|
Water-soluble analogues cc-1065 and their conjugates |
|||||||||||||||||||||||||||||||||||||||||||||||||||||||
IPC classes for russian patent Water-soluble analogues cc-1065 and their conjugates (RU 2489423):
New sulphated oligosaccharide derivatives / 2483074
Present invention refers to new compounds of general formula I [X]n-Y-ZR1R2, wherein the radicals are specified in the description, effective as heparan sulphate-binding protein inhibitors. The invention also refers to a pharmaceutical or veterinary composition having heparan sulphate-binding protein inhibitory activity for preventing or treating a disorder in a mammal, and to the use of these compounds and compositions for antiangiogenic, antimetastatic, anti-inflammatory, antimicrobial, anticoagulant and/or antithrombotic therapy in a mammal.
Monosebacate of pyrazole derivative / 2482122
Present invention provides a novel form of of 3-(3-{4-[3-(b-D-glucopyranosyloxy)-5-isopropyl-1H-pyrazol-4-ylmethyl]-3-methylphenoxy}propylamino)-2,2-dimethylpropionamide (chem. 1) with improved storage stability. Since bis[3-(3-{4-[3-(b-D-glucopyranosyloxy)-5-isopropyl-1H-pyrazol-4-ylmethyl]-3-methylphenoxy}propylamino)-2,2-dimethylpropionamide]monosebacate has extremely excellent storage stability, it can be used as a medicinal substance. Furthermore, it shows extremely good crystalline properties and can be purified by a conventional method, and therefore is suitable for the industrial preparation.
Novel compounds derived from 5-thioxylose and therapeutic application thereof / 2412195
Invention relates to novel derivatives of 5-thioxylose of formula I
Novel fluoroglycoside derivatives of pyrazoles, medicine containing them, and application of derivatives / 2370499
Invention concerns compounds of formula I, where R1, R4, R5, R6, R7, R8 is H; R2 is F; A is O; R3 is (C1-C6)-alkyl, and one, several or all hydrogen atoms can be substituted by fluorine; B is (C0-C15)-alkylene; X is CO or link; L is (C1-C6)-alkylene, where in each case one or two -CH2-groups can be substituted by NH, or (C2-C5)-alkenylene; Y is CO, NHCO or link; R9 is hydrogen, (C1-C6)-alkyl, (C1-C6)-alkylene-OH, (C1-C6)-alkylene-NH2 or (C0-C6)-alkylene-CONH2, where one or more CH2-groups of alkyl residue can be substituted by -OH, -OSO3H, -NH-SO3H, adamantly or sugar residue of formula , or R8 and R9 together with their carrier atom N form 5-7-membered saturated cycle Cyc2, where one or more CH2-groups of the cycle can also be substituted by O, NH, NSO3H, N-(C1-C6)-alkyl, and alkyl group can be substituted by -OH group; and pharmaceutically acceptable salts of the claimed compounds.
Pyrazol derivatives, medical compositions, containing them, their application in medicine and intermediate compounds for their production / 2369613
Invention relates to pyrazol derivatives, represented by general formula: , where R1 - R6 take on values, determined in the invention formula; one of Q and T is a group, presented by the general formula: or a group, presented by the general formula: , whereas the other one represents C1-6 alkyl group, halogen (C1-6 alkyl) group or C3-7 cycloalkyl group; X, Y and Z take on values, determined in the invention formula; to their pharmaceutically acceptable salts that possess inhibitory activity in regard to SGLT1 human being.
Pyrazole derivatieves, medical compositions, containing said derivatives, their application in medicine and intermediate compounds for their obtaining / 2356906
Claimed invention relates to pyrazole derivatives, which are represented by general formula (I), as well as theirpharmacologically acceptable salts, which have inhibiting activity against human SGLT1, to pharmacological composition, inhibitor of human SGLT1 and based on them medications, to their application for producing pharmacologic composition and to intermediate compounds for their obtaining. where R1 represents H, hydroxy(C2-6)alkyl group, one of Q and T represents group, which is presented by general formula: or group, which is presented by general formula while another presents C1-6alkyl group; R2 represents hydrogen atom, C1-6alkyl group or group of formula: -A-R8, where A represents oxygen atom, and R8 represents C6hetherocycloalkyl group, containing oxygen atom as heteroatom; X represents simple bond or oxygen atom, Y represents C1-6alkylene group or C2-6alkylene group; Z represents carbonyl group or sulphonyl group; R4 and R5 are similar or different, and each represents hydrogen atom or C1-6alkyl group, which can have similar or different 1-3 substituents, selected from substituents (i) Values of sunstituents (i) are iven in invention formula.
New heterocyclic fluorglucoside derivatives, medications containing these compounds, and their application / 2339641
Invention concerns substituted heterocyclic fluorglucoside derivatives of the formula (I) , where R1 and R2 are independently F, H, or one of R1 or R2 residues is OH; R3 is OH or F, so that at least one of R1, R2, R3 residues is F; R4 is OH; A is O; X is C or N, so that if Y = S, X should be C; Y is N or S; m = 1; R5 is hydrogen, OH or (C1-C6)-alkyl optionally mono- or polysubstituted by fluorine; R6 is H or (C1-C6)-alkyl, if required; or, if Y = S, R5 and R6 form phenyl together with carbon atoms to which they are linked; B is (C1-C6)-alcandiyl or -CO-NH-CH2-; n = 2 or 3; Cyc1 is phenyl or thiophenyl; R7 is hydrogen, F, CI, Br, J, (C1-C6)-alkyl or (C1-C6)-alkoxy optionally mono- or polysubstituted by fluorine; R8 is hydrogen or halogen; R9 is hydrogen; or R8 and R9 together mean -CH=CH-CH=CH-, -CH=CH-C((C1-C6)-alkoxy)=CH- or -OH=CH-O- and together with carbon atoms to which they are linked form Cyc2, which is phenyl, optionally substituted (C1-C6)-alkoxy or furanyl respectively; and their pharmaceutically acceptable salts.
Derivatives of glucopyranosyloxypyrazole and their using in medicinal agents / 2317302
Invention relates to novel derivatives of glucopyranosyloxypyrazole of the general formula: wherein R means hydrogen atom, lower alkyl, acyl, alkoxycarbonyl, acyloxymethyl or alkoxycarbonyloxymethyl; one of Q and T represents a group of the general formula: wherein P means hydrogen atom, lower acyl or alkoxycarbonyl but other radical means lower alkyl or halogen(lower alkyl); R2 means hydrogen atom, lower alkyl, alkoxy-, alkylthio-group, halogen(lower)alkyl) or halogen atom under condition that P doesn't represent hydrogen atom when R means hydrogen atom or lower alkyl. Also, invention relates to pharmaceutically acceptable salts of these compounds, pharmaceutical compositions used for prophylaxis of renal glucose re-absorption and eliciting the inhibitory activity with respect to human SGLT2, pharmaceutical combinations that are used for prophylaxis or treatment of diseases associated with hyperglycemia.
Glucopyranosyloxy derivatives, pharmaceutical compositions containing these derivatives and intermediate compounds for their production / 2232767
The invention relates to glucopyranosyloxy derivative of the formula (I), where R1represents a hydrogen atom or a lower alkyl group; one of Q1and T1represents a group of formula (II), while the other of them represents a lower alkyl group or a halo(lower alkyl) group; R2represents a hydrogen atom, a lower alkyl group, lower alkoxygroup, lower allylthiourea, halogen(lower alkyl) group or a halogen atom, or its pharmaceutically acceptable salts
-substituted derivatives of carboxylic acids" target="_blank">-substituted derivatives of carboxylic acids" align="left" vspace="30" hspace="30" /> -substituted derivatives of carboxylic acids / 2219172
The invention relates tosubstituted derivatives of carboxylic acids, characterized by the General formula (I), (II), (III) and (IV)
< / BR> < / BR> < / BR> < / BR> or their pharmacologically acceptable (C1-C6)-alkyl esters, or their pharmacologically acceptable Amida, or their pharmacologically acceptable salts
5-substituted indazole as kinase inhibitors / 2487873
Present invention refers to compounds of formula (I) or pharmaceutically acceptable salts thereof wherein A, R1, R2, R3 and m are specified in the patent claim. The present invention also refers to the number of specific compounds, and to a pharmaceutical composition containing the above compounds effective for inhibition of kinases, such as glycogen synthase kinase 3 (GSK-3), Rho kinase (ROCK), Janus kinase (JAK), AKT, PAK4, PLK, CK2, KDR, MK2, JNK1, aurora, pim 1 and nek 2.
3-(1h-indol-3-yl)-4-[2-(4-methylpiperazin-1-yl)quinazolin-4-yl]pyrrole-2,5-dione salts / 2487128
Invention relates to 3-(1H-indol-3-yl)-4-[2-(4-methylpiperazin-1-yl)quinazolin-4-yl]pyrrole-2,5-dione salts in crystalline form, where said salts are formed with an acid selected from hydrochloric acid, maleic acid, malonic acid and methanesulphonic acid. The invention also relates to a method of producing said salts, a pharmaceutical composition containing said salts and a method of treating diseases or disorders mediated by T lymphocytes and/or PKC, which involves addition of said salts.
Chiral cis-imidazolines / 2487127
Described are novel chiral cis-imidazolines selected from a group which includes 2-{4-[(4S,5R)-2-(2-tert-butyl-4-ethoxypyrimidin-5-yl)-4,5-bis-(4-chlorophenyl)-4,5-dimethyl-4,5-dihydroimidazole-1-carbonyl]-piperazin-1-yl}-acetamide, [(4S,5R)-2-(2-tert-butyl-4-ethoxypyrimidin-5-yl)-4,5-bis-(4-chlorophenyl)-4,5-dimethyl-4,5-dihydroimidazol-1-yl]-[4-(1,1-dioxohexahydrothiopyran-4-yl)-piperazin-1-yl]-methanone, [(4S,5R)-2-(2-tert-butyl-4-ethoxypyrimidin-5-yl)-4,5-bis-(4-chlorophenyl)-4,5-dimethyl-4,5-dihydroimidazol-1-yl]-[4-(3-methanesulphonylpropyl)-piperazin-1-yl]-methanone, 2-{4-[(4S,5R)-2-(6-tert-butyl-4-ethoxypyridin-3-yl)-4,5-bis-(4-chlorophenyl)-4,5-dimethyl-4,5-dihydroimidazole-1-carbonyl]-piperazin-1-yl}-N,N-bis-(2-methoxyethyl)-acetamide. 2-{1-[(48;5K)-2-(6-tert-butyl-4-ethoxypyridin-3-yl)-4,5-bis-(4-chlorophenyl)-4,5-dimethyl-4,5-dihydroimidazole-1-carbonyl]-piperidin-4-yl}-acetamide and others described by the general structural formula (I), and pharmaceutical composition containing said compounds.
