaneous leukocytoclastic angiitis, Dego's disease, Dercum's disease, Dermatitis herpetiformis, Dermatomyositis, Diabetes mellitus type 1, Diffuse cutaneous systemic sclerosis, Dressler's syndrome, Discoid lupus erythematosus, Eczema, Endometriosis, Enthesitis-related arthritis, Eosinophilic fasciitis, Epidermolysis bullosa acquisita, Erythema nodosum, Essential mixed cryoglobulinemia, Evan's syndrome, Fibrodysplasia ossificans progressiva, Fibromyalgia, Fibromyositis, Fibrosing aveolitis, Gastritis, Gastrointestinal pemphigoid, Giant cell arteritis, Glomerulonephritis, Goodpasture's syndrome, Graves' disease, Guillain-Barré syndrome, Hashimoto's encephalitis, Hashimoto's thyroiditis, Haemolytic anaemia, Henoch-Schonlein purpura, Herpes gestationis, Hidradenitis suppurativa, Hughes syndrome (See Antiphospholipid syndrome), Hypogammaglobulinemia, Idiopathic Inflammatory Demyelinating Diseases, Idiopathic pulmonary fibrosis, Idiopathic thrombocytopenic purpura (See Autoimmune thrombocytopenic purpura), IgA nephropathy (Also Berger's disease), Inclusion body myositis, Inflammatory demyelinating polyneuopathy, Interstitial cystitis, Irritable Bowel Syndrome , Juvenile idiopathic arthritis, Juvenile rheumatoid arthritis, Kawasaki's Disease, Lambert-Eaton myasthenic syndrome, Leukocytoclastic vasculitis, Lichen planus, Lichen sclerosus, Linear IgA disease (LAD), Lou Gehrig's Disease (Also Amyotrophic lateral sclerosis), Lupoid hepatitis, Lupus erythematosus, Majeed syndrome, Ménière's disease, Microscopic polyangiitis, Miller- Fisher syndrome, Mixed Connective Tissue Disease, Morphea, Mucha-Habermann disease, Muckle–Wells syndrome, Multiple Myeloma, Multiple Sclerosis, Myasthenia gravis, Myositis, Narcolepsy, Neuromyelitis optica (Devic's Disease), Neuromyotonia, Occular cicatricial pemphigoid, Opsoclonus myoclonus syndrome, Ord thyroiditis, Palindromic rheumatism, PANDAS (Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcus), Paraneoplastic cerebellar degeneration, Paroxysmal nocturnal hemoglobinuria, Parry Romberg syndrome, Parsonnage-Turner syndrome, Pars planitis, Pemphigus, Pemphigus vulgaris, Pernicious anaemia, Perivenous encephalomyelitis, POEMS syndrome, Polyarteritis nodosa, Polymyalgia rheumatica, Polymyositis, Primary biliary cirrhosis, Primary sclerosing cholangitis, Progressive inflammatory neuropathy, Psoriasis, Psoriatic Arthritis, Pyoderma gangrenosum, Pure red cell aplasia, Rasmussen's encephalitis, Raynaud phenomenon, Relapsing polychondritis, Reiter's syndrome, Restless leg syndrome, Retroperitoneal fibrosis, Rheumatoid arthritis, Rheumatoid fever, Sarcoidosis, Schizophrenia, Schmidt syndrome, Schnitzler syndrome, Scleritis, Scleroderma, Sjögren's syndrome, Spondyloarthropathy, Sticky blood syndrome, Still's Disease, Stiff person syndrome, Subacute bacterial endocarditis, Susac's syndrome, Sweet syndrome, Sydenham Chorea, Sympathetic ophthalmia, Takayasu's arteritis, Temporal arteritis (giant cell arteritis), Tolosa-Hunt syndrome, Transverse Myelitis, Ulcerative Colitis (a type of idiopathic inflammatory bowel diseases), Undifferentiated connective tissue disease, Undifferentiated spondyloarthropathy, Vasculitis, Vitiligo, Wegener's granulomatosis, Wilson's syndrome, Wiskott-Aldrich syndrome In another specific embodiment, a binding molecule used for the conjugate for the treatment or prevention of an autoimmune disease includes, but are not limited to, anti-elastin antibody; Abys against epithelial cells antibody; Anti-Basement Membrane Collagen Type IV Protein antibody; Anti-Nuclear Antibody; Anti ds DNA; Anti ss DNA, Anti Cardiolipin Antibody IgM, IgG; anti-celiac antibody; Anti Phospholipid Antibody IgK, IgG; Anti SM Antibody; Anti Mitochondrial Antibody; Thyroid Antibody; Microsomal Antibody, T-cells antibody; Thyroglobulin Antibody, Anti SCL-70; Anti-Jo; Anti-U.sub.1RNP; Anti-La/SSB; Anti SSA; Anti SSB; Anti Perital Cells Antibody; Anti Histones; Anti RNP; C-ANCA; P-ANCA; Anti centromere; Anti-Fibrillarin, and Anti GBM Antibody, Anti-ganglioside antibody; Anti- Desmogein 3 antibody; Anti-p62 antibody; Anti-sp100 antibody; Anti-Mitochondrial(M2) antibody; Rheumatoid factor antibody; Anti-MCV antibody; Anti-topoisomerase antibody; Anti- neutrophil cytoplasmic(cANCA) antibody In certain preferred embodiments, the binding molecule for the conjugate in the present invention, can bind to both a receptor or a receptor complex expressed on an activated lymphocyte which is associated with an autoimmune disease. The receptor or receptor complex can comprise an immunoglobulin gene superfamily member (e.g. CD2, CD3, CD4, CD8, CD19, CD22, CD28, CD79, CD90, CD152/CTLA-4, PD-1, or ICOS), a TNF receptor superfamily member (e.g. CD27, CD40, CD95/Fas, CD134/OX40, CD137/4-1BB, INF-R1, TNFR-2, RANK, TACI, BCMA, osteoprotegerin, Apo2/TRAIL-R1, TRAIL-R2, TRAIL-R3, TRAIL-R4, and APO- 3), an integrin, a cytokine receptor, a chemokine receptor, a major histocompatibility protein, a lectin (C-type, S-type, or I-type), or a complement control protein.
In another specific embodiment, useful binding ligands that are immunospecific for a viral or a microbial antigen are humanized or human monoclonal antibodies. As used herein, the term "viral antigen" includes, but is not limited to, any viral peptide, polypeptide protein (e.g. HIV gp120, HIV nef, RSV F glycoprotein, influenza virus neuramimidase, influenza virus hemagglutinin, HTLV tax, herpes simplex virus glycoprotein (e.g. gB, gC, gD, and gE) and hepatitis B surface antigen) that is capable of eliciting an immune response. As used herein, the term "microbial antigen" includes, but is not limited to, any microbial peptide, polypeptide, protein, saccharide, polysaccharide, or lipid molecule (e.g., a bacterial, fungi, pathogenic protozoa, or yeast polypeptide including, e.g., LPS and capsular polysaccharide 5/8) that is capable of eliciting an immune response. Examples of antibodies available l for the viral or microbial infection include, but are not limited to, Palivizumab which is a humanized anti- respiratory syncytial virus monoclonal antibody for the treatment of RSV infection; PRO542 which is a CD4 fusion antibody for the treatment of HIV infection; Ostavir which is a human antibody for the treatment of hepatitis B virus; PROTVIR which is a humanized IgG.sub.1 antibody for the treatment of cytomegalovirus; and anti-LPS antibodies.
The binding molecules –antimitotic agent conjugates of this invention can be used in the treatment of infectious diseases. These infectious diseases include, but are not limited to, Acinetobacter infections, Actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (Acquired immune deficiency syndrome), Amebiasis, Anaplasmosis, Anthrax, Arcanobacterium haemolyticum infection, Argentine hemorrhagic fever, Ascariasis, Aspergillosis, Astrovirus infection, Babesiosis, Bacillus cereus infection, Bacterial pneumonia, Bacterial vaginosis, Bacteroides infection, Balantidiasis, Baylisascaris infection, BK virus infection, Black piedra, Blastocystis hominis infection, Blastomycosis, Bolivian hemorrhagic fever, Borrelia infection, Botulism (and Infant botulism), Brazilian hemorrhagic fever, Brucellosis, Burkholderia infection, Buruli ulcer, Calicivirus infection (Norovirus and Sapovirus), Campylobacteriosis, Candidiasis (Moniliasis; Thrush), Cat-scratch disease, Cellulitis, Chagas Disease (American trypanosomiasis), Chancroid, Chickenpox, Chlamydia, Chlamydophila pneumoniae infection, Cholera, Chromoblastomycosis, Clonorchiasis, Clostridium difficile infection, Coccidioidomycosis, Colorado tick fever, Common cold (Acute viral rhinopharyngitis; Acute coryza), Creutzfeldt-Jakob disease, Crimean-Congo hemorrhagic fever, Cryptococcosis, Cryptosporidiosis, Cutaneous larva migrans, Cyclosporiasis, Cysticercosis, Cytomegalovirus infection, Dengue fever, Dientamoebiasis, Diphtheria, Diphyllobothriasis, Dracunculiasis, Ebola hemorrhagic fever, Echinococcosis, Ehrlichiosis, Enterobiasis (Pinworm infection), Enterococcus infection, Enterovirus infection, Epidemic typhus, Erythema infectiosum (Fifth disease), Exanthem subitum, Fasciolopsiasis, Fasciolosis, Fatal familial insomnia, Filariasis, Food poisoning by Clostridium perfringens, Free-living amebic infection, Fusobacterium infection, Gas gangrene (Clostridial myonecrosis), Geotrichosis, Gerstmann-Sträussler-Scheinker syndrome, Giardiasis, Glanders, Gnathostomiasis, Gonorrhea, Granuloma inguinale (Donovanosis), Group A streptococcal infection, Group B streptococcal infection, Haemophilus influenzae infection, Hand, foot and mouth disease (HFMD), Hantavirus Pulmonary Syndrome, Helicobacter pylori infection, Hemolytic-uremic syndrome, Hemorrhagic fever with renal syndrome, Hepatitis A, Hepatitis B, Hepatitis C, Hepatitis D, Hepatitis E, Herpes simplex, Histoplasmosis, Hookworm infection, Human bocavirus infection, Human ewingii ehrlichiosis, Human granulocytic anaplasmosis, Human metapneumovirus infection, Human monocytic ehrlichiosis, Human papillomavirus infection, Human parainfluenza virus infection, Hymenolepiasis, Epstein-Barr Virus Infectious Mononucleosis (Mono), Influenza, Isosporiasis, Kawasaki disease, Keratitis, Kingella kingae infection, Kuru, Lassa fever, Legionellosis (Legionnaires’ disease), Legionellosis (Pontiac fever), Leishmaniasis, Leprosy, Leptospirosis, Listeriosis, Lyme disease (Lyme borreliosis), Lymphatic filariasis (Elephantiasis), Lymphocytic choriomeningitis, Malaria, Marburg hemorrhagic fever, Measles, Melioidosis (Whitmore’s disease), Meningitis, Meningococcal disease, Metagonimiasis, Microsporidiosis, Molluscum contagiosum, Mumps, Murine typhus (Endemic typhus), Mycoplasma pneumonia, Mycetoma, Myiasis, Neonatal conjunctivitis (Ophthalmia neonatorum), (New) Variant Creutzfeldt-Jakob disease (vCJD, nvCJD), Nocardiosis, Onchocerciasis (River blindness), Paracoccidioidomycosis (South American blastomycosis), Paragonimiasis, Pasteurellosis, Pediculosis capitis (Head lice), Pediculosis corporis (Body lice), Pediculosis pubis (Pubic lice, Crab lice), Pelvic inflammatory disease, Pertussis (Whooping cough), Plague, Pneumococcal infection, Pneumocystis pneumonia, Pneumonia, Poliomyelitis, Prevotella infection, Primary amoebic meningoencephalitis, Progressive multifocal leukoencephalopathy, Psittacosis, Q fever, Rabies, Rat-bite fever, Respiratory syncytial virus infection, Rhinosporidiosis, Rhinovirus infection, Rickettsial infection, Rickettsialpox, Rift Valley fever, Rocky mountain spotted fever, Rotavirus infection, Rubella, Salmonellosis, SARS (Severe Acute Respiratory Syndrome), Scabies, Schistosomiasis, Sepsis, Shigellosis (Bacillary dysentery), Shingles (Herpes zoster), Smallpox (Variola), Sporotrichosis, Staphylococcal food poisoning, Staphylococcal infection, Strongyloidiasis, Syphilis, Taeniasis, Tetanus (Lockjaw), Tinea barbae (Barber’s itch), Tinea capitis (Ringworm of the Scalp), Tinea corporis (Ringworm of the Body), Tinea cruris (Jock itch), Tinea manuum (Ringworm of the Hand), Tinea nigra, Tinea pedis (Athlete’s foot), Tinea unguium (Onychomycosis), Tinea versicolor (Pityriasis versicolor), Toxocariasis (Ocular Larva Migrans), Toxocariasis (Visceral Larva Migrans), Toxoplasmosis, Trichinellosis, Trichomoniasis, Trichuriasis (Whipworm infection), Tuberculosis, Tularemia, Ureaplasma urealyticum infection, Venezuelan equine encephalitis, Venezuelan hemorrhagic fever, Viral pneumonia, West Nile Fever, White piedra (Tinea blanca), Yersinia pseudotuberculosis infection, Yersiniosis, Yellow fever, Zygomycosis.
The binding molecules, proffered antibodies described in this patent that are against pathogenic strains include, but are not limit, Acinetobacter baumannii, Actinomyces israelii, Actinomyces gerencseriae and Propionibacterium propionicus, Trypanosoma brucei, HIV (Human immunodeficiency virus), Entamoeba histolytica, Anaplasma genus, Bacillus anthracis, Arcanobacterium haemolyticum, Junin virus, Ascaris lumbricoides, Aspergillus genus, Astroviridae family, Babesia genus, Bacillus cereus, multiple bacteria, Bacteroides genus, Balantidium coli, Baylisascaris genus, BK virus, Piedraia hortae, Blastocystis hominis, Blastomyces dermatitides, Machupo virus, Borrelia genus, Clostridium botulinum, Sabia, Brucella genus, usually Burkholderia cepacia and other Burkholderia species, Mycobacterium ulcerans, Caliciviridae family, Campylobacter genus, usually Candida albicans and other Candida species, Bartonella henselae, Group A Streptococcus and Staphylococcus, Trypanosoma cruzi, Haemophilus ducreyi, Varicella zoster virus (VZV), Chlamydia trachomatis, Chlamydophila pneumoniae, Vibrio cholerae, Fonsecaea pedrosoi, Clonorchis sinensis, Clostridium difficile, Coccidioides immitis and Coccidioides posadasii, Colorado tick fever virus, rhinoviruses, coronaviruses, CJD prion, Crimean-Congo hemorrhagic fever virus, Cryptococcus neoformans, Cryptosporidium genus, Ancylostoma braziliense; multiple parasites, Cyclospora cayetanensis, Taenia solium, Cytomegalovirus, Dengue viruses (DEN-1, DEN-2, DEN-3 and DEN-4) – Flaviviruses, Dientamoeba fragilis, Corynebacterium diphtheriae, Diphyllobothrium, Dracunculus medinensis, Ebolavirus, Echinococcus genus, Ehrlichia genus, Enterobius vermicularis, Enterococcus genus, Enterovirus genus, Rickettsia prowazekii, Parvovirus B19, Human herpesvirus 6 and Human herpesvirus 7, Fasciolopsis buski, Fasciola hepatica and Fasciola gigantica, FFI prion, Filarioidea superfamily, Clostridium perfringens, Fusobacterium genus, Clostridium perfringens; other Clostridium species, Geotrichum candidum, GSS prion, Giardia intestinalis, Burkholderia mallei, Gnathostoma spinigerum and Gnathostoma hispidum, Neisseria gonorrhoeae, Klebsiella granulomatis, Streptococcus pyogenes, Streptococcus agalactiae, Haemophilus influenzae, Enteroviruses, mainly Coxsackie A virus and Enterovirus 71, Sin Nombre virus, Helicobacter pylori, Escherichia coli O157:H7, Bunyaviridae family, Hepatitis A Virus, Hepatitis B Virus, Hepatitis C Virus, Hepatitis D Virus, Hepatitis E Virus, Herpes simplex virus 1, Herpes simplex virus 2, Histoplasma capsulatum, Ancylostoma duodenale and Necator americanus, Hemophilus influenzae, Human bocavirus, Ehrlichia ewingii, Anaplasma phagocytophilum, Human metapneumovirus, Ehrlichia chaffeensis, Human papillomavirus, Human parainfluenza viruses, Hymenolepis nana and Hymenolepis diminuta, Epstein-Barr Virus, Orthomyxoviridae family, Isospora belli, Kingella kingae, Klebsiella pneumoniae, Klebsiella ozaenas, Klebsiella rhinoscleromotis, Kuru prion, Lassa virus, Legionella pneumophila, Legionella pneumophila, Leishmania genus, Mycobacterium leprae and Mycobacterium lepromatosis, Leptospira genus, Listeria monocytogenes, Borrelia burgdorferi and other Borrelia species, Wuchereria bancrofti and Brugia malayi, Lymphocytic choriomeningitis virus (LCMV), Plasmodium genus, Marburg virus, Measles virus, Burkholderia pseudomallei, Neisseria meningitides, Metagonimus yokagawai, Microsporidia phylum, Molluscum contagiosum virus (MCV), Mumps virus, Rickettsia typhi, Mycoplasma pneumoniae, numerous species of bacteria (Actinomycetoma) and fungi (Eumycetoma), parasitic dipterous fly larvae, Chlamydia trachomatis and Neisseria gonorrhoeae, vCJD prion, Nocardia asteroides and other Nocardia species, Onchocerca volvulus, Paracoccidioides brasiliensis, Paragonimus westermani and other Paragonimus species, Pasteurella genus, Pediculus humanus capitis, Pediculus humanus corporis, Phthirus pubis, Bordetella pertussis, Yersinia pestis, Streptococcus pneumoniae, Pneumocystis jirovecii, Poliovirus, Prevotella genus, Naegleria fowleri, JC virus, Chlamydophila psittaci, Coxiella burnetii, Rabies virus, Streptobacillus moniliformis and Spirillum minus, Respiratory syncytial virus, Rhinosporidium seeberi, Rhinovirus, Rickettsia genus, Rickettsia akari, Rift Valley fever virus, Rickettsia rickettsii, Rotavirus, Rubella virus, Salmonella genus, SARS coronavirus, Sarcoptes scabiei, Schistosoma genus, Shigella genus, Varicella zoster virus, Variola major or Variola minor, Sporothrix schenckii, Staphylococcus genus, Staphylococcus genus, Staphylococcus aureus, Streptococcus pyogenes, Strongyloides stercoralis, Treponema pallidum, Taenia genus, Clostridium tetani, Trichophyton genus, Trichophyton tonsurans, Trichophyton genus, Epidermophyton floccosum, Trichophyton rubrum, and Trichophyton mentagrophytes, Trichophyton rubrum, Hortaea werneckii, Trichophyton genus, Malassezia genus, Toxocara canis or Toxocara cati, Toxoplasma gondii, Trichinella spiralis, Trichomonas vaginalis, Trichuris trichiura, Mycobacterium tuberculosis, Francisella tularensis, Ureaplasma urealyticum, Venezuelan equine encephalitis virus, Vibrio colerae, Guanarito virus, West Nile virus, Trichosporon beigelii, Yersinia pseudotuberculosis, Yersinia enterocolitica, Yellow fever virus, Mucorales order (Mucormycosis) and Entomophthorales order (Entomophthoramycosis), Pseudomonas aeruginosa, Campylobacter (Vibrio) fetus, Aeromonas hydrophila, Edwardsiella tarda, Yersinia pestis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Salmonella typhimurium, Treponema pertenue, Treponema carateneum, Borrelia vincentii, Borrelia burgdorferi, Leptospira icterohemorrhagiae, Pneumocystis carinii, Brucella abortus, Brucella suis, Brucella melitensis, Mycoplasma spp., Rickettsia prowazeki, Rickettsia tsutsugumushi, Clamydia spp.; pathogenic fungi (Aspergillus fumigatus, Candida albicans, Histoplasma capsulatum); protozoa (Entomoeba histolytica, Trichomonas tenas, Trichomonas hominis, Tryoanosoma gambiense, Trypanosoma rhodesiense, Leishmania donovani, Leishmania tropica, Leishmania braziliensis, Pneumocystis pneumonia, Plasmodium vivax, Plasmodium falciparum, Plasmodium malaria); or Helminiths (Schistosoma japonicum, Schistosoma mansoni, Schistosoma haematobium, and hookworms).
