MXPA06005083A - Monomethylvaline compounds capable of conjugationto ligands - Google Patents

Monomethylvaline compounds capable of conjugationto ligands

Info

Publication number
MXPA06005083A
MXPA06005083A MXPA/A/2006/005083A MXPA06005083A MXPA06005083A MX PA06005083 A MXPA06005083 A MX PA06005083A MX PA06005083 A MXPA06005083 A MX PA06005083A MX PA06005083 A MXPA06005083 A MX PA06005083A
Authority
MX
Mexico
Prior art keywords
antibody
alkyl
genbank
drug conjugate
access
Prior art date
Application number
MXPA/A/2006/005083A
Other languages
Spanish (es)
Inventor
D Senter Peter
Polakis Paul
D Spencer Susan
O Doronina Svetlana
E Toki Brian
J Ebens Allen
Beth Kline Toni
X Sliwkowski Mark
Original Assignee
O Doronina Svetlana
J Ebens Allen
Beth Kline Toni
Polakis Paul
Seattle Genetics Inc
D Senter Peter
X Sliwkowski Mark
D Spencer Susan
E Toki Brian
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by O Doronina Svetlana, J Ebens Allen, Beth Kline Toni, Polakis Paul, Seattle Genetics Inc, D Senter Peter, X Sliwkowski Mark, D Spencer Susan, E Toki Brian filed Critical O Doronina Svetlana
Publication of MXPA06005083A publication Critical patent/MXPA06005083A/en

Links

Abstract

Auristatin peptides, including MeVal-Val-Dil-Dap-Norephedrine (MMAE) and MeVal-Val-Dil-Dap-Phe (MMAF), were prepared and attached to Ligands through various linkers, including maleimidocaproyl-val-cit-PAB. The resulting ligand drug conjugates were active in vitro and in vivo.