Pyrimidyl cyclopentanes as akt/protein kinase inhibitors / 2486181
Invention refers to new compounds of formula I, enantiomers and pharmaceutically acceptable salts thereof having the properties of AKT/protein kinase inhibitors. In formula 1 G represents phenyl, naphthalene, 5-member heteroaryl with 1 sulphur atom in a ring or 9-member bicyclic heteroaryl specified in indolyl wherein phenyl, naphthalene, 5-member heteroaryl is optionally substituted by one of three Ra groups; R1 and R1a are independently specified in H, Me, Et, -CH2OH, CF3, CHF2 or CH2F; R2 represents H, -OH, -OMe or F; R2a representsH, Me or F; R3 represents H, Me, Et; R4 represents H, 6-member heterocyclyl containing an oxygen atom as a heteroatom, cyclopropyl methyl or C1-C4 alkyl is optionally substituted F,-OH or -O(C1-C3 alkyl); R5 and R5a are independently specified in H and C1-C4 alkyl, or R5 and R5a together with an atom whereto attached form a carbonyl group or 5-6-member cycloalkyl; each Ra independently represents halogen, C1-C6-alkyl, C3-C6-cycloalkyl,-O-(C1-C6-alkyl), CF3, CN, phenyl, pyrazole, CH2F, CHF2, -OCH2F, -OCHF2, -OH, -SO2(C1-C6-alkyl), C(O)NH2; and j represents 1 or 2; and provided j represents 2, j-ring carbon atom, opposite NR4, may be substituted by heteroatom O.
Pyrimidyl cyclopentanes as akt-protein kinase inhibitors / 2486178
Present invention refers to new compounds of formula I, enantiomers and pharmaceutically acceptable salts thereof which have selective inhibitory action on AKT protein kinase, in particular protein kinase B. In formula I: A represents R1 and R1a are independently specified in H, Me, Et. vinyl, CF3, CHF2 or CH2F:R2 represents H, OH, OMe or F; R2a represents H, Me or F; R3 represents H. Me. Et or CF3; G represents phenyl optionally substituted by one to four groups Rc, or 5-6-member heteroaryl containing one heteroatom specified in sulphur optionally substituted by halogen; R5 and R6 independently represent H, OCH3, C3-C6-cycloalkyl independently substituted by F, OH, C1-C3alkyl or O(C1-C3alkyl), 4-6-member heterocyclyl containing one heteroatom specified in nitrogen optionally substituted by F, OH, C1-C3alkyl, cyclopropylmethyl or -C(=O)(C1-C3alkyl), or C1-C6-alkyl optionally substituted by one or more groups independently specified in OH, oxo O(C1-C6-alkyl), CN, F, NH2. NH(C1-C6-alkyl), O(C1-C6-alkyl)2. cyclopropyl. phenyl, imidazolyl, piperidinyl, pyrrolidinyl, morpholinyl, tetrahydrofuranyl, oxetanil or tetrahydropyranyl. The other radical values are specified in the patent claim.
Method of producing substituted pyrimidin-5-yl carboxylic acids / 2485083
Invention relates to a method of producing substituted pyrimidin-5-yl carboxylic acids of formula I and can be used in organic chemistry. The method is realised by reacting N-substituted guanidines and hetarylamidines with ethoxymethylene derivatives of 1,3-ketoesters according to a scheme given below (where the substitutes are as defined in the claim).
Method of producing benzimidazoles / 2482118
Invention relates to basic organic synthesis and specifically to a method of producing N,N'-bis and N,N,N,N-tetrakis-benzimidazolylmethyl N,N'-bis (piperazinoethyl) ethylenediamines, N,N'-bis and N,N,N,N-tetrakis-benzimidazolylethyl-N,N'-bis (piperazinoethyl) ethylenediamines, which are obtained by reacting carboxylic acids: N,N'-bis and N,N,N,N-tetrakis-carboxyethyl and N,N'-bis and N,N,N,N-tetrakis-carboxyethyl N,N'-bis (piperazinoethyl) ethylenediamine with ortho-phenylenediamine while heating first at temperature of 100-125°C for 0.6-1.5 hours and then at 130-150°C for 2.5-3.4 hours in the medium of an aromatic organic solvent with molar ratio hexamino diacid: phenylenediamine=1:2.1-2.15 and hexamino tetracid:phenylenediamine=1:4.1-4.15. The end product is separated by distilling the reaction mass. The aromatic organic solvent used is ortho-, meta- and para-xylenes or a mixture of ortho-, meta- and para-xylenes.
Crystalline modifications of 3-(1h-indol-3-yl)-4-(4-methylpiperazin-1-yl)quinazolin-4-yl)pyrrol-2,5-dione / 2481341
Present invention refers to novel crystalline forms of 3-(1H-indol-3-yl)-4-[2-(4-methylpiperazin-1-yl)quinazolin-4-yl]pyrrol-2,5-dione acetate salt, their use for the treating the diseases mediated by T-lymphocytes or PKC, to a pharmaceutical compositions thereof, and a method for preparing them. The presented crystalline forms have: a strong diffraction peak at the angle of 2θ making 21.5° for the A form, or a strong diffraction peak at the angle of 2θ making 9.7° for the B form. The mentioned crystalline forms can be prepared by dissolving 3-(1H-indol-3-yl)-4-[2-(4-methylpiperazin-1-yl)quinazolin-4-yl]pyrrol-2,5-dione in 2-propanol at higher temperature, and then cooling after salt formation for preparing the A form, or dissolving 3-(1H-indol-3-yl)-4-[2-(4-methylpiperazin-1-yl)quinazolin-4-yl]pyrrol-2,5-dione in ethyl acetate at higher temperature and then cooling after salt formation for preparing the B form.
Cyclopenta(d)pyrimidines as protein kinase akt inhibitors / 2481336
Invention refers to compounds of formula I , as well as to enantiomers and salts thereof, wherein R1 and R1a are independently specified in H, Me, Et, CH=CH2, CH2OH, CF3, CHF2 or CH2F; R2 and R2a are independently specified in H or F; R5 represents H, Me, Et or CF3; A is presented by formula: ; wherein G, R6, R7, Ra, Rb ,Rc, Rd, R8, m, n and p are presented in cl. 1 of the patent claim.