Other antibodies as a binding ligand in this invention for treatment of viral disease include, but are not limited to, antibodies against antigens of pathogenic viruses, including as examples and not by limitation: Poxyiridae, Herpesviridae, Adenoviridae, Papovaviridae, Enteroviridae, Picornaviridae, Parvoviridae, Reoviridae, Retroviridae, influenza viruses, parainfluenza viruses, mumps, measles, respiratory syncytial virus, rubella, Arboviridae, Rhabdoviridae, Arenaviridae, Non-A/Non-B Hepatitis virus, Rhinoviridae, Coronaviridae, Rotoviridae, Oncovirus [such as, HBV (Hepatocellular carcinoma), HPV (Cervical cancer, Anal cancer), Kaposi's sarcoma- associated herpesvirus (Kaposi's sarcoma), Epstein-Barr virus (Nasopharyngeal carcinoma, Burkitt's lymphoma, Primary central nervous system lymphoma), MCPyV (Merkel cell cancer), SV40 (Simian virus 40), HCV (Hepatocellular carcinoma), HTLV-I (Adult T-cell leukemia/lymphoma)], Immune disorders caused virus: [such as Human Immunodeficiency Virus (AIDS)]; Central nervous system virus: [such as, JCV (Progressive multifocal leukoencephalopathy), MeV (Subacute sclerosing panencephalitis), LCV (Lymphocytic choriomeningitis), Arbovirus encephalitis, Orthomyxoviridae (probable) (Encephalitis lethargica), RV (Rabies), Chandipura virus, Herpesviral meningitis, Ramsay Hunt syndrome type II; Poliovirus (Poliomyelitis, Post-polio syndrome), HTLV-I (Tropical spastic paraparesis)]; Cytomegalovirus (Cytomegalovirus retinitis, HSV (Herpetic keratitis)); Cardiovascular virus [such as CBV (Pericarditis, Myocarditis)]; Respiratory system/acute viral nasopharyngitis/viral pneumonia: [Epstein-Barr virus (EBV infection/Infectious mononucleosis), Cytomegalovirus; SARS coronavirus (Severe acute respiratory syndrome) Orthomyxoviridae: Influenzavirus A/B/C (Influenza/Avian influenza), Paramyxovirus: Human parainfluenza viruses (Parainfluenza), RSV (Human respiratory syncytial virus), hMPV]; Digestive system virus [MuV (Mumps), Cytomegalovirus (Cytomegalovirus esophagitis); Adenovirus (Adenovirus infection); Rotavirus, Norovirus, Astrovirus, Coronavirus; HBV (Hepatitis B virus), CBV, HAV (Hepatitis A virus), HCV (Hepatitis C virus), HDV (Hepatitis D virus), HEV (Hepatitis E virus), HGV (Hepatitis G virus)]; Urogenital virus [such as, BK virus, MuV (Mumps)].
According to a further object, the present invention also concerns pharmaceutical compositions comprising the conjugate of the invention together with a pharmaceutically acceptable carrier for treatment of cancer and autoimmune disorders. The method for treatment of cancer and autoimmune disorders can be practiced in vitro, in vivo, or ex vivo. Examples of in vitro uses include treatments of cell cultures in order to kill all cells except for desired variants that do not express the target antigen; or to kill variants that express undesired antigen. Examples of ex vivo uses include treatments of hematopoietic stem cells (HSC) prior to the performance of the transplantation (HSCT) into the same patient in order to kill diseased or malignant cells. For instance, clinical ex vivo treatment to remove tumor cells or lymphoid cells from bone marrow prior to autologous transplantation in cancer treatment or in treatment of autoimmune disease, or to remove T cells and other lymphoid cells from allogeneic bone marrow or tissue prior to transplant in order to prevent graft-versus-host disease, can be carried out as follows. Bone marrow is harvested from the patient or other individual and then incubated in medium containing serum to which is added the conjugate of the invention, concentrations range from about 1 pM to 0.1 mM, for about 30 minutes to about 48 hours at about 37 ºC. The exact conditions of concentration and time of incubation (=dose) are readily determined by the skilled clinicians.
After incubation the bone marrow cells are washed with medium containing serum and returned to the patient by i.v. infusion according to known methods. In circumstances where the patient receives other treatment such as a course of ablative chemotherapy or total-body irradiation between the time of harvest of the marrow and reinfusion of the treated cells, the treated marrow cells are stored frozen in liquid nitrogen using standard medical equipment.
For clinical in vivo use, the conjugate of the invention will be supplied as solutions or as a lyophilized solid that can be redisolved in sterile water for injection. Examples of suitable protocols of conjugate administration are as follows. Conjugates are given weekly for 8 weeks as an i.v. bolus. Bolus doses are given in 50 to 500 ml of normal saline to which human serum albumin (e.g. 0.5 to 1 mL of a concentrated solution of human serum albumin, 100 mg/mL) can be added. Dosages will be about 50 µg to 20 mg/kg of body weight per week, i.v. (range of 10 µg to 200 mg/kg per injection). 8 weeks after treatment, the patient may receive a second course of treatment. Specific clinical protocols with regard to route of administration, excipients, diluents, dosages, times, etc., can be determined by the skilled clinicians.
Examples of medical conditions that can be treated according to the in vivo or ex vivo methods of killing selected cell populations include malignancy of any types of cancer, autoimmune diseases, graft rejections, and infections (viral, bacterial or parasite).
The amount of a conjugate which is required to achieve the desired biological effect, will vary depending upon a number of factors, including the chemical characteristics, the potency, and the bioavailability of the conjugates, the type of disease, the species to which the patient belongs, the diseased state of the patient, the route of administration, all factors which dictate the required dose amounts, delivery and regimen to be administered.
In general terms, the conjugates of this invention may be provided in an aqueous physiological buffer solution containing 0.1 to 10% w/v conjugates for parenteral administration. Typical dose ranges are from 1 µg/kg to 0.1 g/kg of body weight per day; a preferred dose range is from 0.01 mg/kg to 20 mg/kg of body weight per day or an equivalent dose in a human child. The preferred dosage of drug to be administered is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, the formulation of the compound, the route of administration (intravenous, intramuscular, or other), the pharmacokinetic properties of the compound by the chosen delivery route, and the speed (bolus or continuous infusion) and schedule of administrations (number of repetitions in a given period of time).
The conjugates of the present invention are also capable of being administered in unit dose forms, wherein the term “unit dose” means a single dose which is capable of being administered to a patient, and which can be readily handled and packaged, remaining as a physically and chemically stable unit dose comprising either the active conjugate itself, or as a pharmaceutically acceptable composition, as described hereinafter. As such, typical total daily dose ranges are from 0.01 to 100 mg/kg of body weight. By way of general guidance, unit doses for humans range from 1 mg to 3000 mg per day. Preferably the unit dose range is from 1 to 500 mg administered one to four times a day, and even more preferably from 10 mg to 500 mg, once a day. Conjugatess provided herein can be formulated into pharmaceutical compositions by admixture with one or more pharmaceutically acceptable excipients. Such unit dose compositions may be prepared for use by oral administration, particularly in the form of tablets, simple capsules or soft gel capsules; or intranasally, particularly in the form of powders, nasal drops, or aerosols; or dermally, for example, topically in ointments, creams, lotions, gels or sprays, or via trans-dermal patches. The compositions may conveniently be administered in unit dosage form and may be prepared by any of the methods well known in the pharmaceutical art, for example, as described in Remington: The Science and Practice of Pharmacy, 21 ed.; Lippincott Williams & Wilkins: Philadelphia, PA, 2005.
Preferred formulations include pharmaceutical compositions in which a compound of the present invention is formulated for oral or parenteral administration. For oral administration, tablets, pills, powders, capsules, troches and the like can contain one or more of any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, or gum tragacanth; a diluent such as starch or lactose; a disintegrant such as starch and cellulose derivatives; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, or methyl salicylate. Capsules can be in the form of a hard capsule or soft capsule, which are generally made from gelatin blends optionally blended with plasticizers, as well as a starch capsule. In addition, dosage unit forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents. Other oral dosage forms syrup or elixir may contain sweetening agents, preservatives, dyes, colorings, and flavorings. In addition, the active compounds may be incorporated into fast dissolve, modified-release or sustained-release preparations and formulations, and wherein such sustained-release formulations are preferably bi-modal. Preferred tablets contain lactose, cornstarch, magnesium silicate, croscarmellose sodium, povidone, magnesium stearate, or talc in any combination.
Liquid preparations for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions, and emulsions. The liquid compositions may also include binders, buffers, preservatives, chelating agents, sweetening, flavoring and coloring agents, and the like. Non-aqueous solvents include alcohols, propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and organic esters such as ethyl oleate. Aqueous carriers include mixtures of alcohols and water, buffered media, and saline. In particular, biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be useful excipients to control the release of the active compounds. Intravenous vehicles can include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Other potentially useful parenteral delivery systems for these active compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
Alternative modes of administration include formulations for inhalation, which include such means as dry powder, aerosol, or drops. They may be aqueous solutions containing, for example, polyoxyethylenelauryl ether, glycocholate and deoxycholate, or oily solutions for administration in the form of nasal drops, or as a gel to be applied intranasally. Formulations for buccal administration include, for example, lozenges or pastilles and may also include a flavored base, such as sucrose or acacia, and other excipients such as glycocholate. Formulations suitable for rectal administration are preferably presented as unit-dose suppositories, with a solid based carrier, such as cocoa butter, and may include a salicylate. Formulations for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers which can be used include petroleum jelly, lanolin, polyethylene glycols, alcohols, or their combinations. Formulations suitable for transdermal administration can be presented as discrete patches and can be lipophilic emulsions or buffered, aqueous solutions, dissolved and/or dispersed in a polymer or an adhesive.
In a specific embodiment, a conjugate of the invention is administered concurrently with the other known or will be known therapeutic agents such as the chemotherapeutic agent, the radiation therapy, immunotherapy agents, autoimmune disorder agents, anti-infectious agents or the other antibody-drug conjugates, resulting in a synergistic effect. In another specific embodiment, the synergistic drugs or radiation therapy are administered prior or subsequent to administration of a conjugate, in one aspect at least an hour, 12 hours, a day, a week, a month, in further aspects several months, prior or subsequent to administration of a conjugate of the invention.
In other embodiments, the synergistic drugs include, but not limited to: 1). Chemotherapeutic agents: a). Alkylating agents: such as [Nitrogen mustards: (chlorambucil, cyclophosphamide, ifosfamide, mechlorethamine, melphalan, trofosfamide); Nitrosoureas: (carmustine, lomustine); Alkylsulphonates: (busulfan, treosulfan); Triazenes: (dacarbazine); Platinum containing compounds: (carboplatin, cisplatin, oxaliplatin)]; b). Plant Alkaloids: such as [Vinca alkaloids: (vincristine, vinblastine, vindesine, vinorelbine); Taxoids: (paclitaxel, docetaxol)]; c). DNA Topoisomerase Inhibitors: such as [Epipodophyllins: (9- aminocamptothecin, camptothecin, crisnatol, etoposide, etoposide phosphate, irinotecan, teniposide, topotecan,); Mitomycins: (mitomycin C)]; d). Anti-metabolites: such as {[Anti-folate: DHFR inhibitors: (methotrexate, trimetrexate); IMP dehydrogenase Inhibitors: (mycophenolic acid, tiazofurin, ribavirin, EICAR); Ribonucleotide reductase Inhibitors: (hydroxyurea, deferoxamine)]; [Pyrimidine analogs: Uracil analogs: (5-Fluorouracil, doxifluridine, floxuridine, ratitrexed(Tomudex)); Cytosine analogs: (cytarabine, cytosine arabinoside, fludarabine); Purine analogs: (azathioprine, mercaptopurine, thioguanine)]}; e). Hormonal therapies: such as {Receptor antagonists: [Anti-estrogen: (megestrol, raloxifene, tamoxifen); LHRH agonists: (goscrclin, leuprolide acetate); Anti-androgens: (bicalutamide, flutamide)]; Retinoids/Deltoids: [Vitamin D3 analogs: (CB 1093, EB 1089 KH 1060, cholecalciferol, ergocalciferol); Photodynamic therapies: (verteporfin, phthalocyanine, photosensitizer Pc4, demethoxy- hypocrellin A); Cytokines: (Interferon-alpha, Interferon-gamma, tumor necrosis factor (TNFs), human proteins containing a TNF domain)]}; f). Kinase inhibitors, such as BIBW 2992 (anti- EGFR/Erb2), imatinib, gefitinib, pegaptanib, sorafenib, dasatinib, sunitinib, erlotinib, nilotinib, lapatinib, axitinib, pazopanib. vandetanib, E7080 (anti-VEGFR2), mubritinib, ponatinib (AP24534), bafetinib (INNO-406), bosutinib (SKI-606), cabozantinib, vismodegib, iniparib, ruxolitinib, CYT387, axitinib, tivozanib, sorafenib, bevacizumab, cetuximab, Trastuzumab, Ranibizumab, Panitumumab, ispinesib; g). Others: such as gemcitabine, epoxomicins (e. g. carfilzomib), bortezomib, thalidomide, lenalidomide, pomalidomide, tosedostat, zybrestat, PLX4032, STA-9090, Stimuvax, allovectin-7, Xegeva, Provenge, Yervoy, Isoprenylation inhibitors (such as Lovastatin), Dopaminergic neurotoxins (such as 1-methylphenylpyridinium ion), Cell cycle inhibitors (such as staurosporine), Actinomycins (such as Actinomycin D, dactinomycin), Bleomycins (such as bleomycin A2, bleomycin B2, peplomycin), Anthracyclines (such as daunorubicin, doxorubicin (adriamycin), idarubicin, epirubicin, pirarubicin, zorubicin, mtoxantrone, MDR inhibitors (such as verapamil), Ca ATPase inhibitors (such as thapsigargin), Histone deacetylase inhibitors (Vorinostat, Romidepsin, Panobinostat, Valproic acid, Mocetinostat (MGCD0103), Belinostat, PCI-24781, Entinostat, SB939, Resminostat, Givinostat, AR-42, CUDC-101, sulforaphane, Trichostatin A) ; Thapsigargin, Celecoxib, glitazones, epigallocatechin gallate, Disulfiram, Salinosporamide A. 2). An anti-autoimmune disease agent includes, but is not limited to, cyclosporine, cyclosporine A, aminocaproic acid, azathioprine, bromocriptine, chlorambucil, chloroquine, cyclophosphamide, corticosteroids (e.g. amcinonide, betamethasone, budesonide, hydrocortisone, flunisolide, fluticasone propionate, fluocortolone danazol, dexamethasone, Triamcinolone acetonide, beclometasone dipropionate), DHEA, enanercept, hydroxychloroquine, infliximab, meloxicam, methotrexate, mofetil, mycophenylate, prednisone, sirolimus, tacrolimus. 3). An anti-infectious disease agent includes, but is not limited to, a). Aminoglycosides: amikacin, astromicin, gentamicin (netilmicin, sisomicin, isepamicin), hygromycin B, kanamycin (amikacin, arbekacin, bekanamycin, dibekacin, tobramycin), neomycin (framycetin, paromomycin, ribostamycin), netilmicin, spectinomycin, streptomycin, tobramycin, verdamicin; b). Amphenicols: azidamfenicol, chloramphenicol, florfenicol, thiamphenicol; c). Ansamycins: geldanamycin, herbimycin; d). Carbapenems: biapenem, doripenem, ertapenem, imipenem/cilastatin, meropenem, panipenem; e). Cephems: carbacephem (loracarbef), cefacetrile, cefaclor, cefradine, cefadroxil, cefalonium, cefaloridine, cefalotin or cefalothin, cefalexin, cefaloglycin, cefamandole, cefapirin, cefatrizine, cefazaflur, cefazedone, cefazolin, cefbuperazone, cefcapene, cefdaloxime, cefepime, cefminox, cefoxitin, cefprozil, cefroxadine, ceftezole, cefuroxime, cefixime, cefdinir, cefditoren, cefepime, cefetamet, cefmenoxime, cefodizime, cefonicid, cefoperazone, ceforanide, cefotaxime, cefotiam, cefozopran, cephalexin, cefpimizole, cefpiramide, cefpirome, cefpodoxime, cefprozil, cefquinome, cefsulodin, ceftazidime, cefteram, ceftibuten, ceftiolene, ceftizoxime, ceftobiprole, ceftriaxone, cefuroxime, cefuzonam, cephamycin (cefoxitin, cefotetan, cefmetazole), oxacephem (flomoxef, latamoxef); f). Glycopeptides: bleomycin, vancomycin (oritavancin, telavancin), teicoplanin (dalbavancin), ramoplanin; g).