Description

ZW), Eurasian (AM, AZ, BY, KG, KZ, MD, UK, TJ, TM), Fortwo-letter codes and other abbreviations, referred to the "GuidEuropean (AT, BE, BG, CH, CY, CZ, DE , DK, EE, ES, Fl, ance Notes on Codes and Abbreviations "appearing at the begin- FR, GB, GR, HU, IE, IS, IT, LU, MC, NL, PL, PT, RO, SE, ning regular issue of the PCT Gazette, SI, SK, TR), OAPI (BF, BJ, CF, CG, Cl, CM, GA, GN, GQ, GW, ML, MR, NE, SN, TD, TG). Published: - without intemational search report and to be republished upon receipt of that report COMPOUNDS OF MONOMETILVALINE CAPACES FROM CONJUGATION TO LIGANDS CONTINUITY This application claims the benefit of the Provisional Patent Application of E.U. No. 60 / 518,534, filed on November 6, 2003; Provisional Patent Application of E.U. No. 60 / 557,116, filed on March 26, 2004; Provisional Patent Application No. 60 / 598,899, filed on August 4, 2004; and Provisional Patent Application of E.U. No. 60 / 622,455, filed on October 27, 2004; whose descriptions are incorporated by reference herein. 1. FIELD OF THE INVENTION The present invention is directed to a drug compound and more particularly to drug-linker-ligand conjugates, drug-linker compounds, and drug-ligand conjugates, to compositions including the same, and to methods to use them to treat cancer, an autoimmune disease or an infectious disease. The present invention is also directed to antibody-drug conjugates, to compositions including the same, and to methods for the use thereof to treat cancer, an autoimmune disease or an infectious disease. The invention also relates to methods for the use of an antibody-drug conjugate for in situ and in vivo diagnosis or treatment of mammalian cells or associated pathological conditions. 2. BACKGROUND OF THE INVENTION For many years, it has been the focus of considerable research the improvement of the supply of drugs and other agents to target cells, tissues and tumors, to achieve maximum efficacy and minimal toxicity. Although many attempts have been made to develop effective methods for importing biologically active molecules into cells, both in vivo and in vitro, none has proven completely satisfactory. The optimization of the association of the drug with its intracellular target, while minimizing the intercellular redistribution of the drug, e.g., to neighboring cells, is often difficult or inefficient. Most agents currently administered to a patient parenterally, are not directed, resulting in a systemic delivery of the agent to cells and tissues of the body where they are unnecessary, and often undesirable. This can result in adverse side effects, and often limits the dose of a drug (e.g., chemotherapeutic (anti-cancer), cytotoxic, enzyme inhibiting, and antiviral or antimicrobial drugs) that can be administered. In comparison, although oral drug administration is considered a convenient and economical mode of administration, it shares the same problems of non-specific toxicity to unaffected cells once the drug has been absorbed into the systemic circulation. Additional complications involve problems with oral bioavailability and residence of the drug in the intestine, leading to further exposure of the intestine to the drug and thus to the risk of toxicities in the intestine. Accordingly, a major goal has been to develop methods to specifically target the agents to cells and tissues. The benefits of such treatment include avoiding the general physiological effects of an inappropriate supply of such agents to other cells and tissues, such as uninfected cells. The intracellular direction can be achieved by methods, compounds and formulations that allow the accumulation or retention of biologically active agents, i.e., active metabolites, within the cells. Monoclonal antibody therapy has been established for the targeted treatment of patients with cancer and immunological and angiogenic diseases. The use of antibody-drug conjugates for the local delivery of cytotoxic or cytostatic agents, e.g., drugs to destroy or inhibit tumor cells in the treatment of cancer (Syrigos and Epenetos (1999) Anticancer Research 19: 605-614; Niculescu-Duvaz and Springer (1997) Adv. Drg. Del. Rev., 26: 151-172; Patent of E.U. No. 4975278), theoretically allows a targeted delivery of the drug residue to tumors, and their intracellular accumulation therein, while systemic administration of these unconjugated drug agents can result in unacceptable levels of toxicity to normal cells as well. as for the tumor cells that it is desired to eliminate) Baldwin et al., 1986, Lancet pp. (March 15, 1986): 603-05; Thorpe, 1985, "Antibody Carriers of Cytotoxic Agents in Cancer Therapy: A Review", in Monoclonal Antibodies '84: Biological and Clinical Applications, A. Pinchera et al., (Eds.), Pp. 475-506). Consequently, maximum efficacy is sought with minimal toxicity. Both polyclonal antibodies and monoclonal antibodies have been reported to be useful in these strategies (Rowland et al., 1986, Cancer Immunol., Immunother, 21: 183-87). Drugs used in these methods include daunomycin, doxorubicin, methotrexate and vindesine (Rowland et al., 1968, supra). Toxins used in antibody-toxin conjugates include bacterial toxins such as diphtheria toxin, plant toxins such as ricin, small molecule toxins such as geldanamycin (Kerr et al., 1997, Bioconjugate Chem. 8 (6): 781- 784; Mandler et al., (2000) Journal of the Nat. Cancer Inst., 92 (19): 1573-1581; Mandler et al., (2000) Bioorganic &Med. Chem. Letters, 10: 1025- 1028; Mandler et al., (2000) Bioconjugate Chem. 13: 786-791), maytansinoids (EP 1391213; Liu et al., (1996) Proc. Nati. Acad. Sci. USA 93: 8618-8623) and calicheamicin (Lode et al., (1998) Cancer Res. 58-2928; Hinman et al., (1993) Cancer Res. 53: 3336-3342). Toxins can affect their cytotoxic and cytostatic effects through mechanisms that include tubulin binding, DNA binding, or topoisomerase inhibition (Meyer, DL and Senter, PD "Recent Advances in Antibody Drug Conjugates for Cancer Therapy" in Animal Reports in Medicinal Chemistry "Vol. 38 (2003) Chapter 23, 229-237.) Some cytotoxic drugs tend to be inactive or less active when conjugated to large antibodies or protein receptor ligands.ZEVALIN® (ibritumomab tiuxetan, Biogen / Idec) is a conjugate of antibody-radioisotope composed of a monoclonal murine IgGl kappa antibody directed against the CD20 antigen found on the surface of normal and malignant B lymphocytes and lxlIn or 90Y radioisotopes linked by a thiourea linker-chelator (iseman et al., (2000) Eur Jour Nucí, Med., 27 (7): 766-77; Wiseman et al., (2002) Blood 99 (12): 4336-42; Witzig et al., (2002) J. Clin. Oncol. (10): 2453-63; Witzig et al., (2002) J. Clin. Oncol 20 (15): 3262-69). Although ZEVALIN® has activity against B-cell non-Hodgkin lymphoma (NHL), its administration does not result in severe and prolonged cytopenias in the majority of patients. MYLOTARG® (gemtuzumab ozogamicin, Wyeth Pharmaceuticals), an antibody-drug conjugate composed of a hu CD33 antibody bound to calicheamicin, was approved in 2000 for the treatment of acute myeloid leukemia by injection (Drugs of the Future (2000) 25 ( 7) -.686; US Patents Nos. 4970198; 5079233; 5585089; 5606040; 5793762; 5739116; 5767285; 5773001). Cantuzuman mertansine (Immunogen Inc.), an antibody-drug conjugate composed of hu C242 antibody bound through the disulfide linker SPP to the maytansinoid drug residue, DM1, is progressing in Phase II trials for the treatment of cancers expressing CanAg , such as colon, pancreatic, gastric and others. MLN-2704 (Millennium Pharm., BZL Biologics, Immunogen Inc.), an antibody-drug conjugate composed of the specific anti-prostate membrane antigen monoclonal antibody (PSMA) bound to the maytansinoid drug residue, DM1, is under development for the potential treatment of prostate tumors. The same maytansinoid drug residue, DM1, was linked through a non-disulfide linker, SMCC, to a mouse murine monoclonal antibody, TA.l (Chari et al., (1992) Cancer Research 52: 127-131). This conjugate reported being 200 times less potent than the corresponding disulfide bond conjugate. The SMCC linker was considered "non-divisible". Several short peptidic compounds of the marine mollusk Dolabella auricularia have been isolated and found to have biological activity (Pettit et al., (1993) Tetrahedron 49: 9151; Nakamura et al., (1995) Tetrahedron Letters 36: 5059-5062; Soné et al-, (1995) Jour. Org. Chem. 60: 4474). Analogs of these compounds have also been prepared and some were found to have biological activity (for a review see Pettit et al., (1998) Anti-Cancer Drug Design 13: 243-277). For example, auristatin E (U.S. Patent No. 5635483) is a synthetic analogue of the marine natural product Dolastatin 10, an agent that inhibits the polymerization of tubulin by binding to the same tubulin domain as the anticancer drug vincristine (GR Pettit, (1997) Prog. Chem. Org. Nat. Prod. 70: 1-79). Dolastatin 10, auristatin PE, and auristatin _E, are linear peptides that have four amino acids, three of which are unique to the class of dolastatin compounds, and one termination amide C. The peptides of auristatin, auristatin E (AE) and monomethylauristatin (MMAE), synthetic analogs of dolastatin, were conjugated to: (i) cBR96 chimeric monoclonal antibodies (specific for Lewis Y in carcinomas); (ii) cAVIO that is specific for CD30 in hematological diseases (Kluss an et al., (2004), Bioconjugate Chemistry, 15 (4): 765-773, Doronina et al., (2003) Nature Biotechnology 21 (7) - 778-784; "Monomethyl aline Compounds Capable of Conjugation to Ligands"; Francisco et al., (2002) Blood 102 (4): 1458-1465; EU Publication 2004/0018194; (iii) Anti-CD20 antibodies such as RITUXAN ® (WO 04/032828) for the treatment of cancers expressing CD20 and immune disorders; (iv) antibodies against EphB2 2H9 and IL-8 for the treatment of colorectal cancer (Mao et al., (2004) Cancer Research 64 (3 ): 781-788) (v) antibody E ~ selectin (Bhaskar et al., (2003) Cancer Res. 63: 6387-6394), and (vi) other anti-CD30 antibodies (WO 03/043583). conjugated to monoclonal antibodies is described in Senter et al., Proceedings of the American Association for Cancer Research, Volume 45, Excerpt Number 623, filed on March 28, 2004. Despite the in vitro data for the Dolastatin class compounds and their analogs, the significant general toxicities in the doses required to achieve a therapeutic effect compromise their effectiveness in clinical studies. Accordingly, there is a clear need in the art for dolastatin / auristatin derivatives that have significantly less toxicity, and still a useful therapeutic efficiency. These and other limitations and problems of the past are addressed by the present invention. The ErbB family of tyrosine receptor kinases are important mediators of cell growth, differentiation and survival. The receptor family includes four distinct members including the de facto epidermal growth receptor (EGFR, ErbBl, HERÍ), HER2 (ErbB2 or pl85neu), HER3 (ErbB3) and HER4 (ErbB4 or tyro2). A panel of anti-ErbB2 antibodies has been characterized using the SKBR3 human breast cancer cell line (Hudziak et al., (1989) Mol Cell Biol. 9 (3): 1165-1172) Maximum inhibition was obtained with the antibody called 4D5 which inhibited cell proliferation by 56% Other antibodies in the panel reduced cell proliferation to a lesser degree in this analysis.It was further found that the 4D5 antibody sensitizes breast tumor cell lines overexpressing ErbB2 for the cytotoxic effects of TNF-a (U.S. Patent No. 5677171) The anti-ErbB2 antibodies treated in Hudziak et al. Are further characterized in Fendly et al., (1990) Cancer Research 50: 1550-1558; otts et al., (1990) In Vitro 26 (3): 59a; Sarup et al., (1991) Grouth Regulation 1: 72-82; Shepard et al., (1991) Clin. Immunol. 11 (3) .117-127; Kumar et al., (1991) Mol. Cell Biol. 11 (2): 979-986; Lewis et al., (1993) Cancer Immunol. Immunother. 37: 255-263; Pietras et al., (1994) Oncogene 9: 1929-1838; Vitetta et al., (1994) Cancer Research 54: 5301-5309; Sliwkowski et al., (1994) J. Biol. Chem. 269 (20): 14661-14665; Scott et al., (1991) J. Biol. Chem. 266: 14300-5; D'souza et al., Proc. Nati Acad. Sci. (1994) 91: 7202-7206; Lewis et al., 0 (1996) Cancer Research 56: 1457-1465; and Schaefer et al., (1997) Oncogene 15: 1385-1394. Other anti-ErbB2 antibodies with various properties have been described in Taagliabue et al., Int. J. Cancer 47: 933-937 (1991); McKenzie et al., Oncogene 4: 543-548 (1989); Maier et al., Cancer Res., 51: 5361-5369 (1991); Bácus et al., Molecular Carcinogenesis 3: 350-362 (1990); Stancovski et al., Proc. Nati Acad. Sci. USA 88: 8691-8695 (1991); Bacus et al., Cancer Research 52: 2580-2589 (1992); Xu et al., Int. J. Cancer 53: 401-408 (1993); WO 94/00136; Kasprzyk et al., Cancer Research 52: 2771-2776 (1992); Hancock et al., (1991) Cancer Res. 51: 4575-4580; Shawver et al., (1994) Cancer Res. 54: 1367-1373; Arteaga et al., (1994) Cancer Res. 54: 3758-3765; Harwerth et al., (1992) J. Biol. Chem. 267: 15160-15167; Patent of E.U. No. 5783186; and Klapper et al., (1997) Oncogene 14: 2099-2109. The homology display has resulted in the identification of two other members of the ErbB receptor family; ErbB3 (U.S. Patent No. 5,183,884; U.S. Patent No. 5,480,968; Kraus et al., (1989) Proc. Nati. Acad. Sci. USA 86: 9193-9197) and ErbB4 (EP 599274; Plowman et al. , (1993) Proc. Nati, Acad. Sci. USA 90: 1746-1750, and Plowman et al., (1993) Nature 366: 473-475). Both receptors display an increased expression on at least some breast cancer cell lines. HERCEPTIN® (Trastuzumab) is a recombinant monoclonal antibody derived from DNA that selectively binds with high affinity in a cell-based analysis (Kd = 5 nM) to the extracellular domain of the human epidermal growth factor receptor 3 protein, HER2 (ErbB2) (US Patent No. 5821337; US Patent No. 6054297; US Patent No. 6407213; US Patent No. 6639055; Coussens L., et al., (1985) Science 230: 1132-9; Slamon DJ et al., (1989) Science 244: 707-12). Trastuzumab is an IgGl kappa antibody that contains regions of human structure with the complementarity determining regions of a murine antibody (4D5) that binds to HER2. Trastuzumab binds to the HER2 antigen and therefore inhibits the growth of cancer cells. Because Trastuzumab is a humanized antibody, it minimizes any HAMA response in patients. The humanized antibody against HER2 is produced by a suspension culture of a mammalian cell (Chinese Hamster Ovary, CHO). The proto-oncogene HER2 (or c-erbB2) codes for a transmembrane receptor protein of 185 kDa, which is structurally related to the epidermal growth factor receptor. Overexpression of the HER2 protein is observed in 25% -30% of primary breast cancers and can be determined using an immunohistochemical assessment of fixed tumor blocks (Press MF, et al., (1993) Cancer Res. 53 : 4960-70 Trastuzumab has been shown in both in vitro and animal analyzes to inhibit the proliferation of human tumor cells overexpressing HER2 (Hudziak RM, et al., (1989) Mol. Cell Biol. 9: 1165-72.; Lewis GD, et al., (1993) Cancer Immunol Immunother 37: 255-63; Baselga J. et al., (1998) Cancer Res. 58: 2825-2831) Trastuzumab is a mediator of cellular dependent cytotoxicity of antibody, ADCC (Hotaling TE, et al-, (1996) [abstract], Proc. Annual Meeting Am Assoc. Cancer Res., 37-471; Pegram MD, et al., (1997) [abstract], Proc. Am Assoc. Cancer Res., 38: 602) The in vitro ADCC mediated by Trastuzumab has been shown to be exerted preferentially in cells overexpressing HER2 compared to cancer cells which do not. overexpress HER2. HERCEPTIN® as a single agent is indicated for the treatment of patients with metastatic breast cancer whose tumors overexpress the GER2 protein and who have received one or more chemotherapy regimens for their metastatic disease. HERCEPTIN® in combination with paclitaxel is indicated for the treatment of patients with metastatic breast cancer whose tumors overexpress the HER2 protein and who have not received chemotherapy for their metastatic disease. HERCEPTIN® is clinically active in patients with metastatic breast cancers overexpressing ErbB2 who have received extensive anti-cancer therapy previously (Baselga et al., (1996) J. Clin. Oncol. 14: 737-744). The anti-HER2 monoclonal murine antibody inhibits the growth of breast cancer cell lines that overexpress HER2 at the 2+ and 3+ levels (1-2 x 10e HER2 receptors per cell), but has no activity in cells expressing lower levels from HER2 (Lewis et al., (1993) Cancer Immunol. Immunother., 37: 255-263). Based on this observation, the 4D5 antibody was humanized (huMAb4D5-8, rhuMAb HER2, U.S. Patent No. 5821337; Carter et al., (1992) Proc. Nati Avad. Sci. USA 89: 4285-4289) and was tested in patients with breast cancer whose tumors overexpressed HER2 but had progressed after conventional chemotherapy (Cobleigh et al- (1999) J. Clin Oncol. 17: 2639-2648). Although HERCEPTIN® is an important discovery for the treatment of patients with breast cancers overexpressing ErbB2 who have received extensive anti-cancer therapy, some patients in this population failed to respond or only responded poorly to treatment with HERCEPTIN. Consequently, there is a significant clinical need for the development of additional therapies for cancer directed to HER2 for those patients with tumors that overexpress HER2 or other diseases associated with the expression of HER2 that do not respond, or that respond poorly to treatment with HERCEPTIN. The mention of any reference in this application is not an admission that the reference is the prior art to this request. 3. SUMMARY OF THE INVENTION In one aspect, the present invention provides drug-linker-ligand compounds having the Formula a: or a pharmaceutically acceptable salt or solvate thereof, wherein, L- is a ligand unit; -Aa-Ww-Yy- is a linker unit (LU), where the linker unit includes: -A- is a stretch unit, a is 0 or 1, each -W- is independently an amino acid unit, w is an integer that ranges from 0 to 12, -Y- is a unit of separation, and y is O, 1 or 2; p ranges from 1 to about 20; and -D is a drug unit that has the DE and DE formulas wherein, independently at each location: R2 is selected from H and alkyl is Ca-C8; R3 is selected from H, C?-C8 alkyl, C3-C8 carbocycle, aryl, alkylaryl d-Cs, Ci-Cs alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C?-C8 alkyl (C3-C8 heterocycle) ); R4 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, alkylaryl Ca-C8, alkyl CL-CS (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3-C8 heterocycle) ); R5 is selected from H and methyl; R * and R5 together form a carbocyclic ring and have the formula - (CRaRb) n- wherein Ra and Rb are independently selected from H, Ci-Cs alkyl, and C3-C8 carbocycle, and n is selected from 2, 3, 4 , 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -Cß); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C-C8 alkyl); R9 is selected from H and Ci-Cβ alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is O, S, NH, or NR12, wherein R12 is CL-C8 alkyl; R11 is selected from H, C? -C8 alkyl, C3-C8 heterocycle, - (R130) m? R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or alkyl d.-Cacada occurrence of R1S is independently, H, COOH, - (CH2) nN (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? - C8; each occurrence of R16 is independently H, C? -C8 alkyl, or - (CH2) n -COOH; where n is an integer that fluctuates from 0 to 6; and R18 is selected from aryl -C (R8) 2-C (R8) 2, -C (R8) 2-C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) ) 2- (C3-C8 carbocycle). In another aspect, drug compounds having the Formula Ib are provided: or its salts or pharmaceutically acceptable salts or solvates thereof, wherein; R2 is selected from hydrogen and C? -C8 alkyl; J is selected from hydrogen, C?-C8 alkyl, C3-C8 carbocycle, aryl, C-C8 alkylaryl, C?-C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C?-C8 alkyl (C3-heterocycle) C8); R "is selected from hydrogen, alkyl Ca-C8 / carbocycle C3-Ca, aryl, alkylaryl C? -C8, alkyl C? -C8 (C3-C8 carbocycle), C3-C8 heterocycle and CX-C8 alkyl (C3-C8 heterocycle) wherein R5 is selected from H and methyl; or R4 and R5 together have the formula - (CRaRb) n- wherein Ra and R are independently selected from H, C? -C8 alkyl, and C3-C8 carbocycle, and n is selected from 2, 3, 4, 5, and 6? , and form a ring with the carbon atom to which they are attached; Re is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C-C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from an aryl group or C3-C8 heterocycle; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C8 alkyl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (Rls) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R1G is independently H, CX-C8 alkyl, or - (CH2) n -COOH; and n is an integer ranging from 0 to 6. The compounds of Formula (Ib) are useful for the treatment of cancer, an autoimmune disease or an infectious disease in a patient or useful as an intermediary for the synthesis of a drug linker. , a drug-linker-ligand conjugate, and a drug-ligand conjugate that has a drug unit divisible. In another aspect, compositions are provided which include an effective amount of a drug-linker-ligand conjugate and a pharmaceutically acceptable carrier or vehicle.
In yet another aspect, the invention provides pharmaceutical compositions comprising an effective amount of a drug-linker compound and a pharmaceutically acceptable carrier or vehicle. In yet another aspect, the invention provides compositions comprising an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate and a pharmaceutically acceptable carrier or vehicle. In yet another aspect, the invention provides methods for destroying or inhibiting the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for destroying or inhibiting the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-linker-ligand conjugate. In another aspect, the invention provides methods for destroying or inhibiting the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-ligand conjugate having a drug unit. divisible drug-ligand conjugate.
In yet another aspect, the invention provides methods for treating cancer that include administering to a patient in need thereof an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for treating cancer that include administering to a patient in need thereof an effective amount of a drug-linker-ligand conjugate. In yet another aspect, the invention provides methods for treating cancer that include administering to a patient in need thereof an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate. In yet another aspect, the invention provides methods for destroying or inhibiting the replication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-linker compound. In another aspect, the invention provides methods for destroying or inhibiting the replication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-linker-ligand conjugate. In another aspect, the invention provides methods for destroying or inhibiting the replication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-ligand conjugate having a drug unit. divisible drug-ligand conjugate. In yet another aspect, the invention provides methods for treating an autoimmune disease that include administering to a patient in need thereof an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for treating an autoimmune disease that include administering to a patient in need thereof an effective amount of a drug-linker-ligand conjugate. In yet another aspect, the invention provides methods for treating an autoimmune disease that include administering to a patient in need thereof an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate. In yet another aspect, the invention provides methods for treating an infectious disease, including administering to a patient in need thereof an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for treating an infectious disease, including administering to a patient in need thereof an effective amount of a drug-linker-ligand conjugate.
In yet another aspect, the invention provides methods for treating an infectious disease, including administering to a patient in need thereof an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate. In yet another aspect, the invention provides methods for preventing the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for preventing the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-linker-ligand conjugate. In another aspect, the invention provides methods for preventing the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-ligand conjugate having a drug unit divisible by the drug. conjugate drug-1igando. In yet another aspect, the invention provides methods for preventing cancer that include administering to a patient in need thereof an effective amount of a drug-linker compound.
In yet another aspect, the invention provides methods for preventing cancer that include administering to a patient in need thereof an effective amount of a drug-linker-ligand conjugate. In yet another aspect, the invention provides methods for preventing cancer that include administering to a patient in need thereof an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate. In yet another aspect, the invention provides methods for preventing the multiplication of a cell expressing an autoimmune antibody including administration to a patient in need of an effective amount of a drug-linker compound. In another aspect, the invention provides methods for preventing the multiplication of a cell expressing an autoimmune antibody including administration to a patient in need of an effective amount of a drug-linker-ligand conjugate. In another aspect, the invention provides methods for preventing the multiplication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate.
In yet another aspect, the invention provides methods for preventing an autoimmune disease, including administration to a patient in need thereof of an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for preventing an autoimmune disease, including administering to a patient in need thereof an effective amount of a drug-linker-ligand conjugate. In yet another aspect, the invention provides methods for preventing an autoimmune disease that include administration to a patient in need thereof of an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate. In yet another aspect, the invention provides methods for preventing an infectious disease, including administering to a patient in need thereof an effective amount of a drug-linker compound. In yet another aspect, the invention provides methods for preventing an infectious disease, including administering to a patient in need thereof an effective amount of a drug-linker-ligand conjugate. In yet another aspect, the invention provides methods for preventing an infectious disease including administering to a patient in need thereof an effective amount of a drug-ligand conjugate having a drug unit divisible from the drug-ligand conjugate. In another aspect, a drug compound that can be used as an intermediate for the synthesis of a drug-linker compound having a drug unit divisible from the drug-ligand conjugate is provided. In another aspect, a drug-linker compound that can be used as an intermediate for the synthesis of a drug-linker-ligand conjugate is provided. In another aspect, compounds having the formula 'are provided: or a pharmaceutically acceptable salt or solvate thereof, wherein, Ab includes an antibody including one that binds to CD30, CD40, CD70 and Lewis Y antigen, A is a stretch unit, a is 0 or 1, each W is independently an amino acid unit, w is an integer that ranges from 0 to 12, Y is a unit of separation, and y is 0, 1 or 2; p ranges from 1 to about 20; and D is a drug unit selected from the DE and DF Formulas: wherein, independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8), - R4 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-Ca heterocycle and C? Alkyl? -C8 (C3-C8 heterocycle); Rs is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRaR) n- wherein Ra and R are independently selected from H, C? -C8 alkyl, and C3-C8 carbocycle, and n is selected from 2.3. 4, 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-Ca heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and 0- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is O, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C2o alkyl / -aryl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (Rls) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R16 is independently H, C? -C8 alkyl, or - (CH2) n-C00H; R18 is selected from aryl-C (R8) 2-C (R8) 2, -C (R8) 2-C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) 2- (C3-C8 carbocycle); and n is an integer ranging from 0 to 6. In one embodiment, Ab is not an antibody that binds to an ErbB receptor or binds to one or more of the receptors (1) - (35): (1) BMPR1B (morphogenetic bone protein receptor type IB, access number Genbank NM_001203; (2) E16 (LATÍ, SLC7A5, Accession number Genbank NM_003486); (3) STEAP1 (six prostate transmembrane epithelial antigens, Genbank accession number NM_012449); (4) 0772P (CA125, MUC16, Accession number Genbank AF 361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancement factor, mesothelin, Genbank access number NM_005823); (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, family 34 of soluble carrier (sodium phosphate), member 2, sodium-dependent phosphate transporter 3b type II, Accession number Genbank NM__006424); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thromboespondin repeats (type 1 and type 1 similar), transmembrane domain / TM) and short cytoplasmic domain, (semaphorin) 5B. Genbank access number AB040878); (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA gene 2700050C12, Accession number Genbank AY358628; (9) ETBR (endothelin receptor type B, Accession number Genbank AY275 63); (10) MSG783 (RNF124 , hypothetical protein FLJ20315, Accession number Genbank NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, six epithelial antigens prostate transmembrane 2, six transmembrane prostate proteins, accession number Genbank AF455138); (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, potential cation channel of the transient receptor, subfamily M, member 4, access number Genbank NM_017636 ) (13) CRYPT (CR, CR1, CRGF, CRYPT, TDGF1, growth factor derived from teratocarcinoma, access number Genbank NP_003203 or NM_003212); (14) CD21 (CR2 (complement 2 receptor) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792, Accession number Genbank M26004); (15) CD79b (beta immunoglobulin associated), B29, Accession number Genbank NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing anchial phosphatase protein la), SPAP1B, SPAP1C, Accession number Genbank NM_030764); (17) HER2 (Accession number Genbank M11730); (18) NCA (Accession number Genbank M18728); (19) MDP (Accession number Genbank BC017023); (20) IL20Ra; (Accession number Genbank AF184971); (21) Brevican (Genbank access number AF229053); (22) Ephb2R (Accession number Genbank NM_004442); (23) ASLG659 (Accession number Genbank AX092328), - (24) PSCA (Accession number Genbank AJ297436); (25) GEDA (Access number Genbank AY260763); (26) BAFF-R (Accession number Genbank NP_443177.1); (27) CD22 (Accession number Genbank NO-001762.1); (28) CD79a (CD79A, CD79ce, alpha-associated immunoglobulin, a B cell-specific protein that interacts covalently with beta Ig (CD79B) and forms a complex on the surface with IgM molecules, transduces a signal involved in the differentiation of the cell B, access number Genbank NP_001774.1); (29) CXCR5 (Burkitt's lymphoma receptor 1, a G-protein coupled receptor that is activated by chemokine CXCL13, works in lymphocyte migration and in humoral defense, plays a role in HIV-2 infection and once in the development of AIDS, lymphoma, myeloma and leukemia, access number Genbank NP_001707.1); (30) HLA-DOB (beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4 + T lymphocytes, Genbank accession number NP_002111.1); (31) P2X5 (P2X purinergic receptor input ion channel 5, an extracellular ATP input channel, may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of isiopathic detrusor instability, Genbank access NP_002552.2); (32) CD72 (CD72 antigen of B cell differentiation, Lyb-2, Accession number Genbank NP_001773.1); (33) LY64 (lymphocyte antigen 64 (RP105), membrane protein type I of the leucine-rich repeat family (LRR), regulates B cell activation and apoptosis, loss of function is associated with an activity of increased disease in patients with systemic lupus erythomatosis, Genbank accession number NP_005573.1); (34) FCRH1 (protein 1 similar to the Fe receptor, a putative receptor for the immunoglobulin Fe domain containing Ig-like C2-like domains and ITAM, may have a role in lymphocyte differentiation, Genbank accession number NP_443170.1); or (35) IRTA2 (receptor of the immunoglobulin superfamily associated with translocation 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis; deregulation of the gene by translocation occurs in some B cell diseases, Genbank access NP_112571.1). In yet another aspect, the invention provides pharmaceutical compositions comprising an effective amount of a drug-linker-antibody conjugate and a pharmaceutically acceptable carrier or vehicle.
In yet another aspect, the invention provides compositions comprising an effective amount of a drug-antibody conjugate having a drug unit (residue) of the drug-antibody conjugate and a pharmaceutically acceptable carrier or vehicle. In yet another aspect, the invention provides methods for destroying or inhibiting the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-linker-antibody conjugate. In another aspect, the invention provides methods for destroying or inhibiting the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-antibody conjugate having a drug unit. divisible drug-antibody conjugate. In yet another aspect, the invention provides methods for treating cancer that include administering to a patient in need thereof an effective amount of a drug-antibody conjugate. In yet another aspect, the invention provides methods for treating cancer that include administering to a patient in need thereof an effective amount of a drug-antibody conjugate having a drug unit divisible from the drug-antibody conjugate.
In another aspect, the invention provides methods for destroying or inhibiting the replication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-linker-antibody conjugate. In another aspect, the invention provides methods for destroying or inhibiting the replication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-antibody conjugate having a drug unit. divisible drug-antibody conjugate. In yet another aspect, the invention provides methods for treating an autoimmune disease that include administering to a patient in need thereof an effective amount of a drug-linker-antibody conjugate. In yet another aspect, the invention provides methods for treating an autoimmune disease that include administering to a patient in need thereof an effective amount of a drug-antibody conjugate having a drug unit divisible from the drug-antibody conjugate. In yet another aspect, the invention provides methods for treating an infectious disease, including administering to a patient in need thereof an effective amount of a drug-linker-antibody conjugate. In yet another aspect, the invention provides methods for treating an infectious disease that include administering to a patient in need thereof an effective amount of a drug-antibody conjugate having a drug unit divisible from the drug-antibody conjugate. In yet another aspect, the invention provides methods for preventing the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-linker-antibody conjugate. In another aspect, the invention provides methods for preventing the multiplication of a tumor cell or cancer cell including administration to a patient in need thereof, of an effective amount of a drug-antibody conjugate having a drug unit divisible by the drug. drug-antibody conjugate. In yet another aspect, the invention provides methods for preventing cancer that include administering to a patient in need thereof an effective amount of a drug-linker-antibody conjugate. In yet another aspect, the invention provides methods for preventing cancer that include administering to a patient in need thereof an effective amount of a drug-antibody conjugate having a drug unit divisible from the drug-antibody conjugate. In yet another aspect, the invention provides methods for preventing the multiplication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-linker-antibody conjugate. In another aspect, the invention provides methods for preventing the multiplication of a cell expressing an autoimmune antibody including administration to a patient in need thereof, of an effective amount of a drug-antibody conjugate having a drug unit divisible from the drug-antibody conjugate. In yet another aspect, the invention provides methods for preventing an autoimmune disease, including administration to a patient in need thereof of an effective amount of a drug-linker-antibody conjugate. In yet another aspect, the invention provides methods for preventing an autoimmune disease including administering to a patient in need thereof an effective amount of a drug-antibody conjugate having a drug unit divisible from the drug-antibody conjugate. In yet another aspect, the invention provides methods for preventing an infectious disease including administering to a patient in need thereof an effective amount of a drug-linker-antibody conjugate. Still in another aspect, the invention provides methods for preventing an infectious disease, including administering to a patient in need thereof an effective amount of a drog-antibody conjugate having a drug unit divisible from the drug-antibody conjugate. In another aspect, there is provided a drug compound that can be used as an intermediate for the synthesis of a drug-linker compound having a drug unit divisible from the drug-antibody conjugate. In another aspect, a drug-linker compound that can be used as an intermediate for the synthesis of a drug-linker-antibody conjugate is provided. In one aspect, the present invention provides drug-linker-antibody conjugates (also referred to as antibody-drug conjugates) having the formula: or a pharmaceutically acceptable salt or solvate thereof, wherein, Ab is an antibody that binds to one or more of the antigens (1) - (35): (1) BMPR1B (morphogenetic bone protein receptor type IB, Genbank access NM_001203; (2) El6 (LATÍ, SLC7A5, Accession number Genbank NM_003486); (3) STEAP1 (six prostate transmembrane epithelial antigens, Accession number Genbank NM_012449); (4) 0772P (CA125, MUC16, Number Genbank accession AF 361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancement factor, mesothelin, Genbank access number NM_005823); (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, carrier family 34 soluble (sodium phosphate), member 2, type II sodium-dependent phosphate transporter 3b, Genbank accession number NM_006424); (7) Sema 5b (FLJ10372, KI7AA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, domain sema, seven thromboespondin repeats (type 1 and type 1 similar), transmembrane domain / TM) and domi short cytoplasmic child, (semaphorin) 5B. Accession number Genbank AB040878), - (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, gene RIKEN cDNA 2700050C12, Accession number Genbank AY358628; (9) ETBR (endothelin receptor type B, Accession number Genbank AY275463); (10) MSG783 (RNF124, hypothetical protein FLJ20315, Accession number Genbank NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, six transmembrane epithelial prostate 2 antigens, six transmembrane prostate proteins, Accession number Genbank AF455138); (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, potential cation channel of the transient receptor, subfamily M, member 4, access number Genbank NM_017636), - (13) CRYPT (CR, CR1, CRGF, CRYPT., TDGF1, growth factor derived from teratocarcinoma, access number Genbank NP_003203 or NM_003212); (14) CD21 (CR2 (complement 2 receptor) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792, Accession number Genbank M26004); (15) CD79b (beta immunoglobulin associated), B29, Accession number Genbank NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing anchial phosphatase protein la), SPAP1B, SPAP1C, Accession number Genbank NM_030764); (17) HER2 (Accession number Genbank M11730); (18) NCA (Accession number Genbank M18728); (19) MDP (Accession number Genbank BC017023); (20) IL20Ra (Genbank accession number AF184971); (21) Brevican (Genbank access number AF229053); (22) Ephb2R (Accession number Genbank NM_004442); (23) ASLG659 (Accession number Genbank AX092328); (24) PSCA (Accession number Genbank AJ297436); (25) GEDA (Access number Genbank AY260763); (26) BAFF-R (Accession number Genbank NP_443177.1); (27) CD22 (Accession number Genbank NO-001762.1); (28) CD79a (CD79A, CD79Q !, alpha-associated immunoglobulin, a B cell-specific protein that interacts covalently with beta Ig (CD79B) and forms a complex on the surface with IgM molecules, transduces a signal involved in the differentiation of cell B, Genbank access number NP_001774.1); (29) CXCR5 (Burkitt's lymphoma receptor 1, a G-protein coupled receptor that is activated by chemokine CXCL13, works in lymphocyte migration and in humoral defense, plays a role in HIV-2 infection and once in the development of AIDS, lymphoma, myeloma and leukemia, access number Genbank NP_001707.1); (30) HLA-DOB (beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4 + T lymphocytes, Genbank accession number NP_002111.1); (31) P2X5 (P2X purinergic receptor input ion channel 5, an extracellular ATP input channel, may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of isiopathic detrusor instability, Genbank access NP_002552.2); (32) CD72 (CD72 antigen of B cell differentiation, Lyb-2, Accession number Genbank NP_001773.1); (33) LY64 (lymphocyte antigen 64 (RP105), membrane protein type I of the leucine-rich repeat family (LRR), regulates B cell activation and apoptosis, loss of function is associated with an activity of increased disease in patients with systemic lupus erythomatosis, Genbank accession number NP_005573.1); (34) FCRH1 (protein 1 similar to the Fe receptor, a putative receptor for the immunoglobulin Fe domain containing Ig-like C2-like domains and ITAM, may have a role in lymphocyte differentiation, Genbank accession number NP_443170.1); or (35) IRTA2 (receptor of the immunoglobulin superfamily associated with translocation 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis; deregulation of the gene by translocation occurs in some B cell diseases, Genbank access NP_112571.1). A is a stretch unit, a is 0 or 1, each W is independently an amino acid unit, w is an integer that ranges from 0 to 12, Y is a unit of separation, and y is 0, 1 or 2; p ranges from 1 to about 20; and D is a drug residue selected from the DE and DF Formulas: wherein, the wavy line of DE and DF indicates the site of covalent attachment to A, W, or Y, and independently at each location: R2 is selected from H and alkyl C ± -Ce; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle, and C-C8 alkyl (C3-heterocycle) C8); R 4 is selected from H, C 1 -C 3 alkyl, C 3 -C 8 carbocycle, aryl, C 1 -C 8 alkylaryl, C 1 -C 8 alkyl (C 3 -C 8 carbocycle), C 3 -C 3 heterocycle and C 1 -C 8 alkyl (C 3 heterocycle) C8); R5 is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRaRb) n- wherein R a and R b are independently selected from H, C 1 -C 8 alkyl, and C 3 -C 8 carbocycle, and n is selected from 2, 3, 4, 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C? -C8 alkyl); R9 is selected from H and C-C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is O, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C2o alkyl / aryl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (Rld) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R16 is independently H, Cx-Ca alkyl, or - (CH2) n-C00H; R18 is selected from aryl-C (R8) 2-C (R8) 2, -C (R8) 2-C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) 2- (C3-C8 carbocycle); and n is an integer ranging from 0 to 6. In another aspect, the antibody to the antibody-drug conjugate (ADC) of the invention binds specifically to a receptor encoded by an ErbB2 gene. In another aspect, the antibody of the antibody-drug conjugate is a humanized antibody selected from huMAb4D5-l, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7, huMAb4D5-8, (Trastuzumab). In another aspect, the invention includes an article of manufacture comprising, an antibody-drug conjugate compound of the invention; one container; and a packaging insert or label indicating that the compound can be used to treat cancer, characterized by overexpression of an ErbB2 receptor. In another aspect, the invention includes a method for the treatment of cancer in a mammal, wherein the cancer is characterized by overexpression of a receptor.
ErbB2 and that does not respond, or responds poorly to treatment with an anti-ErbB2 antibody, which comprises administering to a mammal a therapeutically effective amount of an antibody-drug conjugate compound of the invention. In another aspect, a substantial amount of the drug residue is not split from the antibody until the antibody-drug conjugate enters the cell with a cell surface receptor specific for the antibody-drug conjugate antibody, and the residue of The drug is divided from the antibody when the antibody-drug conjugate enters the cell. In another aspect, the bioavailability of the antibody-drug conjugated compound or of an intracellular metabolite of the compound in a mammal, improves compared to a drug compound comprising the drug residue of the antibody-drug conjugate compound, or compared to an analogue of the compound that does not have the drug residue. In another aspect, the drug residue is intracellularly divided in a mammal from the antibody of the compound, or from an intracellular metabolite of the compound. In another aspect, the invention includes a pharmaceutical composition comprising an effective amount of the antibody-drug conjugate compound of the invention, or a pharmaceutically acceptable salt thereof., and a pharmaceutically acceptable diluent, carrier or excipient. The composition may further comprise a therapeutically effective amount of a chemotherapeutic agent such as a tubulin-forming inhibitor, a topoisomerase inhibitor and a DNA linker. In another aspect, the invention includes a method for destroying or inhibiting the proliferation of tumor cells or cancer cells comprising treating the tumor cells or cancer cells with an amount of the antibody-drug conjugate compound of the invention, or a salt thereof. or pharmaceutically acceptable solvate thereof, being effective to destroy or inhibit the proliferation of tumor cells or cancer cells. In another aspect, the invention includes a method for inhibiting cell proliferation comprising exposing mammalian cells in a cell culture medium, to an antibody-drug conjugate compound of the invention, wherein the antibody-drug conjugate compound enters to the cells and the drug is divided from the rest of the antibody-drug conjugate compound; whereby the proliferation of the cells is inhibited. In another aspect, the invention includes a method of treating cancer comprising administering to a patient a formulation of an antibody-drug conjugate compound of the invention and a pharmaceutically acceptable diluent, carrier or excipient. In another aspect, the invention includes an assay for the detection of cancer cells, comprising: (a) exposing the cells to an antibody-drug conjugate compound of the invention; and (b) determining the degree of binding of the antibody-drug conjugate compound to the cells. The invention will be better understood by reference to the following detailed description of exemplary embodiments, taken in conjunction with the accompanying drawings, figures and drawings. The following is descriptive, illustrative and exemplary and should not be taken as limiting the scope defined by any of the appended claims. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 shows an in vivo, single dose efficacy analysis of cAClO-mcMMAF in subcutaneous xenografts Karpas-299 ALCL. Figure 2 shows an in vivo, single dose efficacy analysis of cAClO-mcMMAF in subcutaneous L540cy. For this study, there were 4 mice in the untreated group and 10 in each of the treatment groups. Figures 3a and 3b show the in vivo efficacy of cBR96-mcMMAF in subcutaneous L2987. The field of triangles in Figure 3a and the arrows in Figure 3b indicate the days of therapy. Figures 4a and 4b show the in vitro activity of cACLO-antibody-drug conjugates against CD30 + cell lines.
Figures 5a and 5b show the in vitro activity of cBR96-antibody-drug conjugates against Ley + cell lines. Figures 6a and 6b show the in vitro activity of clF6-antibody-drug conjugates against CD70 + cell lines of renal cell carcinoma. Figure 7 shows an in vitro cell proliferation analysis with SK-BR-3 cells treated with antibody-drug conjugates (ADC): - • - Trastuzumab-MC-vc-PAB-MMAF, 3.8 MMAF / Ab, -o- Trastuzumab-MC-MMAF, 4.1 MMAF / Ab, and -? - Trastuzumab-MC-MMAF, 4.8 MMAF / Ab, measured in units of relative fluorescence (RLU) against the concentration in μg / ml of ADC. H = Trastuzumab where H is linked through a cysteine [cys]. Figure 8 shows an in vitro cell proliferation assay with BT-474 cells treated with ADC: - • -Trastuzumab-MC-vc-PAB-MMAF, 3.8 MMAF / Ab, -o- Trastuzumab-MC-MMAF, 4.1 MMAF / Ab, and -? - Trastuzumab-MC-MMAF, 4.8 MMAF / Ab. Figure 9 shows an analysis of cell proliferation in vitro with MCF-7 cells treated with ADC: - • -Trastuzumab-MC-vc-PAB-MMAF, 3.8 MMAF / Ab, -o- Trastuzumab-MC- (N-Me) vc-PAB-MMAF, 3.9 MMAF / Ab, and -? - Trastuzumab-MC-MMAF, 4.1 MMAF / Ab. Figure 10 shows an in vitro cell proliferation analysis with MDA-MB-468 cells treated with ADC: - • - Trastuzumab-MC-vc-PAB-MMAE, 4.1 MMAE / Ab, -o- Trastuzumab-MC-vc-PAB -MMAE, 3.3 MMAE / Ab, and -? - Trastuzumab-MC-vc-PAB-MMAF, 3.7 MMAF / Ab. Figure 11 shows a plasma concentration clearance study after administration of H-MC-vc-PAB-MMAF-TEG and H-MC-vc-PAB-MMAF to Sprague-Dawley rats. The dose administered was 2 mg of ADC per kg of rat. The concentrations of total antibody and ADC were measured over time. (H = Trastuzumab). Figure 12 shows a plasma concentration clearance study after administration of H-MC-vc-MMAE to Cynomolgus monkeys in different doses: 0.5, 1.5, 2.5 and 3.0 mg / kg administered on day 1 and day 21 The total antibody and ADC concentrations were measured over time. (H = Trastuzumab). Figure 13 shows the change in mean tumor volume over time in athymic mice with mammalian tumor allografts MMTV-HER2 Fo5 dosed on day 0 with: vehicle, Trastuzumab-MC-vc-PAB-MMAE (1250 μg / m2 ) and Trastuzumab-MC-vc-PAB-MMAF (555 μg / m2). (H = Trastuzumab). Figure 14 shows the change in mean tumor volume over time in athymic mice with mammalian tumor allografts MMTV-HER2 Fo5 dosed on day 0 with 10 mg / kg (660 μg / m2) of Trastuzumab-MC-MMAE 1250 μg / m2 of Trastuzumab-MC-vc-PAB-MMAE.
Figure 15 shows the change in mean tumor volume over time in athymic mice with mammalian tumor allografts MMTV-HER2 Fo5 dosed on day 0 with vehicle and 650 μg / m2 of Trastuzumab-MC-MMAF. Figure 16 shows the change in mean tumor volume over time in athymic mice with mammalian tumor allografts MMTV-HER2 Fo5 dosed on day 0 with vehicle and 350 μg / m2 of four Trastuzumab-MC-MMAF conjugates where the ratio of MMAF / trastuzumab (H) is 2, 4, 5.9 and 6. Figure 17 shows the average group change, with error bars, in animal body weights (rat) (Mean ± SD) after the vehicle administration, trastuzumab-MC ~ val-cit-MMAF, Trastuzumab-MC (Me) -val-cit-PAB-MMAF, Trastuzumab-MC-MMAF and Trastuzumab-MC-val-cit-PAB-MMAF. Figure 18 shows the average group change in animal (rat) body weights (Mean ± SD) after administration of 9.94 mg / kg of H-MC-vc-MMAF, 24.90 mg / kg of H-MC-vc -MMAF, 10.69 mg / kg of H-MC (Me) -vc-PAB-MMAF, 26.78 mg / kg of H-MC (Me) -vc-PAB-MMAF, 10.17 mg / kg of H-MC-MMAF, 25.50 mg / kg of H-MC-MMAF, and 21.85 mg / kg of H-MC-vc-PAB-MMAF. H = trastuzumab. The MC linker binds through a trastuzumab cysteine for each conjugate. Figure 19 shows the mean group change, with error bars, in body weights of Sprague-Dawley rat (Mean ± SD) after administration of Trastuzumab / H) -MC-MMAF in doses of 2105, 3158, and 4210 μg / m2. The MC linker binds through a cysteine to Trastuzumab for each conjugate. 4. DETAILED DESCRIPTION OF THE MODALITIES EXAMPLES 4. 1 DEFINITIONS AND ABBREVIATIONS Unless otherwise stated, the following terms and phrases as used herein, are intended to have the following meanings: When used in the present trademarks, applicants intend to independently include the product formulation of trademark, the generic drug, and the active pharmaceutical ingredient (s) of the trademark product. The term "antibody" herein is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (eg, bispecific antibodies) formed of at least two intact antibodies, and antibody fragments, while exhibiting the desired biological activity. An antibody is a protein generated by the immune system, capable of recognizing and binding to a specific antigen. Described in terms of its structure, an antibody typically has a Y-shaped protein consisting of four amino acid chains, two heavy and two light. Each antibody has mainly two regions: a variable region and a constant region. The variable region, located at the ends of the arms of the Y, binds to and interacts with the target antigen. The variable region includes a complementarity determining region (CDR) that recognizes and binds to a specific binding site on a particular antigen. The constant region, located on the stem of the Y, is recognized by and interacts with the immune system (Janeway, C, Travers, P., Walport, M., Shlomchik (2001) Immuno Biology, 5th Ed., Garland Publishing, New York). A target antigen generally has numerous binding sites, also called epitopes, recognized by the CDRs on multiple antibodies. Each antibody that binds specifically to a different epitope has a different structure. Therefore, an antigen can have more than one corresponding antibody. The term "antibody" as used herein, also refers to a full length immunoglobulin molecule or an immunologically active portion of a full length immunoglobulin molecule, ie, a molecule that contains an antigen binding site that immunospecifically linked to an antigen of a target of interest or part thereof, such targets include, but are not limited to, cancer cells or cells that produce autoimmune antibodies associated with an autoimmune disease. The immunoglobulin described herein can be of any type, (eg, IgG, IgE, IgM, IgD and IgA), class (eg, IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2), or subclass of immunoglobulin molecule . Immunoglobulins can be derived from any species. In one aspect, however, the immunoglobulin is of human, murine or rabbit origin. In another aspect, the antibodies are polyclonal, monoclonal, bispecific, human, humanized or chimeric antibodies, onocatenary antibodies, Fv, Fab fragments, F (ab ') fragments, F (ab') 2 fragments, fragments produced by an expression library Fab, anti-idiotypic / anti-Id antibodies), CDRs, and epitope binding fragments of any of the foregoing which immunospecifically bind to cancer cell antigens, viral antigens or microbial antigens. The term "monoclonal antibody" as used herein, refers to an antibody obtained from a population of substantially homogeneous antibodies, ie, the individual antibodies comprising the population are identical, except for possible mutations of natural origin that may be present. in smaller quantities. Monoclonal antibodies are highly specific, being directed against a single antigenic site. In addition, in contrast to polyclonal antibody preparations that include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant in the antigen. In addition to its specificity, monoclonal antibodies are advantageous in that they can be synthesized without being contaminated by other antibodies. The "monoclonal" modifier indicates the character of the antibody obtained from a substantially homogeneous population of antibodies, and is not to be understood as requiring the production of the antibody by any particular method. For example, monoclonal antibodies that are used in accordance with the present invention can be made by the hybridoma method first described by Kohier et al., (1975) Nature 256: 495, or can be made by recombinant DNA methods (see, EU No. 4816567). "Monoclonal antibodies" can also be isolated from phage libraries of antibodies using the techniques described, for example, in Clackson et al., (1991) Nature, 352: 624-628 and Marks et al., (1991) J. Mol. Biol. 222: 581-597. Monoclonal antibodies herein specifically include "chimeric" antibodies in which a portion of the heavy and / or light chain is identical or homologous to the corresponding sequences in antibodies derived from particular species or belonging to a particular class or subclass of antibodies , while the rest of the chain (s) is identical or homologous to the corresponding sequences in antibodies derived from other species or belonging to another class or subclass of antibodies, as well as to fragments of such antibodies, as long as they exhibit the desired biological activity (EU Patent m / 4816567; and Morrison et al., (1984) Proc. Nati Acad. Sci. USA 81: 6851-6855). Various methods have been used to produce monoclonal antibodies (MAbs). Hybridoma technology, which refers to a cloned cell line that produces a single type of antibody, uses cells from various species, including mice (murine), hamsters, rats, and humans. Another method to prepare MAbs uses genetic engineering including recombinant DNA techniques. Monoclonal antibodies made from these techniques include, among others, chimeric antibodies and humanized antibodies. A chimeric antibody combines regions that code for DNA from more than one type of species. For example, a chimeric antibody can derive the variable region of a mouse and the constant region of a human. 'A humanized antibody comes predominantly from a human, although it contains non-human portions. As a chimeric antibody, a humanized antibody can contain a completely human constant region. But, unlike a chimeric antibody, the variable region can be derived partially from a human. The non-human synthetic portions of a humanized antibody frequently come from the CDRs in murine antibodies. In any case, these regions are crucial to allow the antibody to recognize and bind to a specific antigen. As noted, murine antibodies can be used. Although they are useful for diagnosis and in short-term therapies, murine antibodies can not be administered to people in the long term without increasing the risk of a harmful immunogenic response. This response, called human anti-mouse antibody (HAMA), occurs when a human immune system recognizes the murine antibody as foreign and attacks it. A HAMA response can cause a toxic shock or even death. Chimeric and humanized antibodies reduce the likelihood of a HAMA response by minimizing the non-human portions of the antibodies administered. In addition, chimeric and humanized antibodies have the additional benefit of activating human secondary immune responses, such as antibody-dependent cellular cytotoxicity. "Antibody fragments" comprise a portion of an intact antibody that preferably comprises its antigen or variable binding region.
Examples of antibody fragments include Fab, Fab 'F (ab') 2 and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragment (s). An "intact" antibody is one that comprises a variable region of antigen binding, as well as a constant domain of light chain (CL) and heavy chain constant domains, CH1, CH2 and CH3. The constant domains may be constant domains of natural sequence (e.g., constant domains of human natural sequence) or a variant of amino acid sequence thereof. The intact antibody may have one or more "effector functions" that refer to those biological activities attributable to the Fe region (a Fe region of natural sequence or an Fe region of amino acid sequence variant) of an antibody. Examples of antibody effector functions include Clq linkage; Complement-dependent cytotoxicity; Fe receptor binding: antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor, BCR), etc. Depending on the amino acid sequence of the constant domain of their heavy chains, the intact antibodies can be assigned to different "classes". There are five major classes of intact antibodies: IgA, IgD, IgE, IgG and IgM, and several of these can further be divided into "subclasses" (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA and IgA2. The heavy chain constant domains corresponding to different classes of antibodies are termed ce, d, e, y, and ß, respectively. Subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known. The terms "ErbB2" and HER2"are used interchangeably herein and refer to the human HER2 protein described, for example, in Semba et al., Proc. Nati. Acad. Sci. USA 82: 6497-6501 ( 1985) and Yamamoto et al., (1986) Nature, 319: 230-234 (Genbank accession number X03363) The term "ErbB2" refers to the gene encoding human ErbB2 and "neu" refers to the gene encoding for rat pl85neu The preferred ErbB2 is the wild-type human ErbB2 Antibodies to ErbB receptors are commercially available from a number of sources, including, for example, Santa Cruz Biotechnology, Inc., California USA By "ErbB ligand" "is meant a polypeptide that binds to and / or activates an ErbB receptor. The ErbB ligand can be a natural sequence human ErbB ligand such as epidermal growth factor (EGF) (Savage et al., (1972) J. Biol. Chem., 247: 7612-7621), transforming alpha growth factor (TGF-c) (Marquardt et al. t al., (1984) Science 223: 1079-1082); amfiregulin also known as schwannoma or keratinocyte growth factor (Shoyab et al., (1989) Science 243: 1074-1076; Kimura et al., Nature, 348: 257-260 (1990); and Cook et al., Mol. Cell Biol., 11: 2547-2557 (1991), betacellulin (Shing et al., Science, 259: 1604-1607 (1993); and Sasada et al., Biochem. Biophys. Res. Commun., 190: 1173 ( 1993)), heparin-binding epidermal growth factor (HB-EGF) (Higashima et al., Science, 251: 936-939 (1991)), epiregulin (Toyoda et al., J. Biol. Chem. 270: 7495-7500 (1995); and Komurasaki et al., Oncogene, 15: 2841-2848 (1997)), a heregulin (see, below), neuregulin-2 (NRG-2) (Carraway et al., Nature, 387 : 512-516 (1997)), neuregulin-3 (NRG-3) (Zhang et al., Proc. Nati, Acad. Sci. 94: 9562-9567 (1997)), - neuregulin-4 (NRG-4) (Harari et al., Oncogene, 18: 2681-89 (1999)) or crypto (CR-1) (Kannan et al., J. Biol. Chem., 272 (6): 3330-3335 (1997)). ErbB ligands that bind to EGFR include EGF, TGF-a, amfiregulin, betacellulin, HB-EGF and epiregulin. ErbB ligands that bind to ErbB3 include heregulins. ErbB ligands capable of binding to ErbB4 include betacellulin, epiregulin, HB-EGF, NRG-2, NRG-3, NRG-4 and heregulins. The ErbB ligand can also be a synthetic ErbB ligand. The synthetic ligand may be specific for a particular ErbB receptor, or it may recognize particular ErbB receptor complexes. An example of a synthetic ligand is the synthetic heregulin / EGF biregulin chimera (see, for example, Jones et al., (1999) FEBS Letters, 447: 227-231, which is incorporated by reference). "Heregulin" (HRG) refers to a polypeptide encoded by the heregulin gene product as described in the U.S. Patent. No. 5641869 or Marchionni et al., Nature, 362: 312-318 (1993). Examples of heregulins include heregulin-o;, heregulin- / 3l, heregulin- / 32, and heregulin-β3 (Holmes et al., Science, 256-1205-1210 (1992), and U.S. Patent No. 5641869); neu differentiation factor (NDF) (Peles et al., Cell, 69: 205-216 (1992)); activity that induces the acetylcholine receptor (ARIA) (Falls et al., (1993) Cell, 72: 801-815); glial growth factors (GGFs) (Marchionni et al., Nature, 362: 312-318 (1993)); sensory and motor neuron-derived factor (SMDF) (Ho et al., J. Biol. Chem., 270: 14523-14532 (1995)); ? -heregulina (Schaefer et al., Oncogene, 15: 1385-1394 (1997)). The term includes biologically active fragments and / or amino acid sequence variants of a natural sequence HRG polypeptide, such as an EGF-like domain fragment thereof (e.g., HRG / 3all77-244). The "ErbB2 hetero-oligomer is a non-covalently associated oligomer comprising at least two different ErbB receptors.A" ErbB dimer "is a non-covalently associated oligomer comprising two different ErbB receptors.Such complexes can be formed when a cell expressing two or more ErbB receptors are exposed to an ErbB ligand ErbB oligomers, such as ErbB dimers, can be isolated by immunoprecipitation and analyzed by SDS-PAGE as described, for example, in Sliwkowski et al., J. Biol. Chem., 269 (20): 14661-14665 (1994) Examples of such ErbB hetero-oligomers include EGFR-ErbB2 complexes (also referred to as (HER1-HER2), ErbB2-ErbB3 (HER2 / HER3) and ErbB3-ErbB4 (HER3 / HER4). In addition, the ErbB hetero-oligomer may comprise two or more ErbB2 receptors combined with a different ErbB receptor, such as ErbB3, ErbB4 or EGFR (ErbBl). Other proteins such as a rec subunit may be included in the hetero-oligomer. eptor cytosine (e.g., gpl30). A "natural sequence" polypeptide is one that has the same amino acid sequence as a polypeptide, e.g., tumor-associated antigen receptor, derived from nature. Such naturally occurring polypeptides can be isolated from nature or can be produced by recombinant or synthetic means. Thus, a naturally occurring polypeptide may have the amino acid sequence of the naturally occurring human polypeptide, murine polypeptide, or polypeptide of any other mammalian species.
The term "amino acid sequence variant" refers to polypeptides having amino acid sequences that differ to some degree from a naturally occurring polypeptide. Ordinarily, the amino acid sequence variants will possess at least about 70% homology with at least one receptor binding domain of a natural ligand, or with at least one ligand binding domain of a natural receptor, such as an associated antigen to the tumor, and preferably, will be at least about 80%, more preferably, at least about 90% homologous with such receptor or ligand binding domains. The amino acid sequence variants possess substitutions, deletions, and / or insertions at certain positions within the amino acid sequence of the natural amino acid sequence. "Sequence identity" is defined as the percentage of residues in the amino acid sequence variant that is identical after aligning the sequences and entering spaces, if necessary, to achieve maximum percent sequence identity. Computer methods and programs for alignment are well known in the art. Such a computer program is "Align 2", owned by Genentech, Inc., which was submitted with user documentation in the United States Copyright Office, Washington D.C., 20559, on December 10, 1991.
"Antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to a cell-mediated reaction in which non-specific cytotoxic cells expressing Fe (FcRs) receptors (eg, natural killer cells (NK), neutrophils) , and macrophages) recognize the bound antibody in a target cell and subsequently cause lysis of the target cell. The primary cells to mediate ADCC, NK cells, express only FcyRIII, while monocytes express FcyRI, FcyRII and FcyRIII. The expression of FcR in hematopoietic cells is summarized in Table 3 on page 464 of Ravetech and Kinet (1991) Annu. Rev. Immunol., 9: 457-92. To assess the ADCC activity of a molecule of interest, an in vitro ADCC assay, such as that described in the U.S. Patent, can be carried out. No. 5500362 or 5821337. Useful effector cells for such analyzes include peripheral blood mononuclear cells (PBMC) and natural killer (NK) cells. Alternatively, or additionally, the ADCC activity of the molecule of interest can be evaluated in vivo, e.g., in an animal model as described in Clynes et al., Proc. Nati Acad. Sci. USA, 95: 652-656 (1998). The terms "Fe receptor" or "FcR" are used to describe a receptor that binds to the Fe region of an antibody. The preferred FcR is a human FcR of natural sequence. In addition, a preferred FcR is one that binds to an IgG antibody (a gamma receptor) and includes receptors of the subclasses FcyRI, FcyRII and FcyRIII, including allelic variants and alternatively spliced forms of these receptors. FcyRII receptors include FcyRIIA (an "activation receptor") and FcyRIIB (an "inhibition receptor"), which have similar amino acid sequences that differ mainly in their cytoplasmic domains. The activation receptor FcyRIIA contains an activation motif based on tyrosine immunoreceptor (ITAM) in its cytoplasmic domain. The inhibition receptor FcyRIIB contains a motif of inhibition based on tyrosine immunoreceptor (ITIM) in its cytoplasmic domain. (See review M in Daéron, Annu, Rev. Immunol., 15: 203-234 (1997)). The FcRs are reviewed in Ravetech and Kinet, Annu. Rev. Immunol. , 9: 457-92 (1991); Capel et al-, Immunomethods, 4: 25-34 (1994); and Haas et al., J. Lab. Clin. Med., 126-330-41 (1995). Other FcRs, including those to be identified in the future, are encompassed by the term "FcR" herein. The term also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus. (Guyer et al., J. Immunol., 117: 587 (1976) and Kim et al., J. Immunol., 24: 249 (1994)). "Complement-dependent cytotoxicity" or "CDC" refers to the ability of a molecule to lyse a target in the presence of a complement.The complement activation pathway is initiated by linking the first component to the complement system (Clq) to a molecule (eg, an antibody) complexed with a cognate antigen To evaluate complement activation, a CDC assay can be carried out, eg, as described in Gazzano-Santoro et al., J. Immunol. Methods, 202: 163 (1996) The term "variable" refers to the fact that certain portions of the variable domains differ widely in sequence between the antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. It is not uniformly distributed throughout the variable domains of antibodies, it is concentrated in three segments called hypervariable regions in the variable domains of both l chain like heavy chain. The most hy conserved portions of the variable domains are called the structure regions (FRs). The variable domains of heavy and l natural chains each comprise four FRs, which mostly adopt a β-sheet configuration, connected by three hypervariable regions, which form circuits that connect and in some cases form part of the β-sheet structure. The hypervariable regions in each chain are held together in close proximity by the FRs and, with the hypervariable regions of the other chain, contribute to the formation of the antigen binding site of the antibodies (see Kabat et al., (1991), Sequences of Proteins of Immunological Interest, 5th Ed., Public Health Service, National Institutes of Health, Bethesda MD). The constant domains are not directly involved in the binding of an antibody to an antigen, but exhibit various effector functions, such as the participation of the antibody in antibody-dependent cellular cytotoxicity (ADCC). The term "hypervariable regions" when used herein, refers to the amino acid residues of an antibody that are responsible for the antigen binding. The hypervariable region generally comprises amino acid residues from a "complementarity determining region" or "CDR" (eg, residues 24-34 (Ll), 50-56 (L2) and 89-97 (L3) in the variable domain of the chain l and 31-35 (Hl), 50.65 (H2) u 95-102 (H3) in the heavy chain variable domain, - Kabat et al., Supra), and / or those residues of a hypervariable circuit "(eg, residues 26-32 (Ll), 50.52 ( L2) and 91-96 (L3) in the variable domain of l chain and 26-32 (Hl), 53-55 (H2), and 96-101 (H3) in the variable domain of heavy chain; Chothia and Lesk ( 1987) J. Mol. Biol. 196: 901-917) The "region of structure" or "FR" residues are those variable domain residues different from the hypervariable region residues as defined herein.
The papain digestion of the antibodies produces two identical fragments of antigen binding, called "Fab" fragments, each with a single antigen binding site, and a residual "Fe" fragment, whose name reflects its ability to crystallize easily. The pepsin treatment produces an F (ab ') 2 fragment that has two antigen binding sites and is still capable of cross-linking the antigen. "Fv" is the minimum antibody fragment that contains a complete site of antigen recognition and antigen binding. This region consists of a variable domain dimer of a heavy chain and a l chain in close non-covalent association. It is in this configuration that the three hypervariable regions of each variable domain interact to define an antigen binding site on the surface of the VH-VL dimer. Collectively, the six hypervariable regions confer an antigen binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three hypervariable regions specific for an antigen) has the ability to recognize and bind the antigen, albeit at a lower affinity than the entire binding site. The Fab fragment also contains the constant domain of the light chain and the first constant domain (CH1) of the heavy chain. The Fab 'fragments differ from the Fab fragments by the addition of some residues at the carboxy terminus of the heavy chain CH1 domain including one or more cysteines from the antibody's articulation region. Fab '-SH is the designation herein for Fab' in which the cysteine residue (s) of the constant domains contain (n) at least one free thiol group. The F (ab ') 2 antibody fragments were originally produced as pairs of Fab' fragments that have articulation cysteines between them. Other chemical couplings of antibody fragments are also known. The "light chains" of antibodies of any vertebrate species can be assigned to one of two clearly distinct types, called Kappa (n) and lambda (?), Based on the amino acid sequences of their constant domains. The "Fv onocatenary" or "scFv" antibody fragments comprise the VH and VL domains of the antibody, wherein these domains are present in a single polypeptide chain. Preferably, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains that allows the scFv to form the desired structure for antigen binding. For a review of scFv, see Plückthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994).
The term "diabodies" refers to small fragments of antibody with two antigen binding sites, whose fragments comprise a variable heavy domain (VH) connected to a variable light domain (VL) in the same polypeptide chain (VH-VL) . When using a linker that is too short to allow pairing between the two domains in the same chain, the domains are forced to pair with the complementary domains of another chain and to create two antigen binding sites. The diabodies are more fully described, for example, in EP 404,097; WO 93/11161; and Hollinger et al., (1993) Proc. Nati Acad. Sci. USA 90: 6444-6448. The "humanized" forms of non-human (e.g., rodent) antibodies are chimeric antibodies that contain a minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (receptor antibody) in which the residues of a hypervariable region of the receptor are replaced by the residues of a hypervariable region of a non-human species (donor antibody) such as of a mouse, rat , rabbit, or non-human primate that have the specificity, affinity, and desired capacity. In some instances, the residues of the structure region (FR) of the human immunoglobulin are replaced by corresponding non-human residues. In addition, the humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine the performance of the antibody. In general, the humanized antibody will substantially comprise all of at least one, and optionally two variable domains, in which all or substantially all of the hypervariable circuits correspond to those of the human immunoglobulin and all or substantially all of the FRs are those of a sequence of human immunoglobulin, the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fe), typically that of a human immunoglobulin. For additional details, see Jones et al., (1986) Nature, 321: 522-525; Riechmann et al., (1988) Nature 332: 323-329; and Presta (1992) Curr Op. Struct. Biol., 2: 593-596. Humanized anti-ErbB2 antibodies include huMAb4D5, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7, and huMAb4D5-8 (HERCEPTIN®) as described in Table 3 of the US Patent No. 5821337 expressly incorporated herein by reference; and humanized antibodies 520C9 (WO 93/21319) and 2C4 as described herein below. An "isolated" antibody is one that has been identified and separated and / or recovered from a component of its natural environment. The contaminating components of its natural environment are materials that interfere with the diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous and nonproteinaceous solubles. In preferred embodiments, the antibody will be purified (1) to greater than 95% by weight of antibody as determined by the Lowry method, and more preferably to more than 99% by weight, (2) to a sufficient degree to obtain at least 15 N-terminal or internal amino acid sequence residues by use of a rotating cell sequencer, or (3) to homogeneity by SDS-PAGE under reducing or non-reducing conditions using Coomassie blue or preferably silver dye. The antibodies isolated include the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, the isolated antibody will be prepared by at least one purification step. An antibody "that binds" to an antigen of interest is one capable of binding to that antigen with sufficient affinity so that the antibody is useful for directing a cell that expresses the antigen. An antibody that "induces apoptosis" is one that induces the programmed death of the cell as determined by the binding of annexin V, DNA fragmentation, cell shrinkage, endoplasmic reticulum dilatation, cell fragmentation and / or formation of membrane vesicles (called apoptotic bodies). The cell is a tumor cell, e.g., breast, ovarian, stomach, endometrial, salivary gland, pulmonary, renal, colon, thyroid, pancreatic or bladder cell. Several methods are available to evaluate cellular events associated with apoptosis. For example, the translocation of phosphatidyl serine (PS) can be measured by binding of annexin; DNA fragmentation can be assessed through staggered DNA; and nucifear / chromatin condensation in conjunction with DNA fragmentation can be assessed by any increase in hypodiploid cells. A "disorder" is any condition that benefits from the treatment of the present invention. This includes chronic or acute disorders or diseases including those pathological conditions that predispose the mammal to the disorder in question. Non-limiting examples of the disorders that are treated herein include benign and malignant tumors; leukemia and lymphoid diseases, in particular breast, ovarian, stomach, endometrial, salivary gland, pulmonary, renal, colon, thyroid, pancreatic, prostate or bladder cancer; neuronal, gual, astrocytal, hypothalamic and other glandular disorders, macrophages, epithelial, stromal and blastocolic disorders; and inflammatory, angiogenic and immunological disorders. The term "therapeutically effective amount" refers to an amount of a drug effective to treat a disease or disorder in a mammal. In the case of cancer, the therapeutically effective amount of the drug can reduce the number of cancer cells; reduce the size of the tumor; inhibit (i.e., retard to some degree and preferably stop) the infiltration of the cancer cell into peripheral organs; inhibit (i.e., retard to some degree and preferably stop) tumor metastasis; inhibit, to some degree, the growth of the tumor; and / or alleviating to some degree one or more of the symptoms associated with cancer. To the extent that the drug can prevent the growth and / or destroy cancer cells, it can be cytostatic and / or cytotoxic. For cancer therapy, efficacy, for example, can be measured by evaluating the time to disease progression (TTP) and / or by determining the response ratio (RR). The term "substantial amount" refers to a majority, i.e., > 50% of a population of a collection or a sample. The term "intracellular metabolite" refers to a compound resulting from a metabolic process or reaction within a cell in an antibody-drug conjugate (7ADC). The metabolic process or reaction may be an enzymatic process such as the proteolytic cleavage of an ADC peptide linker, or the hydrolysis of a functional group such as hydrazone, ester or amide. The intracellular metabolites include, but are not limited to, antibodies and free drugs that have undergone intracellular cleavage after entry, diffusion, absorption or transport in a cell. The terms "divided intracellularly" and "intracellular division" refer to a metabolic process or reaction within a cell in a drug-ligand conjugate, a drug-linker-ligand conjugate, and an antibody-drug conjugate (ADC) or the like by which the covalent binding is broken, eg, the linker, between the drug residue (D) and the antibody (Ab), resulting in the free drug dissociated from the antibody within the cell. The divided residues of the drug-ligand conjugate, drug-linker-ligand conjugate or ADC are, therefore, intracellular metabolites. The term "bioavailability" refers to the systemic availability (i.e., blood / plasma levels) of a given amount of drug administered to a patient. Bioavailability is an absolute term that indicates the measurement of both the time (relationship) and the total amount (degree) of drug that reaches the general circulation of a dosage form administered.