Phenylpyrazol derivatives / 2480456
Invention refers to a phenylpyrazol derivative presented by formula (1) or to its pharmaceutically acceptable salt: wherein R1 and R2, which may be identical or different, each represents C1-C6 alkyl, or R1 and R2 are coupled together with an adjacent nitrogen atom to form a 5-6-merous saturated heterocylic ring (wherein the mentioned saturated heterocylic ring may be substituted by halogen or C1-C6 alkyl), n represents an integer 0 to 2, T represents a hydrogen atom, halogen or C1-C6 alkyl, and R has one of formulas (I)-(V), (VII) or (VIII):
Bicyclosubstituted pyrazolon azo derivatives, method for production and pharmaceutical use thereof / 2488582
Invention relates to bicyclosubstituted pyrazolon azo derivatives of formula
|
FIELD: biotechnologies. SUBSTANCE: invention relates to new compounds of the formula I: or its pharmaceutically acceptable salts, where values R1, R2, R3, R3', R4, R4', X2, X1, X3, X4, X5, R6, R7, R5', R6', R7', R14', R8, R9, R10, R11a, b, c are specified in the clause 1 of the formula. EFFECT: compounds demonstrate antitumoral activity and may be used to produce a pharmaceutical composition for tumor treatment in a mammal. 14 cl, 18 dwg, 1 tbl, 52 ex
The SCOPE of the PRESENT INVENTION The present invention relates to novel analogs of the DNA-binding alkylating agent CC-1065 and their conjugates. In addition, the present invention relates to intermediate compounds for obtaining these agents and their conjugates. These conjugates are designed to highlight their (many times) useful agents after one or more of the stages of activation and/or the velocity and period of time controlled by the conjugate for selective delivery and/or managed the selection of one or more of these DNA alkylating agents. These agents, conjugates and intermediate compounds can be used to treat diseases which are characterized by undesirable (cell) proliferation. As an example, these agents and these conjugates of the present invention can be used to treat tumors. BACKGROUND of the PRESENT INVENTION Duocarmycin, first isolated from the culture broth of Streptomyces species, are the ancestors of the members of the family of antitumor antibiotics, which also include CC-1065. These are extremely effective agents, as indicated, their biological activity is the ability to consistently-selective alkylation of DNA by N3 of the adenine in the minor grooves that mandatory UN paid which induces a cascade of events, which ends in the death mechanism of apoptotic cells1. Although CC-1065 demonstrate very effective cytotoxicity, they cannot be used in hospitals due to serious delayed hepatotoxicity.2These observations have led to the development of synthetic analogues of CC-1065 (See. CC-1065 derivatives, for example, Aristoff et al., J. Org. Chan. 1992, 57, 6234; Boger et al., Bioorg. Med. Chem. Lett. 1996, 6, 2207; Boger et al., Chem. Rev. 1997, 97, 787; Milbank et al., J. Med. Chem. 1999, 42, 649; Atwell et al., J. Med. Chem. 1999, 42, 3400; Wang et al., J. Med. Chem. 2000, 43, 1541; Boger et al., Bioorg. Med. Chem. Lett. 2001, 11, 2021; Parrish et al., Bioorg. Med. Chem. 2003, 11, 3815; Daniell et al., Bioorg. Med. Chem. Lett. 2005, 15, 111; Tichenor et al., J. Am. Chem. Soc. 2006, 128, 15683; Purnell et al., Bioorg. Med. Chem. 2006, 16, 5677; EP 0154445; WO 88/04659; WO 90/02746; WO 97/12862; WO 97/32850; WO 97/45411; WO 98/52925; WO 99/19298; WO 01/83482; WO 02/067937; WO 02/067930; WO 02/068412; WO 03/022806; WO 2004/101767 and WO 2006/043839), which typically show similar cytotoxicity, but reduced hepatotoxicity. In addition, however, these derivatives do not have sufficient selectivity for tumor cells, because the selectivity of these agents, and cytotoxic agents in General are based largely on differences in the rate of proliferation of tumor cells and normal cells, and therefore, they also affect healthy cells, which show a high rate of proliferation. This usually leads to serious side effects. The concentration of drugs, is the quiet would completely destroy the tumor, it is impossible to achieve due to the limiting dose side effects such as toxicity to the gastrointestinal tract and bone marrow. In addition, tumors can develop resistance to anticancer agents after prolonged treatment. In modern drug development one of the main objectives should recognize the availability of a direct cytotoxic drugs to the site of localization of the tumor. One of the promising approaches to achieving selectivity against tumor cells or tumor tissues is to use the existence of tumour-related antigens, receptors and other sensitive parts, which can serve as a target. Such a target can be configured or she can, to some extent, specifically present in tumor tissue or located in close proximity to tissue, such as neovascular tissue relative to other tissues in order to achieve effective targeting to the target. Identified and justified many targets, and has developed a number of ways to identify and validate targets3. By attaching a ligand, for example, antibodies or fragments of antibodies, such as associated with a tumor antigen, receptor, or other susceptible agent, this agent can be selectively nab is Allen on tumor tissue. Another promising approach to achieve selectivity against tumor cells or tumor tissues is the use of tumour-related enzymes. The relatively high level of tumor-specific enzymes can turn pharmacologically inactive prodrug that comprises an enzyme substrate, either directly or indirectly associated with toxic medication, appropriate medication near or inside the tumor. Using this concept, high concentrations of toxic anti-cancer agent can be selectively created in the tumour site. All tumor cells can be destroyed, if the dose is large enough, which may reduce the development of resistant medicine tumor cells. The enzymes were also transported in proximity to or inside nesenevich cells or nesenevich tissue using, for example, proletarienne therapy with managed antibody enzyme (ADEPT)4, proletarienne therapy with controlled polymer-enzyme (PDEPT) or proletarienne treatment using controlled macromolecule enzyme (MDEPT)5, proletarienne therapy with managed virus enzyme (VDEPT)6or proletarienne therapy with managed genome enzyme (GDEPT)7. Using ADEPT, for example, there is xinou the prodrug converted into a cytotoxic compound on the surface of mistaway cells due to conjugate the antibody-enzyme which was previously focused on the surface of the specified cell. Another promising approach to achieve selectivity against tumor cells or tumor tissues is to use the effect of enhanced permeability and retention (EPR effect). Due to the specified EPR effect macromolecules passively accumulate in solid tumors due to broken pathology angiogenic tumor vasculature with its continuous endothelium, leading to hyperproliferate for large macromolecules and to the absence of the effect of lymphatic drainage of the tumor.8 Attaching therapeutic agent directly or indirectly to the macromolecule, the specified agent can be selectively directed to tumor tissue. In addition to effectively aim at the target, another criterion for successful application directed to conjugates of cytotoxic agents in the treatment of tumors is the fact that one or more of the agents effectively released from the conjugate. The next important criterion is that the conjugate is not toxic or has only a very weak toxicity, while itself cytotoxic agent demonstrates a high level of cytotoxicity. Another important criterion is the fact that the conjugate must have the appropriate pharmacological properties, that is their sufficient stability in the circulation, a slight trend towards aggregation and good solubility. Some conjugates of CC-1065 and their derivatives have been disclosed (see for derivative conjugates CC-1065, for example, Suzawa et al., Bioorg. Med. Chem. 2000, 8, 2175; Jeffrey et al., J. Med. Chem. 2005, 48, 1344; Wang et al., Bioorg. Med. Chem. 2006, 14, 7854; WO 91/16324; WO 94/04535; WO 95/31971; US 5475092; US 5585499; US 5646298; WO 97/07097; WO 97/44000; US 5739350; WO 98/11101; WO 98/25898; US 5843937; US 5846545; WO 02/059122; WO 02/30894; WO 03/086318; WO 2005/103040; WO 2005/112919; WO 2006/002895; and WO 2006/110476). These conjugates do not possess all of these favorable properties. As an illustrative example has been disclosed glycosidic conjugates seco CC-1065 analogues (analogues in which cyclopropyl ring as present in the CC-1065, "solved"), which can be activated at the site of damage, using the ADEPT approach.9The difference in cytotoxicity between these conjugates and related drugs (the ratio of cytotoxicity, here defined as IR50,conjugate/IR50,source drugwas, however, relatively small, and seco CC-1065 analogues were not exceptionally high cytotoxicity. The increase of the ratio of cytotoxicity was performed by creating a glycosidic conjugates to seco CC-1065 derivatives with secondary tsepliaeva group.10Although these conjugates showed high coefficient CIT the toxicity, their pharmacological properties were not optimal. For example, they had poor solubility, as a consequence, the inherent lipophilic nature of CC-1065 class of compounds. Accordingly, certainly there is a need in the conjugates of CC-1065 derivatives, which would demonstrate a high degree of cytotoxicity, containing CC-1065 derivatives, which would have an effective cytotoxicity and favorable pharmacological properties and effectively allocated to CC-1065 derivatives. SUMMARY of the PRESENT INVENTION The present invention satisfies the above requirements, offering the compounds of formula (I) or (II): or their pharmaceutically acceptable salt or solvate, where R1selected from halogen and OSO2Ruwhere Ruselected from optionally substituted C1-6of alkyl, C1-6perhalogenated, benzyl and phenyl; R2selected from H and optionally substituted C1-8of alkyl; R3, R3', R4and R4'independently selected from H and optionally substituted C1-8of alkyl, where two or more of R2, R3, R3', R4and R4'optionally joined to form one or more optionally substituted carbocycles or heterocycles; X2the choice is up from O, C(R14)(R14'and NR14'where R14selected from H and optionally substituted C1-8the alkyl or acyl and R14'may be absent or may be selected from H and optionally substituted C1-8the alkyl or acyl; each R5, R5', R6, R6', R7and R7'independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, Rk, SRk, S(O)Rk, S(O)2Rk, S(O)ORk, S(O)2ORk, OS(O)Rk, OS(O)2Rk, OS(O)ORk, OS(O)2ORk, ORk, OtherkN(RkRL,+N(Rk)(RLRm, P(O)(ORk)(ORL), OP(O)(ORk)(ORL), SiRkRLRmWith(O)RkWith(O)ORkWith(O)N(RLRk, OC(O)Rk, OC(O)ORk, OC(O)N(RkRLN(RL)C(O)RkN(RL)C(O)ORkand N(RL)C(O)N(RmRkwhere Rk, RLand Rmindependently selected from H and optionally substituted C1-4of alkyl, C1-4heteroalkyl, C3-7cycloalkyl, C3-7geterotsiklicheskie, C4-12aryl or C4-12heteroaryl, and two or more of Rk, RLand Rmoptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, the/or R 5+ R5'and/or R6+ R6'and/or R7+ R7'independently represent =O, =S, or =NR12where R12selected from H and optionally substituted C1-6of alkyl, and/or R5'and R6'and/or R6'and R7'and/or R7'and R14'no, that means that a double bond is present between the atoms, which are attached to R5'and R6'and/or R6'and R7'and/or R7'and R14'accordingly, two or more of R5, R5', R6, R6', R7, R7', R14and R14'optionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles; X1selected from O, S, and NR13where R13selected from H and optionally substituted C1-8of alkyl; X3selected from O, S, and NR15where R15selected from H and optionally substituted C1-8the alkyl or acyl, or-X3- represents-X3aand