Glycylcyclines: e. g. tigecycline; g). β-Lactamase inhibitors: penam (sulbactam, tazobactam), clavam (clavulanic acid); i). Lincosamides: clindamycin, lincomycin; j). Lipopeptides: daptomycin, A54145, calcium-dependent antibiotics (CDA); k). Macrolides: azithromycin, cethromycin, clarithromycin, dirithromycin, erythromycin, flurithromycin, josamycin, ketolide (telithromycin, cethromycin), midecamycin, miocamycin, oleandomycin, rifamycins (rifampicin, rifampin, rifabutin, rifapentine), rokitamycin, roxithromycin, spectinomycin, spiramycin, tacrolimus (FK506), troleandomycin, telithromycin; l). Monobactams: aztreonam, tigemonam; m).
Oxazolidinones: linezolid; n). Penicillins: amoxicillin, ampicillin (pivampicillin, hetacillin, bacampicillin, metampicillin, talampicillin), azidocillin, azlocillin, benzylpenicillin, benzathine benzylpenicillin, benzathine phenoxymethylpenicillin, clometocillin, procaine benzylpenicillin, carbenicillin (carindacillin), cloxacillin, dicloxacillin, epicillin, flucloxacillin, mecillinam (pivmecillinam), mezlocillin, meticillin, nafcillin, oxacillin, penamecillin, penicillin, pheneticillin, phenoxymethylpenicillin, piperacillin, propicillin, sulbenicillin, temocillin, ticarcillin; o).
Polypeptides: bacitracin, colistin, polymyxin B; p). Quinolones: alatrofloxacin, balofloxacin, ciprofloxacin, clinafloxacin, danofloxacin, difloxacin, enoxacin, enrofloxacin, floxin, garenoxacin, gatifloxacin, gemifloxacin, grepafloxacin, kano trovafloxacin, levofloxacin, lomefloxacin, marbofloxacin, moxifloxacin, nadifloxacin, norfloxacin, orbifloxacin, ofloxacin, pefloxacin, trovafloxacin, grepafloxacin, sitafloxacin, sparfloxacin, temafloxacin, tosufloxacin, trovafloxacin; q). Streptogramins: pristinamycin, quinupristin/dalfopristin); r). Sulfonamides: mafenide, prontosil, sulfacetamide, sulfamethizole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim, trimethoprim-sulfamethoxazole (co-trimoxazole); s). Steroid antibacterials: e.g. fusidic acid; t).
Tetracyclines: doxycycline, chlortetracycline, clomocycline, demeclocycline, lymecycline, meclocycline, metacycline, minocycline, oxytetracycline, penimepicycline, rolitetracycline, tetracycline, glycylcyclines (e.g. tigecycline); u). Other types of antibiotics: annonacin, arsphenamine, bactoprenol inhibitors (Bacitracin), DADAL/AR inhibitors (cycloserine), dictyostatin, discodermolide, eleutherobin, epothilone, ethambutol, etoposide, faropenem, fusidic acid, furazolidone, isoniazid, laulimalide, metronidazole, mupirocin, mycolactone, NAM synthesis inhibitors (e. g. fosfomycin), nitrofurantoin, paclitaxel, platensimycin, pyrazinamide, quinupristin/dalfopristin, rifampicin (rifampin), tazobactam tinidazole, uvaricin; 4). Anti-viral drugs: a). Entry/fusion inhibitors: aplaviroc, maraviroc, vicriviroc, gp41 (enfuvirtide), PRO 140, CD4 (ibalizumab); b). Integrase inhibitors: raltegravir, elvitegravir, globoidnan A; c). Maturation inhibitors: bevirimat, vivecon; d). Neuraminidase inhibitors: oseltamivir, zanamivir, peramivir; e). Nucleosides & nucleotides: abacavir, aciclovir, adefovir, amdoxovir, apricitabine, brivudine, cidofovir, clevudine, dexelvucitabine, didanosine (ddI), elvucitabine, emtricitabine (FTC), entecavir, famciclovir, fluorouracil (5-FU), 3’-fluoro- substituted 2’, 3’-dideoxynucleoside analogues (e.g. 3’-fluoro-2’,3’-dideoxythymidine (FLT) and 3’-fluoro-2’,3’-dideoxyguanosine (FLG), fomivirsen, ganciclovir, idoxuridine, lamivudine (3TC), l-nucleosides (e.g. β-l-thymidine and β-l-2’-deoxycytidine), penciclovir, racivir, ribavirin, stampidine, stavudine (d4T), taribavirin (viramidine), telbivudine, tenofovir, trifluridine valaciclovir, valganciclovir, zalcitabine (ddC), zidovudine (AZT); f). Non-nucleosides: amantadine, ateviridine, capravirine, diarylpyrimidines (etravirine, rilpivirine), delavirdine, docosanol, emivirine, efavirenz, foscarnet (phosphonoformic acid), imiquimod, interferon alfa, loviride, lodenosine, methisazone, nevirapine, NOV-205, peginterferon alfa, podophyllotoxin, rifampicin, rimantadine, resiquimod (R-848), tromantadine; g). Protease inhibitors: amprenavir, atazanavir, boceprevir, darunavir, fosamprenavir, indinavir, lopinavir, nelfinavir, pleconaril, ritonavir, saquinavir, telaprevir (VX-950), tipranavir; h). Other types of anti-virus drugs: abzyme, arbidol, calanolide a, ceragenin, cyanovirin-n, diarylpyrimidines, epigallocatechin gallate (EGCG), foscarnet, griffithsin, taribavirin (viramidine), hydroxyurea, KP-1461, miltefosine, pleconaril, portmanteau inhibitors, ribavirin, seliciclib.
). Other immunotheraphy drugs: e.g. imiquimod, interferons (e.g. α, β), granulocyte colony-stimulating factors, cytokines, Interleukins (IL-1 ~ IL-35), antibodies (e. g. trastuzumab, pertuzumab, bevacizumab, cetuximab, panitumumab, infliximab, adalimumab, basiliximab, daclizumab, omalizumab), Protein-bound drugs (e.g., Abraxane), an antibody conjugated with drugs selected from calicheamicin derivative, of maytansine derivatives (DM1 and DM4) , CC- 1065 and duocarmycin minor groove binders, potent taxol derivatives, doxorubicin, auristatin antimitotic drugs (e. g. Trastuzumab-DM1, Inotuzumab ozogamicin, Brentuximab vedotin, Glembatumumab vedotin, lorvotuzumab mertansine, AN-152 LMB2, TP-38, VB4-845, Cantuzumab mertansine, AVE9633, SAR3419, CAT-8015 (anti-CD22), IMGN388, milatuzumab-doxorubicin, SGN-75 (anti-CD70), Anti-CD22-MCC-DM1).
According to a still further object, the present invention is also concerned with the process of preparation of the conjugate of the invention. The conjugate and process of the present invention may be prepared in a number of ways well known to those skilled in the art. The antimitotic agents used in the conjugate can be synthesized, for example, by application or adaptation of the methods described below, or variations thereon as appreciated by the skilled artisan. The appropriate modifications and substitutions will be readily apparent and well known or readily obtainable from the scientific literature to those skilled in the art. In particular, such methods can be found in R.C.
Larock, Comprehensive Organic Transformations, 2 Edition, Wiley-VCH Publishers, 1999.
In the reactions described hereinafter, it may be necessary to protect reactive functional groups, for example hydroxy, amino, imino, thio or carboxy groups, where these are desired in the final product, to avoid their unwanted participation in the reactions. Conventional protecting groups may be used in accordance with standard practice, for examples see P. G. Wuts and T.W. Greene, Greene's Protective Groups in Organic Synthesis, Wiley-Interscience; 4th edition (2006). Some reactions may be carried out in the presence of a base, or an acid or in a suitable solvent. There is no particular restriction on the nature of the base, acid and solvent to be used in this reaction, and any base, acid or solvent conventionally used in reactions of this type may equally be used here, provided that it has no adverse effect on other parts of the molecule. The reactions can take place over a wide range of temperatures. In general, we find it convenient to carry out the reaction at a temperature of from - 80°C to 150°C (more preferably from about room temperature to 100°C). The time required for the reaction may also vary widely, depending on many factors, notably the reaction temperature and the nature of the reagents. However, provided that the reaction is effected under the preferred conditions outlined above, a period of from 3 hours to 20 hours will usually suffice.
The work-up of the reaction can be carried out by conventional means. For example, the reaction products may be recovered by distilling off the solvent from the reaction mixture or, if necessary after distilling off the solvent from the reaction mixture, pouring the residue into water followed by extraction with a water-immiscible organic solvent and distilling off the solvent from the extract. Additionally, the product can, if desired, be further purified by various well known techniques, such as recrystallization, reprecipitation or the various chromatography techniques, notably column chromatography or preparative thin layer chromatography. The synthesis of the antimitotic agents and their conjugates of this invention are illustrated in the figures 1 ~28.
The comjugates of binding molecules with potent antimitotic agents are further illustrated but not restricted by the description in the following examples. 6. EXPERIMENTAL MATERIALS: Mass spectra were obtained using a Bruker Esquire 3000 system. NMR spectra were recorded on a Bruker AVANCE300 spectrometer. Chemical shifts are reported in ppm relative to TMS as an internal standard. Ultraviolet spectra were recorded on a Hitachi U1200 spectrophotometer. HPLC was performed using an Agilent 1100 HPLC system equipped with a fraction collector and a variable wavelength detector. Thin layer chromatography was performed on Analtech GF silica gel TLC plates. Aminal acids and their derivatives as well as preloaded resins were either from Merck Chemicals International Co, or Synthetech Co., or Peptides International Inc or Chembridge International Co. or Sigma –Aldrich Co. Some of the linkers, Linkers of NHS ester /Maleimide (AMAS, BMPS, GMBS, MBS, SMCC, EMCS or Sulfo- EMCS, SMPB, SMPH, LC-SMCC, Sulfo-KMUS, SM(PEG)4, SM(PEG)6, SM(PEG)8, SM(PEG)12, SM(PEG)24); NHS ester /Pyridyldithiol (SPDP, LC-SPDP or Sulfo-LC-SPDP, SMPT, Sulfo-LC-SMPT); NHS esters /Haloacetyl (SIA, SBAP, SIAB or Sulfo-SIAB); NHS ester /Diazirine (SDA or Sulfo-SDA, LC-SDA or Sulfo-LC-SDA, SDAD or Sulfo-SDAD); Maleimide /Hydrazide (BMPH, EMCH, MPBH, KMUH); Pyridyldithiol /Hydrazide (PDPH); Isocyanate /Maleimide (PMPI) were purchased from Thermo Fisher Scientific Co. SPDB, SPP linkers were made according to the references (Cumber, A. et al, Bioconjugate Chem., 1992, 3, 397–401). Human anti-CD22 antibody was from Santa Cruz Biotechnology, Inc. and Trastuzumab was from Genentech. All other chemicals or anhydrous solvents were from Sigma- Aldrich International.
Example 1. Methyl 4-(bis(2-hydroxyethyl)amino)oxobutanoate (3) Dimethyl succinate (20.0 g, 136.9 mmol) and dihydroxyethylamine (7.20 g, 68.7 mmol) in the mixture of anhydrous toluene (500 ml) and pyridine (50 ml) were refluxed at 150 °C for 28 h. The mixture was concentrated and purified on SiO column eluted with EtOAc/DCM (5% ~25% EtOAc) to afford the title compound (12.5 g, 83% yield). ESI MS m/z+ for C H NaNO (M + Na) cald 242.2, found 242.4. 9 17 5 Example 2. Methyl 4-(bis(2-((methylsulfonyl)oxy)ethyl)amino)oxobutanoate (4) Methyl 4-(bis(2-hydroxyethyl)amino)oxobutanoate (12.0 g, 49.56 mmol) in anhydrous pyridine (350 ml) was added methanesulfonyl chloride (20.0 g, 175.4 mmol). After stirred overnight the mixture was concentrated, diluted with EtOAc (350 ml), washed with cold 1 M NaH PO (2 × 300ml), dried over MgSO , filtered and evaporated to afford crude product 2 4 4 (~18.8 g, >100% yield). The crude product was used for next step without further purification.
ESI MS m/z+ for C H NaNO S (M + Na) cald 398.2, found 398.4. 11 21 9 2 Example 3. Methyl 4-(bis(2-(acetylthio)ethyl)amino)oxobutanoate (5) Methyl 4-(bis(2-((methylsulfonyl)oxy)ethyl)amino)oxobutanoate (fresh made, 90% pure, 8.5 g, ~20 mmol) in DMA (350 ml) at 0 °C was added thioacetic acid (10 ml, 134 mmol), followed by addtion of Et N (30 ml, 215 mmol). The mixture was then stirred at room temperature overnight, concentrated, diluted with EtOAc (350 ml), washed with NaHCO (sat’ed, 300 ml), NaCl sat’ed solution (300 ml) and 1 M NaH PO (300 ml). The organic layer was dried over Na SO , filtered, evaporated and purified on SiO column eluted with 2 4 2 EtOAc/hexane (10% ~ 25% EtOAc) to afford the title compound (5.1 g, 76% yield). ESI MS m/z+ for 25 C H NaNO S (M + Na) cald 358.1, found 358.2. 13 21 5 2 Example 4. 4-(Bis(2-(pyridinyldisulfanyl)ethyl)amino)oxobutanoic acid (6) Methyl 4-(bis(2-(acetylthio)ethyl)amino)oxobutanoate (5.0 g, 14.9 mmol) in THF (150 ml) was added NaOH (5.0 g, 125 mmol) in water (100 ml). The mixture was stirred at RT for min, neutralized with H PO to pH 7. Then 1,2-di(pyridinyl)disulfane (Aldrithiol-2, 26.0 g, 118 mmol) in THF (100 ml) was added and the mixture was stirred for 4 h, concentrated and purified on SiO column eluted with MeOH/DCM/HOAc (1:20/0.2) to afford the title product (5.8 g, 85.6% yield). ESI MS m/z+ for C H NaN O S (M + Na) cald 478.0, found 478.2. 18 21 3 3 4 Example 5. 2,5-dioxopyrrolidinyl 4-(bis(2-(pyridinyldisulfanyl)ethyl)amino) oxobutanoate (7) 4-(Bis(2-(pyridinyldisulfanyl)ethyl)amino)oxobutanoic acid (5.2 g, 11.5 mmol) in DMA (100 ml) was added NHS (1.6 g, 13.9 mmol) and EDC (5.0 g, 26.1 mmol). The mixture was stirred overnight, evaporated and purified on SiO column eluted with EtOAc/DCM (5% to 15% EtOAc) to afford the title product (5.8 g, 85.6% yield). ESI MS m/z+ for C H NaN O S (M + Na) cald 575.1, found 575.2. 22 24 4 5 4 Example 6. 3,6-endoxo-Δ-tetrahydrophthalimide (12) Maleimide (10.0 g, 103.0 mmol) in toluene (200 ml) was added furan (10.0 ml, 137.4 mmol). The mixture was heated inside a 1 L of autoClave bomb at 100 °C for 8 h. The bomb was cooled to room temperature, and the inside solid was rinsed with methanol, concentrated and crystallized in ethyl acetate/hexane to afford 16.7 g (99%) of the title compound. 1H NMR (CDCl ): 11.12 (s, 1H) (NH), 6.68~6.64 (m, 2H), 5.18~5.13 (m, 2H), 2.97 ~2.92 (m, 2H). MS m/z+ for C H NaNO (M + Na) cald 188.04, found 188.04. 8 7 3 Example 7. Methyl 4-((2-((3aR,4R,7S,7aS)-1,3-dioxo-3a,4,7,7a-tetrahydro-1H-4,7- epoxyisoindol-2(3H)-yl)ethyl)(2-((4R,7S,7aS)-1,3-dioxo-3a,4,7,7a-tetrahydro-1H-4,7- epoxyisoindol-2(3H)-yl)ethyl)amino)oxobutanoate (13) Methyl 4-(bis(2-((methylsulfonyl)oxy)ethyl)amino)oxobutanoate (4, fresh made, 90% pure, 8.5 g, ~20 mmol) in DMA (350 ml) was added 3,6-endoxo-Δ-tetrahydrophthalimide (10.2 g, 61.8 mmol), sodium carbonate (8.0 g, 75.5 mmol) and sodium iodide (0.3 g, 2.0 mmol). The mixture was then stirred at room temperature overnight, concentrated, diluted with EtOAc (350 ml), washed with NaHCO (sat’ed, 300 ml), NaCl sat’ed solution (300 ml) and 1 M NaH PO (300 ml). The organic layer was dried over Na SO , filtered, evaporated, loaded 2 4 2 4 on SiO column and eluted with EtOAc/hexane (10% ~ 30% EtOAc) to afford the title compound (7.9 g, 77% yield). ESI MS m/z+ for C H NaN O (M + Na) cald 536.2, found 27 3 9 536.4.
Example 8. 4-(bis(2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethyl)amino)oxobutanoic acid (14) Compound 13 (3.0 g, 5.8 mmol) and trimethylstannanol (4.8 g, 26.4 mmol) in 1,2- dichloroethane (150 ml) was refluxed at 80 °C for 8 h. It was cooled to room temperature and the residue was passed a short silica gel column and eluted with dichloromethane/methanol to remove the extra trimethyltin hydroxide. Then the pooled fractions were combined, concentrated and diluted with DMA and toluene, refluxed at 120 °C overnight, loaded on SiO column and eluted with MeOH/DCM (5% ~ 10% MeOH) to afford the title compound (1.62 g, 76% yield). ESI MS m/z+ for C H NaN O (M + Na) cald 386.1, found 386.2. 16 17 3 9 Example 9. 2,5-Dioxopyrrolidinyl 4-(bis(2-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)ethyl)amino)oxobutanoate Compound 14 (1.60 g, 4.4 mmol) in DMA (100 ml) was added NHS (0.76 g, 6.61 mmol) and EDC (1.70 g, 8.90 mmol). The mixture was stirred overnight, evaporated and purified on SiO2 column, eluted with EtOAc/DCM (5% to 15% EtOAc) to afford the title product (1.72 g, 85.0% yield). ESI MS m/z+ for C H NaN O (M + Na) cald 483.1, found 483.2. 20 4 9 Example 10. t-Butyl 5-(3’,6’-endoxo-Δ-tetrahydrophthalimido) pentanoate To a solution of t-Butyl -hydroxy pentanote (10.0 g, 57.4 mmol) in pyridine (60 ml) was added mesyl chloride (8.0 ml, 103.3 mmol) and the mixture was stirred for 6 h, evaporated, diluted with EtOAc, washed with cold 1M NaH PO , pH 6, dried over MgSO , filtered and 2 4 4 evaporated to dryness. To a mixture of compound 12 (9.90 g, 60.0 mmol) and Na CO (8.5 g, 80.1 mmol) in DMF (80 ml) was added dry mesylate compound. The mixture was stirred overnight, evaporated, diluted with EtOAc, washed with saline water and 1M NaH PO (pH 6), dried over MgSO , evaporated and purified on SiO column, eluted with EtOAc/dichloromethane (1:12) to afford the title compound (14.01 g, 76%). MS m/z+ for C H NaNO (M + Na) cald 344.16, found 344.16. 17 23 5 Example 11. 5-maleimido-pentanoic acid (21b) Compound 17 (5.0 g, 15.57 mmol) in 1,4-dioxanne (40 ml) was added HCl (10 ml, 36%) at 4 °C and the mixture was stirred for 30 min, evaporated to dryness to form 5-(3’,6’-endoxo- Δ-tetrahydrophthalimido) pentanoic acid (4.08 g, 99%). The dried compound in mixture of DMF/toluene (1:1, 40 ml) was refluxed for 6h, evaporated and crystallized from EtOH/ether/hexane to afford the title compound (2.76 g, 90%). MS m/z+ for C H NO (M + 9 12 4 H) cald 198.07, found 198.07.