The term "cytotoxic activity" refers to an effect of cellular, cytostatic or antiproliferative destruction of an antibody-drug conjugate compound or an intracellular metabolite of an antibody-drug conjugate compound. The cytotoxic activity can be expressed as the IC50 value which is the concentration (molar or mass) per unit volume at which half of the cells survive. The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancer cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid diseases. More particular examples of such cancers include squamous cell cancer, (eg, squamous cell epithelial cancer), lung cancer, including small cell lung cancer, non-small cell lung cancer, ("NSCLC"), lung adenocarcinoma and carcinoma. flaky lung, peritoneal cancer, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, renal or kidney cancer, prostate cancer, vulvar cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer. A "cancer expressing ErbB2" is one that produces sufficient levels of ErbB2 on the surface of its cells, so that an anti-ErbB2 antibody can bind to it and have a therapeutic effect with respect to cancer. A cancer "characterized by excessive activation" of an ErbB2 receptor is one in which the degree of activation of the ErbB2 receptor in cancer cells significantly exceeds the level of activation of that receptor in non-cancerous cells of the same type of tissue. Such excessive activation may result in overexpression of the ErbB2 receptor and / or higher than normal levels of an ErbB2 ligand available to activate the ErbB2 receptor in the cancer cells. Such excessive activation can cause and / or be caused by the malignant state of a cancer cell. In some embodiments, the cancer will undergo a diagnostic or prognostic analysis to determine if an amplification and / or overexpression of an ErbB2 receptor occurs which results in excessive activation of the ErbB2 receptor. Alternatively, or additionally, the cancer may undergo a diagnostic or prognostic analysis to determine if an amplification and / or overexpression of an ErbB2 ligand occurs in cancer, which is attributed to excessive receptor activation. In a subset of such cancers, excessive activation may result from an autocrine stimulatory pathway. A cancer that "overexpresses" an ErbB2 receptor is one that has significantly higher levels of an ErbB2 receptor on the cell surface thereof, compared to a non-cancerous cell of the same type of tissue. Such overexpression can be caused by the amplification of the gene or by increased transcription or translation. Overexpression of the ErbB2 receptor can be determined in a diagnostic or prognostic analysis by evaluating the increased levels of the ErbB2 protein present on the surface of a cell (e.g., through an immunohistochemical analysis; IHC). Alternatively, or additionally, the levels of nucleic acid encoding ErbB2 in the cell can be measured (eg, through fluorescent in situ hybridization (FISH; see WO 98/45479), southern immunoassay, or polymerase chain reaction techniques ( PCR), such as real-time quantitative PCR (RT-PCR) The overexpression of the ErbB2 ligand can be determined diagnostically by evaluating the levels of the ligand (or the nucleic acid encoding it) in the patient, eg, in a tumor or biopsy. by various diagnostic tests such as IHC, FISH, southern immunoassay, PCR or the in vivo analyzes described above.
Overexpression of the ErbB2 receptor can also be studied by measuring the diffused antigen (eg, extracellular domain ErbB2) in a biological fluid such as serum (see, eg, U.S. Patent No. 4933294; WO 91/05264; U.S. Patent No. 5401638;; and Sias et al., (1990) J. Immunol. Methods, 132: 73-80). In addition to the above analyzes, several other in vivo analyzes are available for the expert practitioner. For example, cells within the patient's body can be exposed to an antibody that is optionally labeled with a detectable label, eg, a radioactive isotope, and the binding of the antibody to cells in the patient can be evaluated, eg, by external screening for radioactivity or analyzing a biopsy taken from a patient previously exposed to the antibody. Tumors that overexpress HER2 are assessed by immunohistochemical labels corresponding to the number of copies of HER2 molecules expressed per cell, and can be determined biochemically: 0 = 0.10,000 copies / cell, 1+ = at least approximately 200,000 copies / cell, 2+ = at least about 500,000 copies / cell, 3+ = about 1-2 x 106 copies / cell.
Overexpression of HER2 at level 3+, which leads to ligand-independent activation of tyrosine kinase (Hudziak et al., (1987) Proc. Nati, Acad. Sci. USA 84: 7159-7163), occurs in approximately 30% of breast cancers, and in these patients, relapse-free survival and decreased Total survival (Slamon et al., Science, 244: 707-712; Slamon et al., (1987) Science, 235: 177-182). Conversely, a cancer that "is not characterized by overexpression of the ErbB2 receptor" is one that, in a diagnostic analysis, does not express higher than normal levels of ErbB2 receptor compared to a non-cancerous cell of the same type of tissue. The term "cytotoxic agent" as used herein, refers to a substance that inhibits or prevents the function of cells and / or causes the destruction of cells. The term is intended to include radioactive isotopes (eg, 211At, 131I, 125I, 90Y, 186Re, 188Re, 153Sm, 212Bi, 32P, 60C, and radioactive isotopes of Lu), chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically toxins active bacteria, fungi, plants, or animals, including synthetic analogs and their derivatives. In one aspect, the term is not intended to include radioactive isotopes. A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include alkylating agents such as thiotepa and CYTOXAN® cyclophosphamide; alkyl sulfonates such as busulfan, improsulfan, and piposulfan; aziridines such as benzodopa, carbocuone, meturedopa, and - uredopa; ethylene imines and methylamelamines including altretamine, triethylene methamine, triethylene phosphoramide, triethylene-thiophosphoramide and trimethylolomelamine; TLK 286 (TELCYTA ™); acetogenins (especially bulatacin and bulatacinone); delta-9-tetrahydrocannabinol (dronabinol, MARINOL®); beta-lapacona; lapacol; colcicins; betulinic acid; a camptothecin (including the synthetic analog topotecan (HYCAMTIN®), CPT-11 (irinotecan, CAMPTOSAR®), acetylcamptothecin, scopolectin, and 9-aminocamptothecin); Bryostatin; Callistatin; CC-1065 (including its synthetic analogs of adozelesin, carzelesin and bizelezin); podophyllotoxin; podophyllinic acid; teniposide; cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodicitin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, colofosfamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembicin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimustine; bisphosphonates, such as clodronate; antibiotics such as enediin antibiotics (e.g., calicheamicin, especially gamma II calicheamicin and omega II calicheamicin (see, e.g., Agnew, Chem.
Intl. Ed. Engl., 33: 183-186 (1994)) and anthracyclines such as anamycin, AD 32, alcarubicin, daunorubicin, dextrazoxane, DX-52-1, epirubicin, GPX-100, isarrubicin, KRN5500, menogaril, dynemycin , including dynemycin A, a esperamycin, neocarzinostatin chromophore and chromophores of chromoprotein-related enediin antibiotics, aclacinomisins, actinomycin, autramycin, azaserin, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycins, dactinomycin, detorrubicin, 6-diazo-5 -oxo-L.norleucine, ADRIAMYCIN® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino doxorubicin, liposomal doxorubicin, and deoxidoxorubicin), esorubicin, marcelomycin, mitomycin such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porphyromycin, puromycin, chelamomycin, rodrububin, streptonigrin , streptozocin, tubercidin, ubenimex, zinostatin, and zorrubicin; folic acid analogues, such as denopterin, pteropterin and trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, iamiprin, and thioguanine; pyrimidine analogues such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocythabin and floxuridine; androgens such as calusterone, dromostathlonone propionate, epithiostanol, mepitiostane, and testolactone; anti-adrenals such as aminoglutethimide, mitotane, and trilostane; folic acid filler such as folinic acid (leucovorin); aceglatone; antineoplastic anti-folate agents such as ALIMTA®, LY231514 pemetrexed, dihydrofolate reductase inhibitors such as methotrexate, anti-metabolites such as 5-fluoroacyl (5-FU) and their prodrugs such as UFT, S-1 and capecitabine, and inhibitors of thymidylate synthase and inhibitors of glycinamide ribonucleotide formyltransferase such as raltitrexed (TOMUDEX ™, TDX); inhibitors of dihydropyrimidine dehydrogenase such as eniluracil; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamin; demecolcine; diazicuone; elfonitina; eliptinium acetate; an epothilone; etoglucide; gallium nitrate; hydroxyurea; lentinan; lonidainin; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; fenamet; pirarubicin; losoxantrone; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triazicuone; 2, 2 ', 2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, varracurin A, roridin A and anguidine); uretan; vindesine (ELDISINA®, FILDESIN®) dacarbazine; manomustine; mitobronitol; mitolactol pipobroman; gacitosina; arabinoside ("Ara-C") cyclophosphamide; thiotepa; taxoids and taxanes, eg, TAXOL® paclitaxel (Bristol-Myers Squibb Oncology, Princeton, NJ), cremophor-free ABRAXANE ™, formulation of nanoparticles made of paclitaxel albumin (American Pharmaceutical Partners, Schaumberg, Illinois) and TAXOTERE®, doxetaxel ( Rhone-Poulenc Rorer, Antony, France); chloranbucil; gemcitabine (GEMZAR®); 6-thioguanine; mercaptopurine, platinum; platinum analogues or platinum-based analogues such as cisplatin, oxaliplatin and carboplatin; vinblastine (VELBAN®) etoposide (VP-16), ifosfamide, mitoxantrone, vincristine (ONCOVIN®), vinca alkaloid, vinorrelbine (NAVELBINE®); novantrone; edatrexate; Daunomycin; aminopterin; xeloda; ibandronate; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; pharmaceutically acceptable salts, acids and derivatives, of any of the foregoing; as well as combinations of two or more of the foregoing such as CHOP, an abbreviation for a combination therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone, and FOLFOX, an abbreviation for a treatment regimen with oxaliplatin (ELOXATIN ™) combined with 5- FU and leucovorin. Also included in this definition are anti-hormonal agents that act to regulate or inhibit hormonal action in tumors such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including tamoxifen NOLVADEX®), raloxifene , droloxifene, 4-hydroxy tamoxifen, troxifene, keoxifene, LY117018, onapristone and toremifene FARESTON®; aromatase inhibitors that inhibit the aromatase enzyme, which regulates the production of estrogen in the adrenal glands, such as, for example, 4 (5) -imidazoles, aminoglutethimide, megestrol acetate MEGASE®, AROMASIN® exemestane, formestanie, fadrozolo, vorozolo RIVISOR®, letrozolo FEMARA®, and anastrozolo ARIMIDEX®; and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those that inhibit the expression of genes in signaling pathways involved in aberrant cell proliferation, such as, for example, PKC-alpha, Raf, H-Ras, and the epidermal growth factor receptor (EGF-). R); vaccines such as vaccines for gene therapy, for example, ALLOVECTIN® vaccine, LEUVECTIN® vaccine, and VAXID® vaccine; PROLEUKIN® rIL-2; LURTOTECAN® topoisomerase inhibitor 1; ABARELIX® rmRH; and pharmaceutically acceptable salts, acids or derivatives of any of the foregoing. As used herein, the term "EGFR-targeted drug" refers to a therapeutic agent that binds to EGFR and, optionally, inhibits EGFR activation. Examples of such agents include antibodies and small molecules that bind to EGFR. Examples of antibodies that bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB 8507), MAb 255 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509), (see EU Patent NO. 4943533, Mendelsohn et al.,) And its variants such as 225 chimerized (C255 or Cetuximab; ERBITUX®) and reconfigured human 225 (H225) (see, WO 96/40210, Imclone Systems Inc.); antibodies that bind mutant EGFR, type II (U.S. Patent No. 5,212,290); humanized and chimeric antibodies that bind to EGFR as described in the U.S. Patent. No. 5891996; and human antibodies that bind to EGFR, such as ABX-EGF (see WO 98/50433, Abgenix). The anti-EGFR antibody can be conjugated with a cytotoxic agent, thus generating an immunoconjugate (see, e.g., EP 659,439 A2, Merck Patent GmbH). Examples of small molecules that bind to EGFR include ZD1839 or Gefitinib (IRESSA ™, Astra Zeneca), Erlotinib HCl (CP-358774, TARCEVA ™, Genentech / OSI) and AG1478, AG1571, (SU 5271; Sugen). A "tyrosine kinase inhibitor" is a molecule that inhibits to some degree the tyrosine kinase activity of a tyrosine kinase such as an ErbB receptor. Examples of such inhibitors include the drugs directed to EGFR noted in the preceding paragraph as well as quinazolines such as PD 153035, 4- (3-chloroanilino) quinazoline, pyridopyrimidines, pyrimidopyrimidines, pyrrolopyrimidines, such as CGP 59326, CGP 60261 and CGP 62706, and pyrazolopyrimidines, 4- (phenylamino-7H-pyrrolo [2,3-d] pyrimidines, curcumin (diferuloyl methane, 4, 5-bis (4-fluoroanilono) phthalimide), trifostins containing nitrophenone residues; RD-0183805 (Warner- Lambert), antisense molecules (eg, those that bind to nucleic acid encoding ErbB), quinoxalines (U.S. Patent No. 5,804,396), trifostins (U.S. Patent No. 5804396); ZD 6474 (Astra Zeneca); PTK-787 (Novartis / Schering AG): pan-ErbB inhibitors such as CI-1033 (Pfizer); Affinitac (ISIS 3521; Isis / Lilly); Imatinib mesylate (Gleevac; Novartis); PKI 166 (Novartis); GW 2016 (Glaxo SmithKline); CI-1033 (Pfizer); EKB-569 (Wyeth); Semaxanib (Sugen); ZD 6474 (Astra Zeneca); PTK-787 (Novartis / Schering AG); INC-1C11 (Imclone); or as described in any of the following patent publications: U.S. Patent. No. 5804396; WO 99/09016 (American Cyanamid); WO 98/43960 (American Cyanamid); WO 97/389383 (Warner Lambert); WO 99/06378 (Warner Lambert); WO 99/06396 (Warner Lambert); WO 96/30347 (Pfizer, Inc.); WO 96/33978 (Zeneca); WO 96/3397 (Zeneca); and WO 96/33980 (Zeneca). An "anti-angiogenic agent" refers to a compound that blocks or interferes to some degree with the development of blood vessels. The anti-angiogenic factor, for example, may be a small molecule or antibody that binds to a growth factor or growth factor receptor involved in promoting angiogenesis. In one embodiment, the anti-angiogenic factor is an antibody that binds to vascular endothelial growth factor (VEGF). The term "cytosine" is a generic term for proteins released by a population of cells that act in another cell as intercellular mediators. Examples of such cytosines are lymphosines, monosines, and traditional polypeptide hormones. Included among the cytosines are growth hormone such as human growth hormone, human growth hormone N-methionyl, and bovine growth hormone; parathyroid hormone; thyroxine; insulin; proinsulin; relaxin, -prorrelaxin; glycoprotein hormones such as follicle stimulating hormone (FSH), thyroid stimulating hormone (TSH), and lutenizing hormone (LH); liver growth factor; fibroblast growth factor; prolactin; placental lactogen; factor a and ß of tumor necrosis; Mullerian inhibitory substance; peptide associated with mouse gonadotropin; inhibin, - activin; Vascular endothelial growth factor; integrin; thrombopoietin (TPO); nerve growth factors such as NGF- / S; platelet growth factor: transforming growth factors (TGFs) such as TGF-a and TGF- / 3; factor I and II of insulin-like growth; erythropoietin (EPO); osteoinductive factors; interferons such as interferon ° "1 ß Y?, colony stimulation factors (CSFs) such as macrophage-CSF (M-CSF); granulocyte-macrophage-CSF (GMCSF); and granulocyte-CSF (G-CSF); interleukins (ILs) such as IL-1, IL-la, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL -11, IL-12, a tumor necrosis factor such as TNF-a !, or TNF- / 3, and other polypeptide factors including LIF and ligand kit (KL) As used herein, the term cytosine includes proteins from natural sources or from recombinant cell culture and biological active equivalents of the natural sequence cytosines The term "prodrug" as used in this application refers to a precursor or derivative form of a pharmaceutically active substance that is less cytotoxic than the tumor cells compared to the original drug and is able to be activated enzymatically or hydrolytically or become a more active original form. ., Wilman, "Prodrugs in Cancer Chemotherapy" Biochemical Society Transactions, 14 pp. 375-382, 615a Metting Belfast (1986) and Stella et al. , "Prodrugs: A Chemical Approach to Targeted Drug Delivery ", Directed Drug Delivery, Borchardt et al., (ed.) pp. 247-267, Humana Press (1985). The prodrugs of this invention include, but are not limited to, prodrugs containing phosphate, prodrugs containing thiophosphate, prodrugs containing sulfate, prodrugs containing peptide, prodrugs modified with D-amino acid, glycosylated prodrugs, prodrugs containing β-lactam, prodrugs containing optionally substituted phenoxyacetamide or prodrugs containing optionally substituted phenylacetamide, 5-fluorocytosine and other 5-fluorouridine prodrugs that can become the most active free cytotoxic drug. Examples of cytotoxic drugs that can be derived in a prodrug form for use in this invention include, but are not limited to, the chemotherapeutic agents described above. A "liposome" is a small vesicle composed of various types of lipids, phospholipids and / or surfactant, which is useful for the delivery of a drug (such as including anti-CD30, CD40, CD70 or Lewis Y antibodies, and optionally a chemotherapeutic agent) to a mammal. The components of the liposome are commonly arranged in a bilayer formation, similar to the lipid arrangement of the biological membranes. The term "package insert" is used to refer to instructions commonly included in commercial packages of therapeutic products, which contain information about the indications, use, dosage, administration, contraindications and / or warnings regarding the use of such therapeutic products. An "isolated" nucleic acid molecule is a nucleic acid molecule that is identified and separated from at least one contaminating nucleic acid molecule with which it is commonly associated in the natural source of the antibody nucleic acid. An isolated nucleic acid molecule is a different one from the form or condition in which it is found in nature. Thus, the isolated nucleic acid molecules are distinguished from the nucleic acid molecule as it exists in natural cells. However, an isolated nucleic acid molecule includes a nucleic acid molecule contained in cells that commonly express the antibody wherein, for example, the nucleic acid molecule is in a chromosomal location different from that of natural cells. The term "control sequences" refers to DNA sequences necessary for the expression of an operably linked coding sequence in a particular host organism. Suitable control sequences for prokaryotes, for example, include a promoter, optionally an operating sequence and a ribosome binding site. It is known that eukaryotic cells use promoters, polyadenylation signals and enhancers. A nucleic acid is "operably linked" when placed in a functional relationship with another nucleic acid sequence. For example, DNA for a presequence or secretion guide is operably linked to DNA for a polypeptide if it is expressed as a preprotein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if placed so as to facilitate translation. Generally, "operably linked" means that the DNA sequences that are linked are contiguous, and, in the case of a secretion guide, contiguous and in reading phase. However, breeders do not have to be contiguous. The link can be achieved by binding at convenient restriction sites. If such sites do not exist, synthetic oligogonucleotide linkers or linkers can be used according to conventional practice. As used herein, the terms "cell", "cell line", and "cell culture" are used interchangeably and all such designations include progeny. Thus, the words "transformers" and "transformed cells" include the primary subject cell and cultures derived therefrom without taking into account the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity explored in the originally transformed cell are included. When different designations are intended, it will be clear from the context. An "autoimmune disease" herein is a disease or disorder that arises from and is directed against an individual's tissues or a co-segregated manifestation thereof or that results from a condition thereof. Examples of autoimmune diseases or disorders include, but are not limited to arthritis (rheumatoid arthritis, juvenile rheumatoid arthritis, osteoarthritis, psoriatic arthritis, and ankylosing spondylitis), psoriasis, dermatitis including atopic dermatitis; chronic idiopathic urticaria, including chronic autoimmune urticaria, polymyositis / dermatomyositis, toxic epidermal necrolysis, systemic scleroma and sclerosis, responses associated with inflammatory bowel disease (IBD) (Crohn's disease, ulcerative colitis), and IBD with co-secreted pyoderma gangrenosum, erythema nodosum, primary sclerosing cholangitis, and / or episcleritis), respiratory distress syndrome, including adult respiratory distress syndrome (ARDS), meningitis, IgE-mediated diseases such as anaphylaxis and allergic rhinitis, encephalitis such as Rasmussen encephalitis, uveitis, colitis such as microscopic colitis and collagenous colitis, glomerunephritis (GN) such as membranous GN, idiopathic membranous GN, proliferative membranous GN (MPGN), including GM Type I and Type II and rapidly progressive, allergic conditions, eczema, asthma, conditions involving infiltration of T cells and chronic inflammatory responses, arteriesclerosis, autoimmune myocarditis, leukocyte adhesion deficiency, systemic lupus erythematosis (SLE) such as cutaneous SLE, lupus (including nephritis, cerebritis, pediatric, non-renal, discoid, alopecia), established juvenile diabetes, multiple sclerosis (MS) such as Spino-optic MS, allergic encephalomyelitis, immune responses associated with acute and delayed hypersensitivity mediated by cytosine and T lymphocytes, tuberculosis, sarcoidosis, granulomatosis including Wegener's granulomatosis, granulocytosis, vasculitis, (including vasculitis of large vessels (including polymyalgia rheumatica and arteritis de giant cell (from Takayasu), middle vessel vasculitis (including Kawasaki disease and polyarteritis nodosa), CNS vasculitis, and ANCA-associated vasculitis, such as Churg-Strauss vasculitis or syndrome (CSS)), aplastic anemia, Coombs positive anemia, Diamond Blackfan anemia, immune haemolytic anemia including autoimmune hemolytic anemia (AIHA), pernicious anemia, pure red cell aplasia (PRCA), Factor VIII deficiency, hemophilia A, autoimmune neutropenia, pancytopenia, leukopenia, diseases involving leukocyte diapedesis, inflammatory CNS disorders, multiple organ damage syndrome, myasthenia gravis, diseases mediated by the antigen-antibody complex, anti-glomerular membrane, anti-phospholipid antibody syndrome, allergic neuritis, Bechet's disease, Castleman's syndrome, Goodpasture's syndrome, Lambert-Eaton myasthenic syndrome, Reynaud's syndrome, Sjorgen's syndrome, Stevens-Johnson syndrome, transplant rejection solid organ (including pretreatment for titrations of high-panel reactive antibody, IgA deposition in tissues, and rejection arising from renal transplantation, liver transplantation, intestinal transplantation, cardiac transplantation, etc.), graft-versus-host disease (GVHD) ), bullous pemphigoid, pemfigus (including pemfigus vulgaris, foliaceus, and mucous membrane) sa pemfingoide), autoimmune polyendocrinopathies, Reiter's disease, rigidity syndrome, immune complex nephritis, polyneuropathies IgM or IgM-mediated neuropathy, idiopathic thrombocytopenic purpura (ITP), thrombotic thrombocytopenic purpura (TTP), thrombocytopenia (developed by patients with infarction to the myocardium, for example), including autoimmune thrombocytopenia, testis and ovarian autoimmune disease including orchitis or autoimmune oophoritis, primary hypothyroidism; autoimmune endocrine diseases including autoimmune thyroiditis, chronic thyroiditis (Hashimoto's thyroiditis), subacute thyroiditis, idiopathic hypothyroidism, Addison's disease, Grave's disease, autoimmune polyglandular syndromes, (or polyglandular endocrinopathy syndromes), Type I diabetes also referred to as diabetes mellitus dependent of insulin (IDDM), including pediatric IDDM, and Sheehan syndrome; autoimmune hepatitis, insterstitial lymphoid pneumonitis (HIV), bronchiolitis obliterans (not transplant), vs NSIP, Guillian-Barré syndrome, Berger's disease (IgA nephropathy), primary biliary cirrhosis, celiab sprue (gluten enteropathy), refractory sprue with dermatitis secreted herpetiformis co, cryoglobulinemia, amilotropic lateral sclerosis (ALS; Lou Gehrig's disease), coronary artery disease, autoimmune inner ear disease (AIED), autoimmune hearing loss, opsoclonus myoclonus syndrome (WHO), polychondritis such as refractory polychondritis, pulmonary alveolar proteinosis, amyloidosis, hepatitis giant cell, scleritis, monoclonal gammopathy of uncertain / unknown significance (MGUS), peripheral neuropathy, paraneoplastic syndrome, channelopathies such as epilepsy, migraine, arrhythmia, muscular disorders, deafness, blindness, periodic paralysis and CNS channelopathies; Autism, inflammatory myopathy, and focal segment glomerulosclerosis (FSGS). "Alkyl" is C? -C? 8 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms. Examples are methyl (Me, -CH3), ethyl (Et, CH2CH3), 1-propyl (n-Pr, n-propyl, -CH2CH2CH3), 2-propyl (i-Pr, i-propyl, -CH (CH3) 2), 1-butyl (n-Bu, n-butyl , CH2CH2CH2CH3), 2-methyl-1-propyl (i-Bu, i-butyl, CH2CH (CH3) 2), 2-butyl (s-Bu, s-butyl, -CH (CH3) CH2CH3), 2-methyl -2-propyl (t-Bu, t-butyl, -C (CH3) 3), 1-pentyl (n-pentyl, -CH2CH2CH2CH2CH3), 2-pentyl (-CH (CH3) CH2CH2CH3), 3-pentyl (- CH (CH 2 CH 3) 2), 2-methyl-2-butyl (-C (CH 3) 2 CH 2 CH 3), 3-methyl-2-butyl (-CH (CH 3) CH (CH 3) 2), 3-methyl-1-butyl (-CH2CH2CH (CH3) 2), 2-methyl-l-butyl (-CH2CH (CH3) CH2CH3), 1-hexyl (-CH2CH2CH2CH2CH2CH3), 2-hexyl (-CH (CH3) CH2CH2CH2CH3), 3-hexyl (-CH (CH2CH3) (CH2CH2CH3)), 2-methyl-2-pentyl (-C (CH3) 2CH2CH2CH3), 3-methyl-2-pentyl (-CH (CH3) CH (CH3) CH2CH3), 4-methyl-2-pentyl (-CH (CH3) CH2CH (CH3) 2), 3-methyl-3-pentyl (-C (CH3) (CH2CH3) 2), 2-methyl-3 -pentyl (-CH (CH2CH3) CH (CH3) 2), 2,3-dimethyl-2-butyl (-C (CH3) 2CH (CH3) 2) , 3,3-dimethyl-2-butyl (-CH (CH3) C (CH3) 3. "Alkenyl" is C? -C18 hydrocarbon containing normal, secondary, tertiary or cyclic carbon atoms with at least one carbon dioxide unsaturation, ie, a carbon-carbon double bond sp2 Examples include, but are not limited to: ethylene or vinyl (-CH = CH2), allyl (-CH2CH = CH2), cyclopentenyl (-C5H7), and 5-hexenyl (- CH2CH2CH2CH2 = CH2) "Alkynyl" is hydrocarbon xC e containing normal, secondary, tertiary or cyclic carbon atoms with at least one unsaturation site, ie, a triple carbon-carbon bond sp. Examples include, but they are not limited to: acetylenic (~ C = CH) and propargyl (-CH2C = CH). "Alkylene" refers to a saturated, branched or straight hydrocarbon radical or cyclic of 1-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different carbon atoms of an alkane of origin. Typical alkylene radicals include, but are not limited to: methylene (-CH2-), 1,2-ethyl (-CH2CH2-), 1,3-propyl (-CH2CH2CH2-), 1,4-butyl (-CH2CH2CH2CH2 -) and the similar. "Alkenylene" refers to an unsaturated, branched chain or straight or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different atoms carbon of an alkene of origin. Typical alkenylene radicals include, but are not limited to: 1,2-ethylene (-CH = CH-). "Alkynylene" refers to an unsaturated, branched or straight or cyclic hydrocarbon radical of 2-18 carbon atoms, and having two monovalent radical centers derived by the removal of two hydrogen atoms from the same or two different atoms carbon of an origin alkyne. Typical alkynylene radicals include, but are not limited to: acetylene (-C = C-), propargyl (-CH2C = C), and 4-pentynyl (-CH2CH2CH2C = CH-). "Aryl" means a monovalent radical of aromatic hydrocarbon of 6-20 carbon atoms derived by the removal of a hydrogen atom from a single carbon atom of an aromatic ring system of origin. Some aryl groups are represented in the exemplary structures as "Ar". Typical aryl groups include, but are not limited to, radicals derived from benzene, substituted benzene, naphthalene, anthracene, biphenyl and the like. "Arylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with an aryl radical. Typical arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, 2-phenyleth-1-yl, naphthylmethyl, 2-naphthyletan-1-yl, 2-naphthyleten-1-yl, naphthobenzyl, 2-naptophenyletan-1-yl and the like. The arylalkyl group comprises 6 to 20 carbon atoms, e.g., the alkenyl residue, including alkanyl, alkenyl or alkynyl groups, the arylalkyl group is 1 to 6 carbon atoms and the aryl residue is from 5 to 14 carbon atoms. "Heteroarylalkyl" refers to an acyclic alkyl radical in which one of the hydrogen atoms bonded to a carbon atom, typically a terminal or sp3 carbon atom, is replaced with a heteroaryl radical. Typical heteroarylalkyl groups include, but are not limited to, 2-benzimidazolylmethyl, 2-furylethyl and the like. The heteroarylalkyl group comprises 6 to 20 carbon atoms, eg, the alkyl residue, including alkanyl, alkenyl or alkynyl groups, of the heteroarylalkyl group is from 1 to 6 carbon atoms and the heteroaryl residue is from 5 to 14 carbon atoms and to 3 heteroatoms selected from N, O, P, and S. The residue of the heteroarylalkyl group can be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms) or a bicyclic having 7 to 10 members of ring (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S), for example: a bicyclo [4,5], [5,5], [5,6], or [ 6,6]. "Substituted alkyl", "substituted aryl" and "substituted arylalkyl" means alkyl, aryl, and arylalkyl respectively, in which one or more hydrogen atoms are each independently replaced with a substituent. Typical substituents include, but are not limited to, -X, -R, -O ", -OR-SR, -S", -NR2, = NR, -CX3, -CN, -OCN, -SCN, -N = C = 0, -NCS, -NO, -N02, = N2, -N3, NC (= 0) R, -C (= 0) NR2, -S03", -S03H, -S (= 0) 2, -OS (= 0) 2OR, -S (= 0) 2NR, S (= 0) R, -OP (= 0) (0R) 2, -P (= 0) 0R) 2, -P0'3, - P03H2, -C (= 0) R, -C (= 0) X, -C (= S) R, -C02R, -C02", -C (= S) 0R, -C (= 0) SR, - C (= S) SR, -C (= 0) NR2, -C (= S) NR2, -C (= NR) NR2, wherein each X is independently a halogen; F, Cl, Br, or í; and each R is independently -H, C2-C18 alkyl, C6-C2o aryl, C3-C1 heterocycle, protective group or prodrug residue. Alkylene, alkenylene and alkynylene groups can also be substituted in a similar manner as described above. "Heteroaryl" and "heterocycle" refer to a ring system in which one or more ring atoms is a heteroatom, e.g., nitrogen, oxygen, and sulfur. The heterocycle radical comprises 1 to 20 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S. A heterocycle may be a monocycle having 3 to 7 ring members (2 to 6 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S) or a bicyclic having 7 to 10 ring members (4 to 9 carbon atoms and 1 to 3 heteroatoms selected from N, O, P and S), for example: a bicyclo system [4,5], [5,5], [5,6], or [6,6]. Heterocycles are described in Paquette, Leo A.,; "Principies of Modern Heterocyclic Chemistry" (W. A. Benjamin, New York, 1968), particularly in chapters 1, 3, 4, 6, 7 and 9; "The Chemistry of Heterocyclic Compounds, A series of Monographs", (John Wiley &Sons, New York, 1950 to the present), in particular volumes 13, 14, 16, 19, and 28; and J. Am. Chem. Soc. (1960) 82: 5566. Examples of heterocycles include, by way of example and not limitation, pyridyl, dihydropyridyl, tetrahydropyridyl (piperidyl), thiazolyl, tetrahydrothiophenyl, tetrahydrothiophenyl, oxidized with sulfur, pyrimidinyl, furanyl, thienyl, pyrrolyl, pyrazolyl, imidazolyl, tetrazolyl, benzofuranyl, tianaphthalenyl, indolyl, indolenyl, quinolinyl, isoquinolinyl, benzaimidazolyl, piperidinyl, 4-piperidonyl, pyrrolidinyl, 2-pyrrolidodonyl, pyrrolinyl, tetrahydrofuranyl, bis-tetrahydrofuranyl, tetrahydropyranyl, bis-tetrahydropyranyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, octahydroisoquinolinyl, azocinyl, triazinyl, 6H-1, 2,5-thiadiazinyl, 2H, 6H-1, 5, 2-dithiazinyl, thienyl, thianthrenyl, pyranyl, isobenzofuranyl, chromenyl, xanthenyl, phenoxatinyl, 2H-pyrrolyl, isothiazolyl, isoxazolyl, pyrazinyl, pyridazinyl, indolizinyl, isoindolyl, 3H- indolyl, lH-indazolyl, purinyl, 4H-quinolizinyl, phthalazinyl, naphthyridinyl, quinoxalini it, quinazolinyl, cinolinyl, pteridinyl, 4aH-carbazolyl, carbazolyl, / 3-carbolinyl, phenanthridinyl, acridinyl, pyrimidinyl, phenanthroline, phenanzinyl, phenothiazinyl, furazanyl, phenoxazinyl, isochromanyl, chromanyl, imidazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperazinyl, indolinyl. , isoindolinyl, quinuclidinyl, morpholinyl, oxazolidinyl, benzotriazolyl, benzisoxazolyl, oxindolyl, benzoxazolinyl, and isatinoyl. By way of example and not limitation, the carbon-bound heterocycles are linked in the 2, 3, 4, 5 or 6 position of a pyridine, in the 3, 4, 5 or 6 position of a pyridazine, in the 2-position. , 4, 5 or 6 of a pyrimidine, in position 2, 3, 5 or 6 of a pyrazine, in position 2, 3, 4 or 5 of a furan, tetrahydrofuran, thiofuran, thiophene, pyrrolo, or tetrahydropyrrolo, in position 2, 4, or 5 of an oxazolo, imidazolo or thiazolo, in position 3, 4, or 5 of an isoxazolo, pyrazolo, or isothiazolo, in the 2 or 3 position of an aziridine, in position 2, 3 or 4 of an azetidine, in the 2, 3, 4, 5, 6, 7, or 8 position of a quinoline or in the 1, 3 position, 4, 5, 6, 7 or 8 of an isoquinoline. Even more typically, carbon-linked heterocycles include, 2-pyridyl, 3-pyridyl, 4-pyridyl, 5-pyridyl, 6-pyridyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2-pyrimidinyl. , 4-pyrimidinyl, 5-pyrimidinyl, 6-pyrimidinyl, 2-pyrazinyl, 3-pyrazinyl, 5-pyrazinyl, 6-pyrazinyl, 2-thiazolyl, 4-thiazolyl, or 5-thiazolyl. By way of example, and not as limitation, the nitrogen-linked heterocycles are linked at position 1 of an aziridine, azetidine, pyrrolo, pyrrolidine, 2-pyrroline, 3-pyrroline, imidazole, imidazolidine, 2-imidazoline, 3-imidazoline , pyrazolo, pyrazoline, 2-pyrazoline, 3-pyrazoline, piperidine, piperazine, indole, indoline, IH-indazolo, in position 2 of an isoindol, or isoindoline, in position 4 of a morphillin, and in position 9 of a carbazolo, or / 3-carboline. Even more typically, nitrogen-linked heterocycles include 1-aziridyl, 1-azetidyl, 1-pyrrolyl, 1-imidazolyl, 1-pyrazolyl and 1-piperidinyl. "Carbocycle" means a saturated or unsaturated ring having from 3 to 7 carbon atoms as a monocycle or from 7 to 12 carbon atoms as a bicyclo. Monocyclic carbocycles have from 3 to 6 ring atoms, even more typically 5 or 6 ring atoms. The bicyclic carbocycles have from 7 to 12 ring atoms, e.g., arranged as a bicyclo system [4,5], [5,5], [5,6], or [6,6], or 9 or 10 ring atoms arranged as a bicyclo system [5.6] or [6,6]. Examples of monocyclic carbocycles include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex- 2-enyl, 1-cyclohex-3-enyl, cycloheptyl, and cyclooctyl. "Linker", "linker unit", or "link" means a chemical residue comprising a covalent bond or a chain of atoms that covalently binds an antibody to a drug residue. In various modalities, a linker is specified as LU. The linkers include a divalent radical such as an alkyldiyl, an aryldiyl, a heteroaryldiyl residue, such as: - (CR 2) n 0 (CR 2) n-, repeat units of alkyloxy (eg, polyethyleneoxy, PEG, polymethyleneoxy) and alkylamino (eg , polyethyleneamine, Jeffamine ™), - and diacid esters and amides including succinate, succinamide, diglycolate, malonate and caproamide. The term "chiral" refers to molecules that have the property of non-superimposability of the mirror image partner, while the term "achiral" refers to molecules that are superimposable in their mirror image partner. The term "stereoisomers" refers to compounds that have an identical chemical constitution, but differ with respect to the arrangement of atoms or groups in space. "Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g., melting points, boiling points, spectral properties, and reactivities. Mixtures of diastereomers can be separated under high resolution analytical procedures such as electrophoresis and chromatography. "Enantiomers" refer to two stereoisomers of a compound that are mirror images not superimposable to one another. The stereochemical definitions and conventions generally used herein follow S.P. Parker, Ed., McGraw Hill Dictionary of Chemical Terms (1984) McGraw Hill Book Company, New York; and Eliel E. and Wilen, S. Stereochemistry of Organic Compounds (1994) John Wiley & Sons, Inc., New York. There are many organic compounds in optically active forms, i.e., they have the ability to rotate the flat plane of polarized light. When describing an optically active compound, the prefixes D and L, or R and S are used to denote the absolute configuration of the molecule around its chiral center (s). The prefixes d and 1 or (+) and (-) are used to designate the sign of rotation of plane polarized light by the compound, meaning (-) or 1 that the compound is levogyrator. A compound with the prefix (+) or d is dextrogiratorio. For a given chemical structure, these stereoisomers are identical except that they are mirror images of each other. A specific stereoisomer may also be referred to as an enantiomer, and a mixture of such isomers is often referred to as an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or racemate, which may occur when there has been no stereoselection or stereospecificity in a chemical reaction or process. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomeric species, devoid of optical activity. Examples of a "patient" include, but are not limited to, a human, a rat, a mouse, a guinea pig, a monkey, a pig, a goat, a cow, a horse, a dog, a cat, a bird, and farm birds. In an exemplary embodiment, the patient is a human. "Aryl" refers to a carbocyclic aromatic group. Examples of aryl groups include, but are not limited to, phenyl, naphthyl and anthracenyl. A carbocyclic aromatic group or a heterocyclic aromatic group can be unsubstituted or substituted with one or more groups including, but not limited to, C?-C8 alkyl, -O- (C?-C8 alkyl), aryl, -C (0) ) R ', -OC (0) R'-, -C (0) R'-, -C (0) NH 2, -C (0) NHR', -C (0) N (R ') 2-, -NHC (0) R'-, -S (0) 2R'-, -S (0) R'-, -OH, halogen, -N3, -NH2, -NH (R '), -N (R' ) 2 and -CN; wherein each R 'is independently selected from H, C? -C8 alkyl, and aryl. The term "C?-C8 alkyl" as used herein refers to a straight or branched chain saturated or unsaturated hydrocarbon having from 1 to 8 carbon atoms. Representative "C alquilo-C8 alkyl" groups include, but are not limited to, -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, -n-hexyl, -n-heptyl, n-octyl, -n-nonyl and -n-decyl; while branched C?-C8 alkyls include, but are not limited to, -isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, 2-methylbutyl, unsaturated C?-C8 alkyls include, but are not limited to -vinyl, -alyl, -1-butenyl, -2-butenyl, -isobutenyl, -1-pentenyl, -2-pentenyl, -3-methyl-1-butenyl, -2-methyl-2-butenyl , -2,3-dimethyl-2-butenyl, 1-hexyl, 2-hexyl, 3-hexyl, acetylenyl, -propynyl, -1-butynyl, -2-butynyl, -1-pentynyl, -2-pentynyl, - 3-methyl-l-butynyl, methyl, ethyl, propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tertbutyl, n-pentyl, isopentyl, neopentyl, n-hexyl, isohexyl, 2-methylphenyl, 3-methylphenyl, 2, 2-dimethylbutyl, 2,3-dimethylbutyl, 2,2-dimethylpentyl, 2,3-dimethyl penyl, 3,3-dimethylpentyl, 2,3-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? -C8 alkyl group may be unsubstituted or substituted with one or more groups including, but not limited to, C? -C8 alkyl, -O- (C? -C8 alkyl), aryl, -C (0) R ', -OC (0) R'-, -C (0) R'-, -C (0) NH 2, -C (0) NHR', -C (O) N (R ') 2-, -NHC (0) R'-, -S (0) 2R'-, -S (0) R'-, -OH, halogen, -N3, -NH2, -NH (R '), -N (R') 2 and -CN; wherein each R 'is independently selected from H, C? -C8 alkyl, and aryl. A "C3-C8 carbocycle" is a saturated or unsaturated, 3-, 4-, 5-, 6-, 7-, or 8-member non-aromatic carbocyclic ring. Representative C3-C8 carbocycles include, but are not limited to, -cyclopropyl, -cyclobutyl, cyclopentyl, -cyclopentadienyl, cyclohexyl, cyclopentyl, 1,3-cyclohexadienyl, -1, 4-cyclohexadienyl, -cyticheptyl, -1, 3-cycloheptadienyl, -1,3,5-cycloheptatrienyl, -cyclooctyl, and -cyclooctadienyl. A C3-C8 carbocycle group may be unsubstituted or substituted with one or more groups including, but not limited to, C?-C8 alkyl, -0- (C?-C8 alkyl), aryl, -C (0) R ' , -0C (0) R'-, -C (0) R'-, -C (0) NH2, -C (0) NHR ', -C (O) N (R') 2-, -NHC ( 0) R'-, -S (0) 2R'-, -S (0) R'-, -OH, halogen, -N3, -NH2, -NH (R '), -N (R') 2 and -CN; wherein each R 'is independently selected from H, C? -C8 alkyl, and aryl. A "C3-C8 carbocycle" refers to a C3-C8 carbocycle group defined above wherein one of the hydrogen atoms of the carbocycle groups is replaced with a bond. A "C? -C10 alkylene" is a saturated straight-chain hydrocarbon group of the formula - (CH2) _? 0-. Examples of a C? -C? 0 alkylene include methylene, ethylene, propylene, butylene, pentylene, hexylene, heptylene, octylene, nonylene, and decalene. An "arylene" is an aryl group that has two covalent bonds and can be found in the ortho, meta or para configurations as shown in the following structures: - in which the phenyl group can be unsubstituted or substituted with up to four groups including, but not limited to C-C8 alkyl, -O- (C? -C8 alkyl), aryl, -C (0) R ', -0C (0) R'-, -C (0) R'-, -C (0) NH 2, -C (0) NHR ', -C (O) N (R') 2-, NHC (0) R'-, -S (0) 2R'-, -S (0) R'-, -OH, halogen, -N3, -NH2, -NH (R '), -N (R') 2 and -CN; wherein each R 'is independently selected from H, C? -C8 alkyl, and aryl. A "C3-C8 heterocycle" refers to an aromatic or non-aromatic C3-C8 carbocycle in which one to four ring carbon atoms are independently replaced with a heteroatom of the group consisting of O, S, and N. The examples Representative of C3-C8 heterocycle include, but are not limited to, benzofuranyl, benzothiophene, indolyl, benzopyrazolyl, coumarinyl, isoquinolinyl, pyrrolyl, thiophenyl, furanyl, thiazolyl, imidazolyl, pyrazolyl, triazolyl, quinolinyl, pyrimidinyl, pyridinyl, pyridonyl, pyrazinyl, pyridazinyl, isothiazolyl, isoxazolyl, and tetrazolyl. A C3-C8 heterocycle can be unsubstituted or substituted with up to seven groups including, but not limited to, C-C8 alkyl, -O- (C-C8 alkyl), aryl, -C (0) R ', -0C ( 0) R'-, -C (0) R'-, -C (0) NH 2, -C (0) NHR ', -C (0) N (R') 2-, -NHC (0) R ' -, -S (0) 2R'-, -S (0) R'-, -OH, halogen, -N 3, -NH 2, -NH (R '), -N (R') 2 and -CN; wherein each R 'is independently selected from H, C? -C8 alkyl, and aryl. "C3-C8 heterocycle" refers to a C3-C8 heterocycle group defined above wherein one of the hydrogen atoms of the heterocycle groups is replaced with a bond. A C3-C8 heterocycle can be unsubstituted or substituted with up to six groups including, but not limited to, C -C8 alkyl, -O- (C? -C8 alkyl), aryl, C (0) R ', -0C ( 0) R'-, -C (0) R'-, -C (0) NH 2, -C (0) NHR ', -C (0) N (R') 2-, -NHC (0) R ' -, -S (0) 2R'-, -S (0) R'-, -OH, halogen, -N 3, -NH 2, -NH (R '), -N (R') 2 and -CN; wherein each R 'is independently selected from H, C? -C8 alkyl, and aryl. An "exemplary compound" is a drug compound or a drug-linker compound. An "exemplary conjugate" is a drug-ligand conjugate that has a drug unit divisible from the drug-ligand conjugate or a drug-linker-ligand conjugate. In some embodiments, exemplary compounds and exemplary conjugates are found in isolated or purified form. As used herein, "isolated" means separate from other components of (a) a natural source, such as a plant or animal cell or cell culture, or (b) a synthetic organic chemical reaction mixture. As used herein "purified" means that upon isolation, the isolate contains at least 95%, and in another aspect at least 98% exemplary conjugate or exemplary conjugate by weight of the isolate. Examples of a "hydroxyl protecting group" include, but are not limited to, methoxymethyl ether, 2-methoxyethoxymethyl ether, tetrahydropyranyl ether, benzyl ether, p-methoxybenzyl ether, trimethylsilyl ether, triethylsilyl ether, triisopropyl silyl ether, t-butyldimethyl silyl ether. , triphenylmethyl silyl ether, acetate ester, substituted acetate esters, pivaloate, benzoate, methanesulfonate and p-toluenesulfonate. "Omission group" refers to a functional group that can be replaced by another functional group. Such omission groups are well known in the art, and examples include, but are not limited to, a halide (eg, chloride, bromide, iodide), methanesulfonyl (mesyl), p-toluensolfonyl (tosyl), trifluoromethylsulfonyl (triflate) and trifluoromethylsulfonate. The phrase "pharmaceutically acceptable salt" as used herein, refers to pharmaceutically acceptable organic or inorganic salts of an exemplary compound or exemplary conjugate. Exemplary exemplary and conjugated compounds contain at least one amino group, and thus acid addition salts with this amino group can be formed. Exemplary salts include, but are not limited to, sulfate, citrate, acetate, oxalate, chloride, bromide, iodide, nitrate, bisulfate, phosphate, acid phosphate, isonicotinate, lactate, salicylate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gantisinate, fumarate, gluconate, glucuronate, saccharate, formate, benzoate, glutamate, matanosulfonate, ethanesulfonate , benzenesulfonate, p-toluenesulfonate, and pamoate salts (ie, 1,1 '-methylene-bis- (2-hydroxy-3-naphthoate)). A pharmaceutically acceptable salt can involve the inclusion of another molecule such as an acetate ion, a succinate ion or another counterion. The counterion can be any organic or inorganic residue that stabilizes the charge in the parent compound. In addition, a pharmaceutically acceptable salt can have more than one charged atom in its structure. Examples where multiple charged atoms are part of the pharmaceutically acceptable salt can have multiple counterions. Thus, a pharmaceutically acceptable salt can have one or more charged atoms and / or one or more counterions. "Pharmaceutically acceptable solvate" or "solvate" refers to an association of one or more solvent molecules and a compound of the invention, e.g., an exemplary compound or an exemplary conjugate. Examples of solvents forming pharmaceutically acceptable solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The following abbreviations are used herein and have the stated definitions: AE is auristatin E, Boc is N- (t-butoxycarbonyl), cit is citrulline, dap is dolaproine, DCC is 1,3-dicyclohexylcarbodiimide, DCM is dichloromethane, SEA is diethylamine, DEAD is diethylazodicarboxylate, DEPC is diethylphosphorylcyanate, DIAD is diisopropylazodicarboxylate, DIEA is N, N-diisopropylethylamine, dil is dolaisoleucine, DMAP is 4-dimethylaminopyridine, DME is ethylene glycol dimethyl ether (or 1,2-dimethoxyethane), DMF is N , N-dimethylformamide, DMSO is dimethisulfoxide, doe is dolafenin, dov is N, N-dimethylvaline, DTNB is 5, 5-dithiobis (2-nitrobenzoic acid), DTPA is diethylenetria inapentaacetic acid, DTT is dithiothreitol, EDCl is hydrochloride 1 - (3-dimethylaminopropyl) -3-ethylcarbodiimide, EEDQ is 2-ethoxy-1-ethoxycarbonyl-1,2-dihydroquinoline, ES-MS is electroaspersion mass spectrometry, EtOAc is ethyl acetate, Fmoc is N- (9-fluorenylmethoxycarbonyl ), gly is glycine, HATU is O- (7-azabenzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium hexafluorophosphate, HOBt is 1-hydroxybenzotriazolo, HPLC is liquid chromatography at high pressure, ile is isoleucine, lys is lysine, MeCN (CH3CN ) is acetonitrile, MeOH is methanol, Mtr is 4-anisyldiphenylmethyl (or 4-methoxytrityl), ñor is (ÍS, 2R) - (+) - norephedrine, PAB is p-aminobenzyl, PBS is buffered saline in phosphate (pH 7.4) , PEG is polyethylene glycol, Ph is phenyl, Pnp is p-nitrophenyl, MC is 6-maleimidocaproyl, phe is L-phenylalanine, PyBrop is bromo tris-pyrrolodino phosphonium hexafluorophosphate, SEC is size exclusion chromatography, Su is succinimide, TBTU is O-benzotriazol-1-yl-N, N, N, N-tetramethyluronium tetrafluoroborate, TFA is trifluoroacetic acid, TLC is thin layer chromatography, UV is ultraviolet and val is valine. The following linker abbreviations are used herein and have the indicated definitions: Val Cit is valine-citrulline, dipeptide site in divisible protease linker; PAB is p-aminobenzylcarbamoyl; (Me) ve is N-methyl-valine citrulline, where the binding of the linker peptide has been modified to prevent its division by cathepsin B; MC (PEG) 6-OH is maleimidocaproyl-polyethylene glycol; SPP is N-succinimidyl-4- (2-pyridylthio) pentanoate; and SMCC is N-succinimidyl-4- (N-maleimidomethyl) cyclohexane-1 carboxylate. The terms "treat" or "treatment" unless otherwise indicated by the context, refer to both therapeutic treatment and prophylactic or preventive measures, where the goal is to prevent or delay (decrease) an unwanted physiological change or disorder, such as the development or spread of cancer. For the purposes of this invention, the desired beneficial or clinical outcomes include, but are not limited to, relief of symptoms, decrease in the extent of the disease, stable (ie, not impaired) state of the disease, delay or delay of the disease. progress of the disease, improvement or palliation of the disease state, and remission (either partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival compared to the expected survival if treatment is not received. Those in need of treatment include those already with the condition or disorder as well as those prone to having the condition or disorder or those in which the condition or disorder is to be prevented. In the context of cancer, the term "treating" includes any or all of: prevention of the growth of tumor cells, cancer cells, or of a tumor; prevention of replication of tumor cells or cancer cells, decrease in total tumor burden or decrease in the number of cancer cells, and improvement of one or more symptoms associated with the disease. In this context of an autoimmune disease, the term "treating" includes any or all of: prevention of replication of cells associated with an autoimmune disease state including, but not limited to, cells that produce an autoimmune antibody, decrease in load of the autoimmune antibody, and improvement of one or more symptoms of an autoimmune disease. In the context of an infectious disease, the term "treating" includes any or all of: prevention of growth, multiplication or replication of the pathogen that causes the infectious disease, and improvement of one or more associated symptoms of an infectious disease. The following abbreviations of cytotoxic drugs are used herein and have the stated definitions: MMAE is mono-methyl auristatin E (MW 718); MMAF is N-methylvaline-valine-dolaisoleucine-dolaproine-phenylalanine (MW 731. 5); MMAF-DMAEA is MMAF with DMAEA (dimethylaminoethylamine) in an amide bond to the C-terminating phenylalanine (MW 801. 5); MMAF-TEG is MMAF with tetraethylene glycol esterified to phenylalanine; MMAF-NtBu is N-t-butyl, linked as an amide to the C-terminus of MMAF: AEVB is auristatin R-valeryl benzylhydrazone, acid-labile linker through the C-terminus of AE (MW 732); and AFP is p-phenylene diamine monoamide with C-termination phenylalanine of auristatin F (MW 732). 4.2 COMPOUNDS OF THE INVENTION 4.2.1 COMPOUNDS OF THE FORMULA (la) In one aspect, the invention provides drug-linker-ligand conjugates having the formula: or a pharmaceutically acceptable salt or solvate thereof, wherein L is a ligand unit; -Aa-Ww-Yy is a linker unit (LU), where the linker unit includes: -A- is a stretch unit, a is 0 or 1, each -W- is independently an amino acid unit, w is an integer that fluctuates from 0 to 12, -Y- is a unit of separation, and y is 0, 1 or 2; p ranges from 1 to about 20; and -D is a drug unit that has the DE and DF Formulas: wherein, independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C-C8 alkyl (C3-heterocycle) C8), - R4 is selected from H, C-C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3-C8 heterocycle), - Rs is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRaRb) n- wherein R a and R b are independently selected from H, C 1 -C 8 alkyl, and C 3 -C 8 carbocycle, and n is selected from 2, 3, 4, 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C-C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) C8); each R8 is independently selected from H, OH, C-C8 alkyl, C3-C8 carbocycle, and 0- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is O, S, NH, or NR, 1A2 wherein? Is C? -C8 alkyl; R is selected from H, C-C8 alkyl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (Rls) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R 14 is H or C 1 -C 8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of Rls is independently H, C? -C8 alkyl, or - (CH2) n -COOH; R18 is selected from aryl -C (R8) 2-C (R8) 2, -C (R8) 2-C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) 2- (carbocycle C3-CB), and n is an integer that ranges from 0 to 6. In another embodiment, the present invention provides drug compounds having the Formula Ib: Ib or its salts or pharmaceutically acceptable salts or solvates thereof, wherein; R2 is selected from hydrogen and C-C8 alkyl; RJ is selected from hydrogen, C? -C8 alkoyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); R4 is selected from hydrogen, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3-C8 heterocycle) wherein R5 is selected from H and methyl; or R4 and R5 together have the formula - (CRaRb) n- wherein Ra and Rb are independently selected from H, C? -C8 alkyl, and C3-C8 carbocycle, and n is selected from 2, 3, 4, 5, and 6? , and form a ring with the carbon atom to which they are attached; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from an aryl group or C3-C8 heterocycle; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C8 alkyl, C3-C8 heterocycle, - (Ra30) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R1S is independently H, C? -C8 alkyl, or - (CH2) n-C00H; and n is an integer ranging from 0 to 6. In yet another embodiment, the invention provides drug-linker-ligand conjugates having the formula: Formula the ' or a pharmaceutically acceptable salt or solvate thereof, wherein, Ab is an antibody, A is a stretch unit, a is 0 or 1, each W is independently an amino acid unit, w is an integer ranging from 0 to 12 , Y is a unit of separation, and y is 0, 1 or 2; p ranges from 1 to about 20; and D is a drug unit selected from the DE and DF Formulas: - wherein, independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl- (heterocycle) C3-C8), - R4 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle, and C-alkyl ? -C8 (C3-C8 heterocycle); R5 is selected from H and methyl; or R4 and Rs together form a carbocyclic ring and have the formula - (CRaRb) n- wherein R a and R b are independently selected from H, C 1 -C 8 alkyl, and C 3 -C 8 carbocycle, and n is selected from 2, 3, 4, 5, and 6; Rd is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C2o alkyl / aryl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 3 alkyl; R 14 is H or C 1 -C 8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (Rls) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R16 is independently H, C? -C8 alkyl, or - (CH2) n -COOH; R18 is selected from aryl-C (R8) 2-C (R8) 2, -C (R8) 2-C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) 2- (C3-C8 carbocycle); and n is an integer that ranges from 0 to 6. Ab is any antibody covalently linked to one or more drug units. Ab includes an antibody that binds to the antigen CD30, CD40, CD70, Lewis Y. In another embodiment, Ab does not include an antibody that binds to an ErbB receptor or to one or more of the receptors (1) - (35): - (1) BMPR1B (morphogenetic bone protein receptor type IB, Accession number Genbank NM_001203; (2) E16 (LATÍ, SLC7A5, Accession number Genbank NM_003486); (3) STEAP1 (six prostate transmembrane epithelial antigens, Genbank access NM_012449); (4) 0772P (CA125, MUC16, Accession number Genbank AF 361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancement factor, mesothelin, Genbank access number NM_005823); ) Napi3b (NAPI-3B, NPTIIb, SLC34A2, family 34 of soluble carrier (sodium phosphate), member 2, sodium-dependent phosphate transporter 3b type II, Accession number Genbank NM_006424); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thromboespondin repeats (type 1 and type 1 similar), transmembrane domain / TM) and short cytoplasmic domain, (semaphorin) 5B Genbank accession number AB040878 ); (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA gene 2700050C12, Accession number Genbank AY358628; (9) ETBR (endothelin receptor type B, Accession number Genbank AY275463); (10) MSG783 (RNF124, Hypothetical protein FLJ20315, Accession number Genbank NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, six transmembrane prostate epithelial 2 antigens, six transmembrane prostate proteins, Genbank accession number AF455138); (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, potential cation channel of the transient receptor, subfamily M, member 4, Accession number Genbank NM_017636); (13) CRYPT (CR, CR1, CRGF, CRYPT, TDGF1, growth factor derived from teratocarcinoma, access number Genbank NP_003203 or NM_003212); (14) CD21 (CR2 (complement 2 receptor) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792, Accession number Genbank M26004); (15) CD79b (beta immunoglobulin associated), B29, Genbank access number NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein la), SPAP1B, SPAP1C, Genbank access number NM_030764); (17) HER2 (Accession number Genbank M11730); (18) NCA (Accession number Genbank M18728); (19) MDP (Accession number Genbank BC017023); (20) IL20Ra (Genbank accession number AF184971); (21) Brevican (Genbank access number AF229053); (22) Ephb2R (Accession number Genbank NM_004442); (23) ASLG659 (Accession number Genbank AX092328); (24) PSCA (Accession number Genbank AJ297436); (25) GEDA (Access number Genbank AY260763); (26) BAFF-R (Accession number Genbank NP_443177.1); (27) CD22 (Accession number Genbank NO-001762.1); (28) CD79a (CD79A, CD79a, alpha-associated immunoglobulin, a B cell-specific protein that interacts covalently with beta Ig (CD79B) and forms a complex on the surface with IgM molecules, transduces a signal involved in the differentiation of the cell B, access number Genbank NP_001774.1); (29) CXCR5 (Burkitt's lymphoma receptor 1, a G-protein coupled receptor that is activated by chemokine CXCL13, works in lymphocyte migration and in humoral defense, plays a role in HIV-2 infection and once in the development of AIDS, lymphoma, myeloma and leukemia, access number Genbank NP__001707.1); (30) HLA-DOB (beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4 + T lymphocytes, Genbank accession number NP_002111.1); (31) P2X5 (P2X purinergic receptor input ion channel 5, an extracellular ATP input channel, - may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of isiopathic detrusor instability, of access Genbank NP_002552.2); (32) CD72 (CD72 antigen of B cell differentiation, Lyb-2, Accession number Genbank NP_001773.1); (33) LY64 (lymphocyte antigen 64 (RP105), membrane protein type I of the leucine-rich repeat family (LRR), t regulates B cell activation and apoptosis, loss of function is associated with an activity of increased disease in patients with systemic lupus erythomatosis, Genbank access number NP_005573.1); (34) FCRH1 (protein 1 similar to the Fe receptor, a putative receptor for the immunoglobulin Fe domain containing Ig-like C2-like domains and ITAM, may have a role in lymphocyte differentiation, Genbank accession number NP_443170.1); or (35) IRTA2 (receptor of the immunoglobulin superfamily associated with translocation 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis; deregulation of the gene by translocation occurs in some B cell diseases, Genbank access NP_112571.1). In another embodiment, R3, R4 and R7 are independently isopropyl or sec-butyl and R5 is -H. In an exemplary embodiment, R3 and R4 are each isopropyl, R5 is -H, and R7 is sec-butyl. In yet another embodiment, R2 and R6 are each methyl, and R9 is -H. In yet another embodiment, each occurrence of R8 is -OCH3. In an exemplary embodiment, R3 and R4 are each isopropyl, R2 and R6 are each methyl, R5 is -H, R7 is sec-butyl, each occurrence of R8 is -OCH3 and R9 is -H. In a Z mode it is -0- or -NH-. In one embodiment, R10 is aryl. In an exemplary embodiment, R10 is -phenyl. In an exemplary embodiment, when Z is -O-, R 11 is -H, methyl or t-butyl. In one embodiment, when Z is -NH, R11 is -CH (R15) 2, wherein R15 is - (CH2) n-N (R16) 2, and R16 is C? -C8 alkyl or - (CH2) n-C00H. In another embodiment, when Z is -NH, R11 is -CH (R15) 2, where R15 is - (CH2) n-S03H. In one aspect, Ab is CAC10, cBR96, cS2C6, clF6, C2F2, hAClO, hBR96, hS2C6, hlF6 and h2F2. The exemplary modalities of the Formula have the following structures: L -MC - ve - PAB -MMAF -MC - e - PAB -MMAE L-MC-MMAE L-MC-MMAF wherein L is an antibody, Val is valine and Cit is citrulline. Drug loading is represented by p, the average number of drug molecules per antibody in a molecule (e.g., of the Formula la, la 'and le). The drug loading can range from 1 to 20 drugs (D) per ligand (e.g., Ab or mAb).
- The compositions of the Formula Ia and the Formula include antibody conjugated collections with a range of drugs, from 1 to 20. The average number of drugs per antibody in the preparation of conjugation reactions can be characterized by conventional means such as spectroscopy of mass, ELISA and HPLC analysis. The quantitative distribution of the ligand-drug conjugates can also be determined in terms of p. In some instances, the separation, purification and characterization of homogeneous ligand-drug conjugates may be achieved wherein p is a certain value of ligand-drug conjugates with other drug charges, by means such as reverse phase HPLC or electrophoresis. 4.2.2. THE DRUG COMPOUNDS OF THE FORMULA (Ib) In another aspect, the present invention provides drug compounds having the Formula Ib: or their salts or pharmaceutically acceptable salts or solvates thereof, wherein; R2 is selected from hydrogen and C? -C8 alkyl; R3 is selected from hydrogen, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); R4 is selected from hydrogen, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3-C8 heterocycle) wherein R5 is selected from H and methyl; or R4 and R? together they have the formula - (CRaRb) n- wherein Ra and Rb are independently selected from H, C? -C8 alkyl, and C3-C8 carbocycle, and n is selected from 2, 3, 4, 5, and 6, and form a ring with the carbon atom to which they are attached; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C? Ca alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from an aryl or heterocycle group C3-C8; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C8 alkyl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; - R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R16 is independently H, C? -C8 alkyl, or - (CH2) n -COOH; and n is an integer ranging from 0 to 6. In one embodiment, R3, R4 and R7 are independently isopropyl or sec-butyl and R5 is -H. In an exemplary embodiment, R3 and R4 are each isopropyl, R5 is -H, and R7 is sec-butyl. In yet another embodiment, R2 and R6 are each methyl, and R9 is -H. In yet another embodiment, each occurrence of R8 is -0CH3. In an exemplary embodiment, R3 and R4 are each isopropyl, R2 and R6 are each methyl, R5 is -H, R7 is sec-butyl, each occurrence of R8 is -OCH3 and R9 is -H. In a Z mode it is -O- or -NH-. In one embodiment, R10 is aryl. In an exemplary embodiment, R10 is -phenyl. In an exemplary embodiment, when Z is -0-, R11 is -H, methyl or t-butyl. In one embodiment, when Z is -NH, R11 is -CH (R15) 2, wherein R15 is - (CH2) n-N (Rls) 2, and R16 is C? -C8 alkyl or - (CH2) n -COOH. In another embodiment, when Z is -NH, R11 is -CH (R15) 2, where R15 is - (CH2) n-S03H. Illustrative compounds of Formula (Ib), which may each be used as drug residues (D) in ADC, include compounds having the following structures: F and pharmaceutically acceptable salts or solvates thereof. THE COMPOUNDS OF THE FORMULA (le) In another aspect, the invention provides antibody-drug conjugated compounds (ADCs) having the formula le: comprising an antibody covalently linked to one or more drug units (residues). Conjugated antibody-drug compounds include pharmaceutically acceptable salts or solvates thereof.
Compounds of Formula I are defined where: Ab is an antibody that binds to one or more of the tumor-associated antigen receptors (1) - (35): (1) BMPR1B (morphogenetic bone protein receptor type IB, Genbank access number NM_001203; (2) El6 (LATÍ, SLC7A5, Genbank access number NM_003486); (3) STEAP1 (six prostate transmembrane epithelial antigens, Genbank access number NM_012449); (4) 0772P (CA125, MUC16 , Accession number Genbank AF 361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancement factor, mesothelin, Genbank access number NM_005823); (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, family 34 of soluble carrier (sodium phosphate), member 2, sodium-dependent phosphate transporter 3b type II, Accession number Genbank NM_006424); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thromboespondin repeats (type 1 and type 1 similar), transmembrane domain / TM) and short cytoplasmic domain, (semaphorin) 5B. Genbank access number AB040878); (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA gene 2700050C12, Accession number Genbank AY358628; (9) ETBR (endothelin receptor type B, Accession number Genbank AY275463); (10) MSG783 (RNF124, Hypothetical protein FLJ20315, Genbank access number NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, six transmembrane epithelial prostate 2 antigens, six transmembrane prostate proteins, of access Genbank AF455138); (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, potential cation channel of the transient receptor, subfamily M, member 4, Accession number Genbank NM_017636); (13) CRYPT (CR, CR1, CRGF, CRYPT, TDGF1, growth factor derived from teratocarcinoma, access number Genbank NP_003203 or NM_003212); (14) CD21 (CR2 (complement 2 receptor) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792, Accession number Genbank M26004); (15) CD79b (beta immunoglobulin associated), B29, Accession number Genbank NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein la), SPAP1B, SPAP1C, accession number Genbank NM 030764); (17) HER2 (Accession number Genbank M11730); (18) NCA (Accession number Genbank M18728); (19) MDP (Accession number Genbank BC017023); (20) IL20Ra! (Accession number Genbank AF184971); (21) Brevican (Genbank access number AF229053); (22) Ephb2R (Accession number Genbank NM_004442); (23) ASLG659 (Accession number Genbank AX092328); (24) PSCA (Accession number Genbank AJ297436); (25) GEDA (Access number Genbank AY260763); (26) BAFF-R (Accession number Genbank NP_443177.1); (27) CD22 (Accession number Genbank NO-001762.1); (28) CD79a (CD79A, CD79a;, alpha-associated immunoglobulin, a B cell-specific protein that covalently interacts with Ig beta (CD79B) and forms a complex on the surface with IgM molecules, transduces a signal involved in the differentiation of cell B, Genbank access number NP_001774.1); (29) CXCR5 (Burkitt's lymphoma receptor 1, a G-protein coupled receptor that is activated by chemokine CXCL13, works in lymphocyte migration and in humoral defense, plays a role in HIV-2 infection and once in the development of AIDS, lymphoma, myeloma and leukemia, access number Genbank NP_001707.1); (30) HLA-DOB (beta subunit of MHC class II molecule (la antigen) that binds peptides and presents them to CD4 + T lymphocytes, Genbank accession number NP_002111.1); (31) P2X5 (P2X purinergic receptor input ion channel 5, an extracellular ATP input channel, may be involved in synaptic transmission and neurogenesis, deficiency may contribute to the pathophysiology of isiopathic detrusor instability, Genbank access NP_002552.2); (32) CD72 (CD72 antigen of B cell differentiation, Lyb-2, Accession number Genbank NP_001773.1); (33) LY64 (lymphocyte antigen 64 (RP105), membrane protein type I of the leucine-rich repeat family (LRR), regulates B cell activation and apoptosis, loss of function is associated with an activity of increased disease in patients with systemic lupus erythomatosis, Genbank accession number NP_005573.1); (34) FCRH1 (protein 1 similar to the Fe receptor, a putative receptor for the immunoglobulin Fe domain containing Ig-like C2-like domains and ITAM, may have a role in lymphocyte differentiation, Genbank accession number NP_443170.1); or (35) IRTA2 (receptor of the immunoglobulin superfamily associated with translocation 2, a putative immunoreceptor with possible roles in B cell development and lymphomagenesis; deregulation of the gene by translocation occurs in some B cell diseases, Genbank access NP_112571.1). A is a stretch unit, a is 0 or 1, each W is independently an amino acid unit, w is an integer that ranges from 0 to 12, Y is a unit of separation, and y is 0, 1 or 2; p ranges from 1 to about 20; and D is a drug residue selected from the DE and DF Formulas: wherein, the wavy line of DE and DF indicates the covalent binding site to A, W, or Y, and independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, CX-C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) C8); R4 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); R5 is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRaRb) n- wherein R a and R b are independently selected from H, C 1 -C 8 alkyl, and C 3 -C 8 carbocycle, and n is selected from 2, 3, 4, 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and O- (C? -C8 alkyl); R9 is selected from H and CX-C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C20 alkyl, aryl, C3-C8 heterocycle, - (RX30) m-R14, or - (R130) m-CH (R1B) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R1S is independently H, C? -C8 alkyl, or - (CH2) n -COOH; R18 is selected from aryl-C (R8) 2-C (R8) 2, -C (R8) 2- C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) 2- (C3-C8 carbocycle); and n is an integer that fluctuates from 0 to 6. In a -Ww- mode it is -Val-Cit-. In another embodiment, R3, R4 and R7 are independently isopropyl or sec-butyl and R? it's H. In an exemplary embodiment, R3 and R4 are each isopropyl, R5 is -H, and R7 is sec-butyl. In yet another embodiment, R2 and R6 are each methyl, and R9 is -H. In yet another embodiment, each occurrence of R8 is -OCH3. In an exemplary embodiment, R3 and R4 are each isopropyl, R2 and R6 are each methyl, R5 is -H, R7 is sec-butyl, each occurrence of R8 is -OCH3 and R9 is -H. In a Z mode it is -O- or -NH-. In one embodiment, R10 is aryl. In an exemplary embodiment, Rxo is -phenyl. In an exemplary embodiment, when Z is -O-, R 11 is -H, methyl or t-butyl.
In one embodiment, when Z is -NH, R11 is -CH (R15) 2, wherein R15 is - (CH2) n-N (R16) 2, and R16 is C? -C8 alkyl or - (CH2) n -COOH. In another embodiment, when Z is -NH, R11 is -CH (R15) 2, where R15 is - (CH2) n-S03H. In one aspect, Ab is cACLO, CBR96, cS2C6, clF6, c2F2, hACLO, hBR96, hS2C6, hlF6 and h2F2. The exemplary modalities of the Formula ADC have the following structures: Ab-MC-vc-PAB-MMAF Ab-MC-vc-PAB-MMAE Ab-MC-MMAE Ab-MC-MMAF wherein Ab is an antibody that binds to one or more tumor-associated antigen receptors (1) - (35); Val is valine and Cit is citrulline. The drug load is represented by p, the average number of drugs per antibody in a molecule of Formula I. The drug load can range from 1 to 20 drugs (D) per antibody (Ab or mAb). The ADC compositions of Formula I include collections of antibodies conjugated with a range of drugs, from 1 to 20. The average number of drugs per antibody in ADC preparations of conjugation reactions can be characterized by conventional means such as visible spectroscopy by UV, mass spectrometry, ELISA and HPLC analysis. The quantitative distribution of ADC can also be determined in terms of p. In some instances, separation, purification and characterization of homogeneous ADC conjugates may be achieved where p is a certain ADC value with other drug loads, by means such as reverse phase HPLC or electrophoresis. For some antibody-drug conjugates, p may be limited by the number of binding sites in the antibody. For example, when the binding is a thiol cysteine, as in the above exemp embodiments, an antibody may have only one or more cysteine thiol groups, or may have only one or more sufficiently reactive thiol groups through which a linker may be attached . Typically, less than the maximum theoretical drug residues are conjugated to an antibody during a conjugation reaction. An antibody may contain, for example, many lysine residues that do not react with the drug-linker intermediate or linker reagent. Only the most reactive lysine groups can react with an amine-reactive linker. Generally, antibodies do not contain many, if any, free cysteine thiol groups and reagents that can bind to a drug residue. Most of the cysteine thiol residues in the antibodies of the compounds of the invention exist as disulfide bridges and must be reduced with a reducing agent such as dithiothreitol (DDT). Additionally, the antibody must be subjected to denaturing conditions to reveal reactive nucleophilic groups such as lysine or cysteine. The charge (drug / antibody ratio) of an ADC can be controlled in several different ways, including: (i) limiting the molar excess of drug-linker intermediary or linker reagent relative to the antibody, (ii) limiting the conjugation reaction time or temperature, and (iii) limiting partially reductive conditions for the modification of cysteine thiol . It will be understood that when more than one nucleophilic group reacts with a drug-linker intermediate, or linker reagent followed by the drug residue reagent, then the resulting product is a mixture of ADC compounds with a distribution of one or more drug residues. attached to an antibody. The average number of drugs per antibody can be calculated from the mixture by means of dual ELISA analysis of the antibody, specific for the antibody and specific for the drug. The individual ADC molecules can be identified in the mixture by mass spectroscopy, and separated by HPLC, eg, hydrophobic interaction chromatography ("Effect of drug loading on the pharmacology, pharmacokinetics and toxicity of an anti-CD30 antibody-drug conjugate", Hamblett, KJ et al., Excerpt No. 624, American Association for Cancer Research, 2004 Annual Meeting, March 27-31, 2004, Proceedings of the AACR, Volume 45, March 2004, "Controlling the Location of Drug Attachment in Antibody- Drug Conjugates ", Alley, SC, et al., Extract No. 627, American Association AACR, Volume 45, March 2004). Therefore, a homogeneous ADC with a single charge value can be isolated from the conjugation mixture by electrophoresis or chromatography. 4.3 THE LINKER UNIT A "linker unit" (LU) is a bifunctional compound that can be used to link a drug unit and a ligand unit to form drug-linker-ligand conjugates, or that is useful in the formation of immunoconjugates directed against antigens associated with tumor. Such immunoconjugates allow the selective delivery of toxic drugs to tumor cells. In one embodiment, the linker unit of the drug-linker compound and the drug-linker-ligand conjugate has the formula: - Aa- ww-? Y- where: A- is a stretch unit, a is 0 or 1, each -W- is independently an amino acid unit, w is an integer that ranges from 0 to 12, -Y- is a unit of separation, and y is 0, 1 or 2. In the drug-linker-ligand conjugate , the linker is able to bind the drug residue and the ligand unit. 4.3.1. THE STRETCHING UNIT The stretching unit (-A-), if present, is capable of binding a ligand unit to an amino acid unit (-W-). In this regard, a ligand (L) has a functional group that can form a bond with a functional group of a stretching unit. Useful functional groups that may be present in a ligand, either naturally or through chemical manipulation include, but are not limited to sulfhydryl (-SH), amino, hydroxyl, carboxy, the anomeric hydroxyl group of a carbohydrate and carboxyl. In one aspect, the ligand functional groups are sulfhydryl and amino. Sulfhydryl groups can be generated by reduction of an intramolecular disulfide bond of a ligand. Alternatively, sulfhydryl groups can be generated by the reaction of an amino group of a lysine residue of a ligand using 2-iminothiolane (Traut's reagent) or another reagent that generates sulfhydryl. In one embodiment, the stretching unit forms a bond with a sulfur atom of the ligand unit. The sulfur atom can be derived from a sulfhydryl group of a ligand. The stretching units representative of this embodiment are illustrated within the square brackets of the Formulas Illa and Illb, where L-, -W-, -Y-, -D, wyy are as defined above, and R17 is selected from C 1 -C 10 alkylene, C 3 -C 8 carbocycle, -O-C 8 -C 8 alkyl) -, -arylene-, C 1 -C 0 alkylene-arylene, C 1 -C 0 alylene-C 1 -C 0 alkylene, C 1 -C 10 alkylene - (C3-C8 carbocycle) -, - (C3-C8 carbocycle) - C? -C? 0- alkylene, C3-C8 heterocycle, C? -C10-alkylene (C3-C8 heterocycle) -, - (C3 heterocycle) -C8) -alkylene C? -C10-, - (CH2CH20) r-, and - (CH2CH20) r -CH2-; and r is an integer that fluctuates from 1-10. It should be understood from all exemplary embodiments of Formula la, such as III-VI, that even when not expressly denoted, they are linked to a ligand of 1 to 20 drug residues (p = 1-20).