X3b-where X3aconnected to the carbon atom that is attached to X4and X3battached to the phenyl ring in ortho-position to R10where X3aindependently selected from H and optionally substituted C1-8the alkyl or acyl and X3bselected from the same group of substituents, and R8; X4the choice is up from N and CR 16where R16selected from H and optionally substituted C1-8the alkyl or acyl; X5selected from O, S, and NR17where R17selected from H and optionally substituted C1-8the alkyl or acyl; R8, R9, R10and R11each independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, RX, SRx, S(O)Rx, S(O)2Rx, S(O)ORx, S(O)2ORx, OS(O)Rx, OS(O)2Rx, OS(O)ORx, OS(O)2ORx, ORx, OtherXN(RxRy,+N(Rx)(RyRz, P(O)(ORx)(ORy), OP(O)(ORx)(ORy), SiRxRyRzWith(O)RxWith(O)ORxWith(O)N(RyRx, OC(O)Rx, OC(O)ORx, OC(O)N(RxRyN(Ry)C(O)RxN(Ry)C(O)ORxN(Ry)C(O)N(RzRxand water-soluble group, where Rx, Ryand Rzindependently selected from H and optionally substituted C1-15of alkyl, C1-15heteroalkyl, C3-15cycloalkyl, C3-15geterotsiklicheskie, C4-15aryl or C4-15heteroaryl, one or more optional substituents in Rx, Ryand Rzoptional is a water-soluble group, and two or more of Rx, Ryand Rznot necessarily connected with the formation of a single and more optionally substituted aliphatic or aromatic carbocycles or heterocycles, and at least one of R8, R9, R10and R11includes at least one water-soluble group, two or more of R8, R9, R10, R11or X3boptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles; a and b independently are selected from 0 and 1; c is chosen from 0, 1 and 2; provided that in the compound of formula (I), at least one of R2, R3, R3', R4, R4', R5and R5'is not hydrogen. In another aspect under consideration, the invention relates to the compound of formula (III): or its pharmaceutically acceptable salt or MES, where V2or is absent or represents a functional fragment; each L2independently absent or represents a linking group that binds V2with L or V1or Y, if L is absent; each L is independently absent or represents a linking group that links L2or V2if L2no, with one or more of V1and/or Y; each V1independently represents H or conditionally ottsepleny or conditionally-transformed fragment, which can be derived or transformed in accordance with the ATA chemical, photochemical, physical, biological, or enzymatic process; each Y is independently absent or is self-destructing spacer elements of the system, which consists of 1 or more of the self-destructing spacers and connected with V1not necessarily L, and one or more Z; each of p and q are numbers representing degree of branching and each independently is a positive integer; z represents a positive integer equal to or less than the total number of connection points to Z in one or more of V1-Y fragments; each Z independently represents a compound of formula (I) or (II)as defined above where one or more of the X1, R6, R7, R8, R9, R10and R11may, optionally, additionally be substituted by the Deputy of the formula (V): where each V2'L2', L', V1', Y', Z', p', q' and z' has the values specified for V2L2, L, V1, Y, Z, p, q and z, where one or more of the substituents of formula (V) is independently connected to one or more of X1, R6, R7, R8, R9, R10and R11through the Y'or V1'if Y'no, each Z is connected with the Y or V1if Y is absent, or through the X1or via an atom in R6, R7, R8, R , R10or R11; provided that at least one of the one or more of V1and one or more of V1'does not represent H. It is noted that z does not mean degree of polymerization; z indicates that the number of fragments Z are connected to one another. The present invention also relates to the compound of formula (IV): or its pharmaceutically acceptable salt or MES, where RM represents a reactive fragment and L, V1, Y, Z, p and z have the above values, except that L now connects RM with one or more of V1and/or Y, and V1, Y, and Z may contain a protective group, and one or more of V2'-L2'fragments, not necessarily present in Z, as described above, may optionally and independently be replaced by RM'that is a reactive fragment, and where, if (IV) there is more than one reactive fragment, these reactive fragments of the same or different. These conjugates linker agent may be (or may not) be considered as intermediate compounds for the compounds of formula (III). In addition, the present invention relates to containing cyclopropene ring analogs of the compounds of formulas (I) and (II)that is formed is the result of the rearrangement and the concomitant removal of the H-R 1from the corresponding seco compounds of formulas (I) and (II) (figure 1). Consider that these contain cyclopropane ring analogues are active compounds that are believed to be formed from compounds of formulas (I) and (II) in vivo in the specified rearrangement. The present invention relates to enantiomerically pure and/or diastereoisomers pure compounds of formulas (I)-(IV), as well as enantiomeric and/or diastereoisomeric mixtures of compounds of formulas (I)-(IV). It has been unexpectedly discovered that compounds of the formula (III) demonstrate a high indicator of cytotoxicity, and in addition, it was shown that the related compounds of formulas (I) and (II) have high cytotoxicity and higher solubility in water than previously known similar compounds. These properties make the compounds of formula (III) is very suitable for the purposes of drug delivery, including such applications as drug delivery to the target and controlled allocation of drugs. BRIEF DESCRIPTION of DRAWINGS Figure 1 illustrates the rearrangement seco connection containing cyclopropyl connection. Figure 2 illustrates the receipt of some agents by attaching alkylating DNA fragment and DNA binding of the fragment. Figure 3 illustrates the receipt of some β-galactopyranose conjuga the s of the present invention. Figure 4 illustrates the obtaining of DNA binding compounds 13. Figure 5 illustrates obtaining a DNA binding compound 19. 6 illustrates obtaining a DNA binding compound 27. Fig.7 illustrates obtaining a DNA binding compound 30. Fig illustrates the formation of compounds 37-39 of DNA binding compounds 41. Fig.9 illustrates the synthesis agent 45. Figure 10 illustrates the synthesis agent 33. 11 illustrates obtaining the conjugate 36. Fig illustrates the synthesis of conjugates of linker-agent 47a-f. Fig illustrates the synthesis of conjugates of linker-agent 48c-d. Fig illustrates obtaining conjugates linker-agent 50a-c. Fig illustrates the synthesis of activated linkers 57-60. Fig illustrates the in vitro cytotoxicity of conjugates 8a and 8a' in relation to cell lines of human lung carcinoma A. Fig is the treatment for in vivo experiment according to the scheme ADEPT. Fig illustrates the volume of tumors in patients under-treated and control mice in ADEPT in vivo experiment. DESCRIPTION THIS ISBREENE Below a detailed description is given for a more complete understanding of the present invention. Description Unless otherwise noted, all used here technical and synthetic terms typically have the same values, which usually meant umelaut specialists in this field. The term "antibody", as used herein, refers to full length immunoglobulin molecule, to an immunological active part of the full-length immunoglobulin molecules, or to the derived full-length immunoglobulin molecules or active part of the molecule that contains an antigen binding site that immunospecificity binds the antigen of interest target or its parts, and such targets include, but are not limited to, tumor cells. The immunoglobulin may be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgG2, IgG3, IgG4, IgAl and IgA2)or subclass of immunoglobulin molecules. The immunoglobulin can be obtained from any species, such as humans, rodents (e.g. mice, rats or hamsters), monkeys, sheep, rabbits, goats, Guinea pigs, camelids, horses, cows, or chickens, but preferably, the immunoglobulin is of human, murine or rabbit origin. Antibodies that can be used in the present invention include, but are not limited to, monoclonal, polyclonal, bespecifically, human, gumanitarnye or chimerical antibodies, single-chain antibodies, Fv fragments, Fab fragments, F(ab') fragments, F(ab')2fragments, fragments produced using libraries Fab expressed sequence is of linota, anti-idiotypical antibodies, CDR, and epilepsyusa fragments of any of the above, which immunospecificity associated with interest antigen. The term "tsepliaeva group" refers to a group which can be substituted by another group. Such otsepleniya groups are well known in the art, and examples include, but are not limited to, halides (fluorides, chlorides, bromides, iodides), sulfonates (for example, methanesulfonate, para-toluensulfonate and triftorbyenzola), succinimide-N-oxide, para-nitrophenoxide, Pantothenate, tetraterpenoids, carboxylate and alkoxycarbonyl. The term "water-soluble group" refers to a functional group, which is well collaterals in the aquatic environment and which gives greater solubility in water connection, to which it is attached. Examples of water-soluble groups include, but are not limited to, alcohol and polyalcohol, linear or cyclic saccharides, primary, secondary, tertiary or Quaternary amines and polyamine, sulfate groups, carboxylate groups, phosphate groups, phosphonate groups, ascorbate groups, glycols, including polyethylene glycols and polyesters. The term "substituted", when used as an adjective to the terms "alkyl", "heteroalkyl", "cycloalkyl", "heteroseksualci", "aryl", "heteroa the l", and so on, indicates that the above "alkyl", "heteroalkyl", "cycloalkyl", "heterocytolysine", "aryl" or "heteroaryl" group contains one or more of the substituents that include, but are not limited to, OH, =O, =S, =NRh, =N-ORh, SH, NH2, NO2, NO, N3, CF3, CN, OCN, SCN, NCO, NCS, C(O)NH2With(O)H, C(O)OH, halogen, Rh, SRh, S(O)Rh, S(O)ORh, S(O)2Rh, S(O)2ORh, OS(O)Rh, OS(O)ORh, OS(O)2Rh, OS(O)2ORhOP(O)(ORh)(ORi), P(O)(ORh)(ORi), ORh, OtheriN(RhRi,+N(Rh)(RiRjSi(Rh)(Ri)(Rj), C(O)RhWith(O)ORhWith(O)N(RiRh, OC(O)Rh, OC(O)ORh, OC(O)N(RhRiN(Ri)C(O)RhN(Ri)C(O)ORhN(Ri)C(O)N(RjRhand thio derivatives of these substituents, or a protonated or deprotonated forms of any of these substituents, where Rh, Riand Rjindependently selected from H and optionally substituted C1-15of alkyl, C1-15heteroalkyl, C3-15cycloalkyl, C3-15geterotsiklicheskie, C4-15aryl or C4-15heteroaryl or combinations thereof, and two or more of Rh, Riand Rjcan be optionally connected to form one or more of carbon is (or heterocycles. The term "aryl", in the sense used here, refers to carbocyclic aromatic Deputy, which may consist of one ring, or two or more rings fused together. Examples of aryl groups include, but are not limited to, phenyl, naphthyl and anthracene. The term "heteroaryl", in the sense used here, refers to carbocyclic aromatic Deputy, which may consist of one ring, or two or more rings fused together, where at least one carbon atom in one of the rings is replaced by a heteroatom. Examples of heteroaryl groups include, but are not limited to, pyridinyl, furanyl, pyrrolyl, triazolyl, pyrazolyl, imidazolyl, thiophenyl, indolyl, benzofuranyl, benzimidazolyl, indazoles, benzotriazolyl, benzisoxazoles and chinoline. The term "alkyl", in the sense here used, refers to a linear or branched, saturated or unsaturated hydrocarbon substituent. Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl, butyl, pentyl, hexyl, octyl, decyl, isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, 2-methylbutyl, vinyl, allyl, 1-butenyl, 2-butenyl, isobutylene, 1-pentenyl, and 2-pentenyl. The term "heteroalkyl", in the sense used here, refers to the LINEST is mu or branched, saturated or unsaturated hydrocarbon Deputy, in which at least one carbon atom is replaced by a heteroatom. Examples include, but are not limited to, methoxymethyl, ethoxymethyl, methoxyethyl, ethoxyethyl, methylaminomethyl, dimethylaminomethyl, methylaminomethyl, dimethylaminomethyl, methylthiomethyl, ethylthiomethyl, ethylthioethyl and methylthioethyl. The term "cycloalkyl", in the sense here used, refers to a saturated or unsaturated non-aromatic carbocyclic Deputy, which may consist of one ring, or two or more rings fused together. Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexyl, 1,3-cyclohexadienyl, decaline, and 1,4-cyclohexadienyl. The term "heteroseksualci" in the sense used here, refers to a saturated or unsaturated non-aromatic cyclic hydrocarbon Deputy, which may consist of one ring, or two or more rings fused together, where at least one carbon atom in one of the rings is replaced by a heteroatom. Examples include, but are not limited to, tetrahydrofuranyl, pyrrolidinyl, piperidinyl, 1,4-dioxane, decahydroquinoline, piperazinil, oxazolidinyl and morpho is inil. The ending "-ilen," as opposed to "-Il", for example, in "alkylene" as opposed to "alkyl" indicates that the specified example "alkylene" represents a bivalent fragment connected with one or two other fragments via two covalent simple connection or through one of the double bond in contrast to the variant, when the monovalent group is connected to one segment through one simple covalent bond, as specified in the example of "alkyl". The term "alkylene" therefore refers to an unbranched or branched, saturated or unsaturated hydrocarbon fragment; the term "heteroalkyl" in the sense used here, refers to an unbranched or branched, saturated or unsaturated hydrocarbon fragment, in which at least one carbon atom is replaced by a heteroatom; the term "arisen" in the sense used here, refers to aromatic hydrocarbon fragment, which may consist of one ring, or two or more rings fused together; the term "heteroaryl" in the sense used here refers to aromatic hydrocarbon fragment, which may consist of one ring, or two or more rings fused together, where at least one carbon atom in one of the rings of replacing the Yong heteroatom; the term "cycloalkyl" in the sense used here, refers to a saturated or unsaturated non-aromatic carbocyclic portion, which may consist of one ring, or two or more rings fused together; the term "heterocyclochain" in the sense used here, refers to a saturated or unsaturated non-aromatic cyclic hydrocarbon fragment, which may consist of one ring, or two or more rings fused together, where at least one carbon atom in one of the rings is replaced by a heteroatom. Examples of the divalent fragments include those examples which are given here for monovalent groups in which removed one carbon atom. The prefix "poly" in terms of "polyalkylene", "polyheteroarylenes", "polyarylene", "polyheteroarylenes", polycyclohexylene", "polyheterocycles", etc. indicates that two or more of these "-ilen," fragments, for example, alkilinity fragments, together with the formation of branched or unbranched multivalent fragment containing one or more of the sites attach to neighboring fragments. Some compounds of the present invention have chiral centers and double bonds; mixtures of enantiomeric, diastereomeric and geometric two or the more isomers, in any combination, as well as individual isomers included in the scope of the present invention. Compounds of the present invention can also be characterized not natural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. Assumes that all isotopic variants of the above compounds of the present invention, regardless of whether they are radioactive or not, are included in the scope of the present invention. The phrase "pharmaceutically active salt" in the sense used here, refers to pharmaceutically acceptable organic or inorganic salts of the compounds of the present invention. For compounds containing one or more basic groups, for example, the amino group, can form a salt accession acid. For compounds containing one or more of the acid groups such as carboxylic acid group, can be used to form the salt of the attaching base. For compounds containing, acidic and basic groups can form, in addition, zwitterion in the form of salts. In those cases, when atoms with multiple charges are part pharmaceutically acceptable salt, it can have many counterions. The phrase "pharmaceutically acceptable MES" refers to the Association of one or more of the molecules of the solvent the connection of the present invention. Examples of solvents which form pharmaceutically acceptable solvate, include, but are not limited to, water, isopropyl alcohol, ethanol, methanol, DMSO, ethyl acetate and acetic acid. The term "conjugate" hereinafter refers to the compound of formula (III). The term "conjugate linker-agent" hereinafter refers to the compound of formula (IV). The term "agent" hereinafter refers to compounds of formula (I), (II), (VII), (VIII), (I') or (II'). The term "guide to the target fragment" refers to any molecule that specifically binds or reaction associated, or complexesa fragment, specific or relative abundance present on or near mesiniaga site, in or near mistaway cells, or (close) mistaway tissue or misheneva the body, for example, receptor, receptor complex, the substrate, antigenic determinant, or other receptive fragment or molecule, which may send the specified conjugate to the specified micheneau site using other mechanisms, due to their nature, for example, for the expense of the EPR effect. Examples of the sending to the target fragment include, but are not limited to, aptamer, antibody, or antibody fragment, a polymer, a dendrimer, lectin, responsive biological modifier, an enzyme, a vitamin, a growth factor, a steroid, saarnisto, oligosaccharide residue, a carrier protein and a hormone, or any combination thereof. The phrase "fragment, which improves the pharmacokinetic properties of the compound"refers to a fragment, which alters the pharmacokinetic properties of the compounds of the present invention in such a way that can be achieved in an improved therapeutic effect. The selection may, for example, to increase the solubility, increase circulation time, or reduced immunogenicity. The term "linking group" refers to a structural element of the connection that binds one structural element of the specified compounds with one or more of the other structural elements of the specified same connection. The phrase "a number representing the degree of branching" is used to denote that a number is a subscript that follows the closing parenthesis shows how many units of the snippet inside the brackets directly attached to the fragment located immediately to the left of the corresponding opening bracket. For example, A-(B)bwhere b is a number representing the degree of branching, means that each of b units of B are directly connected to A. This, in turn, means that if b=2, the formula reduces to B-A-B. The phrase "a number representing the degree of polymers the tion" is used to refer to that subscript following the closing parenthesis shows how many units of the snippet within the braces are connected to each other. For example, -(B)bwhere b denotes the number representing the degree of polymerization, means that if b=2, the formula reduces to A-B-B. The term "spacer single allocation" refers to the self-destructing spacer that can distinguish one piece after the destruction. The term "spacer multiple selection" refers to the self-destructing the spacer, which may allocate two or more components after repeated self-destruct. The term "electronic cascade spacer" refers to a self-destructing the spacer, or a branched or unbranched, which can self-destruct as a result of one or more of the 1.2+2n e cascade of destruction (n≥1). The term "ω-amino aminocarbonyl cyclization spacer" refers to a self-destructing the spacer, which may be disposed in cyclization process in the formation of a cyclic derivative of urea. The term "spacer elements system" refers to one piece of spacer elements, or two or more of the same or different spacer elements fragments connected together. Spacer elements the system can be branched or nonbranched and may contain one or more of the sites attach to Z, as for V1and not necessarily for L. In this paper and in the claims, the verb "include", "have", "include" and their combined use options in their limitiruyushchem sense, which implies that the subject, subsequent or preceding the verb, are included, but items not mentioned specifically, is not excluded. In addition, a link to an element with an indefinite article "a" or "an" does not exclude the possibility that there is more than one item, unless the context specifically does not follow that there is one and only one element. The indefinite article "a" or "an" so "means " at least one". In General the structures over the description and in the claims the letters used to define structural elements. Some of these letters may coincide with the designation of an atom such as C, N, O, P, K, B, F, S, U, V, W, I, and Y. In order to avoid misunderstandings, it is necessary to distinguish refer to the structural elements and atoms. If there is one or more adjectives and/or determining the expression the noun, which is a first in the list of nouns or b) which is anywhere in the middle of a list of nouns, the specified noun and adjective together preceded by the word "and", prilagatel the e refers not only to the specified noun but to all of the following noun separately, unless the context States otherwise. This means, for example, that the phrase "optionally substituted C1-4alkyl, C1-4heteroalkyl, C3-7cycloalkyl, or C3-7heteroseksualci" should be read as "optionally substituted C1-4alkyl, optionally substituted C1-4heteroalkyl, optionally substituted C3-7cycloalkyl or optionally substituted C3-7heteroseksualci". Throughout the description and claims of the invention presents images of molecular structures or their parts. As usual in such images of the bonds between atoms are represented by lines, in some cases, to indicate stereochemistry, bold or dashed, or wedge-shaped lines. Usually the line that ends in a space ("free" end), i.e. with one end missing another line or a specific atom, connected with it, is CH3group. This is true for figures representing the compounds in accordance with this invention. For such structures, representing the structural connection element in accordance with this invention, the line ending in space, may indicate the position of the accession of another structural element of the specified connection. This is about nachatsja wavy line, perpendicular to and crossing the free line in most images. In addition, these structures or their parts were depicted assuming that patterns read from left to right, which means, for example, that the figures of the compounds of the formula (III) V2always located to the left (if present), and Z is always right in these structures. In the description uses the following abbreviations and they have these definitions: Aloc: allyloxycarbonyl; Asc: ascorbate; Boc=tert-butyloxycarbonyl; DCC=N,N'-dicyclohexylcarbodiimide; DIPEA: diisopropylethylamine; DME: 1,2-dimethoxyethane; DMF=N,N-dimethylformamide; Fmoc=9-fluorenylmethoxycarbonyl; HOBt: 1-hydroxybenzotriazole; HOSu=N-hydroxysuccinimide; NMM: N-methylmorpholine; PABA=para-aminobenzoyl alcohol; PBS: saline, superyoung phosphate; PNP=pair-nitrophenoxide; PNPC1=pair-nitrophenylphosphate; PNP2O=bis(para-nitrophenyl)carbonate; SCID: severe complicated immunodeficiency; TFA: triperoxonane acid; THF: tetrahydrofuran. Agents, conjugates of linker-agent conjugates In the present invention proposed new agents that can be classified as belonging to the class of binding DNA alkylating agent CC-1065 and duocarmycin. Moreover the present invention relates to novel conjugates of these agents and conjugates of the linker-the Gent, which may or may not serve as intermediates for obtaining the conjugates. Assume that these agents of the present invention can be used to treat diseases which are characterized by undesirable (cell) proliferation. For example, the agent of the present invention can be used for the treatment of tumors, cancer, autoimmune disease or infectious disease. Assume that the conjugates of the present invention in one aspect should be applicable for referral agents of formulas (I) and (II) to the specific micheneau the site where the specified conjugate can be transformed into one or more of the agents or it can be induced to transform into one or more of these agents. The present invention can also find application in (nonspecific) controlled the selection of one or more of these agents from the conjugate, with the aim, for example, improved pharmacokinetic properties. It has been unexpectedly discovered that compounds of the formula (III) demonstrate a high degree of cytotoxicity, and, in addition, it was shown that the related compounds of formulas (I) and (II) are highly cytotoxic and dissolve better in water than previously known similar compounds. Not communicating with any theory, it is possible before ologit, what is the explanation for the high ratio of cytotoxicity may be the presence of a water-soluble group such agent in combination with several shielded carbon to which is attached tsepliaeva group. The increased solubility of the agent may lead to the fact that the connection will be easier to achieve their (intracellular) the target and, thus, increase their cytotoxicity after separation of the conjugate, although it also can reduce the aggregation of the conjugate and reduce side effects conjugate associated with premature release of agent from the conjugate, since the released agent may be less effective in penetration (not miseriya) cells due to its high polarity. Steric hindrance can reduce direct alkylation of biomolecule conjugate and non-DNA alkylation agent, thereby reducing specificescuu (cyto)toxicity. Thus, inventiona the concept of the unification water solubility and steric shielding in the compounds of formulas (I) and (II) may result in agents that dissolve better in water and are more selective and have a high rate of cytotoxicity of conjugates of the present invention. Agents In one aspect of this invention proposed a compound of formula (I) or (II): or its pharmaceutically acceptable salt, or MES, where R1selected from halogen and OSO2Ruwhere Ruselected from optionally substituted C1-6of alkyl, C1-6perhalogenated, benzyl and phenyl; R2selected from H and optionally substituted C1-8of alkyl; R3, R3', R4and R4'independently selected from H and optionally substituted C1-8of alkyl, where two or more of R2, R3, R3', R4and R4'optionally joined to form one or more optionally substituted carbocycles or heterocycles; X2selected from O, C(R14)(R14'and NR14'where R14selected from H and optionally substituted C1-8the alkyl or acyl and R14'may be absent or may be selected from H and optionally substituted C1-8the alkyl or acyl; each of R5, R5', R6, R6', R7and R7'independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, Rk, SRk, S(O)Rk, S(O)2Rk, S(O)ORk, S(O)2ORk, OS(O)Rk, OS(O)2Rk, OS(O)ORk, OS(O)2ORk, ORk, OtherkN(RkRL,+N(Rk)(RLRm, P(O)(ORk)(ORL), OP(O)(ORk)(ORLkRLRmWith(O)RkWith(O)ORkWith(O)N(RLRk, OC(O)Rk, OC(O)ORk, OC(O)N(RkRLN(RL)C(O)RkN(RL)C(O)ORkand N(RL)C(O)N(RmRkwhere Rk, RLand Rmindependently selected from H and optionally substituted C1-4of alkyl, C1-4heteroalkyl, C3-7cycloalkyl, C3-7geterotsiklicheskie, C4-12aryl or C4-12heteroaryl, and two or more of Rk, RLand Rmoptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, and/or R5+ R5'and/or R6+ R6'and/or R7+ R7'independently represent =O, =S, or =NR12where R12selected from H and optionally substituted C1-6of alkyl, and/or R5'and R6'and/or R6'and R7'and/or R7'and R14'no, that means that a double bond is present between the atoms, which are attached to R5'and R6'and/or R6'and R7'and/or R7'and R14'accordingly, two or more of R5, R5', R6, R6', R7, R7', R14and R14'optionally joined to form one or more optionally substituted aliphatic or aromatic Carbo is eklow or heterocycles; X1selected from O, S, and NR13where R13selected from H and optionally substituted C1-8of alkyl; X3selected from O, S, and NR15where R15selected from H and optionally substituted C1-8the alkyl or acyl, or-X3- represents-X3aand X3b-where X3aconnected to the carbon atom that is attached to X4and X3battached to the phenyl ring in ortho-position to R10where X3aindependently selected from H and optionally substituted C1-8the alkyl or acyl and X3bselected from the same group of substituents, and R8; X4selected from N and CR16where R16selected from H and optionally substituted C1-8the alkyl or acyl; X5selected from O, S, and NR17where R17selected from H and optionally substituted C1-8the alkyl or acyl; R8, R9, R10and R11each independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, RX, SRx, S(O)Rx, S(O)2Rx, S(O)ORx, S(O)2ORx, OS(O)Rx, OS(O)2Rx, OS(O)ORx, OS(O)2ORx, ORx, OtherXN(RxRy,+N(Rx)(RyRz, P(O)(ORx)(ORy), OP(O)(ORx)(ORy), SiRxRyRzWith(O)RxWith(the)OR xWith(O)N(RyRx, OC(O)Rx, OC(O)ORx, OC(O)N(RxRyN(Ry)C(O)RxN(Ry)C(O)ORxN(Ry)C(O)N(RzRxand water-soluble group, where Rx, Ryand Rzindependently selected from H and optionally substituted C1-15of alkyl, C1-15heteroalkyl, C3-15cycloalkyl, C3-15geterotsiklicheskie, C4-15aryl or C4-15heteroaryl, one or more of the optional substituents in Rx, Ryand Rzoptionally present water-soluble group, and two or more of Rx, Ryand Rzoptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, and at least one of R8, R9, R10and R11includes at least one water-soluble group, two or more of R8, R9, R10, R11or X3boptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles; a and b independently are selected from 0 and 1; c is chosen from 0, 1 and 2; provided that in the compound of formula (I), at least one of R2, R3, R3', R4, R4', R5and R5'is not hydrogen. Should understand is, what the present invention relates to enantiomerically pure and/or diastereomers pure compounds of formulas (I) and (II), as well as enantiomeric and/or diastereoisomeric mixtures of compounds of formulas (I) and (II). Consideration of the effects of substituents in the compounds of formulas (I) and (II) and containing cyclopropyl equivalents presented in the document presented without the assumption of a specific mechanism of action of the compounds of formulas (I) and (II) and containing cyclopropyl-analogues. R1in the compounds of formula (I) or (II) is tsepliaeva group. In one embodiment of the present invention tsepliaeva group R1in the compounds of formula (I) or (II) represents a halide. In another embodiment, R1choose from chloride (Cl), bromide (Br) and iodide (I). In another embodiment, R1is chloride (Cl). In another embodiment, R1represents a bromide (Br). Varying tsepliaeva group, R1you can adjust the alkylating activity of seco agents and influence the rate of transformation seco agent containing cyclopropyl agent. If the ability to chipped off group, R1too high, it can cause seco agent will also alkylating agent that can reduce the ratio of cytotoxicity of the conjugates of the compounds of formulas (I) and (II)because the specified agent may be able to alkilirovanii, the ri still staying connected in the conjugate. On the other hand, if R1is too bad tsepliaeva group, seco agent will not be able to provide the closure with the formation containing cyclopropyl agent, which is reputed to be an active element, which can reduce its cytotoxicity and, most likely, reduce the ratio of cytotoxicity. Other means for regulating the alkylating activity of seco agents and their containing cyclopropyl derivatives may be some shielding of the carbon that is attached tsepliaeva group, or which may be nucleophilic attack by selecting at least one of the present R2, R3, R3', R4, R4', R5and R5'that are different from hydrogen. Shielding the specified carbon can reduce specific alkylation of compounds of formulas (I) and (II) and containing cyclopropyl analogs and their conjugates. Although the introduction of steric hindrance may also affect the degree of alkylation of DNA, and it may be reasonable to assume that spetsificheskoe alkylation may be affected by the relatively stronger than the alkylation of DNA, as the latter occurs, apparently, after the agent is perfectly located for nucleophilic attack, being associated with the minor groove of the DNA. Carbon, the which is attached R 1in the compound of formula (II), and which is a secondary carbon atom, already to some extent shielded compared to the carbon attached to R1in the compound of formula (I), if R2represents H. In this respect, the compound of formula (II) can be compared with the compound of the formula (I)in which R2different from hydrogen. Further, the shielding can, however, be accomplished by selecting one or more of R3, R3', R4, R4', R5and R5'that must be different from hydrogen. In one embodiment, R2in the compound of formula (I) represents an optionally substituted C1-8alkyl. In another embodiment, R2is optionally substituted linear C1-8alkyl. In another embodiment, R2represents unsubstituted linear C1-8alkyl. In another embodiment, R2represents methyl. Alternative or simultaneous steric shielding of the carbon attached to R1you can enter by selecting one or more of R3, R3', R4and R4'different from hydrogen. In one embodiment, R3, R3', R4and R4'each represents H. In another embodiment, R3and R3'both represent H. In another embodiment, R4and R4'both represent H. In another embodiment, one of R3and R3'PR is dstanley C 1-8alkyl while the other is H. In another embodiment, one of R4and R4'represents C1-8alkyl while the other is H. In another embodiment, one of R3and R3' represents C1-8alkyl and one of R4and R4'represents C1-8alkyl, while others are H. In another embodiment, both R3and R3'independently selected from C1-8the alkyl. In another embodiment, both R4and R4'independently selected from C1-8the alkyl. In another embodiment, R2is H and one of R3, R3', R4and R4'selected from C1-8the alkyl. In another embodiment, R2is H and one of R3, R3', R4and R4'choose from bromide. In another embodiment, R2is H and two of R3, R3', R4and R4'independently selected from C1-8the alkyl. In another embodiment, R2is H and two of R3, R3', R4and R4'represent methyl. On the alkylating activity of the compounds of formula (I) or (II) or their cyclopropylacetic analogues may be affected by the nature of X1. The nature of X1can affect the speed with which and the conditions under which the closure ring seco agents with education cyclopropyl analogues and/or the speed with which cyclopropene ring raskryvayut is the result of nucleophilic attack DNA, and thus affect the nature of the alkylation. In one embodiment, X1represents O. The substituents R5, R5', R6, R6', R7, R7'and X2as well as the size of the ring (rings)connected with the left side of the ring containing X1may , for example, each independently or two or more together, to influence the pharmacokinetic properties of the agent to affect the solubility, to influence the nature of aggregation, to