Example 12. N-succinimidyl 5-maleimido-pentanoate (23b) (DMPS linker) To a solution of 5-maleimido-pentanoic acid 21b (2.0 g, 10.1 mmol) in dichloromethane (20 ml) was added N-hydroxysuccimide (1.50 g, 13.0 mmol) and EDC (7.0 g, 36.4 mmol) and the mixture was stirred overnight, evaporated and purified on SiO column (EtOAc/dichloromethane, 1:10) to afford the title compound 23b (2.43 g, 82%). MS m/z+ for C H NaN O (M + Na) cald 317.09, found 317.09. 13 14 2 6 Example 13. t-Butyl 5-(3’,6’-endoxo-Δ-tetrahydrophthalimido)pentanoyl hydrazine carboxylate (25a-a) -(3’,6’-endoxo-Δ-tetrahydrophthalimido) pentanoic acid (1.0 g, 3.77 mmol) in DMF (30 ml) was added tert-butyl carbazate (0.60 g, 4.53 mmol) and EDC (2.0 g, 10.4 mmol). The mixture was stirred overnight, evaporated and purified on SiO column (EtOAc/dichloromethane, 1:10) to afford the title compound (1.18 g, 83%). MS m/z+ for C H NaN O (M + Na) cald 402.17, found 402.18. 18 25 3 6 Example 14. 5-maleimido-pentanoic acid hydrazide (25a-b) Compound 25a-a (1.18 g, 3.11 mmol) was dissolved in the mixture of DMF/toluene (1:1, ml), refluxed for 6 h and evaporated. The residue was dissolved in 1,4-dioxanne (20 ml) and HCl (5 ml, 36%) was added at 4 °C and the mixture was stirred for 30 min, evaporated to dryness and crystallized from EtOH/ether/hexane to afford the title compound (577 mg, 88%).
MS m/z+ for C H N O (M + H) cald 212.10, found 212.10. 9 14 3 3 Example 15. General procedure for 3’-bromo-maleimdo compounds 39 and 40, and 3’,4’- dibromo-maleimdo compounds 43 and 44.
To amine 37 or 38 (~ 6 g) in DMF (60 ml) was added bromomaleic anhydride (1 eq) or 2,3-dibromomaleic anhydride (1 eq) and the mixture was stirred overnight, evaporated on an oil pump to dryness to afford the crude enoic acids. To the crude enoic acids were added HOAc (~50 ml) and Ac O (2 ~ 4 g) and the reaction mixture was fluxed at 120 °C for 6 ~ 12 h, concentrated and purified on SiO column, eluted with EtOAc/dichloromethane (1:10 ~ 1:1) to afford (61% ~ 87% yield) of the 3’-bromo-maleimdo compounds 39 and 40, and 3’,4’- dibromomaleimdo compounds 43 and 44 respectively. -(3-bromo-2,5-dioxo-2,5-dihydro-1H-pyrrolyl)pentanoic acid MS m/z+ for C H BrNO (M + H) cald 275.98, found 275.98. 9 11 4 3-(2-(2-(2-(3-bromo-2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy)ethoxy)ethoxy)- propanoic acid MS m/z+ for C H BrNO (M + H) cald 380.03, found 380.03. 13 19 7 -(3,4-dibromo-2,5-dioxo-2,5-dihydro-1H-pyrrolyl)pentanoic acid MS m/z+ for C H Br NO (M + H) cald 353.89, found 353.89. 9 10 2 4 3-(2-(2-(2-(3,4-dibromo-2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy)ethoxy)ethoxy)- propanoic acid MS m/z+ for C H Br NO (M + H) cald 457.94, found 457.94. 13 18 2 7 Example 16. General procedure for NHS ester of 3’-bromo-maleimdo compounds 41 and 42, and NHS ester of 3’,4’-dibromo-maleimdo compounds 45 and 46.
To the solution of 3’-bromo-maleimdo compounds 39 and 40 (1 eq), or 3’,4’- dibromomaleimdo compounds 43 and 44 in DMA (~0.15 M) were added N- hydroxysuccinimide (1.1 eq) and EDC (2~4 eq) and the mixture was stirred overnight, concentrated and purified on SiO column, eluted with EtOAc/dichloromethane (1:20 ~ 1:5) to afford (70% ~ 93% yield) of the 3’-bromomaleimdo linkers 41 and 42, and 3’,4’-dibromo- maleimdo linkers 45 and 46 respectively. 2,5-dioxopyrrolidinyl 5-(3-bromo-2,5-dioxo-2,5-dihydro-1H-pyrrol yl)pentanoate MS m/z+ for C H BrN NaO (M + Na) cald 395.00, found 395.00. 13 13 2 7 2,5-dioxopyrrolidinyl 5-(3,4-dibromo-2,5-dioxo-2,5-dihydro-1H-pyrrol yl)pentanoate MS m/z+ for C H Br N NaO (M + Na) cald 472.91, found 472.91. 13 12 2 2 6 2,5-dioxopyrrolidinyl 3-(2-(2-(2-(3-bromo-2,5-dioxo-2,5-dihydro-1H-pyrrol yl)ethoxy)ethoxy)ethoxy)propanoate MS m/z+ for C H BrN NaO (M + Na) cald 499.04, 17 21 2 9 found 499.04. 2,5-dioxopyrrolidinyl 3-(2-(2-(2-(3,4-dibromo-2,5-dioxo-2,5-dihydro-1H-pyrrol yl)ethoxy)ethoxy)ethoxy)propanoate MS m/z+ for C H Br N NaO (M + Na) cald 576.95, 17 20 2 2 9 found 576.95.
Example 17. 4-(2-Pyridyldithio)methylpentanoic acid 4-Mercaptomethylpentanoic Acid (Goff, D. et al, BioConjugate Chem. 1990, 1, 381- 386) (4.67 g, 31.5 mmol) in methanol (15 ml) was added the solution of 2,2’-Dithiodipyridine (30.0 g, 10 136.2 mmol) in the mixture of methanol (80 ml) and 100 mM sodium phosphate buffer solution (pH 7.5, 70 ml). After stirred for 6 h, the mixture was concentrated, extracted with EtOAc/Hexane (1:1). The aqueous solution was adjusted to pH 3 and extracted with EtOAc (3 × 100 ml). The organic layers were combined, dried over Na SO , filtered, evaporated and purified on SiO column (MeOH/dichloromethane/HOAc, 1:15:0.01) to afford the title compound (7.05 g, 87%). MS m/z+ for C H NO S (M + H) cald 258.05, found 11 16 2 2 258.05.
Example 18. N-Succinimidyl 4-(2-pyridyldithio) methylpentanoate (243) (SMDP linker) 4-(2-pyridyldithio) methylpentanoic acid (2.0 g, 7.78 mmol) in dichloromethane (20 ml) was added N-hydroxysuccimide (1.10 g, 9.56 mmol) and EDC (4.0 g, 20.8 mmol) and the mixture was stirred overnight, evaporated and purified on SiO column (EtOAc/dichloromethane, 1:10) to afford the title compound (2.48 g, 90%). MS m/z+ for C H NaN O S (M + Na) cald 377.07, found 377.08. 18 2 4 2 Example 19. (3aR,4R,6S,6aR)((R)-2,2-dimethyl-1,3-dioxolanyl)-2,2- dimethyltetrahydro-furo[3,4-d][1,3]dioxolol (62) To a stirred slurry of D-gulonic-lactone (20.01 g, 112.37 mmol) and anhydrous CuSO (25.0 g, 157.22 mmol) in dry acetone (450 mL) was added conc. H SO (1.6 mL), the mixture was then stirred at room temperature for 24 h. The pH of the solution was adjusted to 7 with Ca(OH) , and the resulting slurry was filtered and evaporated in vacuo to afford a diacetonide (2,3:5,6-O-diisopropylidene-D-gulono-1,4-lactone) as a light-yellow syrup which was used in the next step without further purification. To a stirred solution of diacetonide in THF (300 mL) at -78 °C was added slowly 1 M solution of DIBAL-H (180 ml, 180 mM) in toluene. After being stirred for 1 h at -78 °C, the reaction mixture was quenched with water (50 mL) and filtered through Celite. The organic layer was separated, dried (Na SO ), and concentrated in vacuo. The residue was purified by silica gel chromatography with hexane/ethyl acetate (5:1) to give the title compound (25.27 g, 83% two steps) as colorless syrup. ESI MS m/z+ for C H NaO (M +Na) cald 283.12, found 283.12. 12 20 6 Example 20. (3aR,4R,6S,6aR,Z)((R)-2,2-dimethyl-1,3-dioxolanyl)-2,2-dimethyl-N-(2- methylpropylidene)tetrahydrofuro[3,4-d][1,3]dioxolamine oxide (64) A mixture of 2,3:5,6-O-diisopropylidene-D-gulofuranose (62) (10.0 g, 38.4 mmol) and hydroxylamine hydrochloride (25.01 g, 360.87 mmol) in pyridine (150 mL) was stirred at room temperature for 1 h. The reaction mixture was concentrated in vacuo, added water (250 mL) and extracted with dichloromethane. The combined organic extracts were washed with brine, dried (MgSO ), concentrated in vacuo and filtered through short silica gel column eluted with ethyl acetate to give 2,3:5,6-O-diisopropylidene-D-gulose oxime (63) (10.34 g, 98%) as a colorless vitreous substance which was used directly without further purification. A mixture of this crude oxime (10.30 g, 37.43 mmol), isobutyraldehyde (3.00 g, 41.66 mmol), and MgSO (3 g, 20 mmol) was stirred at the room temperature overnight. The mixture was filtered through a pad of Celite. The filtered was concentrated in vacuo and the residue was passed through a short SiO column and eluted with ethyl acetate to afford the title compound (11.57 g, 94% yield) as white solid. ESI MS m/z+ for C H NO cald 329.18., found 329.18. 16 27 6 Example 21. (3R,5R)((3aR,4R,6S,6aR)((R)-2,2-dimethyl-1,3-dioxolanyl)-2,2- dimethyltetrahydrofuro[3,4-d][1,3]dioxolyl)[(-)-10',2'-camphorsultam]- Npropylisoxazolidinecarboxamide (65) A mixture of (3aR,4R,6S,6aR,Z)((R)-2,2-dimethyl-1,3-dioxolanyl)-2,2-dimethyl-N- (2-methylpropylidene)tetrahydrofuro[3,4-d][1,3]dioxolamine oxide (6.00 g 18.22 mmol) and (2R)-N-(acryloyl) bornane-10,2-sultams (5.10 g, 18.95 mmol) in dichloromethane (50 mL) was heated under reflux for 37 h. After concentration, the residue was recrystallized from EtOH (30 mL) to give the title compound (8.72 g, 80% yield) as a colorless solid. Flash chromatography (silica gel, hexane/AcOEt 7:3) of the mother liquor gave further (0.47 g, 4%) of the title compound as a colorless solid. MS ESI: m/z: [M+Na] , calcd for C H N NaO S 29 46 2 9 621.28, found 621.28.
Example 22. (3R,5R)(5 tert-butoxycarbonyl)isopropylisoxazolidinecarboxylic acid (67) A solution of LiOH (5.0 g, 208.7 mmol) in water (60 mL) at 45 °C was added to a solution of (3R,5R)((3aR,4R,6S,6aR)((R)-2,2-dimethyl-1,3-dioxolanyl)-2,2- dimethyltetrahydrofuro[3,4-d][1,3]dioxolyl)[(-)-10',2'-camphorsultam]- Npropylisoxazolidinecarboxamide (30.0 g, 48.2 mmol) in THF (100 mL) and MeOH (60 mL). After stirred for 1 h, the mixture was concentrated, poured into H O (150 mL) and the mixture was adjusted to pH 9 with 4 M HCl. The mixture was extracted with EtOAc, then the aqueous layer was adjusted to pH 3 with 4 M HCl, extracted with EtOAc, the organic extract was washed with brine, dried over Na SO , filtered, and concentrated in vacuo. The residue was triturated with hexane to give (3R,5R){(3aR,4R,6S,6aR)[(R)-2,2-dimethyl-1,3- dioxolanyl]-2,2-dimethyltetrahydrofuro[3,4-d,1,3]dioxolyl}isopropylisoxazolidine carboxylic acid (66a, 17.1 g 88%) as a colorless solid. This material was used for the next step without further purification. MS ESI: m/z: [M+Na] , calcd for C H NNaO , 424.19, found 19 31 8 424.19.
To a solution of this material (8.0 g, 19.95 mmol) in MeCN (80 mL) was added 60% HClO aq. (6.0 mL, 35.77 mmol) at 45 °C, and the mixture was stirred at RT for 1 h. After concentration, the residue was dissolved in 1,4-dioxane (40 mL), and then a suspension of NaHCO (25 g, 297 mmol) in H O (32 mL) and Boc O (4.80 g, 22.00 mmol) was added at 3 2 2 4 °C. The mixture was stirred at RT for 4 h, concentrated, diluted with H O and EtOAc/Hexane (1:1) and separated. The aqueous layer was adjusted to pH 3 with 4 M HCl aq.
The mixture was extracted with EtOAc, the organic layers were washed with brine, dried over Na SO , filtered, and evaporated in vacuo. The residue was triturated with hexane to give (3R,5R)(tertbutoxycarbonyl)- 3-isopropylisoxazolidinecarboxylic acid (4.91 g, 95%) as a colorless amorphous solid. MS ESI: m/z: [M+Na] , calcd for C H NNaO , 282.13, found 12 21 5 282.13.
Example 23. Methyl (R)[(3R,5R)[(t-butyl-yl)methoxycarbonyl] isopropylisoxazolidinecarboxamido](triphenylmethylthio)propionate (68) Pr NEt (0.75 mL, 4.31 mmol) and TBTU (2.50 g, 7.78 mmol) were added to a solution of (3R,5R)(tert-butoxycarbonyl)isopropylisoxazolidinecarboxylic acid (1.01 g, 3.89 mmol) and l-(S)-Tr-cysteine methyl ester hydrochloride (1.76 g, 4.27 mmol) in dichloromethane (15 mL) at 4 °C and the mixture was stirred at RT overnight. The mixture was poured into NaHCO (sat.) solution, extracted with dichloromethane, dried over Na SO , 3 2 4 filtered, concentrated and silica gel chromatography (hexane/AcOEt 1:2) to give the title compound (2.10 g, 87%) as colorless amorphous solid. MS ESI: m/z: [M+Na] , calcd for C H N NaO S, 641.27, found 641.26. 42 2 6 Example 24. (3R,5R)-tert-butyl 3-isopropyl(4-(methoxycarbonyl)thiazol yl)isoxazolidinecarboxylate (69) Tf O (2.0 mL, 12.0 mmol) was added to a solution of Ph P=O (4.10 g, 14.74 mmol) in dichloromethane (40.0 mL) and the mixture was stirred at -10 °C for 1 h. A solution of methyl (R)[(3R,5R)[(t-butyl-yl)methoxycarbonyl]isopropylisoxazolidinecarboxamido] (triphenylmethylthio)propionate (68) (4.00 g, 6.47 mmol) in dichloromethane (20 mL) was added to the reaction mixture at -10 °C and the mixture was stirred at RT for 6 h, added to NaHCO saturated solution at 4 °C and extracted with dichloromethane. The organic extracts were washed with brine, dried over Na SO , filtered, concentrated in vacuo and SiO 2 4 2 chromatography (hexane/AcOEt 3:2) to give the corresponding thiazolidine derivative as a yellow amorphous solid. MnO (5.80 g, 66.7 mmol) was added to a solution of this material in dichloromethane (60 mL) and the mixture was stirred at RT for 24 h. The mixture was filtrated via Celite, concentrated in vacuo and SiO chromatography (hexane/AcOEt 3:2) to give the title compound (1.75 g, 75%) as a colorless amorphous solid. ESI: m/z: [M+Na] , calcd for C H N NaO S, 379.13, found 379.14. 16 24 2 5 Example 25. Methyl 2-((1R,3R)(tert-butoxycarbonylamino)hydroxy methylpentyl)-thiazolecarboxylate (70) Mo(CO) (1.10 g, 3.12 mmol) was added to a solution of (3R,5R)-tert-butyl 3- isopropyl(4-(methoxycarbonyl)thiazolyl)isoxazolidinecarboxylate (69) (1.00 g, 2.81 mmol) in CH CN (20 ml) and H O (2 ml) and the mixture was stirred at 70 °C for 16 h. After concentration, the residue was diluted with EtOAc (50 mL) and a 10% aq. solution of citric acid (50 ml). NaIO was added to the mixture until the aqueous layer became clear, and extracted with EtOAc. The organic extracts were washed with a 10% aq. Na S O and brine, dried over Na SO , filtered, concentrated and SiO 2 2 3 2 4 2 chromatography (hexane/EtOAc 3:2) to give the title compound (906 mg, 90%) as a colorless solid. MS ESI: m/z: [M+Na] , calcd for C H N NaO S, 381.14, found 381.14. 16 26 2 5 Example 26. Methyl 2-((1R,3R)acetoxy(tert-butoxycarbonyl-amino) methylpentyl)-5 thiazolecarboxylate (71) Compound 70 (900 mg, 2.51 mmol) in pyridine (15 ml) was added Ac O (0.5 ml, 5.29 mmol) and the mixture was stirred overnight, concentrated, SiO chromatography (hexane/EtOAc 4:1) to give the title compound (950 mg, 95%) as a colorless solid. MS ESI: m/z: [M+Na] , calcd for C H N NaO S, 423.15, found 423.16. 18 28 2 6 Example 27. 2-((1R,3R)acetoxy(tert-butoxycarbonylamino)methylpentyl)thiazole- 4-carboxylic acid (72) Compound 71 (940 mg, 2.35 mmol) in THF (15 ml) was added NaH (120 mg, 3.0 mmol, 60% in oil) at 4 °C and the mixture was stirred for 2 h, then CH I (0.155 ml, 2.49 mmol) was added. After stirred overnight, concentrated, re-dissolved in EtOAc, filtered through short SiO column and evaporated to dryness to give a crude 2-((1R,3R)acetoxy(tert- butoxycarbonyl-(methyl)amino)methylpentyl)thiazolecarboxylic acid (73a). To the dry compound (73a) in 1,2-dichloroethane (20 ml) was added trimethyltin hydroxide (620 mg, 3.43 mmol). The mixture was stirred at 80 °C overnight, concentrated, re-dissolved MeOH/dichloromethane/HOAc (1:5:0.01, 20 ml), filtered through short SiO column, concentrated and co-evaporated with toluene to dryness. To the dry compound in pyridine (15 ml) was added Ac O (0.4 ml, 4.23 mmol). After stirred overnight, the mixture was concentrated, purified by SiO chromatography (MeOH/dichloromethane/HOAc (1:10:0.01) to give the title compound (735 mg, 78%) as a colorless solid. ESI: m/z: [M+Na] , calcd for C H N NaO S 423.15, found 423.16. 18 28 2 6 Example 28. Methyl 2-((1R,3R)(tert-butoxycarbonyl(methyl)amino)(3-(1,3- dioxoisoindolinyl)propoxy)methylpentyl)thiazolecarboxylate (86) Compound 70 (850 mg, 2.37 mmol) in THF (15 ml) at -20 °C was added NaH (100 mg, 2.5 mmol, 60% in oil). After stirred for 30 min at -20 °C, N-(3-Bromopropyl)phthalimide (655 mg, 2.4 mmol) was added and the mixture was stirred at -20 °C for 30 min and then warmed up to room temperature in 4 h. The reaction mixture was quenched with methanol (0.5 ml), diluted with dichloromethane (60 ml), filtered through a short silica gel column, evaporated to dryness to provide crude methyl 2-((1R,3R)(tert-butoxycarbonyl-amino)(3-(1,3- dioxoisoindolinyl)propoxy)methylpentyl)thiazolecarboxylate (85) which was used directly without further purification. To the crude compound 85 in THF (25 ml) at room temperature was added NaH (170 mg, 4.25 mmol, 60% in oil). After stirred for 45 min, CH I (0.20 ml, 3.21 mmol) was added. The mixture was stirred at room temperature overnight, quenched with NaH PO (2.0 M, 2 ml). The mixture was diluted with DMA (5ml), evaporated in vacuo, purified by SiO chromatography with EtOAc/dichloromethane (1:10 ~ 1:4) to afford the title compound (921 mg, 69%). ESI: m/z: [M+Na] , calcd for C H N NaO S 582.22, 28 37 3 7 found 582.22.