• CH -CONH- R17-C (0) - -Ww-Yu- D pib An illustrative stretch unit is that of Formula Illa where R17 is - (CH2) 5-: Another illustrative stretch unit is that of Formula Illa wherein R is - (CH2CH20) r -CH2-; and r is 2: Still another illustrative stretch unit is that of Formula Illb where R17 is - (CH2) 5-: In another embodiment, the stretching unit is linked to the ligand unit through a disulfide bond between a sulfur atom of the ligand unit and a sulfur atom of the stretching unit. A representative stretching unit in this embodiment is illustrated within the square brackets of Formula IV, where R17, L-, -W-, -Y-, -D. W and y are as defined above.
In yet another embodiment, the reactive group of the stretch unit contains a reactive site that can form a bond with a primary or secondary amino group of a ligand. Examples of these reactive sites include, but are not limited to, activated esters such as succinimide esters, 4-nitrophenyl esters, pentafluorophenyl esters, tetrafluorophenyl esters, anhydrides, acid chlorides, sulfonyl chlorides, isocyanates and isothiocyanates. Representative stretching units in this embodiment are illustrated within the square brackets of Formulas Va and Vb, where R17, L-, -W-, -Y-, -D. W and y are as defined above. t "-C (0) NH-R17 ~ C (0) - Ww-Y-D Va In yet another aspect, the reactive group of the stretch unit contains a reactive site that is reactive to a modified carbohydrate group (-CHO) that may be present in a ligand. For example, a carbohydrate can be partially oxidized using a reagent such as sodium periodate and the resulting unit (-CHO) of the oxidized carbohydrate can be condensed with a stretching unit containing a functionality - such as a hydrazide, an oxime, a primary amine or secondary, a hydrazine, a thiose icarbazone, a hydrazine carboxylate, and an aryl hydrazide such as those described by Kaneko, T. Et al., (1991) Bioconjugate Chem., 2: 133-41. Representative stretching units in this embodiment are illustrated within the square brackets of the Formulas Via, VIb, and VIc, where R17, L-, -W-, -Y-, -D. W and y are as defined above.
N-NH-R 7-C (0) f-W 'wW-Y' vy-D Via : N-0- 17-C (0) - * W 'WW-Yy-D VIb 4. 3.2 THE AMINO ACID UNIT The amino acid unit (-W-) when present, links the stretch unit to the separation unit if the separation unit is present, - links the stretch unit to the drug residue if the separation unit is absent, and links the ligand unit to the drug unit if the stretch unit and the separation unit are absent. Ww is a dipeptide, tripeptide, tetrapeptide, pentapeptide, hexapeptide, heptapeptide, octapeptide, nonapeptide, decapeptide, undecapeptide, or dodecapeptide unit. Each unit -W- has independently the formula denoted below in the square brackets and w is an integer that fluctuates from 0 to 12: wherein R19 is hydrogen, methyl, isopropyl, isobutyl, sec-butyl, benzyl, p-hydroxybenzyl, -CH20H, -CH (0H) CH3, CH2CH2SCH3, -CH2CONH2, -CH2COOH, -CH2CH2C0NH2. -CH2CH2C00H, - (CH2) 3NHC (= NH) NH2, - (CH2) 3NH2, - (CH2) 3NHC0CH3, - (CH2) 3NHCHO, - (CH2) 4NHC (= NH) NH2, - (CH2) 4NH2, - (CH2) 4NHC0CH3, - (CH2) 4NHCH0, - (CH2) 3NHC0NH2, - (CH) 4NHCONH2, -CH2CH2CH (OH) CH2NH2, 2-pyridylmethyl, 3-pyridylmethyl, 4-pyridylmethyl, phenyl, cyclohexyl, - The amino acid unit can be enzymatically divided by one or more enzymes, including a tumor-associated protease, to release the drug unit (-D), which in one embodiment is protonated in vivo upon release to provide a drug (D). Illustrative Ww units are represented by formulas (VII) - (IX): (VH) wherein R20 and R21 are as follows R20 R21 Benzyl (CH2) 4NH2; Methyl (CH2) 4 NH2; Isopropyl (CH2) 4NH2; Isopropyl (CH2) 3NHCONH2; Benzyl (CH2) 3NHCONH2; Isobutyl (CH2) 3NHC0NH2; Sec-butyl (CH2) 3NHC0NH2; benzyl methyl; and benzyl (CH2) 3 NHC (= NH) NH2; where R, and R are as follows: 20 ^ 21 Benzyl benzyl (CH 2) 4 NH 2; Isopropyl benzyl (CH 2) 4 NH 2; and H benzyl (CH 2) 4 NH 2; wherein R, R, R and R are as follows: R20 R21 R22 R23 H benzyl isobutyl H; and Methyl isobutyl methyl isobutyl Exemplary amino acid units include but are not limited to, units of the formula (VII) wherein: R20 is benzyl and R21 is - (CH2) NH2; R20 is isopropyl and R21 is - (CH2) 4NH2; R20 is isopropyl and R21 is - (CH2) 3NHC0NH2. Another exemplary amino acid unit is a unit of the formula (VIII) wherein R20 is benzyl, R21 is benzyl and R22 is - (CH2) 4NH2. Useful units -Ww- can be designed and optimized in their selectivity for enzymatic cleavage by particular enzymes, for example, a tumor-associated protease. In one embodiment, a -Ww ~ unit is one whose division is catalyzed by cathepsin B, C and D, or a plasmin protease. In one embodiment, -Ww- is a dipeptide, tripeptide, tetrapeptide or pentapeptide. Where R19, R20, R21, R22 or R23 is different from hydrogen, the carbon atom to which R19, R20, R21, R22 or R23 is attached is chiral. Each carbon atom to which R19, R20, R21, R22 or R23 is attached is independently in the (S) or ® configuration. In one aspect of the amino acid unit, the amino acid unit is valine-citrulline. In another aspect, the amino acid unit is phenylalanine-lysine (i.e., fk). In yet another aspect of the amino acid unit, the amino acid unit is N-methylvaline-citrulline. In yet another aspect, the amino acid unit is 5-aminovaleric acid, homo-phenylalanine-lysine, tetraisoquinolinecarboxylate-lysine, cyclohexylalanine-lysine, isonopecotic-lysine, beta-alanine-lysine, glycine-serine-valine-glutamine, and isonopecotic acid. In certain embodiments, the amino acid unit may comprise natural amino acids. In other embodiments, the amino acid unit may comprise non-natural amino acids. 4.3.3 THE SEPARATION UNIT The separation unit (-Y-) when present, binds the amino acid unit to the drug residue if the amino acid unit is present. Alternatively, the separation unit links the stretch unit to the drug residue if the amino acid unit is absent. The separation unit also binds the drug residue to the ligand unit when both the amino acid unit and the stretch unit are absent. The separation units are of two general types: self-immolative and not self-immolative. A self-immolative separation unit is one in which part or all of the separation unit remains bound to the drug residue after division, particularly enzymatic cleavage of an amino acid unit of a drug-linker-ligand conjugate or the compound of drug-linker. Examples of a non-self-immolative separation unit include, but are not limited to, a glycine-glycine separation unit and a glycine separation unit (both illustrated in Scheme 1 (infra) .When an exemplary compound containing a unit of glycine-glycine separation or a glycine separation unit, undergoes enzymatic cleavage through a tumor-associated protease, a cancer cell-associated protease or a lymphocyte-associated protease, a glycine-glycine drug residue or a glycine drug residue, L-Aa-Ww- In one embodiment, the independent hydrolysis reaction takes place within the target cell, dividing the glycine-drug residue link and releasing the drug. -Yy- is a p-aminobenzyl alcohol (PAB) unit (see Schemes 2 and 3) whose phenylene portion is substituted with Qm where Q is C?-C8 alkyl, -0- (C?-C8 alkyl), - halogen, -nitro or -ciano; and m is an ent ero that fluctuates from 0-4. Scheme 1 Ab- -Aa-Wvt-Giy-D Ab ~ f-Aa-Ww-G) y-Gly] -D splitting enzymatic cleavage + enzymatic í Gly-D Gly-Gly-D hydrolysis hydrolysis Drug Drug In one modality, a non-self-immolative separation unit (-Y-) is -Gly-Gly-. In another modality a non-self-immolative separation unit (-Y-) is -Gly-. In one embodiment, a drug-linker compound or a drug-linker-ligand conjugate is provided in which the separation unit is absent (y = o), or a pharmaceutically acceptable salt or solvate thereof. Alternatively, an exemplary compound containing a self-immolative separation unit can release -D without the need for a separate hydrolysis step. In this embodiment, -Y- is a PAB group linked to -Ww- through the amino nitrogen atom of the PAB group, and directly connected to -D through a carbonate, carbamate or ether group. Without joining any particular theory or mechanism, Scheme 2 illustrates a possible drug release mechanism of a PAB group that is directly linked to -D through a carbonate or carbamate group disclosed by Toki et al., (2002) J. Org. Chem., 67: 1866-1872. Scheme 2 1, 6- Elimination Drug wherein Q is C? -C8 alkyl, -O- (Cx-C8 alkyl), -halogen, -nitro or -cyano; m is an integer that fluctuates from 0-4; and p ranges from 1 to about 20. Without joining any particular theory or mechanism, Scheme 2 illustrates a possible drug release mechanism of a PAB group that is directly linked to -D through an ether or amine bond.
Scheme 3 enzymatic unfolding 1, 6- elimination Drug wherein Q is C? -C8 alkyl, -O- (C? -C8 alkyl), -halogen, -nitro or -cyano; m is an integer that fluctuates from 0-4; and p ranges from 1 to about 20. Other examples of self-immolative separators include, but are not limited to, aromatic compounds that are electronically similar to the PAB group such as 2-aminoimidazole-5-methanol derivatives (Hay et al., ( 1999) Bioorg, Med. Chem. Lett. 9: 2237) and ortho or for aminobenzylacetals. Separators that undergo cyclization to amide bond hydrolysis, such as substituted and unsubstituted 4-aminobutyric acid amides (Rodrigues et al., Chemistry Biology, 1995, 2, 223), bicyclo ring systems [2.2.1 ] and bicyclo [2.2.2] appropriately substituted (Storm et al., J. Amer. Chem. Soc., 1972, 94, 5815) and 2-aminophenyl propionic acid amides (Amsberry et al., J. Org. Chem. , 1990, 55, 5867). Also exemplary of self-immolative spacer useful in the exemplary compounds are the elimination of amine-containing drugs that are substituted at the glycine α-position (Kingsbury et al., J. Med. Chem., 1984, 27 1447). In one embodiment, the separation unit is a branched bis (hydroxymethyl) styrene (BHMS) unit as illustrated in Scheme 4 that can be used to incorporate and release multiple drugs. Scheme 4 enzymatic splitting 2 drugs wherein Q is C? -C8 alkyl, -0- (C? -C8 alkyl), -halogen, -nitro or -cyano; m is an integer that fluctuates from 0-4; and p - - ranges from 1 to approximately 20. In one embodiment, the -D residuals are the same. Even in another modality, the -D residuals are different. In one aspect, the separation units (-Yy-) are represented by the Formulas (X) - (XII): wherein Q is C? -C8 alkyl, -O- (C? -C8 alkyl), -halogen, nitro or -cyano; m is an integer that fluctuates from 0-4; Y NHCH2C (0) -NHCH2C (0) -xp.
The embodiments of Formula IA 'and the conjugated antibody-drug compounds include: - w and y are each 0, 4. 4 THE DRUG UNIT (RESIDUAL) The drug residue (D) of the antibody-drug conjugates (ADC) is of the dolastatin / auristatin type (US Patents Nos. 5635483; 5780588) which have been shown to interfere with microtubule dynamics, GTP hydrolysis, and nuclear and cell division (Woyke et al., (2001) Antimicrob.
Agents and Chemother., 45 (12): 3580-3584) and have anti-cancer activity (U.S. Patent No. 5663149) and anti fungal (Pettit et al., (1998) Antimicrob Agents Chemother., 42: 2961-2965).
- D is a drug unit (residue) having a nitrogen atom that can form a bond with the separation unit when y = lo 2, with the carboxyl terminating group C of an amino acid unit when y = o, with the carboxyl group of a stretch unit when w and y = 0, and with the carboxyl group of a drug unit when a, w and y = 0. It should be understood that the terms "drug unit" and "drug residue" are synonymous and are used interchangeably herein. In one modality, -D is either the DE or DF formula: wherein, independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); R4 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, alkylaryl Ca-C8, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) C8); R5 is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRRb) n- wherein R a and R b are independently selected from H, C 1 -C 8 alkyl, and C 3 -C 8 carbocycle, and n is selected from 2, 3, 4, 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) -C8); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-Ca carbocycle, and 0- (Ca-C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is O, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C8 alkyl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C1-Cß; each occurrence of R1S is independently H, C? -C8 alkyl, or - (CH2) n -COOH; n is an integer ranging from 0 to 6. In one embodiment, R3, R4 and R7 are independently isopropyl or sec-butyl and R5 is -H. In an exemplary embodiment, R3 and R4 are each isopropyl, R5 is -H, and R7 is sec-butyl. In another embodiment, R2 and R6 are each methyl, and R9 is -H. In yet another embodiment, each occurrence of R8 is -OCH3. In an exemplary embodiment, R3 and R4 are each isopropyl, R2 and R6 are each methyl, R5 is -H, R7 is sec-butyl, each occurrence of R8 is -0CH3 and R9 is -H. In a Z mode it is -0- or -NH-. In one embodiment, R10 is aryl. In an exemplary embodiment, R10 is -phenyl. In an exemplary embodiment, when Z is -O-, R 11 is -H, methyl or t-butyl. In one embodiment, when Z is -NH, R11 is -CH (R15) 2, wherein R15 is - (CH2) nN (R? E) 2, and R16 is C? -C8 alkyl or - (CH2) n- C00H. In another embodiment, when Z is -NH, R11 is -CH (R15) 2, where R15 is - (CH2) n-S03H. Exemplary drug units (-D) include drug units that have the following structures: and pharmaceutically acceptable salts and solvates thereof. In one aspect, hydrophilic groups, such as, but not limited to triethylene glycol esters (TEG), as shown above, can be attached to the drug unit in R11. Without joining any theory, the hydrophilic groups help in the internalization and non-agglomeration of the drug unit. 4.5 THE LINKING UNIT The ligand unit (L-) includes within its scope any unit of a ligand (L) that is linked or reactively associated or complexed with a receptor, antigen or other receptive residue associated with a population of given target cell. A ligand is a molecule that binds to, complexes with, or reacts with a residue of a cell population that is sought to be modified biologically therapeutically or otherwise. In one aspect, the ligand unit acts to deliver the drug unit to a particular target cell population with which the ligand unit reacts. Such ligands include, but are not limited to, high molecular weight proteins such as, for example, full-length antibodies, antibody fragments, lower molecular weight proteins, polypeptides or peptides, lectins, glycoproteins, non-peptides, vitamins, molecules nutrient transporters (such as, but not limited to, transferrin), or any other molecule or cell-binding substance. A ligand unit can form a link to a stretch unit, an amino acid unit, a separation unit, or a drug unit. A ligand unit can form a link to a linker unit through a heteroatom of the ligand. Heteroatoms which may be present in a ligand unit include sulfur (in one embodiment, of a sulfhydryl group of a ligand), oxygen (in one embodiment, of a carbonyl, carboxyl or hydroxyl group of a ligand) and nitrogen (in one embodiment). mode of a primary or secondary amino group). These heteroatoms may be present in the ligand in the natural state of the ligand, for example an antibody of natural origin, or they may be introduced into the ligand through chemical modification. In one embodiment, a ligand has a sulfhydryl group and the ligand is linked to the linker unit through the sulfur atom of the sulfhydryl group. In yet another aspect, the ligand has one or more lysine residues that can be chemically modified to introduce one or more sulfhydryl groups. The ligand unit is linked to the linker unit through the sulfur atom of the sulfhydryl group. Reagents that can be used to modify plants include, but are not limited to, N-succinimidyl-S-acetylthioacetate (SATA) and 2-iminothiolane hydrochloride (Traut's reagent). In another embodiment, the ligand may have one or more carbohydrate groups that can be chemically modified to have one or more sulfhydryl groups. The ligand unit is linked to the linker unit, such as the stretching unit, through the sulfur atom of the sulfhydryl group. In yet another embodiment, the ligand may have one or more carbohydrate groups that can be oxidized to provide an aldehyde group (-CH0) (see, eg, Laguzza, et al., J. Med. Chem., 1989, 32 (3) , 548-55). The corresponding aldehyde can form a bond with a reactive site in a stretching unit. Reactive sites in a stretch unit that can react with a carbonyl group in a ligand include, but are not limited to, hydrazine and hydroxylamine. Other protocols for the modification of proteins for the union or association of the drug units are described in Coligan et al. , Current Protocols in Protein Science, vol. 2, John Wiley &; Sons (2002), incorporated herein by reference. Non-immunoreactive proteins, polypeptides, or useful peptide ligands, include, but are not limited to, transferrin, epidermal growth factors ("EGF"), bombesin, gastrin, gastrin-releasing peptide, platelet-derived growth factor, IL -2, IL-6, transforming growth factors ("TGF"), such as TGF-a; and TGF- / 3, vaccine growth factor ("VGF"), insulin and insulin-like growth factors I and II, lectins and low density lipoprotein apoprotein. Useful polyclonal antibodies are heterogeneous populations of antibody molecules derived from the serum of immunized animals. Various methods well known in the art can be used for the production of polyclonal antibodies to an antigen of interest. For example, for the production of polyclonal antibodies, several host animals can be immunized by injection with an antigen of interest or a derivative thereof, including but not limited to rabbits, mice, rats and guinea pigs. Various adjuvants can be used to increase the immune response, depending on the host species, and including, but not limited to Freund's adjuvant (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols , polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (Bacillus Calmette-Guerin) and corynebacterium parvura. Such adjuvants are also well known in the art. Useful monoclonal antibodies are homogeneous populations of antibodies to a particular antigenic determinant (eg, a cancer cell antigen, a viral antigen, a microbial antigen, a protein, a peptide, a carbohydrate, a chemical, nucleic acid or its fragments. monoclonal (Ab) for an antigen of interest can be prepared using any technique known in the art that provides for the production of antibody molecules by continuous cell lines in culture, These include, but are not limited to, the hybridoma technique originally described by Kohier et al. al., (1975) Nature 256: 495, the human B-cell hybridoma technique (Kozbor et al., 1983, Immunology Today, 4:72), and the EBV-hybridoma technique (Cole et al., 1985 , Monoclonal Antibodies and Cancer Therapy, Alan R. Liss Inc., pp. 77-96) Such antibodies can be of any kind of immunoglobulin including IgG, IgM, IgE, IgA and IgD and which any subclass of it. The hybridoma that produces the mAbs useful in this invention can be cultured in vitro or in vivo. Useful monoclonal antibodies include, but are not limited to, human monoclonal antibodies, humanized monoclonal antibodies, antibody fragments or human-mouse chimeric monoclonal antibodies (or other species). Human monoclonal antibodies can be made by any of a number of techniques known in the art (eg, Teng et al., 1983, Proc Nati Acad Sci USA 80, 7308-7312 Kozbor et al., 1983, Immunology Today 4 , 72-79; and Olsson et al., 1982, Meth. Enzymol., 92, 3-16). The antibody can also be a bispecific antibody. Methods for making bispecific antibodies are well known in the art. The traditional production of full-length bispecific antibodies is based on the co-expression of two heavy chain-immunoglobulin light chain pairs, where the two chains have different specificities (Milstein et al., 1983, Nature 305: 537-539 ). Due to the random selection of heavy and light immunoglobulin chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Similar procedures are described in International Publication No. WO 93/08829 and in Traunecker et al., EMBO J. 10: 3655-3659 (1991). According to a different procedure, antibody variable domains are fused with the desired binding specificities (antibody-antigen combining sites) to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy chain constant domain comprising at least part of the joint CH2 and CH3 regions. It is preferred that the first heavy chain constant region (CH1) contains the necessary site for the link of the light chain, present in at least one of the mergers. Nucleic acids with sequences encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and co-transfected into a suitable host organism. This provides great flexibility to adjust the mutual proportions of the three polypeptide fragments in modalities where the unequal ratios of the three polypeptide chains used in the construction provide optimal yields. However, it is possible to insert the coding sequences of two or all three polypeptide chains into an expression vector, when the expression of at least two polypeptide chains in equal ratios results in higher yields when the ratios are not greater meaning. In one embodiment of this procedure, the specific antibodies have an immunoglobulin hybrid heavy chain with a first binding specificity in one arm, and an immunoglobulin heavy chain-light chain hybrid pair (providing a second binding specificity) in the other arm. This asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, since the presence of an immunoglobulin light chain in only half of the bispecific molecule provides an easy way of separation (International Publication No. WO 94). / 04690 which is incorporated herein by reference in its entirety). For further details for the generation of bispecific antibodies, see, for example, Suresh et al., Methods in Enzymology, 1986, 121: 210; Rodrigues et al ,. 1993 J. of Immunology 151: 6954-6961; Cárter et al., 1992, Bio / Technology 10: 163-167; Carter et al., 1995, J. of Hematotherapy 4: 463-470; Merchant et al., 1998, Nature Biotechnology 16: 677-681. Using such techniques, bispecific antibodies can be prepared for use in the treatment or prevention of disease as defined herein. Bifunctional antibodies are also described in European Patent Publication No. EPA 0 105 360. As described in this reference, hybrid or bifunctional antibodies can be derived either biologically, ie, by cell fusion techniques, or chemically, especially with cross-linking agents or disulfide bridge-forming reagents, and may comprise whole antibodies or fragments thereof. Methods for obtaining such hybrid antibodies are described, for example, in International Publication WO 83/03679, and European Patent Publication No. EPA 0 217 577, both incorporated herein by reference. The antibody can be a functionally active fragment, derivative or analogue of an antibody that immunospecifically binds to cancer cell antigens, viral antigens, or microbial antigens or other antibodies bound to cells or tumor matrix. In this regard, "functionally active" means that the fragment, derivative or analog is capable of emitting anti-idiotypic antibodies that recognize the same antigen that recognized the antibody from which the fragment, derivative or analog is derived. Specifically, in an exemplary embodiment, the antigenicity of the idiotype of the immunoglobulin molecule can be improved by deleting the structure and CDR sequences that are C-terminating for the CDR sequence that specifically recognizes the antigen. To determine which CDR sequences are linked to the antigen, synthetic peptides containing the CDR sequences can be used in antigen binding assays by any method of linkage analysis known in the art (eg, BIAcore analysis) (see, eg, Kabat et al., (1991), Sequences of Proteins of Immunological Interest, 5th Ed., Public Health Service, National Institutes of Health, Bethesda MD; Kabat et al., 1980 J. of Immunology, 125 (3): 961-969). Other useful antibodies include antibody fragments such as, but not limited to, F (ab ') 2 fragments containing the variable region, the light chain constant region and the CH1 domain of the heavy chain that can be produced by pepsin digestion. the antibody molecule, and Fab fragments that can be generated by reducing the disulfide bridges of the F (ab ') 2 fragments. Other useful antibodies are dimers of heavy chain and light chain antibodies, or any minimal fragment thereof such as Fvs or single chain antibodies (SCAs). (e.g., as described in U.S. Patent No. 4946778; Bird 1988, Science 242: 423-42; Houston et al., 1988, Proc.
Nati Acad. Sci. USA 85: 5879-5883; and Ward et al., 1989, Nature, 334: 544-54, or any other molecule with the same specificity as the antibody. Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are useful antibodies. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from constant regions of a murine monoclonal and human immunoglobulin. (See, e.g., Cabilly et al., U.S. Patent No. 4816567, and Boss et al., U.S. Patent No. 4,816,397, which are incorporated herein by reference in their entirety). Humanized antibodies are antibody molecules of non-human species that have one or more complementarity determining regions (CDRs) of non-human species and a framework region of a human immunoglobulin molecule. (See, e.g., Queen, U.S. Patent No. 5,585,089, which is incorporated herein by reference in its entirety). Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using the methods described in International Publication No. WO 87/02671; European Patent Publication No. 184,187; European Patent Publication No. 171496; European Patent Publication No. 173494; International Publication No. WO 86/01533; Patent of E.U. No. 4816567; European Patent Publication No., 12,023; Berter et al., 1988, Science 240: 1041-1043; Liu et al., 1987, Proc. Nati Acad. Sci., USA 84: 3439-3443; Liu et al., 1987, J. Immunol. , 139: 3521-3526; Sun et al., 1987, Proc. Nati Acad. Sci. USA 84: 214-218; Nishimura et al., 1987, Cancer Res. 47: 999-1005; Wood et al., 1985, Nature 314: 446-449; and Shaw et al., 1988, J. Nati. Cancer Inst. , 80: 1553-1559; Morrison, 1985, Science 229: 1202-1207; Oi et al., 1986, BioTechniques 4: 214; Patent of E.U. No. 5225539; Jones et al., 1986, Nature 321: 552-525; Verhoeyen et al., (1988) Science 239: 1534; and Beidler et al., 1988, J. Immunol. 141: 4053-4060; each of which is incorporated herein by reference in its entirety. Fully human antibodies are particularly desirable and can be produced using transgenic mice that are unable to express endogenous immunoglobulin heavy and light chain genes, but can express human heavy and light chain genes. The transgenic mice are immunized in the normal manner with a selected antigen, e.g., all or part of a polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice are redistributed during B cell differentiation, and subsequently undergo class exchange and somatic mutation. Thus, by using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. See Lonberg and Huszar (1995, Int. Rev. Immunol., 13: 65-93). For a detailed discussion of this technology for the production of human antibodies and human monoclonal antibodies and protocols for the production of such antibodies, see, e.g., US Patents. Nos. 5625126; 5633425; 5569825; 5661016; 5545806; each of which is incorporated herein by reference in its entirety. Other human antibodies can be obtained commercially, for example, from Abgenix, Inc., (Freemont, CA) and Genpharm (San Jose, CA). Fully human antibodies that recognize a selected epitope can be generated using a technique referred to as "guided selection". In this procedure, a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a fully human antibody that recognizes the same epitope. (Jespers et al., (1994) Biotechnology 12: 899-903). Human antibodies can also be produced using various techniques known in the art, including phage display libraries (Hoogenboom and Winter J. Mol. Biol., 227: 381 (1991)).; Marks et al., J. Mol. Biol., 222: 581 (1991); Quan, M. P. and Cárter, P. 2002. The rise of monoclonal antibodies as therapeutics, in Anti-lgE and Allergic Disease, Jardieu, P. M. and Fick Jr., R. B. eds., Marcel Dekker, New York, N. Y., Chapter 20 pp. 427-469). In other embodiments, the antibody is a fusion protein of an antibody, or a functionally active fragment thereof, for example, in which the antibody is fused through a covalent bond (eg, a peptide bond), either at the N terminus or at the C terminus, to an amino acid sequence of another protein (or portion thereof, preferably at least the amino acid portion 10, 20 or 50 of the protein) that is not the antibody. Preferably, the antibody or its fragment is covalently bound to the other protein at the N terminus of the constant domain. Antibodies include analogs and derivatives that are modified either i.e., by the covalent attachment of any type of molecule while such covalent attachment allows the antibody to retain its immunospecificity of antigen binding. For example, but not by way of limitation, the derivatives and analogs of the antibodies include those that have been further modified, eg, by glycosylation, acetylation, pegylation, phosphorylation, amidation, derivatization by known protecting / blocking groups, proteolytic cleavage, linkage to a cell antibody or other protein unit, etc. Any of numerous chemical modifications can be carried out by known techniques, including, but not limited to, specific chemical division, acetylation, formylation, metabolic synthesis in the presence of tunicamycin, etc. Additionally, the analog or derivative may contain one or more non-natural amino acids. The antibodies include antibodies that have modifications (e.g., substitutions, deletions or additions) in amino acid residues that interact with Fe receptors. In particular, antibodies include antibodies that have modifications to amino acid residues identified as being involved in the interaction between the anti-Fe domain and the FcRn receptor (see, eg, International Publication No. WO 97/34631, which is incorporated herein by reference). the reference in its entirety). Immunospecific antibodies to a cancer cell antigen can be obtained commercially, for example from Genentech (San Francisco, CA) or produced by any method known to those skilled in the art such as, e.g., chemical synthesis or recombinant expression techniques. The nucleotide sequence encoding immunospecific antibodies to a cancer cell antigen can be obtained, e.g., from the GenBank database or a similar database, literary publications or by routine cloning and sequencing. In a specific embodiment, known antibodies for the treatment or prevention of cancer can be used. Immunospecific antibodies to a cancer cell antigen can be obtained commercially or produced by any method known to the person skilled in the art such as, e.g., recombinant expression techniques. The nucleotide sequence coding for immunospecific antibodies to a cancer cell antigen can be obtained, e.g., from the GenBank database or a similar database, literary publications or by routine cloning and sequencing. Examples of antibodies available for the treatment of cancer include, but are not limited to, humanized anti-HER2 monoclonal antibody, HERCEPTIN® (trastuzumab, Genentech) for the treatment of patients with stastatic breast cancer; TOTUXAN® (rituximab; Genentech) which is a chimeric anti-CD20 monoclonal antibody for the treatment of patients with non-Hodgkin's lymphoma; OvaRex (AltaRex Corporation, MA) which is a murine antibody for the treatment of ovarian cancer; Panorex (Glaxo Wellcome, NC) which is a murine IgG2a antibody for the treatment of colorectal cancer; Cetuximab Erbitux (Inclone Systems Inc., NY) which is an ani-EGFR IgG chimeric antibody for the treatment of epidermal growth factor-positive cancers, such as head and neck cancer; Vitaxin (Medl mune, Inc., MD) which is a humanized antibody for the treatment of sarcoma; Campath 1 / H (Leukosite, MA) which is a humanized IgGl antibody for the treatment of chronic lymphocytic leukemia (CLL); Smart M195 (Protein Design Labs., Inc., CA) which is a humanized anti-CD33 IgG antibody for the treatment of acute myeloid leukemia (AML); LymphoCide (Immunomedics, Inc., NJ) which is a humanized anti-CD22 antibody for the treatment of non-Hodgkin's lymphoma: Smart ID10 (Protein Design Labs., Inc., CA) which is a humanized anti-HLA-DR antibody for the treatment of non-Hodgkin's lymphoma; Oncolym (Technoclone, Inc., CA) which is a murine anti-HLA-DR10 radiolabelled antibody for the treatment of non-Hodgkin's lymphoma; Allomune (BioTransplant, CA) which is a humanized anti-CD2 mAb for the treatment of Hodgkin's disease or non-Hodgkin's lymphoma; Avastin (Genentech, Inc., CA) which is a humanized anti-VEGF antibody for the treatment of pulmonary and colorectal cancers; Apratuzamab (Immunomedics, Inc., NJ and Amgen, CA) which is an anti-CD22 antibody for the treatment of non-Hodgkin's lymphoma; and CEAcide (Immunomedics, NJ) which is a humanized anti-CEA antibody for the treatment of colorectal cancer. Other antibodies useful in the treatment of cancer include, but are not limited to, antibodies against the following antigens: CA125 (ovarian), CA15-3 (carcinomas), CA19-9 (carcinomas), L6 (carcinomas), Lewis Y (carcinomas), Lewis X (carcinomas), alpha fetoprotein (carcinomas), CA 242 (colorectal), alkaline placental phosphatase (carcinomas), prostate specific antigen (prostate), prostatic acid phosphatase (prostate), epidermal growth factor (carcinomas), MAGE-1 (carcinomas), MAGE-2 (carcinomas), MAGE-3 (carcinomas), MAGE-4 (carcinomas), anti-transferrin receptor (carcinomas), p97 (melanoma), MUCI-KLH (breast cancer), CEA (colorectal), gp 100 (melanoma), MARTI (melanoma), PSA (prostate), IL-2 receptor (T cell leukemia and lymphomas), CD20 (lymphoma not of Hodgkin), CD52 (leukemia), CD33 (leukemia), CD22 (lymphoma), human chorionic gonadotropin (carcinoma), CD39 (multiple myeloma), CD40 (lymphoma), mucin (carcinomas), P21 (carcinomas), MPG (melanoma) and oncogene product Neu (carcinomas). Some useful specific antibodies include, but are not limited to, BR96 mAb (Trail P. A., Willner, D. Lasen, SJ, Henderson, AJ, Hofstead, SJ, Casazza, AM, Firestone, RA, Hellstro, I., Hellstrom, KE, "Cure of Xenografted Human Carcinomas by BR96-Doxorubicin Immunoconjugates" Science 1993, 261, 212-215), BR64 (Trail PA, Willner, D., Knipe J., Henderson, AJ, Lasch, SJ, Zoeckler, ME, Trailsmith, MD, Doyle, TW, King, HD, Casazza AM, Braslawsky, GR, Brown J.
P., Hofstead, SJ, (Greenfield, RS, Firestone RA, Mosure K., Kadow DF, Yang MB, Hellstrom, KE, Hellstrom, I., "Effect of Linker Variation on the Stability, Potency and Efficacy of Carcinoma-reactive BR64-Doxorubicin Immunoconjugates "Cancer Research 1997, 57, 100-105, mAbs against the CD40 antigen, such as S2C6 mAb (Francisco, JA, Donaldson, KL, Chace, D., Siegall, CB, and Wahl, AF," Agonistic properties and in vivo antitumor activity of the anti-CD40 antibody, SGN-14"Cancer Res., 2000, 60, 3225-3231), mAbs against the CD70 antigen, such as 1F6 mAb and 2F2 Ab, and mAbs against the CD30 antigen , such as AC10 (Bowen, MA, Olsen, KJ, Cheng, L., Avila, D., and Podack, ER, "Functional effects of CD30 on a large granular lymphoma cell line YT", J. I munol., 151 , 5896-5906, 1993; Wahl et al., 2002 Cancer Res., 62 (13): 3736-42.) Many other internalizing antibodies that bind to tumor-associated antigens may be used and may be used. evised (Franke A.E., Sievers E.L., and Schienberg D.A., "Cell surface receptor-targeted therapy of acute myeloid leukemia: a review" Cancer Biother. Radiopharm. , 2000, 15, 459-76; Murray J. L., "Monoclonal antibody treatment of solid tumors: a coming of age" Semin Oncol., 2000, 27, 64-70; Breitling F., and Dubel S., Recombinant Antibodies, John Wiley and Sons, New York, 1998). In certain embodiments, the antibody is not Trastuzumab (full-length humanized anti-HER2 (MW 145167)), Herceptin F (ab ') 2 (derived from anti-HER2 enzymatically (MW 1000000)), 4D5 (murine anti-HER2 from total length, hybridoma), rhu 4D5 (full-length humanized antibody transiently expressed), rhu Fab 4D5 (recombinant humanized Fab (MW 47738)), 4D5 Fc8 (full-length murine anti-HER2, with mutated FcRn binding domain) , Hg (full-length humanized "4D5" without joint), with heavy chain articulated cysteines mutated to serines expressed in E. coli (consequently non-glycosylated) In another specific modality, the known antibodies for the treatment or prevention of a disease autoimmune antibodies are used according to the compositions and methods of the invention Immunospecific antibodies to an antigen of a cell responsible for the production of autoimmune antibodies can be obtained from any organization ( e.g., a university scientist or a company) or produced by any method known to the person skilled in the art such as, e.g., chemical synthesis or recombinant expression techniques. In another embodiment, useful antibodies that are immunospecific for the treatment of autoimmune diseases include, but are not limited to, anti-nuclear antibody; Anti-ds DNA, anti-ss DNA, anti-cardiolipin IgM antibody; anti-phospholipid antibody IgMm IgG; anti-SM antibody; anti-mitochondrial antibody; thyroid antibody; microsomal antibody; thyroglyphine antibody; Anti-SCL-70; anti-Jo; anti-U1RNP; anti-La / SSB; anti-SSA; anti-SSB; antibody perital cells; anti-histones; anti-RNP; C-ANCA; P-ANCA; anti-centomer; anti-fibrillarin and anti-GBM antibody. In certain embodiments, useful antibodies can bind to both a receptor and a receptor complex expressed on an activated lymphocyte. The receptor or receptor complex can comprise a member of the immunoglobulin gene superfamily, a member of the TNF receptor superfamily, an integrin, a cytosine receptor, a chemosin receptor, a major histocompatibility protein, a lectin, or a complement control. Non-limiting examples of suitable members of the immunoglobulin superfamily are CD2, CD3, CD4, CD8, CD19, CD22, CD28, CD79, CD152 &CTLA-4, PD-1, and ICOS. Non-limiting examples of suitable members of the TNF receptor superfamily are CD27, CD40, CD95 / Fas, CD134 / OX40, CD137 / 4-1BB, TNF-RI, TNFR-2, RANK, TACI, BCMA, osteoprotegerin, Apo2 / TRAIL-RI, TRAIL-R2, TRAIL-R3, TRAIL-R4 and APO-3. Non-limiting examples of suitable integrins are CDlla, CDllb, CDllc, CD18, CD29, CD41, CD49a, CD49c, CD49e, CD49e, CD49f, CD103 and CD104. Non-limiting examples of suitable lectins are type C lectin, type S, and type I. In one embodiment, the ligand binds to an activated lymphocyte that is associated with an immune disease. In another specific embodiment, the useful immunospecific ligands for a microbial antigen are monoclonal antibodies. The antibodies can be chimeric, humanized or human monoclonal antibodies. As used herein, the term "viral antigen" includes, but is not limited to, any viral peptide, polypeptide protein (eg, HIV gpl20, HIV nef, RSV F glycoprotein, influenza virus neuraminidase, hemagglutinin virus), influenza, HTLV tax, herpes simplex virus glycoprotein (eg, gB, gC, gD, and gE) and hepatitis B surface antigen that is capable of emitting 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 (eg, a bacterial, fungal, pathogenic protozoan polypeptide) , or yeast including, eg, LPS and capsular polysaccharide 5/8) that is capable of emitting an immune response. Immunospecific antibodies to a viral or microbial antigen can be obtained commercially, for example, from BD Biosciences (San Francisco, CA), Chemicon International, Inc., (Temecula, CA), or Vector Laboratories, Inc., (Burlingame, CA) , or produced by any method known to the person skilled in the art such as, eg, chemical synthesis or recombinant expression techniques. The nucleotide sequence encoding antibodies that are immunospecific for a viral or microbial antigen can be obtained, e. g., from the GenBank database or a similar database, literary publications or through routine cloning and sequencing. In a specific embodiment, useful ligands are those useful for the treatment or prevention of viral or microbial infection according to the methods described herein. Examples of available antibodies useful for the treatment of viral infection or microbial infection, include, but are not limited to, SUNAGIS (Medlmmune, Inc., MD) which is a humanized anti-respiratory syncytial virus (RSV) monoclonal antibody useful for the treatment of patients with RSV infection; PR0542 (Progenies) which is a CD4 fusion antibody useful for the treatment of HIV infection; OSTAVIR (Protein Design Labs., Inc., CA) which is a human antibody useful for the treatment of hepatitis B virus; PROTOVIR (Protein Design Labs., Inc., CA) which is a humanized IgGl antibody useful for the treatment of cytomegalovirus (CMV); and anti-LPS antibodies. Other antibodies useful in the treatment of infectious diseases include, but are not limited to, antibodies against the antigens of pathogenic bacteria species (Streptococcus pyogenes, Streptococcus pneumoniae, Neisseria gonorrheae, Neisseria meningitidis, Corynebacterium diphtheriae, Clostridium Botulinum, Clostridium perfingens, Clostriodium tetani, Hemophilus influenzae, Klebsiella pneumoniae, Klebsiella ozaenas, Klebsiella rhinoscleromotis, Staphylococcus aureus, Vibrio colerae, Escherichia coli, Pseudomonas aeruginosa, Campylobacter (Vibrio) fetus, Aeromonas hydrophila, Bacillus cereus, Edwardsiella tarda, Yersinia enterocolitica, Yersinia pestis, Yersinia pseudotuberculosis, Shigella dysenteriae, Shigella flexneri, Shigella sonnei, Salmonella typhimurium , Treponema pallidum, Treponema pertenue, Treponema carateneum, Borrelia vincentii, Borrelia burgdorferi, Leptospira icterohemorrhagiae, Mycobacterium tuberculosis, Pneumocystis carinii, Francisella turarensis, Brucella abortus, Brucella suis, Brucella melitensis, Mycoplasma spp, Rickettsia prowa zeki, Rickettsia tsutsugumishio, Chlamidia spp); pathogenic fungi (Coccidioides immitis, Aspergillus fumigatus, Candida albicans, Blastomyces dermatidis, Crytococcus neoformans, Histoplasma capsulatum); protozoa (Entamoeba histolytica, Toxoplasma gondii, Trichomonas tenas, Trichomonas hominis, Trichomonas vaginalis, Trypanosoma gambiense, Trypanosoma rhodesiense, Trypanosoma cruzi, Leishmania donovani, Leishmania tropic, Leishmania braziliensis, Pneumocystis pneumonia, Plasmodium vivax, Plasmodium falciparum, Plasmodium malaria); or Helminiths (Enterobius vermicualris, Trichuris trichiura, Ascaris lumbricoides, Trichinella spiralis, Strongyloides stercoralis, Schistosoma japonicum, Schistosoma mansonium, Schistosoma haematobium, and hookworms). Other antibodies useful in the invention for the treatment of viral disease include, but are not limited to, antibodies to antigens from pathogenic viruses, including as examples but not limited to: Poxviridae, Herpesviridae, Hepes Simplex virus 1, Herpes Simplex virus 2, Adenoviridae, Papoviridae, Enteroviridae, Picornaviridae, Parvoviridae, Reoviridae, Retroviridae, Influenza virus, Parainfluenza virus, Mumps virus, Measles virus, Respiratory syncytial virus, Rubella, Arboviridae, Rhabdoviridae, Arenaviridae, Hepatitis A virus, Hepatitis B virus, hepatitis C virus, hepatitis E virus, non-A / non-B hepatitis virus, Rhinoviridae, Coronaviridae, Rotoviridae, and human immunodeficiency virus. In attempts to discover effective cellular targets for cancer diagnosis and therapy, researchers have sought to identify transmembrane or other tumor-associated polypeptides that are specifically expressed on the surface of one or more particular types of cancer cells as compared to a or more normal non-cancerous cells. Frequently such tumor-associated polypeptides are more abundantly expressed on the surface of cancer cells as compared to on the surface of non-cancerous cells. The identification of such tumor-associated cell surface antigen polypeptides has resulted in the ability to specifically target cancer cells for destruction through antibody-based therapies. Antibodies comprising Ab in antibody-drug conjugates (ADCs) of the Formula I and which may be useful in the treatment of cancer include, but are not limited to, antibodies against tumor-associated antigens (TAA). Such tumor-associated antigens are known in the art, and can be prepared for use in the generation of antibodies using methods and information well known in the art. examples of TAA include (1) - (35), but are not limited to TAA (1) - (35) listed below. For convenience, information regarding these antigens, of which all are known in the art, are listed below and include names, alternative names, access numbers of the GenBank main references. The tumor-associated antigens directed by antibodies include all amino acid sequence variants and isoforms that possess at least about 70%, 80%, 85%, 90% or 95% sequence identity in relation to the sequences identified in the sequences corresponding ones (SEQ ID NOS: 1-35) or the sequences identified in the references cited. In some embodiments, TAAs having amino acid sequence variants exhibit substantially the same properties or biological characteristics as TAAs having the sequence found in the corresponding sequences listed (SEQ ID NOS: 1-35). For example, a TAA having a variant sequence is generally capable of specifically binding to an antibody that binds specifically to TAA with the corresponding sequence listed. The sequences and description specifically mentioned herein are expressly incorporated by reference. ANTIGENS ASSOCIATED WITH TUMOR (1) - (35): (1) BMPR1B (type IB bone morphogenetic protein receptor, GenBank accession number NM_001203.
Dijke, P., et al., Science 264 (5155).-101-104 (1994), Oncogene 14 (11): 1377-1382 (1997)); WO 200463362 (Claim 2); WO 2003042661 (Claim 12); US 2003134790-A1 (Page 38-39); WO 2002102235 (Claim 13: Page 296); WO 2003055443 (Page 91-92); WO 2002 99122 (Example 2; Page 528-530); WO 2003029421 (Claim 6; WO 2003024392 (Claim 2, Figure 112); WO 200298358 (Claim 1; Page 183); WO 200254940 (Page 100-101); WO200259377 (page 349-350); WO 200230268 (Claim 27; Page 376); WO 200148204 (Example; Fig. 4) NP_001194, type IB bone morphogenetic protein receptor / pid = NP_001194.1- cross-references: MIM: 603248; NP_001194.1; NM_001203_1 502 aa MLLRSAGKLNVGTKKEDGESTAPTPRPKVLRCKCHHHCPEDSVNNICSTDGYCFTMIEED DSGLPWTSGCLGLEGSDFQCRDTPIPHQRRSIECCTERNECNKDLHPTLPPLKNRDFVD GPIHHRALLISVTVCSLLLVLIILFCYFRYKRQETRPRYSIGLEQDETYIPPGESLRDLI EQSQSSGSGSGLPLLVQRTIAKQIQMVKQIGKGRYGEVWMGKWRGEKVAVKVFFTTEEAS WFRETEIYQTVLMRHENILGFIAADIKGTGSWTQLYLITDYHENGSLYDYLKSTTLDAKS MLKLAYSSVSGLCHLHTEIFSTQGKPAIAHRDLKSKNILVKKNGTCCIADLGLAVKFISD TNEVDIPPNTRVGTKRYMPPEVLDESLNRNHFQSYIMADMYSFGLILWEVARRCVSGGIV EEYQLPYHDLVPSDPSYEDMREIVCIKKLRPSFPNRWSSDECLRQMGKLMTECWAHNPAS RLTALRVKKTLAKMSESQDIKL (SEO ID NO: 1) (2) E16 (LATI, SLC7A5, Genbank Accession No.
NM_003486); Biochem. Biophys. Res. Commun. 255 (2), 283-288 (1999), Nature 395 (6699): 288-291 (1998), Gaugitsch, H.W., et al. (1992) J. Biol. Chem. 267 (16) .11267-11273); WO2004048938 (Example 2); WO2004032842 (Example IV); WO2003042661 (Claim 12); WO2003016475 (Claim 1); WO200278524 (Example 2); WO200299074 (Claim 19; Page 127-129); WO200286443 (Claim 27; Pages 222, 393); WO2003003906 (Claim 10; Page 293); WO200264798 (Claim 33; Page 93-95); WO200014228 (Claim 5; Page 133-136); US2003224454 (Figure 3); - WO2003025138 (Claim 12; Page 150); NP_003477 vehicle family of solute 7 (cationic amino acid transporter, and + system), member 5 / pid = NP_003477.3 - Homo sapiens Cross-references: MIM: 600182; NP_003477.3; NM_015923; NM_003486_1 507 aa MAGAGPKRRALAAPAAEEKEEAREKMLAAKSADGSAPAGEGEGVTLQRNITLLNGVAIIV GTIIGSGIFVTPTGVLKEAGSPGLALWWAACGVFSIVGALCYAELGTTISKSGGDYAYM LEVYGSLPAFLKLWI? LLIIRPSSQYIVALVFATYLLKPLFPTCPVPEEAAKLVACLCVL LLTAVNCYSVKAATRVQDAFAAAKLLALALIILLGFVQIGKGWSNLDPNFSFEGTKLDV GNIVLALYSGLFAYGGWNYLNFVTEEMINPYRNLPLAIIISLPIVTLVYVLTNLAYFTTL STEQMLSSEAVAVDFGNYHLGVMSWIIPVFVGLSCFGSVNGSLFTSSRLFFVGSREGHLP SILSMIHPQLLTPVPSLVFTCVMTLLYAFSKDIFSVINFFSFFNWLCVALAIIGMIWLRH RKPELERPIKVNLALPVFFILACLFLIAVSFWKTPVECGIGFTIILSGLPVYFFGVWWKU KPKWLLQGIFSTTVLCQKLMQWPQET (SEQ ID NO. 2) (3) ETAPAl (six transmembrane epithelial antigen of prostate, Genbank Accession No. NM_012449 Cancer Res. 61 (15), 5857-5860 (2001), Hubert, RS, et al (1999) Proc. Nati Acad. Sci. USES. 96 (25): 14523-14528) WO2004065577 (Claim 6); WO2004027049 (Figure 1L) EP1394274 (Example 11); WO2004016225 (Claim 2) WO2003042661 (Claim 12); US2003157089 (Example 5) US2003185830 (Example 5); US2003064397 (Figure 2) WO200289747 (Example 5; Page 618-619); WO2003022995 (Example 9; Figure 13A, Example 53; Page 173, Example 2; Figure 2A); NP__036581 six transmembrane epithelial antigens of the prostate Cross-references: MIM: 604415; NP_036581.1; NM_012449_1 339 aa MESRKDITNQEELWKMKPRRNLEEDDYLHKDTGETSMLKRPVLLHLHQTAHADEFDCPSE LQHTQELFPQWHLPIKIAAIIASLTFLYTLLREVIHPLATSHQQYFYKIPILVINKVLPM VSITLLALVYLPGVIAAIVQLHNGTKYKKFPHWLDKWMLTRKQFGLLSFFFAVLHAIYSL SYPMRRSYRYKLLNWAYQQVQQNKEDAWIEHDVWRMEIYVSLGIVGLAILALLAVTSIPS VSDSLTWREFHYIQSKLGIVSLLLGTIHALIFAWNKWIDIKQFVWYTPPTFMIAVFLPIV VLIFKSILFLPCLRKKILKIRHGWEDVTKINKTEICSQL (SEQ ID NO 3) (4) 0772P (CA125, MUC16, Genbank Access No. AF361486 J. Biol. Chem. 276 (29): 27371-27375 (2001)); WO2004045553 (Claim 14); WO200292836 (Claim 6; 12); WO200283866 (Claim 15; Page 116-121); US2003124140 (Example 16); US2003091580 (Claim 6); WO200206317 (Claim 6; Page 400-408); Cross references: GI: 34501467; AAK74120.3; AF361486_1 6995 aa PVTSLLTPGLVITTDRMGISREPGTSSTSNLSSTSHERLTTLEDTVDTEAMQPSTHTAVT NVRTSISGHESQSSVLSDSETPKATSPMGTTYTMGETSVSISTSDFFETSRIQIEPTSSL TSGLRETSSSERISSATEGSTVLSEVPSGATTEVSRTEVISSRGTSMSGPDQFTISPDIS TRAITRLSTSPIMTESAESAITIETGSPGATSEGTLTLDTSTTTFWSGTHSTASPGFSHS EMTTLMSRTPGDVPWPSLPSVEEASSVSSSLSSPAMTSTSFFSTLPESISSSPHPVTALL TLGPVKTTDMLRTSSEPETSSPPNLSSTSABILATSEVTKDREKIHPSSNTP, WNVGTVI YKHLSPSSVLADLVTTKPTSPMATTSTLGNTSVSTSTPAFPETMMTQPTSSLTSGLREIS TSQETSSATERSASLSGMPTGATTKVSRTEALSLGRTSTPGPAQSTISPEISTETITRIS TPLTTTGSAEMTITPKTGHSGASSQGTFTLDTSSPASWPGTHSAATHRSPHSGMTTPMSR GPEDVSWPSRPSVEKTSPPSSLVSLSAVTSPSPLYSTPSESSHSSPLRVTSLFTPVMMKT TDMLDTSLEPVTTSPPSMNITSDESLATSKATMETEAIQLSENTAVTQMGTISARQEFYS SYPGLPEPSKVTSPWTSSTIKDIVSTTIPASSEITRIEMESTSTLTPTPRETSTSQEIH SATKPSTVPYKALTSATIEDSMTQVMSSSRGPSPDQSTMSQDISTEVITRLSTSPIKTES TEMTITTQTGSPGATSRGTLTLDTSTTFMSGTHSTASQGFSHSQMTALMSRTPGEVPWLS HPSVEEASSASFSLSSPVMTSSSPVSSTLPDSIHSSSLPVTSLLTSGLVKTTELLGTSSE PETSSPPNLSSTSAEIl ^ TTEVTTDTEKLEMTNVVTSGYTHESPSSVLADSVTTKATSSM GITYPTGDTNVLTSTPAFSDTSRIQTKSKLSLTPGLMETSISEETSSATEKSTVLSSVPT GATTEVSRTEAISSSRTSIPGPAQ STMSSDTSMETITRISTPLTRKESTDMAITPKTGPS GATSQGTFTLDSSSTASWPGTHSATTQRFPRSWTTPMSRGPEDVSWPSPLSVEKNSPPS SLVSSSSVTSPSPLYSTPSGSSHSSPVPVTSLFTSIMMKATDMLDASLEPETTSAPNMNI TSDESLAASKATTETEAIHVFENTAASHVETTSATEELYSSSPGFSEPTKVISPWTSSS IRDNMVSTTMPGSSGITRIEIESMSSLTPGLRETRTSQDITSSTETSTVLYKMPSGATPE VSRTEVMPSSRTSIPGPAQSTMSLDISDEWTRLSTSPIMTESABITITTQTGYSLATSQ VTLPLGTSMTFLSGTHSTMSQGLSHSEMTNLMSRGPESLSWTSPRFVETTRSSSSLTSLP LTTSLSPVSSTLLDSSPSSPLPVTSLILPGLVKTTEVLDTSSEPKTSSSPNLSSTSVEIP ATSEIMTDTEKIHPSSNTAVAKVRTSSSVHESHSSVLADSETTITIPSMGITSAVEDTTV FTSNPAPSETRRIPTEPTFSLTPGFRETSTSEETTSITETSAVLFGVPTSATTEVSMTEI MSSNRTHIPDSDQSTMSPDIITEVITRLSSSSMMSESTQMTITTQKSSPGATAQSTLTLA TTTAPLARTHSTVPPRFLHSEMTTLMSRSPENPSWKSSPFVEKTSSSSSLLSLPVTTSPS VSSTLPQSIPSSSFSVTSLLTPGMVKTTDTSTEPGTSLSPNLSGTSVEILAASEVTTDTE KIHPSSSMAVTNVGTTSSGHELYSSVSIHSEPSKATYPVGTPSSMAETSISTSMPANFET TGFEAEPFSHLTSGLRKTNMSLDTSSVTPTNTPSSPGSTHLLQSSKTDFTSSAKTSSPDW PPASQYTEIPVDIITPFNASPSITESTGITSFPESRFTMSVTESTHHLSTDLLPSAETIS TGTVMPSLSEAMTSFATTGVPRAISGSGSPFSPTESGPGDATLSTIAESL PSSTPVPFSS STFTTTDSSTIPALHEITSSSATPYRVDTSLGTESSTTEGRLVMVSTLDTSSQPGRTSSS PILDTRMTESVELGTVTSAYQVPSLSTRLTRTDGIMEHITKIPNEAAHRGTIRPVKGPQT STSPASPKGLHTGGTKRMETTTTALKTTTTALKTTSRATLTTSVYTPTLGTLTPLNASMQ MASTIPTEMMITTPYVFPDVPETTSSLATSLGAETSTALPRTTPSVFNRESETTASLVSR SGAERSPVIQTLDVSSSEPDTTASWVIRPASTIPTVSKTTPNFFHSELDTVSSTATSHGA DVSSAIPTNISPSELDALTPLVTISGTDTSTTFPTLTKSPHETETRTTWLTHPAETSSTI PRTIPNFSHHESDATPSIATSPGAETSSAIPIMTVSPGAEDLVTSQVTSSGTDRNMTIPT LTLSPGEPKTIASLVTHPEAQTSSAIPTSTISPAVSRLVTSMVTSLAAKTSTTNRALTNS PGEPATTVSLVTHSAQTSPTVPWTTSIFFHSKSDTTPSMTTSHGAESSSAVPTPTVSTEV PGWTPLVTSSRAVISTTIPILTLSPGEPETTPSMATSHGEEASSAIPTPTVSPGVPGW TSLVTSSRAVTSTTIPILTFSLGEPETTPSMATSHGTEAGSAVPTVLPEVPGMVTSLVAS SRAVTSTTLPTLTLSPGEPETTPSMATSHGAEASSTVPTVSPEVPGWTSLVTSSSGVNS TSIPTLILSPGELETTPSMATSHGAEASSAVPTPTVSPGVSGWTPLVTSSRAVTSTTIP ILTLSSSEPETTPSMATSHGVEASSAVLTVSPEVPGMVTFLVTSSRAVTSTTIPTLTISS DEPETTTSLVTHSEAKMISAIPTLGVSPTVQGLVTSLVTSSGSETSAFSNLTVASSQPET IDSWVAHPGTFASSWPTLTVSTGEPFTNISLVTHPAESSSTLPRTTSRFSHSELDTMPS TVTSPEAESSSAISTTIS PGIPGVLTSLVTSSGRDISATFPTVPESPHESEATASWVTHP AVTSTTVPRTTPNYSHSEPDTTPSIATSPGAEATSDFPTITVSPDVPDMVTSQVTSSGTD TSITIPTLTLSSGEPETTTSFITYSETHTSSAIPTLPVSPDASKMLTSLVISSGTDSTTT FPTLTETPYEPETTAIQLIHPAETNTMVPRTTPKFSHSKSDTTLPVAITSPGPEASSAVS TTTISPDMSDLVTSLVPSSGTDTSTTFPTLSETPYEPETTATWLTHPAETSTTVSGTIPN FSHRGSDTAPSMVTSPGVDTRSGVPTTTIPPSIPGWTSQVTSSATDTSTAIPTLTPSPG EPETTASSATHPGTQTGFTVPIRTVPSSEPDTMASWVTHPPQTSTPVSRTTSSFSHSSPD ATPVMATSPRTEASSAVLTTISPGAPEMVTSQITSSGAATSTTVPTLTHSPGMPETTALL STHPRTETSKTFPASTVFPQVSETTASLTIRPGAETSTALPTQTTSSLFTLLVTGTSRVD LSPTASPGVSAKTAPLSTHPGTETSTMIPTSTLSLGLLETTGLLATSSSAETSTSTLTLT VSPAVSGLSSASITTDKPQTVTSWNTETSPSVTSVGPPEFSRTVTGTTMTLIPSEMPTPP KTSHGEGVSPTTILRTTMVEATNLATTGSSPTVAKTTTTFNTLAGSLFTPLTTPGMSTLA SESVTSRTSYNHRSWISTTSSYNRRYWTPATSTPVTSTFSPGISTSSIPSSTAATVPFMV PFTLNFTITNLQYEEDMRHPGSRKFNATERELQGLLKPLFRNSSLEYLYSGCRLASLRPE KDSSATAVDAICTHRPDPEDLGLDRERLYWELSNLTNGIQELGPYTLDRNSLYVNGFTHR SSMPTTSTPGTSTVDVGTSGTPSSSPSPTTAGPLLMPFTLNFTITNLQYEEDMRRTGSRK FNTMESVLQGLLKPLFKNTSVGPLYSGCRLTLLRPEKDGAATGVDAICTHRLDPKSPGLN REQLYWELSKLTNDIEELGPYTLDRNSLYVNGFTHQSSVSTTSTPGTSTVDLRTSGTPSS LSSPTIMAAGPLLVPFTLNFTITNLQYGEDMGHPGSRKFNTTERVLQGLLGPIFKNTSVG PLYSGCRLTSLRSEKDGAATGVDAICIHHLDPKSPGLNRERLYWELSQLTNGIKELGPYT LDRNSLYVNGFTHRTSVPTTSTPGTSTVDLGTSGTPFSLPSPATAGPLLVLFTLNFTITN LKYEEDMHRPGSRKFNTTERVLQTLVGPMFKNTSVGLLYSGCRLTLLRSEKDGAATGVDA ICTHRLDPKSPGVDREQLYWELSQLTNGIKELGPYTLDRNSLYVNGFTHWIPVPTSSTPG TSTVDLGSGTPSSLPSPTSATAGPLLVPFTLNFTITNLKYEEDMHCPGSRKFNTTERVLQ SLLGPMFKNTSVGPLYSGCRLTLLRSEKDGAATGVDAICTHRLDPKSPGVDREQLYWELS QLTNGIKELGPYTLDRNSLYVNGPTHQTSAPNTSTPGTSTVDLGTSGTPSSLPSPTSAGP LLVPFTLNFTITNLQYEEDMHHPGSRKFNTTERVLQGLLGPMFKNTSVGLLYSGCRLTLL RPEKNGAATGMDAICSHRLDPKSPGLNREQLYWBLSQLTHGIKELGPYTLDRNSLYVNGF THRSSVAPTSTPGTSTVDLGTSGTPSSLPSPTTAVPLLVPFTLNFTITNLQYGEDMRHPG SRKFNTTERVLQGLLGPLFKNSS VGPLYSGCRLISLRSEKDGAATGVDAICTHHLNPQSP GLDREQLYWQLSQMTNGIKELGPYTLDRNSLYVNGFTHRSSGLTTSTPWTSTVDLGTSGT PSPVPSPTTAGPLLVPFTLNFTITNLQYEEDMHRPGSRKFNATERVLQGLLSPIFKNSSV GPLYSGCRLTSLRPEKDGAATGMDAVCLYHPNPKRPGLDREQLYWELSQLTRNITELGPY SLDRDSLYVNGFTHQNSVPTTSTPGTSTVYWATTGTPSSFPGHTEPGPLLIPFTFNFTIT NLHYEENMQHPGSRKFNTTERVLQGLLKPLFXNTSVGPLYSGCRLTLLRPEKQEAATGVD TICTHRVDPIGPGLDRERLYWELSQLTNSITELGPYTLDRDSLYVNGFNPWSSVPTTSTP GTSTVHLATSGTPSSLPGHTAPVPLLIPFTLNFTITNLHYEENMQHPGSRKFNTTERVLQ GLLKPLFKSTSVGPLYSGCRLTLLRPEKHGAATGVDAICTLRLDPTGPGLDRERLYWELS QLTNSVTELGPYTLDRDSLYVNGFTHRSSVPTTSIPGTSAVHLETSGTPASLPGHTAPGP LLVPFTLNFTITNLQYEEDMRHPGSRKFNTTERVLQGLLKPLFKSTSVGPLYSGCRLTLL RPEKRGAATGVDTICTHRLDPLNPGLDREQLYWELSKLTRGIIELGPYLLDRGSLYVNGF THRNFVPITSTPGTSTVHLGTSETPSSLPRPIVPGPLLVPFTLNFTITNLQYEEAMRHPG SRKFNTTERVLQGLLRPLFKNTSIGPLYSSCRLTLLRPEKDKAATRVDAICTHHPDPQSP GLNREQLYWELSQLTHGITELGPYTLDRDSLYVDGFTHWSPIPTTSTPGTSIVNLGTSGI PPSLPETTATGPLLVPFTLNFTTTNLQYEENMGHPGSRKFNIT? SVLQGLLKPLFKSTSV GPLYSGCRLTLLRPEKDGVATRVDAICTHRPDPKIPGLDRQQLYWEL SQLTHSITELGPY TLDRDSLYVNGFTQRSSVPTTSTPGTFTVQPETSETPSSLPGPTATGPVLLPFTLNFTII NLQYEEDMHRPGSRKFNTTERVLQGLLMPLFKNTSVSSLYSGCRLTLLRPEKDGAATRVD AVCTHRPDPKSPGLDRERLYWKLSQLTHGITELGPYTLDRHSLYVNGFTHQSSMTTTRTP DTSTMHLATSRTPASLSGPTTASPLLVLFTINFTITNLRYEENMHHPGSRKFNTTERVLQ GLLRPVFKNTSVGPLYSGCRLTLLRPKKDGAATKVDAICTYRPDPKSPGLDREQLYWELS QLTHSITELGPYTLDRDSLYVNGFTQRSSVPTTSIPGTPTVDLGTSGTPVSKPGPSAASP LLVLFTLNFTITNLRYEENMQHPGSRKFNTTERVLQGLLRSLFKSTSVGPLYSGCRLTLL RPEKDGTATGVDAICTHHPDPKSPRLDREQLYWELSQLTHNITELGPYALDNDSLFVNGF THRSSVSTTSTPGTPTVYLGASKTPASIFGPSAASHLLILFTLNFTITNLRYEENMWPGS RKFNTTERVLQGLLRPLFKNTSVGPLYSGCRLTLLRPEKDGEATGVDAICTHRPDPTGPG LDREQLYLELSQLTHSITELGPYTLDRDSLYVNGFTHRSSVPTTSTGWSEEPFTLNFTI NNLRYMADMGQPGSLKFNITDNVMQHLLSPLFQRSSLGARYTGCRVIALRSVKNGAETRV DLLCTYLQPLSGPGLPIKQVFHELSQQTHGITRLGPYSLDKDSLYLNGYNEPGPDEPPTT PKPATTFLPPLSFATTAMGYHLKTLTLNFTISNLQYSPDMGKGSATFNSTEGVLQHLLRP LFQKSSMGPFYLGCQLISLRPEKDGAATGVDTTCTYHPDPVGPGLDIQQLYWELSQLTHG VTQLGFYVLDRDSLFINGYAPQNLSIRGEYQINFHIVNWNLSNPDPTSSEYITLLRDIQD KVTTLYKGSQ LHDTFRFCLVTNLTMDSVLVTVKALFSSNLDPSLVEQVFLDKTLNASFHW LGSTYQLVDIHVTEMESSVYQPTSSSSTQHFYLNFTITNLPYSQDKAQPGTTNYQRNKRN IEDALNQLFRNSSIKSYFSDCQVSTFRSVPNRHHTGVDSLCNFSPLARRVDRVAIYEEFL RMTRNGTQLQNFTLDRSSVLVDGYSPNRNEPLTGNSDLPFWAVTLIGLAGLLGLITCLIC GVLVTTRRRKKEGEYNVQQQCPGYYQSHLDLEDLQ (SEQ ID NO.4) (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancement factor, mesothelin, Genbank Access No. NM_005823 Yamaguchi, N., et al., Biol. Chem. 269 (2), 805-808 (1994), Proc. Nati Acad. Sci. USA.96 (20): 11531-11536 (1999), Proc. Nati Acad. Sci. USA.93 (1): 136-140 (1996), J. Biol. Chem. 270 (37): 21984-21990 (1995)); WO2003101283 (Claim 14); (WO2002102235 (Claim 13; Page 287-298); WO2002101075 (Claim 4; Page 308-309);, WO200271928 (Page 320-321); WO9410312 (Page 52-57); Cross references: MIM: 601051; NP 005814.2; NM 005823 1 - - 622 aa MALPTARPLLGSCGTPALGSLLFLLFSLGWVQPSRTLAGETGQEAAPLDGVLANPPNISS LSPRQLLGFPCAEVSGLSTERVRELAVALAQKNVKLSTEQLRCLAHRLSEPPEDLDALPL DLLLFLNPDAFSGPQACTRFPSRITKANVDLLPRGAPERQRLLPAALACWGVRGSLLSEA DVRALGGLACDLPGRFVAESAEVLLPRLVSCPGPLDQDQQEAARAALQGGGPPYGPPSTW SVSTMDALRGLLPVLGQPIIRSIPQGIVAAWRQRSSRDPSWRQPERTILRPRFRREVEKT ACPSGKKAREIDESLIFYKKWELEACVDAALLATQMDRVNAIPFTYEQLDVLKHKLDELY PQGYPESVIQHLGYLFLKMSPEDIRKWNVTSLETLKALLEVNKGHEMSPQVATLIDRFVK GRGQLDKDTLDTLTAFYPGYLCSLSPEELSSVPPSSIWAVRPQDLDTCDPRQLDVLYPKA RLAFQNMNGSEYFVKIQSFLGGAPTEDLKALSQQNVSMDLATFMKLRTDAVLPLTVAEVQ KLLGPHVEGLKARERHRPVRDWILRQRQDDLDTLGLGLQGGIPNGYLVLDLSMQEALSGT PCLLGPGPVLTVLALLLASTLA (SEQ ID NO: 5) (6) Napi3b (NAPI-3B, NPTIIb, Slc34a2, family vehicle solute 34 (sodium phosphate), member 2, conveyor sodium phosphate 3b type II, Genbank Access No. NM_006424, J. Biol. Chem. 277 (22): 19665-19672 (2002), Genomics 62 (2): 281-284 (1999), Feild, JA, et. al. (1999) Biochem. Biophys. Res. Commun. 258 (3): 578- 582); WO2004022778 (Claim 2); EP1394274 (Example 11); WO2002102235 (Claim 13; Page 326); EP875569 (Claim 1; Page 17-19); WO200157188 (Claim 20; Page 329); WO2004032842 (Example IV); WO200175177 (Claim 24; Page 139-140); Cross-references: MIM: 604217; NP 006415.1; NM 006424 1 690 aa MAPWPELGDAQPNPDKYLEGAAGQQPTAPDKSKETNKTDNTFAPVTKIELLPSYSTATLI DEPTEVDDPWNLPTLQDSGIKWSERDTKGKILCFFQGIGRLILLLGFLYFFVCSLDILSS AFQLVGGKMAGQFFSNSSIMSNPLLGLVIGVLVTVLVQSSSTSTSIWSMVSSSLLTVRA AIPIIMGANIGTSITNTIVALMQVGDRSEFRRAFAGATVHDFFNWLSVLVLLPVEVATHY LEIITQLIVESFHFKNGEDAPDLLKVITKPFTKLIVQLDKKVISQIAMNDEKAKNKSLVK IWCKTFTNKTQINVTVPSTANCTSPSLCWTDGIQNWTMKNVTYKENIAKCQHIFVNFHLP DLAVGTILLILSLLVLCGCLIMIVKILGSVLKGQVATVIKKTINTDFPFPFAWLTGYLAI LVGAGMTFIVQSSSVFTSALTPLIGIGVITIERAYPLTLGSNIGTTTTAILAALASPGNA LRSSLQIALCHFFFNISGILLWYPIPFTRLPIRMAKGLGNISAKYRWFAVFYLIIFFFLI PLTVFGLSLAGWRVLVGVGVPWFIIILVLCLRLLQSRCPRVLPKKLQNWNFLPLWMRSL KPWDAWSKFTGCFQMRCCYCCRVCCRACCLLCGCPKCCRCSKCCEDLERAQEGQDVPVK APETFDNITISREAQGEVPASDSKTECTAL (SEQ ID NO: 6) (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaforin 5b Hlog, sema domain, seven repeats of thrombospondin (type 1 and similar to type 1), transmembrane domain (TM) and cytoplasmic domain or, (semaphorin) 5B, Genbank Access No. ABO40878, Nagase T., et al (2000) DNA Res. 7 (2) .143-150); WO2004000997 (Claim 1); WO2003003984 (Claim 1); WO200206339 (Claim 1; Page 50); WO200188133 (Claim 1; Page 41-43, 48-58); WO2003054152 (Claim 20); WO2003101400 (Claim 11); Access: Q9P283; EMBL; AB040878; BAA95969.1.Genew; HGNC: 10737; 1093 TYPGARDFSQLALDPSGNQLIVGARNYLFRLSLANVSLLQATEWASSEDTRRSCQSKGKT aa MVLAGPLAVSLLLPSLTLLVSHLSSSQDVSSEPSSEQQLCALSKHPTVAFEDLQPWVSNF EEECQNYVRVLIVAGRKVFMCGTNAFSPMCTSRQVGNLSRTTEKINGVARCPYDPRHNST AVISSQGELYAATVIDFSGRDPAIYRSLGSGPPLRTAQYNSKWLNEPNFVAAYDIGLFAY FFLRENAVEHDCGRTVYSRVARVCKNDVGGRFLLEDTWTTFMKARLNCSRPGEVPFYYNE LQSAFHLPEQDLIYGVFTTNVNSIAASAVCAFNLSAISQAFNGPFRYQENPRAAWLPIAN PIPNFQCGTLPETGPNENLTERSLQDAQRLFLMSEAVQPVTPEPCVTQDSVRFSHLWDL VQAKDTLYHVLYIGTESGTILKALSTASRSLHGCYLEELHVLPPGRREPLRSLRILHSAR ALFVGLRDGVLRVPLERCAAYRSQGACLGARDPYCGWDGKQQRCSTLEDSSNMSLWTQNI TACPVRNVTRDGGFGPWSPWQPCEHLDGDNSGSCLCRARSCDSPRPRCGGLDCLGPAIHI ANCSRNGAWTPWSSWALCSTSCGIGFQVRQRSCSNPAPRHGGRICVGKSREERFCNENTP CPVPIFWASWGSWSKCSSNCGGGMQSRRRACENGNSCLGCGVEFKTCNPEGCPEVRRNTP WTPWLPVNVTQGGARQEQRFRPTCRAPLADPHGLQFGRRRTETRTCPADGSGSCDTDALV EDLLRSGSTSPHTVSGGWAAWGPWSSCSRDCELGFRVRKRTCTNPEPRNGGLPCVGDAAE YQDCNPQACPVRGAWSCWTSWSPCSASCGGGHYQRTRSCTSPAPSPGEDICLGLHTEEAL CATQACPEGWSPWSEWSKCTDDGAQSRSRHCEELLPGSSACAGNSSQSRPCPYSEIPVIL PASSMEEATGCAGFNL IHLVATGISCFLGSGLLTLAVYLSCQHCQRQSQESTLVHPATPN HLHYKGGGTPKNEKYTPMEFKTLNKNNLIPDDRANFYPLQQTNVYTTTYYPSPLNKHSFR PEASPGQRCFPNS (SEQ ID NO: 7) (8) PSCA hlg (2700050C12Rik, C530008O16RÍk, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Access Genbank No. AY358628); US2003129192 (Claim 2); US2004044180 (Claim 12); US2004044179 (Claim 11); US2003096961 (Claim 11); US2003232056 (Example 5); W02003105758 (Claim 12); US2003206918 (Example 5); EP1347046 (Claim 1); W02003025148 (Claim 20); Cross references: GI: 37182378; AAQ88991.1; AY358628_1 141 aa MWVLGIAATPCGLFLLPGFALQIQCYQCEEFQLNNDCSSPEFIVNCTVNVQDMCQKEVME QSAGIMYRKSCASSAACLIASAGYQSFCSPGKLNSVCISCCNTPLCNGPRPKKRGSSASA LRPGLRTTILFLKLALFSAHC (SEQ ID NO: 8) (9) ETBR (endothelin type B receptor, Genbank Access No.
AY275463); Nakamuta M., et al. Biochem. Biophys. Res. Commun. 177, 34-39, 1991; Ogawa Y., et al. Biochem. Biophys. Res. Commun. 178, 248-255, 1991; Arai H., et al. Jpn. Circ. J. 56, 1303-1307, 1992; Arai H., et al J. Biol. Chem. 268, 3463-3470, 1993; Sakamoto A., Yanagisawa M., et al Biochem. Biophys. Res. Commun. 178, 656-663, 1991; Elshourbagy N.A. , et al J.
Biol. Chem. 268, 3873-3879, 1993; Haendler B., et al. J.
Cardiovasc. Pharmacol. 20, sl-S4, 1992; Tsutsumi M., et al Gene 228, 43-49, 1999; Strausberg R.L., et al. , Proc. Nati Acad. Sci. USA. 99, 16899-16903, 2002; Bourgeois C, et al. J. Clin. Endocrinol Metab. 82, 3116-3123, 1997; Okamoto Y., et al Biol. Chem. 272, 21589-21596, 1997; Verheij J.B., et al Am. J. Med. Genet. 108, 223-225, 2002; Hofstra R.M.W. , et al. Eur. J. Hum. Genet 5, 180-185, 1997; Puffenberger E. G., et al Cell 79, 1257-1266, 1994; Attie T., et al, Hum. Mol. Genet 4, 2407-2409, 1995; Auricchio A., et al Hum. Mol. Genet 5: 351-354, 1996; Amiel J., et al Hum. Mol. Genet 5, 355-357, 1996; Hofstra R.M.W. , et al Nat. Genet. 12, 445-447, 1996; Svensson P.J., et al Hum. Genet 103, 145-148, 1998; Fuchs S., et al Mol. Med. 7, 115-124, 2001; Pingault V., et al. (2002) Hum. Genet 111, 198-206; WO2004045516 (Claim 1); W02004048938 (Example 2); WO2004040000 (Claim 151); WO2003087768 (Claim 1); WO2003016475 (Claim 1); WO2003016475 (Claim 1); WO200261087 (Figure 1); WO2003016494 (Figure 6); WO2003025138 (Claim 12; Page 144); WO200198351 (Claim 1; Page 124-125); EP522868 (Claim 8; Figure 2); WO200177172 (Claim 1; Page 297-299); US2003109676; US6518404 (Figure 3); US5773223 (Claim la; Col 31-34); WO2004001004; 442 aa MQPPPSLCGRALVALVLACGLSRIWGEERGFPPDRATPLLQTAEIMTPPTKTLWPKGSNA SLARSLAPAEVPKGDRTAGSPPRTISPPPCQGPIEIKETFKYINTWSCLVFVLGIIGNS TLLRIIYKNKCMRNGPNILIASLALGDLLHIVIDIPINVYKLLAEDWPFGAEMCKLVPFI QKASVGITVLSLCALSIDRYRAVASWSRIKGIGVPKWTAVEIVLIWWSWLAVPEAIGF DIITMDYKGSYLRICLLHPVQKTAFMQFYKTAKDWWLFSFYFCLPLAITAFFYTLMTCEM LRKKSGMQIALNDHLKQRREVAKTVFCLVLVFALCWLPLHLSRILKLTLYNQNDPNRCEL LSPLLVLDYIGINMASLNSCINPIALYLVSKRFKNCFKSCLCCWCQSFEEKQSLEEKQSC LKFKANDHGYDNFRSSNKYSSS (SEQ ID NO.9) (10) .MSG783 (RNF124, hypothetical protein FLJ20315, Genbank Accession No NM_017763.); WO2003104275 (Claim 1); WO2004046342 (Example 2); WO2003042661 (Claim 12); WO2003083074 (Claim 14; Page 61); WO2003018621 (Claim 1); WO2003024392 (Claim 2, Figure 93); WO200166689 (Example 6); Cross references: Locus ID: 54894; NP_060233.2; NM_017763_1 783 aa MSGGHQLQLAALWPWLLMATLQAGFGRTGLVLAAAVESERSAEQKAIIRVIPLKMDPTGK LNLTLEGVFAGVAEITPAEGKLMQSHPLYLCNASDDDNLEPGFISIVKLESPRRAPRPCL SLASKARMAGERGASAVLFDITEDRAAAEQLQQPLGLTWPWLIWGNDAEKLMEFVYKNQ KAHVRIELKEPPAWPDYDVWILMTWGTIFVIILASVLRIRCRPRHSRPDPLQQRTAWAI SQLATRRYQASCRQARGEWPDSGSSCSSAPVCAICLEEFSEGQELRVISCLHEFHRNCVD PWLHQHRTCPLCVFNITEGDSFSQSLGPSRSYQEPGRRLHLIRQHPGHAHYHLPAAYLLG PSRSAVARPPRPGPFLPSQEPGMGPRHHRFPRAAHPRAPGEQQRLAGAQHPYAQGWGMSH LQSTSQHPAACPVPLRRARPPDSSGSGESYCTERSGYLADGPASDSSSGPCHGSSSDSW NCTDISLQGVHGSSSTFCSSLSSDFDPLVYCSPKGDPQRVDMQPSVTSRPRSLDSWPTG ETQVSSHVHYHRHRHHHYKKRFQWHGRKPGPETGVPQSRPPIPRTQPQPEPPSPDQQVTG SNSAAPSGRLSNPQCPRALPEPAPGPVDASSICPSTSSLFNLQKSSLSARHPQRKRRGGP SEPTPGSRPQDATVHPACQIFPHYTPSVAYPWSPEAHPLICGPPGLDKRLLPETPGPCYS NSQPVWLCLTPRQPLEPHPPGEGPSEWSSDTAEGRPCPYPHCQVLSAQPGSEEELEELCE QAV (SEQ ID NO: 10) (11) STAGE2 (HGNC_8639, IPCA-1, PCANAP1, ESTAMPA1, STAGE2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, six transmembrane epithelial prostate 2 antigens, six prostate proteins Transmembrane, Access Genbank No. AF455138, Lab. Invest. 82 (11): 1573-1582 (2002)); WO2003087306; US2003064397 (Claim 1, Figure 1), WO200272596 (Claim 13; Page 54-55); WO200172962 (Claim 1, Figure 4B); WO2003104270 (Claim eleven); WO2003104270 (Claim 16); US2004005598 (Claim 22); WO2003042661 (Claim 12); US2003060612 (Claim 12, Figure 10); WO200226822 (Claim 23, Figure 2); WO200216429 (Claim 12; Figure 10); Cross references: GI: 22655488; AAN04080.1; AF455138_1 490 aa MESISMMGSPKSLSETVLPNGINGIKDARKVTVGVIGSGDFAKSLTIRLIRCGYHWIGS RNPKFASEFFPHWDVTHHEDALTKTNIIFVAIHREHYTSLWDLRHLLVGKILIDVSNNM RINQYPESNAEYLASLFPDSLIVKGFNWSAWALQLGPKDASRQ "VYICSNNIQARQQVIE LARQLNFIPIDLGSLSSAREIENLPLRLFTLWRGPVWAISLATFFFLYSFVRDVIHPYA RNQQSDFYKIPIEIVNKTLPIVAITLLSLVYLAGLLAAAYQLYYGTKYRRFPPWLETWLQ CRKQLGLLSFFFAMVHVAYSLCLPMRRSERYLFLNMAYQQVHANIENSWNEEEVWRIEMY ISFGIMSLGLLSLLAVTSIPSVSNALNWREFSFIQSTLGYVALLISTFHVLIYGWKRAFE EEYYRFYTPPNFV1ALVLPSIVILGKIILFLPCISQKLKRIKKGWEKSQFLEEGIGGTIP HVSPERVTVM (SEQ ID NO: 11) 5 (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, potential cation channel of the transient receptor, subfamily M, member 4, Genbank Access No. NM_017636 Xu, XZ, et al., Proc. Nati, Acad. Sci. USA, 98 (19) -, 10692-10697 (2001), Cell 109 (3): 397-407 (2002), J. Biol. Chem. 278 0 (33) -.30813-30820 (2003)); US2003143557 (Claim 4); WO200040614 (Claim 14; Page 100-103); WO200210382 (Claim 1, Figure 9A); WO2003042661 (Claim 12); WO200230268 (Claim 27; Page 391); US2003219806 (Claim 4); WO200162794 (Claim 5 14; Figure 1A-D); Cross references: MIM: 06936; NP_060106.2; NM_017636_1 1214 MWPEKEQSWIPKIFKKKTCTTFIVDSTDPGGTLCQCGRPRTAHPAVAMEDAFGAAWTV aa WDSDAHTTEKPTDAYGELDFTGAGRKHSNFLRLSDRTDPAAV? SLVTRTWGFRAPNLWS or VLGGSGGPVLQTWLQDLLRRGLVRAAQSTGAWIVTGGLHTGIGRHVGVAVRDHQMASTGG TKWAMGVAPWGWRNRDTLINPKGSFPARYRWRGDPEDGVQFPLDYNYSAFFLVDDGTH GCLGGENRFRLRLESYISQQKTGVGGTGIDIPVLLLLIDGDEKMLTRIENATQAQLPCLL VAGSGGAADCLAETLEDTLAPGSGGARQGEARDRIRRFFPKGDLEVLQAQVERIMTRKEL LTVYSSEDGSEEFETIVLK? LVKACGSSEASAYLDELRLAVAWNRVDIAQSELFRGDIQW RSFHLEASLMDALLNDRPEFVRLLISHGLSLGHFLTPMRLAQLYSAAPSNSLIRNLLDQA SHSAGTKAPALKGGAAELRPPDVGHVLRMLLGKMCAPRYPSGGAWDPHPGQGFGESMYLL SDKATSPLSLDAGLGQAPWSDLLLWALLLNRAQMAMYFWEMGSNAVSSALGACLLLRVMA RLEPDAEEAARRKDLAFKFEGMGVDLFGECYRSSEVRAARLLLRRCPLWGDATCLQIÍAMQ ADARAFFAQDGVQSLLTQKWWGDM? STTPIWALVLAFFCPPLIYTRLITFRKSEEEPTRE ELEFDMDSVINGEGPVGTADPAEKTPLGVPRQSGRPGCCGGRCGGRRCLRRWFHFWGAPV TIPMGNWSYLLFLLLFSRVLLVDFQPAPPGSLELLLYFWAFTLLCEELRQGLSGGGGSL ASGGPGPGRASLSQRLRLYLADSWNQCDLVALTCFLLGVGCRLTPGLYHLGRTVLCIDFM VFTVRLLHIFTVNKQLGPKIVIVSKMMKDVFFFLFFLGVWLVAYGVATEGLLRPRDSDFP SILRRVFYRPYLQIFGQIPQEDMDVALMEHSNCSSEPGFWAHPPGAQAGTCVSQYANWLV VLLLVIFLLVANILLVNLLIAMFSYTFGKVQGNSDLYWKAQRYRLIREFHSRPALAPPFI VISHLRLLLRQLCRRPRSPQPSSPALEHFRVYLSKFAERKLLTWESVHKENFLLARARDK RESDSERLKRTSQKVDLALKQLGHIREYEQRLKVLEREVQQCSRVLGWVARALSRSALLP PGGPPPPDLPGSKD (SEQ ID NO: 12) (13) CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, derived growth factor teratocarcinoma, Genbank Accession No.
NP_003203 or NM_003212, Ciccodicola, A., et al., EMBO J. 8 (7): 1987-1991 (1989), Am.
J. Hum. Genet 49 (3): 555-565 (1991)); US2003224411 (Claim 1); WO2003083041 (Example 1); WO2003034984 (Claim 12); WO200288170 (Claim 2; Page 52-53); WO2003024392 (Claim 2, Figure 58); WO200216413 (Claim 1; Page 94-95, 105); WO200222808 (Claim 2, Figure 1); US5854399 (Example 2; Col 17-18); US5792616 (Figure 2); Cross-references: MIM: 187395; NP_003203.1; NM_003212_1 188 aa MDCRKMARFSYSVIWIMAISKVPELGLVAGLGHQEFARPSRGYLAFRDDSIWPQEEPAIR PRSSQRVPPMGIQHSKELNRTCCLNGGTCMLGSFCACPPSFYGRNCEHDVRKENCGSVPH DTWLPKKCSLCKCWHGQLRCFPQAFLPGCDGLVMDEHLVASRTPELPPSARTTTFMLVGI CLSIQSYY (SEQ ID NO: 13) (14) CD21 (CR2 (Complement receptor 2) or C3DR (C3d / receiver Epstein Barr virus) or Hs.73792 Genbank Access No. M26004, Fujisaku et al (1989) J. Biol. Chem. 264 (4): 2118-2125); Weis J.J.,. et al. , J. Exp. Med. 167.1047-1066, 1988, Moore M., et al. , Proc. Nati Acad. Sci. USA. 84, 9194-9198, 1987; Barel M., et al. , Mol. Immunol. 35.1025-1031, 1998; Weis J.J., et al Proc. Nati Acad. Sci. USA. 83, 5639-5643, 1986; Sinha S.K., et al. (1993) J. I munol. 150, 5311-5320; WO2004045520 (Example 4); US2004005538 (Example 1); WO2003062401 (Claim 9); WO2004045520 (Example 4); WO9102536 (Figure 9.1-9.9); WO2004020595 (Claim 1); Access: P20023; Q13866; Q14212; EMBL; M26004; AAA35786.1. 1033 KSLLCITKDKVDGTWDKPAPKCEYFNXYSSCPEPIVPGGYKIRGSTPYRHGDSVTFACKT aa MGAAGLLGVFLALVAPGVLGISCGSPPPILNGRISYYSTPIAVGTVIRYSCSGTFRLIGE NFSMNGNXSVWCQANNMWGPTRLPTCVSVFPLECPALPMIHNGHHTSENVGSIAPGLSVT YSCESGYLLVGEKIINCLSSGKWSAVPPTCEEARCKSLGRFPNGKVKEPPILRVGVTANF FCDEGYRLQGPPSSRCVIAGQGVAWTKMPVCEEIFCPSPPPILNGRHIGNSLANVSYGSI VTYTCDPDPEEGVNFILIGESTLRCTVDSQKTGTWSGPAPRCELSTSAVQCPHPQILRGR MVSGQKDRYTYNDTVIFACMFGFTLKGSKQIRCNAQGTWEPSAPVCEXECQAPPNILNGQ KEDRHMVRFDPGTSIKYSCNPGYVLVGEESIQCTSEGVWTPPVPQCKVAACEATGRQLLT KPQHQFVRPDVNSSCGEGYKLSGSVYQECQGTIPWPMEIRLCKEITCPPPPVIYNGAHTG SSLEDFPYGTTVTYTCNPGPERGVEFSLIGESTIRCTSNDQERGTWSGPAPLCKLSLLAV QCSHVHIANGYYISGKEAPYPYNDTVTFKCYSGPTLKGSSQIRCYADNTWDPEIPVCEKE TCQHVRQSLQELPAGSRVELVNTSCQDGYQLTGHAYQMCQDAENGIWFKKIPLCKVIHCH PPPVIVNGKHTGMMAENFLYGNEVSYECDQGFYLLGEKKLQCRSDSKGHGSWSGPSPQCL RSPPVTRCPNPEVKHGYKLNKTHSAYSHNDIVYVDCNPGFIMNGSRVIRCHTDNTWVPGV PTCIKKAFIGCPPPPKTPNGNHTGGNIARFSPGMSILYSCDQGYLLVGEALLLCTHEGTW SQPAPHCKEVNCSSPADMDGIQKGLEPRKMYQYGAWTLECEDGYMLEGSPQSQCQSDHQ WNPPLAVCRSRSLAPV LCGIAAGLILLTFLIVITLYVISKHRERNYYTDTSQKFAPHLEA RE "VYSVDPYNPAS (SEQ ID NO: 14) (15) CD79b (CD79B, CD79β, IGb (beta associated with immunoglobulin), B29, Genbank Access No. NM_000626 or 11038674, Proc. Nati Acad. Sci. USA. (2003) 100 (7): 4126-4131, Blood (2002) 100 (9): 3068-3076, Muller et al (1992) Eur. J. Immunol. 22 (6) A621-1625); WO2004016225 (claim 2, Figure 140); WO2003087768, US2004101874 (claim 1, page 102); WO2003062401 (claim 9); WO200278524 (Example 2); US2002150573 (claim 5, page 15); US5644033; WO2003048202 (claim 1, pages 306 and 309); WO 99/558658, US6534482 (claim 13, Figure 17A / B), - WO200055351 (claim 11, pages 1145-1146); Cross-references: MIM: 147245; NP_000617.1; NM_00062__1 229 aa MARLALSPVPSHWMVALLLLLSAEPVPAARSEDRYRNPKGSACSRIWQSPRFIARKRGFT VKMHCYMNSASGNVSWLWKQEMDENPQQLKLEKGRMEESQNESLATLTIQGIRFEDNGIY FCQQKCNNTSEVYQGCGTELRVMGFSTLAQLKQRNTLKDGIIMIQTLLIILFIIVPIFLL LDKDDSKAGMEEDHTYEGLDIDQTATYEDIVTLRTGEVKWSVGEHPGQE (SEQ ID NO -.15) (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing protein anchor the phosphatase), SPAP1B , SPAP1C, Genbank Access No. NM_030764, Genome Res. 13 (10): 2265-2270 (2003), Immunogenetics 54 (2): 87-95 (2002), Blood 99 (8).-2662-2669 (2002) , Proc. Nati, Acad. Sci. USA, 98 (17): 9772-9777 (2001), Xu, MJ, et al (2001) Biochem Biophys., Res. Commun. 280 (3): 768-775; WO2004016225 (Claim 2); WO2003077836; WO200138490 (Claim 5; Figure 18D-1-18D-2); WO2003097803 (Claim 12); WO2003089624 (Claim 25); Cross-references: MIM-.606509; NP_110391.2; NM__030764__1 508 aa MLLWSLLVIFDAVTEQADSLTLVAPSSVFEGDSIVLKCQGEQNWKIQKMAYHKDNKEL SV FKKFSDFLIQSAVLSDSGNYFCSTKGQLFLWDKTSNIVKIKVQELFQRPVLTASSFQPIE GGPVSLKCETRLSPQRLDVQLQFCFFRENQVLGSGWSSSPELQISAVWSEDTGSYWCKAE TVTHRIRKQSLQSQIHVQRIPISNVSLEIRAPGGQVTEGQKLILLCSVAGGTGNVTFSWY REATGTSMGKKTQRSLSAELEIPAVKESDAGKYYCRADNGHVPIQSKWNIPVRIPVSRP VLTLRSPGAQAAVGDLLELHCEALRGSPPILYQFYHEDVTLGNSSAPSGGGASFNLSLTA EHSGNYSCEANNGLGAQCSEAVPVSISGPDGYRRDLMTAGVLWGLFGVLGFTGVALLLYA LFHKISGESSATNEPRGASRPNPQEFTYSSPTPDMEELQPVYVNVGSVDVDWYSQVWSM QQPESSANIRTLLENKDSQVIYSSVKKS (SEQ ID NO: 16) (17) HER2 (ErbB2, Genbank Accession No. M11730, Coussens L., et al Science (1985) 230 (4730): 1132-1139); Yamamoto T., et al Nature 319, 230-234, 1986; Semba K., et al Proc. Nati Acad. Sci. USA. 82, 6497-6501, 1985; Swiercz J.M. , et al J. Cell Biol. 165, 869-880, 2004; Kuhns J.J., et al J, Biol. Chem. 274, 36422-36427, 1999; Cho H.-S., et al Nature 421, 756-760, 2003; Ehsani A., et al (1993) Genomics 15, 426-429 WO2004048938 (Example 2); WO2004027049 (Figure 11) WO2004009622; WO2003081210; WO2003089904 (Claim 9) WO2003016475 (Claim 1); US2003118592; WO2003008537 (Claim 1); WO2003055439 (Claim 29; 1A-B); WO2003025228 (Claim 37; Figure 5C), - WO200222636 (Example 13; Page 95-107); WO200212341 (Claim 68, Figure 7); WO200213847 (Page 71-74); WO200214503 (page 114-117); WO200153463 (Claim 2; Page 41-46); WO200141787 (Page 15); WO200044899 (Claim 52, Figure 7); WO200020579 (Claim 3; Figure 2); US5869445 (Claim 3; Col 31-38); WO9630514 (Claim 2; Page 56-61); EP1439393 (Claim 7); WO2004043361 (Claim 7); WO2004022709; WO200100244 (Example 3, Figure 4); Access: P04626; EMBL; M11767; AAA35808.1.EMBL; M11761; AAA35808.1. 1255 aa MELAALCRWGLLLALLPPGAASTQVCTGTDMKLRLPASPETHLDMLRHLYQGCQWQGNL ELTYLPTNASLSFLQDIQEVQGYVLIAHNQVRQVPLQRLRIVRGTQLFEDNYALAVLDNG DPLNNTTPVTGASPGGLRELQLRSLTEILKGGVLIQRNPQLCYQDTILWKDIFHKNNQLA LTLIDTNRSRACHPCSPMCKGSRCWGESSEDCQSLTRTVCAGGCARCKGPLPTDCCHEQC AAGCTGPKHSDCLACLHFNHSGICELHCPALVTYNTDTPESMPNPEGRYTFGASCVTACP YNYLSTDVGSCTLVCPLHNQEVTAEDGTQRCEKCSKPCARVCYGLGMERLREVRAVTSAN IQEFAGCKKIFGSLAFLPESFDGDPASNTAPLQPEQLQVFETLEEITGYLYISAWPDSLP DLSVFQNLQVIRGRILHNGAYSLTLQGLGISWLGLRSLRELGSGLALIHHMTRLCFVHTV PWDQLFRNPHQALLHTANRPEDECVGEGLACHQLCARGHCWGPGPTQCVNCSQFLRGQEC VERCRVLQGLPREYVNARHCLPCHPECQPQNGSVTCPGPEADQCVACAHYKDPPFCVARC PSGVKPDLSYMPIWKFPDEEGACQPCPINCTHSCVDLDDKGCPAEQRASPLTSIISAWG ILLV LGWFGILIKRRQQKIRKYTMRRLLQETELVEPLTPSGAMPNQAQNRILKETEL RKVKVLGSGAFGTVYKGIWIPDGENVKIPVAIKVLRENTSPKANKEILDRAYVMAGVGSP YVSRLLGICLTSTVQLVTQLMPYGCLLDHVRENRGRLGSQDLLNWCMQIAKGMSYLEDVR LVHRDIJ ^ JINVLVKSPNHVKITDFGLARLLDIDETEYHADGGKVPIKWMALESILRRRPT HQSDVWSYGVTVWELMTFGAKPYDGIPAREIPDLLEKGERLPQPPICTIDVYMIMVKCWM IDSECRPRFRELVSEFSR MARDPQRFWIQNEDLGPASPLDSTFYRSLLEDDDMGDLVDA EEYLVPQQGFFCPDPAPGAGGMVHHRHRSSSTRSGGGDLTLGLEPSEEEAPRSPLAPSEG AGSDVFDGDLGMGAAKGLQSLPTHDPSPLQRYSEDPTVPLPSETDGYVAPLTCSPQPEYV NQPDVRPQPPSPREGPLPAARPAGATLERPKTLSPGXNGWKDVFAFGGAVENPEYLTPQ GGAAPQPHPPPAFSPAFDNLYYWDQDPPERGAPPSTFKGTPTAENPEYLGLDVPV (SEQ ID NO: 17) (18) NCA (CEACAM6, Genbank Accession No. M18728); Barnett T., et al. , Genomics 3, 59-66, 1988; Tawaragi Y., et al Biochem. Biophys. Res. Commun. 150, 89-96, 1988; Strausberg R.L. , et al. Proc. Nati Acad. Sci. USA. 99: 16899-16903, 2002; WO2004063709; EP1439393 (Claim 7); WO2004044178 (Example 4); WO2004031238; WO2003042661 (Claim 12); WO200278524 (Example 2); WO200286443 (Claim 27; Page 427); WO200260317 (Claim 2); Access: P40199; Q14920; EMBL; M29541; AAA59915.1. EMBL; M18728; 344 aa MGPPSAPPCRLHVPWKEVLLTASLLTFWNPPTTAKLTIESTPFNVABGKEVLLLAHNLPQ NRIGYSWYKGERVDGNSLIVGYVIGTQQATPGPAYSGRETIYPNASLLIQNVTQNDTGFY TLQVIKSDLVNEEATGQFHVYPELPKPSISSNNSNPVEDKDAVAFTCEPEVQNTTYLWWV NGQSLPVSPRLQLSNGNMTLTLLSVKRNDAGSYECEIQNPASANRSDPVTLNVLYGPDVP TISPS ANYRPGENLNLSCHAASNPPAQYSWFINGTFQQSTQELFIPNITVNNSGSYMCQ AHUSATGLNRTTVTMITVSGSAPVLSAVATVGITIGVLARVALI (SEQ ID NO: 18) (19) MDP (DPEP1, Genbank Access No. BC017023, Proc. Nati, Acad. Sci. USA, 99 (26): 16899-16903 (2002)); WO2003016475 (Claim 1); WO200264798 (Claim 33; Page 85-87); JP05003790 (Figure 6-8); W09946284 (Figure 9); Cross references: MIM: 179780; AAH17023.1; BC017023_1 411 aa - MWSGWWLWPLVAVCTADFFRDFAERIMRDSPVIDGHNDLPWQLLDMFNNRLQDERANLTT LAGTHTNIPKLPAGFVGGQFWSVYTPCDTQNKDAVRRTLEQMDWHPMCRMYPETFLYVT SSAGIRQAFREGKVASLIGVEGGHSIDSSLGVLRALYQLGMRYLTLTHSCNTPWADNWLV DTGDSEPQSQGLSPFGQRWKELNRLGVLIDLARVSVATMKATLQLSRAPVIFSHSSAYS VCASRRNVPDDVLRLVKQTDSLVNVNFYNNYISCTNK NLSQVADHLDHIKEVAGARAVG FGGDFDGVPPVPEGLEDVSKYPDLIAELLRRNWTEAEVKGALADNLLRVFEAVEQASNLT QAPEEEPIPLDQLGGSCRTHYGYSSGASSLHRHWGLLLASLAPLVLCLSLL (SEQ ID NO -.19) (20) 20RA (20RA, zcytor7, Genbank Accession No. AF184971); Clark H.F., et al Genome Res. 13, 2265-2270, 2003; Mungall A.J., al. Nature 425, 805-811, 2003; Blumberg H., et al Cell 104, 9-19, 2001; Dumoutier L., et al. , J. Immunol. 167, 3545-3549, 2001; Parrish-Novak J., et al J. Biol. Chem. 277, 47517-47523, 2002; Pletnev S., et al (2003) Biochemistry 42: 12617-12624; Sheikh F., et al (2004) J. Immunol.172, 2006-2010; EP1394274 (Example 11); US2004005320 (Example 5); WO2003029262 (Page 74-75); WO2003002717 (Claim 2; Page 63); WO200222153 (Page 45-47); US2002042366 (Page 20-21); WO200146261 (Page 57-59); WO200146232 (Page 63-65); W09837193 (Claim 1; Page 55-59); Access: Q9UHF4; Q6UWA9; Q96SH8; EMBL; AF184971; AAF01320.1. 553 aa MRAPGRPALRPLPLPPLLLLLLAAPWGRAVPCVSGGLPKPANITFLSINMKNVLQWTPPE GLQGVKVTYTVQYFIYGQKKWLNKSECRNINRTYCDLSAETSDYEHQYYAKVKAIWGTKC SKWAESGRFYPFLETQIGPPEVALTTDEKSISWLTAPEKWKRNPEDLPVSMQQIYSNLK YNVSVLNTKSNRTWSQCVTNHTLVLTWLEPNTLYCVHVESFVPGPPRRAQPSEKQCARTL KDQSSEFKAKIIFWYVLPISITVFLFSVMGYSIYRYIHVGKEKHPANLILIYGNEFDKRF FVPAEKIVINFITLNISDDSKISHQDMSLLGKSSDVSSLNDPQPSGNLRPPQEEEEVKHL GYASHLMEIFCDSEENTEGTSFTQQESLSRTIPPDKTVIEYEYDVRTTDICAGPEEQELS LQEEVSTQGTLLESQAALAVLGPQTLQYSYTPQLQDLDPLAQEHTDSEEGPEBEPSTTLV DWDPQTGRLCIPSLSSFDQDSEGCEPSEGDGLGEEGLLSRLYEEPAPDRPPGENETYLMQ FMEEWGLYVQMEN (SEQ ID NO: 20) (21) Brevican (BCAN, BEHAB, Genbank No. AF229053 Access) Gary S.C., et al. Gene 256, 139-147, 2000; Clark H.F., et al Genome Res. 13, 2265-2270, 2003; Strausberg R.L., et al.
Proc. Nati Acad. Sci. USA. 99, 16899-16903, 2002; US2003186372 (Claim 11); US2003186373 (Claim 11); US2003119131 (Claim 1, Figure 52); US2003119122 (Claim 1, Figure 52); US2003119126 (Claim 1); US2003119121 (Claim 1; 52), - US2003119129 (Claim 1); US2003119130 (Claim 1); US2003119128 (Claim 1; 52); US2003119125 (Claim 1); WO2003016475 (Claim 1); WO200202634 (Claim 1); 911 aa MAQLFLPLLAALVLAQAPAALADVLEGDSSEDRAFRVRIAGDAPLQGVLGGALTIPCHVH YLRPPPSRRAVLGSPRVKWTFLSRGREAEVLVARGVRVKVNEAYRFRVALPAYPASLTDV SLALSELRPNDSGIYRCEVQHGIDDSSDAVEVKVKGWFLYREGSARYAFSFSGAQRACA RIGAHIATPEQLYAAYLGGYEQCDAGWLSDQTVRYPIQTPREACYGDMDGFPGVRNYGW DPDDLYDVYCYAEDLNGELFLGDPPEKLTLEEARAYCQERGAEIATTGQLYAAWDGGLDR CSPGWLADGSWYPIVTPSQRCGGGLPGVKTLFLFPNQTGFPNKHSRFNVYCPRDSAQPS AIPEASNPASNPASDGLEAIVTVTETLEELQLPQEATESESRGAIYSIPIMEDGGGGSST PEDPAEAPRTLLEFETQSMVPPTGFSEEEGKALEEEEKYEDEEEKEEEEEEERVEDEALW AWPSELSSPGPEASLPTEPAAQEKSLSQAPARAVLQPGASPLPDGESEASRPPRVHGPPT ETLPTPRERNLASPSPSTLVEAREVGEATGGPELSGVPRGESEETGSSEGAPSLLPATRA PEGTRELEAPSEDNSGRTAPAGTSVQAQPVLPTDSASRGGVAWPASGDCVPSPCHNGGT CLEEEEGVRCLCLPGYGGDLCDVGLRFCNPGWDAFQGACYKHFSTRRSWEEAETQCRMYG AHLASISTPEEQDFINNRYREYQWIGLNDRTIEGDFLWSDGVPLLYENWNPGQPDSYFLS GENCVVMWHDQGQWSDVPCNYHLSYTCKMGLVSCGPPPELPLAQVFGRPRLRYEVDTVL RYRCREGLAQRNLPLIRCQENGRWEAPQISCVPRRPARALHPBEDPEGRQGRLLGRWKAL LIPPSSPMPGP (SEQ ID NO: 21) (22) EphB2R (DRT, ERK, Hek5, EPHT3, Tyro5, Genbank Access No. NM_004442) Chan, J. and Watt, V.M. , Oncogene 6 (6), 1057-1061 (1991) Oncogene 10 (5): 897-905 (1995), / Annu. Rev. Neurosci. 21: 309-345 (1998), Int. Rev. Cytol. 196: 177-244 (2000)); WO2003042661 (Claim 12); WO200053216 (Claim 1; Page 41); WO2004065576 (Claim 1); WO2004020583 (Claim 9); WO2003004529 (Page 129-132), -WO200053216 (Claim 1; Page 42); Cross references: MIM: 600997; NP_004433.2; NM_004442_1 987 aa MALRRLGAALLLLPLLAAVEETLMDSTTATAELGWMVHPPSGWEEVSGYDENMNTIRTYQ VCNVFESSONNWLRTKFIRRRGAHRIHVEMKFSVRDCSSIPSVPGSCKETFNLYYYEADF - DSATKTFPNWMENPWVKVDTIAADESFSQVDLGGRVMKINTEVRSFGPVSRSGFYLAFQD YGGCMSLIAVPVFYRKCPRIIQNGAIFQETLSGAESTSLVAARGSCIANAEEVDVPIKLY CNGDGEWLVPIGRCMCKAGFEAVENGTVCRGCPSGTFKANQGDEACTHCPINSRTTSEGA TNCVCRNGYYRADLDPLDMPCTTIPSAPQAVISSVNETSLMLEWTPPRDSGGREDLVYNI ICKSCGSGRGACTRCGDNVQYAPRQLGLTEPRIYISDLLAHTQYTFEIQAVNGVTDQSPF SPQFASVNITTNQAAPSAVSIMHQVSRTVDSITLSWSQPDQPNGVILDYELQYYEKELSE YNATAIKSPTNTVTVQGLKAGAIYVPQVRAPTVAGYGRYSGKMYFQTMTEAEYQTSIQEK LPLIIGSSAAGLVFLIAVWIAIVCNRRRGFERADSEYTDKLQHYTSGHMTPGMKIYIDP FTYEDPNEAVREFAKEIDISCVKIEQVIGAGEFGEVCSGHLKLPGKREIFVAIKTLKSGY TEKQRRDFLSEASIMGQFDHPNVIHLEGWTKSTPVMIITEFMENGSLDSFLRQNDGQFT VIQLVGMLRGIAAGMKYLADMNYVHRDLAARNILVNSNLVCKVSDFGLSRFLEDDTSDPT YTSALGGKIPIRWTAPEAIQYRKFTSASDVWSYGIVMWEVMSYGERPYWDMTNQDVINAI EQDYRLPPPMDCPSALHQLMLDCWQKDRNHRPKFGQIVNTLDKMIRNPNSLKAMAPLSSG INLPLLDRTIPDYTSFNTVDEWLEAIKMGQYKESFANAGFTSFDWSQMMMEDILRVGVT LAGIIQKKILNSIQVMRAQMNQIQSVEV (SEQ ID NO: 22) (23) ASLG659 (B7h, Genbank Accession No. AX092328) US20040101899 (Claim 2); WO2003104399 (Claim 11); WO2004000221 (Figure 3); US2003165504 (Claim 1); US2003124140 (Example 2); US2003065143 (Figure 60); WO2002102235 (Claim 13; Page 299); US2003091580 (Example 2); WO200210187 (Claim 6; Figure 10); WO200194641 (Claim 12, Figure 7b); WO200202624 (Claim 13; Figure 1A-1B); US2002034749 (Claim 54; Page 45-46); WO200206317 (Example 2; - - Page 320-321, Claim 34; Page 321-322); WO200271928 (Page 468-469); WO200202587 (Example 1, Figure 1); WO200140269 (Example 3; Pages 190-192); WO200036107 (Example 2; Page 205-207); WO2004053079 (Claim 12); WO2003004989 (Claim 1); WO200271928 (Page 233-234,452-453); WO 0116318; 282 aa MASLGQILFWSIISIIIILAGAIALIIGFGISGRHSITVTTVASAGNIGEDGILSCTFEP DIKLSDIVIQWLKEGVLGLVHEFKEGKDELSEQDEMPRGRTAVFADQVIVGNASLRLKNV QLTDAGTYKCYIITSKGKKNANLEYKTGAFSMPEVNVDYNASSETLRCEAPRWFPQPTW WASQVDQGANFSEVSNTSFELNSENVTMKWSVLYNVTINNTYSCMIENDIAKATGDIKV TESEIKRRSHLQLLNSKASLCVSSFFAISWALLPLSPYLMLK (SEQ ID NO: 23) (24) PSCA (Precursor germ cell antigen prostate, Genbank Accession No. AJ297436) Reiter R.E., et al Proc. Nati, Acad. Sci. USA. 95, 1735-1740, 1998; Gu Z., et al Oncogene 19, 1288-1296, 2000; Biochem. Biophys. Res. Commun. (2000) 275 (3): 783-788; WO2004022709; EP1394274 (Example 11); US2004018553 (Claim 17); WO2003008537 (Claim 1); WO200281646 (Claim 1; Page 164), - WO2003003906 (Claim 10; Page 288) WO200140309 (Example 1; Figure 17); US2001055751 (Example 1 Figure Ib); WO200032752 (Claim 18; Figure 1) WO9851805 (Claim 17; Page 97); W09851824 (Claim 10; Page 94); WO9840403 (Claim 2; Figure IB); Access: 043653; EMBL; AF043498; AAC39607.1. 123 aa MKAVLLALLMAGLALQPGTALLCYSCKAQVSNEDCLQVENCTQLGEQCWTARIRAVGLLT VISKGCSLNCVDDSQDYYVGKKNITCCDTDLCNASGARALQPAAAILALLPALGLLLWGP GQL (SEQ ID NO: 24) (25) GEDA (Genbank Access No. AY260763); protein similar to lipoma fusion partner AAP14954 HMGIC / pid = AAP14954.1 - Homo sapiens Species: Homo sapiens (human) WO2003054152 (Claim 20); WO2003000842 (Claim 1); WO2003023013 (Example 3, Claim ), - US2003194704 (Claim 45); Cross references: GI: 30102449; AAP14954.1; AY260763_1 236 aa MPGAAAAAAAAAAAMLPAQEAAKLYHTNYVRHSRAI GVLWAI FTI CFAI VNWCF I QP YW IGDGVDTPQAGYFGLPHYCIGNGFSRELTCRGSFTDFSTLPSGAFKAASFFIGLSMMLI I ACIICFTLFFFCNTATWKICAWMQLTSAACLVLGCMIFPDGWDSDEVKRMCGEKTDKYT LGACSVRWAYILAIIGILDALILSFLAFVLGNRQDSLMAEELKAENKVLLSQYSLE (SEQ ID NO: 25) (26) BAFF-R (cell activation factor receptor B, receiver 3 BLyS, BR3, Genbank Access No. NP_443177.1); NP_443177 BAFF receptor / pid = NP_443177.1 - Homo sapiens Thompson, J.S., et al Science 293 (5537), 2108-2111 (2001); WO2004058309; WO2004011611; WO2003045422 (Example; Page 32 33); WO2003014294 (Claim 35; Figure 6B) WO2003035846 (Claim 70; Page 615-616) WO200294852, (Col 136-137); WO200238766 (Claim 3 Page 133); WO200224909 (Example 3, Figure 3); Cross references: MIM: 606269; NP_443177.1; NM_052945_1 184 aa MRRGPRSLRGRDAPAPTPCVPAECFDLLVRHCVACGLLRTPRPKPAGASSPAPRTALQPQ ESVGAGAGEAALPLPGLLFGAPALLGLALVLALVLVGLVSWRRRQRRLRGASSABAPDGD KDAPEPLDKVIILSPGISDATAPAWPPPGEDPGTTPPGHSVPVPATELGSTELVTTKTAG PEQQ (SEQ ID NO: 26) (27) CD22 (CD22-B isoform receptor cell B, Genbank No. NP-access 001762.1); Stamenkovic, I. and Seed, B., Nature 345 (6270), 74-77 (1990); US2003157113; US2003118592; WO2003062401 (Claim 9); WO2003072036 (Claim 1, Figure 1); WO200278524 (Example 2); Cross-references: MIM: 107266; NP_001762.1; NM_001771_1 847 aa MHLLGPWLLLLVLEYLAFSDSSKWVFEHPETLYAWEGACVWIPCTYRALDGDLESFILFH NPEYNKNTSKFDGTRLYESTKDGKVPSEQKRVQFLGDKNKNCTLSIHPVRLNDSGQLGLR MESKTEKWMERIHLNVSERPFPPHIQLPPEIQESQEVTLTCLLNFSCYGYPIQLQWLLEG VPMRQAAVTSTSLTIKSVPTRSELKFSPQWSHHGKIVTCQLQDADGKFLSNDTVQLNVKH TPKLEIKVTPSDAI EGDSVTMTCEVSSSNPEYTTVSWLKDGTSLKKQNTFTLNLREVT KDQSGKYCCQVSNDVGPGRSEEVFLQVQYAPEPSTVQILHSPAVEGSQVEFLCMSLANPL PTNYTWYHNGKEMQGRTEEKVHIPKILPWRAGTYSCVAENILGTGQRGPGAELDVQYPPK KVTTVIQNPMPIREGDTVTLSCNYNSSNPSVTRYEWKPHGAWEEPSLGVLKIQNVGWDNT TIACARCNSWCSWASPVALNVQYAPRDVRVRKIKPLSEIHSGNSVSLQCDFSSSHPKEVQ FFWEKNGRLLGKESQLNFDSISPEDAGSYSCWVNNSIGQTASKAWTLEVLYAPRRLRVSM SPGDQVMEGKSATLTCESDANPPVSHYTWFDWNNQSLPHHSQKLRLEPVKVQHSGAYWCQ GTNSVGKGRSPLSTLTVYYSPETIGRRVAVGLGSCLAILILAICGLKLQRRWKRTQSQQG LQENSSGQSFFVRNKKVRRAPLSEGPHSLGCYNPMMEDGISYTTLRPPEMNIPRTGDAES SEMQRPPRTCDDTVTYSALHKRQVGDYENVIPDFPEDEGIHYSELIQFGVGERPQAQENV DYVILKH (SEQ ID NO: 27) (28) CD79a (CD79A, CD79, alpha-associated immunoglobulin, a specific protein d e cell B that interacts covalently with beta Ig (CD79B) and forms a complex on the surface with Ig M molecules, transduces a signal involved in the differentiation of cell B) PROTEIN SEQUENCE Complete mpggpgv ... dvqlekp (1. .226: 266 aa), pl: 4.84, MW: 25028 TM: 2 [P] Gen Chromosome: 19ql3.2, Genbank Access No. NP_0017741; WO2003088808, US20030228319; WO2003062401 (claim 9); US2002150573 (claim 4, pages 13-14); W09958658 (claim 13, Figure 16); WO9207574 (Figure 1); US5644033; Ha et al. (1992) J. Immunol. 148 (5) .1526-1531; Mueller et al (1992) Eur. J. Biochem. 22: 1621-1625; Hashimoto et al (1994) Immunogenetics 40 (4): 287-295; Preud'homme et al (1992) Clin. Exp. Immunol. 90 (1) .141-146; Yu et al. (1992) J. Immunol. 148 (2) 633-637; Sakaguchi et al. (1988) EMIBO J. 7 (11): 3457-3464; 226 aa MPGGPGVLQALPATIFLLFLLSAVYLGPGCQALWMHKVPASLMVSLGEDAHFQCPHNSSN NANVTWWRVLHGNYTWPPEFLGPGEDPNGTLIIQNWNKSHGGIYVCRVQEGNESYQQSCG TYLRVRQPPPRPFLDMGEGTKNRIITAEGIILLFCAWPGTLLLFRKRWQNEKLGLDAGD EYEDENLYEGLNLDDCSMYEDISRGLQGTYQDVGSLNIGDVQLEKP (SEQ ID NO: 28) (29) CXCR5 (receptor 1 Burkitt lymphoma, a receptor coupled to the G protein is activated by the chemokine, functions in the migration of lymphocytes and humoral defense plays a role in HIV-2 infection and perhaps the development of AIDS, lymphoma, myeloma and leukemia) PROTEIN SEQUENCE Complete mnypltl ... atslttf (1..372; 372aa), pl: 8.54 MW: 41959 TM: 7 [P] Gen Chromosome: llq23.3, Genbank Access No. NP_001707.1; WO2004040000; WO2004015426; US2003105292 (Example 2); US6555339 (Example 2); WO200261087 (Figure 1); WO200157188 (Claim 20, page 269); WO200172830 (pages 12-13); WO200022129 (Example 1, pages 152-153, Example 2, pages 254-256); W09928468 (claim 1, page 38); US5440021 (Example 2, col 49-52); W09428931 (pages 56-58); W09217497 (claim 7, Figure 5); Dobner et al (1992) Eur. J. Immunol. 22: 2795-2799; Barella et al. (1995) Biochem. J. 309: 773-779; 372 aa MNYPLTLEMDLENLEDLFWELDRLDNYNDTSLVENHLCPATEGPLMASFKAVFVPVAYSL IFLLGVIGNVLVLVILERHRQTRSSTETFLFHLAVADLLLVFILPFAVAEGSVGWVLGTF LCKTVIALHKVNFYCSSLLLACIAVDRYLAIVHAVHAYRHRRLLSIHITCGTIWLVGFLL ALPEILFAKVSQGHHNNSLPRCTFSQENQAETHAWFTSRFLYHVAGFLLPMLVMGWCYVG WHRLRQAQRRPQRQKAVRVAILVTSIFFLCWSPYHIVIFLDTLARLXAVDNTCKLNGSL PVAITMCEFLGLAHCCLNPMLYTFAGVKFRSDLSRLLTKLGCTGPASLCQLFPSWRRSSL SESENATSLTTF (SEQ ID NO: 29) (30) HLA-DOB (Beta subunit of the MHC class II molecule (antigen la) that binds the peptides and presents them for CD4 + T lymphocytes) PROTEIN SEQUENCE Complete mgsgwwp ... vllpqsc (1.273; 273 aa, pl: 6.56 MW: 30820 TM: 1 [P] Chromosome Gene: 6p21.3, Genbank Access No. NP_002111.1; Tonnelle et al. (1985) EMBO J. 4 (11): 2839-2847; Jonsson et al. (1989) Immunogenetics 29 (6): 411-413; Beck et al (1992) J.
Mol. Biol. 228: 433-441; Strausberg et al (2002) Proc. Nati Acad. Sci USA 99: 16899-16903; Servenius et al (1987) J. Biol.
Chem. 262: 8759-8766; Beck et al (1996) J. Mol. Biol. 255: 1-13; Naruse et al (2002) Tissue Antigens 59: 512-519; W09958658 (claim 13, Figure 15); US6153408 (Col 35-38); US5976551 (col 168-170); US6011146 (col 145-146); Kasahara et al (1989) Immunogenetics 30 (1): 66-68; Larhammar et al. (1985) J. Biol. Chem. 260 (26): 14111-14119; 273 aa MGSGWVPWWALLVNLTRLDSSMTQGTDSPEDFVIQAKADCYFTNGTEKVQFWRFIFNL EEYWFDSDVGMFVALTKLGQPDAEQWNSRLDLLERSRQAVDGVCRHNYRLGAPFTVGRK VQPEVTVYPERTPLLHQHNLLHCSVTGFYPGDIKIKWPLNGQEERAGVMSTGPIRNGDWT FQTVVMLEMTPELGHVYTCLVDHSSLLSPVSVEWRAQSEYSWRKMLSGIAAFLLGLIFLL VGIVIQLRAQKGYVRTQMSGNEVSRAVLLPQSC (SEQ ID NO: 30) (31) P2X5 (Pcanal 5 ion confined in the ligand P2X the purigénico receptor, an ion channel confined by extracellular ATP, may be involved in synaptic transmission and neurogenesis , deficiency may contribute to the pathophysiology of idiopathic detrusor instability) PROTEIN SEQUENCE Complete mgqagck ... lephrst (1.4222; 422 aa), pl: 7.63, MW: 47206 TM: 1 [P] Chromosome Gene : 17pl3.3, Genbank Access No. NP_002552.2; Le et al. (1997) FEBS Lett. 418 (1-2): 195-199; WO2004047749; WO2003072035 (claim 10); Touchman et al (2000) Genome Res. 10: 165-173; WO200222660 (claim 20); WO2003093444 (claim 1); WO2003087768 (claim 1); WO2003029277 (page 82), -422aa MGQAGCKGLCLSLFDYKTEKYVIAKNKKVGLLYRLLQASILAYLVVWVPLIKKGYQDVDT SLQSAVITKVKGVAFTNTSDLGQRIWDVADYVIPAQGENVFFWTNLIVTPNQRQNVCAE NEGIPDGACSKDSDCHAGRAVTAGNGVKTGRCLRRENLARGTCEIFAWCPLETSSRPEEP FLKEAEDFTIFIKNHIRFPKFNFSKSNVMDVKDRSFLKSCHFGPKNHYCPIFRLGSVIRW AGSDFQDIALEGGVIGINIEWNCDLDKAASECHPHYSFSRLDNKLSKSVSSGYNFRFARY YRDAAGVEFRTLMKAYGIRPDVMVNGKGAFFCDLVLIYLIKKREFYRDKKYEEVRGLEDS SQEAEDEASGLGLSEQLTSGPGLLGMPEQQELQEPPEAKRGSSSQKGNGSVCPQLLEPHR ST (SEQ ID NO: 31) (32) CD72 (CD72 antigen of B cell differentiation, Lyb-2) PROTEIN SEQUENCE Complete aeaity ... tafrfpd (1.359; 359 aa), pl: 8.66, MW: 40225 TM: 1 [P] Gen Chromosome: 9pl3.3, Genbank Access No. NP_001773.1; WO2004042346 (claim 65); WO2003026493 (pages 51-52, 57-58); WO200075655 (pages 105-106); Von Hoegen et al (1990) J. Immunol. 144 (12): 4870-4877; Strausberg et al (2002) Proc. Nati Acad. Sci USA 99: 16899-16903; 359 aa MAEAITYADLRPVKAPLKKSISSRLGQDPGADDDGEITYENVQVPAVLGVPSSLASSVLG DKAAVKSEQPTASWRAVTSPAVGRILPCRTTCLRYLLLGLLLTCLLLGVTAICLGVRYLQ VSQQLQQTNRVLEVTNSSLRQQLRLKITQLGQSAEDLQGSRRELAQSQEALQVEQRAHQA AEGQLQACQADRQKTKETLQSEEQQRRALEQKLSNMENRLKPFFTCGSADTCCPSGWIMH QKSCFYISLTSKNWQESQKQCETLSSKLATFSEIYPQSHSYYFLNSLLPNGGSGNSYWTC LSSNKDWKLTDDTQRTRTYAQSSKCNKVHKTWSWWTLESESCRSSLPYICEMTAFRFPD (SEQ ID NO: 32) (33) LY64 (lymphocyte antigen 64 (RPI05), membrane protein type I of the leucine family rich in the repeat family (LRR), which regulates B cell activation and apoptosis, loss of function is associated with the increased activity of the disease in patients with systemic lupus erythematosis) PROTEIN SEQUENCE Complete mafdvsc ... rwkyqhi (1.661; 661 aa), pl: 6.20, MW: 74147 TM: 1 [P] Chromosome of Gen: 5ql2, Genbank Access No. NP_005573. 1; US2002193567; WO9707198 (claim 11, pages 39-42); Miura et al. (1996) Geno ics 38 (3): 299-304; Miura et al. (1998) Blood 92: 2815-2822; WO2003083047; W09744452 (claim 8, pages 57-61); WO200012130 (pages 24-26); 661 aa FDVSCFFWVVLFSAGCKVITSWDQMCIEKEANKTYNCENLGLSEIPDTLPNTTEFLEF SFNFLPTIHNRTFSRLMMLTFLDLTRCQINKIHEDTFQSHHQLSTLVLTGNPLIFMAETS LNGPKSLKRLFLIQTGISNLEFIPVHNLENLESLYLGSNHISSIKFPKDFPARNLKVLDF QNNAIHYISREDMRSLEQAINLSLNFNGMNVKGIELGAFDSTVFQSLNFGGTPNLSVIFN GLQNSTTQSLWLGTFEDIDDEDISSAMLKGLCEMSVESLNLQEHRFSDISSTTFQCFTQL QELDLTATHLKGLPSGMKGLNLLKKLVLSVNHFDQLCQISAANFPSLTHLYIRGNVKKLH LGVGCLEKLGNLQTLDLSHNDIEASDCCSLQLKULSHLQTLNLSHNEPLGLQSQAFKECP QLELLDLLHINAPQSPFQNLHFLQVLNLTYCFLDTSNQHLLAGLPVLRHLNLKGNH FQDGTITKTNLLQTVGSLEVLILSSCGLLSIDQQAFHSLGKMSHVDLSHNSLTCDSIDSL SULKGIYLNLAANSINIISPRLLPILSQQSTINLSHNPLDCTCSNIHFLTWYKENLHKLE GSEETTCANPPSLRGVKLSDVKLSCGITAIGIFFLIVFLLLLAILLFFAVKYLLRWKYQH I (SEQ ID NO: 33) (34) FCRH1 (protein 1 similar to Fe receptor, a putative receptor for the domain Fe immunoglobulin containing Ig-like domains type C2 and ITAM, may have a role in the differentiation of B lymphocyte) PROTEIN SEQUENCE Complete mlprlll ... vdyedam (1..429; 429 aa), pl: 5.28, MW: 46925 TM: 1 [P] Gen Chromosome: Iq21-lq22, Genbank Access No. NP_443170. 1; WO2003077836; WO200138490 (claim 6, Figure 18E-1-18-E-2); Davis et al (2001) Proc. Nati Acad. Sci USA 98 (17) -.9772-9777; WO2003089624 (claim 8); EP1347046 (Claim 1); WO2003089624 (claim 7); 429 aa MLPRLLLLICAPLCEPAELFLIASPSHPTEGSPVTLTCKMPFLQSSDAQFQFCPFRDTRA LGPGWSSSPKLQIAAMWKEDTGSYWCEAQTMASKVLRSRRSQINVHRVPVADVSLETQPP GGQVMEGDRLVLICSVAMGTGDITFLWYKGAVGLNLQSKTQRSLTAEYEIPSVRESDAEQ YYCVAENGYGPSPSGLVSITVRIPVSRPILMLRAPRAQAAVEDVLELHCEALRGSPPILY WFYHEDITLGSRSAPSGGGASFNLSLTEEHSGNYSCEANNGLGAQRSEAVTLNFTVPTGA RSNHLTSGVXEGLLSTLGPATVALLFCYGLKRKIGRRSARDPLRSLPSPLPQEFTYLNSP TPGQLQPIYENVNWSGDEVYSIAYYNQPEQESVAAETLGTHMEDKVSLDIYSRLRKANI TDVDYEDAM (SEQ ID NO: 34) (35) IRTA2 (translocation 2 of the immunoglobulin superfamily receptor, a putative immunoreceptor with possible roles in B-cell development and lymphomagenesis; gene deregulation through trans-location occurs in some B cell malignancies) PROTEIN SEQUENCE Complete mllwvil ... assaphr (1.977; 977 aa), pl: 6.88 MW: 106468 TM: 1 [P] Gen Chromosome: lq2l, Genbank Access No. NP_112571.1; WO2003024392 (claim 2, Figure 97); Nakayama et al. (2000) Biochem. Biophys. Res. Commun. 277 (1): 124-127; WO2003077836; WO200138490 (claim 3, Figure 18B-1-18B-2); 977 aa MLLWVILLVLAPVSGQFARTPRPIIFLQPPWTTVFQGERVTLTCKGFRFYSPQKTKWYHR YLGKEILRETPDNILEVQESGEYRCQAQGSPLSSPVHLDFSSASLILQAPLSVFEGDSW LRCRAKAEVTLNNTIYKNDNVLAFLNKRTDFHIPHACLKDNGAYRCTGYKESCCPVSSNT VKIQVQEPFTRPVLRASSFQPISGNPVTLTCETQLSLERSDVPLRFRFFRDDQTLGLGWS LSPNFQITAMWSKDSGFYWCKAATMPHSVISDSPRSWIQVQIPASHPVLTLSPEKALNFE GTKVTLHCETQEDSLRTLYRFYHEGVPLRHKSVRCERGASISFSLTTENSGNYYCTADNG LGAKPSKAVSLSVTVPVSHPVLNLSSPEDLIFEGAKVTLHCEAQRGSLPILYQFHHEDAA LERRSANSAGGVAISFSLTAEHSGNYYCTADNGFGPQRSKAVSLSITVPVSHPVLTLSSA EALTPEGATVTLHCEVQRGSPQILYQFYHEDMPLWSSSTPSVGRVSFSFSLTEGHSGNYY CTADNGFGPQRSEWSLFVTVPVSRPILTLRVPRAQAWGDLLELHCEAPRGSPPILYWF YHEDVTLGSSSAPSGGEASFNLSLTAEHSGNYSCEANNGLVAQHSDTISLSVIVPVSRPI LTFRAPRAQAWGDLLELHCEALRGSSPILYWPYHEDVTLGKISAPSGGGASFNLSLTTE HSGIYSCRADNGPEAQRSEMVTLKVAVPVSRPVLTLRAPGTHAAVGDLLELHCEALRGSP LILYRFFHEDVTLGNRSSPSGGASLNLSLTAEHSGNYSCEADNGLGAQRSETVTLYITGL TANRSGPPATGVAGGLLSIAGLAAGALLLYCWLSRKAGRKPASDPARSPPDSDSQEPTYH NVPAWEELQPVYTNANPRGENWYSEVRIIQBKKKRAVASDPRHLRNKGSP11YSEVKVA STPVSGSLPLASSAPHR (S EQ ID NO: 35) See also: WO04 / 045516 (03 Jun 2004); WO03 / 000113 (03 Jan 2003); WO02 / 016429 (Feb 28, 2002); WO02 / 16581 (Feb 28, 2002); WO03 / 024392 (Mar 27, 2003); WO04 / 016225 (Feb 26, 2004); WO01 / 40309 (07 Jun 2001), and Provisional patent application of E.U. Series No. 60/520842"COMPOSITIONS AND METHODS FOR TREATMENT OF TUMOR OF HEMATOPOYETIC ORIGIN ", presented on November 17, 2003, all of which is incorporated herein by reference in its entirety., the Ligand-Linker-Drug Conjugate has the Formula Illa, wherein the Ligand is an Ab antibody including one that binds at least one of CD30, CD40, CD70, Lewis antigen Y, w = 0, y = 0, and D has the Formula Ib. Exemplary conjugates of the Illa Formula include wherein R17 is - (CH2) 5-. Also included are such Conjugates of the Formula Illa in which D has the structure of compound 2 in Example 3 and esters thereof. Also included are such Conjugates of Formula Illa containing about 3 to about 8, in one aspect, about 3 to about 5 D residues of Drug, ie, Conjugates of the Formula wherein p is a value in the range of about 3-8, for example approximately 3-5. Conjugates containing combinations of annotated structural features - in this paragraph are also contemplated as being within the scope of the compounds of the invention. In another embodiment, the Ligand-Linker-Drug Conjugate has the Formula Illa, wherein the Ligand is an Ab Antibody that binds one of CD30, CD40, CD70, Lewis antigen Y, w = l, y = 0 and D has the Formula Ib. Such Conjugates of the Formula Illa in which R17 is - (CH2) 5- are included. Also included are such Conjugates of the Formula Illa in which W is -Val-Cit-, and / or wherein D has the structure of Compound 2 in Example 3 and esters thereof. Also included are such Conjugates of Formula Illa containing about 3 to about 8, preferably about 3 to about 5 Drug D Residues, that is, Conjugates of the Formula wherein p is a value in the range of about 3-8. , preferably about 3-5. The conjugates containing combinations of the structural features noted in this paragraph are also exemplary. In one embodiment, the Ligand-Linker-Drug Conjugate has the Formula Illa, wherein the Ligand is an Ab Antibody that binds one of CD30, CD40, CD70, Lewis Antigen Y, w = l, y = ly D has the Formula Ib. The conjugates of the Formula Illa in which R17 is - (CH2) 5- are included. Also included are such Conjugates of the Illa Formula wherein: W is Val-Cit-; And he has the Formula X; D has the - structure of Compound 2 in Example 3 and esters thereof; p is about 3 to about 8, preferably about 3 to about 5 D residues of Drug. The conjugates contain combinations of the structural features noted in this paragraph that are also contemplated within the scope of the compounds of the invention. An additional embodiment is a drug to antibody conjugate (ADC), or a pharmaceutically acceptable salt or solvate thereof, wherein Ab is an antibody that binds one of the tumor-associated antigens (1) - (35) noted above ( "TAA compound"). Another embodiment is the TAA Compound or pharmaceutically acceptable salt or solvate thereof, that is, it is in an isolated and purified form. Another embodiment is a method for eliminating or inhibiting the multiplication of a tumor cell or cancer cell comprising administering to a patient, for example a human with a hyperproliferative disorder, an amount of the TAA Compound or a pharmaceutically acceptable salt or solvate thereof, said amount being effective to eliminate or inhibit the multiplication of a tumor cell or cancer cell. Another embodiment is a method of treating cancer which comprises administering to a patient, for example a human - with a hyperproliferative disorder, an amount of the TAA Compound or a pharmaceutically acceptable salt or solvate thereof, said amount being effective to treat cancer, alone or together with an effective amount of an additional anticancer agent. Another embodiment is a method for treating an autoimmune disease, comprising administering to a patient, for example a human with a hyperproliferative disorder, an amount of the TAA Compound or a pharmaceutically acceptable salt or solvate thereof, said amount effective to treat a disease. autoimmune Suitable antibodies for use in the invention can be produced by a method known in the art for the synthesis of antibodies, in particular, by chemical synthesis or by recombinant expression, and are preferably produced by recombinant expression techniques. 4.5.1 PRODUCTION OF RECOMBINANT ANTIBODIES The antibodies of the invention can be produced using any method known in the art to be useful for the synthesis of antibodies, in particular, by chemical synthesis by recombinant expression. The recombinant expression of the antibodies or fragment, derivative or analog thereof, regulates the construction of a nucleic acid encoding the antibody. If the nucleotide sequence of the antibody is known, a nucleic acid encoding the antibody can be assembled from the chemically synthesized oligonucleotides (eg, as described in Kutmeier et al, 1994, BíoTechniques 17: 242), which involves the synthesis of overlaying the oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligation of these oligonucleotides, and then amplifying the bound oligonucleotides, eg, by PCR. Alternatively, a nucleic acid molecule encoding an antibody can be generated from a suitable source. If a clone is not available it contains the nucleic acid encoding the particular antibody, but the antibody sequence is known, a nucleic acid encoding the antibody can be obtained from a suitable source (eg, an antibody from the cDNA library, or of cDNA library generated from any tissue or cells expressing immunoglobulin) b and, eg, PCR amplification using synthetic hybridizable primers for the 3 'and 5' ends of the sequence or by cloning using an oligonucleotide test specific for the sequence of particular gene. If an antibody that specifically recognizes a particular antigen is not commercially available (or a source for a cDNA library for cloning a nucleic acid encoding such an immunoglobulin), antibodies specific for a particular antigen can be generated by any method known in the art, for example, by immunizing a patient or suitable animal model such as a rabbit or mouse, to generate polyclonal antibodies or more preferably by generating monoclonal antibodies, eg, as described by Kohier and Milstein (1975, Nature 256: 495-497) or, as described by Kozbor et al. (1983, I munology Today 4:72) or Cole et al (1985 in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). Alternatively, a clone encoding at least the Fab portion of the antibody can be obtained by examining the Fab expression libraries (eg, as described in Huse et al, 1989, Science 246: 1275-1281) for the clones of the Fab fragments that bind to the specific antigen or when examining antibody libraries (See, e.g., Clackson et al, 1991, Nature 352: 624; Hane et al, 1997 Proc. Nati Acad. Sci. USA 94: 4937). Once the nucleic acid sequence encoding at least the variable domain of the antibody is obtained, it can be introduced into a vector containing the nucleic acid sequence encoding the constant regions of the antibody (see, eg, International Publication No. WO). 86/05807; WO 89/01036; and U.S. Patent No. 5122464). Vectors are available that have complete light or heavy chain that are allowed for the expression of a complete antibody molecule. Then, the nucleic acid encoding the antibody can be used to introduce the nucleotide substitutions or omission necessary to replace (or delete) the one or more variable region cysteine residues that participate in an intrachain disulfide bond with an amino acid residue that does not contain a sulfhydryl group. Such modifications can be carried out by a method known in the art, but are not limited to chemical mutagenesis and site-directed in vitro mutagenesis (Hutchinson et al, 1978, J. Biol. Chem. 253: 6551). In addition, the development techniques for the production of "chimeric antibodies" can be used (Morrison et al., 1984, Proc. Nati Acad. Sci. 81: 851-855; Neuberger et al, 1984, Nature 312: 604-608; Takeda et al. , 1995, Nature 314: 452-454) by connecting genes from a mouse antibody molecule of the appropriate antigen specificity together with the genes from a human antibody molecule of appropriate biological activity. A chimeric antibody is a molecule in which different portions are derived from different animal species and the immunoglobulin constant region - Alternatively, the techniques described for the production of single chain antibodies can be adapted (US Patent 4,694,778; Bird, 1988, Science 242: 423-42; Huston et al, 1988, Proc. Nati Acad. Sci. USA 85: 5879 -5883; and Ward et al, 1989, Nature 334: 544-54) to produce single chain antibodies. The single chain antibodies are formed by binding heavy and light chain fragments of the Fv region through an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in E. coli can also be used (Skerra et al, 1988, Science 242: 1038-1041). Antibody fragments that recognize specific epitopes can be generated by known techniques. For example, such fragments include but are not limited to the F (ab ') 2 fragments that can be produced by pepsin digestion of the antibody molecule and the Fab fragments that can be generated by reducing the disulfide bridges of the F (ab) fragments. ')2. Once the nucleic acid sequence encoding an antibody has been obtained, the vector for antibody production can be produced by recombinant DNA technology using techniques well known in the art. Methods that are well known to those skilled in the art can be used to construct expression vectors containing the appropriate antibody coding sequence and transcriptional and translational control signals. These methods include, for example, recombinant DNA in vi tro techniques, synthetic techniques and in vivo genetic recombination. See, for example, the techniques described in Sambrook et al (1990, Molecular Cloning, A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, NY) and Ausubel et al (eds., 1998, Current Protocols in Molecular Biology, John Wiley &Sons, NY). An expression vector comprising the nucleotide sequence of an antibody or the nucleotide sequence of an antibody can be transferred to a host cell by standard techniques (eg, electroporation, liposomal transfection and calcium phosphate precipitation) and transfected cells from culture. then by conventional techniques to produce the antibody. In specific embodiments, the expression of the antibody is regulated by a constitutive, a specific inducible or tissue promoter. The host cells used to express the recombinant antibody can be either bacterial cells such as Escherichia coli or preferably, eukaryotic cells, especially for the expression of the complete recombinant immunoglobulin molecule. In particular, mammalian cells such as Chinese hamster ovary (CHO) cells, together with a vector such as the promoter element of the early gene of the major intermediate from human cytomegalovirus is an effective expression system for immunoglobulins (Foecking et al., 198, Gene 45: 101, Cockett et al, 1990, BioTeclinology 8: 2). A variety of host expression vector systems can be used to express the immunoglobulin antibodies. Such host expression systems represent carriers whereby antibody coding sequences can be produced and subsequently purified, but also represent cells that can be transformed or transfected with the appropriate nucleotide coding sequences, expressing an antibody immunoglobulin molecule in situ . this includes, but is not limited to, microorganisms such as bacteria (eg, E. coli and B. subtilis) transformed with the vectors, expression of the recombinant bacteriophage DNA, plasmid DNA or cosmid DNA containing coding sequences of immunoglobulin; yeast (e.g., Saccharomyces pichia) transformed with recombinant yeast expression vectors containing immunoglobulin coding sequences; insect cell systems infected with expression vectors of the recombinant virus (e.g., baculovirus) containing the immunoglobulin coding sequences; plant cell systems infected with expression vectors of the recombinant virus (eg, cauliflower mosaic virus (CaMV) and tobacco mosaic virus (TMV)) or transformed with the expression vectors of the recombinant plasmid (e .g., plasmid Ti) containing immunoglobulin coding sequences; or mammalian cell systems (e.g., COS, CHO, BH, 293, 293T, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (eg, metallothionein promoter). ) or from mammalian virus (eg, the late adenovirus promoter, the 7.5K promoter of the vaccine virus). In bacterial systems, a number of expression vectors may be advantageously selected depending on the intended use for the antibody to be expressed. For example, when a large amount of such a protein is to be produced, vectors that direct expression at high levels of the fusion protein products that are easily purified may be desirable. Such vectors include, but are not limited to, the expression vector pUR278 of E. coli (Ruther et al, 1983, EMBO J. 2: 1791), in which the antibody encoding the sequence can be ligated individually into the vector in the structure with the lac Z coding region whereby the fusion protein is produced, - the pIN vectors (Inouye &Inouye, 1985, Nucleic Acids Res. 13: 3101-3109; Van Heeke &Schuster, 1989, J Biol Chem 24: 5503-5509), and the like The PGEX vectors can also be used to express foreign bodies as fusion proteins with glutathione S-transferase (GST) In general, such fusion proteins are soluble and can easily purified from cells used by adsorption and binding to glutathione-agarose beads followed by elution in the presence of free glutathione.PGEX vectors are designed to include thrombin binding sites or factor Xa protease so that the product of the target gene clo Swimming can be released from the GST residue. In an insect system the nuclear pyrolysis virus Autographa californica (AcNPV) or the analogous virus from Drosophila Melanogaster is used as a vector to express foreign genes. The virus grows in the cells Spodoptera f ugiperda. The sequence encoding the antibody can be cloned individually into the non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter). In mammalian host cells, a number of viral based expression systems can be used. In cases where an adenovirus is used as an expression vector, the sequence encoding the antibody of interest can be ligated to a transcription / translation control complex, e.g., the late promoter and the tripartite loader sequence. This chimeric gene can then be inserted into the adenovirus genome by in vitro or in vivo recombination. Insertion into a non-essential region of the viral genome (e.g., El region or E3) results in a recombinant virus that is viable and capable of expressing the immunoglobulin molecule in infected hosts. (e.g., see Logan &Shenk, 1984, Proc. Nati, Acad. Sci. USA 81: 355-359). Specific initiation signals may also be required for the efficient translation of inserted antibody coding sequences. These signals include the ATG start codon and the adjacent sequences. In addition, the initiation codon must be in the phase with the reading structure of the desired coding sequence to ensure translation of the complete insert. These exogenous translational control signals and initiation codons can be from a variety of sources, both natural and synthetic. The efficiency of expression can be improved by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (See Bittner et al., 1987, Methods in Enzymol, 153: 51-544). In addition, a host cell strain can be selected to modulate the expression of the inserted sequences or modify or process the gene product in the specific manner desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have specific characteristics and mechanisms for post-translational processing and modification of the proteins and gene products. Appropriate cell lines or host systems can be selected to ensure correct modification and processing of the expressed foreign protein. For this purpose, eukaryotic host cells possessing the cellular machinery for the proper processing of transcription, glycosylation and major phosphorylation of the gene product can be used. Such mammalian host cells include but are not limited to CHO, VERY, BH, Hela, COS, MDCK, 293,293T, 3T3, W138, BT483, Hs578T, HTB2, BT20 and T47D, CRL7030 and Hs578Bst. For the long term high yield production of the recombinant proteins, stable expression is preferred. For example, cell lines that stably express an antibody can be designed. Instead of using expression vectors containing viral origins of replication, host cells can be transformed with controlled DNA by appropriate expression control elements (eg, promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.) and a selectable marker. After the introduction of the foreign DNA, the designed cells can be grown for 1-2 days in an enriched medium and then connected to a selective medium. The selectable marker in the recombinant plasmid confers resistance to the selection and allows the cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into the cell lines. This method can be used advantageously to design cell lines expressing the antibody. Such designed cell lines may be particularly useful in the systematic screening and evaluation of tumor antigens that interact directly or indirectly with the antibody. Various screening systems can be used, including but not limited to thymidine kinase of herpes simplex virus (Wigler et al., 1977, Cell 11: 223), hypoxanthine-guanine phosphoribotransferase (Szybalska & amp;; Szybalski, 192, Proc. Nati Acad. Sci. USA 48: 202) and phosphoribosyltransferase adenine (Lowy et al, 1980, Cell 22: 817) genes that can be used in the tk-, hgprt- or aprt- cells respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: DHFR that confer resistance to methotrexate (Wigler et al, 1980, Proc Nati Acad Sci USA 77: 357; O'Hare et al. 1981, Proc. Nati, Acad. Sci. USA 78: 1527); gpt, which confers resistance to mycophenolic acid (Mulligan &Berg, 1981, Proc Nati Acad Sci USA 78: 2072); neo, which confers resistance to the aminoglycoside G-418 (Clinical Pharmacy 12: 488-505; Wu and Wu, 1991, Biotherapy 3: 87-95; Tolstoshev, 1993, 'Ann.Rev.Pharmacol.Toxicol., 32: 573-596; Mulligan, 1993, Science 260: 926-932; and Morgan and Anderson, 1993, Ann. Rev. Biochem. 62: 191-217; May, 1993, TIB TECH 11 (5).-155-215) and hygro, that confers resistance to hygromycin (Santerre et al, 1984, Gene 30: 147). The methods commonly known in the art of recombinant DNA technology that can be used are as described in Ausubel et al (eds., 1993, Current Protocols in Molecular Biology, John Wiley &Sons, NY; Kriegler, 1990, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY, and Chapters 12 and 13, Dracopoli et al (eds), 1994, Current Protocols in Human Genetics, John Wiley &Sons, NY; Colberre-Garapin et al, 1981, J. Mol Biol. 150: 1). The levels of expression of an antibody can be increased by amplification of the vector (for a review, see Bebbington and Hentschel, The use of vector based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol. Academic Press, New York, 1987) When a marker in the marker system expressing an antibody is amplified, an increase in the level of the inhibitor present in the culture of the host cell will increase the copy number of the marker gene. the amplified region is associated with the nucleotide sequence of the antibody, the antibody production will also increase (Crouse et al, 1983, Mol Cell, Biol 3: 257) The host cell can be co-transfected with two expression vectors, the first vector encoding a heavy chain derived from polynucleotide and the second vector encoding a light chain derived from polypeptide.The two vectors may contain identical selectable markers that allow equal expression of heavy and light chain polypeptides. Alternatively, a single vector can be used to encode both heavy and light chain polypeptides. In such situations, the light chain must be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, 1986, Nature 322: 52, Kohier, 1980, Proc Nati, Acad Sci. USA 77: 2197). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA. Once the antibody has been expressed recombinantly it can be purified using any method known in the art for purification of an antibody, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after administration). Protein A, and column chromatography classification), centrifugation, differential solubility, or any other standard technique for protein purification. In yet another exemplary embodiment, the antibody is a monoclonal antibody. In any case, the hybrid antibodies have a dual specificity, preferably with one or more specific binding sites for the selection hapten or one or more specific binding sites for a target antigen, for example, an antigen associated with a tumor, a autoimmune disease, an infectious organism, or another disease state. 4.5.2 PRODUCTION OF ANTIBODIES The production of antibodies will be illustrated with reference to anti-CD30 antibodies but it will be apparent to those skilled in the art that antibodies to other members of the TNF receptor family can be produced and modified in a similar manner. The use of CD30 for the production of antibodies is only exemplary and is not intended to be limiting. The CD30 antigen to be used in the production of antibodies can be, e .g. , a soluble form of the extracellular domain of CD30 or a portion thereof, containing the desired epitope. Alternatively, cells expressing CD30 on its cell surface (eg, L540 (Hodgkin lymphoma derived from the cell line with a T cell phenotype) and L428 (Hodgkin lymphoma derived from the cell line with a B cell phenotype) can Other forms of CD30 useful for generating antibodies will be apparent to those of skill in the art In another exemplary embodiment, the ErbB2 antigen can be used for the production of antibodies, perhaps be, e.g., a soluble from the extracellular domain of ErbB2 or a portion thereof, containing the desired epitope.Alternatively, cells that express ErbB2 on its cell surface (e.g., transformed NIH-3T3 cells to overexpress ErbB2, - or a carcinoma cell line such as SK-BR-3 cells, see Stancovski et al., Proc. Nati, Acad. Sci. USA 88: 8691-8695 (1991) can be used to generate antibodies Other forms of ErbB2 useful for generating antibodies will be apparent to those of skill in the art. (i) Polyclonal antibodies Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (se) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e .g. , Californian limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional agent or derivatizing agent, eg, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (a through lysine residues), glutaraldehyde, succinic anhydride, SOC] 2, or RXN = C = NR, wherein R and R1 are different alkyl groups. Animals were immunized against the antigen, immunogenic conjugates, or derivatives by combining, eg, 100 μg or 5 μg of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally in multiple sites One month later the animals were boosted with 1/5 to 1/10 of the original amount of peptide or conjugate in complete Freund's adjuvant by subcutaneous injection at multiple sites. Seven to 14 days later the animals were bled and the serum was analyzed for the antibody titer. The animals were reinforced until the title was leveled. Preferably, the animals were boosted with the conjugate of the same antigen, but conjugated to a different protein and / or through a different cross-linking reagent. The conjugates can also be made in recombinant cell cultures as protein fusions. Also, aggregation agents such as alum are suitably used to improve the immune response. (ii) Monoclonal Antibodies Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may occur in minor amounts. Thus, the "monoclonal" modifier indicates the character of the antibody as being not a mixture of separate antibodies. For example, monoclonal antibodies can be made using the hybridoma method first described by Kohier et al. , Nature, 256,495 (1975), or can be made by recombinant DNA methods (U.S. Patent No. 4816567). In the hybridoma method, a mouse or other appropriate host animal, such as a hamster, is immunized as described above to produce lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes can be immunized in vi tro. The lymphocytes then fuse with myeloma cells, - - using a suitable fusion agent such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp.59-103 (Academic Press, 1986) .The hybridoma cells thus prepared are seeded and they grow in a suitable culture medium which preferably contains one or more substances that inhibit the growth or survival of the original unfused myeloma cells, for example, if the original myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT). or HPRT), the culture medium for the hybridomas will typically include hypoxanthine, aminopterin, and thymidine (HAT medium), whose substances prevent the growth of HGPRT-deficient cells.The preferred myeloma cells are those that fuse efficiently, support high stable levels of antibody production by the cells that produce selected antibodies, and are sensitive to a medium such as the HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from mouse tumors MOPC-21 and MPC-1 1 available from the Salk Institute Cell Distribution Center, San Diego, California USA, and SP-cells. 2 or X63-Ag8-653 available from the American Type Culture Collection, Rockville, Maryland USA. Human myeloma and mouse-human heteromyeloma cell lines have also been described for the production of human monoclonal antibodies (Kozbor, J. "Immunol., 133: 3001 (1984); and Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987). The ure medium in which the hybridoma cells grow is analyzed for the production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of the monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunosorbent assay (ELISA). The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al, Anal Biochem. , 107: 220 (1980). After the hybridoma cells are identified to produce antibodies of the desired specificity, affinity and / or activity, the clones can be subcloned by limiting the dilution and growth procedures by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, p. 59-103 (Academic Press, 1986) The ure medium suitable for this purpose includes, for example, D-MEM or RPMI-1640 medium.In addition, the hybridoma cells can grow in vivo as ascites tumors in a animal The monoclonal antibodies secreted by the subclones are suitably separated from the ure medium, ascites fluid, or serum by conventional methods of antibody purification such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography. The DNA encoding the monoclonal antibodies is easily isolated and sequenced using conventional procedures (e. G., By using oligonucleotide probes that are capable of specifically binding to genes encoding the heavy and light chains of murine antibodies). Hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA can be placed in expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary cells (CHO), or myeloma cells that do not otherwise produce antibody protein, to obtain the synthesis of the monoclonal antibodies in the recombinant host cells. Revised articles on recombinant expression in bacteria of DNA encoding the antibody include Skerra et al, Curr. Opinion in Immunol. , 5: 256-262 (1993) and Plückthun, Immunol Revs. , 130: 151-188 (1992). In a further embodiment, the monoclonal antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al, Nature, 348: 552-554 (1990). Clackson et al. , Nature, 352: 624-628 (1991) and Marks et al. , J. Mol. Biol. , 222: 581-597 (1991) describes the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity human antibodies (nM range) by redistribution (Marks et al, BiolTechnology, 10: 779-783 (1992), as well as combinational infection and in vivo recombination as a strategy to construct very large phage libraries (Waterhouse et al, Nuc.Acids.Res., 21: 2265-2266 (1993). Thus, these techniques are viable alternatives to traditional monoclonal antibody hybridoma techniques for the isolation of monoclonal antibodies. DNA can also be modified, for example, by substituting the coding sequence for human heavy chain and light chain constant domains instead of homologous murine sequences (U.S. Patent No. 4816567; and Morrison, et al., (1984) Proc. Nati, Acad. Sci. USA 81: 6851), or by covalently linking to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
Typically such non-immunoglobulin polypeptides are replaced by constant domains of an antibody, or are substituted by variable domains of an antigen combining site of an antibody to create a chimeric bivalent antibody comprising a combination site having specificity for an antigen and another antigen combining site that has specificity for a different antigen. (iii) Humanized Antibodies A humanized antibody can have one or more amino acid residues introduced therein from a non-human source. These non-human amino acid residues are often referred to as "imported" residues, which are typically taken from an "imported" variable domain. Humanization can be performed essentially following the method of Winter et al. (Jones et al., Nature 321: 522-525 (1986); Riechmann et al. , Nature, 332: 323-327 (1988); Verhoeyen et al. , Science 239: 1534-1536 (1988), by substituting the hypervariable region sequences for the corresponding sequences of a human antibody. Accordingly, such "humanized" antibodies are chimeric antibodies (U.S. Patent No. 4,816,567) wherein substantially less than an intact human variable domain has been replaced by the corresponding sequence of a non-human species. In practice, humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some FR residues are replaced by residues from analogous sites in rodent antibodies. In the choice of human variable domains, both light and heavy, which can be used to make humanized antibodies, it is very important to reduce the antigenicity. According to the so-called "best adapted" method, the variable domain sequence of the rodent antibody is systematically detected against the entire library of known human variable domain sequences. The human sequences that are closest to that of the rodent are then accepted as the human structure region (FR) for the humanized antibody (Sims et al., J. Immunol, 151: 2296 (1993); Chothia et al., J. Mol. Biol., 196: 901 (1987) Another method uses a region of particular structure derived from the consensus sequence of all human antibodies of a particular subgroup of light and heavy chains. for several different humanized antibodies (Cárter et al., Proc Nati Acad Sci USA, 99: 4285 (1992), Presta et al., J. Immunol, 151: 2623 (1993)). Antibodies can be humanized with high affinity retention for antigen and other favorable biological properties Humanized antibodies can be prepared by processes of analysis of the original sequences and several conceptual humanized products using three-dimensional models of the original sequences. and humanized. Three-dimensional immunoglobulin models are commonly available and are familiar to those of skill in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of the selected candidate immunoglobulin sequences. The inspection of these deployments allows the analysis of the probable role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of the residues that influences the ability of the candidate immunoglobulin to bind to its antigen. In this form, the FR residues can be selected and combined from the receptor and import sequences in order to achieve the desired characteristic of the antibody, such as increased affinity for the target antigen (s). the hypervariable region are directly and more substantially involved in influencing antigen binding Various forms of the humanized antibody are contemplated For example, the humanized antibody may be an antibody fragment, such as a Fab.
Alternatively, the humanized antibody can be an intact antibody, such as an IgGl antibody. The Examples describe the production of an exemplary humanized anti-ErbB2 antibody. The humanized antibody can, for example, comprise non-human hypervariable region residues incorporated in a human variable heavy domain and can further comprise a structure region (FR) substitution at a position selected from the group consisting of 69H, 71H and 73H using the variable domain numbering system established in Kabat et al. , Sequences of Proteins of Immunological Interest, 5a. Ed. Public Health Service, National Institutes of Health, Bethesda, MD (1991). In another embodiment, the humanized antibody comprises FR substitutions at two or all positions 69H, 71H and 73H. Another Example describes the preparation of the purified trastuzumab antibody of the HERCEPTIN® formulation. (iv) Human Antibodies As an alternative to humanization, human antibodies can be generated. For example, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of production of endogenous immunoglobulins. For example, it has been described that the homozygous deletion of the region gene linked to the antibody heavy chain (JH) in chimeric and germline mutant mice results in complete inhibition of the production of endogenous antibodies. The transfer of such a genetic ordering of human germline immunoglobulins in such a germline mutant mouse will result in the production of human Antibodies upon challenge of the antigen. See, e .g. , Jakobovits et al. , Proc. Nati Acad. Sci. USA, 90: 2551 (1993); Jakobovits et al. , Nature, 362-255-258 (1993); Bruggermann et al, Year in Immuno. , 7:33 (1993); and US Patents. Nos. 5,591,669, 5,589,369 and 5,545,807. Alternatively, the phage display technology (McCafferty et al, Nature 348: 552-553 (1990) can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable domain (V) gene repeats of immunized donors. According to this technique, the V domain genes of the antibody are cloned into the structure in either a major or minor protein coat gene of a filamentous bacteriophage, such as M13 or fd, and are deployed as functional antibody fragments over the surface of the phage particle, because the filamentous particles contain a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in the selection of the gene encoding the antibody that exhibits those properties Thus, the phage mimic some of the properties of the B cell. The phage display can be performed in a variety of e formats; for review see, e.g., Johnson, Kevin S. and Chiswell, David J., Current Opinion in Structural Biology 3: 564-571 (1993). Several sources of V-gene segments can be used for phage display. Clackson et al, Nature, 352,624-628 (1991) isolated a diverse array of anti-oxazolone antibodies from a small randomized combinational library of V genes derived from the spleens of immunized mice. A repertoire of V genes from immunized human donors can be constructed and antibodies can be isolated for a diverse array of antigens (including autoantigens) essentially following the techniques described by Marks et al, J. Mol. Biol. 222: 581-597 (1991), or Griffith et al, EMBO J 12: 725-734 (1993). See also, the Patents of E.U. Nos. 5565332 and 5573905. As discussed above, human antibodies can also be generated by activated B cells in vitro (see U.S. Patent Nos. 5567610 and 5229275). Human anti-CD30 antibodies are described in the U.S. Patent Application. Series No. 10 / 338,366. (v) Antibody Fragments Several techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived through the proteolytic digestion of insect antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24: 107-117 (1992)).; and Brennan et al, Science, 229: 81 (1985). However, these fragments can now be produced directly by recombinant host cells. For example, the antibody fragments can be isolated from antibody phage libraries, discussed above. Alternatively, Fab '-SH fragments can be recovered directly from E. coli and chemically coupled to form F (ab') 2 fragments (Cárter et al, Bio / Technology 10: 163-167 (1992). According to another procedure, the fragments F (ab ') 2 can be isolated directly from cultures of recombinant host cells. Other techniques for the production of antibody fragments will be apparent to expert practitioners. In other embodiments, the antibody of choice is a Fv fragment of a strand (scFv). See WO 93/16185; the Patent of E.U. No. 5,571,894; and the U.S. Patent. No. 5,587,458. The antibody fragment can also be a "linear antibody", e.g., as described in the U.S. Patent. No. 5,641,870 for example. Such linear antibody fragments may be monospecific or bispecific. (vi) Bispecific Antibodies Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes. Exemplary bispecific antibodies can bind to two different epitopes of the CD30 protein. Alternatively, an anti-CD30 arm can be combined with an arm that binds to Fe receptors for IgG (Fc? R), such as Fc? RI (CD64), Fc? RII (CD32) and Fc? RIII (CD 16) a In order to focus the cellular defense mechanisms towards the cell expressing CD30. Bispecific antibodies can also be used to localize cytotoxic agents to cells expressing CD30. The traditional production of full-length bispecific antibodies is based on the co-expression of two heavy chain-immunoglobulin light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305: 537-539 (1983). Due to the randomization of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mre of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather difficult, and the yield of the product is low. Similar procedures are described in WO 93/09829, and in Traunecker et al. , EMBO J., 10: 3655-3659 (1991). According to a different procedure, the variable domains of antibody with the desired binding specificities (antibody-antigen combining sites) are fused to the immunoglobulin constant domain sequences. The fusion preferably meets an immunoglobulin heavy chain constant domain, comprising at least part of the joint, CH2, and CH3 regions. it is preferred to have the first heavy chain constant region (CH1) containing the site necessary for the light chain linkage present in at least one of the fusions. The DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain, are inserted into the separate expression vectors, and co-transfected into a suitable host organism. This provides for greater flexibility in adjusting the mutual proportions of the three polypeptide fragments in embodiments where unequal proportions of the three polypeptide chains used in the construction provide optimal yields. However, it is possible to insert the coding sequence for two or all three polypeptide chains into an expression vector when the expression of at least two polypeptide chains in equal proportions results in high yields or when the proportions are not particular meaning.
In one embodiment of this method, the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a binding specificity in one arm, and a hybrid immunoglobulin light chain-heavy chain pair (which provide a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations., as the presence of a light chain of immunoglobulin in only one half of the bispecific molecule provides an easy form of separation. This procedure is described in WO 94/04690. For further details to generate bispecific antibodies, see, for example, Suresh et al. , Methods in Enzymology, 121: 210 (1996). According to another procedure described in the U.S. Patent. No. 5,731,168, the interface between a pair of antibody molecules can be designed to maximize the percentage of heterodimers that are recovered from the culture of recombinant cells. The preferred interface comprises at least part of the CH3 domain of an antibody constant domain. In this method, one or more small amino acid side chains of the interface of the first antibody molecule is replaced with larger side chains (e.g., tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large lateral chain (s) are created at the interface of the second antibody molecule by replacing the large amino acid side chains with small ( e .g., alanine or threonine). This provides a mechanism to increase the performance of the heterodimer over other unwanted byproducts such as homodimers. Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical bonds. Brennan et al. , Science, 229: 81 (1985) describes a procedure in which intact antibodies are cleaved proteolytically to generate F (ab ') 2 fragments. These fragments are reduced in the presence of sodium arsenite of the complexing agent to stabilize neighboring dithiols and prevent the formation of intramolecular disulfide. The generated Fab 'fragments are then converted to thionitrobenzoate derivatives (TNB). One of the Fab' -TNB derivatives is then recovered for the Fab '-thiol by reduction with mercaptoethylamine and mixed with an equimolar amount of the other Fab' derivative - TNB to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes. Recent advances have facilitated the direct recovery of Fab'-SH fragments from E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al. , J. Exp. Med., 175: 217-225 (1992) describes the production of an F (ab ') 2 molecule of fully humanized bispecific antibody. Each Fab 'fragment was secreted separately from E. coli and subjected to directed chemical coupling in vi tro to form the bispecific antibody. Various techniques for making and isolating bispecific antibody fragments directly from the culture of recombinant cells have also been described. For example, bispecific antibodies have been produced using leucine closure. Kostelny et al. , J. Immunol. , 148 (5) .1547-1553 (1992). Peptides from the leucine lock of the Fos and Jun proteins were linked to the Fab 'portions of two different antibodies by genetic fusion. The antibody homodimers were reduced in the region of articulation to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be used for the production of antibody homodimers. The "dicuerpo" technology described by Hollinger et al. , Proc. Nati Acad. Sci. USA, 90: 6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a variable domain of heavy chain - (VH) connected to a light chain variable domain (VL) by a link that is too short to allow pairs to be formed between the two domains in the same chain. According to the above, the VH and VL domains of one fragment are forced to pair with the VL and VH domains of another fragment, thereby forming two antigen binding sites. Another strategy for making bispecific antibody fragments by using Fv dimer (sFv) of a chain has also been reported. See, Gruber et al. , J. Immunol. , 152: 5368 (1994). Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al. , J. Immunol. 147: 60 (1991). (vii) Other modifications of amino acid sequence. Modification (s) of amino acid sequence of the antibodies described herein are contemplated. For example, it may be desirable to improve the binding affinity and / or other biological properties of the antibody. The amino acid variants of the antibodies are prepared by introducing appropriate nucleotide changes in the antibody nucleic acid or by peptide synthesis. Such modifications include, for example, cancellations and / or insertions and / or substitutions of residues within the amino acid sequences of the antibody. Any combination of cancellation, insertion and replacement is done to reach the final construction, provided that the final construction has the desired characteristics. The amino acid changes can also alter the post-translational processes of the antibody, such as changing the number or position of the glycosylation sites. A useful method for the identification of certain residues or regions of the antibody are the favored locations for mutagenesis is called "alanine scanning mutagenesis" as described by Cunningham and Wells Science, 244: 1081-1085 (1989). Here, a waste or group of the target waste is identified. { e .g. , charged residues such as arg, asp, his, lys, and glu) and are replaced by a neutral or negatively charged amino acid (more preferably alanine or polyalanine) to affect the interaction of the amino acids with the antigen. These amino acid locations demonstrate the functional sensitivity for corrected substitutions then when introducing in addition or other variants into, or for, substitution sites. Thus, while the site is predetermined to introduce an amino acid sequence variation, the nature of the mutation per se need not be predetermined. For example, to analyze the performance of a mutation at a given site, ala, scanning or random mutagenesis is conducted at the target codon or region and the expressed variants of the antibody are screened for the desired activity. The amino acid sequence insertions include amino and / or carboxy terminal fusions that vary in length from a residue to polypeptides containing one hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an amino acid with a residue or an antibody fused to a cytotoxic polypeptide. Other insertional variants of the antibody molecule include fusion of N or C terminals of the antibody to an enzyme. { e. g. , for ADEPT) or a polypeptide that increases the half-life of the antibody serum. Another type of variants is a variant of amino acid substitution. These variants have at least one amino acid residue in the antibody molecule replaced by a different residue. These sites of greatest interest for substitutional mutations include hypervariable regions, but FR alterations are also contemplated. Substantial modifications are made in the biological properties of the antibody by selecting substitutions that differ significantly in their effect in maintaining (a) the structure of the polypeptide backbone in the area of substitution, for example, as a non-helical sheet conformation, (b) the change or hydrophobicity of the molecule at the target site, or (c) the volume of the side chain. The residues that occur naturally are divided into groups based on the common properties of the side chain: (1) hydrophobic: norleucine, met, ala, val, leu, ile; (2) neutral hydrophilic: cys, ser, thr; (3) acidic: asp, glu; (4) basic: asn, gln, his, lys, arg; (5) residues that influence the orientation of the chain: gly, pro; and (6) aromatic: trp, tyr, phe. Non-conservative substitutions will cause the exchange of a member of one of these classes by another class. A particularly preferred type of substitution variants include substituting one or more hypervariable region residues of an original antibody (eg, a human or humanized antibody). In general, the resulting variant (s) selected for further development will have improved biological properties relative to the original antibody from which they are generated. A convenient way to generate such substitutional variants includes affinity maturation using phage display. Briefly, several hypervariable region sites (e.g., 6-1 sites) are mutated to generate all possible amino acid substitutions at each site. The antibody variants thus generated are deployed in a monovalent form from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage display variants are then systematically detected for their biological activity. { e .g. , link affinity) as described herein. In order to identify candidate hypervariable region sites for modification, alanine scanning mutagenesis can be performed to identify the hypervariable region residues that contribute significantly to antigen binding. Alternatively, or additionally, it may be beneficial to analyze a crystal structure of an antigen-antibody complex to identify the points of contact between the antibody and the antigen. Such contact residues and the surrounding residues are candidates for substitution according to the techniques elaborated herein. One such variant is generated, the panel of variants is screened as described herein and antibodies with superior properties in one or more relevant assays can be selected for further development. It may be desirable to modify the antibody of the invention with respect to the function of the effector, e. g. , in order to improve the median cytotoxicity by the antigen-dependent cell (ADCC) and / or complement-dependent cytotoxicity (CDC) of an antibody. This can be achieved by introducing one or more substitutions in a Fe region of the antibody. Alternatively or additionally, the cysteine residue (s) may be introduced into the Fe region, thereby allowing the formation of the interchain chain disulfide bond in this region. The homodimeric antibody thus generated can have enhanced internalization and / or complement-mediated cell lysate and improved antibody-dependent cell cytotoxicity (ADCC). See Caron et al., J. Exp Med. 176: 1191-1195 (1992) and Shopes, B. J. Immunol. 148: 2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional crosslinking as described in Wolff et al. Cancer Research 53: 2560-2565 (1993). Alternatively, an antibody can be designed having dual FC regions and thereby can have improved complement lysis and enhanced ADCC capabilities. See Stevenson et al. Anti-Cancer Drug Design 3: 219-230 (1989). To increase the half-life of the antibody serum, a binding epitope to the salvage receptor can be incorporated into the antibody (especially an antibody fragment) for example, as described in the U.S. Patent. No. 5739277. As used herein, the term "salvage receptor binding epitope" refers to an epitope of the Fe region of an IgG molecule. (e.g., IgG ?, IgG2, IgG3, or IgG) which is responsible for increasing the serum half-life of the IgG molecule in vivo. (viii) Glycosylation variants Antibodies in the ADC of the invention can be glycosylated in conservative positions in their constant regions (Jefferis and Lund, (1997) Chem. Immunol. 65: 111-128; Wright and. Morrison, (1997) TibTECH 15: 26-32). the oligosaccharide side chains of the immunoglobulins affect the fusion of the protein (Boyd et al., (1996) Mol Immunol., 32: 1311-1318; Wittwe and Howard, (1990) Biochem. 29: 4175-4180), and the intramolecular interaction between the portions of the glycoprotein that can affect the conformation and the presented three-dimensional surface of the glycoprotein (Hefferis and Lund, supra; Wyss and Wagner, (1996) Current Opin. Bíotech 7: 409-416). Oligosaccharides can also serve to direct a given protein towards certain molecules based on specific recognition structures. For example, it has been reported that in agalactosylated IgG, the oligosaccharide residue exits out of the interspace. -CH2 and the acetylglucosamine residues at the N-terminus become available to bind to the protein that binds to the mannose - (Malhotra et al., (1995) Nature Med. 1: 237-243). Removed by glycopeptidase from the oligosaccharides of CAMPATH-1H (a recombinant humanized murine monoclonal IgGl antibody that recognizes the CDw52 antigen of human lymphocytes) produced in Chinese Hamster Ovary (CHO) cells results in a complete reduction in lysis-mediated lysis. the complement (CMCL) (Boyd et al., (1996) Mol.Immunol., 32: 1311-1318), although the selective removal of the residues of the sic acid using neurominidase results in the loss of DMCL. The glycosylation of antibodies that affects antibody-dependent cellular cytotoxicity (ADCC) has also been reported. In particular, CHO cells with the expression regulated by tetracycline of β (1,4) -N-acetylglucosaminyltransferase III (GnTIII), a glycosyltransferase that catalyzes the formation of bisection GlcNAc, was reported to have enhanced the activity of ADCC (Umana et al., (1999) Mature Biotech 17: 176-180). The glycosylation of antibodies which is typically either linked to N or linked to O, refers to the attachment of the carbohydrate residue to the side chain of an asparagine residue. The tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for the enzymatic binding of the carbohydrate residues to the asparagine side chain. Thus, the presence of any of these triplet sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the binding of one of the sugars N-acetylgalactosamine, galactose or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine can also be used. Antibody glycosylation variants are variants in which the glycosylation pattern of an antibody is altered. Altering means canceling one or more carbohydrate residues found in the antibody, adding one or more carbohydrate residues to the antibody, changing the composition of the glycosylation (glycosylation pattern), the degree of glycosylation, etc. The addition of glycosylation sites to the antibody is conveniently performed by altering the amino acid sequence such that it contains one or more of the triplet sequences described above (for the N-linked glycosylation sites). The alteration can be made by adding or replacing one or more serine or threonine residues to the original antibody sequence (for glycosylation sites linked to O). Similarly, removal of the glycosylation sites can be accomplished by altering amino acids within the natural glycosylation sites of the antibody. The amino acid sequence is usually altered by altering the underlying nucleic acid sequence. These methods include, but are not limited to, isolation from a natural source (in the case of variants of amino acid sequences that occur naturally) or preparation by oligonucleotide-mediated mutagenesis, PCR mutagenesis and cassette mutagenesis of a previously prepared variant or a version no antibody variant. Glycosylation (including the glycosylation pathway of the antibodies can be altered without altering the amino acid sequence or the underlying nucleotide sequence.) Glycosylation depends largely on the host cells used to express the antibody, since the type of cells used for the expression of recombinant glycoproteins, e.g., antibodies, as potential therapeutics, is rarely the native cell, significant variations in the glycosylation pattern of antibodies can be expected. See, e .g., Hse et al., (1997) J. Biol. Chem. 272: 9062-9070. In addition, for the choice of host cells, the factors that affect glycosylation during the recombinant production of antibodies include the mode of growth, the medium formulation, the density of the culture, oxygenation, pH, purification scheme and the like. Several methods have been proposed for altering the glycosylation pattern achieved in a particular host organism including introducing or overexpressing certain enzymes included in the production of oligosaccharides (U.S. Patent Nos. 5047335, 5510261, 5278299). Glycosylation, or certain types of glycosylation, can be enzymatically removed from the glycoprotein, for example, using endoglycosidase H (Endo H). In addition, the recombinant host cell can be genetically engineered, e. g. , making certain types of polysaccharides defective in processing. These and similar techniques are well known in the art. The glycosylation structure of the antibodies can be easily analyzed by conventional carbohydrate analysis techniques, including lectin chromatography, NMR, mass spectrometry, HPLC, GPC, compositional analysis of polysaccharides, sequential enzymatic digestion and HPAEC-PAD, which utilizes ion exchange at high pH to separate the oligosaccharides based on the change. Methods for releasing oligosaccharides for analytical purposes are also known, including, but not limited to, enzymatic treatment (commonly performed using peptide-N-glycosidase F / endo-β-galactosidase), elimination, using severe alkaline environment to release mainly the structures linked to 0, and chemical methods using anhydrous hydrazine to release oligosaccharides both linked to N and 0. - 4. 5.2a SYSTEMATIC DETECTION OF ANTIBODY-DRUG CONJUGATES (ADC) Transgenic animals and cell lines are particularly useful for systematically detecting drug antibody conjugates (ADCs) that have potential as prophylactic or therapeutic treatments of diseases or disorders including overexpression of proteins including Lewis Y, CD30, CD40, and CD70. Transgenic animals and cell lines are particularly useful in the routine screening of drug antibody conjugates (ADCs) that have potential as prophylactic or therapeutic treatments of diseases or disorders including HER2 overexpression (US6632979). The routine screening of a useful ADC may include administering ADC candidates through a range of doses to the transgenic animal, and analyzing at various time points for the effect (s) of the ADC on the disease or disorder being evaluated. . Alternatively or additionally, the drug may be administered before or simultaneously with exposure to an inducer of the disease, if applicable. The candidate ADC can be detected systematically in a serial and individual way or in parallel in a medium or high performance systematic detection format. The rate at which the ADC can be systematically detected by utility for prophylactic or therapeutic treatment of diseases or disorders is limited only by the speed of the methodology of synthesis or systematic screening, including the detection / measurement / analysis of data. One embodiment is a method of screening which comprises (a) transplanting cells from a stable renal cell cancer cell line to a non-human animal, (b) administering an ADC drug candidate to the non-human animal and (c) determining the ability of the candidate to inhibit the formation of tumors of the transplanted cell line. Another embodiment is a method of screening which comprises (a) contacting cells of a stable Hodgkin's disease cell line with a candidate ADC drug and (b) evaluating the ability of the ADC candidate to block activation of the CD40 ligand. Another embodiment is a method of screening which comprises (a) contacting cells of a stable Hodgkin's disease cell line with a candidate drug ADC and (b) evaluating the candidate's ability ADC to induce cell death. In one modality, the ability of the ADC candidate to induce apoptosis is evaluated. Another embodiment is a method of screening which comprises (a) transplanting cells from a stable cancer cell line to a non-human animal, (b) - - administering a candidate ADC drug to. non-human animal, and (c) determining the ability of the candidate to inhibit tumor formation of the transplanted cell line. The invention also relates to a method of systematically screening ADC candidates for the treatment of diseases or disorders characterized by overexpression of HER2 comprising (a) contacting cells of a stable breast cancer cell line with a candidate drug and (b) assess the ability of the ADC candidate to inhibit growth and (b) assess the ability of the ADC candidate to inhibit the growth of the stable cell line. Another embodiment is a method of screening which comprises (a) contacting cells of a stable cancer cell line with a candidate drug ADC and (b) evaluating the ability of the ADC candidate to block the activation of the HER2 ligand. In another modality, the ability of the ADC candidate to block the link to the heregulin is evaluated. In another modality, the ability of the ADC candidate to block tyrosine phosphorylation stimulated by the ligand is evaluated. Another embodiment is a method of screening which comprises (a) contacting cells from a stable cancer cell line with a candidate ADC drug and (b) evaluating the ability of the ADC candidate to induce cell death. In one modality, the ability of the ADC candidate to induce apoptosis is evaluated. Another embodiment is a method of screening which comprises (a) administering a candidate ADC drug to a transgenic non-human mammal overexpressing in its mammary gland cells a natural human HER2 protein or fragment thereof, wherein said transgenic mammal has stably integrated into its genome is a nucleic acid sequence that encodes a natural human HER2 protein or a fragment thereof that has the biological activity of a natural human HER2, operably linked to the transcriptional regulatory sequences that direct its expression to the gland mammary tumor, and develop a mammary tumor that does not respond or responds sparingly to the treatment of the anti-HER2 antibody, or to a non-human mammal that carries a tumor transplant from said transgenic non-human mammal; and (b) evaluating the effect of the ADC candidate on the target disease or disorder. Without limitation, the disease or disorder may be a cancer that overexpresses HER2, such as a breast cancer. ovary, stomach, endothelial, salivary gland, lung, kidney, colon, thyroid, pancreatic and bladder. The cancer is preferably breast cancer which expresses HER2 in at least about 500,000 copies per cell, more preferably, at least 2,000,000 copies per cell. The candidate ADC drugs may, for example, be evaluated for their ability to induce cell death and / or apoptosis, using assay methods known in the art and described below. In one embodiment, the ADC candidate is systematically detected upon administration to the transgenic animal over a range of doses, and to evaluate the physiological response of the animal to the compound over time. Administration can be oral, or by appropriate injection, depending on the chemical nature of the compound being evaluated. In some cases, it may be appropriate to administer the compound together with co-factors that could improve the efficiency of the compound. If the cell line derived from the target transgenic animal is used for the systematic screening of compounds useful for treating various disorders, test compounds are added to the cell culture medium at an appropriate time., and the cellular response to the compound is evaluated over time using the appropriate biochemical and / or histological analyzes. In some cases, it may be appropriate to apply the compound of interest to the culture medium together with co-factors that could improve the efficiency of the compound. Thus, an analysis is provided herein to identify ADC which targets and binds specifically to an objective protein, the presence of which correlates with an abnormal cellular function, and the pathogenesis of cell proliferation and / or differentiation that is causally related to the development of tumors. To identify an ADC that blocks the activation of the ligand of an ErbB receptor. { e .g. , ErbB2) can be determined the ability of the compound to block the binding of the ErbB ligand to the cells that express the ErbB (ErbB2) receptor (eg, conjugation with another ErbB receptor with which the ErbB receptor of interest forms a hetero -ErbB oligomer). For example, cells isolated from the transgenic animal overexpressing HER2 can be incubated and transfected to express another ErbB receptor (with which HER2 forms the hetero-oligomer), with the ADC and then exposed to labeled ErbB ligands. The ability of the compound to block the binding of the ligand to the ErbB receptor in the ErbB hetero-oligomer can then be evaluated. For example, inhibition of heterogulin binding (HRG) to breast tumor cell lines, which overexpress HER2 and are established from transgenic non-human mammals (eg mouse) in the present, can be performed by the ADC candidate using monostratified cultures on ice in a 24-well plate format. Anti-ErbB monoclonal antibodies can be added to each well and incubated for 30 minutes. Then rHRGßl? 77-224 labeled with 125I (25,000 cpm) can be added, and incubation can be continued for 4 to 6 hours. Dose response curves can be prepared and an IC50 value (cytotoxic activity) can be calculated for the compound of interest. Alternatively or additionally, the ability of an ADC to block tyrosine phosphorylation stimulated by the ErbB ligand of an ErbB receptor present in the ErbB hetero-oligomer can be assessed. For example, cell lines established from transgenic animals herein can be incubated with at least one ADC and then titrated for tyrosine phosphorylation activity dependent on the ErbB ligand using an anti-phosphotyrosine monoclonal antibody (which is optionally conjugated with a detectable label), The activation kinase receptor assay described in the US Patent No. 5766863 is also available to determine activation of the ErbB receptor and blocking that activity by the compound. In one embodiment, ADC that inhibits HRG stimulation of p80 tyrosine phosphorylation in MCF7 cells can be detected systematically as described below. For example, a cell line established from HER2 transgenic animals can be plated on 24-well plates and the compound can be added to each well and incubated for 30 minutes at room temperature.; then rHRGßl177_224 is added to each well to a final concentration of 0.2 nM, and the incubation can continue for approximately 8 minutes. The medium can be aspirated from each well, and the reactions can be stopped by the addition of 100 μl of SDS sample buffer (5% SDS, 25 mM DTT, and 25 mM Tris-HCl, pH 6.8). Each sample (25 μl) can be electrophoresed in a gradient gel of 4-12% (Novex) and then transferred to the polyvinylidene disulfide membrane. Antiphosphotyrosine immunoassays (at 1 μg / ml) can be developed and the intensity of the predominant reactive band at Mr -180,000 can be quantified by reflectance densitometry. An alternative method to evaluate the inhibition of receptor phosphorylation is the KIRA (receptor kinase activation) analysis of Sadick et al. , (1998) Jour. of Pharm. and Biomed. Anal. Some of the established monoclonal antibodies against HER2 that are known to inhibit the stimulation of HRG by the tyrosine phosphorylation pl80 can be used as a positive control in this assay. A dose response curve can be prepared for the inhibition of HRG stimulation of p8080 tyrosine phosphorylation as determined by reflectance densitometry and an IC50 can be calculated for the compound of interest. The effects of inhibition of growth of a test ADC on cell lines derived from a transgenic animal HER2 can be assessed, e. g. , essentially as described in Schaefer et al. , (1997) Oneogene 15: 1385-1394. According to this assay, the cells can be treated with a test compound at various concentrations for 4 days and stained with crystal violet or the Blue Alamar redox dye. Incubation with the compound may show a growth inhibitory effect on this cell line similar to that displayed by the monoclonal antibody 2C4 in NMA-NM-175 cells (Schaefer et al., Supra). In additional modalities, an endogenous HRG will not significantly reverse this inhibition. To identify growth inhibitory compounds that specifically target an antigen of interest, compounds that inhibit the growth of cancer cells overexpressing the antigen of interest derived from transgenic animals can be systematically detected, the assay described in the U.S. Patent may be performed. No. 5677171. According to this assay, the cancer cells which overexpress the antigen of interest are grown in a 1: 1 mixture of F12 medium and DMEM supplemented in 10% fetal bovine serum, glutamine and streptomycin penicillin. Cells are plated at 20,000 cells in a 35 mm cell culture dish (2 mls / 35 mm dish) and the test compound is added at various concentrations. After six days, the number of cells was counted in comparison to the untreated cells using a COULTER ™ cell counter. Those compounds that inhibit cell growth by about 20-100% or about 50-100% can be selected as growth inhibitory compounds. To select compounds that induce cell death, loss of membrane integrity as indicated by the absorption of e. g. , Pl, blue trípano or 77AAD can be evaluated in relation to the control. The absorption analysis of Pl uses cells isolated from tumor tissue of interest of a transgenic animal. According to this assay, the cells are cultured in Dulbecco's Modified Eagle's Medium (D-MEM): Ham's F-12 (50:50) supplemented with 10% inactivated heart FBS (Hyclone) and 2 mM L-glutamine. Thus, the assay is performed in the absence of complement and immune effector cells. The cells are seeded at a density of 3 x 10s per dish in 100 x 20 mm dishes and allowed to bind overnight. The medium is then removed and replaced with fresh medium only or medium containing various concentrations of the compound. The cells are incubated for a period of 3 days. After each monostratified treatment is washed with PBS and discarded by trypsination. The cells are then centrifuged at 1200 rpm for 5 minutes at 4 ° C, the pellets resuspended in 3 ml of cold Ca 2+ binding buffer (10 mM Hepes, pH 7.4, 140 mM -NaCl, 2.5 mM CaCl2) and aliquots in 12 x 75 mm tubes capped with 35 mm scrubber (1 ml per tube, group of 3 tubes per treatment) for removal of cell clusters. The tubes then receive Pi (10 μg / ml). Samples can be analyzed using a FACSCAN ™ flow cytometer and FACSCONVERT ™ CellQuest software (Becton Dickinson). Those compounds that induce statistically significant levels of cell death as determined by the absorption of Pl can be selected as compounds that induce cell death. In order to select compounds that induce apoptosis, an annexin binding assay can be performed using cells established from the tumor tissue of interest of the transgenic animal. The cells are cultured and seeded in dishes as discussed in the previous paragraph. The medium is then removed and replaced with fresh medium only or medium containing 10 μg / ml of drug antibody conjugate (ADC). After a three-day incubation period, the monostrates are washed with PBS and discarded by trypsinization. The cells are then centrifuged, resuspended in a Ca 2+ -linker, and aliquots are made in the tubes as discussed above for the cell death assay. The tubes then receive labeled annexin (e.g., annexin V-FITC) (1 μg / ml). Samples can be analyzed using a FACSCAN ™ flow cytometer and FACSCONVERT ™ CellQuest software (Becton Dickinson). Those compounds that induce statistically significant levels of annexin binding to the control can be selected as compounds that induce apoptosis. 4.5.3 IN VITRO CELLULAR PROLIFERATION TESTS In general, the cytotoxic or cytostatic activity of a drug antibody conjugate (ADC) is measured by: exposing the mammalian cells that have receptor proteins to the ADC antibody in a cell culture medium; culturing the cells for a period of about 6 hours to about 5 days; and measure cell viability. In vitro assays with base cells were used to measure the viability (proliferation), cytotoxicity and induction of apoptosis (caspase activation) of the ADC of the invention. The potency in vi tro of the drug antibody conjugate was measured by a cell proliferation assay (Example 18, Figures 7-10). The CellTiter-Glo® Luminescent Cell Viability Assay is a commercially available homogeneous assay method (Promega Corp., Madison, Wi), based on the recombinant expression of the luciferae Coleoptera.