Example 29. 2-((1R,3R)(tert-butoxycarbonyl(methyl)amino)(3-(1,3-dioxoisoindolin- 2-yl)propoxy)methylpentyl)thiazolecarboxylic acid (87) To the dry compound 86 (910 mg, 1.63 mmol) in 1,2-dichloroethane (20 ml) was added trimethyltin hydroxide (400 mg, 2.21 mmol). The mixture was stirred at 80 °C overnight, concentrated, purified on SiO column, eluted with CH OH/dichloromethane/HOAc (1:10:0.01) to afford the title compound (756 mg, 85%). ESI: m/z: [M+Na] , calcd for C H N NaO S 27 36 3 7 546.22, found 546.22.
Example 30. Methyl 2-((1R,3R)acetoxy(tert-butoxycarbonyl(3-(1,3-dioxoisoindolin- 2-yl)propyl)amino)methylpentyl)thiazolecarboxylate (89) To the compound 71 (800 mg, 2.00 mmol) in THF (30 ml) at room temperature was added NaH (150 mg, 3.75 mmol, 60% in oil). After stirred for 45 min, N-(3- Bromopropyl)phthalimide (655 mg, 2.4 mmol) was added. The mixture was stirred at room temperature overnight, quenched with NaH PO (2.0 M, 2 ml). The mixture was added DMA (5 ml), evaporated and purified on SiO column, eluted with EtOAc/dichloromethane (1:10 ~ 1:4) to afford the title compound (971 mg, 82%). ESI: m/z: [M+Na] , calcd for C H N NaO S 610.22, found 610.22. 29 37 3 8 Example 31. 2-((1R,3R)acetoxy(tert-butoxycarbonyl(3-(1,3-dioxoisoindolin yl)propyl)amino)methylpentyl)thiazolecarboxylic acid (90) To the dry compound 89 (900 mg, 1.53 mmol) in 1,2-dichloroethane (35 ml) was added trimethyltin hydroxide (400 mg, 2.21 mmol). The mixture was stirred at 80 °C overnight, concentrated. Then the mixture in pyridine (20 ml) was added Ac O (3 ml) and stirred overnight, evaporated, purified on SiO column (MeOH/dichloromethane/HOAc, 5 1:10:0.01) to afford the title compound (755 mg, 86%). ESI: m/z: [M+Na] , calcd for C H N NaO S 28 35 3 8 596.20, found 596.20.
Example 32. (S)-Ethyl 5-(4-(benzyloxy)phenyl)(tert-butoxy-carbonylamino) methylpentenoate (185) (S)-Methyl 3-(4-(benzyloxy)phenyl)(tert-butoxy-carbonylamino)propanoate (184, 8.00 g, 20.76 mmol) in dichloromethane (250 ml) at -78 °C was added dropwise DIBAL (40 ml, 40 mmol, 1.0 M) in dichloromethane. After stirred at -78 °C for 2 h, the reaction was quenched with addition of MeOH (5 ml). The mixture was warmed to RT, acidified with 1 M HCl to pH 4 and separated. The aqueous layer was extracted with DCM (2 × 150 ml). The organic layers were washed with water, dried over Na SO , filtered and evaporated to dryness to form cude aldehyde intermediate. Then the crude intermediate aldehyde was dissolved in DCM, the ylide solution prepared from 1-(1-ethoxycarbonyl ethyl)-triphenylphosphonium bromide (18.0 g, 40.64 mmol) and KO Bu (5.00g, 44.64 mmol) in dichloromethane (80 ml) was added at RT.
After stirred at RT over night, the mixture was extracted concentrated and purified by SiO chromatography (EtOAc/Hexane, 1:8 ~ 1:4) to afford (6.90 g, 76%) of the title compound. ESI: m/z: [M+Na] , calcd for C H NNaO 462.22, found 462.22. 26 33 5 Example 33. (4R)-ethyl 4-((tert-butoxycarbonyl)amino)(4-hydroxyphenyl) methylpentanoate (186) (S)-Ethyl 5-(4-(benzyloxy)phenyl)(tert-butoxy-carbonylamino)methylpentenoate (185, 6.70 g, 15.26 mmol) in a hydrogenation bottle was charged methanol (150 ml), Pd/C (0.3 g, 10% Pd). The hydrogenation reaction was conducted at 30 psi for 6 h. The mixture was filtered through Celite, evaporated and crystallized with EtOH/hexane to the title compound (4.61 g, 86% yield). ESI: m/z: [M+Na] , calcd for C H NNaO 374.20, found 374.30. 19 29 5 Example 34. (4R)-ethyl 4-((tert-butoxycarbonyl)amino)(4-hydroxynitrophenyl) methylpentanoate (187) To a solution of compound 186 (4.50 g, 12.81 mmol) in anhydrous dichloromethane (200 ml) was added Ac O (2 ml, 21.16 mmol) and fuming HNO (0.65 ml, 14.07 mmol). The mixture was stirred at RT for 4 h, diluted with water (150 ml), separated and the aqueous layer was extracted with EtOAc. The organic layers were combined, dried over Na SO , evaporated and purified on SiO column (EtOAc/DCM, 1:10) to afford (4.21 g, 83%) of the title compound. ESI: m/z: [M+Na] , calcd for C H N NaO , 419.19, found 419.20. 19 28 2 7 Example 35. ethyl 4-((tert-butoxycarbonyl)amino)methyl(3-nitro (phosphonooxy)phenyl)pentanoate (188) Compound 187 (4.00 g, 10.09 mmol) in a mixture of CH CN (70 ml) and DMA (30 ml) was added DIPEA (4.00 ml, 23.00 mmol) at 0 °C. After stirred for 2 min, POCl (2.00 ml, 21.45 mmol) was added dropwise at 0 °C. The mixture was stirred at RT for 8 h, and quenched with slowly addition of NaHCO (3.5 g, 41.60 mmol) in water (20 ml) at 0 °C. After stirred at 0 °C overnight, the mixture was concentrated and purified on C-18 cartridge (20 x 4 cm) eluted with gradient mixture, 25 ml/min, A: 0.5% HOAc, B: CH OH, from 100% A in 10 min, then to 75% A and 25% B in 45 min. The fractions containing the product was pooled and evaporated to afford the title compound (3.89 g, 81% yield). ESI: m/z: [M-H] , calcd for C H N O P 19 28 2 10 475.16, found 475.20.
Example 36. (4R)((tert-butoxycarbonyl)amino)methyl(3-nitro (phosphonooxy)phenyl)pentanoic acid (189) To a solution of LiOH (5.0 g, 208.7 mmol) in water (60 mL) was added to a solution of compound (188) (3.75 g, 7.87 mmol) in THF (100 mL). After stirred for 4 h at 0 °C, the mixture was adjusted to pH ~6 with 4 M HCl, concentrated, and purified by C-18 chromatography, eluted with gradient mixture, 25 ml/min, A: 0.5% HOAc, B: CH OH, from 100% A in 10 min, then to 75% A and 25% B in 45 min. The fractions containing the product was pooled and evaporated to afford the title compound (2.82 g, 80% yield). ESI: m/z: [M-H] , calcd for C H N O P 447.12, found 447.20. 17 24 2 10 Example 37. (4R)(3-amino(phosphonooxy)phenyl)((tert-butoxycarbonyl)amino)- 2-methylpentanoic acid (190) Compound 189 (2.60 g, 5.80 mmol) in a hydrogenation bottle was charged methanol (80 ml), Pd/C (0.2 g, 10% Pd). The hydrogenation reaction was conducted at 35 psi of H for 6 h.
The mixture was filtered through Celite, evaporated to afford the crude title compound (190) (2.18 g, 90% yield), which was used directly without further purification. ESI: m/z: [M+Na] , calcd for C H N O P 417.15, found 417.15. 17 26 2 8 Example 38. (S)-methyl 2-((tert-butoxycarbonyl)amino)(4-hydroxy nitrophenyl)propanoate (196) To a solution of (S)-methyl 2-((tert-butoxycarbonyl)amino)(4-hydroxyphenyl) propanoate (195, 4.5 g, 15.24 mmol) in anhydrous dichloromethane (240 ml) was added Ac O (4 ml, 42.32 mmol) and fuming HNO (0.85 ml, 18.40 mmol). The mixture was stirred at RT for 4 h, diluted with water (150 ml), separated and the aqueous layer was extracted with EtOAc.
The organic layers were combined, dried over Na SO , evaporated and purified on SiO 2 4 2 column (EtOAc/DCM, 1:10) to afford (4.30 g, 83%) of the title compound. ESI: m/z: [M+Na] , calcd for C H N NaO , 363.13, found 363.20. 20 2 7 Example 39. (S)-methyl 2-((tert-butoxycarbonyl)amino)(3-nitro (phosphonooxy)phenyl)propanoate (197) To a solution of compound 196 (4.10 g, 12.05 mmol) in a mixture of CH CN (90 ml) was added DIPEA (4.00 ml, 23.00 mmol) at 0 °C. After stirred for 2 min, POCl (2.00 ml, 21.45 mmol) was added dropwise at 0 °C. The mixture was stirred at RT for 8 h, and quenched with slowly addition of NaHCO (3.5 g, 41.60 mmol) in water (20 ml) at 0 °C. After stirred at 0 °C overnight, the mixture was concentrated and purified on C-18 cartridge (20 × 4 cm) eluted with gradient mixture, 25 ml/min, A: 0.5% HOAc, B: CH OH, from 100% A in 10 min, then to 75% A and 25% B in 45 min. The fractions containing the product was pooled and evaporated to afford (4.20 g, 83%) the title compound. ESI: m/z: [M-H] , calcd for C H N O P 419.08, 20 2 10 found 419.10.
Example 40. 3-[3-amino(phosphonooxy)phenyl]-(2R){[(tert- butoxy)carbonyl]amino}-propanoic acid (198) To the dry compound 197 (4.0 g, 9.52 mmol) in the mixture of 1,2-dichloroethane (50 ml) and DMA (60 ml) was added trimethyltin hydroxide (4.00 g, 22.1 mmol). The mixture was stirred at 80 °C for 6h, evaporated, filtered through short SiO column eluted with water/MeCN (1:4). The fractions containing the product were pooled, concentrated to generate (S)((tert-butoxycarbonyl)amino)(3-nitro(phosphonooxy)phenyl)propanoic acid. In a hydrogenation bottle was charged DMA (70 ml), Pd/C (0.3 g, 10% Pd), followed by the addition of the prepared propanoic acid. The hydrogenation reaction was conducted at 30 psi of hydrogen for 6 h. The mixture was filtered through Celite, evaporated, and crystallized to afford (2.86 g, 80% yield) of the title compound (198), which was used directly without further purification. ESI: m/z: [M-H] , calcd for C H N O P 375.10, found 375.10. 14 20 2 8 Example 41. Benzyl 3-[4-(benzyloxy)phenyl]-(2R){[(tert-butoxy)carbonyl]-(methyl)- amino}-propanoate (200) A solution of benzyl 3-[4-(benzyloxy)phenyl]-(2R){[(tert-butoxy)carbonyl]- amino}propanoate (4.0 g, 8.67 mmol) in THF (60 ml) was added NaH (430 mg, 10.75 mmol, 60% in oil). After stirred at RT for 1 h, CH I (1.82 g, 12.82 mmol) was added, and the mixture was stirred overnight, quenched with CH OH (0.5 ml), evaporated and purified on SiO column (EtOAc/dichloromethane, 1:10) to afford the title compound (3.83 g, 93%). MS ESI: m/z: [M+Na] , calcd for C H NNaO 498.24, found 498.24. 29 33 5 Example 42. (2R){[(tert-butoxy)carbonyl](methyl)amino}(4-hydroxy nitrophenyl)propanoic acid (201) Compound 200 (3.80 g, 8.00 mmol) in a hydrogenation bottle was charged methanol (80 ml), Pd/C (0.3 g, 10% Pd). The hydrogenation reaction was conducted at 30 psi of hydrogen for 6 h. The mixture was filtered through Celite, evaporated to afford crude (2R) {[(tertbutoxy)carbonyl](methyl)amino}(4-hydroxyphenyl)propanoic acid (201a), which was used directly without further purification. To the compound 201a in anhydrous dichloromethane (240 ml) at -25 °C was added dropwise a mixture of SnCl4 (1.5 ml, 12.75 mmol) and fuming HNO (0.60 ml, 12.98 mmol) in dichloromethane (40 ml). The mixture was stirred at -25 °C for 75 min, quenched with saturated NaHCO to pH 3~4, separated and the aq. layer was extracted with EtOAc. The organic layers were combined, dried over Na SO , concentrated and purified on SiO2 column (MeOH/DCM/HOAc 1:8:0.01) to afford (1.98 g, 73%) of the title compound. ESI: m/z: [M+Na] , calcd for C H N NaO 363.13, found 20 2 7 363.13.
Example 43. (2R){[(tert-butoxy)carbonyl](methyl)amino}[3-nitro(phosphonooxy)- phenyl]propanoic acid (202) To a solution of compound 201 (1.98 g, 5.82 mmol) in a mixture of CH3CN (30 ml) and DMA (30 ml) was added DIPEA (2.00 ml, 11.50 mmol) at 0 °C. After stirred for 2 min, POCl (1.10 ml, 11.79 mmol) was added dropwise at 0 °C. The mixture was stirred at RT for 8 h, and quenched with slowly addition of NaHCO (2.0 g, 23.80 mmol) in water (10 ml) at 0 °C. After stirred at 0 °C overnight, the mixture was concentrated and purified on C-18 cartridge (20 × 4 cm) eluted with gradient mixture, 25 ml/min, A: 0.5% HOAc, B: CH OH, from 100% A in 10 min, then to 75% A and 25% B in 45 min. The fractions containing the product was pooled and evaporated to afford (1.96, 80%) the title compound. ESI: m/z: [M-H] , calcd for C H N O P 419.09, found 419.09. 20 2 10 Example 44. 3-[3-amino(phosphonooxy)phenyl]-(2R)[(tert-butoxycarbonyl) (methyl)-amino]-propanoic acid (203) Compound 202 (1.96 g, 4.67 mmol) in a hydrogenation bottle was charged DMA (60 ml), Pd/C (0.2 g, 10% Pd). The hydrogenation reaction was conducted at 30 psi of hydrogen for 6 h.
The mixture was filtered through Celite, evaporated to dryness to afford (1.74 g, 95%) of the title compound, which was used directly without further purification. ESI: m/z: [M-H] , calcd for C H N O P 389.12, found 389.12. 22 2 8 Example 45. tert-butyl N-(1-oxophenylpropan(2R)-yl)carbamate (204) (1S,2R)-(+)-Norephedrine (7.0 g, 46.29 mmol) in the mixture of THF (40 ml) and 1M NaHCO (100 ml) at 4 °C was added dropwise Boc O (10.15 g, 46.53 mmol) in THF (60 ml) in 45 min. The mixture then stirred at RT for 6 h, concentrated, extracted with EtOAc, dried over Na SO , concentrated and filtered through short SiO column eluted with EtOAc/Hexane 2 4 2 (1:2), concentrated to afford the crude tert-butyl N-((1S)-hydroxyphenylpropan(2R)- yl)carbamate (204b) ( 10.81, 93%). MS ESI: m/z+: [M+Na] , calcd for C H NaNO 274.15, 14 21 3 found 274.15.
The crude compound was used directly without further purification. Compound 204b in dichloromethane (50 ml) was added Dess-Martin periodinane solution in dichloromethane (180 ml, 0.3 M). After stirred for 1h, the mixture was added ice cold NaOH (1 M, 100 ml), separated and the organic layers were washed with 1M NaH PO , pH 6 (100 ml), dried over Na SO , evaporated and purified on SiO column (EtOAc/hexane 1:5) to afford the title 2 4 2 compound (9.34 g, 81% in two steps). MS ESI: m/z+: [M+Na] , calcd for C H NaNO 14 19 3 272.14, found 272.14.