(U.S. Patent No. 5583024; 5674713 and 5700670). This cell proliferation assay determines the number of viable cells in the culture based on the quantification - of the ATP present, an indicator of metabolically active cells (Crouch et al., (1993) J. Immunol., Meth. 160: 81- 88, U.S. Patent No. 6602677). The CellTiter-Glo® assay was conducted in a 96 well format, making it suitable for automated high throughput screening (HTS) (Cree et al., (1995) AntiCancer Drugs 6: 398-404). The homogeneous assay procedure includes adding the reagent alone (CellTiter-Glo® Reagent) directly to the cell culture in medium suspended in serum. The washed cells are removed from the medium and the multiple pipette steps are not required. The system detects as few as 12 cells / well in a 384-well format in 10 minutes after adding the reagent and mixing. The cells can be treated continuously with ADC, or they can be treated and separated from the ADC. In general, the cells briefly treated, i. e. 3 hours, show the potency effects of the cells treated continuously. The homogeneous "add-mix-measure" format results in cell lysis and the generation of a luminescent signal proportional to the amount of ATP present. The amount of ATP is directly proportional to the number of cells present in the culture. The CellTiter-Glo® assay generates a "brightness-type" luminescent signal, produced by the luciferase reaction, which has a half-life generally greater than five hours, depending on the type of cell and medium - used. Viable cells are reflected in relative luminescent units (RLU). The Beetle Luciferin substrate, is oxidized to the decarboxylate by the recombinant luciferaza of the firefly with the concomitant conversion of ATP to AMP and the digestion of the photons. The extended half-life eliminates the need for the use of reagent injectors and provides flexibility for the processing of batch or continuous modes of multiple plates. This cell proliferation assay can be used with several multi-well formats, e. g. , formats of 96 or 384 wells. The data can be registered by luminometer or CCD camera imaging device. The production of luminescence is present as relative units of light (RLU), measured over time.
Luciferase ATP + Luciferin + O2 Oxyluciferin + AMP + Ppi + CO2 + light The anti-proliferative effect of the drug antibody conjugates was measured by the cell proliferation assay, cell death in vitro against four different breast tumor cell lines (Figures 7-10). The IC 50 values were established for SK-BR-3 and BT-474 which are known to over-express the HER2 receptor protein. Table 2 shows the power measurements (IC50) of the exemplary drug antibody conjugate in the cell proliferation assay against SK-BR-3 cells. Table 2b shows the potency measurements (IC50) of the exemplary drug antibody conjugates in cell proliferation assays against BT-474 cells. Drug antibody conjugates: Trastuzumab-MC-vc-PAB-MMAF, 3.8 MMAF / Ab; Trastuzumab-MC- (N-Me) vc-PAB-MMAF, 3.9 MMAF / Ab; Trastuzumab-MC-MMAF, 4.1 MMAF / Ab; Trastuzumab-MC-vc-PAB-MMAE, 4.1 MMAE / Ab; Trastuzumab-MC-vc-PAB-MMAE, 3.3 MMAE / Ab; and Trastuzumab-MC-vc-PAB-MMAF, 3.7 MMAF / Ab do not inhibit the proliferation of MCF-7 cells (Figure 9). Antibody conjugates: Trastuzumab-MC-vc-PAB-MMAE, 4.1 Ab; Trastuzumab-MC-vc-PAB-MMAE, 3.3 MMAE / Ab; Trastuzumab-MC-vc-PAB-MMAF, 3.7 MMAF / Ab; Trastuzumab-MC-ve-PAB-MMAF, 3.8 MMAF / Ab; Trastuzumab-MC- (N-Me) vc-PAB-MMAF, 3.9 MMAF / Ab; and Trastuzumab-MC-MMAF, 4.1 MMAF / Ab do not inhibit the proliferation of MDA-MB-468 cells (Figure 10). The MCF-7 and MDA-MB-468 cells do not overexpress the HER2 receptor protein. Therefore, the anti-HER2 antibody drug conjugate of the invention shows selectivity for cells expressing HER2.
Table 2a SK-BR-3 cells Antibody Drug Conjugate IC50 (μg ADC / ml) H = trastuzumab bound through a cysteine [cys] except where noted - - Table 2b cells BT474 H = trastuzumab 7C2 = murine anti-HER2 antibody that binds to epitopes other than trastuzumab Fc8 = mutant that does not bind to FcRn Hg = "No joint" 4D5 humanized full-length, with chain-link cysteines heavy mutated to serinas. Expressed in E.coli (therefore not glycosylated). Anti-TF Fab = anti-tissue factor antibody fragment * activity against MDA-MB-468 cells In a surprising and unexpected discovery, the results of cell proliferation activity within the ADC in Tables 2a and 2b show in general that ADC with a low average number of drug residues per antibody showed efficiency, e .g. , IC50 < 0.1 μg ADC / ml. The results suggest that at least for the ADT trastuzumab, the optimal ratio of drug residues per antibody may be less than 8, and may be approximately 2 to approximately 5. 4.5.4 DEPURATION AND STABILITY OF PLASMA JN VIVO Purification was investigated and stability of the pharmacokinetic plasma of ADC in rats and cynomolgus monkeys. The plasma concentration was measured over time. Table 2c shows the pharmacokinetic data of drug conjugates of the antibody and other samples dosed in rats. The rats are non-specific models for the ErbB receptor antibodies, since the rat is not known to express the HER2 receptor proteins.
Table 2c Pharmacokinetics in Rats H = trastuzumab bound via a cysteine [cys] except where 2 mg / kg dose was noted except where noted - - AUC inf is the area under the plasma concentration-time curve from the hour of dosing to infinity and is a measure of the total exposure to the measured entity (ADC drug). CL is defined as the volume of purified plasma of the entity measured in unit of time and is expressed when normalizing to body weight. The term Tl / 2 is the average life of the drug in the body measured during its phase of elimination. The term% Conj. is the relative amount of ADC compared to the total antibody detected, by separating the ELISA immunoaffinity tests ("Analytical Methods for Biotechnology Products", Ferraiolo et al., p85-98 in Drug Pharmacokinetics (1994) PG Welling - - and LP Balant, Eds., Handbook of Experimental Pharmacology, Vol. 110, Springer-Verlag.The% Conj. calculation is simply AUCinf of ADC - = - AUCinf total Ab and is a general indicator of linker stability although other factors and Mechanisms may be in effect Figure 11 shows a graph of a plasma concentration clearance study after administration of the antibody drug conjugates: H-MC-vc-PAB-MMAF-TEG and H-MC- vc-PAB-MMAF for Sprague-Dawley rats Antibody and total ADC concentrations were measured over time Figure 12 shows a graph of a two-stage plasma concentration clearance study where ADC was administered at different concentrations. doses and concentrations of the antibody and total ADC that were measured over time. IN VIVO EFFICACY The in vivo efficacy of the ADC of the invention was measured by a high expression mouse model of transgenic explant HER2. An allograft was propagated from the transgenic Fo5 mmtv mouse that does not respond, or responds poorly, to HERCEPTIN® therapy. The subjects were treated once with ADC and monitored for 3-6 weeks to measure the time in which the tumor grows twice, cell deletion log and tumor shrinkage. Then, dose-response and multiple-dose experiments were conducted. The tumors originated rapidly in transgenic mice that expressed a mutationally activated form of neu, the HER2 rat homologue, but the HER2 that was overexpressed in breast cancers did not mutate and the tumor formation is much less robust in transgenic mice than they express HER2 without mutating (Webster et al., (1994) Semin. Cancer Biol. 5: 69-76). To improve tumor formation with untreated HER2, transgenic mice were produced using a HER2 cDNA plasmid in which an upstream ATG was surmounted in order to prevent the initiation of translation to such upstream ATG codons, which could otherwise thus reducing the frequency of translation initiation from the authentic downstream HER2 start codon (e.g., see Child et al., (1999) J. Biol. Chem. 274: 24335-24341). Additionally, a chimeric intron was added at the 5 'end which will also improve the level of expression as reported previously (Neuberger and Williams (1988) Nucleic Acids Res. 16: 6713; Buchman and Berg (1988) Mol. Cell. Biol. 8: 4395; Brinster et al. (1988) Proc. Nati, Acad. Sci. USA 85: 836). The chimeric intron was derived from a Promega vector, the mammalian expression vector pCI-neo (bp 890-1022). The 3 'cDNA end is flanked by human growth hormone exons 4 and 5 and polyadenylation sequences. In addition, FVB mice were used because this strain is more susceptible to tumor development. The promoter was used from MMTV-LTR to ensure the expression of tissue-specific HER2 in the mammary gland. The animals were fed the AIN 76A diet in order to increase the susceptibility to tumor formation (Rao et al (1997) Breast Cancer Res. And Treatment 45: 149-158). Table 2d Tumor measurements in allograft mouse model - MMTV-HER2 Fo5 Breast tumor, nude nude mouse single dose at day 1 (T = 0) except when entering H = bound trastuzumab via a cysteine [cys] except when write down 7c2 = murine anti HER2 antibody that binds to a different epitope to trastuzumab. Fe8 = mutant that does not bind to FcRn Hg = "Without articulation" 4D5 humanized of total length, with cysteines of heavy chain articulation mutated to serines. Expressed in E. coli (therefore not glycosylated) 2H9 = Anti-EphB2R 11D10 = Antí-0772P The term Ti is the number of animals in the study group with tumor at T = 0 -e- the total number of animals in the group. The term PR is the number of animals that achieve a partial remission of the tumor -J- animals with tumor at T = 0 in the group. The term CR is the number of animals that achieves a complete remission of the tumor - = - animals with tumor at T = 0 in the group. The term cell deletion Log is the time in days for the tumor volume to double-the time in days for the volume of the control tumor to be doubled by the 3.32X time for the tumor volume to double in the animals of control (dosed with the Vehicle). The calculation for cell deletion log takes into account the delay resulting from tumor growth from the treatment and the time at which the tumor volume of the control group is doubled. The anti-tumor activity of ADC is classified with the values of cellular elimcuon log of: ++++ > 3.4 (highly active) +++ = 2.5-3.4 ++ = 1.7-2.4 + 1.0-1.6 inactive = 0 Figure 13 shows the mean change in tumor volume over time in athymic nude mice with allografts of the mammary tumor MMTV-HER2 Fo5 dosed on Day 0 with: Vehicle, Trastuzumab-Mc-vc-PAB-MMAE (1250 μg / m2) and Trastuzumab-MC-vc-PAB-MMAF (555 μg / m2). (H = Trastuzumab). The growth of tumors was retarded by treatment with ADC compared to the control growth level (Vehicle). Figure 14 shows the mean tumor volume change over time in athymic nude mice with mammary tumor allografts MMTV-HER2 Fo5 dosed on Day 0 with 10 mg / kg (660 μg / m2) of Trastuzumab-MC- MMAE and 1250 μg / m2 of Trastuzumab-MC-vc-PAB-MMAE. Figure 15 shows the mean tumor volume change over time in nude - - nude mice with mammary tumor allografts MMTV-HER2 Fo5 dosed with 650 μg / m2 of Trastuzumab-MC-MMAF. Table 2d and Figures 13-15 show that ADC has strong anti-tumor activity in the allograft of a HER2 positive tumor (Fo5) that originally originated in a MMTV-HER2 transgenic mouse. The antibody alone (e.g., Trastuzumab) has no significant anti-tumor activity in this model (Erickson et al, U.S. Patent No. 6632979). As illustrated in Figures 13-15, the growth of the tumors was retarded by treatment with ADC compared to the growth level of the control (Vehicle). In a surprising and unexpected discovery, the anti-tumor activity in vivo resulting from the ADC in Table 2d generally shows that ADC with a low average number of drug residues per antibody showed efficacy, eg, time when the tumor is doubled > 15 days and cellular removal log media > 1.0. Figure 16 shows for the antibody drug conjugate, trastuzumab-MC-vc-PAB-MMAF, the mean tumor volume decreased and did not progress where the MMAF: trastuzumab ratio was 2 and 4, while the tumor progressed to a ratio of 5.9 and 6 but at a lower rate than the Vehicle (shock absorber). The rate of tumor progression in this mouse xenograft model was approximately the same, i.e., 3 days, for the Vehicle - and trastuzumab. The results suggest that at least for ADT trastuzumab, the optimal ratio of drug residues per antibody can be from less than about 8 and can be from about 2 to about 4. 4.5.5 TOXICITY IN RODENTS Antibody and antibody drug conjugates a control without ADC, "Vehicle" were evaluated in a rat model of acute toxicity. The toxicity of ADC was investigated by the treatment of male and female Sprague-Dawley rats with the ADC and the subsequent inspection and analysis of the effects on various organs. The total observations included changes in body weights and signs of injuries and hemorrhages. The clinical parameters of pathology (serum chemistry and hematology), histopathology and necropsy were conducted on dosed animals. It should be considered that the weight loss or weight change relative to animals dosed with the Vehicle, in animals after dosing with ADC is a complete and general indicator of systemic or localized toxicity. Figures 17-19 show the effects of several ADCs and controls (Vehicle) after dosing on the body weight of the rats. Hepatotoxicity was measured by elevated liver enzymes, increased numbers of mitotic and apoptotic figures and hepatocyte necrosis. Hematolymphoid toxicity was observed by decreasing leukocytes, mainly granulocytes (neutofilis) and / or platelets and the involvement of the lymphoid organ, i.e., atrophy or apoptotic activity. Toxicity was also noted by lesions of the gastrointestinal tract such as increased numbers of mitotic and apoptotic figures and degenerative enterocolitis. Enzymes indicative of liver injury that were studied include: AST (aspartate aminotransferase) Location: cytoplasmic: liver, heart, skeletal muscle, kidney Liver: plasma ratio of 7000: 1 Tl / 2: 17 hours ALT (alanine aminotransferase) Location: cytoplasmic: liver, kidney, heart, skeletal muscle Liver: plasma ratio of 3000 -.1 Tl / 2: 42 hours; diurnal variation GGT (g-glutamyl transferase) Location-. plasma cell membrane with high secretory or absorbing capacity; liver, kidney, intestine Deficient Predictor of liver damage; commonly elevated in bile duct disorders The toxicity profiles of trastuzumab-MC-val-cit-MMAF, trastuzumab-MC (Me) -val-cit-PAB-MMAF, trastuzumab-MC-MMAF and trastuzu ab-MC-val -cit-PAB-MMAF were studied in female Sprague-Dawley rats (Example 19). The humanized trastuzumab antibody does not bind appreciably to rat tissue and any toxicity could be considered non-specific. Variants at dose levels of 840 and 2105 μg / m2 MMAF were compared with trastuzumab-MC-val-cit-PAB-MMAF at 2105 μg / m2. Animals in groups 1, 2, 3, 4, 6 and 7 (Vehicle, 9.94 &24.90 mg / kg trastuzumab-MC-val-cit-MMAF, 10.69 mg / kg trastuzumab-MC (Me) -val -cit-PAB-MMAF and 10.17 &25.50 mg / kg of trastuzumab-MC-MMAF respectively) gained weight during the study. The animals in groups 5 and 8 (26.78 mg / kg trastuzumab-MC (Me) -val-cit-PAB-MMAF and 21.85 mg / kg trastuzumab-MC-val-cit-PAB-MMAF, respectively) lost weight during the study. On Day 5 of the Study, the change in the body weights of the animals in groups 2, 6 and 7 were not significantly different from that of the animals in group 1. The change in body weights of the animals in groups 3, 4, 5 and 8 were statistically different from the animals of group 1 (Example 19). Rats treated with trastuzumab-MC-MMAF (groups 6 and 7) were indistinguishable from the control animals treated with the vehicle at both dose levels; i.e., this conjugate showed a higher safety profile in this model. Rats treated with trastuzumab-MC-val-cit-MMAF (without the self-immolative PAB residue, groups 2 and 3) showed typical dose-dependent changes for MMAF conjugates; the degree of the changes was lower compared to a full-length MC-val-cit-PAB-MMAF conjugate (group 8). Platelet counts on day 5 were approximately 30% of the baseline values in animals of group 3 (high dose trastuzumab-MC-val-cit-MMAF) compared to 15% in animals of group 8 (high dose of trastuzumab-MC-val-cit-PAB-MMAF). Elevation of the liver enzymes AST and ALT of bilirubin and the degree of thrombocytopenia was most evident in animals treated with trastuzumab-MC (Me) -val-cit-PAB-MMAF (groups 4 and 5) in a form dependent on the dose; Group 5 animals (high dose group) showed 5 ALT levels of approximately 10x the baseline value on day 5, and platelets were reduced by approximately 90% at the time of necorpsy. Female Sprangue Dawley rats were also dosed at high levels (Example 19, High Dose study: Groups 2, 3, 4) with trastuzumab-MC-MMAF and Vehicle control (Group 1). Signs of moderate toxicity were observed, including a dose-dependent elevation of liver enzymes ALT, AST and GGT. On day 5 the animals in the highest dose group show a 2-fold elevation of ALT and a 5-fold elevation of AST; GGT also rose (6U / 1). Enzyme levels show a trend toward normalization on day 12. There was moderate granulocytosis in all three dose groups on day 5, the amount of platelets remaining essentially unchanged in all animals. The morphological changes were moderate; the animals treated at the 4210 μg / m2 / Group 2 dose level showed no remarkable histology of the liver, spleen, thymus, intestines and spinal cord. Modified apoptotic and mitotic activity was observed in the thymus and liver respectively in animals treated at the dose level of 5500 μg / m2 (Group 3). The spinal cord was normocellular, but showed evidence of granulocytic hyperplasia, which is consistent with the absolute granulocytosis observed in the peripheral blood counts in these animals. The animals in the highest dose in group 4 showed qualitatively the same characteristics; the mitotic activity in the liver appears slightly increased compared to the animals in Group 3. Also, hematopoiesis was observed extramedullary in the spleen and liver. EphB2R is a Type 1 receptor tyrosine kinase - - TM with close homology between the mouse and the human and is over-expressed in colorectal cancer cells. 2H9 is an antibody against EphB2R. The pure antibody has no effect on tumor growth, but 2H9-val-cit-MMAE removes cells expressing EphB2R and showed efficacy in a mouse xenograft model using human colon tumors CXF1103 (Mao et al (2004) Cancer Res. 64: 781-788). 2H9 and 7C2 are both mouse anti-HER2 IgGl antibodies. The toxicity profiles of 2H9-MC-val-cit-PAB-MMAF (3.7 MMAF / Ab), 7C2 -MC-val-cit-PAB-MMAF (4 MMAF / Ab) and trastuzu ab-MC-val- were compared. cit-PAB-MMAF (5.9 MMAF / Ab). The differences in the structure of each immunoconjugate or the drug portion of the immunoconjugate can affect the pharmacokinetics and finally the safety profile. The humanized trastuzumab antibody does not bind appreciably to rat tissue and any toxicity would be considered non-specific. TOXICITY / SECURITY IN THE MONO CYNOMOLGUS Similar to the toxicity / safety study in rats, cynomolgus monkeys were treated with ADC followed by measurements of liver enzymes, and the inspection and analysis of the effect on various organs. General observations included changes in body weights and signs of injury and hemorrhage. Clinical pathological parameters (serum chemistry and hematology), histopathology, and necropsy in dosed animals were conducted (Example 19). The antibody drug conjugate, H-MC-vc-PAB-MMAE (H trastuzumab-linked via cysteine) showed no evidence of liver toxicity at any of the dose levels tested. Peripheral blood granulocytes showed decrease after a single dose of 1100 μg / m2 with full recovery at 14 days post-dose. The antibody drug conjugate H-MC-ve-PAB-MMAF showed elevation of liver enzymes at the dose level of 550 (transient) and 880 mg / m2, without evidence of granulocytopenia, and a platelet-dependent decrease of the dose, transient (groups 2 and 3). 4.6 SYNTHESIS OF THE COMPOUNDS OF THE INVENTION Exemplary Compounds and Exemplary Conjugates may be made using the synthetic procedures delineated below in Schemes 5-16. As described in more detail below, Exemplary Exemplary or Conjugated Compounds can conveniently be prepared using a Linker that has a reactive site for linking to the Drug and the Ligand. In one aspect, a Linker has a reactive site having an electrophilic group that is reactive to a nucleophilic group present in a ligand, such as but not limited to an antibody. Nucleophilic groups useful in an antibody include but are not limited to sulfhydryl, hydroxyl and amino groups. The heteroatom of the nucleophilic group of an antibody is reactive to an electrophilic group in a linker and forms a covalent bond to a linker unit. Useful electrophilic groups include, but are not limited to, maleimide and haloacetamide groups. The electrophilic group provides a convenient site for antibody binding. In another embodiment, a linker has a reactive site having a nucleophilic group that is reactive to an electrophilic group present in an antibody. Electrophilic groups useful in an antibody include, but are not limited to, carbonyl groups of aldehyde and ketone. The heteroatom of a nucleophilic group of a linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit. Useful nucleophilic groups in a linker include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and aryl hydrazide. The electrophilic group in an antibody provides a convenient site for binding to a linker. The carboxylic acid functional groups and the chloroformate functional groups are also useful reactive sites for a linker because they can react with secondary amino groups of a drug to form an amide bond. Also useful as a reactive site is a functional group of carbonate in a linker, such as but not limited to p-nitropenyl carbonate, which can react with an amino group of a drug, such as but not limited to N-methyl valine, to form a carbamate bond. Typically, peptide-based drugs can be prepared by forming a peptide bond between two or more amino acids and / or peptide fragments. Such peptide bonds can be prepared, for example, according to the liquid phase synthesis method (see E. Schroder and K. Lübke, "The Peptides", volume 1, pp. 76-136, 1965, Academic Press). This is well known in the field of peptide chemistry. The synthesis of an Illustrative Dilator having an electrophilic maleimide group is illustrated below in Schemes 8-9. Synthetic methods useful for the synthesis of a Linker are described in Scheme 10. Scheme 11 shows the construction of a Linker unit that has a val-cit group., an electrophilic maleimide group and a PAB group Self-immolative spacer. Scheme 12 represents the synthesis of a Linker having a phe-lys group, an electrophilic maleimide group, with and without the PAB group Auto-immolative spacer. Scheme 13 presents a general profile for the synthesis of a Drug-Linker Compound, while Scheme 14 presents an alternate route for preparing a Drug-Linker Compound. Scheme 15 represents the synthesis of a branched linker containing a BHMS group. Scheme 16 delineates the binding of an antibody to a Drug-Linker Compound to form a Drug-Linker-Antibody Conjugate, and Scheme 14 illustrates the synthesis of the Drug-Linker-Antibody Conjugates that they have, for example but without limited to, 2 or 4 drugs per Antibody. As described in more detail below, the exemplary Conjugates are conveniently prepared using a Linker that has two or more Reagent Sites to link to the Drug and a Ligand. In one aspect, a Linker has a reactive site having an electrophilic group that is reactive to a nucleophilic group present in a Ligand, such as an antibody. Nucleophilic groups useful in an antibody include but are not limited to, sulfhydryl, hydroxyl and amino groups. The heteroatom of the nueleophilic group of an antibody is reactive to an electrophilic group in a linker and forms a covalent bond to a linker unit. Useful electrophilic groups include, but are not limited to, maleimide and haloacetamide groups. The electrophilic group provides a convenient site for antibody binding. In another embodiment, a Linker has a Reagent site having a nucleophilic group that is reactive to an electrophilic group present in a Ligand, such as an antibody. Electrophilic groups useful in an antibody include, but are not limited to, carbonyl groups of aldehyde and ketone. The heteroatom of a nueleophilic group of a linker can react with an electrophilic group on an antibody and form a covalent bond to an antibody unit. Useful nucleophilic groups in a linker include, but are not limited to, hydrazide, oxime, amino, hydrazine, thiosemicarbazone, hydrazine carboxylate, and aryl hydrazide. The electrophilic group in an antibody provides a convenient site for binding to a linker. 4.6.1 SYNTHESIS OF DRUG RESIDUE Typically, peptide-based drugs can be prepared by forming a peptide bond between two or more amino acids and / or peptide fragments. Such peptide bonds can be prepared, for example, according to the liquid phase synthesis method (see E. Schroder Arad K. Lübke, "The Peptides", volume 1, pp. 76-136, 1965, Academic Press). well known in the field of peptide chemistry. The auristatin / dolastatin drug residues can be prepared according to the general methods of: U.S. Patent. No. 5635483; Patent of E.U. No. 5780588; Pettit et al. , (1989) J. Am. Chem. Soc. 111; 5463-5465; Pettit et al. (1998) Anti-Cancer Drug Design 13: 243-277; and Pettit et al. , (1996) J. Chem. Soc. Perkin Trans. 1 5: 859-863. In one embodiment, a Drug is prepared by combining approximately one stoichiometric equivalent of a dipeptide and tripeptide, preferably in a container reaction under suitable condensation conditions. This procedure is illustrated in Schemes 5-7, below. Scheme 5 illustrates the synthesis of an N-terminal tripeptide unit F which is a useful intermediate for the synthesis of the drug compounds of Formula Ib.
Scheme 5 D As illustrated in Scheme 5, a protected amino acid A (wherein PG represents a protecting amine group, R4 is selected from hydrogen, C? -C8 alkyl, C3-C8 carbocycle, O- (C? -C8 alkyl) , -aryl, alkyl-aryl, alkyl- (C3-Ca carbocycle), C3-C8 heterocycle, alkyl- (C3-C8 heterocycle) wherein R5 is selected from H and methyl; or R4 and R5 together, have the formula - (CRaRb) n- wherein Ra and Rb are independently selected from hydrogen, C al-C8 alkoyl and C3-C8 carbocycle and n is selected from 2, 3, 4, 5 and 6, and form a ring with the atom to which it is attached. unite) is coupled to t-butyl ester B (wherein R6 is selected from -H and alkyl-Cx-C8; and R7 is selected from hydrogen, C? -C8 alkyl, C3-C8 carbocycle, -0- (C? -C8), -aryl, alkyl-aryl, alkyl- (C3-C8 carbocycle, -), C3-C8 heterocycle and alkyl- (C3-C8 heterocycle) under suitable coupling conditions, eg, in the presence of PyBrop and diisopropylethylamine , or using DCC (see, for example, Miyazaki, K. e t. Chem. Pharm. Bull. 1995, 43 (10), 1706-1718). Suitable Protective Groups PG, and suitable synthetic methods to protect an amino group with a protecting group are well known in the art. I will see. g. , Greene, T.W. and Wuts, P.G.M., Protective Groups in Organic Synthesis, 2nd Edition, 1991, John Wiley & Sons. Exemplary protected amino acids A are PG-Ile and, particularly, PG-Val, while other suitable protected amino acids include, without limitation: PG-cyclohexylglycine, PG-cyclohexylalanine, PG-aminociclopropane-1-carboxylic acid, PG-aminoisobutyric acid, PG phenylalanine, PG-phenylglycine, and PG-tert-butylglycine. Z is an exemplary protection group. Fmoc is another exemplary protection group. An exemplary t-buty ester B is dolaisoleuin t-butyl ester. The dipeptide C can be purified, e.g., using chromatography, and subsequently deprotect it, e.g., using H2 and 10% Pd-C in ethanol when PG is benzyloxycarbonyl, or using diethylamine for removal of a Fmoc protection group. The resulting amine D readily forms a peptide bond with an amino acid BB (wherein R 1 is selected from -H, C 1 -C 8 alkyl, and C 3 -C 8 carbocycle; and R 2 is selected from -H and C 1 -C 8 alkyl; or R1 and R2 together, have the formula - (CRaRb) n- wherein Ra and Rb are independently selected from -H, -C8 alkyl and C3-C8 carbocycle and n is selected from 2, 3, 4, 5 and 6, and forms a ring with the nitrogen atom to which they are attached, and R3 is selected from hydrogen, C? -C8 alkyl, C3-C8 carbocycle, -O- (CX-C8 alkyl), -aryl, alkyl-aryl , alkyl- (C3-C8 carbocycle), C3-C8 heterocycle and alkyl- (C3-C8 hetercycle)). N, N-dialkyl amino acids are exemplary amino acids for BB, such as commercially available N, N-dimethyl valine. Other N, N-dialkyl amino acids can be prepared by reductive biskylation using known procedures (see, eg, Bowman, R.E, Stroud, H.H J *. Chem. Soc., 1950, 1442-1340). Fmoc-Me-L-Val and Fmoc-Me-L-glycine are two exemplary amino acids BB useful for the synthesis of N-monoalkyl derivatives. The amine D and the amino acid BB react to provide the tripeptide E using the coupling reagent DEPC with triethylamine as the base. The C-terminal protection group of E is subsequently deprotected using HCl to provide the tripeptide compound of Formula F. The illustrative DEPD coupling methodology and the PyBrop coupling methodology shown in Scheme 5 are outlined below in General Procedure A and General Procedure B, respectively. The illustrative methodology for the deprotection of a Z-protected amine through catalytic hydrogenation is outlined below in General Procedure C. General Procedure A: Synthesis of peptides using DEPC. The N-protected or?,? -disubstituted amino acid or peptide D (1.0 eq.) And an amine BB (11 eq.) Are diluted with an aprotic organic solvent, such as dichloromethane (0.1 to 0.5 M). An organic base such as triethylamine or diisopropylethylamine (1.5 eq.) Is then added, followed by DEPC (1.1 eq.). The resulting solution is stirred, preferably under argon, for up to 12 hours while monitored by HPLC or TLC. The solvent is removed in vacuo at room temperature, and the crude product is purified using, for example, HPLC or flash column chromatography (silica gel column). The relevant fractions were combined and concentrated in vacuo to yield tripeptide E which was dried under vacuum overnight. General Procedure B: Synthesis of Peptides using PyBrop. Amino acid B (1.0 eq.), Optionally having a protective carboxyl group, is diluted with an aprotic organic solvent such as dichloromethane or DME to provide a solution of a concentration between 0.5 and 1.0 mM, then diisopropylethylamine (1.5 eq. .). Fmoc-, or Z-protected amino acid A (1.1 eq.) Was added as a solid in one portion, then PyBrop (1.2 eq.) Was added to the resulting mixture. The reaction was monitored by TLC or H-PLC, followed by a preparation procedure similar to that described in General Procedure A. General Procedure C: Z removal through catalytic hydrogenation. The Z-protected amino acid or C-peptide was diluted with ethanol to provide a solution of a concentration between 0.5 and 1.0 mM in a suitable vessel, such as a thick-walled round bottom flask. 10% palladium on carbon (5-10% w / w) was added and the reaction mixture was placed under a hydrogen atmosphere. The progress of the reaction was monitored using HPLC and was completed generally within 1-2 h. The reaction mixture was filtered through a pre-washed celite pad and the celite was washed again with a polar organic solvent, such as methanol after filtration. The eluent solution was concentrated in vacuo to yield a residue which was diluted with an organic solvent, preferably toluene. The organic solvent was then removed in vacuo to yield the deprotected amine C. Scheme 6 shows a useful method for making a C-terminal Dipeptide of Formula K and a method for coupling the dipeptide of Formula K with the tripeptide of Formula F to make the drug compounds of Formula Ib.
The dipeptide K can be easily prepared by the condensation of the amino acid Boc-Dolaproine G (see, for example, Pettit GR, et al., Synthesis, 1996, 719-725), with an amine of Formula H using well known condensing agents for the chemistry of peptides, such as, for example, DEPC in the presence of triethylamine, as shown in scheme 5. The dipeptide of formula K can then be coupled with a tripeptide of formula F using General Procedure D to prepare the drug compounds protected by Fmoc of the formula L which can subsequently be deprotected using General Procedure E in order to provide the drug compounds of the formula (Ib). General procedure D: Drug synthesis. A mixture of dipeptide K (1.0 eq.) And tripeptide F (1 eq.) Is diluted with an aprotic organic solvent, such as dichloromethane, to form a 0.1 M solution, then a strong acid, such as trifluoroacetic acid, is added ( 1/2 v / v) and the resulting mixture is stirred under a nitrogen atmosphere for two hours at 0 ° C. The reaction can be monitored using TLC, or preferably, HPLC. The solvent is removed in vacuo and the resulting residue is azeotropically dried twice, preferably using toluene. The resulting residue is dried under high vacuum for 12 hours and then diluted with an aprotic organic solvent, such as dichloromethane. An organic base such as triethylamine or diisopropylethylamine is then added. (1.5 eq.) Depending on the chemical functionality in the waste. The reaction mixture is monitored either by TLC or HPLC and upon completion of the reaction is subjected to a processing procedure similar or identical to that described in General Procedure A. General procedure E: Removal of Fmoc using diethylamine. A drug Fmoc protected L is diluted with an aprotic organic solvent such as dichloromethane and diethylamine (1/2 v / v) is added to the resulting solution. The reaction progress is monitored by TLC or HPLC and is typically completed in 2 hours. The reaction mixture is concentrated in vacuo and the resulting residue is azeotropically dried, preferably using toluene, then dried under high vacuum to produce the drug Ib having an unprotected amino group. Scheme 7 shows a useful method for making MMAF derivatives of the formula (Ib).
Scheme 7 (Ib) when Z -O- and R11 is t-butyl (Ib) when Z -O- and R "is t-H The dipeptide 0 can be easily prepared by the condensation of the amino acid Boc-Dolaproine G (see, for example, Pettit GR, et al., Synthesis, 1996, 719-725), with a protected amino acid of the Formula M using highly condensing agents. known for peptide chemistry, such as, for example, DEPC in the presence of triethylamine, as shown in schemes 5 and 6. The dipeptide of formula O can then be coupled with a tripeptide of formula F using General Procedure D for making Fmoc-protected MMAF compounds of the formula P which can subsequently be deprotected using General Procedure E in order to provide the MMAF compounds of the formula (Ib).
Thus, the above methods are useful for making drugs that can be used in the present invention. 4.6.2 SYNTHESIS OF DRUG-LINKER To prepare the drug-linker compound of the present invention, the drug is reactivated with a reactive site in the linker. In general, the linker can have the structure: Reagent Site 2 - Aaa-W »ww YJ-vy '[Reagent Site 1 when both the separation unit (-Y-) and the stretching unit (-A-) are present. Alternatively, the linker can have the structure: Reagent Site 2 - a-Ww / Reagent Site 1 when the separation unit (-Y-) is absent. The linker can also have the structure: Reagent Site 2 Ww Reagent Site 1 when both the stretching unit (-A-) and the separation unit (-Y-) are absent. The linker can also have the structure: Reactive Site 2 | Aa Isitio Reactive ll when both the amino acid unit (W) and the separation unit (Y) are absent. In general, a suitable linker has an amino acid unit linked to an optional stretching unit and an optional separation unit. The reactive site 1 is present at the termination of the separation, and the reactive site 2 is present at the completion of the stretch. If a separation unit is not present, the reactive site 1 is present at the C terminus of the amino acid unit. In an exemplary embodiment of the invention, the reactive site No. 1 is reactive to a nitrogen atom of the drug, and the reactive site No. 2 is reactive to a sulfhydryl group in the ligand. The reactive sites 1 and 2 can be reactive to different functional groups.
In one aspect of the invention, the reactive site In another aspect of the invention, the reactive site or. 1 is In yet another aspect of the invention, the reactive site No. 1 is a p-nitrophenyl carbonate having the formula: In one aspect of the invention, reactive site No. 2 is a thiol accepting group. Suitable thiol acceptance groups include haloacetamide groups having the formula wherein X represents an omission group, preferably O-mesyl, O-tosyl, -Cl-, Br, or -I; or a maleimide group that has the formula Useful linkers can be obtained through commercial sources, such as Molecular Biosciences, Inc. (Boulder, CO), or prepared as summarized in Schemes 8-10 below.
Scheme 8 wherein X is -CH2- or -CH2OCH2-; and n is an integer that fluctuates from 0.10 when C is -CH2-; or from 1-10 when X is -CH2OCH2-. The method shown in Scheme 9 combines maleimide with glycol, under Mitsunobu conditions to make a polyethylene glycol maleimide extruder (see for example, Walker, MA, J. Org. Chem., 1995, 60, 5352-5), followed by the installation of a reactive p-nitrophenyl carbonate site group.
Scheme 9 wherein E is -CH2- or -CH2OCH2-; and e is an integer that fluctuates from 0-8; Alternatively, PEG-maleimide and PEG-haloacetamide stretchers can be prepared as described by Frisch et al., Bioconjugate Chem. 1996, 7, 180-186. Scheme 10 illustrates a general synthesis of an illustrative linker unit containing a maleimide stretch group and optionally a self-immolative p-aminobenzyl ether separator.
Scheme 10 wherein Q is C? -C8 alkyl, -O- (C? -C8 alkyl), -halogen, -nitro or -cyano; m is an integer that fluctuates from 0-4; and n is an integer that fluctuates from 0-10. Useful stretchers can be incorporated into a linker using the commercially available intermediates from Molecular Biosciences (Boulder, CO) described below, using known techniques of organic synthesis. The extenders of the formula (Illa) can be introduced into a linker by reactivating the following intermediates with the N-terminus of an amino acid unit as illustrated in Schemes 11 and 12: where n is an integer that fluctuates from 1-10 and T is -H or S03Na; where n is an integer that fluctuates from 0-3; ; Y The stretch units of the formula (Illb) can be introduced into a linker by reactivating the following intermediates with the N-terminus of an amino acid unit: where X is -Br or -I; Y The stretching units of formula (IV) can be introduced into a linker by reactivating the following intermediates with the N-terminus of an amino acid unit: The stretch units of the formula (Va) can be introduced into a linker by reactivating the following intermediates with the N-terminus of an amino acid unit: Other extruders can be synthesized according to known procedures. Aminooxy extruders of the formula shown below can be prepared by treating alkyl halides with N-Boc-hydroxylamine according to the procedures described in Jones, D. S., et al., Tetrahedron Letters, 2000, 41 (10), 1531-1533; and Gilon C, et al., Tetrahedron, 1967, 23 (11), 4441-4447. wherein -R 17 is selected from-Cquile-C?-C ?alkylene, -C 3 -C 8 -carbocycle-, -0- (C?-C 8 alkyl) -, -arylene-, -alkylene-arylene-C? -C ? or ~ / -arlene-alkylene-C? -C10-, -alkylene C? -C10- (C3-C8 carbocycle) -, - (C3-C8 carbocycle) -C? -C10 alkylene-, C3-C8 heterocycle? , -alkylene C? -C10- (C3-C8 heterocycle) -, - (C3-C8 heterocycle) -C? -C? alkylene? -, - (CH2CH20) r, - (CH2CH20) r-CH2-; and r is an integer that fluctuates from 1-10. The isothiocyanate stretchers of the formula shown below can be prepared from isothiocyanatocarboxylic acid chlorides as described in Angew. Chem., 1975, 87 (14): 517.
S = C = N-R17-C (O > where -R17 is as described herein. Scheme 11 shows a method for obtaining a di-peptide val-cit linker having a maleimide extruder and optionally a self-immolative p-a inobenzyl separator. Scheme 11 p-nitrophenyl-OCOO-p-nitrophenyl p-nitrophenyl-OCOO-p-nyl-phenyl DIEA (1.5 eq.), DMF DIEA (1.5 eq.), DMF wherein Q is C? -C8 alkyl, -O- (C? -C8 alkyl), -halogen, -nitro or -cyano; m is an integer that fluctuates from 0-4. Scheme 12 illustrates the synthesis of a phe-lys dipeptide linker unit (Mtr) having a maleimide stretcher unit and a self-immolative p-aminobenzyl separation unit. The starting material AD (lys (Mtr)) is commercially available from Bachem, Torrance, CA or can be prepared according to Dubowchik, et al., Tetrahedron Letters (1997) 38: 5257-60. Scheme 12 wherein Q is C? -C8 alkyl, -O- (C? -C8 alkyl), -halogen, nitro or -cyano; m is an integer that fluctuates from 0-4.
As shown in scheme 13, a linker can be reactivated with an amino group of a drug compound of Formula (Ib) to form a drug-linker compound containing an amide or carbamate group, linking the drug unit to the linker unit. When the reactive site No. 1 is a carboxylic acid group, as in the linkage AJ, the coupling reaction can be carried out using HATU or PyBrop and an appropriate amine base, resulting in a drug-linker compound AK, which contains an amide bond between the drug unit and the linker unit. When the reactive site No. 1 is a carbonate, as in the AL linker, the linker can be coupled to the drug using HOBt in a mixture of DMF / pyridine to provide a drug-linker compound AM, which contains a carbamate link between the drug unit and the linker unit. Alternatively, when the reactive site No. 1 is an obstruction group, such as in the AN linker, the linker can be coupled to an amine group of a drug through a substitution nucleophilic process to provide a drug-linker compound that has an amine bond (AO) between the drug unit and the linker unit. Illustrative methods useful for linking a drug to a ligand to form a drug-linker compound are illustrated in Scheme 13 and are underlined in General Procedures G-H. Scheme 13 HATU Drug + Linker-COOH * - Drug-NH-C (0) -linker (Ib) AJ AK - Linker Drug + Linker-X b- ^ Drug- N-Linker (Ib) AN AO General Procedure G: Amide formation using HATU. A drug (Ib) (1.0 eq.) And a protected N linker containing a carboxylic acid reactive site (1.0 eq.), Are diluted with a suitable organic solvent, such as dichloromethane, and the resulting solution is treated with HATU (1.5 eq.) And an organic base, preferably pyridine (1-5 eq.). The reaction mixture is allowed to stir under an inert atmosphere, preferably argon, for 6 hours, during which time the reaction mixture is monitored using HPLC. The reaction mixture is concentrated and the resulting residue is purified using HPLC to yield the amide of the formula AK. Method H: Formation of carbamate using HOBt. A mixture of an AL linker having a reactive site of p-nitrophenyl carbonate (1.1 eq.) And drug (Ib) (1.0 eq.) Is diluted with an appropriate organic solvent such as DMF, to provide a solution having a concentration of 50-100 mM, and the resulting solution is treated with HOBt (2.0 eq.) and placed under an inert atmosphere, preferably argon. The reaction mixture is allowed to stir for 15 minutes and then an organic base, such as pyridine (1/4 v / v) is added and the progress of the reaction monitored using HPLC. The linker is typically consumed in 16 hours. The reaction mixture is then concentrated in vacuo and the resulting residue is purified using, for example HPLC, to produce the carbamate AM. An alternative method for preparing drug-linker compounds is outlined in Scheme 14. Using the method of Scheme 14, the drug binds to a partial linker unit (ZA, for example), which does not have a stretch unit attached. This provides an intermediate AP, which has an amino acid unit that has an N terminus protected by Fmoc. The Fmoc group is then removed and the resulting amine intermediate AQ is then attached to a stretch unit through a catalyzed coupling reaction using PyBrop and DEPC. The construction of drug-linker compounds containing either a bromoacetamide AR extruder or a PEG maleimide extruder AS is illustrated in Scheme 14.
Scheme 14 z + D Diethylamine wherein Q is C-C8 alkyl, -0- (C-C8 alkyl), -halogen, -nitro or -cyano; m is an integer that fluctuates from 0-4. The useful methodology for the preparation of a linker unit containing a branched separator is shown in Scheme 15.
Scheme 15 1. 1 HCi, THF 2. Raney Ni, hydrazine MeOH-THF Scheme 15 illustrates the synthesis of a val-cit linker having a maleimide stretch unit and a bis (4-hydroxymethyl) styrene (BHMS) unit. The synthesis of the intermediary of BHMS / AW) has been improved by previous procedures in the literature (see International Publication No. WO 9813059 for Firestone et al., And Crozet, MP; Archaimbault G.; Vanelle, P.; Nougier R., Tetrahedron Lett. (1985) 26: 5133-5134) and utilizes commercially available starting materials, diethyl (4-nitrobenzyl) phosphonate (AT) and commercially available 2,2-dimethyl-1,3-dioxan-5-one (AU ). The linkers AY and BA can be prepared from the AW intermediate using the methodology described in Scheme 9. 4.6.3 DENDRITE LINKS The linker can be a dendritic linker for the covalent attachment of more than one drug residue through a residue functional branch ligand to a ligand, such as, but not limited to an antibody (Sun et al., (2002) Bioorganic &Medicinal Chemistry Letters 12: 2213-2215; Sun et al., (2003) Bioorganic & Medical Chemistry 11: 1761-1768). The dendritic linkers can increase the molar ratio of drug to antibody, i.e., the charge, which is related to the potency of the drug-linker-ligand conjugate. Thus, when an antibody made of cysteine contains only a reactive group of cysteine thiol, a multitude of drug residues can be linked through a dendritic linker. The following exemplary embodiments of dendritic linker reagents allow to conjugate up to nine nucleophilic drug residue reagents by reaction with the functional groups of chloroethyl nitrogen mustard: o O li CH2OCH2CH2CNHCH2CY3 4. 6.4 CONJUGATION OF DRUG RESIDUES TO ANTIBODIES Scheme 16 illustrates the useful methodology for making drug-linker-ligand conjugates having from about 2 to about 4 drugs per antibody. An antibody is treated with a reducing agent such as dithiothreitol (DTT) to reduce some or all of the disulfide cysteine residues to form highly nucleophilic cysteine thiol groups (-CH2SH). The partially reduced antibody then reacts with drug-linker compounds, or linker reagents, with electrophilic functional groups such as maleimide or α-halo carbonyl, according to the conjugation method on page 766 of Klussman et al., (2004). ), Bioconjugate Chemistry 15 (4): 765-773.
Scheme 16 Drug-Linker DTT Compound Antibody "Partially Reduced Antibody" Drug-Linker Conjugate- Ligand with Reduced Drug Load For example, an antibody, e.g., AC10, dissolved in 500 mM sodium borate and 500 mM sodium chloride at pH 8.0, is treated with an excess of 100 mM dithiothreitol (DTT). After incubation at 37 ° C for approximately minutes, the buffer is exchanged by elution on Sephadex G25 resin and eluted with PBS with 1 mM DTPA. The thiol / Ab value is verified by determining the concentration of reduced antibody from the absorbance at 280 nm of the solution and the concentration of thiol by reaction with DNTB (Aldrich, Milwaukee, Wl) and the determination of absorbance at 412 nm. The reduced antibody dissolved in PBS is frozen on ice. The drug linker, e.g., MC-val-cit-P7AB-MMAE in DMSO, dissolved in acetonitrile and water at a known concentration, is added to the reduced antibody frozen in PBS. After about one hour, an excess of maleimide is added to complete the reaction and cap all thiol group of unreacted antibody. The reaction mixture is concentrated by centrifugal ultrafiltration and the ADC, e.g., AC10-MC-vc-PAB-MMAE, is purified and desalted by elution through G25 resin in PBS, filtered through 0.2 um filters under sterile conditions and frozen for storage. A variety of drug conjugates (ADCs) were prepared, with a variety of linkers, and drug residues, MMAE and MMAF. The following table is an exemplary ADC group that was prepared following the protocol of the Example 27, and characterized by HPLC and drug loading analysis.
Target ADC amount ratio (antigen) isolated (mg) drug / Ab 0772P 16E12-Mc-vc-PAB-MMAE 1.75 4 0772P HDlO-MC-vc-PAB-MMAE 46.8 4.4 0772P llDlO-MC-vc-PAB-MMAF 54.5 3.8 Brevican Brevican-MC-MMAF 2 6 Brevican Brevican-MC-MMAF 2 6 Brevican-MC-vc-PAB-MMAF 1.4 6 CD21 CD21-MC-VC-PAB-MMAE 38.1 4.3 CD21 CD21-MC-vc-PAB-MMAF 43 4.1 CRYPTUM 11F4-MC-vc-PAB-MMAF 6 4.8 CRYPTUM 25G8-MC-VC-PAB-MMAF 7.4 4.7 E16 12G12-MC-VC-PAB-MMAE 2.3 4.6 E16 3B5-MC-VC-PAB-MMAE 2.9 4.6 E16 12B9-MC-VC-PAB-MMAE 1.4 3.8 E16 12B9 -MC-VC-PAB-MMAE 5.1 4 E16 12G12-MC-VC-PAB-MMAE 3 4.6 E16 3B5-MC-VC-PAB-MMAE 4.8 4.1 E16 3B5-MC-VC-PAB-MMAF 24.7 4.4 EphB2R 2H9-MC-VC-PAB-MMAE 29.9 7.1 EphB2R 2H9-MC-fk-PAB-MMAE 25 7.5 EphB2R 2H9-MC-VC-PAB-MMAE 175 4.1 EphB2R 2H9-MC-VC-PAB-MMAF 150 3.8 EphB2R 2H9 -MC-VC-PAB-MMAF 120 3.7 EphB2R 2H9-MC-VC-PAB-MMAE 10.7 4.4 IL-20Ra IL20Ra-fk-MMAE 26 6.7 IL-20Ra IL20Ra-vc-MMAE 27 7.3 EphB2 IL8-MC-VC-PAB -MMAE 251 3.7 MDP MDP-vc-MMAE 32 MPF 19C3 -vc-MMAE 1.44 6.5 MPF 7D9-vc-MMAE 4.3 3.8 MPF 19C -vc-MMAE 7.9 3 MPF 7D9-MC-vc-PAB-MMAF 5 4.3 Napi3b lOHl-vc-MMAE 4.5 4.6 Napi3b 4C9-VC-MMAE 3.0 5.4 Napi3b 10H1-vc-MMAE 4.5 4.8 Napi3b 10H1-VC-MMAF 6.5 4 NCA 3E6-MC-fk-PAB-MMAE 49.6 5.4 NCA 3E6-MC-VC-PAB-MMAE 56.2 6.4 PSCA PSCA-fk-MMAE 51.7 8.9 PSCA PSCA-vc-MMAE 61.1 8.6 Napi3b lOHl-MC-vc-PAB-MMAE 75 4.2 Napi3b lOHl-MC-vc-PAB-MMAF 95 4.4 Napi3b 10H1-MC-MMAF 92 4 EphB2R 2H9-MC-ve-PAB-MMAE 79 5 EphB2R 2H9-MC-MMAF 92 4.9 0772P 11D10 (chimera Fe) -MC-vc- 79 4.3 PAB-MMAE 0772P 11D10 (chimera Fe) -MC-vc- 70 4.5 PAB-MMAF 072P 11D10 (chimera Fe) -MC- 23 4.5 MMAF Brevican 6D2-MC-ve -PAB-MMAF 0.3 4.5 Brevican 6D2-MC-MMAF 0.36 4.5 EphB2R 2H9 (chimera Fe) -MC-vc- 1983 4.3 PAB-MMAE E16 12B9-MC-VC-PAB-MMAE 14.1 4.6 E16 12B9-MC-VC-PAB -MMAF 16.4 4.5 E16 12G12-MC-VC-PAB-MMAE 10.5 4.1 E16 12G12-MC-vc-PAB-MMAF 10.2 3.8 E16 3B5-MC-VC-PAB-MMAE 58.6 3.8 E16 3B5-MC-VC-PAB-MMAF 8 3.1 0772P 11D10 (chimera Fe) -MC-vc 340 3.9 -PAB-MMAE Steapl (Steapl-92) -MC-vc-PAB- 3.5 MMAE Steapl (Steapl-92) -MC-vc-PAB- 4.7 MMAF Steapl ( Steapl-120) -MC-vc-PAB-2 MMAE Steapl (Steapl-120) -MC-vc-PAB-2.3 MMAF E16 3B5-MC-VC-PAB-MMAF 52.2 4.5 4.7 COMPOSITIONS AND METHODS OF ADMINISTRATION In other modalities, A composition is described that includes an effective amount of an exemplary compound and / or exemplary conjugate and a pharmaceutically acceptable carrier or vehicle. For convenience, drug units and drug-linker compounds may be referred to as exemplary compounds, while drug-ligand conjugates and drug-linker-ligand conjugates may be referred to as exemplary conjugates. The compositions are suitable for veterinary or human administration. The present compositions may be in any form that allows the composition to be administered to a patient. For example, the composition can be in the form of a solid, liquid or gas (aerosol). Typical routes of administration include, without limitation, oral, topical, parenteral, sublingual, rectal, vaginal, ocular, intra-tumor, and intranasal. Parenteral administration includes subcutaneous, intravenous, intramuscular injections, intrasternal injection or infusion techniques. In one aspect, the compositions are administered parenterally. In yet another aspect, exemplary compounds and / or exemplary conjugates or compositions are administered intravenously. The pharmaceutical compositions can be formulated so as to allow an exemplary compound and / or exemplary conjugate to be bioavailable when administering the composition to a patient. The compositions can take the form of one or more dose units, where, for example, a tablet can be a single dose unit, and a container of an exemplary compound and / or an exemplary aerosol conjugate can contain a plurality of dose units. The materials used to prepare the pharmaceutical compositions can be non-toxic in the amounts used. It will be apparent to those of ordinary skill in the art that the optimal dose of the active ingredient (s) in the pharmaceutical composition, it will depend on a variety of factors. Relevant factors include, without limitation, the type of animal (e.g., human), the particular form of the exemplary compound or exemplary conjugate, the manner of administration and the composition employed. The pharmaceutically acceptable carrier or vehicle can be particulate, so that the compositions are, for example, in the form of a tablet or powder. The vehicle (s) can be liquid (s), the compositions being, for example, an oral syrup or liquid injectable. Additionally, the carrier (s) can be gaseous (s) or particulate (s) in order to provide a useful aerosol composition, e.g., in administration by inhalation. When proposed for oral administration, the composition is preferably in solid or liquid form, wherein the semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as solid or liquid.
As a solid composition for oral administration, the composition can be formulated as a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like. Such a solid composition typically contains one or more inert diluents. Additionally, one or more of the following may be present: linkers such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, corn starch and the like; lubricants such as magnesium stearate or Sterotex; sliders such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin, a flavoring agent such as peppermint, methyl salicylate or orange flavor, and a coloring agent. When the composition is in the form of a capsule, e.g., a gelatin capsule, it may contain, in addition to the above types, a liquid carrier such as polyethylene glycol, cyclodextrin or a fatty oil. The composition may be in the form of a liquid, e.g., an elixir, syrup, solution, emulsion or suspension. The liquid may be useful for oral administration or for injection delivery. When proposed for oral administration, a composition may comprise one or more of a sweetening agent, preservatives, dye / dye and a flavor improver. In a composition for administration by injection, one or more of a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and an isotonic agent may also be included. The liquid compositions, whether in solutions, suspensions or other similar form, may also include one or more of the following: sterile diluents such as water for injection, saline, preferably physiological saline, Ringer's solution, isotonic sodium chloride, oils fixed such as mono or synthetic diglycerides which can serve as a solvent or suspension medium, polyethylene glycols, glycerin, cyclodextrin, propylene glycol, or other solvents; antibacterial agents such as benzyl alcohol, or methyl paraben; antioxidants such as ascorbic acid, or sodium bisulfite: chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and tonicity adjusting agents such as sodium chloride or dextrose. A parenteral composition can be enclosed in an ampule, a disposable syringe or a multiple dose vial made of glass, plastic or other material. Physiological saline is an exemplary adjuvant. An injectable composition is preferably sterile. The amount of the exemplary compound and / or the exemplary conjugate, effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard techniques. Additionally, in vitro or in vivo assays may optionally be employed to assist in the identification of optimal dose ranges. The precise dose to be employed in the compositions will also depend on the route of administration, and on the severity of the disease or disorder, and should be decided according to the judgment of the physician and the patient's circumstances. The compositions comprise an effective amount of an exemplary compound and / or an exemplary conjugate in order to obtain a suitable dose. Typically, this amount is at least about 0.01% of an exemplary compound and / or of an exemplary conjugate by weight of the composition. When proposed for oral administration, this amount may vary to range from about 0.1% to about 80% by weight of the composition. In one aspect, the oral compositions may comprise from about 4% to about 50% of the exemplary compound and / or the exemplary conjugate by weight of the composition. Still in another aspect, the present compositions are prepared so that a parenteral dosage unit contains from about 0.01% to about 2% by weight of the exemplary compound and / or the exemplary conjugate by weight. For intravenous administration, the composition may comprise from about 0.01 to about 100 mg of an exemplary compound and / or the exemplary conjugate per kg of the animal's body weight. In one aspect, the composition may include from about 1 to about 100 mg of the exemplary compound and / or the exemplary conjugate per kg of the animal's body weight. In another aspect, the amount administered will be in the range of from about 0.1 to about 25 g / kg body weight, of the exemplary compound and / or the exemplary conjugate. Generally, the dose of the exemplary compound and / or exemplary conjugate administered to a patient is typically from about 0.01 mg / kg to about 2000 mg / kg of the animal's body weight. In one aspect, the dose administered to a patient is between about 0.01 mg / kg to about 10 mg / kg of the animal's body weight, in another aspect, the dose administered to a patient is between about 0.1 mg / kg to about 250 mg / kg of the body weight of the animal, even in another aspect, the dose administered to a patient is between approximately 0.1 mg / kg to approximately 20 mg / kg of the body weight of the animal, even in another aspect, the administered dose is it ranges from about 0.1 mg / kg to about 10 mg / kg of the animal's body weight, and in yet another aspect the administered dose is between about 1 mg / kg to about 10 mg / kg of the animal's body weight. Exemplary exemplary compounds and / or conjugates can be administered by any convenient route, for example by infusion or rapid injection, by absorption through the epithelial or mucocutaneous layers (eg, oral mucosa, rectal and intestinal mucosa, etc.) . The administration can be systemic or local. Various delivery systems are known, e.g., encapsulation in liposomes, microparticles, microcapsules, capsules, etc., and can be used to administer an exemplary compound and / or exemplary conjugate or composition. In certain embodiments, more than one exemplary compound and / or exemplary conjugate or composition is administered to the patient. In specific embodiments, it may be desirable to administer one or more exemplary compounds and / or exemplary conjugates or compositions locally to the area in need of treatment. This can be achieved, for example, and not by way of limitation, by local infusion during surgery; topical application, e.g., in conjunction with a wound dressing after surgery; by injection; by means of a catheter; by means of a suppository; or by means of an implant, the implant being a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. In one embodiment, administration can be by direct injection into the site (or anterior site) of a cancerous, tumor or neoplastic or pre-neoplastic tissue. In another embodiment, administration can be by direct injection into the site (or anterior site) of a manifestation of autoimmune disease. In certain embodiments, it may be desirable to introduce one or more exemplary compounds and / or exemplary conjugates or compositions into the central nervous system by any suitable route, including intraventricular and intrathecal injection. Intraventricular injection can be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration, e.g., can also be employed by the use of an inhaler or nebulizer, and the formulation with an aerosol agent, or by perfusion in a fluorocarbon or synthetic pulmonary surfactant. In yet another embodiment, exemplary exemplary compounds and / or conjugates, or compositions, may be delivered in a controlled release system, such as, but not limited to, a pump of various polymeric materials. In yet another embodiment, a controlled release system can be placed in proximity to the target of the exemplary compounds and / or exemplary conjugates or compositions, eg, the brain, thus requiring only a fraction of the systemic dose (see, eg, Goodson in Medical Applications of Controlled Relay, supra, vol 2, pp. 115-138 (1984)). Other controlled release systems discussed in the Langer review may be used (Science 249: 1527-1533 (1990) .The term "vehicle" refers to a diluent, adjuvant, excipient, with which the exemplary and / or conjugated exemplary compound Such pharmaceutical carriers can be liquids, such as water or oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. The vehicles can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea and the like, In addition, auxiliary agents, stabilizers, thickeners, lubricants and colorants can be used. a patient, the exemplary compound and / or exemplary conjugate or composition and the pharmaceutically acceptable carriers are sterile.Water is an exemplary vehicle when the exemplary compounds and / or conjugates are administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be used as liquid carriers, particularly for injectable solutions. Acceptable pharmaceutical carriers also include excipients such as starch, glucose, lactose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, skimmed dry milk, glycerol, propylene. glycol, water ethanol and the like. The present compositions, if desired, may also contain minor amounts of wetting agents or emulsifiers or buffering agents. The present compositions may take the form of solutions, suspensions, emulsions, tablets, pills, lozenges, capsules, capsules containing liquids, powders, sustained release formulations, suppositories, emulsions, aerosols, sprays, suspensions or any other form suitable for use. Other examples of suitable pharmaceutical vehicles are described in Remington's Pharmaceutical Sciences by E. W., Martin. In one embodiment, exemplary exemplary and / or conjugated compounds are formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to animals, particularly human beings. Typically, carriers or carriers for intravenous administration are sterile, aqueous isotonic buffer solutions. If necessary, the compositions may also include a solubilizing agent. Compositions for intravenous administration may optionally comprise a local anesthetic such as lignocaine to relieve pain at the site of injection. Generally the ingredients are supplied either separately or mixed together in a single dose form, for example, as a freeze-dried dry powder or water-free concentrate in a hermetically sealed container such as a vial or sachette indicating the amount of the active ingredient . When an exemplary compound and / or exemplary conjugate is to be administered by infusion, it may be dispensed, for example, with an infusion bottle containing water or saline of pharmaceutically sterile grade. When the exemplary exemplary compound and / or conjugate is administered by injection, sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration. Compositions for oral delivery can be in the form of tablets, dragees, aqueous suspensions. or oleaginous, granules, powders, emulsions, capsules, syrups, or elixirs, for example. The orally administered compositions may contain one or more agents optionally, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, wintergreen or cherry oil; coloring agents and preservatives to provide a pharmaceutically palatable preparation. In addition, when in the form of a tablet or pill, the compositions can be coated to retard disintegration and absorption in the gastrointestinal tract thereby providing sustained action over an extended period of time. The selectively permeable membranes surrounding an osmotically active conducting compound are also suitable for orally administered compounds. In these latter platforms, the fluid from the environment surrounding the capsule is imbibed by the conductive compound, which dissolves to displace the agent or agent composition through an opening. These delivery platforms can provide a supply profile of an essentially zero order opposite to the acute profiles of immediate release formulations. A time delay material such as glycerol monostearate or glycerol stearate may also be used. The compositions may be proposed for topical administration, in which case the carrier may be in the form of a solution, emulsion, ointment or gel base. If proposed for transdermal administration, the composition may be in the form of a transdermal patch or an iontophoresis device. Topical formulations may comprise a concentration of exemplary and / or exemplary conjugate of from about 0.05% to about 50% w / v (weight per unit volume of the composition), in another aspect, from 0.1% to 10% by weight /volume. The composition can be proposed for rectal administration, in the form, e.g., of a suppository that will melt in the rectum and release the exemplary and / or exemplary conjugate. The composition may include various materials that modify the physical form of a solid or liquid dosage unit. For example, the composition may include materials that form a coating protection around the active ingredients. The materials forming the coating protection are typically inert and can be selected from, for example, sugar, shellac and other enteric coating agents. Alternatively, the active ingredients can be enclosed in a gelatin capsule. The compositions may consist of gaseous dose units, e.g., they may be in the form of an aerosol. The term aerosol is used to denote a variety of systems that vary from those of colloidal nature to systems consisting of pressurized packaging. The supply may be a liquefied or compressed gas or by a suitable pump system that supplies the active ingredients. Either in solid, liquid or gaseous form, the present compositions may include a pharmacological agent used in the treatment of cancer, autoimmune disease or an infectious disease. Four . 8 THERAPEUTIC USES OF EXEMPLIFICATION CONJUGATES Exemplary Compounds and / or Exemplary Conjugates are useful for the treatment of cancer, an autoimmune disease or an infectious disease in a patient. Four . 