Example 46. (1R,3R)((2S,3S)-N-(methyl)methyl((R)methylpiperidine carboxamido)-pentamido)methyl(4-( (1-oxophenylpropan yl)carbamoyl)thiazolyl)-pentyl acetate (205) Compound 204 (180 mg, 0.722 mmol) in 4 ml of dioxane was added HCl conc. (1.0 ml, 37%) at 4 °C and the mixture was stirred at RT for 30 min, evaporated and coevaporated with toluene to dryness. Then to the dryness solid in DMA (7 ml) were added compound 106 (251 mg, 0.466 mmol), EDC (305 mg, 1.56 mmol) and DIPEA (0.13 ml, 0.747 mmol) and the mixture was stirred for 8h, evaporated and purified on SiO column (EtOAc/dichloromethane, 1:4) to afford the title compound (255.3 mg, 82%). ESI: m/z+: [M+Na] , calcd for C H NaN O S 692.36, found 692.36. 51 5 6 Example 47. (1R,3R)((2S,3S)-N,3-dimethyl((R)methylpiperidinecarboxamido)- pentanamido)(4-((R)(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)oxophenyl-3,6,9- trioxa-13,14-diazaheptadecenylcarbamoyl)thiazolyl)methylpentyl acetate (206) Compound 205 (75 mg, 0.112 mmol) in methanol (5 ml) was added compound 12 (50 mg in HCl salt, 0.126 mmol) and HOAc (3 ul, 0.052 mmol). The mixture was stirred overnight, neutralized with DIPEA (23 ul, 0.132 mmol), evaporated and purified on SiO cartridge (4 g, EtOAc/dichloromethane, 1:5 ~1:3) to afford the title compound (79.3 mg, 70%). MS ESI: m/z+: [M+Na] , calcd for C H NaN O S 1033.51, found 1033.50. 50 74 8 12 Example 48. (1S,2R)(2-((1R,3R)acetoxy((2S,3S)-N,3-dimethyl((R) methylpiperidinecarboxamido)pentanamido)methylpentyl)thiazole carboxamido)phenyl-propyl 3-(2-(2 5 -(2-(2,5-dioxo-2,5-dihydro-1H-pyrrol yl)ethoxy)ethoxy)ethoxy)-propanoate (211) Compound 208a (95 mg, 0.141 mmol) and 3-(2-(2-(2-(2,5-dioxo-2,5-dihydro-1H-pyrrol- 1-yl)ethoxy)ethoxy)ethoxy)propanoic acid (55 mg, 0.182 mmol) in dichloromethane (5 ml) were added DCC (122 mg, 0.591 mmol) and DMAP (25 mg, 0.204 mmol) and the mixture was stirred overnight, evaporated and purified with SiO chromatography (EtOAc/dichloromethane, 1:3) to afford the title compound (95.1 mg, 71%). MS ESI: m/z+: [M+Na] , calcd for C H NaN O S 977.47, found 977.47. 48 70 6 12 Example 49. 2,5-dioxopyrrolidinyl 2-((1R,3R)acetoxy((2S,3S)-N,3-dimethyl ((R)methylpiperidinecarboxamido)pentanamido)methylpentyl)thiazole carboxylate (234) Compound 106 (788.1 mg, 1.464 mmol) in DMF (10 ml) were added NHS (202.0 mg, 1.756 mmol) and EDC (980 mg, 5.104 mmol) and the mixture was stirred overnight, evaporated and purified with SiO chromatography (EtOAc/dichloromethane, 1:3) to afford the title compound (762.8 mg, 82%). MS ESI: m/z+: [M+Na] , calcd for C H NaN O S 658.30, 45 5 8 found 658.30.
Example 50. (4R)(tert-butoxycarbonylamino)(3-(5-(2,5-dioxo-2,5-dihydro-1H- pyrrolyl)pentanamido)(phosphonooxy)phenyl)methylpentanoic acid (235) Compound 190 (825.1 mg, 1.973 mmol) in DMF (7 ml) was added 2,5-dioxopyrrolidin yl 5-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)pentanoate (23d) (711 mg, 2.417 mmol) and DIPEA (0.250 ml, 1.438 mmol), and the mixture was stirred overnight, evaporated and purified with C-18 chromatography (4×25 cm, v = 15 ml/min, 100% of 1% HOAc to 75% of 1% HOAc/25% MeOH in 45 min ) to afford the title compound (895.7 mg, 76%). MS ESI: m/z-: [M-H] , calcd for C H N O P 596.21, found 596.21. 26 35 3 11 Example 51. (4R)(tert-butoxycarbonylamino)(3-(3-(2-(2-(2-(2,5-dioxo-2,5-dihydro- 1Hpyrrolyl)ethoxy)ethoxy)ethoxy)propanamido)(phosphonooxy)phenyl) methylpentanoic acid (236) Compound 190 (632.5 mg, 1.512 mmol) in DMF (7 ml) were added 2,5-dioxopyrrolidin- 1-yl 3-(2-(2-(2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy)ethoxy)ethoxy)propanoate (24c) (727 mg, 1.826 mmol) and DIPEA (0.250 ml, 1.438 mmol), and the mixture was stirred overnight, evaporated and purified with C-18 chromatography (4 × 25 cm, v = 15 ml/min, 100% of 1% HOAc to 75% of 1% HOAc/25% MeOH in 45 min ) to afford the title compound 236 (763.2 mg, 72%). MS ESI: m/z-: [M-H] , calcd for C H N O P 700.25, found 700.25. 44 3 14 Example 52. (4R)(2-((1R,3R)acetoxy((2S,3S)-N,3-dimethyl((R) methylpiperidinecarboxamido)pentanamido)methylpentyl)thiazole carboxamido)(3-(5-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)pentanamido) (phosphonooxy)phenyl)methylpentanoic acid (239) Compound 235 (102 mg, 0.171 mmol) in 1,4-dioxane (4 ml) was added conc. HCl (1 ml, 37%) and the mixture was stirred for 30 min, evaporated to dryness to afford crude compound 237. To the crude compound in DMA (5 ml) were added compound 234 (110 mg, 0.173 mmol) and DIPEA (30 µl, 0.172 mmol), and the mixture was stirred for overnight, evaporated and purified by SiO chromatography (1% HOAc in water/acetone, 1:9 ~ 1:4) to afford the title compound (123.2 mg, 71%). MS ESI: m/z-: [M-H] , calcd for C H N O PS 1016.42, found 47 67 7 14 1016.42.
Example 53. (4R)(2-((1R,3R)acetoxy((2S,3S)-N,3-dimethyl((R) methylpiperidinecarboxamido)pentanamido)methylpentyl)thiazole carboxamido)(3-(3-(2-(2-(2-(2,5-dioxo-2,5-dihydro-1H-pyrrolyl)ethoxy)ethoxy)- ethoxy)-propanamido)(phosphonooxy)phenyl)methylpentanoic acid (240) Compound 236 (108 mg, 0.154 mmol) in 1,4-dioxane (4 ml) was added conc. HCl (1 ml, 37%) and the mixture was stirred for 30 min, evaporated to dryness to afford the crude compound 238. To the crude compound in DMA (5 ml) were added compound 234 (110 mg, 0.173 mmol) and DIPEA (30 µl, 0.172 mmol), and the mixture was stirred for overnight, evaporated and purified by SiO chromatography (1% HOAc in water/acetone, 1:9 ~ 1:4) to afford the title compound (131.2 mg, 76%). MS ESI: m/z-: [M-H] , calcd for C H N O PS 51 75 7 17 1120.47, found 1120.48.
Example 54. (4R)(tert-butoxycarbonylamino)methyl(4-(phosphonooxy)(4- (pyridinyldisulfanyl)butanamido)phenyl)pentanoic acid (244) To a solution of compound 190 (548.3 mg, 1.311 mmol) in DMF (10 ml) were added succinimidyl 4-(pyridinyl)disulfanyl)-butyrate (550.2mg, 1.687 mmol) and DIPEA (0.18 ml, 1.03 mmol). The mixture was stirred overnight, evaporated and purified by SiO chromatography (1% HOAc in water/acetone, 1:9 ~ 1:4) to afford the title compound (660.2 mg, 80%). MS ESI: m/z-: [M-H] , calcd for C H N O PS 628.16, found 628.16. 26 36 3 9 2 Example 55. (4R)(2-((1R,3R)acetoxy((2S,3S)-N,3-dimethyl((R) methylpiperidinecarboxamido)pentanamido)methylpentyl)thiazole carboxamido)methyl(4-(phosphonooxy)(4-(pyridin yldisulfanyl)butanamido)phenyl)pentanoic acid(248) Compound 244 (110.5 mg, 0.175 mmol) in 1,4-dioxane (4 ml) was added conc. HCl (1 ml, 37%) and the mixture was stirred at 4 °C for 30 min, evaporated to dryness to afford the crude compound 246. To the crude compound in DMA (5 ml) were added compound 234 (110 mg, 0.173 mmol) and DIPEA (30 ul, 0.172 mmol). The mixture was stirred overnight, evaporated and purified by SiO chromatography (1% HOAc in water/acetone, 1:9 ~ 1:4) to afford the title compound (129.1 mg, 71%). MS ESI: m/z-: [M-H] , calcd for C H N O PS 1048.38, found 47 68 7 12 3 1048.38.
Example 56. (4R)(2-((1R,3R)acetoxy((2S,3S)-N,3-dimethyl((R) methylpiperidinecarboxamido)pentanamido)methylpentyl)thiazole carboxamido)(3-(4-mercaptobutanamido)(phosphonooxy)phenyl) methylpentanoic acid (248b) Compound 248 (30 mg, 0.0285 mmol) in a mixture of DMA (2 ml) and NaH PO (0.1 M, pH 7) was added dithiothreitol (20 mg, 0.129 mmol). The mixture was stirred for 2 h, evaporated and purified by SiO chromatography (1% HOAc in water/CH CN, 1:9 ~ 1:4) to afford the title compound (22 mg, 85%). MS ESI: m/z-: [M-H] , calcd for C H N O PS 42 64 6 12 2 939.38, found 939.38.
Example 57. 4-(4-bromobutyl)oxaazatricyclo[5.2.1.0{2,6}]decene-3,5-dione (271) -oxaazatricyclo[5.2.1.0{2,6}]decene-3,5-dione (6.0 g, 36.35 mmol) and NaH (1.50 g, 37.50 mmol, 60% in oil) were stirred in DMA (60 ml) for 1 h, then 1,4-dibromobutane (35.0 g, 162.10 mmol) and NaI (0.50 g, 3.33 mmol) were added. The mixture was stirred overnight, quenched with CH OH (0.5 ml), evaporated, purified on SiO column (EtOAc/Hexane, 1:8) to afford the title compound (9.34 g, 86%). MS ESI: m/z+: [M+Na] , calcd for C H BrNaNO 322.02, found 322.02. 12 14 3 Example 58. Methyl 2-((1R,3R)(tert-butoxycarbonylamino)[4’-(3’’,6’’-endoxo-Δ- tetrahydrophthalimido)butyloxy]methylpentyl)thiazolecarboxylate (272) Methyl 2-((1R,3R)(tert-butoxycarbonylamino)hydroxymethylpentyl)thiazole carboxylate (70) (1.0 g, 2.79 mmol) and NaH (120 mg, 3.00 mmol, 60% in oil) were stirred in THF (30 ml) for 20 min and compound 271 (1.00 g, 3.34 mmol) and NaI (50 mg, 0.33 mmol) were added. The mixture was stirred overnight, quenched with MeOH (0.5 ml), evaporated and purified on SiO column (EtOAc/dichloromethane, 1:10) to afford the title compound (1.36 g, 84%). MS ESI: m/z+: [M+Na] , calcd for C H NaN O S 600.25, found 600.25. 28 39 3 8 Example 59. Methyl 2-((1R,3R)(N,N-tert-butoxycarbonylmethylamino)[4’-(3’’,6’’- endoxo-Δ-tetrahydrophthalimido)butyloxy]methylpentyl)thiazolecarboxylate (273) Compound 272 (1.30 g, 2.25 mmol) and NaH (108 mg, 2.70 mmol, 60% in oil) stirred in DMF (80 ml) for 1 h was added CH I (460 mg, 3.24 mmol). The mixture was stirred overnight, evaporated and purified on SiO column (EtOAc/dichloromethane, 1:12 ~ 1:8) to afford the title compound (1.01 g, 76%). MS ESI: m/z+: [M+Na] , calcd for C H NaN O S 614.26, 29 41 3 8 found 614.26.
Example 60. 2-((1R,3R)(N,N-tert-butoxycarbonylmethyl-amino)(4’- maleimidobutyloxy)methylpentyl)thiazolecarboxylic acid (274) To compound 273 (900 mg, 1.52 mmol) in a mixture of 1,2-dichloroethane (30 ml) and toluene was added trimethyltin hydroxide (400 mg, 2.21 mmol). The mixture was stirred at 100 °C overnight, concentrated, purified on SiO column eluted with MeOH/dichloromethane/HOAc (1:10:0.01) to afford the title compound (730 mg, 94%). ESI: m/z: [M+Na] , calcd for C H N NaO S 532.22, found 532.22. 24 35 3 7 Example 61. Methyl 2-((1R,3R)acetoxy(N,N-(tert-butoxycarbonyl)(4’-(3’’,6’’- endoxo-Δ-tetrahydrophthalimido)butyl)amino)methylpentyl)-thiazolecarboxylate (275) Compound 71 (1.50 g, 3.74 mmol) and NaH (180 mg, 4.50 mmol, 60% in oil) stirred in DMF (80 ml) for 1 h was added compound 271 (1.48 g, 4.94 mmol) and NaI (70 mg, 0.467 mmol). The mixture was stirred overnight, evaporated and purified on SiO column (EtOAc/dichloromethane, 1:10 ~ 1:6) to afford the title compound (1.60 g, 69%). MS ESI: m/z+: [M+Na] , calcd for C H NaN O S 642.26, found 642.26. 41 3 9 Example 62. 2-((1R,3R)acetoxy(N,N-(tert-butoxycarbonyl)(4’- maleimidobutyl)amino)methylpentyl)-thiazolecarboxylic acid (276) To compound 275 (800 mg, 1.29 mmol) in a mixture of 1,2-dichloroethane (40 ml) and toluene was added trimethyltin hydroxide (400 mg, 2.21 mmol). The mixture was stirred at 100 °C overnight, filtered through a short SiO column, washed the column with MeOH/dichloromethane/HOAc (1:5:0.01) and evaporated to dryness. To the crude dryness mixture in pyridine (15 ml) was added Ac O (0.3 ml, 3.17 mmol) at 0 °C. The mixture was stirred at RT overnight, evaporated and purified on SiO column eluted with MeOH/dichloromethane/HOAc (1:10:0.01) to afford the title compound (578.4 mg, 74%). ESI: m/z: [M+Na] , calcd for C H N NaO S 628.24, found 628.24. 29 39 3 9 Example 63. Phenylalanine-ketoepoxide N-Boc phenylalanine-ketoepoxide (Sun, L. et al, J. Mol. Catalysis A: Chem., 2005, 234 (1-2), 29-34) (300 mg, 1.08 mmol) in 1,4-dioxane (8 mL) at 0 °C was added hydrochloric acid (37%, 2 mL). The mixture was stirred for 1 hour at which time TLC showed complete consumption of starting material. The resulting solution was diluted with toluene (10 ml), evaporated and crystallized with EtOH/hexanes to yield HCl salt of the title compound (201 mg, 87%). ESI: m/z: [M+H] , calcd for C H NO 178.12, found 178.12. 11 16 Example 64. (S, E)-Ethyl 5-phenyl(tert-butoxy-carbonylamino)methylpentenoate (327) L-t-Boc-phenylalanine methyl ester 326 (5.60 g, 20.05 mmol) in dichloromethane (80 ml) at -78 °C was added dropwise DIBAL (40 ml, 40 mmol, 1.0 M) in dichloromethane. After stirred at -78 °C for 45 min, the ylide solution prepared from 1-(1-ethoxycarbonyl ethyl)- triphenylphosphonium bromide (18.0 g, 40.64 mmol) and KO Bu (5.00g, 44.64 mmol) in dichloromethane (80 ml) at RT was added at -78 °C. After stirred at -78 °C for 1 h and RT over night, the mixture was poured into 1 L of NaH PO (sat’ed.) solution with vigorously stirring. Separated and the aqueous phase was extracted with dichloromethane. The organic layers were dried over Na SO , concentrated and purified by SiO chromatography 2 4 2 (EtOAc/Hexane, 1:7 ~ 1:5) to afford 5.50 g (83% yields) of the title compound. ESI: m/z: [M+Na] , calcd for C H NNaO 356.19, found 356.20. 19 27 4 Example 65. Ethyl 3-((S)(tert-butoxycarbonylamino)phenylethyl)methyloxirane- 2-carboxylate (328) The compound 327 (5.0 g, 15.0 mmol) in dichloromethane (80 ml) was added 3- chloroperoxybenzoic acid (5.5 g, 22.3 mmol) and the mixture was stirred overnight, diluted with NaHCO (25 ml, sat’ed), separated and extracted the aqueous solution with dichloromethane. The organic layers were combined, dried over Na SO , filtered, evaporated and purified on SiO column (1:4 EtOAc/Hexane) to afford 4.71 g (90% yield) of the title compound. ESI: m/z: [M+Na] , calcd for C H NNaO 372.19, found 372.20. 19 27 5 Example 66. 3-((S)(tert-butoxycarbonylamino)phenylethyl)methyloxirane carboxylic acid (329) To a solution of LiOH (5.0 g, 208.7 mmol) in water (60 mL) was added to a solution of compound (328) (4.70 g, 13.45 mmol) in THF (100 mL). After stirred for 1 h, the mixture was concentrated, poured into water (150 mL) and the mixture was adjusted to pH ~ 4 with 4 M HCl aq. The mixture was extracted with EtOAc, dried over Na SO , concentrated and purified by SiO chromatography (MeOH/DCM/HOAc 1:10:0.01) to afford (3.97 g, 92%) of the title compound. ESI: m/z: [M+Na] , calcd for C H NNaO 344.16, found 344.16. 17 23 5 Example 67. 3-((S)(((9H-fluorenyl)methoxy)carbonylamino)phenylethyl) methyloxiranecarboxylic acid (331) To a solution of compound (329) (3.90g, 12.14 mmol) in dichloromethane (40 ml) at 0 °C, was added TFA (10 ml) and the mixture was stirred at 0 °C for 30 min, diluted with toluene, evaporated to dryness to form the crude TFA salt of compound 330. In a solution of Na CO (5.0 g, 47.16 mmol) in mixture of water (60 mL) and ethanol (30 ml) were added the crude compound 330 and Fmoc-Cl (3.70 g, 14.30 mmol). After stirred for 6 h, the mixture was concentrated, poured into water (150 mL) and the mixture was adjusted to pH ~ 4 with 4 M HCl aq. The mixture was extracted with EtOAc, dried over Na SO , concentrated and purified by SiO chromatography (MeOH/DCM/HOAc 1:10:0.01) to afford 3.87 g (72% yields over 2 steps) of the title compound. ESI: m/z: [M+Na] , calcd for C H NNaO 466.17, found 466.17. 27 25 5 Example 68. General Solution Peptide Coupling Procedure: The HCl salt of an amine (1 eq) was dissolved in dichloromethane or DMF (0.2 M) and cooled to 4 °C in an ice bath, followed by the addition of the appropriate Boc-protected amino acid (1.3 eq), EDC (2 eq), or TBTU (2 eq), or PyBrOP (2 eq), HOBt (1.5 eq) and DIPEA (3.5 eq). The reaction was allowed to slowly warm to room temperature and stirred for 15 h, after which it was diluted with EtOAc and washed successively with aqueous solutions of 1M HCl, saturated sodium bicarbonate, water and saturated sodium chloride. The organic layer was dried with Na SO , filtered and concentrated under reduced pressure. Purification by column chromatography (0% to 20% MeOH:dichloromethane) yielded Boc protected peptide.
Example 69. General Boc Deprotection Procedure: The Boc protected amino acid was dissolved in 20% TFA in dichloromethane or 4 M HCl in dioxane and stirred 30 min, or until the reaction was deemed complete by TLC. The solution was then concentrated under reduced pressure to give the TFA or HCl salt of the peptide. The TFA salt of the peptide was co-evaporated with 2% HCl in dichloromethane/toluene for 3~ 4 times to generate the HCl salt.
Example 70. General Solid Phase Peptide Synthesis (SPPS) Procedure: Boc SPPS was used Merrifield resin or modified 5 PAM resin or MBHA resin. Fmoc SPPS was used Wang resin, or 2-chlorotrityl resin, or HMPB, MBHA resin. The pre-treatment of the resin (pre-swell) and the first loading of an amino compound were followed the manufacture’s labels or notes. Resin bound Boc protected amino acids were deprotected with % TFA in dichloromethane or 3 M HCl in dioxane for 30 minutes and washed with DMF, MeOH, 50% DIPEA ( Pr NEt) in dichloromethane and dichloromethane. For steps involving the deprotection of multiple free amines, this step was repeated once before acylation to ensure completeness of reaction. Resin bound Fmoc protected amino acids were deprotected with % piperidine in DMF for 30 minutes and washed with DMF, MeOH and dichloromethane.
For steps involving the deprotection of multiple free amines, this step was repeated once before acylation to ensure completeness of reaction. The free amine beads were then suspended in a solution of the protected amino acid (3 eq per eq of free amine), TBTU or PyBrOP (3 eq per eq of free amine) and DIPEA (5 eq per eq of free amine) and mixed for 4 h, and then washed with DMF, MeOH and dichloromethane. For steps involving the acylation of multiple free amines, the coupling pr°Cedure was repeated once before deprotection to ensure completeness of reaction. These steps were repeated until the desired peptide was synthesized.
Example 71. General Cleavage of Peptides from Wang Resin or 2-Chlorotrityl resin: The peptide bound with Wang resin was mixed with 50% TFA in dichloromethane and triisopropylsilane (1~5%) or the peptide bound with 2-Chlorotrityl resin was mixed with 1% TFA in dichloromethane. Mixed for 2 h and then filtered. The resin was washed with dichloromethane (3×30 ml), methanol (3×30 ml), which were combined with the filtrate and evaporated to almost dryness. Cold Et O was then added to precipitate the desired deprotected peptide.
Example 72. General Cleavage of Peptides from Merrifield, MBHA or PAM resin: The peptide bound with the resins was mixed with HF/Me S/anisole (10:1:1) or CH SO H/Me S/ anisole (20:1:1), or HF/anisole/Me S/p-thioCresol (10:1:1:0.2) for peptide 3 3 2 2 containing Cys mixed for 2 h, concentrated under a stream of N , diluted with TFA and filtered.
The resin was washed with dichloromethane (3×30 ml), Methanol (3×30 ml), which were combined with the filtrate and condensed under reduced pressure to almost dryness. Cold Et O was then added to precipitate the desired deprotected peptide.
Example 73. Chromatographic purification: The crude peptide mixture was then purified through SiO column chromatography (10% to 25% MeOH in dichloromethane) or by a reverse phase HPLC eluted with gradient from 100% of water (optionally containing 1% HOAc) to 30% of water (containing 1% HOAc)/70% methanol in 1h, pooled the fraction, evaporated to give the desired protected.
Example 74. Conjugate Preparation.
A binding molecule, preferably an antibody can be conjugated to an antimitotic agents of this prevention through amide, or thiol ether or disulfide bond linkage. In a experiment of generating free thiols on an antibody, the antibodies (mAbs) (>5 mg/mL) in PBS containing 50 mM sodium borate, pH 8.0, were treated with dithiothreitol (10 mM final) at 35 °C for 30 min.
After gel filtration (G-25, PBS containing 1mM EDTA), thiol determination using Ellman's reagent [5,5’-dithiobis(2-nitrobenzoic acid)] indicated that there were approximately eight thiols per mAb. The free thiols were also introduced through conjugation of Traut’s Reagent (2-Iminothiolane) (Jue, R., et al. BioChem. 1978, 17 (25): 5399-5405), or through conjugation of SATP (N-succinimidyl-S-acetylthiopropionate) or SAT(PEG) linkers at pH 7~8, followed by releasing SH group with hydroxylamine treatment (Duncan, R, et al, Anal. BioChem. 1983, 132, 68-73; Fuji, N. et al, Chem. Pharm. Bull. 1985, 33, 362-367). On average, 5 ~ 8 of free thiols were introduced on mAbs.
To the mAbs containing free thiols at 4 °C were added the drug bearing maleimide or bromoacetamide (0.5 M sodium borate buffer pH 9 was required to promote mAb alkylation with bromoacetamide) (1.2~ 1.5 equiv of drug derivatives /SH group ratio) in cold DMA (2~20% v/v). After 1~2 h, the reactions were quenched with excess cysteine; the conjugates were concentrated by centrifugal ultrafiltration, gel filtered (G-25, PBS), and sterile filtered.
Protein concentration and drug loading were determined by spectral analysis at 280 and 252 nm, respectively. Size-exclusion HPLC was used to determine percent monomer of each conjugate prepared, and RP-HPLC established that there was less than 0.5% unconjugated cysteine quenched drug. The resulting conjugate was monomeric and contained, on the average, 3.2−4.2 antimitotic agents linked per antibody molecule for these thiol ether linked conjugation.
For the conjugation through DMPS, SMDP, SMPT, SPP, SPDP, SPDB, SMCC, or SM(PEG)n linkers. A solution of mAb (>5 mg/mL) in aqueous buffer (50 mM PBS, 50 mM NaCl, 1 mM EDTA) at pH 6.5~7.5 was incubated for 2 h with a 6 to 10-fold molar excess of a linker. The reaction mixture was purified via a Sephadex G25 gel filtration column to remove low molecular weight material. (The concentration of the antibody was determined spectrophotometrically if the linkers contained a pyridylthio. The coefficients for the antibody -1 -1 ε =2067550 M cm . An aliquot of the modified antibody was treated with an excess (>20 280nm equiv) of dithiothreitol and the release of pyridinethione determined using the known -1 -1 -1 -1 extinction coefficients of ε = 8080 M cm and ε = 5100 M cm for pyridine 343nm 280nm thione). The modified antibody was treated with 1.2 ~ 1.5 equiv of an antimitotic agent bearing a thiol group.
The reaction mixture was incubated for 5~18 h at RT. The reaction mixture was purified via a Sephadex G25 gel filtration column to remove the unconjugated drug and other low molecular weight species. The concentration of the conjugate was determined spectrophotometrically at 280 and 252 nm. The resulting conjugate was monomeric and contained, on the average, 3.2−4.5 antimitotic agent molecules linked per antibody molecule.
Example 75. In Vitro Cytotoxicity Assays BJAB (Burkitt’s lymphoma), BT-474 (breast carcinoma) cells, Namalwa (human Burkitt’s lymphoma), Ramos (human Burkitt’s lymphoma), COLO 205 (human colon aden°Carcinoma), and A375 (human malignant melanoma,) were from ATCC. The breast tumor line KPL-4 was from Dr. J. Kurebayashi (Kurebayashi, J. et al. Br J Cancer 1999; 79: 707–17).The cultures were maintained in RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum (FBS). All cell lines were cultured in a humidified incubator at 37°C, 6% CO . The cytotoxicity study was performed using a clonogenic assay similar to a reference described (Franken, et al, Nature Protocols 1, 2315-2319 (2006)). The test cell lines were plated into 6- well culture dishes at a constant number of 5000 cells per well. The cells were incubated with varying concentrations (1 pM to 50 nM) of the test-agent (the antimitotic agents or their conjugates) for 72 h. The medium was then aspirated from the plates and replaced with fresh medium. The cultures were allowed to grow and form colonies for a total of 7 to 10 days after plating. The cultures were then fixed and stained with 0.2% crystal violet in 10% formalin /PBS, and the colonies were counted. Plating efficiency of non-treated cells (medium alone) was determined by dividing the number of colonies counted by the number of cells plated. The surviving fraction of cells exposed to a toxic agent was determined by dividing the number of colonies in wells that were exposed to the agent by the number of colonies in the control wells.
References: Zanda, M. ; et al, Can. Pat. Appl. CA 2710693 (2011).
Chai, Y.; et al. Eur. Pat. Appl. 2174947 (2010), PCT WO 2010034724.
Leamon, C.; et al, PCT WO 2010033733, WO 2009002993..
Ellman, J.; et al, PCT WO 2009134279; PCT WO 2009012958 Matschiner, G.; et al, PCT WO 2009095447.
Vlahov, I.; et al, PCT WO 2009055562, WO 2008112873.
Low, P.; et al, PCT WO 2009026177.
Richter, W., PCT WO 2008138561.
Kjems, J.; et al, PCT WO 2008125116.
Davis, M.; et al, PCT WO 2008076333.
Diener, J.; et al, U.S. Pat. Appl. 20070041901, WO 2006096754 Matschiner, G.; et al, PCT WO 2006056464.
Vaghefi, F.; et al, PCT WO 2006033913 Doemling, A., Ger. Offen. DE 102004030227; PCT WO 2004005327; WO 2004005326; WO 2004005269.
Stanton, M.; et al, U.S. Pat. Appl. Publ. 20040249130.
Hoefle, G.; et al, Ger. Offen. DE 10254439 ; DE 10241152; DE 10008089.
Leung, D.; et al, PCT WO 2002077036.
Reichenbach, H.; et al, Ger. Offen. DE 19638870 Shibue, T., et al., Bioorg Med Chem Lett, 2011. 21(1): p. 431-4.
Floyd, W.C., 3rd, et al., ChemMedChem, 2011. 6(1): p. 49-53.
Shibue, T., et al., Total Syntheses of Tubulysins. Chemistry, 2010.
Kubicek, K., et al., Angew Chem Int Ed Engl, 2010. 49(28): p. 4809-12.
Chai, Y., et al., Chem Biol, 2010. 17(3): p. 296-309.
Chandrasekhar, S.,et al, J Org Chem, 2009. 74(24): p. 9531-4.
Pando, O., et al., Org Lett, 2009. 11(24): p. 5567-9.
Reddy, J.A., Mol Pharm, 2009. 6(5): p. 1518-25.
Ullrich, A., et al., Angew Chem Int Ed Engl, 2009. 48(24): p. 4422-5.
Ullrich, A.; et al, European J. Org. Chem. 2009, 36, 6367-6378 Schluep, T., et al., Clin Cancer Res, 2009. 15(1): p. 181-9.
Balasubramanian, R., et al., J Med Chem, 2009. 52(2): p. 238-40.
Leamon, C.P., et al.,. Cancer Res, 2008. 68(23): p. 9839-44.
Vlahov, I.R., et al., Bioorg Med Chem Lett, 2008. 18(16): p. 4558-61.
Patterson, A.W., et al, J Org Chem, 2008. 73(12): p. 4362-9.
Balasubramanian, R., et al., Bioorg Med Chem Lett, 2008. 18(9): p. 2996-9.
Raghavan, B., et al., J Med Chem, 2008. 51(6): p. 1530-3.
Richter, C.D., et al., Nat Chem Biol, 2008. 4(1): p. 75-81.
Patterson, A.W., et al., Chemistry, 2007. 13(34): p. 9534-41.
Wang, Z., et al., Chem Biol Drug Des, 2007. 70(2): p. 75-86.
Sani, M., et al., Angew Chem Int Ed Engl, 2007. 46(19): p. 3526-9.
Wipf, P. et al, Org Lett, 2007. 9(8): p. 1605-7.
Sasse, F., et al, Nat Chem Biol, 2007. 3(2): p. 87-9.
Peltier, H.M., et al., J Am Chem S°C, 2006. 128(50): p. 16018-9.
Domling, A., et al., Angew Chem Int Ed Engl, 2006. 45(43): p. 7235-9.
Khalil, M.W., et al., Chembi°Chem, 2006. 7(4): p. 678-83.
Kaur, G., et al., Bi°Chem J, 2006. 396(2): p. 235-42.
Wipf, P., et al, Org Lett, 2004. 6(22): p. 4057-60.
Steinmetz, H., et al., Angew Chem 5 Int Ed Engl, 2004. 43(37): p. 4888-92.
Friestad, G.K., et al, Org Lett, 2004. 6(19): p. 3249-52.
Sandmann, A., et al, Chem Biol, 2004. 11(8): p. 1071-9.
Sasse, F., et al., J Antibiot (Tokyo), 2000. 53(9): p. 879-85.

Claims (16)

What is claimed is:
1. A conjugate having a structure of Formula (I): 9 12 4 13 R R R R N 11 5 R 10 and pharmaceutical acceptable salts and solvates thereof, 5 wherein T is a targeting or binding ligand; L is a releasable linker; is a linkage bond that L connects to an atom inside the bracket independently; n is 1-20 and m is 1-10, wherein the cell-binding ligand T is selected from an antibody; a single chain antibody; an antibody fragment that binds to the target cell; a monoclonal antibody; a single chain monoclonal antibody; or a monoclonal antibody fragment that binds to the target cell; a 10 chimeric antibody; a chimeric antibody fragment that binds to the target cell; a domain antibody; a domain antibody fragment that binds to the target cell; adnectins that mimic antibodies; DARPins; a lymphokine; a hormone; a vitamin; a growth factor; a colony stimulating factor; a nutrient-transport molecule (a transferrin); or a binding peptide, protein, small molecule attached on albumin, a polymer, a dendrimer, a liposome, a nanoparticle, a 15 vesicle, or a (viral) capside, wherein the releasable linker L has the formula of: , wherein: - W- is a stretcher unit, which links a targeted binding molecular unit (T) to an amino acid unit (Aa) when presents, or links V when an Aa is not present, and W may independently contain a self-immolative spacer, a peptidyl unit, a hydrazone, a disulfide, a thioether, an ester, or an 20 amide bond; w is 0 or 1; Wherein Aa is independently a natural or unnatural amino acid unit; r is independently an integer ranging from 0 to 12; (Aa) represents a natural or unnatural amino acid, dipeptide, tripeptide, tetrapeptide, pentapeptide, hexapeptide, heptapeptide, octapeptide, nonapeptide, decapeptide, undecapeptide or dodecapeptide unit; Wherein V is a spacer unit and is independently H, O, NH, S, C -C of alkyl; C -C of heteroalkyl, alkenyl, 1 8 2 8 25 alkynyl; C -C of aryl, heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl, or one to four amino acid units, or (CH CH O) r is an integer ranging from 0 to 12; and v is 0, 1 or 2, 2 2 r, 1 2 3 4 inside the bracket is a potent antimitotic agent wherein R , R R and R are independently C -C of alkyl; C -C of heteroalkyl; C -C of aryl, Ar-alkyl, heterocyclic, carbocyclic, 1 8 2 8 3 8 30 alkylcycloalkyl, heterocycloalkyl heteroalkylcycloalkyl, alkylcarbonyl; or two R's, such as R R 2 3 3 4 5 6 12 13 R R R R , R R , and R R can be 3-7 members of a carbocyclic, cycloalkyl, or heterocyclic, 1 3 4 2 heterocycloalkyl ring system; Y is N or CH. In addition, R , R , and R can be H, and R can be absent, 5 6 8 10 wherein R R , R and R are independently H and C -C of alkyl or C -C heteroalkyl, , 1 4 2 4 7 14 14 1 15 14 1 15 1 wherein R is independently selected from H, R or -R C(=O)X R ; -R X R ; X is 5 selected from O, S, S-S, NH, or NR 9 14 14 14 wherein R is independently H, -O-, -OR , -OC(=O)R -, -OC(=O)NHR -, -OC(=O) 14 15 14 14 15 R SSR -, OP(=O)(OR )-, or OR OP(=O)(OR ), 11 14 14 16 14 2 16 14 2 2 wherein R is independently H, R , -R C(=O)R , -R X R , -R C(=O)X , wherein X 14 14 14 14 14 is -O-, -S-, -NH-, -N(R )-, -O-R -, -S-R -, -S(=O)-R -, or -NHR ; 12 14 14 14 10 wherein R is independently R , -O-, -S-, -N-, =N-, =NNH-, -NH(R ), -OR , - 16 14 14 14 16 C(O)O-, -C(O)OR -,C(O)NH-, C(O)NHR , -SR , -S(=O)R , -P(=O)(OR ) , - 16 16 16 OP(=O)(OR ) , -CH OP(=O)(OR ) , -SO R ; wherein R is C -C of alkyl, heteroalkyl, alkyl acid, alkyl amide, alkyl amine; C -C 1 10 2 10 of heteroalkyl; or Ar; Ar refers to an aromatic or hetero aromatic group, composed of one or 15 several rings, comprising four to ten carbon atoms, preferentially four to six carbon atoms, the term of hetero aromatic group refers to an aromatic group that has one or several carbon atoms replaced by hetero atoms, preferentially one, two or three carbon atoms replaced by O, N, Si, Se, P or S, more preferentially O, S, N, the term aryl or Ar also refers to an aromatic group, 17 16 16 wherein one or several H atoms can be replaced independently by R , F, Cl, Br, I, OR , SR , 16 17 16 16 16 17 16 17 16 16 16 16 17 20 NR R , N=NR , N= R , NR R , NO SOR R , SO R , SO R OSO R , PR R , 2, , 2 3 3 16 17 16 17 17 17 17 POR R , PO R R , OP(O)(OR ) , OCH OP(O)(OR ) , OC(O)OP(O)(OR ) , 2 2 2 2 2 16 17 17 17 17 17 PO(OR )(OR ), OP(O)(OR )OP(O)(OR ) , OC(O)R or OC(O)NHR , 14 15 wherein R and R are independently selected from C -C of alkyl; C -C of heteroalkyl 1 8 2 8 alkenyl, alkynyl; C -C of aryl, cycloalkyl, heterocyclic, carbocyclic, balkylcycloalkyl, 25 heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl, 16 17 wherein R , R are independently H, C -C of alkyl; C -C of heteroalkyl, alkenyl, 1 8 2 8 alkynyl; C -C of aryl, cycloalkyl, heterocyclic, carbocyclic, alkylcycloalkyl, heterocycloalkyl, heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl or C -C glycosides, or pharmaceutical 4 12 salts, 12 10 13 30 in addition, R can be H when R is not H, or when R is: k=0~4 , , , wherein Z 18 18 18 18 18 18 is H, CH OP(O)(OR ) , C(O)OP(O)(OR ) , PO(OR ) , P(O)(OR )OP(O)(OR ) , C(O)R , 2 2 2 2 2 18 18 C(O)NHR , SO (OR ), C -C glycosides, or C -C of alkyl, or C -C of carboxyalkyl, 2 4 12 1 8 3 8 heterocyclic; wherein R is H, C -C of alkyl; C -C of heteroalkyl, alkenyl, alkynyl; C -C of 1 8 2 8 3 8 2 18 19 aryl, carboxyalkyl, heterocyclic or alkylcarbonyl; where Z is NHR wherein R is H, OH, 18 18 18 18 18 NH , OSO (OR ), XCH OP(O)(OR ) , XPO(OR ) XC(O)OP(O)(OR , XC(O) R , 2 2 2 2 2 2 XC(O)NHR C -C of alkyl, carboxylic acid; C -C of alkenyl, alkynyl, carboxyalkyl; C -C 1 8 2 8 3 8 5 of aryl, heterocyclic, alkylcarbonyl; or pharmaceutical salts; X is O, S, NH; Y and Y are N or CH, independently, 12 11 or R can be H when R is: 2 8 8 or wherein X is O, S, N-R ; R is H, C -C of alkyl or C -C of heteroalkyl, 10 wherein the said conjugate of the invention specifically excludes the following structures: 24 25 26 wherein R is H or CH ; R is alkyl or C(O)alkyl or C(O)aryl; R is H, C -C alkyl, C - 3 1 5 2 28 27 C alkenyl, C(=O)C -C alkyl, or C(=O)C -C alkenyl; R is H or C -C alkyl; R is H, 1 to 3 5 1 5 2 5 1 5 15 substituents selected from halo, nitro, or carboxylate, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, 29 29 30 haloalkoxy, phenol protecting groups, or OR , wherein R is substituted aryl, C(O)R , 30 30 30 P(O)(OR )2, or SO R , wherein R is selected from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl or a metal cation; T is a targeting or binding ligand; L is a releasable linker.