8 .1 TREATMENT OF CANCER Exemplary conjugates and / or exemplary compounds are useful for inhibiting the multiplication of a tumor cell or cancer cell, causing apoptosis in a tumor or cancer cell, or for treating cancer in a patient. Exemplary Exemplary and / or Conjugated Compounds may be used in accordance with a variety of facilities for the treatment of animal cancers. The Drug-Linker-Ligand Conjugates can be used to deliver a Drug or Drug unit to a tumor cell or cancer cell. Without being rounded by theory, in one embodiment, the Ligand unit of a Exemplifying Conjugate binds to or is associated with an antigen associated with a cancer cell or a tumor cell and the Exemplary Conjugate may be taken within a tumor cell or cell carcinogenicity through endocytosis mediated by the receptor. The antigen can bind to a tumor cell or cancer cell or can be an extracellular matrix protein associated with the tumor cell or cancer cell. Once inside the cell, one or more specific peptide sequences within the Linker Unit are hydrolytically divided by one or more proteases associated with the tumor cell or cancer cell, resulting in the release of a drug or a Drug-Linker Compound. The released drug or Drug-Linker Compound is then free to migrate into the cell and induce cytotoxic or cytostatic activities. In an alternative embodiment, the Drug or Drug unit is divided from the Exemplifying Conjugate outside the tumor cell or cancer cell and the Drug or Drug-Binding Compound subsequently penetrates the cell. In one embodiment, the ligand unit binds to the tumor cell or cancer cell. In another embodiment, the ligand unit binds to a tumor cell or cancer cell antigen that is on the surface of the tumor cell or cancer cell. In another embodiment, the ligand unit binds a tumor cell antigen or cancer cell which is an extracellular matrix protein associated with the tumor cell or cancer cell. The specificity of the Ligand unit for a particular tumor cell or cancer cell may be important in determining those tumors or cancers that are treated more effectively. For example, Exemplary Conjugates having a Ligand BR96 Unit may be useful for treating antigen-positive carcinomas including those of the lung, breast, colon, ovaries and pancreas. Exemplary Conjugates having an Anti-CD30 Ligand or an anti-CD40 Unit may be useful for treating hematologic malignancies. Other particular types of cancers that may be treated with Exemplary Conjugates include, but are not limited to, those described in Table 3. TABLE 3 Solid tumors, including but not limited to: myxosarcoma myxosarcoma, lyosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, angiosarcoma, endotheliosarcoma, lymphaniosarcoma, lymphangioendotheliosarcoma, sinotornona mesothelioma Ewing's tumor 1iomiosareorna rhabdomyosarcoma colon cancer colorectal cancer kidney cancer pancreatic cancer bone cancer breast cancer ovarian cancer prostate cancer esophogeal cancer stomach cancer oral cancer nasal cancer throat cancer squamous carcinoma basal cell carcinoma adenocarcinoma carcinoma sudoríparo carcinoma sebaceous carcinoma papilar papillary adenocarcinomas cystadenocarcinoma medullary carcinoma bronchial carcinoma renal carcinoma hepatoma biliary carcinoma choriocarcinoma seminoma embryonal carcinoma Wilms tumor cervic cancer to uterine cancer testicular cancer small cell carcinoma of lung carcinoma of vej iga lung cancer epithelial carcinoma glioma glioblastoma multiforme astrocytoma medulloblastoma craniofaringioma ependymoma pinealoma hemangioblastoma acoustic neuroma oligodendroglioma meningioma skin cancer melanoma neuroblastoma retinoblastorna blood cancers, including but not limited to: ALL acute lymphoblastic leukemia acute lymphoblastic B cell leukemia acute lymphoblastic T cell leukemia acute myeloblastic leukemia "AML" acute promyelocytic leukemia "APL" acute monoblastic leukemia acute erythroleukaemic leukemia megakaryoblastic leukemia guda acute myelomonocytic leukemia acute non-lymphocytic leukemia acute undifferentiated leukemia acute myelocytic leukemia "CML" chronic lymphocytic leukemia "CLL" tricholeukemia multiple myeloma Acute and chronic leukemias: lymphoblastic myeloid lymphocytic myelocytic leukemias Lymphomas: Hodgkin's disease Non-Hodgkin's lymphoma Multiple myeloma Waldenstrom's macroglobulinemia heavy-chain disease Polycythemia vera Exemplary conjugates provide tumor or cancer-specific targeting of conjugation, thus reducing the overall toxicity of these compounds. The Linker Units stabilize the Exemplary Conjugates in the blood, they are still divided by the specific proteases of the tumor inside the cell, releasing a Drug. 4.8.2 MULTI-MODALITY THERAPY FOR CANCER Cancers, including but not limited to, a tumor, metastasis or other disease or disorder characterized by uncontrolled cell growth, can be treated or prevented by the administration of an Exemplifying Conjugate and / or an Exemplary Compound. In other embodiments, methods for treating or preventing cancer are provided, including administering to a patient in need thereof an effective amount of a Exemplary Conjugate and a chemotherapeutic agent. In one embodiment, the chemotherapeutic agent is one with which the cancer treatment has not been found to be refractory. In another embodiment, the chemotherapeutic agent is one with which the cancer treatment has been found to be refractory. Exemplary Conjugates can be administered to a patient who has undergone surgery as a treatment for cancer. In one modality, the additional method of treatment is radiation therapy. In a specific embodiment, the Exemplary Conjugate is administered concurrently with the chemotherapeutic agent or with radiation therapy. In another specific embodiment, the chemotherapeutic agent or radiation therapy is administered before or subsequent to the administration of Exemplary Conjugates, in one aspect at least one hour, five hours, 12 hours, one day, one week, one month, in aspects additional several months (eg, up to three months), before or after the administration of an Exemplary Conjugate. A chemotherapeutic agent can be administered through a series of sessions. Any or a combination of chemotherapeutic agents listed in Table 4 may be administered. With respect to radiation, any radiation therapy protocol may be used depending on the type of cancer to be treated. For example, but not by way of limitation, x-ray radiation can be administered; in particular, high energy megavoltage (radiation greater than 1 MeV energy) can be used for deep tumors and electron beam and orthovoltage x-ray radiation can be used for skin cancers. Radioisotopes that emit gamma rays, such as radioactive isotopes, can also be administered. radio, cobalt and other elements. Additionally, methods for the treatment of cancer with a Exemplary Compound and / or Exemplary Conjugate are provided as an alternative for Chemotherapy or radiation therapy wherein the chemotherapy or radiation therapy has been proven or can be proved too toxic, eg, the results in lateral effects not acceptable or not bearable, for the subject to be treated. The animal that is treated can, optionally, be treated with another cancer treatment such as surgery, radiation therapy or chemotherapy., depending on which treatment is found that is acceptable or bearable. Exemplary Exemplary and / or Conjugated Compounds may also be used in an in vitro or ex vivo form, such as for the treatment of certain cancers, including, but not limited to leukemias and lymphomas, such treatment involving autologous strain cell transplants. This may involve a multi-step process in which the animal's autologous hematopoietic strain cells are harvested and purged from all cancer cells, the remaining medullary cell population of the animal is then eradicated through the administration of a high dose of a Exemplary Compound and / or Exemplary Conjugate with or without accompanying radiation therapy at high doses and the cell graft of the strain is re-merged into the animal. Supportive care is then provided while core function is restored and the animal is recovered. 4.8.3 MULTI-DRUG THERAPY FOR CANCER Methods for treating cancer include administering to a patient in need thereof an effective amount of a Exemplary Conjugate and another therapeutic agent this is an anti-cancer agent. Suitable anticancer agents include, but are not limited to, methotrexate, taxol, L-asparaginase, mercaptopurine, thioguanine, hydroxyurea, cytarabine, cyclophosphamide, ifosfamide, nitrosoureas, cisplatin, carboplatin, mitomycin, dacarbazine, procarbicine, topotecan, nitrogen mustards, cytoxane, etoposide, 5-fluorouracil, BCNU, irinotecan, camptothecins, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, plicamycin, mitoxantrone, asparaginase, vinblastine, vincristine, vinorelbine, paclitaxel and docetaxel. In one aspect, the anticancer agent includes, but is not limited to, a drug listed in Table 4. TABLE 4 - - Thioguanine Hormone therapies: Antagonists receptors Anti-estrogen tamoxifen raloxifene megestrol LHRH agonists: goscrclin leuprolide acetate Anti-androgens flutamide bicalutamide Retoniodes / Deltoides Analogs of vitamin D3 EB 1089 CB 1093 KH 1060 Photopinamic therapies vertoporfin (BDP-MA) phthalocyanine photosensitizer Pc4 demethoxy-hypocrellin A (2BA-2-DMHA) Interferon- Interferon-cytokines? tumor necrosis factor Other Gemcitabine Velcade Revamid Talamid Isoprenilation inhibitors: Lovastatin Dopaminergic neurotoxins; l-methyl-4-phenylpyridinium ion Four . 8 4 TREATMENT OF AUTOIMMUNIC DISEASES Exemplary conjugates are useful for eliminating or inhibiting the reproduction of a cell that produces an autoimmune disease or for treating an autoimmune disease. Exemplary Conjugates may be used in accordance with a variety of facilities for the treatment of an autoimmune disease in a patient. The Drug-Linker-Ligand Conjugates can be used to deliver a Drug to a target cell. Without being rounded by the theory, in one embodiment, the Drug-Linker-Ligand Conjugate is associated with an antigen on the surface of a target cell and the Exemplary Conjugate is then taken into a target cell through the endocytosis mediated by the receiver. Once inside the cell, one or more specific peptide sequences within the Linker Unit are divided enzymatically or hydrolytically, resulting in the release of a Drug. The released drug is then free to migrate to the cytosol and induce cytotoxic or cytostatic activities. In an alternative embodiment, the Drug is divided from the Exemplifying Conjugate outside the target cell and the Drug subsequently penetrates the cell. In one embodiment, the Ligand unit binds to an autoimmune antigen. In one aspect, the antigen is on the surface of a cell involved in an autoimmune condition. In another embodiment, the Ligand unit binds to an autoimmune antigen that is on the surface of a cell. In one embodiment, the Ligand binds to activated lymphocytes that are associated with the autoimmune disease state. In a further embodiment, the Exemplary Conjugates eliminate or inhibit the multiplication of cells that produce an autoimmune antibody associated with a particular autoimmune disease. Particular types of autoimmune diseases that can be treated with Exemplary Conjugates include, but are not limited to, disorders related to Th2 lymphocyte (eg, atopic dermatitis, atopic asthma, rhinoconjunctivitis, allergic rhinitis, Omenn syndrome, systemic sclerosis, and graft versus host disease); disorders related to the Thl lymphocyte (eg, rheumatoid arthritis, multiple sclerosis, soriásis, Sjorgren's syndrome, Hashimoto's thyroiditis, Grave's disease, primary biliary cirrhosis, Wegener's granulomatosis and tuberculosis), - disorders related to the activated B lymphocyte (eg , systemic lupus erythematosus, Goodpasture syndrome, rheumatoid arthritis and type 1 diabetes); and those described in Table 5. TABLE 5 Active Chronic Hepatitis Addison's Disease Allergic Alveolitis Allergic Reaction Allergic Rhinitis Alport Syndrome Anaphylaxis Ankylosing Spondylitis Syndromes Anti-Phospholipids Arthritis Ascariasis Aspergillosis Atopic Allergy Atrophic Dermatitis Atropic Rhinitis Behcet's Disease Allergic Alveolitis of Poultry Farmers Bronchial asthma Caplan syndrome Cardiomyopathy Celiac disease Chagas disease Chronic gromerulonephritis Cogan syndrome Cryoglutinin Disease Congenital Rubella Infection CREST Syndrome Crohn's Disease Cryoglobulinemia Cushing's Syndrome Dermatomyositis Systemic Lupus Dressler's Syndrome Eaton-Lambert ECHO virus infection Encephalomyelitis Endocrine ophthalmopathy Epstein-Barr virus infection Echinodermal erythematosis Evan's syndrome Felty's syndrome Fibromyalgia Fuch's cyclitis Gastrointestinal atrophy Gastrointestinal allergy Giant-cell arthritis Glomerulonephritis Goodpasture's syndrome Graft-versus-host disease Graves disease de Guillain-Barre Hashimoto's Thyroiditis Henoch-Schonlein's Hemolytic Anemia Idiopathic Adrenal Idiopathic Pulmonary Fibrosis IgA Nephropathy Inflammatory Bowel Diseases Insulin-dependent Diabetes Mellitus Juvenile Arthritis Juvenile Diabetes Mellitus (Type 1) Lambert-Eaton Syndrome Laminitis Flat Lichen Hepatitis Lupoid Lupus Lymphopenia Meniere's Disease Mixed Connective Tissue Disease Multiple Sclerosis Myasthenia Grave Pernicious Anemia Polyglandular Syndromes Presenile Dementia Primary Agammaglobulinemia Primary Biliary Cirrhosis Psoriasis Sciatic Arthritis Raynauds Phenomenon Recurrent Absorption Reiter Syndrome Rheumatic Fever Rheumatoid Arthritis Syndrome of Sampter Schistosomiasis Schmidt's Syndrome Schloderma Sclerosis Syndrome Stiff-Man's Syndrome Sympathetic Ophthalmosis Systemic Lupus Erythematosis Takayasu's Arteritis Temporary Arteritis Thrombocytopenia Thyrotoxicosis Toxic Epidermal Necrolysis Insulin Resistance Type B Diabetes Mellitus Type 1 Ulcerative Colitis Uveitis Vitiligo Macroglobulinemia of Waldenstrom Wegener's Granulomatosis 4.8.5 MULTI-DRUG THERAPY OF AUTOIMMUNE DISEASES describe methods for treating an autoimmune disease including administering to a patient in need thereof an effective amount of an Exemplifying Conjugate and another therapeutic agent known for the treatment of an autoimmune disease. In one embodiment, the anti-autoimmune disease agent includes, but is not limited to, the agents listed in Table 6. Table 6 Cyclosporin Cyclosporin A Mycophenolate Mycophenolate Sirolimus Tacrolimus Enanercept Prednisone Azathioprine Methotrexate Cyclophosphamide Prednisone Aminocaproic acid Chloroquine Hydroxychloroquine Hydrocortisone Dexamethasone Chlorambucil DHEA danazol bromocriptine meloxicam infliximab 4. 8 6 TREATMENT OF INFECTIOUS DISEASES Exemplary Conjugates are useful for eliminating or inhibiting the multiplication of a cell that produces an infectious disease or for treating an infectious disease. Exemplary Conjugates can be used in accordance with a variety of facilities for the treatment of an infectious disease in a patient. Drug-Linker-Ligand Conjugates can be used to deliver a Drug to a target cell. In one embodiment, the Ligand unit binds to the infectious disease cell. In one embodiment, the conjugates eliminate or inhibit the multiplication of cells to produce a particular infectious disease. Particular types of infectious diseases that can be treated with the Exemplary Conjugates include, but are not limited to, those described in Table 7. TABLE 7 Bacterial Diseases: Diphtheria Whooping Cure Hidden Bacteremia Urinary Tract Infection - - Gastroenteritis Cellulitis Epiglottitis Tracheitis Adenoid Hypertrophy Retrofaringeal abscess Impetigo Ectima Pneumonia Endocarditis Septic Arthritis Pneumocrosis Peritonitis Bacteriaemia Meningitis Acute Purulent Meningitis Urethritis Cervicitis Rectitis Pharyngitis Salpingitis Epididymitis Gonorrhea Syphilis Listeriosis Carbuncle Nocardiosis Salmonella Typhoid Fever Dysentery Conjunctivitis Sinusitis Brucellosis Tullaremia Cholera Bubonic Plague Tetanus Necrotizing Enteritis Cactinomycosis Mixed Anaerobic Infections Syphilis Borreliosis Leptospirosis Lyme Borreliosis Fever by Rat Bite Tuberculosis Lymphodenitis Leprosy Chlamydial Chlamydial pneumonia - - Trachoma Conjunctivitis of Inclusion Systemic Fungal Diseases: Histoplamósis Coccidiodomicosis Blastomycosis Esporotricósis Cryptococcosis Systemic Candidosis Aspergillosis Mucormycosis Micetorna Chromomycosis Diseases Rickettsiosis: Typhus Exentectic Fever Erythematosis Rickettsiosis Transmitted by Garrapata Rickettsiosis Exantemática Fever Q Bartonelosis Parasitic diseases: Malaria Babesiosis African trypanosomosis Chagas disease Leishmaniosis Visceral leishmaniosis Toxoplasmosis Meningoencephalitis Keratitis Amebiasis Giardiasis Cryptosporidiasis Isosporiasis Cyclosporiasis Microsporidiosis Ascariasis Infection of Trichocephalus Hookworm Infection Infection of Nematode Toxocariosis Ocular Trichinosis Dracunculus Disease Lymphatic filariasis Loasis Onchocercosis - - Infection Dirofilaria immitis Schistosomiasis Cucariasis Cutaneous Paragonimus westermani Trematode Hepatic Fasciolosis Fasciolopsosis Opisthorchiasis Taenia infections Hydatidosis Alveolar hydatidosis Viral Diseases: Measles Sclerosus Subcutaneous Panencephalitis Common Cold Mumps Rubella Roseola Erythema Infectious Chickenpox Varicella Respiratory Syncytial Virus Diphtheria Bronchiolitis Infectious Mononucleosis Poliomelitis Herpangina Exanthema Viruses of the hands and feet and mouth Bornholm's disease Genital herpes Condyloma acuminatum Aseptic meningitis Myocarditis Pericarditis Gastroenteritis Acquired Immunodeficiency Syndrome (AIDS) Human Immunodeficiency Virus (HIV) Reye Syndrome Kawasaki Syndrome Gripa Bronchitis Viral Pneumonia "Andante" Acute Respiratory Disease Acute Faringoconjunctival Fever Keratoconjunctivitis Epidemic Herpes Simplex Virus (HSV-1) Herpes Simplex Virus 2 (HSV-2) Zoster Cytomegalovirus Rabies Progressive Multifocal Leukoencephalopathy Curu Insomnia Fatal Family Crazy Cow Disease Spongiform Encephalopathy Tropical Spasmodic Paraparesis Equine Encephalitis California Encephalitis Streptococcus. Louis Fever Yellow Dengue Lymphocytic Choriomeningitis Lassa Fever Hemorrhagic Fever Hantvirus Pulmonary Syndrome Marburg Virus Infections Ebola Virus Infections Smallpox 4.8.7 MULTI-DRUG THERAPY OF INFECTIOUS DISEASES Methods for treating an infectious disease are described, including administration to a patient in need thereof an Exemplifying Conjugate and another therapeutic agent which is an anti-disease disease agent. In one embodiment, the anti-infectious disease agent is, but is not limited to, the agents listed in Table 8.
TABLE 8 Antibiotics ß-Lactam: Penicillin G Penicillin V Cloxacillin Dicioxacillin Methicillin Nafcillin Oxacillin Ampicillin Amoxicillin Bacampicillin Azlocillin Carbenicillin Mezlocillin Piperacillin Ticarcillin Aminoglycosides: Amicacin Gentamicin Kanamycin Neomycin Netilmicin Streptomycin Tobramycin Macrolides: Azithromycin Clarithromycin Erythromycin Lincomycin Clindamycin Tetracyclines: Demeclocycline Doxycycline Minocycline Oxytetracycline Tetracycline Quinolones: Cinoxacin Nalidixic Acid Fluoroquinolones: - Ciprofloxacin Enoxacin Grepafloxacin Levofloxacin Lomefloxacin orfloxacin Ofioxacin Esparfloxacin Trovafloxiciña Polypeptides: Bacitracin Colistin Polymyxin B Sulfonamides: Sulfisoxazole Sulfamethoxazole Sulfadiazine Sulfametizole Sulfacetamide Various Antibacterial Agents Trimethoprim Sulfamethazole Cloramfenicol Vaneomicin Metronidazole Quinupristin Dalfopristin Rifampin Spectinomycin Nitrofurantoin Antiviral Agents General Antiviral Agents: Idoxuradine Vidarabine Trifluridine Acyclovir Famciciclovir Penciciclovir Valaciclovir Ganciciclovir Foscarnet Ribavitin Amantadine Rimantadine Cidofovir Oligonucleotides Antisense Immunoglobulins Inteferons Drugs for HIV infection: Tenofovir Emtricitabine Zidovudine Didanosine Zalcitabine Stavudine Lamivudine Nevirapine Delavirdine Saquinavir Ritonavir Indinavir Nelfinavir EXAMPLES Example 1 - Preparation of Compound AB AB Fmoc-val-cit-PAB-OH (14.61 g, 24.3 mmol 1.0 eq., Patent No.6214345 from Firestone et al.) Was diluted with DMF (120 ml, 0.2 M) and a diethylamine (60 mg) was added to this solution. ml). the reaction was monitored by HPLC and found to be complete in 2 h. The reaction mixture was concentrated and the resulting residue was precipitated using ethyl acetate (ca. 100 ml) under sonication for 10 min. Ether (200 ml) was added and the precipitate was sonically added for 5 min. The solution was allowed to settle for 30 min. without stirring and then filtered and dried under high vacuum to provide Val-cit-PAB-OH, which was used in the next step, without further purification.
Yield: 8.84 g (96%). Val-cit-PAB-OH (8.0 g, 21 mmol) was diluted with DMF (110 mL) and the resulting solution was treated with MC-OSU (Willner et al., (1993) Bioconjugate Chem. 4:52 1; 6.5 g, 21 mmol, 1.0 eq.). The reaction was completed according to HPLC after 2 h. The reaction mixture was concentrated and the resulting oil was precipitated using ethyl acetate (50 ml). After sonication for 15 min, ether (400 ml) was added and the mixture was sonically added until all the large particles broke. The solution was then filtered and the solid dried to provide an off-white solid intermediate. Rendimeinto: 11.63 g (96%); ES-MS m / z 757.9 [MH] Fmoc-val-cit-PAB-OH (14.61 g, 24.3 mmol, 1.0 eq., U.S. Patent No. 6214345 of Firestone et al.,) Was diluted with DMF (120 ml , 0.2 M) and a diethylamine (60 ml) was added to this solution. The reaction was monitored by HPLC and found complete in 2 h. The reaction mixture was concentrated and the resulting residue was precipitated using ethyl acetate (ca. 100 ml) under sonication for 10 min. Ether (200 ml) was added and the precipitate was further sonicated for 5 min. The solution was allowed to settle for 30 min. without stirring and then filtered and dried under high vacuum to provide Val-cit-PAB-OH, which was used in the next step without further purification. Rendimeinto: 8.84 g (96%). Val-cit-PAB-OH (8.0 g, 21 mmol) was diluted with DMF (110 mL) and the resulting solution treated with MC-OSu (Willner et al., (1993) Bioconjugate Chem. 4: 521; 6.5 g , 21 mmol, 1.0 eq.). The reaction was completed according to HPLC after 2 h. The reaction mixture was concentrated and the resulting oil was precipitated using ethyl acetate (50 ml). After sonication for 15 min. , ether (400 ml) was added and the mixture was sonically added until all the large particles broke. The solution was then filtered and the solid dried to provide an off-white solid intermediate. Rendimination: 11.63 g (96%); ES-MS m / z 757.9 [M-H]. The off-white solid intermediate (8.0 g, 14.0 mmol) was diluted with DMT (1 20 ml, 0.12 M) and to the resulting solution was added bis (4-nitrophenyl) carbonate (8.5 g, 28.0 mmol, 2.0 eq.) And DIEA (3.66 ml, 21.0 mmol, 1.5 eq.). The reaction was completed in 1 h according to IHPLC. The reaction mixture was concentrated to give an oil which was precipitated with EtOAc, and triturated with EtOAc (ca 25 ml). The solute was further precipitated with ether (ca 200 ml) and triturated for 15 min. The solid was filtered and dried under high vacuum to provide Compound AB which was 93% pure according to HPLC and was used in the next step without further purification. Rendimeinto: 9.7 g (94%).
Example 2 - Preparation of Compound 1 Phenylalanine salt t-butyl ester HCl (868 mg, 3 mmol), N-Boc-Dolaproin (668 mg, 1 eq.), DEPC (820 μL, 1.5 eq.), And DIEA (1.2 ml) were diluted with dichloromethane (3 ml). After 2 hours (h) at room temperature (approximately 28 degrees Celsius), the reaction mixture was diluted with dichloromethane (20 ml), washed successively with saturated aqueous aHC03 (aq.) (2 x 10 ml),? saturated aqueous aCl (2 x 10 ml). The organic layer was separated and concentrated. The resulting residue was resuspended in ethyl acetate and purified by flash chromatography on ethyl acetate. The relevant fractions were charged and concentrated to provide the dipeptide as a white solid: 684 mg (46%). ES-MS m / z 491.3 [M + H] A For Boc selective cleavage in the presence of t-butyl ester, the above dipeptide (500 mg, 1.28 mmolj was diluted with dioxane (2 ml), 4M HCl / dioxane was added. (960 μl, 3 eq.), And the reaction mixture was stirred overnight at room temperature.The almost complete deprotection was observed by RP-HPLC with minimal amount of cleavage of t-butyl ester.The mixture was cooled in a ice bath, and triethylamine (500 μl) was added.After 10 min, the mixture was removed from the cooling bath, diluted with dichloromethane (20 ml), washed successively with saturated aq NaHCO 3 (2 x 10 ml). ), Saturated NaCl aq. (2 x 10 ml) The organic layer was concentrated to give a yellow foam: 287 mg (57%) The intermediate was used without further purification The tripeptide Fmoc-Meval-val-dil-Ot -Bu (prepared as described in WO 02/088172, entitled "Pentapeptide Compounds and Uses Related Thereto"; 0.73 mmol) was treated with TFA (3 ml), dichloromethane (3 ml) for 2 h at room temperature. The mixture was concentrated to dryness, the residue was co-evaporated with toluene (3 x 20 ml), and dried in vacuo overnight. The residue was diluted with dichloromethane (5 ml) and added to the deprotected dipeptide (287 mg, 0.73 mmol), followed by DIEA (550 μl, 4 eq.), DEPC (201 μl, 1.1 eq.). After 2 h at ambient temperature the reaction mixture was diluted with ethyl acetate (50 ml), washed successively with 10% aq. of citric acid (2 x 20 ml), saturated NaHCO3 aq. (2 x 10 ml), saturated NaCl aq (10 ml). The organic layer was separated and concentrated. The resulting residue was re-suded in ethyl acetate and purified by flash chromatography on ethyl acetate. The relevant fractions were charged and concentrated to provide Fmoc-Meval-vai-dil-dap-phe-O-t-Bu as a white solid: 533 mg (71%). Rf 0.4 (EtOAc). ES-MS m / z 1010.6 [M + H] +. The product (200 mg, 0.2 mmol) was diluted with dichloromethane (3 ml), diethylamine (1 ml). The reaction mixture was stirred overnight at room temperature. The solvents were removed to provide an oil which was purified by flash chromatography on silica gel in the gradient from step 0-10% MEOH in dichloromethane to provide Compound 1 as a white solid: 137 mg (87%). Rf 0.3 (10% MeOH / CH 2 Cl 2) - ES-MS m / z 788.6 [M + H] A Example 3 - Preparation of compound 2 Compound 2 was prepared from compound 1 (30 mg, 0.038 mmol) by treatment with 4M HCl / dioxane (4 ml) for 7 h at room temperature. The solvent was removed, and the residue was dried in vacuo overnight to give compound 2 as a white hydroscopic solid: 35 mg (120% calculated by HCl salt). ES-MS m / z 732.56 [M + H] Example 4 - Preparation of compound 3 Fmoc-Meval-val-dil-dap-phe-O-t-Bu (Example 2, 50 mg) was treated with 4M HCl / dioxane (4 ml) for 16 h at room temperature. The solvent was removed, and the residue was dried in vacuo overnight to give 50 mg of a white solid hydroscopic intermediate. The white solid intermediate (20 mg, 0.02 mmol) was diluted with dichloromethane was added (1 ml); DEPC (5 μl, 0.03 mmol, 1.5 eq.) Followed by DIEA (11 μl, 0.06 mmol, 3 eq.), And t-butylamine (3.2 μl, 0.03 mmol, 1.5 eq.). After 2 h at room temperature, the reaction was found to be incomplete by RP-HPLC. More DEPC (10 μl) and t-butylamine (5 μl) were added and the reaction was stirred for 4 h. additional The reaction mixture was diluted with dichlorornethane (15 ml), washed successively with water (5 ml), 0.1 M HCl aq. (10 ml), saturated NaCl aq. (10 ml). The organic layer was separated and concentrated. The resulting residue was diluted with dichloromethane and purified by flash chromatography on a gradient of the 0-5% MeOH step in dichloromethane. The relevant fractions were collected and concentrated to provide the protected intermediary Fmoc as a white solid: 7.3 mg (36%). Rf 0.75 (10% MeOH / CH2Cl2). The protected intermediate Fmoc was diluted with dichloromethane (0.5 ml) and treated with diethylamine (0.5 ml) for 3 h at room temperature. The reaction mixture was concentrated to dryness. The product was isolated by flash chromatography on silica gel in a gradient from step 0-10% MEOH in dichloromethane to provide compound 3 as a white solid: 4 mg (70%). Rf 0.2 (10% MeOH / CH2Cl2). ES-MS m / z 787 [M + H] A 809 [M + Na] +. Example 5 - Preparation of compound 4 Boc-L-Phenylalanine (265 mg, 1 mmol, 1 eq.) And triethylene glycol monomethyl ether (164 μl, 1 mmol, 1 eq.) Were diluted with dichloromethane (5 ml). Then, DCC (412 mg, 2 mmol, 2 eq.) Was added, followed by DMAP (10 mg). The reaction mixture was stirred overnight at room temperature. The prescipitate leaked. The solvent was removed in vacuo, the residue was diluted with ethyl acetate, and purified by flash chromatography on silica gel in ethyl acetate. The fractions containing product were extracted, concentrated and dried in vacuo to give a white solid: 377 mg (91%). Rf 0.5 (EtOAc). ES-MS m / z 434 [M + Na] +. Removal of the Boc Protection Group was carried out by treating the above material in dioxane (10 ml) with 4M HCl / dioxane (6 ml) for 6 h at room temperature. The solvent was removed in vacuo, the residue was dried in vacuo to give a white solid. The HCl Salt of Phenylalanine-triethylene glycol monomethyl ether ester (236 mg, 0.458 mmol, leq.) And N-Boc-Dolaproin (158 mg, 0.55 mmol, 1.2 eq.) Were diluted with dichloromethane (3 mL). DEPC (125 μl, 1.5 eq.) And added to the mixture followed by DIEA (250 μl, 3 eq.). After 2 h at room temperature the reaction mixture was diluted with ethyl acetate (30 ml), washed successively with saturated aq NaHCO 3. (2 1 0 ml), 10% citric acid aq. (2 x 10 ml), saturated aqueous NaCl (10 ml). The organic layer was separated and concentrated. The resulting residue was re-suspended in ethyl acetate and purified by flash chromatography on silica gel in ethyl acetate. The relevant fractions were charged and concentrated to provide a white foam intermediate: 131 mg (50%). Rf 0.25 (EtOAc). ES-MS m / z 581.3 [M + H] Boc deprotection was done in dichloromethane (2 ml), TFA (0.5 ml) at room temperature for 2 h. The solvent was removed in vacuo, and the residue was evaporated with toluene (3 x 25 ml), then dried in vacuum to give 138 mg of dipeptide TFA salt. Fmoc-Meval-val-dil-OH (Example 2,147 mg, 0.23 mmol, 1 eq.), And dipeptide TFA salt (138 mg) were diluted with dichloromethane (2 ml). To the mixture was added DEPC (63 μl, 1.5 eq.), Followed by DIEA (160 μl, 4 eq.). After 2 h at room temperature the reaction mixture was diluted with dichloromethane (30 ml), washed successively with 10% citric acid aq. (2 x 20 ml), saturated aqueous NaCl (20 ml). The organic layer was separated and concentrated. The resulting residue was re-suspended in dichloromethane and purified by flash chromatography on silica gel in a gradient from step 0-5% MEOH in dichloromethane. The relevant fractions were coalesced and concentrated to provide white foam: 205 mg (81%). Rf 0.4 (10% MeOH / CH2Cl2). ES-MS m / z 1100.6 [M + H] A 1122.4 [M + Na] A The Fmoc protection group was removed by treatment with diethylamine (2 ml) in dichloromethane (6 ml). After 6 h at room temperature the solvent was removed in vacuo, the product was isolated by flash chromatography on silica gel in a gradient from the 0-10% MEOH in dichloromethane stage. The relevant fractions were collected and concentrated. After evaporation from dichloromethane / hexane, 1: 1, compound 4 was obtained as a white foam: 133 mg (80%). Rf 0. 15 (10% MeOH / CH2Cl2) ES-MS m / z 878. 6 [M + H] Example 6 - Preparation of compound 5 Fmoc-Meval-val-dil-OH (Example 2.0.50 g, 0.78 mmol) and dap-phe-OMe • HCL (0.3 g, 0.78 mmol, prepared according to Pettit, GR, et al., Anti-Cancer Drug Design 1998, 13, 243-277) were dissolved in CH2C12 (10 ml) followed by the addition of diisopropylethylamine (0.30 ml, 1.71 mmol, 2.2 eq.). DEPC (0.20 ml, 1.17, 1.5 eq.) Was added and the contents were maintained on Ar. The reaction was completed in accordance with HPLC in 1 h. The mixture was concentrated to an oil and purified by Si02 chromatography (300 x 25 mm column) and eluted with 100% EtOAc. The product was isolated as a white foam solid. Rendimeinto: 0.65 g (87%). ES-MS m / z 968.35 [M + H] A 991.34 [M + Na] +; UV? 215, 265 nm. The Fmoc-protected peptide (0.14 g, 0.14 mmol) in methylene chloride (5 ml) was treated with diethylamine (2 ml) and the contents were kept at room temperature for 2 h. The reaction was completed by HPLC, concentrated to an oil, absorbed in 2 ml of DMSO and injected into a preparative HPLC (column of C12-RP, 5 μ, 100 A, linear gradient of MeCN in water (containing 0.1 % TFA) from 10 aa to 100% in 40 min followed by 20 min to 100%, at a flow rate of 25 ml / min). The fractions containing the product were evaporated to yield a white powder for the trifluoroacetate salt. Rendimeinto: 0.126 g (98%). Rf 0.28 (100% EtOAc); ES-MS m / z 746.59 [M + H] A 768.51 [M + Na] +; UV? ^ Ax 215 nm. Example 7 - Preparation of compound 6 The trifluoroacetate salt of Compound 5 (0.11 g, 0.13 mmol), Compound AB (0.103 g, 0.14 mmol, 1.1 eq.) And HOBt (3.4 mg, 26 μmol, 0.2 eq.) Were suspended in DMF / piyridine (2). ml / 0.5 ml, respectively). Diisopropylethylamine (22.5.μl, 0.13 mmol, 1.0 eq.) Was added and the yellow solution was stirred while under argon. After 3 h, 1.0 eq. of additional DIEA. 24 Hours later, 0.5 eq. of the activated linker was included in the reaction mixture. After a total of 40 h, the reaction was complete. The contents were evaporated, absorbed in DMSO and injected into a prep-HPLC column (C? 2-RP column, 5 μ, 100 Á, linear gradient of MeCN in water (containing 0.1% TFA) from 10 to 100% in 40 min followed by 20 min at 100%, at a flow rate of 50 ml / min). The desired fractions were evaporated to give the product as a yellow oil. Methylene chloride (ca 2 ml) and large amount of ether were added to provide compound 6 as a white precipitate which was filtered and dried. Rendimeinto: 90 mg (52%). ES-MS m / z 1344.32 [M + H] A 1366.29 [M + Na] +; UV? 215,248 nm. Example 8 - Preparation of compound 7 Compound 4 (133 mg, 0.15 mmol, 1 eq.), Compound AB, (123 mg, 0.167 mmol, 1.1 eq.), And HOBt (4 mg, 0.2 eq.) Were diluted with DMF (1.5 ml). After 2 min, pyridine (5 ml) was added and the reaction was monitored using RP-HPLC. The reaction showed to be complete within 18 h.
The reaction mixture was diluted with dichloromethane (20 ml), washed successively with citric acid aq. 10% (2 x 10 ml), water (10 ml), saturated aqueous NaCl (10 ml). The organic layer was separated and concentrated. The resulting residue was re-suspended in dichloromethane and purified by flash chromatography on silica gel in a gradient from the 0-10% MEOH step in dichloromethane. The relevant fractions were charged and concentrated to provide compound 7 as a white foam: 46 mg (21%). Rf 0.15 (10% MeOH / CH2Cl2). ES-MS m / z 1476.94 [M + H] A Example 9 - Preparation of MC-Val-Cit-PAB-MMAF t-butyl ester 8 Compound 1 (83 mg, 0.11 mmol), Compound AB (85 mg, 0.12 mmol, 1.1 eq.), And HOBT (2.8 mg, 21 mmol, 0.2 eq.) Were absorbed in dry DMT (1.5 ml) and pyridine (0.3 ml) while under argon. After 30 h, the reaction was found to be essentially complete by HPLC. The mixture was evaporated, absorbed in a minimal amount of DMSO and purified by prep-HPLC (column of C12-RP, 5 μ, 100 Á, linear gradient of MeCN in water (containing 0.1% TFA) from 10 to 100% at 4Omin followed by 2O min at 100%, at a flow rate of 25 ml / min) to provide compound 8 as a white solid. Rendimement: 103 mg (71%). ES-MS m / z 1387.06 [M + H] A 1409.04 [M + Na] +; UV ax 205.248 nm. Example 10 - Preparation of MC-val-cit-PAB-MMAF 9 Compound 8 (45 mg, 32 μmol) was suspended in methylene chloride (6 ml) followed by the addition of TFA (3 ml). The resulting solution was maintained for 2 h. The reaction mixture was concentrated in vacuo and purified by prep-HPLC (C12-RP column, 5 μ, 100 A, linear gradient of MeCN in water (containing 0.1% TFA) from 10 to 100% in 40 min followed by 20 to 100%, at a flow rate of 25 ml / min). The desired fractions were concentrated to provide maleimidocaproyl-valine-citrulline-p-hydroxymethylaminobenzene-MMAF (MC-val-cit-PAB-MMAF) 9 as a white solid. Rendimeinto: 11 mg (25%). ES-MS m / z 1330.29 [M + H] A 1352.24 [M + Na] +; UV 205.248 nm. Example 11 - Preparation of MC-val-cit-PAB-MMAF tert-butyl amide 10 Compound 3 (217 mg, 0.276 mmol, 1.0 eq.), Compound AB (204 mg, 0.276 mmol, 1.0 eq.), And HOBt (11 mg, 0.0828 mmol, 0.3 eq.) Were diluted with pyridine / DMF ( 6 ml). DIEA (0.048 ml) was added to this mixture, and the mixture was stirred ca. 16 hr. The volatile organics were evaporated in vacuo. The crude residue was purified by Chromatotron® (radial thin layer chromatography) with a gradient in the step (0-5-10% methanol in DCM) to provide MC-val-cit-PAB-MMAF ter-butyl amide 10. Rendimement: 172 mg (45%); ES-MS m / z 1386.33 [M + H] A 1408.36 [M + Na] +; UV 215, 248 nm. Example 12 - Preparation of AC10-MC-MMAE by conjugation of AC10 and MC-MMAE AC10 was treated, dissolved in 500 mM sodium borate and 500 mM soldio chloride at pH 8.0 with more than 100 mM dithiothreitol (DTT). After incubation at 37 ° C for approtely 30 minutes, the buffer was changed by elution on Sephadex G25 resin and eluted with PBS with 1 mM DTPA. The thiol / Ab value was determined by determining the concentration of reduced antibody from the absorbance at 280 nm of the solution and the concentration of thiol by reaction with DTNB (Aldrich, Milwaukee, Wl) and the determination of the absorbance at 412 nm. The reduced antibody dissolved in PBS was frozen on ice. The linker reagent of the drug, maleimidocaproyl-monomethyl auristatin E, i.e. MC-MMAE was dissolved in DMSO, diluted in acetonitrile and water to a known concentration, and added to the cooled antibody AC10 in PBS. After about one hour, an excess of maleimide was added to quench the reaction and cover any of the thiol groups of the unreacted antibody. The reaction mixture was concentrated by centrifugal ultrafiltration and AC10-MC-MMAE was purified and desalted by elution through G25 resin in PBS, filtered through 0.2 μm filters under sterile conditions, and frozen for storage. Example 13 - Preparation of AC10-MC-MMAF by conjugation of AC10 and MC-MMAF AC10-MC-MMAF was prepared by conjugation of AC10 and MC-MMAF following the procedure of Example 12. Example 14 - Preparation of AC10-MC-val -cit-PAB-MMAE by conjugation of AC10 and MC-val-cit-PAB-MMAE AClO-MC-val-cit-PAB-MMAE was prepared by conjugation of AC10 and MC-val-cit-PAB-Mmae following the procedure of Example 12. Example 15 - Preparation of AClO-MMC-val-cit-PAB-MMAF by conjugation of AC10 and MC-val-cit-PAB-MMAF (9) AClO-MC-val-cit-PAB-MMAF was prepared by conjugation of AC10 and MC-val-cit-PAB-MMAF (9) following the procedure of Example 12. Example 16 - Determination of the cytotoxicity of the selected compounds The cytotoxic activity of MMAF and Compounds 1-5 was evaluated on the lines Lewis Y positive OVCAR-3 cells, H3396 breast carcinoma, L2987 lung carcinoma, and LS174t colon carcinoma positive cell lines. Lewis Y can be tested for their cytotoxicity. . To evaluate the cytotoxicity of Compounds 1-5, the cells can be seeded at about 5-10,000 per well in 150 μl of culture medium then treated with graded doses of Compounds 1-5 in quadruplicates at the start of the assay. Cytotoxicity assays are commonly carried out for 96 hours after the addition of the test compounds. Fifty μl of resazurin tincture can be added to each well during the last 4 to 6 hours of incubation to assess the variable cells at the end of the culture. The dye reduction can be determined by fluorescent spectrometry using the excitation and emission wavelengths of 535nm and 590nm, respectively. For the analysis, the degree of reduction of resazurin by the treated cells can be compared to that of the untreated control cells. For the 1 h assays the cells can be pulsed with the drug for 1 h and then washed; the effect of cytotoxicity can be determined after 96 h of incubation. EXAMPLE 17 - In Vitro Cytotoxicity Tasting for the Selected Compounds Table 10 shows the cytotoxic effect of the Conjugates of Compounds 7-10 cILO, tested as described in General Procedure 1 on a CD30 + Karpas 299 cell line. present the data from two separate experiments. The cACLO conjugates of Compounds 7 and 9 were found to be slightly more active than cACLO-val-cit-MMAE: TABLE 10 - In other experiments, BR96-val-cit-MMAF was at least 250 times more potent than free MMAF. General Procedure I - Determination of cytotoxicity. To evaluate the cytotoxicity of Exemplary Conjugates 7-10, cells were cultured at approximately 5-10,000 per well in 150 μl of culture medium then treated with graded doses of Exemplary Conjugates 7-10 in quadruplicates at the start of the assay. The cytotoxicity tests were carried out for 96 hours after the addition of the test compounds. Fifty μl of the resazurin dye was added to each well during the last 4-6 hours of the incubation to evaluate the viable cells at the end of the culture. The dye reduction was determined by fluorescence spectrometry using the excitation and emission wavelengths of 535nm and 590nm, respectively. For the analysis, the amount of resazurin reduction by the treated cells was compared to that of the untreated control cells. Exarnple 18 - In Vitro Cell Proliferation Assay The efficacy of ADC can be measured by a cell proliferation assay using the following protocol (Promega Corp. Technical Bulletin TB288; Mendoza et al., (2002) Cancer Res. 62: 5485-5488 ): 1. An aliquot of 100 μl of cell culture containing approximately 104 cells (SKBR-3, BT474, MCF7 or MDA-MB-468) in one medium was deposited in each well of a 96-well opaque wall plate. 2. The control wells were prepared containing medium and without cells. 3. ADC was added to the experimental wells and incubated for 3-5 days. 4. The plates were equilibrated at room temperature for approximately 30 minutes. 5. One volume of CellTiter-Glo Reagent was added equal to the volume of the cell culture medium present in each well. 6. The contents were mixed for 2 minutes in an orbital shaker to induce cell proliferation. 7. The plate was incubated at room temperature for 10 minutes to stabilize the luminescence signal. 8. The luminescence was recorded and reported in graphs as RLU = units of relative luminescence. Example 19 - Plasma purification in rats Plasma purification pharmacokinetics of antibody drug conjugates and total antibody were studied in Sprague-Dawley rats (Charles River Laboratories, 250-275 gms each). The animals were dosed by injection into the vein of the bolus tail (IV Push). Approximately 300 μl of whole blood was collected through the jugular cannula, or by pricking the tail, in recipients of lithium / heparin anticoagulant at each time point: 0 (predose), 10, and 30 minutes; 1, 2, 4, 8, 24 and 36 hours; and 2, 3, 4, 7, 14, 21, 28 days post dose. The total antibody was measured by ELISA-ECD / GxhuFc-HRP. The antibody drug conjugate was measured by ELISA MMAE / MMAF / ECD-Bio / SA-HRP. Example 20 - Purification of Plasma in Monkeys Plasma purification pharmacokinetics of antibody drug conjugates and total antibody can be studied in cynomolgus monkeys. Figure 12 shows a purification study of the two-stage plasma concentration after administration of H-MC-vc-MMAE to Cynomolgus monkeys at different doses: 0.5, 1.5, 2.5, and 3.0 mg / kg, administered in the day 1 and day 21. Total antibody and ADC concentrations were measured over time.
(H = Trastuzumab). Example 21 - In Vivo Efficacy of Tumor Volume in Transgenic Expression Mice Animals suitable for transgenic experiments can be obtained from standard commercial sources such as Taconic (Germantown, N.Y.). Many species are suitable, but female FVB mice are preferred because of their greater susceptibility to tumor formation.
FVB males can be used for coupling and vasectomizing CDs. 1 spike can be used to stimulate pseudopregnancy. Vasectomized mice can be obtained from any commercial provider. The founders can be bred either with FVB mice or with heterozygous 129 / BL6 x FVB p53 mice. Mice with heterozygosity in the p53 allele can be used to potentially increase tumor formation. Some Fl tumors are of mixed species. Founder tumors can only be FVB. Animals that have tumors (allograft propagated from Fo5 mmtv transgenic mice) can be treated with a single dose or multiple doses by IV injection of ADC. The volume of the tumor can be assessed at several time points after the injection. Example 22 - Synthesis of MC-MMAF through t-butyl ester Synthesis 1: MßVal-Val-Dil-Dap-Ph? -OlBu, 1001 MC-MeVal-Val-Dn-Dap-Phß-OtBu MC-MMAF MeVal-Val-Dil-Dap-Phe-OtBu (compound 1, 128.6 mg, 0.163 mmol) was suspended in CH2C12 (0.500 mL). 6-Maleimidocaproic acid (68.9 mg, 0.326 mmol) and 1,3-diisopropylcarbodiimide (0.0505 mL, 0.326 mmol) were added followed by pyridine (0.500 mL). The reaction mixture was allowed to stir for 1.0 hr. HPLC analysis indicated the complete consumption of the starting compound 1. The volatile organics were evaporated under reduced pressure. The product was isolated through rapid column chromatography, using a stage glycerin of 0 to 5% methanol in CH2C12. A total of 96 mg of pure MC-MeVal-Val-Dil-Dap-Phe-OtBu (12) (60% yield) was recovered. ES-MS m / z 981.26 [M + H] A 1003.47 [M + Na] +; 979.65 [M-H] A MC-MeVal-Val-Dil-Dap-Phe-OtBu (Compound 12, 74 mg, 0.0754 mmol) was suspended in CH2C12 (2.0 mL) and TFA (1 mL) at room temperature. After 2.5 hr, the HPLC analysis indicated the complete consumption of the starting material. The volatile organics were evaporated under reduced pressure, and the product was isolated through preparative RP-HPLC, using a Phenomenex C12 Synergi Max-RP 80A Column (250 x 21.20 mm). Eluent: linear gradient 10% to 90% MeCN / 0.05% TFA (aq) for 30 minutes, then isocratic 90% MeCN / 0.05% TFA (aq) for about 20 additional minutes. ES-MS m / z 925.33 [M + H] A 947.30 [M + Na] +; 923.45 [M-H] A Example 23a - Synthesis of MC-MMAF (11) through dimethoxybenzyl ester Synthesis 2: Fmoc-Oap-Pfie-ODMB Dap-PheOODMB Fmoc-MeVal-Val-DR-Dap.pi e-ODMB M & -MBVat-Val-D? L-Dap-Phß-ODMB MC-MMAF Preparation of Fmoc-L-Phenylalanine-2,4-dimethoxybenzyl ester (Fmoc-Phe-ODMB) A round bottom flask of 5-1 of 3 necks was charged with Fmoc-L-Phenylalanine (200 g, 516 mmol Bachem), 2,4-dimethoxybenzyl alcohol (95.4 g, 567 mmol, Aldrich), and CH2C12 (2.0 1). N, N-dimethylfomamide t-butyl acrylate (155 ml, 586 mmol, Fluka) was added to the resulting suspension for 20 min under N2, which resulted in a clear solution. The reaction was then stirred at room temperature overnight, after which time the TLC analysis (0.42, Heptane / EtOAc = 2: 1) indicated that the reaction was complete. The reaction mixture was concentrated under reduced pressure to give a light yellow oil, which was redissolved in CH2C12 (200 ml) and purified through a short plug of silica gel (25 cm x 25 cm, CHC12) to give a spuna without color (250 g). MeCN (1 L) was added to the resulting foam, which completely dissolved the batch. It was then concentrated to dryness and redissolved in MeCN (11) and the resulting suspension was stirred for 1 h, filtered and the filtered slurry was rinsed with MeCN (2 x 200 ml) to give Fmoc-L-phenylalanine-2, 4-dimethoxybenzyl ester as a white solid (113.58 g, 41%, 95.5% AUC by HPLC analysis). Data: HPLC. Preparation L-Phenylalanine-2,4-dimethoxybenzyl ester (Phe-ODMB) A 500 ml round bottom flask was charged with Fmoc-L-phenylalanine-2, -dimethoxybenzyl ester (26.00g, 48.3 mmol), CH2C12 (150 ml) and diethylamine (75 ml, Acros). The mixture was stirred at room temperature and the termination was monitored by HPLC. After 4 h, the mixture was concentrated (bath at temp < 30 ° C). The residue was resuspended in CHC12 (200 ml) and concentrated. This was repeated once. To the residue was added MeOH (20 ml), which caused the formation of a gel. This residue was diluted with CH2C12 (200 ml), concentrated and the turbid oil was left under vacuum overnight. The residue was suspended in CH2C12 (100 ml), then toluene (1 20 ml) was added. The mixture was concentrated and the residue was left under vacuum overnight. Data: HPLC, ÍH NMR. Preparation of Fmoc-Dolaproin (Fmoc-Dap) Boc-Dolaproin (58.8 g, 0.205 mol) was suspended in 4 N HCL in 1,4-dioxane (256 ml, 1.02 mol, Aldrich). After stirring for 1.5 hours, TLC analysis indicated that the reaction was complete (10% MeOH / CH2CL2) and the mixture was concentrated to dryness wax. 1,4-dioxane was charged (50 ml) was added and the mixture was concentrated to dryness and dried under vacuum overnight. The resulting white solid was dissolved in H20 (400 ml) and transferred to a 3-1 three-necked round bottom flask with a mechanical stirrer and temperature probe. N, N-diisopropylethylamine (214.3 ml, 1.23 mol, Acros) was added for one minute, causing an exotherm from 20.5 to 28.2 ° C (intena). The mixture was cooled in an ice bath and 1,4-dioxane (400 ml) was added. A solution of Fmoc-OSu (89.90 g, 0.267 mol, Advanced ChemTech) in 1,4-dioxane (400 ml) was added from an addition funnel for 15 minutes, keeping the reaction temperature below 9 ° C. The mixture was allowed to warm to room temperature and was stirred for 19 hours, after which the mixture was concentrated by rotary evaporation to an aqueous mixture (390 g). The suspension was diluted with H20 (750 ml) and Et20 (750 ml), causing a copious white precipitate to form. The layers are separated, keeping the solids with the organic layer. The aqueous layer was acidified using conc. HCL. (30 ml) and extracted with EtOAc (3 x 500 ml). The condensed extracts were dried over MgSO4, filtered and concentrated to give 59.25 g of a yellow oil A. The Et20 extract was extracted once with sat. NaHCO3. (200 ml), keeping the solids with the aqueous layer. The aqueous suspension was acidified using HCL conc. (50 ml) and extracted with Et20 (50 ml) maintaining the solids with the organic layer. The organic layer was filtered and concentrated to give 32.33 g of a yellow oil B. The two oils (A and B) were combined and purified by flash chromatography on silica gel eluting with CH2C12 (3.5 1), then 3% MeOH / CH2C12 (9 1) to give 68.23 g of Fmoc-dolaproine as a white foam (81%, 97.5% pure by HPLC (AUC) Preparation of Fmoc-Dap-Phe-ODMB Crude Phe-ODMB (48.3 mmol) was suspended in anhydrous DMF (105 ml, Acros) for 5 minutes and Fmoc-Dap (19.80 g, 48.3 mmol) was added.The mixture was cooled in an ice bath and TBTU (17.08 g, 53.20 mmol, Matrix Innovations) was added. N, N-diisopropylethylamine (25.3 mL, 145.0 mmol, Acros) was added via geringa for 3 min.After lh, the ice bath was removed and the mixture allowed to warm for 30 min. The mixture was emptied in water (11) and extracted with ethyl acetate (300 ml) After separation, the aqueous layer was re-extracted with ethyl acetate (2 x 150 ml). The combined layers were washed with brine (150 ml), dried (MgSO 4) and filtered (filter paper) to remove insolubles (inorganic and some dibenzofulvene). After concentration, the residue (41 g) was absorbed onto silica (41 g) and purified by chromatography (22 cm x 8 cm column, 65% Heptane / EtOAc (2.5 1), 33% Heptane / EtOAc (3.8 1), to give 29.4 g of the product as a white foam (86%, 92% purity by HPLC) Data: HPLC, 1 H NMR, TLC (1: 1 EtOAc / Heptane Rf = 0.33, red dye in vanillin). Preparation of Dap-Phe-ODMB A 1-1 round bottom flask was charged with Fmoc-Dap-Phe-ODMB (27.66 g), CH2CL2 (122 ml) and diethylamine (61 ml, Acros). The temperature was monitored and the termination was monitored by HPLC.After 7 h, the mixture was concentrated (bath at temp <30 ° C) .The residue was suspended in CH2C12 (300 ml) and concentrated.This was repeated two times. times to the residue was added MEOH (20 ml) and CH2C12 (300 ml), and the solution was concentrated.The residue was suspended in CH2C12 (100 ml) and toluene (400mL), concentrated, and the residue was left under empty during night to give a residue like cream. Data: HPLC, ÍH NMR, MS. Preparation of Fmoc-MeVal-Val-Dil-Dap-Phe-ODMB Crude Dap-Phe-ODMB (39.1 mmol) was suspended in anhydrous DMF (135 mL, Acros) for 5 minutes and Fmoc-MeVal-Val-Di 1 was added. -OH (24.94g, 39.1 mmol, see Example 2 for the preparation). The mixture was cooled in an ice bath and TBTU (13.81g) was added., 43.0 mmol, Matrix Innovations). NN-Diisopropylethylamine (20.5 ml, 1 17.3 mmol, Acros) was added via syringe for 2 minutes. After 1 hour, the ice bath was removed and the mixture allowed to warm for 30 min. The mixture was poured into water (1.5 1) and diluted with ethyl acetate (480 ml). After standing for 15 minutes, the layers were separated and the aqueous layer was extracted with ethyl acetate (300 ml). The combined organic layers were washed with brine (200 ml), dried (MgSO4) and filtered (filter paper) to remove insolubles (inorganic and some dibenzofulvene). After concentration, the residue (49 g) was removed from the flask and absorbed onto silica (49 g) and purified by chromatography (day column of 15 cm x 10 cm; 2: 1 EtOAc / Heptane (3 1), EtOAc (5 1); 250 ml fractions) to give 31.84 g of Fmoc-MeVal-Val-Dil-Dap-Phe-ODMB as a white foam (73%, 93% pure by HPLC (AUC) Data: HPLC, TLC (2: 1 EtOAc / heptane, Rf = 0.21, red dye in vanillin.) Preparation of MeVal-Val-Dil-Dap-Phe-ODMB A 1-1 round-bottomed flask was charged, with Fmoc-MeVal-Val-Dil-Dap-Phe- ODMB (28.50 g), CH2C12 (80 ml) and diethylamine (40 ml) The mixture was stirred at room temperature overnight and then concentrated under reduced pressure.The residue was absorbed onto silica (30 g) and purified by rapid chromatography (column 15 cm x 8 cm, 2% MeOH / DCM (2 1), 3% MeOH / DCM (11), 6% MeOH / DCM (4 1), 250 ml fractions) to give 15.88 g of MeVal-Val-Dil-Dap-Phe-ODMB as a white foam (69%, 96% purity by HPLC (AUC)) Data: HPLC, TLC (6% MeOH / DCM, Rf = 0.24, red dye in vanillin) Preparation of MC-MoVal-Val-Dil-Dap-Phe-ODMB A 50-ml round bottom matrix was loaded with MeVal-Val-Dil-Dap-Phe-ODMB (750 mg, 0.85 mmol), anhydrous DMF (4 ml), maleimidocaproic acid (180 mg, 0.85 mmol), and TBTU (300 mg, 0.93mrnol, Matrix Innovations) at temperature - - ambient . N was added, N-Diisopropylethylamine (450 μl, 2.57 mmol) through geringa. After 1.5 hours, the mixture was poured into water (50 ml) and diluted with ethyl acetate (30 ml). NaCl was added to improve the separation. After separation of the layers, the aqueous layer was extracted with ethyl acetate (25 ml). The combined organic layers were dried (MgSO 4), filtered and concentrated. The resulting oil (1 g) was purified by flash chromatography [100 ml silica; 25% Heptane / EtOAc (100 mL), 10% Heptane / EtOAc (200 mL), EtOAc (1.5 1)] to give MC-MeVal-Val-Dil-Dap-Phe-ODMB (13) as a white foam (521) mg, 57%, 94% purity by HPLC (AUC) Data: H NMR, HPLC Preparation of MC-MeVal-Val-Dil-Dap-Phe-OH (MC-MMAF) (11) A flask was charged 50-ml round bottom, with MC-MeVal-Val-Dil-Dap-Phe-ODMB (Compound 13, 428 mg, 0.39 mmol) and dissolved in 2.5% TFA / CH2C12 (20 ml) The solution turned pink- purple for 2 min. The termination was monitored by HPLC and TLC (6% MeOH / DCM, KMn04 staining.) After 40 min, three drops of water were added and the turbid pink-purple mixture was concentrated to give 521 mg of a pink residue Purification by chromatography (15% IPA / DCM) gave 270 mg of MC-MMAF (73%, 92% purity by HPLC) as a white solid.
Example 23b - Synthesis of the mc-MMAF analog MB- eJ? -Val-Dil-Dap-Phe -OtBu B- MAF MeVal-Val-Dil-Dap-Phe-OtBu (compound 1, 35 mg, 0.044 mmol) was suspended in DMF (0.250 mL). 4- (2,5-Dioxo-2, 5-dihydro-pyrrol-1-yl) -benzoic acid (11 mg, 0.049 mmol) and HATU (17 mg, 0.044 mmol) was added followed by DIEA (0.031 mL, 0 17 mmol). This reaction mixture was allowed to stir for 2.0 hr. The HPLC analysis indicated the complete consumption of the initial compound 1. The product was isolated through preparative RP-HPLC, using a Phenornenex C12 Synergi Max-RP 80Á Column (250 x 21.20 mm). Eluent: linear gradient 10% to 80% MeCN / 0.05% TFA (aq) for 8 minutes, then isocratic 80% MeCN / 0.05% TFA (aq) for about 12 additional minutes. A total of 20 mg of pure product (14) (0.02 mmol, 46% yield) was isolated. ES-MS m / z 987.85 [M + H] -1019.41 [M + Na] +; 985.54 [M-H] A MB-MeVal-Vai-Dil-Dap-Phe-OtBu (compound 14.38 mg, 0.0385 mmol) was suspended in CH2C12 (1 mL) and TFA (1 mL). The mixture was stirred for 2.0 hr, and then the volatile organics were evaporated under reduced pressure. The product was purified by preparative RP-HPLC, using a Phenomenex C12 Synergi Max-RP 80Á Column (250 x 21.20 mm). Eluent: linear gradient 10% to 80% MeCN / 0.05% TFA (aq) for 8 minutes, then isocratic 80% MeCN / 0.05% TFA (aq) for 12 additional minutes. A total of 14.4 mg of the MB-IWAF product was isolated (0.015 mmol, 40% yield). ES-MS m / z 930.96 [M + H] + 952.98 [M + Na] +; 929.37 [M-H] "Example 23c - Preparation of MC-MeVal-Cit-PAB-MMAF (16) To a room temperature suspension of Fmoc- MeVal-OH (3.03 g, 8.57 mmol) and N, N '-disuccimidyl carbonate (3.29 g, 12.86 mmol) in CH2C12 (80 mL) was added DIEA (4.48 mL, 25.71 mmol) . This reaction mixture was allowed to stir for 3.0 hr, and then it was emptied into a separatory funnel where the organic mixture was extracted with 0.1 M HCl (aq). The crude organic residue was concentrated under reduced pressure, and the product was isolated by flash column chromatography on silica gel using a linear gradient of 20-100% ethyl acetate / hexanes. A total of 2.18 g of pure Fmoc-MeVal-OSu was recovered (4.80 mmoles, 56% yield). To a room temperature suspension of Fmoc-MeVal-OSu (2.18 g, 4.84 mmol) in DME (13 mL) and THF (6.5 mL) was added a solution of L-citrulline (0.85 g, 4.84 mmol) and NaHCO 3 (0.41 g). g, 4.84 mmol) in H20 (13 ml). The suspension was allowed to stir at room temperature for 16 hr, then extracted in ter-BuOH / CHCl3 / H20, acidified to pH = 2-3 with 1 M HCl. The organic phase was separated, dried and concentrated under reduced pressure. The residue was triturated with diethyl ether resulting in 2.01 g of Fmoc-MeVal-Cit-COOH which was used without further purification. The crude Fmoc-MeVal-Cit-COOH was suspended in 2: 1 CH2Cl / MeOH (100 ml), and p-aminobenzyl alcohol (0.97 g, 7.9 mmol) and EEDQ (1.95 g, 7.9 mmol) were added thereto. This suspension was allowed to stir for 125 hr, then the volatile organics were removed under reduced pressure, and the residue was purified by flash column chromatography on silica gel using 10% MeOH / CH2Cl2. Fmoc-MeValCit-PAB-OH Puree (0.55 g, 0.896 mmol, 18.5% Rendimeinto) was recovered. ES-MS m / z 616.48 [M + H] A To a suspension of Fmoc-MeVal-Cit-PAB-OH (0.55 g, 0. 896 mmol) in CH2C12 (40 ml) was added STRATOSPHERES ™ (piperizine resin bond) (> 5 mmol / g, 150 mg). After being stirred at room temperature for 16 hr the mixture was filtered through celite (pre-washed with MeOH), and concentrated under reduced pressure. The residue was triturated with diethyl ether and hexanes. The resulting solid material, MeVal-Cit-PAB-OH, was suspended in CH2C12 (20 ml), and to this was added MC-OSU (0.28 g, 0.896 mmol), DIEA (0.17 ml, 0.99 mmol), and DMF (15 ml). This suspension was stirred for 16 hr, but HPLC analysis of the reaction mixture indicated incomplete reaction, thus, the suspension was concentrated under reduced pressure to a volume of 6 ml, then a solution of 10% NaHCO 3 (aq) was added and the suspension was stirred for an additional 16 hr. The solvent was removed under reduced pressure, and the residue was purified by rapid column chromatography on silica gel using a gradient of 0-10% MeOH / CH2Cl2, resulting in 42 mg (0.072 mmol, 8% yield) of MC -MeVal-Cit-P7? B-0H. To a suspension of MC-MeVal-Cit-PAB-OH (2.37 g, 4.04 mmol) and bis (nitrophenyl) carbonate (2.59 g, 8.52 mmol) in CH2C12 (10 mL) was added DIEA (1.06 mL, 6.06 mmol). This suspension was stirred for 5.5 hr, concentrated under reduced pressure and purified by trituration with diethyl ether. MC-MeVal-Cit-PAB-OCO-pNP (147 mg, 0.196 mmol) was suspended in a solution of 1: 5 pyridine / DMF (3 ml), and to this was added HOBT (5 mg, 0.039 mmol), DIEA ( 0.17 ml, 0.978 mmol) and MMAF (Cornpound 2, 150 mg, 0.205 mmol). This reaction mixture was stirred for 16 hr at room temperature, and then purified by preparative RP-HPLC (x3), using a Column Phenornenex C12 Synergi Max-RP 80A (250 x 21.20 mm). Eluent: linear gradient 10% to 90% MeCN / 0.05% TFA (aq) for 30 minutes, then isocratic 90% MeCN / 0.05% TFA (aq) for about 20 additional minutes. MC-MeVal-Cit-PAB-MMAF (16) was obtained as a yellowish solid (24.5 mg, 0.0182,0.45% yield). ES-MS m / z 1344.95 [M + H] A 1366.94 [M + Na] Example 23d - Preparation of succinimide ester of suberyl-Val-Cit-PAB-MMAF (17) Compound 17 Compound 1 (300 mg, 0.38 mmol), Fmoc-Val-Cit-PAB-pNP (436 mg, 0.57 mmol, 1.5 eq.) Were suspended in anhydrous pyridine, 5 ml. HOBT (10 mg, 0.076 mmol, 0.2 eq.) Was added followed by DIEA (1 99 μL, 14 mmol, 3 eq.). The reaction mixture was sonicated for 10 min, and then stirred overnight at room temperature. The pyridine was removed under reduced pressure, the residue was re-suspended in CH2CL2. The mixture was separated by flash chromatography on silica gel in a gradient of the MeOH step, from 0 to 10%, in CH2C12. The fractions containing product were removed, concentrated, dried in vacuo overnight to give 317 mg (59% yield) of Fmoc-Val-Cit-PAB-MMAF-OtBu. ES-MS m / z 1415.8 [M + H] A Fmoc-Val-Cit-PAB-MMAF-OtBu (100 mg) was stirred in 20% TFA / CH2C12 (10 mL), for 2 hrs. The mixture was diluted with CH2C12 (50 mL). The organic layer was washed successively with water (2 x 30 ml) and brine (1 x 30 ml). The organic phase was concentrated, loaded onto a pad of silica gel in 10% MeOH / CH2Cl2. The product was eluted with 30% MeOH / CH2Cl2. After drying in vacuum overnight, Fmoc-Val-Cit-PAB-MMAF was obtained as a white solid, 38 mg, 40% yield. ES-MS m / z 1357.7 [M-H] A Fmoc-Val-Cit-P7AB-MMAF, 67 mg, was suspended in CH2CL2 (2 ml), diethylamine (2 ml) and DMF (2 ml). The mixture was stirred for 2 hrs at room temperature. The solvent was removed under reduced pressure. The residue was co-evaporated with pyridine (2 ml), then with toluene (2 x 5 ml), dried in vacuum. Val-Cit-PAB-MMAF was obtained as a brown oil, and was used without further purification. All Val-Cit-PAB-MMAF prepared from 67 mg of Fmoc-Val-Cit-PAB-MMAF, was suspended in pyridine (2 ml), and added to a disuccinimidyl suberate solution (74 mg, 0.2 mmol, 4 eq.), in pyridine (1 ml). The reaction mixture was stirred at room temperature. After 3 hrs ether (20 ml) was added. The precipitate was collected, washed with an additional amount of ether. The reddish solid was suspended in 30% MeOH / CH2Cl2, filtered through a pad of silica gel with 30% MeOH / CH2Cl2 as an eluent. Compound 17 was obtained as a white solid, 20 mg (29% yield). ES-MS m / z 1388.5 [MH] "Example 24 - Efficacy and vivo of the Antibody-Drug Conjugates mcMMAF E. Fficacy of cA ClO-mcMMAF in Karpas-299 xenografts ALCL: To evaluate the in vivo efficacy of cAClO-mcMMAF with an average of 4 drug residues per antibody (cAClO-mcF4), human ALCL Karpas-299 cells were implanted subcutaneously in immunodeficient mice CB-17 SCID (5x1O6 cells per mouse). Tumor volumes were calculated using the formula (0.5xLxW2) where L and W the largest and the shortest of two bidirectional measurements.
When the average tumor volume in the study animals reached approximately 100 mm3 (range 48-162) the mice were divided into 3 groups (5 mice per group) and left either untreated or given a single intravenous injection. through the tail vein of either 1 or 2 mg / kg cAC10-mcF4 (Figure 1). Tumors in the untreated mice grew rapidly to an average volume of > 1,000 mm3 within 7 days of the start of therapy. In contrast, all tumors treated with cAC10-mcF4 showed rapid regression with 3/5 in the group of 1 mg / kg and 5/5 in the group of 2 mg / kg obtaining complete tumor response. Although the tumor in one of the complete responses in the 2 mg / kg group recurred approximately 4 weeks later, no tumors detectedblocks in the 4/5 responders remaining in this group and in the 3 full response in the group of 1 mg / kg at 10 weeks post therapy. Efficiency of cBR96-mcMMAF in L2987 NSCLC xenografts: cBR96 is a chimeric antibody that recognizes the Le antigen. To evaluate the in vivo efficacy of cBR96-mcMMAF with 4 drugs per antibody (cBR96-mcF4) L2987 non-small cell lung cancer (NSCLC) tumor fragments were implanted in athymic nude mice. When the tumors averaged approximately 100 mm3 the mice were divided into 3 groups: untreated groups and 2 therapies. For the therapy, as shown in Figure 3a, the mice were administered eBR96-mcF4 at either 3 or 10 mg / kg / injection every 4 days for a total of 4 injections (q4dx4). As shown in Figure 3b, the mice were administered cBR96-mcF4 or a non-binding control conjugate, cAC10-mcF4, at 10 mg / kg / injection every 4 days for a total of 4 injections (q4dx4). As shown in Figures 3a and 3b, BR96-mcF4 produced a pronounced growth retardation compared to the controls. Figure 2 shows a single-dose, in vivo efficacy test of cAClO-mcMMAF in subcutaneous L540CY. For this study there were 4 mice in the untreated group and 10 in each of the treatment groups. Example 25 - In Vitro Efficacy of MC-MMAF Antibody-Drug Conjugates Activi ty and cACLO-antibody-drug conjugates against CD30 + cell lines. Figures 4a and 16b show the dose response curves of a representative experiment where the cultures of Karpas 299 (anaplastic large cell lymphoma) and LA28, (Hodgkin's Lymphoma) were incubated with serially diluted cACLO-mcMMAF (Figure 4a ) or cACLO-vcMMAF (Figure 4b) for 96 hours. The cultures were labeled for 4 hours with 50 μM of resazurin [7-hydroxy-3H-phenoxazine-3-one 10-oxide] and fluorescent measurement. The data was reduced in GraphPad Prism version 4.00 using the adjustment procedure of the 4-parameter dose response curve. The IC 50 values were defined as the concentration where the growth is reduced by 50% compared to untreated control cultures. Each concentration was treated in quadruplicate. Activi ty of the cBR96-antibody conjugates -drug against Le cell lines? . Figures 5a and 5b show the dose response curves of a representative experiment in which cultures of H3396 (breast carcinoma) and L2987 (non-small cell lung carcinoma) were incubated with serially diluted cBR96-mcMMAF (Figure 5a ) or -vcMMAF (Figure 5b) for 96 hours. Cultures were labeled for 4 hours with 50 μM resazurin and fluorescent measurement. The data were reduced in GraphPad Prism version 4.00 using the adjustment procedure to the 4-parameter dose response curve. IC50 values were defined as the concentration where the growth was reduced by 50% in comparison with untreated control cultures. Each concentration was tested in quadruplicate. Activity of clF6-antibody-drug conjugates against cell lines of CD70 + renal cell carcinoma. Figures 6a and 6b show dose response curves of a representative experiment in which cultures of Caki-1 and 786-0 cells were incubated with serially diluted clF6-mcMMAF (Figure 6a) or -vcMMAF (Figure 6b) for 96 hours The cultures were labeled for 4 hours with 50 μM resazurin and the measurement of fluorescence. The data was reduced in GraphPad Prism version 4, 00 using the adjustment procedure of the 4-parameter dose response curve. IC50 values were defined as the concentration where growth was reduced by 50% compared to untreated control cultures. Each concentration was tested in quadruplicate. Example 26 - Purification of trastuzumab A vial containing 440 mg HERCEPTIN® (huMAb4DS-8, rhuMAb HER2, US Patent No. 5821337) antibody) was dissolved in 50 ML MES buffer (25 mM MES, 50 mM NaCl, pH 5.6) and was loaded onto a cation exchange column (Sepharose S, 15 cm x 1.7 cm) which has been balanced in the same regulator. The column was then washed with the same buffer (5 column volumes). Trastuzumab was eluted by raising the NaCl concentration of the buffer to 200 mM. Fractions containing the antibody were pooled, diluted to 10 mg / ml, and dialyzed into a buffer containing 50 mm of potassium phosphate, 50 mM NaCl, 2 mM EDTA, pH 6.5, Example 27 - Preparation of trastuzumab-MC-MMAE by conjugation of trastuzumab and MC-MMAE Trastuzumab, dissolved in 500 mM sodium borate and 500 mM sodium chloride at pH 8.0 was treated with an excess of 100 mM dithiothreitol (DTT). After incubation at 37 ° C for approximately 30 minutes, the buffer was changed by elution on Sephadex G25 resin and eluted with PBS with 1 mM DTPA. The thiol / Ab value was verified by determining the concentration of the reduced antibody from the absorbance at 280 nm of the solution and the thiol concentration by the reaction with DTNB (Aldrich, Milwaukee, Wl) and the determination of the absorbance at 412 nm. The reduced antibody was dissolved in PBS and cooled on ice. The linker reagent of the drug, maleimidocaproyl-monomethyl auristatin E (MMAE), i.e. MC-MMAE was dissolved in DMSO, diluted in acetonitrile and water to a known concentration, and added to the reduced antibody trastuzumab cooled in PBS. After about one hour, an excess of maleimide was added to cool the reaction and plug any of the thiol groups of unreacted antibody. The reaction mixture was concentrated by centrifugal ultrafiltration and the trastuzumab-MC-MMAE was purified and desalted by elution through G25 resin in PBS, filtered through 0.2 μm filters under sterile conditions, and frozen for storage. Example 28 - Preparation of trastuzumab-MC-MMAF by conjugation of trastuzumab and MC-MMAF Trastuzumab-MC-MMAF was prepared by conjugation of trastuzumab and MC-MMAF following the procedure of Example 27. Example-29 - Preparation of trastuzumab-MC -val-cit-PAB-MMAE by the conjugation of trastuzumab and MC-val-cit-PAB-MMAE Trastuzumab-MC-val-cit-PAB-MMAE was prepared by conjugation of trastuzumab and MC-val-cit-PAB- MMAE following the procedure of Example 27. Example. 30 - Preparation of trastuzumab-MC-va-cit-PAB-MMAF by conjugation of trastuzumab and MC-val-cit-PAB-MMAF 9 Trastuzumab-MC-val-cit-PAB -MMAF was prepared by conjugation of trastuzumab and MC-val-cit-PAB-MMAF 9 following the procedure of Example 27. Example 31 r Toxicity in rats The acute toxicity profile of free drugs and ADC was evaluated in adolescent Sprague rats -Dawley (75-125 gms each, Charles River Laboratories (Hollister, CA) .The animals are injected On day 1, complete chemistry and hematology profiles were obtained at baseline, day 3 and day 5 and complete necrosis was performed on day 5. Liver enzyme measurements were made on all animals and routine histology as performed in three random animals for each group for the following tissues: sternum, liver, kidney, thymus, spleen, large and small intestine. The experimental groups were as follows: Administration mg / kg MMAF / Group MMAF / MAb N / sex m2 1 Vehicle 0 0 0 2 / F 2 trastuzumab-MC-val- 9.94 840 4.2 6 / F cit-MMAF 3 trastuzumab-MC-val- 24.90 2105 4.2 6 / F cit-MMAF 4 trastuzumab-MC (Me) - 10.69 840 3.9 6 / F val-cit- PAB-MMAF 5 trastuzumab-MC (Me) - 26.78 2105 3.9 6 / F val-cit-PAB-MMAF 6 trastuzumab-MC-MMAF 10.17 840 4.1 6 / F 7 trastuzumab-MC-MMAF 25.50 2105 4.1 6 / F 8 trastuzumab-MC-val- 21.85 2105 4.8 6 / F cit-PAB-MMAF For trastuzumab-MC-val-cit-MMAF, trastuzumab-MC (Me) -val-cit-PAB-MMAF, trastuzumab-MC-MMAF and trastuzumab-MC-val-cit-PAB-MMAF, the μg MMAF / m2 was calculated using 731.5 as the MW of MMAF and 145167 as the MW of Herceptin. The body surface area was calculated as follows: [. { (body weight in grams for 0.667 of power) x 11.8} / 10000]. (Guide for Industry and Reviews, 2002). The dose solutions were administered by a single injection into the intravenous bolus tail vein on Day 1 of the Study at a dose volume of 10 ml / kg. The body weights of the animals were measured pre-dose on Day 1 of the Study and daily after it. Whole blood was collected in tubes containing EDTA for hematology analysis. Whole blood was collected in serum separator tubes for clinical chemistry analysis. Pre-dose blood samples were collected on Day 4 of the Study, on Day 3 of Study and Day 5 of Study. Complete blood was also collected in tubes containing sodium heparin at necropsy and the plasma was frozen at -70 ° C for possible further analysis. The following tissues were collected and placed in neutral buffer of formalin at necropsy: liver, kidneys, heart, thymus, spleen, brain, sternum and sections of the GI tract, including the stomach and the large and small intestines. The sternum, small intestine, large intestine, liver, thymus, spleen and kidney were examined. The serum enzyme levels associated with the liver at each time point were compared to a range (5th and 95th percentiles) of normal female Sprague-Dawley rats. The cell and platelet counts at each time point were compared to a range (5th and 95th percentiles) of normal female Sprague-Dawley rats. High dose study in normal female Sprague-Dawley rats: Group 1 Vehicle Group 2 trastuzumab-MC-MMAF, 52.24mg / kg, 4210μg / m2 Group 3 trastuzumab-MC-MMAF, 68.25mg / kg, 5500μg / m2 Group 4 trastuzumab-MC-MMAF, 8600mg / kg, 6930μg / m2 Tissues from 11 animals were submitted to routine histology. These animals had been part of an acute mixed dose toxicity study using a trastuzumab-MC-MMAF immunoconjugate. The animals were followed for 12 days after dosing. Example 32 - Toxicity / Safety for the Cynomolgus Monkey Three groups of four (2 males, 2 females) of native Macaca fascicularis (cynomolgus monkey) were studied for trastuzumab-MC-vc-PAB-MMAE and trastuzumab-MC-vc-PAB- MMAF Intravenous administration was conducted on days 1 and 22 of the study.
- - H = trastuzumab The dosage is expressed in the surface area of an animal in order to be relevant to other species, i.e. the dosage at μg / m2 is independent of the species and thus comparable among the species. The ADC formulations contain PBS, 5.4 mM, sodium phosphate, 4.2 mM potassium phosphate, 140 mM sodium chloride, pH 6.5. Blood was collected for predose wing of hematology analysis and at 5 min, 6 hr, 10 hr and 1, 3, 5, 7, 14, 21 days after each dose. The erythrocyte (RBC) and platelet (PLT) counts were measured by the photodispersion method. The leukocyte counts (WBC) were measured by the peroxides / basophils method. The reticulocyte counts were measured by the photodispersion method with cationic dye. The cell counts were measured in the Advia 120 device. ALT (alanine aminotransferase) and AST (aspartate aminotransferase) were measured in U / L using UV / NADH methodology; IFCC on an Olympus AU400 device, and using ELISA Ab Total ECD / GxhuFc-HRP. Joint Tests Ab ELISA - MMAE / MMAF // ECD- - - Bio / SA-HRP. Example 33 - Production, Characterization and Humanization of Monoclonal Anti-ErbB2 4D5 Antibody The murine monoclonal antibody 4D5 that specifically binds the extracellular domain ErbB2 was produced as described in Fendly et al. , (1990) Cancer Research 50: 1550-1558. Briefly, NIH 3T3 / HER2-340o cells (expressing approximately 1 x 105 ErbB2 molecules / cells) produced as described in Hudziak et al. Proc. Nati Acad. Sci. (USA) 84: 7158-7163 (1987) were cultured with phosphate buffered saline (PBS) containing 25mM EDTA and used to immunize BALB / c mice. The mice were administered i.p. of 107 cells in 0.5ml PBS at weeks 0, 2, 5 and 7. Mice with antisera that were in unoprecipitated ErbB2 labeled 32P were given in i.p injections. of an ErbB2 membrane extract purified from wheat germ agglutinin-Sepharose (WGA) at weeks 9 and 13. This was followed by an i.v. of 0.1 ml of the preparation of ErbB2 preparation and the spherocytes were fused with the mouse myeloma line X63-Ag8.653. Hybridoma supernatants were systematically detected by ErbB2 binding by ELISA and radioimmunoprecipitation. Representation and characterization of the Epitope The binding of the ErbB2 epitope by the monoclonal antibody 4D5 was determined by competitive binding analysis (Fendly et al., Cancer Research 50: 1550-1558 (1990).) Cross-block studies were done by direct fluorescence in intact cells using PAND? XTM Screen Machine to quantify fluorescence The monoclonal antibody was conjugated to fluorescein isothiocyanate (FITC), using established procedures (Wofsy et al., Selected Methods in Cellular Immunology, p.287, Mishel and Schiigi (eds. ) San Francisco: WJ Freeman Co. (1980) Confluent monolayers of NIH 3T3 / HER2-340o cells were trypsinized, washed once, and reslurried at 1.75 x 106 cells / ml in cold PBS containing 0.5% albumin. of bovine serum (BSA) and 0.1% NaN3 A final concentration of 1% latex particles (IDC, Portland, OR) was added to reduce the coagulation of the membranes of PANDEX ™ plate Cells in suspension, 20 μl, and 20 μl of purified monoclonal antibodies (100 μg / ml to 0.1 μg / ml) were added to the wells of the PANDEX ™ plate and incubated on ice for 30 minutes. A predetermined dilution of the monoclonal antibody labeled FITC- in 20 μl was added to each well, incubated for 30 minutes, washed and the fluorescence was quantified by PANDEX ™. Monoclonal antibodies were considered to share an epitope if each blocks the binding of the other by 50% or more compared to an irrelevant monoclonal antibody control. In this experiment, monoclonal antibody 4D5 was assigned an epitope I (amino acid residues from about 529 to about 625, inclusive within the estrabular domain ErbB2.) The growth inhibitory characteristics of monoclonal antibody 4D5 were assessed using the breast tumor cell line , SK-BR-3 (see Hudziak et al., (1989) Molec. Cell, Biol. 9 (3): 1165-1172) In summary, the SK-BR-3 cells were detached using 0.25% (vol. / vol) of trypsin and suspended in a complete medium at a density of 4 x 10 5 cells per ml The aliquots of 100 μl (4 x 104 cells) were plated in 96-well microtiter plates, the cells were allowed to adhere, and then 100 μl of a medium alone or a medium containing monoclonal antibody (final concentration 5 μg / ml) was added. After 72 hours, the plates were washed twice with PBS (pH 7.5), stained with crystal violet (0.5% in methanol), and analyzed for relative cell proliferation as described in Sugarman et al. , (1985) Science 230: 943-945. Monoclonal antibody 4D5 inhibited retype cell proliferation of SK-BR-3 by approximately 56%. The 4D5 monoclonal antibody was also evaluated for its ability to inhibit tyrosine phosphorylation stimulated by HRG of the proteins in the Mr 180 range, 000 cell lysates compelts of MCF7 cells (Lewis et al., (1996) Cancer Research 56: 1457-1465). MCF7 cells were reported to express all known ErbB receptors, but at relatively low levels. Since ErbB2, ErbB3, and ErbB4 have almost identical molecular sizes, it is not possible to discern which protein is becoming phosphorylated tyrosine when whole cell lysates are evaluated by Western immunoassay. However, these cells are ideal for tyrosine HRG phosphorylation analysis because under the assay conditions used, in the absence of exogenously added HRG, they exhibit low to undetectable levels of tyrosine phosphorylation proteins in the range of 180,000. MCF7 cells were placed in 24-well plates and monoclonal antibodies were added to ErbB2 in each well and incubated for 30 minutes at room temperature.; then rHRGßl? 77_244 was added to each well for a final concentration of 0.2 nM, and the incubation was continued for 8 minutes. The medium was carefully aspirated from each well, and the reactions were stopped by the addition of 100 μl of the SDS sample buffer (5% SDS, 25 mM DTT, and 25 mM Tris-HCl, pH 6.8). Each sample (25 μl) was electrophoresed on a 4-12% gradient gel (Novex) and then electrophoretically transferred to a polyvinylidene difluoride membrane. Immunoassays of antiphosphotyrosine (4G10, from UBI, used at 1 μg / ml) were developed, and the intensity of the predominant reactive band at Mr, 180,000 was quantified by reflectance densitometry, as previously described (Holmes et al., (1992) Science 256: 1205-1210; Sliwkowski et al J ". Biol. Chem. 269: 14661-14665 (1994) Monoclonal antibody 4D5 significantly inhibited the generation of a tyrosine phosphorylation signal induced by HRG at M 180,000.In the absence of HRG, but was incapable of stimulating the tyrosine phosphorylation of proteins in the range of Mx 180,000.This antibody also did not cross-react with EGFR (Fendly et al .. Cancer Research 50: 1550-1558 (1990), ErbB3, or ErbB4. The monoclonal antibody 4D5 was able to block the stimulation of HRG by tyrosine phosphorylation by 50%. The growth inhibitory effect of monoclonal antibody 4D5 on MDA-MB175 and SK-BR-3 cells was assessed in the presence or absence of exogenous rHRGßl (Schaefer et al., Oncogene 15: 1385-1394 (1997).) ErbB2 levels in MDA-MB-175 cells are 4-6 times higher than the level found in normal breast epithelial cells and the ErbB2-ErbB4 receptor phosphorylates tyrosine constitutively in MDA-MB-175 cells.The monoclonal antibody 4D5 was able to inhibit Cell proliferation of MDA-MB-175 cells, both in the presence and absence of exogenous HRG The inhibition of cell proliferation by 4DS is independent of the expression level of ErbB2 (Lewis et al., Immunol. Immunotherm Cancer, 37: 255-263 (1993) A maximum inhibition of 66% in SK-BR-3 cells could be detected, however this effect can be overcome by exogenous HRG.The murine monoclonal antibody 4D5 was humanized, using a "mutagenesis genetic conversion" strategy, as is described in the U.S. Patent. No. 5821337, the full description of which is expressly incorporated herein by reference. The humanized monoclonal antibody 4D5 used in the following experiments was designated huMAb4D5-8. This antibody is of the IgGl isotype. REFERENCES CITED The present invention is not limited in scope by the specific embodiments described in the examples that are proposed as illustrations of some aspects of the invention and any of the embodiments that are functional equivalents are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will be apparent to those skilled in the art and are intended to fall within the scope of the appended claims. All references cited herein are incorporated by reference in their entirety and for all purposes to the same extent as if each publication or patent or individual patent application was indicated to be incorporated specifically and individually by reference in its entirety for all purposes .

Claims (10)

  1. CLAIMS A compound that has the formula DF or a conjugate thereof, or a pharmaceutically acceptable salt or solvate of the compound of the formula DF or the conjugate thereof, wherein the sinuous line of DF indicates H or a covalent bound site, and independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C-C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3 heterocycle) C8); R4 is selected from H, alkyl d-Cs, C3-C8 carbocycle, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and CX-C8 alkyl (C3-C8 heterocycle) ); R5 is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRaRb) n- wherein R and Rb are independently selected from H, C? -C8 alkyl, and C3-C8 carbocycle, and n is selected from 2, 3, 4, 5, and 6; R6 is selected from H, and C? -C8 alkyl; R7 is selected from H, alkyl QL-CS, carbocycle C3- - C8, aryl, C? -C8 alkylaryl, C? -C8 alkyl (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl (C3-C8 heterocycle); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle, and 0- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C? -C8 alkyl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that fluctuates from 1-1000; R 13 is C 2 -C 8 alkyl / -R 14 is H or C 1 -C 8 alkyl; each occurrence of R15 is independently, H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or alkyl - (CH2) n-S03-C? -C8; each occurrence of R16 is independently H, C? -C8 alkyl, or - (CH2) n-C00H; where n is an integer that fluctuates from 0 to 6.
  2. 2. A conjugate of the compound of claim 1 having the formula: L- (LU-DF) P or a pharmaceutically acceptable salt or solvate thereof, wherein L- is a Ligand unit, LU is a Linker unit, and p varies from 1 to about 20; u the Linker Unit may be present or absent.
  3. 3. The conjugate of claim 2 having the formula: L- (DF) p or a pharmaceutically acceptable salt or solvate thereof, wherein - is a Ligand unit, and p ranges from 1 to about 20.
  4. 4. A conjugate of the compound of claim 1 having the formula: LU-DF or a pharmaceutically acceptable salt or solvate thereof wherein -LU- is a Linker unit.
  5. 5. The conjugate of claim 2, wherein the conjugate is an antibody-drug conjugate having the formula: Ab - (- Aa-Ww -Yy-DF) P 'or a pharmaceutically acceptable salt or solvate thereof, wherein: Ab is an antibody, -Aa-Ww-Yy- comprises a Linker unit, • A is an Stretch unit, a is 0 or 1, each W is independently an Amino Acid unit, w is an integer that varies from 0 to 12, Y is a Swarming unit, and y is 0, 1 or 2, and p varies from 1 to approximately twenty.
  6. 6. The antibody-drug conjugate of claim 5 having the formula: A ±) - (DF) p wherein a, w, and y are each 0.
  7. 7. The antibody-drug conjugate of claim 5 wherein the antibody binds to the linker unit or to the drug through a cysteine residue of the antibody.
  8. 8. The antibody-drug conjugate of claim 7 wherein p is from 2 to 5.
  9. 9. The antibody-drug conjugate of claim 5 or 7 having the formula: ? N-R17-C (0H-WWw-Y 'Vy "DF wherein L is an antibody and R17 is C 1 -C 8 alkylene, C 3 -C 8 -carbocycle, -O- (C 1 -C 8 alkyl), 1-arylene, C 1 -C 10 alkylene-arylene, arylene-C? -C10 alkylene-, -C? -C10 alkylene- (C3-C8 carbocycle) -, - (C3-C8 carbocycle) -C? -C10 alkylene-, -C3-C8- heterocycle, -Cx- alkylene? C? 0- (C3-C8 heterocycle) -, - (C3-C8 heterocycle) -C? -C10 alkylene -, - (CH2CH20) r-, or - (CH2CH20) r-CH-; and r is an integer that varies from 1-10.
  10. 10. The antibody-drug conjugate of claim 9 having the formula: where w and y are each 0. 12. The antibody-drug conjugate of claim 5 wherein w is an integer ranging from 2 to 12. 13. The antibody-drug conjugate of claim 12 wherein w is 2. 14. The antibody-drug conjugate of claim 13 wherein Ww is -valin-citrulline-, phenylalanine-lysine-, or -N-methyl valine-citrulline-. 15. The antibody-drug conjugate of claim 12 wherein Ww is 5-aminovaleric acid, homo-phenylalanine lysine, tetraisoquinolinecarboxylate lysine, cyclohexylalanine lysine, isonepecotic acid lysine, beta- 19. The antibody-drug conjugate of claim 5 having the formula: wherein each occurrence of D is independently a compound selected from the DE Formulas. and DF: wherein R18 is selected from -aryl C (R8) 2-C (R8) 2-, -heterocycle C (R8) 2-C (R8) 2- (C3-C8), and -carbocycle C (R8) 2- C (R8) 2- (C3-C8), - and at least one of D is a compound of formula DF. 20. The antibody-drug conjugate of any of claims 5-19 wherein DF has the formula selected from the group consisting of: H 21. The antibody-drug conjugate of claim 5 wherein the antibody is selected from the group consisting of a monoclonal antibody, a bispecific antibody, a chimeric antibody, a humanized antibody, a diabody, and an antibody fragment. 22. The antibody-drug conjugate of claim 5, which has the formula: Ab-MC-ve-PAB-MMAF Ab-MC-MMAF, wherein Ab is an antibody, Val is valine, and Cit is citrulline. 23. The antibody-drug conjugate of claim 22 wherein the antibody binds to CD30, CD20, CD33, CD40 or the Lewis antigen Y. 24. A pharmaceutical composition comprising an effective amount of the conjugate of any of claims 2-23, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable diluent, carrier or excipient. 25. The pharmaceutical composition of claim 24 further comprising a therapeutically effective amount of chemotherapeutic agent selected from a tubulin-forming inhibitor, a topoisomerase inhibitor, and a / DNA linker. 26. The use of the conjugate of any of claims 2-23, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament. 27. The use of the conjugate of any of claims 2-23, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for treating a cancer. 28. The use of claim 27 for the treatment of cancer with an additional anti-cancer agent, an immunosuppressive agent or an anti-infective agent. 29. The use of the conjugate of any of claims 2-23, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for treating an autoimmune disease. 30. The use of the conjugate of any of claims 2-23, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for treating an infectious disease. 31. A method of treating cancer comprising administering to a patient an amount of the antibody-drug conjugate of any of claims 5-23, or a pharmaceutically acceptable salt or solvate thereof, said amount being effective to treat cancer, wherein the antibody of the Conjugate antibody-drug binds to an antigen expressed by a cancer cell. 32. The method of claim 31, further comprising administering an effective amount of an additional anti-cancer agent, an immunosuppressive agent or an anti-infective agent. 33. A method for treating an autoimmune disease, comprising administering to a patient an amount of the antibody-drug conjugate of any of claims 5-23, or a pharmaceutically acceptable salt or solvate thereof, the amount effective to treat the autoimmune disease, wherein the antibody-drug conjugate antibody binds to an antigen expressed by a cell of the autoimmune disease. 34. A method for treating an infectious disease, comprising administering to a patient an amount of the antibody-drug conjugate of any of claims 5-23, or a pharmaceutically acceptable salt or solvate thereof, the amount being effective to treat the infectious disease, wherein the antibody-drug conjugate antibody binds to an antigen expressed by an infectious disease cell. 35. The conjugate of claim 2 or 4, wherein the Linker unit (LU) comprises: -Aa-Ww-Yy-, where -A- is a Stretch unit, a is 0 or 1, each -W- is independently an Amino Acid unit, w is an integer that varies from 0 to 12, -Y- is a Swarming unit, and y is 0, 1 or 2. 36. The compound of claim 1 having the formula selected from the group consisting of: HN yA rAAAo or a pharmaceutically acceptable salt or solvate thereof. 37. The antibody-drug conjugate of claim 5 or 23 wherein the antibody is not an antibody that binds to an ErbB receptor or to one or more of the (l) - (35) receptors: (1) BMPR1B (Morphogenetic protein receptor of bone Type IB, Access of Genbank No. NM_001203); (2) El6 (LATÍ, SLC7A5, Genbank Access No. NM_003486); (3) STEAP1 (transmembrane epithelial antigen six of - prostate, Genbank Access No. NM_012449), - (4) 0772P (CA125, MUC16, Genbank Access No. AF361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancing factor, mesothelin, Genbank Access No. NM_005823); (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, family 34 of solute vehicle (potassium phosphate), member 2, type II sodium phosphate transporter 3b-dependent, Genbank Access No. NM__006424); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, sema domain, seven thrombospondin repeats (type 1 and similar type-1), transmembrane domain (TM) and short cytoplasmic domain, (semaforin) 5B, Genbank Access No. AB040878); (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, gene RIKEN cDNA 2700050C12, Genbank Access No. AY358628); (9) ETBR (Endothelin type B receptor, Genbank Access No. AY275463); (10) MSG783 (RNF124, hypothetical protein FLJ20315, Genbank Access No. NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, transmembrane six epithelial antigen of prostate 2, six transmembrane prostate protein , Access of Genbank No. AF455138); (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, M subfamily, member 4, Genbank Access No. NM_017636); (13) CRYPT (CR, CR1, CRGF, CRYPT, TDGF1, growth factor derived from teratocarcinum a, Genbank Accession No. NP_003203 or NM_003212); (14) CD21 (CR2 (Complementary Receiver 2) or C3DR (C3d / Epstein Barr Virus Receptor) or Hs.73792 Access Genbank No. M26004); (15) CD79b (IGb (beta-associated immunoglobulin), B29, Genbank Access No. NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein), SPAP1B, SPAP1C, Access of Genbank No. NM_030764); (17) HER2 (Access of Genbank No. M11730); (18) NCA (Genbank Accession No. M18728); (19) MDP (Genbank Accession No. BC017023); (20) IL20Ra (Access of Genbank No. AF184971); (21) Brevican (Genbank Accession No. AF229053); (22) Ephb2R (Genbank Access No. NM_004442); (23) ASLG659 (Genbank Accession No. AX092328), - (24) PSCA (Genbank Accession No. AJ297436); (25) GEDA (Access of Genbank No. AY260763); (26) BAFF-R (Genbank Accession No. NP_443177.1); (27) CD22 (Genbank Access No. NP-001762.1); (28) CD79a (CD79A, CD79a, alpha-associated immunoglobulin, Genbank Access No. NP_001774.1); (29) CXCR5 (Burkitt Lymphoma Receptor 1, Genbank Access No. NP_001707.1); (30) HLA-DOB (beta subunit of the MHC class II molecule (la antigen) that binds peptides and presents them with CD4 + T lymphocytes, Genbank Access No. NP_002111.1); (31) P2X5 (P2X ligand of purinergic receptor-ion channel 5 with input, Genbank Access No. NP_002552.2); (32) CD72 (CD72 Antigen of B cell differentiation, Lyb-2, Genbank Access No. NP_001773.1); (33) LY64 (Lymphocyte Antigen 64 (RP105), Leucine-rich Family Type I Membrane Protein (LRR), Genbank Accession No. NP_005573.1), - (34) FCRH1 (Fe Receptor Similar to protein 1, Genbank Access No. NP_443170.1); and (35) IRTA2 (Associated 2 translocation of the immunoglobulin superfamily receptor Genbank Access No. NP_112571.1). 38. A conjugate that has the formula: or a pharmaceutically acceptable salt or solvate thereof, wherein L- is a unit of Ligand; A -A- is a Stretch unit; a is 0 or 1; each -W- is independently an Amino acid unit; w is an integer that varies from 0 to 12, -each n is independently 0 or 1; p varies from 1 to about 20; and each occurrence of D is independently a drug residue selected from Formulas DE and DF: - wherein at least one of D is a drug residue of Formula DF and independently at each location: R 2 is selected from H and C 1 -C 8 alkyl; R3 is selected from H, C? -C8 alkyl, C3-C8 carbocycle, aryl, C? -C8 alkyl-aryl, C? -C8 alkyl- (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl - (C3-C8 heterocycle); R 4 is selected from H, alkyl QL-C8, carbocycle C3-C8, aryl, alkyl-aryl C? -C8 alkyl C? -8- (C3-C8 carbocycle), C3-C8 heterocycle and C? -C8 alkyl- ( C3-C8 heterocycle); R5 is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRaRb) n- wherein Ra and Rb are independently selected from H, C? -C8 alkyl and C3-C8 carbocycle and n is selected from 2, 3, 4, 5 and 6; R6 is selected from H and alkyl Ca-C8; R7 is selected from H, QL-C8 alkyl, C3-C8 carbocycle, aryl, C?-C8 alkyl-aryl, Ca-C8 alkyl- (C3-C8 carbocycle), C3-C8 heterocycle and C?-C8 alkyl- ( C3-C8 heterocycle); each R8 is independently selected from H, OH, C? -C8 alkyl / C3-C8 carbocycle and 0- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; R10 is selected from aryl or C3-C8 heterocycle; Z is O, S, NH, or NR, wherein R12 is CL-C8 alkyl; R11 is selected from H, C1-C20 alkyl / aryl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that varies from 1-1000; R13 is C2-C8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently H, COOH, - (CH2) n -N (R16) 2, - (CH2) n-S03H, or - (CH2) n-S03-C? -C? alkyl; each occurrence of R16 is independently H, alkyl Q? .- C8, or - (CH2) n -COOH; R18 is selected from aryl-C (R8) 2-C (R8) 2-, -heterocycle-C (R8) 2-C (R8) 2- (C3-C8), and carbocycle -C (R8) 2-C (R8) 2- (C3-C8); and n is an integer that varies from 0 to 6. 39. The conjugate of claim 38 wherein w is an integer ranging from 2 to 12. 40. The conjugate of claim 38 having the formula DF selected from the group consisting of: or a pharmaceutically acceptable salt or solvate thereof. 41. The conjugate of claim 38, wherein the antibody is not an antibody that binds to an ErbB receptor or one or more of the (1) - (35) receptors: (1) BMPR1B (Type IB bone morphogenetic protein receptor , Access of Genbank No. NM_001203); (2) E16 (LATÍ, SLC7A5, Genbank Access No. NM_003486); (3) STEAP1 (transmembrane epithelial antigen six of prostate, Genbank Access No. NM_012449); (4) 0772P (CA125, MUC16, Genbank Access No. AF361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancing factor, mesothelin, Genbank Access No. NM_005823); (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, family 34 of solute vehicle (potassium phosphate), member 2, type II sodium phosphate-dependent phosphate carrier 3b, Genbank No. NM_006424); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaforin 5b Hlog, sema domain, thrombospondin repeats seven (type 1 and type 1-like), transmembrane domain (TM) and short cytoplasmic domain, (semaforin) 5B, Genbank Access No. AB040878); (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genbank Access No. AY358628); (9) ETBR (Endothelin type B receptor, Genbank Access No. AY275463); (10) MSG783 (RNF124, hypothetical protein FLJ20315, Genbank Access No. NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, transmembrane six epithelial antigen of prostate 2, six transmembrane prostate protein , Genbank Accession No. AF455138), - (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, M subfamily, member 4, Genbank Access No. NM_017636); (13) CRYPT (CR, CR1, CRGF, CRYPT, TDGF1, growth factor derived from teratocarcinoma, Genbank Access No. NP_003203 or NM_003212); (14) CD21 (CR2 (Complementary Receiver 2) or C3DR (C3d / Epstein Barr Virus Receptor) or Hs.73792 Access Genbank No. M26004); (15) CD79b (IGb (beta-associated immunoglobulin), B29, Access of Genbank No. NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchor protein), SPAP1B, SPAP1C, Access of Genbank No. NM_030764); (17) HER2 (Access of Genbank No. M11730), - (18) NCA (Access of Genbank No. M18728), - (19) MDP (Access of Genbank No. BC017023); (20) IL20Ra (Access of Genbank No. AF184971); (21) Brevican (Genbank Accession No. AF229053); (22) Ephb2R (Genbank Access No. NM_004442), - (23) ASLG659 (Genbank Accession No. AX092328); (24) PSCA (Access of Genbank No. AJ297436); (25) GEDA (Access of Genbank No. AY260763); (26) BAFF-R (Genbank Accession No. NP_443177.1); (27) CD22 (Genbank Access No. NP-001762.1); (28) CD79a (CD79A, CD79a, alpha-associated immunoglobulin, Genbank Access No. NP_001774.1); (29) CXCR5 (Burkitt Lymphoma Receptor 1, Genbank Access No. NP_001707.1); (30) HLA-DOB (beta subunit of the MHC class II molecule (la antigen) that binds peptides and presents them with CD4 + T lymphocytes, Genbank Access No. NP_002111.1); (31) P2X5 (P2X ligand of the purinergic receptor-channel 5 of the ion with entry, Genbank Access No. NP_002552.2), - (32) CD72 (CD72 Antigen of B cell differentiation, Lyb-2, Access of Genbank No NP_001773.1); (33) LY64 (Lymphocyte Antigen 64 (RP105), Leucine-rich Family Type I Membrane Protein (LRR), Genbank Accession No. NP_005573.1); (34) FCRH1 (Fe receptor similar to protein 1, Access - of Genbank No. NP_443170.1), - and (35) IRTA2 (Translocation of the associated immunoglobulin superfamily receptor Genbank Access No. NP_112571.1). 42. The conjugate or compound or a pharmaceutically acceptable salt or solvate of the conjugate or compound thereof of any of claims 1-23 and 35-41 which is in an isolated and purified form. 43. An antibody-drug conjugate comprising an antibody covalently linked to one or more drug residues, the antibody-drug conjugate having the Formula Is: or a pharmaceutically acceptable salt or solvate thereof, wherein: Ab is an antibody that binds to one or more antigens associated with the tumor (1) - (35): (1) BMPR1B (bone morphogenetic protein receptor Type IB, Access of Genbank No. NM__001203); (2) El6 (LATÍ, SLC7A5, Genbank Access No. NM_003486); (3) STEAP1 (transmembrane epithelial antigen six of prostate, Genbank Access No. NM_012449); (4) 0772P (CA125, MUC16, Genbank Access No. AF361486); (5) MPF (MPF, MSLN, SMR, megakaryocyte enhancing factor, mesothelin, Genbank Access No. NM_005823); (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, family 34 of solute vehicle (potassium phosphate), member 2, sodium phosphate-dependent phosphate type 3b transporter, Genbank Access No. NM_006424); (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaforin 5b Hlog, sema domain, thrombospondin repeats seven (type 1 and type 1-like), transmembrane domain (TM) and short cytoplasmic domain, (semaforin) 5B, Genbank Access No. AB040878); (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genbank Access No. AY358628); (9) ETBR (Endothelin type B receptor, Genbank Access No. AY275463); (10) MSG783 (RNF124, hypothetical protein FLJ20315, Genbank Access No. NM_017763); (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, gene 1 associated with prostate cancer, protein 1 associated with prostate cancer, transmembrane six epithelial antigen of prostate 2, six transmembrane prostate protein , Access of Genbank No. AF455138); (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, M subfamily, - member 4, Genbank Access No. NM_017636); (13) CRYPT (CR, CR1, CRGF, CRYPT, TDGF1, growth factor derived from teratocarcinoma, Genbank Access No. NP_003203 or NM_003212); (14) CD21 (CR2 (Complementary Receiver 2) or C3DR (C3d / Epstein Barr Virus Receiver) or Hs.73792 Access from the Genbank No. M26004); (15) CD79b (IGb (beta-associated immunoglobulin), B29, Genbank Access No. NM_000626); (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing the phosphatase anchor protein), SPAP1B, SPAP1C, Genbank Access No. NM_030764); (17) HER2 (Access of Genbank No. M11730); (18) NCA (Genbank Accession No. M18728); (19) MDP (Genbank Accession No. BC017023); (20) IL20Ra (Genbank Accession No. AF184971), - (21) Brevican (Access of Genbank No. AF229053); (22) Ephb2R (Genbank Access No. NM_004442), - (23) ASLG659 (Genbank Accession No. AX092328); (24) PSCA (Access of Genbank No. AJ297436); (25) GEDA (Access of Genbank No. AY260763); (26) BAFF-R (Genbank Accession No. NP_443177.1), - (27) CD22 (Genbank Accession No. NP-001762.1); (28) CD79a (CD79A, CD79a, alpha-associated immunoglobulin, Genbank Access No. NP 001774.1); - (29) CXCR5 (Burkitt Lymphoma Receptor 1, Genbank Access No. NP_001707.1); (30) HLA-DOB (beta subunit of the MHC class II molecule (la antigen) that binds peptides and presents them with CD4 + T lymphocytes, Genbank Access No. NP_002111.1); (31) P2X5 (P2X ligand of the purinergic receptor-channel 5 of the ion with entry, Genbank Access No. NP_002552.2), - (32) CD72 (CD72 Antigen of B cell differentiation, Lyb-2, Access of Genbank No NP_001773.1); (33) LY64 (Lymphocyte Antigen 64 (RP105), Leucine-rich Family Type I Membrane Protein (LRR), Genbank Accession No. NP_005573.1); (34) FCRH1 (Fe receptor similar to protein 1, Genbank Access No. NP_443170.1); and (35) IRTA2 (Associated 2 translocation receptor for the immunoglobulin superfamily Genbank Accession No. NP_112571.1), A is a Stretch unit, a is 0 or 1, each W is independently an Amino Acid unit, w 'is an integer that varies from 0 to 12, Y is a Espandora unit, yy is 0, 1 or 2, p varies from 1 to 20, and D is a drug residue selected from the group that - - consists of the DE formulas. and DF: wherein the sinuous line of DE and DF indicates the site of covalent attachment to A, W, or Y, and independently at each location: R2 is selected from H and C? -C8 alkyl; R3 is selected from H, C?-C8 alkyl, C3-C8 carbocycle, aryl, C?-C8 alkyl-aryl / C?-C8 alkyl- (C3-C8 carbocycle), C3-C8 heterocycle and C?-C8 alkyl - (C3-C8 heterocycle); R 4 is selected from H, CL-C8 alkyl, C3-C8 carbocycle, aryl, C?-C8 alkyl-aryl, C?-C8 alkyl- (C3-C8 carbocycle), C3-C8 heterocycle and C?-C8 alkyl (C3-C8 heterocycle); R5 is selected from H and methyl; or R4 and R5 together form a carbocyclic ring and have the formula - (CRRb) n- wherein Ra and Rb are independently selected from H, C? -C8 alkyl and C3-C8 carbocycle and n is selected from 2, 3, 4, 5 and 6; R6 is selected from H and C? -C8 alkyl; R7 is selected from H, alkyl CL-C8, carbocycle C3-C8, aryl, alkyl-aryl C -Ca, C alquilo-C8 alkyl- (C3-C8 carbocycle), C3-C8 heterocycle and C?-C8 alkyl- ( C3-C8 heterocycle); each R8 is independently selected from H, OH, C? -C8 alkyl, C3-C8 carbocycle and 0- (C? -C8 alkyl); R9 is selected from H and C? -C8 alkyl; and R10 is selected from aryl or C3-C8 heterocycle; Z is 0, S, NH, or NR12, wherein R12 is C? -C8 alkyl; R11 is selected from H, C1-C20 alkyl, aryl, C3-C8 heterocycle, - (R130) m-R14, or - (R130) m-CH (R15) 2; m is an integer that varies from 1-1000; R13 is C2-C8 alkyl; R14 is H or C? -C8 alkyl; each occurrence of R15 is independently H, COOH, - (CH2) n-N (R16) 2, - (CH2) n-S03H, or - (CH2) n-S03-C-L-C8 alkyl; each occurrence of R16 is independently H, C? -C8 alkyl, or - (CH2) n-C00H; R18 is selected from aryl-C (R8) 2-C (R8) 2-, -C (R8) 2-C (R8) 2- (C3-C8 heterocycle), and -C (R8) 2-C (R8) ) 2- (C3-C8 carbocycle); and n is an integer that varies from 0 to 6. 44. The antibody-drug conjugate of claim 43 wherein D is Formula DE: 45. The antibody-drug conjugate of claim 43, wherein D is Formula DF: 46. The antibody-drug conjugate of claim 43, wherein the antibody binds to the drug residue through a cysteine residue of the antibody. 47. The antibody-drug conjugate of claim 46, wherein p is 1 to 4. 48. The antibody-drug conjugate of claim 43 wherein p is 2 to 8. 49. The antibody-drug conjugate of claim 43 wherein p is 2 to 5. 50. The antibody-drug conjugate of claim 46 having the formula: 51. The antibody-drug conjugate of claim 50 having the formula: where w and y are each 0. 52. The antibody-drug conjugate of claim 51 wherein D is Formula DE. 53. The antibody-drug conjugate of claim 52 wherein Formula DE has the formula: 54. The antibody-drug conjugate of claim 51 wherein D is Formula DF. 55. The antibody-drug conjugate of claim 54 wherein Formula DF has the formula: 56. The antibody-drug conjugate of claim 43 having the formula: 57. The antibody-drug conjugate of claim 56 having the formula: 58. The antibody-drug conjugate of claim 57 having the formula: 59. The antibody-drug conjugate of claim 58 wherein D is the Formula DE. 60. The antibody-drug conjugate of claim 59 wherein Formula DE has the formula: 61. The antibody-drug conjugate of claim 57 wherein D is Formula DF 62. The antibody-drug conjugate of claim 61 wherein Formula DF has the formula: 63. The antibody-drug conjugate of claim 43 having the formula: 64. The antibody-drug conjugate of claim 43 wherein w is an integer ranging from 2 to 12. 65. The antibody-drug conjugate of claim 43 wherein w is 2. 66. The antibody-drug conjugate of claim 45 wherein Ww is -valin-citrulline. 67. The antibody-drug conjugate of claim 43 wherein D is selected from the group consisting of the Formulas: 68. The antibody-drug conjugate of claim 43 wherein the antibody specifically binds to a HER2 receptor. 69 The antibody-drug conjugate of claim 68 which specifically binds to the extracellular domain of the HER2 receptor and inhibits the growth of tumor cells that overexpress the HER2 receptor. 70. The antibody-drug conjugate of claim 43 wherein the antibody is selected from the group consisting of a monoclonal antibody, a bispecific antibody, a chimeric antibody, a humanized antibody, and an antibody fragment. 71. The antibody-drug conjugate of claim 70 wherein the antibody fragment is a Fab fragment. 72. The antibody-drug conjugate of claim 70 wherein the humanized antibody is selected from the group consisting of huMAb4D5-l, huMAb4D5-2, huMAb4D5-3, huMAb4D5-4, huMAb4D5-5, huMAb4D5-6, huMAb4D5-7 and huMAb4D5 -8 (trastuzumab). 73. The antibody-drug conjugate of claim 72 wherein the antibody is huMAb4D5-8 (trastuzumab). 74. The antibody-drug conjugate of claim 43 selected from the group consisting of the Formulas: A -MC- c-PAB -MMAF; Ab-MC-vc-PAB-MMAE, - A -MC-MMAE; Y A -MC-MMAF, where Val is valine, and Cit is citrulline. 75 The anti-body-drug conjugate of claim 74 wherein the antibody is huMAb4D5-8 (trastuzumab). 76. A pharmaceutical composition comprising an effective amount of the antibody-drug conjugate of any of claims 43-75, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable diluent, carrier or excipient. 77. The pharmaceutical composition of claim 76 further comprising a therapeutically effective amount of chemotherapeutic agent selected from a tubulin-forming inhibitor, a topoisomerase inhibitor, and a 7DNA linker. 78. A method for eliminating or inhibiting the proliferation of tumor cells or cancer cells comprising treating the tumor cells or cancer cells with an amount of the antibody-drug conjugate of any of claims 43-75, said amount being effective to eliminate or inhibit the proliferation of tumor cells or cancer cells. 79. A method of treating cancer comprising administering to a patient with a hyperproliferative disorder an amount of the antibody-drug conjugate of any of claims 43-75, said amount being effective for treating the hyperproliferative disorder. 80. The method of claim 79 wherein the hyperproliferative disorder is cancer selected from the group consisting of breast, ovarian, stomach, endothelial, salivary gland, lung, kidney, colon, colorectal, thyroid, pancreatic, prostate and bladder cancers. 81. The method of claim 79 further comprising administering an effective amount of an additional agent selected from the group consisting of an anti-cancer agent, an immunosuppressive agent, and an anti-infective agent. 82. A method for treating an autoimmune disease, comprising administering to a patient an amount of the antibody-drug conjugate of any of claims 43-75, said amount being effective for treating an autoimmune disease. 83. A method for treating an infectious disease, comprising administering to a patient an amount of the antibody-drug conjugate of any of claims 43-75, said amount being effective for treating an infectious disease. 84. A method for inhibiting cell proliferation comprising: exposing mammalian cells in a cell culture medium to the antibody-drug conjugate of any of claims 43-75, and measuring the cytotoxic activity of the antibody-drug conjugate, whereby inhibits cell proliferation. 85. A method of treating cancer comprising administering to a patient a formulation of an antibody-drug conjugate of any of claims 43-75 and a pharmaceutically acceptable diluent, carrier or excipient. 86. The method of claim 85 wherein the antibody-drug conjugate specifically binds to a receptor encoded by an ErbB2 gene. 87. The method of claim 85 wherein the amount of the antibody-drug conjugate administered to the patient is in the range of about 0.1 to about 10 mg / kg of the patient's weight. 88. The method of claim 85 wherein the antibody-drug conjugate is administered at three week intervals. 89. The method of claim 85 wherein said antibody-drug conjugate is formulated with a pharmaceutically acceptable parenteral vehicle and administered parenterally. 90. The method of claim 85 wherein said antibody-drug conjugate is formulated in a unit dose injectable form. 91. The method of claim 85 wherein said antibody-drug conjugate is administered intravenously. 92. The method of claim 85 further comprising administering a second antibody that binds to a tumor associated antigen selected from (D- (35). 93. The method of claim 92 wherein said second antibody is conjugated with a cytotoxic agent. 94. A method for inhibiting the growth of tumor cells overexpressing a tumor-associated antigen comprising administering to a patient an antibody-drug conjugate of any of claims 43-75 that specifically binds said tumor-associated antigen, and to an agent chemotherapeutic wherein said antibody-drug conjugate and said chemotherapeutic agent are each administered in effective amounts to inhibit the growth of the tumor cells in the patient. 95. The method of claim 94 wherein said antibody-drug conjugate sensitizes the tumor cells to said chemotherapeutic agent. 96. A method for the treatment of a human patient susceptible or diagnosed with a disorder characterized by overexpression of the ErbB2 receptor, which comprises administering an effective amount of a combination of an antibody-drug conjugate of any of claims 43-75, and to a chemotherapeutic agent . 97. An assay for detecting cancer cells comprising: exposing the cells to an antibody-drug conjugate of any of claims 43-75, and determining the degree of binding of the antibody-drug conjugate to the cells. 98. The assay of claim 97 wherein the cells are breast tumor cells. 99. The assay of claim 97 wherein the degree of binding is determined by measuring the levels of the nucleic acid encoding the tumor-associated antigen by fluorescence hybridization in si tu (FISH). 100. The assay of claim 97 wherein the degree of binding is determined by immunohistochemistry (IHC). 101. An article of manufacture comprising: an antibody-drug conjugate of any of claims 43-75; a container; and a packaging insert or label indicating that the antibody-drug conjugate can be used to treat cancer. 102. The article of manufacture of the re-excitation 101 wherein said insert or packaging label indicates that the antibody-drug conjugate can be used to treat cancer characterized by overexpression of an ErbB2 receptor. 103. The article of manufacture of the reification 101 where cancer is breast cancer. 104. A method for the treatment of cancer in a mammal, wherein the cancer is characterized by overexpressing a receptor ErbB2 (HER2) and does not respond, or responds poorly, to treatment with an anti-ErbB2 antibody, which comprises administering to the mammal a Therapeutically effective amount of an antibody-drug conjugate of any of claims 43-75. 105. The method of claim 104 wherein the mammal is human. 106. The use of an antibody-drug conjugate of any of claims 43-75, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament. 107. The use of an antibody-drug conjugate of any of claims 43-75, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for eliminating or inhibiting the proliferation of tumor cells or cancer cells. 108. The use of an antibody-drug conjugate of any of claims 43-75, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for treating a hyperproliferative disorder. 109. The use of claim 108 wherein the hyperproliferative disorder is cancer selected from the group consisting of breast, ovarian, stomach, endothelial, salivary gland, lung, kidney, colon, colorectal, thyroid, pancreatic, prostate and bladder cancers. 110. The use of claim 108 wherein the medicament further comprises an additional agent selected from an anti-cancer agent, an immunosuppressive agent, and an anti-infective agent. 111. The use of an antibody-drug conjugate of any of claims 43-75, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for treating an autoimmune disease. 112. The use of an antibody-drug conjugate of any of claims 43-75, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for treating an infectious disease.
MXPA/A/2006/005083A 2003-11-06 2006-05-04 Monomethylvaline compounds capable of conjugationto ligands MXPA06005083A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60/518,534 2003-11-06
US60/557,116 2004-03-26
US60/598,899 2004-08-04
US60/622,455 2004-10-27

Publications (1)

Publication Number Publication Date
MXPA06005083A true MXPA06005083A (en) 2006-12-13

Family

ID=

Similar Documents

Publication Publication Date Title
US10808039B2 (en) Monomethylvaline compounds capable of conjugation to ligands
MXPA06005083A (en) Monomethylvaline compounds capable of conjugationto ligands