2. A conjugate according to claim 1 having Formula (II): 4 R R 1 11 R R n (II) and pharmaceutical acceptable salts and solvates thereof, 9 14 14 14 wherein R is independently H, -OH, -OR , -OC(=O)R -, -OC(=O)NHR -, -OC(=O) 14 15 14 14 15 R SSR , OP(=O)(OR ) , or OR OP(=O)(OR ) , 12 14 14 14 wherein R is independently R , -OH, -SH, -NH , =NH, =NNH , -NH(R ), -OR , - 16 14 14 14 14 16 16 COR , -COOR -, C(O)NH , C(O)NHR , -SR , -S(=O)R , -P(=O)(OR ) , -OP(=O)(OR ) , 2 2 2 16 16 -CH OP(=O)(OR ) , -SO R , 12 10 13 5 in addition, R can be H when R is not H, or when R is: k=0~4 k=0~4 , , , wherein Z is H, 18 18 18 18 18 18 CH OP(O)(OR ) , C(O)OP(O)(OR ) , PO(OR ) , P(O)(OR )OP(O)(OR ) , C(O)R , 2 2 2 2 2 18 18 C(O)NHR , SO (OR ), C -C glycosides, or C -C of alkyl; C -C of carboxyalkyl, C -C of 2 4 12 1 8 2 8 3 8 2 18 18 heterocyclic; where Z is NHR or OPO(OR ) 12 11 10 or R can be H when R has: or group , wherein is attachment that is H, C -C of alkyl, C -C of alkyl carboxylic acid, C -C of alkyl carboxylic amide, C -C of alkyl carboxylic 1 6 2 6 2 6 2 8 8 ester, or C -C of heteroalkyl; wherein X is O, S, CH , N-R ; R is H, C -C of alkyl or C -C 2 6 2 1 6 2 6 of heteroalkyl. 15
3. The conjugate according to claim 1 having the structure of Formula (III): 12 13 O 5 6 R R R (III) and pharmaceutical acceptable salts and solvates thereof, 7 14 14 1 15 14 1 15 wherein R is independently selected from R or -R C(=O)X R -, or -R X R - in addition, R can be absent.
4. The conjugate according to claim 1, having the structure of Formula (IV): 9 12 O 5 6 R R R (IV) and pharmaceutical acceptable salts and solvates thereof, 9 14 14 14 wherein R is independently -O-, -OR -, -OC(=O)R -, -OC(=O)NHR -, -OC(=O) 14 15 14 9 R SSR -, OP(=O)(OR )O, in addition, R can be absent.
5. A conjugate according to claim 1 having a structure of Formula (V): 9 12 4 13 N 11 5 10 R R (V) and pharmaceutical acceptable salts and solvates thereof, 11 14 14 17 14 2 17 14 2 wherein R is independently -R -, -R C(=O)R -, -R X R -, -R C(=O)X -; 10 wherein R is independently H, OH, C -C of alkyl; C -C of alkenyl, alkynyl, 1 8 2 8 heteroalkyl; C -C of aryl, arylene, heterocyclic, carbocyclic, heterocycloalkyl, or an 2 14 14 14 14 amino acid, or two amino acid; X is -O-, -S-, -NH-, -N(R )-, -O-R -, -S-R -, -S(=O)-R -, 14 14 or -NHR -; R is C -C of alkyl; C -C of alkenyl, alkynyl, heteroalkyl C -C of aryl, 1 8 2 8 3 8 heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, 15 heteroalkylcycloalkyl, heteroaralkyl, alkylcarbonyl, wherein said conjugate of the Formula (V) specifically excludes the following structures: 25 26 wherein R is alkyl or C(O)alkyl or C(O)aryl; R is H, C -C alkyl, C -C alkenyl, C(=O)C - 1 5 2 5 1 C alkyl, or C(=O)C -C alkenyl; R is H, or 1 to 3 substituents selected from halo, nitro, 5 2 5 20 carboxylate or a derivative thereof, cyno, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, 29 29 30 30 phenol protecting groups, and OR , wherein R is substituted aryl, C(O)R , P(O)(OR ) , 30 30 or SO R , wherein R is selected from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl or a metal cation; T is a targeting or binding ligand; L is a releasable linker. 25
6. A conjugate according to claim 1 having a structure of Formula (VI): (VI) and pharmaceutical acceptable salts and solvates thereof, 12 14 14 14 wherein R is independently R , -O-, -S-, -NH-, =N-, =NNH-, -N(R )-, -OR -, - 14 14 14 14 15 C(O)O-, -C(O)NH-, C(O)NR -, -SR -, -S(=O)R -, -NHR -, -CH OP(=O)(OR )-, - 15 15 15 14 14 15 5 P(=O)(OR ) -, -C(O)OP(=O)(OR )-, -OP(=O)(OR )O , -SO R , wherein R and R are independently C -C of alkyl; C ~C of alkenyl, alkynyl, heteroalkyl; C -C of aryl, 1 8 2 8 3 8 heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl.
7. A conjugate according to claim 2 having a structure of Formula (VII): 10 (VII) and pharmaceutical acceptable salts and solvates thereof, wherein R is C -C of alkyl, alkyl acid, alkyl amide, alkyl amine; C -C of 1 10 2 10 heteroalkyl; or Ar; Ar refers to an aromatic or hetero aromatic group, composed of one or several rings, comprising four to ten carbon atoms, preferentially four to six carbon atoms, 15 the term of hetero aromatic group refers to an aromatic group that has one or several carbon atoms replaced by hetero atoms, preferentially one, two or three carbon atoms replaced by O, N, Si, Se, P or S, preferentially O, S, N, the term aryl or Ar also refers to an aromatic 18 16 group, wherein one or several H atoms are replaced independently by R , F, Cl, Br, I, OR , 16 16 17 16 16 16 18 16 18 16 16 16 SR , NR R , N=NR , N= R , NR R , NO SOR R , SO R , SO R OSO R , 2, , 2 3 3 16 18 16 18 16 18 16 18 16 18 20 PR R , POR R , PO R R , OPO OR R , CH OPO R R , 16 18 16 18 16 18 16 18 OP(O)(OR )OP(O)(OR ) , C(O)OPO R R , or PO R R , wherein R R are 3 3 , independently H, C ~C of alkyl; C ~C of alkenyl, alkynyl, heteroalkyl; C ~C of aryl, 1 8 2 8 3 8 heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, heteroaralkyl, heteroalkylcycloalkyl, alkylcarbonyl; or C ~C glycosides; or pharmaceutical salts, 4 12 25 wherein said conjugate of the formula (VII) specifically excludes the following structures: 24 25 26 wherein R is H or CH ; R is alkyl or C(O)alkyl or C(O)aryl; R is H, C -C alkyl, C -C 3 1 5 2 5 alkenyl, C(=O)C -C alkyl, or C(=O)C -C alkenyl; R is H or C -C alkyl; T is a targeting 1 5 2 5 1 5 or binding ligand; L is a releasable linker.
8. The conjugate according to claim 1, having the structure of Formula (VIII): OAc O (VIII) and pharmaceutical acceptable salts and solvates thereof; wherein T is a targeting or binding ligand; L is a releasable linker that connects to a 10 molecule inside the bracket independently; n is 1-20 and m is 1-10; q = 1-5; 1 18 18 18 wherein Z is H, CH OP(O)(OR , PO(OR ) C(O)OP(O)(OR ) , 2 2 2 2 18 18 18 18 18 P(O)(OR )OP(O)(OR ) , C(O)R , C(O)NHR , SO (OR ), C -C glycosides, or C -C 2 2 4 12 1 8 of alkyl, C -C carboxyalkyl, C -C heterocyclic; R is H, C -C of alkyl, heteroalkyl; of of 2 8 3 8 1 8 C -C of alkenyl, alkynyl, carboxyalkyl; C -C of aryl, alkylcarbonyl, heterocyclic; R is H or 2 8 3 8 15 CH .
9. The conjugate according to any one of claim 1, 2, 3, 4, 5, 6, 7 or 8 having a structure according to the formula IIa, IIb, IIg, IIh, IIi, , IIk, IIo, IIp, IIq, IIIa, , IIIc, IIIh, IIIi, IIIj, IVa, IVb, IVc, IVd, Va, Vb, Vc, Ve, Vh,VIa, VIb, VIc, VId, VIf, VIg, VIh, VIi, VIIa, VIIb, VIId, VIIe, VIIf, VIIg, VIIh, VIIi, VIIj,VIIk, VIIl, VIIm, and VIIn: 20 IIa OPO(OH) H X' O HN H O R N N N O Y' S NH Z' 5 IIk IIIa IIIc IIIh IIIi 5 IIIj 5 Va VIIa 5 VIIb VIId VIIe VIIf VIIg VIIh VIIi 5 VIIj VIIk VIIl VIIm VIIn ’ ’ ’ wherein Aa is a natural or unnatural amino acid; Ar is aryl; n is 1-20; q = 1-5; X , Y and Z are 22 22 23 independently CH, O, S, NH, or NR ; R and R are independently C -C of alkyl; C -C of 1 8 2 8 5 alkenyl, alkynyl, heteroalkyl; C -C of aryl, heterocyclic, carbocyclic, cycloalkyl, alkylcycloalkyl, heterocycloalkyl, Ar-alkyl, heteroalkylcycloalkyl, heteroaralkyl, or - (OCH CH ) -; R , R’ and R’’ are independently H or CH . Inside the round brackets are the 2 2 n 3 antimitotic drugs and the inside square brackets are the antimitotic drug conjugated to a cell- binding agent T via a linker.
10 10. The conjugate of the cytotoxic agents according to any one of claims 1, 2, 3, 4, 5, 6, 7 or 8 comprises antimitotic agents and linkers having structures of 232a, 232b, 232c, 232d, 232e, 232f, 232g, 232h, 232i, 232j, 232k, 232l, 232m, 232n, 232o, 232p, 232q, 232r, 232s, 232t, 221a, 221b, 233a, 233b, 233c, 233d, 222a, 290, 299a, 299b, 241, 242, 247, 248, 206, 211, 346, 350, 354, 357, 361, 366, 371, and 376 which are illustrated below: 15 232a 232b 232c 232d 232e 232f 232g 5 232h 232i 232j 232k 232l 232m 232n 5 232o 232p 232q 232r 232s 232t 221a 5 221b 233a 233b 233c 233d 222a 5 299a 299b 5 350
11. The conjugate of the cytotoxic agents according to any one of claim 1, 2, 3, 4, 5, 6, 7 5 or 8 comprises Formula of: mAb-TZ01, mAb-TZ04, mAb-TZ05, mAb-TZ06, mAb-TZ07, mAb-TZ08, mAb-TZ10a, mAb-TZ10b, mAb-5 TZ11, mAb-TZ12, mAb-TZ13, mAb-TZ14, mAb-TZ17, mAb-TZ18, mAb-TZ19, mAb-TZ20, mAb-TZ21, mAb-TZ22, mAb-TZ23, mAb- TZ24, and mAb-TZ25 which are illustrated below: mAb- 10 TZ01 mAb-TZ04 mAb- TZ05 mAb-TZ06 mAb-TZ07 mAb- TZ08 mAb- 5 mAb-TZ10a mAb-TZ10b mAb-TZ11 mAb-TZ12 mAb- 10 TZ13 mAb- TZ14 mAb-TZ17 5 mAb-TZ18 mAb- TZ19 mAb- TZ20 mAb-TZ21 mAb-TZ22 mAb-TZ23 mAb- 5 TZ24 mAb- TZ25 wherein mAb is an antibody.
12. The conjugate of the cytotoxic agents according to any one of claims 1, 2, 3, 4, 5, 6, 10 7, 8, 9, 10, or 11, for use in the manufacture of a medicament to target cells selected from tumor cells, virus infected cells, microorganism infected cells, parasite infected cells, autoimmune cells, activated cells, myeloid cells, activated T-cells, B cells, or melanocytes, cells expressing one or more of Apo2, ASLG659, BAFF-R, BMPR1B (bone morphogenetic protein receptor), IGF-IR, CA125, CanAg, E16, EGFR, EphA2 receptor, ErbB2, 15 interleukin,MUC1, MUC16, Napi3b (NAPI-3B, NPTIIb, SLC34A2, solute carrier family 34, member 2, type II sodium –dependent phosphate transporter 3b), VEGF, TF, MY9, anti-B4, EpCAM, FcRH2, C242, CD2, CD3, CD4, CD5, CD6, CD11, CD 11a, CD18, CD19, CD20, CD21, CD22, CD26, CD30, CD33, CD37, CD38, CD40, CD44, CD56, CD70, CD72, CD79, CD79a, CD79b, CD90, CD105, CD138, CRIPTO (CR, CR1, CRGF, 20 CRIPTO, CXCR5, LY64, TDGF1, Tetratocarcinoma-derived growth factors), endothelin B receptor (ETBR), EphA receptors, EphB receptors, EGFr, EGFRvIII, ETBR (Endothelin), FCRH1, HER2, HER2/neu, HER3, HLA-DOB (MHC class II molecule Ia antigen), integrins, IRTA2, LIV-1, MPF (MPF, MSLN, SMR, megakaryocyte potentiating factor, mesothelin), GEDA, Mesothelin, NaPi2b, PDL1, Sema 5b (FLJ10372, KIAA1445, 5 Mm42015, SEMA5B, 5EMAG, semaphoring 5 bHlog, sdema domain, seven thrombospondin repeats, cytoplasmic domain), PSCA, PSMA, STEAP1 (six transmembrane epithelial antigen of prostate 1), and STEAP2 (HGNC 8639, IPCA-1, PCANP1, STAMP1, STEAP2, STMP, prostate) antigens; and cells expressing insulin growth factor receptor, epidermal growth factor receptor, or folate receptor.
13. A linker when used to conjugate two cytotoxic agents of Formula (I), (II), (III), (IV),(V), (VI), (VII) or (VIII) per linker according to any one of claims 1, 2, 3, 4, 5, 6, 7, or 8 having a 3- bromo-maleimido, or 3, 4-dibromo-maleimido terminus of the formula: Wherein R is C -C alkyl or C H (OC H ) , n is 1-20; X is H or Br. 1 8 2 4 2 4 n 15
14. A cytotoxic agent- binding molecule conjugate of any one of claims 1, 2, 3, 4, 5, 6, 7, or 8, wherein two cytotoxic agents of Formula (I), (II), (III), (IV), (V), (VI), (VII) or (VIII) incorporated per linker, having the formula: Wherein R is C -C alkyl or C H (OC H ) , n is 1-20; D is a cytotoxic agent/drug of 1 8 2 4 2 4 n 20 Formula (I), (II), (III), (IV), (V), (VI), (VII) or (VIII), T is a cell binding molecule.
15. A cytotoxic agent- binding molecule conjugate any one of claims 1, 2, 3, 4, 5, 6, 7, or 8, wherein when two cytotoxic agents of Formula (I), (II), (III), (IV), (V), (VI), (VII) or (VIII), incorporated per linker, having the formula: 25 or Wherein R is C -C alkyl or C H (OC H ) , n is 1-20; D is a cytotoxic agent/drug of Formula (I), 1 8 2 4 2 4 n (II), (III), (IV), (V), (VI), (VII) or (VIII), T is a cell binding molecule.
16. A pharmaceutical composition comprising a therapeutically effective amount of the antimitotic agents-cell binding molecule conjugates of any one of claims 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11, or its pharmaceutically acceptable salt, and carrier, diluent, or excipient therefore, or 5 a combination therefore, for the treatment or prevention of cancer, autoimmune disease, or infectious disease.
NZ719369A 2012-07-12 Conjugates of Cell Binding Molecules with Cytotoxic Agents NZ719369B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ719369A NZ719369B2 (en) 2012-07-12 Conjugates of Cell Binding Molecules with Cytotoxic Agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
NZ70348712 2012-07-12
NZ719369A NZ719369B2 (en) 2012-07-12 Conjugates of Cell Binding Molecules with Cytotoxic Agents

Publications (2)

Publication Number Publication Date
NZ719369A NZ719369A (en) 2017-04-28
NZ719369B2 true NZ719369B2 (en) 2017-08-01

Family

ID=

Similar Documents

Publication Publication Date Title
US11767294B2 (en) Conjugates of cell binding molecules with cytotoxic agents
US10293055B2 (en) Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
US10265291B2 (en) Disulfur bridge linkers for conjugation of a cell-binding molecule
US10696700B2 (en) Charged linkers and their uses for conjugation
US9988408B2 (en) Cytotoxic agents for conjugation to a cell binding molecule
US10131682B2 (en) Hydrophilic linkers and their uses for conjugation of drugs to a cell binding molecules
AU2015242213A1 (en) Bridge linkers for conjugation of cell-binding molecules
NZ719369B2 (en) Conjugates of Cell Binding Molecules with Cytotoxic Agents