influence the process of alkylation of DNA, or to affect the strength of binding DNA. In addition, especially R5and R5'may also affect the degree of shielding of the carbon, which may be a nucleophilic attack. In one embodiment, R5and R5'both represent H. In another embodiment, at least one of R5and R5'is not hydrogen. In another embodiment, R5is not hydrogen. In another embodiment, R2represents hydrogen and at least one of R5or R5'not represent hydrogen. In another embodiment, R5selected from nitro, halogen, amino, hydroxy, and optionally substituted, alkylamino, alkylcarboxylic, alkoxycarbonyl, acyloxy, alkylcarboxylic, alkylaminocarbonyl or1-4the alkyl. In another embodiment, R5is optionally substituted With linear1-4Alki is. In another embodiment, R5represents unsubstituted linear C1-4alkyl. In another embodiment, R5represents methyl. Although the rate of alkylation and the effectiveness of compounds similar to compounds of formulas (I) and (II), can be adjusted in various ways, in one aspect of the present invention this is achieved by introducing spatial screening, selecting for compounds of formula (I), at least one of present R2, R3, R3', R4, R4', R5and R5'different from hydrogen and the compounds of formula (II) optionally one or more of those present R2, R3, R3', R4, R4', R5and R5'different from hydrogen. In one aspect of the present invention compounds of formula (I) and (II) can be represented by compounds of formulas (Ia) and (IIa), respectively: In one embodiment, R6and R7in (Ia) and (IIa), both represent N. In another aspect of the present invention compounds of formula (I) and (II) can be represented by compounds of formulas (Ib) and (IIb), respectively: In one embodiment, X2in (Ib) or (IIb) is N. In one embodiment, X2in (Ib) or (IIb) is CH. In one embodiment, R5, R6and R7in (Ib) or (IIb) each represents H. what one embodiment, R 5, R6and R7in (Ib) or (IIb) each represents H and X2is CH. In another embodiment, R5and R7in (Ib) or (IIb) each represents H and R6is CO2Me. In another embodiment, R5and R7in (Ib) or (IIb) each represents H and R6is OMe. In another embodiment, R5and R7in (Ib) or (IIb) each represents H and R6is CN. In another embodiment, R5in (Ib) or (IIb) is selected from nitro, halogen, amino, hydroxy, and optionally substituted, alkylamino, alkylcarboxylic, alkoxycarbonyl, acyloxy, alkylcarboxylic, alkylaminocarbonyl or1-4the alkyl. In another embodiment, R5in (Ib) or (IIb) is optionally substituted With linear1-4alkyl. In another embodiment, R5in (Ib) or (IIb) is unsubstituted linear C1-4alkyl. In another embodiment, R5in (Ib) or (IIb) is methyl. In yet another aspect, compounds of formulas (I) and (II) can be represented by compounds of formulas (Ic) and (IIc), respectively: In one embodiment, X2in (Ic) or (IIc) is NH. In another embodiment, R6and R7in (Ic) or (IIc) are H and CO2CH3respectively, and X2represents NH. In another embodiment, R7and R6in (Ic) or (IIc) are H and CO CH3respectively, and X2represents NH. In another embodiment, R6in (Ic) or (IIc) is CH3and X2represents NH. In yet another aspect, compounds of formulas (I) and (II) can be represented by compounds of formulas (Id) and (IId), respectively: In one embodiment, X2in the (Id) or (IId) is NH. In one embodiment, R6and R6'in the (Id) or (IId) together represent =O. In another embodiment, R7and R7'in the (Id) or (IId) are CO2CH3and CH3respectively. In another embodiment, in compounds of formula (Id) or (IId), X2is NH, R6and R6'together represent =O, and R7and R7'represent CO2CH3and CH3respectively. In one embodiment, X3represents NH. In another embodiment, X3represents O. In one embodiment, X4is CH. In one embodiment, X5represents O. Water-soluble group is a group which imparts increased solubility of the compounds of formulas (I) and (II) in comparison with previously known similar compounds. This group may also prevent or reduce aggregation of the compounds of the present invention or to reduce side effects. Examples of water-soluble groups include, but are not limited to, -NH2-NH-, -Othera, -NRa-, -N(Ra)(Rb), -+N(Ra)(Rb)-, -+N(Ra)(Rb)(Rc), -COOH, -COORa, -OP(O)(OH)2, -OP(O)(OH)O-, -OP(O)(ORa)O-, -OP(O)(OH)ORa, -OP(O)(ORbORa, -P(O)(OH)2, -P(O)(OH)O-, -P(O)(ORa)OH, -P(O)(ORa)O-, -P(O)(ORa)(ORb), -OS(O)2OH, -OS(O)2O-, -OS(O)2ORa, -S(O)2OH, -S(O)2O-, -S(O)2ORa, -OS(O)OH, -OS(O)O-, -OS(O)ORa, -S(O)OH, -S(O)O-, -OS(O)-, -S(O)ORa, -OS(O)2-, -OS(O)2Ra, -S(O)2-, -S(O)2Ra, -OS(O)Ra, -S(O)- -S(O)Ra, -OH, -SH, -(OCH2CH2)vOH, -(OCH2CH2)V'O-, -(OCH2CH2)v'ORasugar fragment, oligosaccharide fragment and Oligopeptide fragment, or protonated, or deprotonirovannoi form, and in addition, any combination thereof, where Ra, Rband Rcindependently selected from H and optionally substituted C1-3of alkyl, where two or more of Ra, Rband Rcnot necessarily connected with the formation of one or more of carbocycles or heterocycles, and v'represents an integer selected from 1-100. Water-soluble group can be in any position inside R8, R9, R10and/or R11or can entirely be R8, R9, R10or R11the fragment. Water-soluble group may, for example, the R, to be located in an internal position, may be part of the main circuit, part of a ring structure may be a functional group, is suspended from the main chain or ring, or may be in a position, in which R8, R9, R10or R11Deputy attached to the rest of the molecule. In one embodiment, one of R8, R9, R10and R11contains water-soluble group. In another embodiment, one of R8, R9and R10contains water-soluble group. In another embodiment, R8contains water-soluble group. In another embodiment, R9contains water-soluble group. In another embodiment, R10contains water-soluble group. In one embodiment, the water-soluble group is a carboxylic acid group. In another embodiment, a water-soluble group is the amino group. In yet another embodiment, a water-soluble group is a primary amino group. In another embodiment, a water-soluble group is a secondary amino group. In another embodiment, a water-soluble group is a tertiary amino group. In another embodiment, a water-soluble group is a Quaternary amino group. In one embodiment, the water-soluble group is dimethylaminopropyl. In another the embodiment, a water-soluble group is morpholinopropan. In yet another embodiment, a water-soluble group is a group l-methylpiperidin-4-yl. In yet another embodiment, a water-soluble group is methylaminopropyl. In yet another embodiment, a water-soluble group is a group of the piperidine-4-Il. In yet another embodiment, a water-soluble group is amino (NH2group. In yet another embodiment, a water-soluble group is N-methyl-N-(carboxymethyl)amino group. In yet another variant, the water-soluble group is N-methyl-N-(2-methoxy-2-oxoethyl)amino group. In yet another embodiment, a water-soluble group is not the ether group (-O-, -ORa), regardless of whether it oligoamine or polyether group. In yet another embodiment, a water-soluble group is not an amide group, regardless of whether it Oligopeptide or polypeptide group. In yet another embodiment, a water-soluble group is not the primary, secondary, or tertiary amino group, the nitrogen of which is directly connected (and thus anywhereman) with aromatic fragment, or secondary or tertiary amino group is part of the aromatic fragment, where the nitrogen atom is an atom in the aromatic ring system. In yet another embodiment, a water-soluble group is not hydroc the ilen group, combined with the aromatic ring system. In one embodiment, R8, R9, R10or R11selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R8, R9or R10selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R8selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R9selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1- alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R10selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In one embodiment, R8, R9, R10or R11choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy and 2-(N,N-dimethylamino)acetylamino. In another embodiment, R8, R9or R10choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy and 2-(N,N-dimethylamino)acetylamino. In another embodiment, R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy and 2-(N,N-dimethylamino)acetylamino. In one embodiment, R8is 2-(morpholine-4-yl)ethoxy. In another embodiment, R8represents (1 methylpiperidin-4-yl)methoxy. Even the bottom option R 8is 2-(N,N-dimethylamino)ethoxy. In another embodiment, R8is 2-(N,N-dimethylamino)acetylamino. In another embodiment, R9is 2-(N,N-dimethylamino)ethoxy. In another embodiment, R10is 2-(N,N-dimethylamino)ethoxy. In another embodiment, R8, R9, R10or R11choose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R8, R9, R10or R11choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R8, R9or R10choose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy, and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R8choose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-meth is l-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N - dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R9choose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy, and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R10choose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R8is 2-(methylamino)ethoxy. In another embodiment, R8is 2-aminoethoxy. In another embodiment, R8is 2-(N-methyl-N-(carboxymethyl)amino)ethoxy. In another embodiment, R8is 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. If one of R8, R9, R10or R11contains water-soluble group, the other substituents may each independently represent either hydrogen, or Deputy, containing another odor storemay group or Deputy, containing no water-soluble group. In one embodiment, R9, R10and R11each represents hydrogen. In one embodiment, R8contains water-soluble group, and R9, R10and R11each represents hydrogen. In another embodiment, R8contains water-soluble group, and R9represents methoxy. In another embodiment, R9is methoxy and R10and R11each represents hydrogen. In another embodiment, R8contains water-soluble group, and R9is methoxy and R10and R11each represents hydrogen. In the following embodiment, R8is 2-(N,N-dimethylamino)ethoxy, R9is methoxy and R10and R11each represents hydrogen. In another embodiment, R9or R10present Deputy containing a water-soluble group, and R8is not hydrogen. In another embodiment, R9is Deputy containing a water-soluble group, and R8is not hydrogen. In another embodiment, R10is Deputy containing a water-soluble group, and R8is not hydrogen. In another embodiment, R8choose from where X33selected from O, S and NR15where R15has the above mn of the treatment, Raand Rbhave the above values, and NRaRbconnected to the phenyl ring via any of the carbon atoms of 1-4. In another embodiment, NRaRbconnected to the phenyl ring through the carbon atoms of 2 or 4. R8can also be the Deputy of the formula: where X3'X4'Xs', R8', R9', R10'and R11'have the above values for X3X4Xs, R8, R9, R10and R11accordingly, except that R8', R9', R10'and R11'must not contain a water-soluble group, if the other water-soluble group is already present in R8, R9, R10or R11and where R8"can be selected from H and optionally substituted C1-5the alkyl or C1-5heteroalkyl and not necessarily connected with R9or R11education optionally substituted aliphatic or aromatic heterocycle. In one embodiment, R8or R8"and R11and/or R8'and R11'can be connected with the formation, together with the linking atoms optionally substituted dihydropyrrolo. Compounds of formula (I) and (II) as one of the advantages have increased solubility in comparison with previously known similar with what disiniame. The presence of, for example, carboxylic acid group or a secondary aliphatic amino group can significantly increase the solubility. At the same time, such a group may hinder the penetration of the compounds of formula (I) or (II) through a biological barrier, especially if there is a non-polar barrier, such as the cell membrane. This can be an advantage, especially if the compound of formula (I) or (II) deliver in micheneau cell using conjugation with guides to the target fragment before he would release from the conjugate. If the compound of formula (I) or (II) prematurely released from the conjugate, for example, when the circulation, it may be unable or too little can penetrate (not miseriya) cells specificeski as its ability to membrane translocation may be impaired. This can lead to increased selectivity, and therefore reduce side effects. In one embodiment, R2is not hydrogen. In another embodiment, R2is H and at least one of present R3, R3', R4, R4', R5and R5'does not represent H. In another embodiment, R2represents H, X1is O, and at least one of R3, R3', R4, R4', R5and R5'not submitted the H. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14and at least one of present R3, R3', R4, R4', R5and R5'does not represent H. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X5is About, and at least one of present R3, R3', R4, R4', R5and R5'does not represent H. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14Xsis O, X3is NR15and at least one of present R3, R3', R4, R4', R5and R5'does not represent H. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2represents CH, X5is O, X3represents NH, and at least one of present R3, R3', R4, R4', R5and R5'does not represent H. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X5is O, X3is NR at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8, R9, R10or R11selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8, R9or R10selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14 X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R9selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14 X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R10selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8, R9, R10or R11choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR1 X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8, R9or R10choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14Xsis O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X4is CR16X5/sup> is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14X4is CR16X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'is not H, R10and R11both represent H and R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, in compounds of formula (Ib) or (IIb) R2represents H, X1is O, X2is CR14 X4is CR16X5is O, X3is NR15at least one of present R3, R3', R4, R4', R5and R5'is not H, R10and R11both represent N, R9selected from H and OMe, and R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, R2is H, at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R2is H, at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R9selected from-O-C1-6alkylene-N(R )2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O and (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R2is H, at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R10selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. In another embodiment, R2is H, at least one of present R3, R3', R4, R4', R5and R5'does not represent H, and R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In one Varian is e R 5is not NO2. In another embodiment, at least one of the substituents R1, R2, R3, R3', R4, R4', R5, R5', R6, R6', R7, R7', R8, R9, R10, R11, R12, R13, R14, R15, R16and R17contains or is part of a COOH. In another embodiment, at least one of the substituents R1, R2, R3, R3', R4, R4', R5, R5', R6, R6', R7, R7', R8, R9, R10, R11, R12, R13, R14, R15, R16and R17contains or is a fragment COOH, and if there is only one COOH fragment and the specified COOH fragment is contained in R8, R9, R10or R11there is at least another water-soluble group present in R8, R9, R10or R11. In another embodiment, at least one of the substituents R8, R9, R10and R11contains or is part of a COOH. In another embodiment, at least one of the substituents R8, R9, R10and R11contains or is part of COOH and there is at least another water-soluble group present in R8, R9, R10or R11. In another embodiment, IU the greater extent, one of the water-soluble groups in R8, R9, R10and R11represents aliphatic fragment of a secondary amine, which is not anywhereman with aromatic fragment or a carbonyl group. In another embodiment, R8choose from In yet another variant, at least one water-soluble groups in R8, R9, R10and R11represents aliphatic fragment of a secondary amine, which is not anywhereman with aromatic fragment or a carbonyl group and at least one of the substituents R1, R2, R3, R3', R4, R4', R5, R5', R6, R6', R7, R7', R8, R9, R10, R11, R12, R13, R14, R15, R16and R17contains or is part of a COOH. In yet another variant, at least one water-soluble groups in R8, R9, R10and R11represents aliphatic fragment of a secondary amine, which is not anywhereman with aromatic fragment or a carbonyl group and at least one of the substituents R8, R9, R10and R11contains or is part of a COOH. In yet another variant, at least one of the substituents R8, R9, R10and R11contains COOH fragment and Ali is eticheski fragment of a secondary amine, not anywhereman with aromatic fragment or a carbonyl group. In one embodiment, the compound of the present invention represented by formula (Ib1): or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R8ais Deputy containing a water-soluble group, and R9arepresents H or optionally substituted C1-15alkoxy. In another embodiment, the compound of the present invention represented by formula (Ib2): or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R8a'is Deputy containing a water-soluble group, and R9a'represents H or optionally substituted C1-15alkoxy. In one embodiment, the present invention relates to compounds of formula (Ib1) and (Ib2), where R8aor R8a', respectively, are selected from-O-C1-6alkylene-N(R100)2, -NC(O)-C1-5alkylene-N(R100)2, (1-(R100)piperidine-4-yl)-C1-5alkylen-O - (morpholine-4-yl)-C1-8alkylen-O-, where each R100independently selected from H and C1-3the alkyl, the latter being optionally substituted by COOH or COOR300where R300is1-4alkyl. <> In one embodiment, the present invention relates to compounds of formula (Ib1) and (Ib2), where R8aor R8a'accordingly, selected from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy, and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy.In another embodiment, the present invention relates to the compound of formula (Ib1), where R8achoose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy. In another embodiment, the present invention relates to the compound of formula (Ib1) where R8achoose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy and R9arepresents H. In another embodiment, the present invention relates to the compound of formula (Ib1), where R8achoose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2 aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl--(2-methoxy-2-oxoethyl)amino)ethoxy and R 9arepresents methoxy. In one embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R1ais chlorine (Cl) or bromine (Br), R2aand R5brepresent methyl and H, respectively, or H and methyl, respectively, R8dchoose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy and R9dselected from H and methoxy. In another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention represented by the formula: and the and (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In yet another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In yet another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In yet another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In yet another embodiment, the compound of the present invention represented by the formula: or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention represented by formula (Ib3) or (1R) isomer or a mixture of two isomers, where R5ahave the above values for R5except that it cannot represent hydrogen. In one embodiment, R5ain the compound of formula (Ib3) are selected from nitro, halogen, amino, hydroxy, optionally substituted, alkylamino, optionally substituted, alkylcarboxylic, optionally substituted, alkoxycarbonyl, optionally substituted, alkyloxy, optionally substituted, alkylcarboxylic, optionally substituted, alkylaminocarbonyl and optionally substituted C1-4the alkyl. In another embodiment, R5ain the compound of formula (Ib3) is optionally substituted With linear1-4alkyl. In another embodiment, R5ain the compound of formula (Ib3) is unsubstituted linear C1-4alkyl. In another embodiment, R5ain the compound of formula (Ib3) is methyl. In another embodiment, the compound of the present invention represented by formula (Ib3-1): or (1R) isomer, or a mixture of two isomers, where each R100aindependently represents methyl, carboxymethyl, 2-methoxy-2-oxoethyl or hydrogen. In another embodiment, the compound of the present invention represented by the formula: or (1R) what Zomer, or a mixture of two isomers. In another aspect the present invention relates to compounds of formula (I') or (II'): where all substituents have the values indicated above for the compounds of formulas (I) and (II). Assume that the compounds of formulas (I) and (II) must be transformed in vivo into compounds (I') and (II'), respectively, with concomitant removal of the H-R1that is schematically presented in figure 1 for the compounds of formula (I). Therefore, the present invention relates to compounds of formula (I') or (II'), and these compounds contain cyclopropyl group that formed as a result of regrouping and concomitant removal of the H-R1of the compounds of formula (I) or (II). It should be understood that in this description and the claims when referring to compounds of formula (I) or (II) are also included reference to compounds of formula (I') or (II'), respectively, if only we are not talking about structural parts (I) and (II)that are not present in (I) and (II'), or if the context is not there are other indications. Similarly, if there is a link to the structural part (fragment), conjugate linker agent or conjugate of formula (I) or (II)they include links to similar structural parts (fragments), the conjugated linker agent, or conjugates of formula (I') or (II'), respectively, if only we are not talking about the structure of the business parts of the compounds (I) and (II), not present in (I) and (II'), or if in the context of other instructions. In another aspect, the present invention relates to compounds of formula (VII) or (VIII): or their pharmaceutically acceptable salts or solvate, where R1selected from halogen and OSO2Ruwhere Ruselected from optionally substituted C1-6of alkyl, C1-6perhalogenated, benzyl and phenyl; R2selected from H and optionally substituted C1-8of alkyl; R3, R3', R4and R4'independently selected from H and optionally substituted C1-8of alkyl, where two or more of R2, R3, R3', R4and R4'optionally joined to form one or more optionally substituted carbocycles or heterocycles; X2selected from O, C(R14)(R14'), and NR14'where R14selected from H and optionally substituted C1-8the alkyl or acyl and R14'may be absent or may be selected from H and optionally substituted C1-8the alkyl or acyl; each R5, R5', R6, R6', R7and R7'independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, Rk, SRk, S(O)Rk, S(O)2Rk, S(O)ORk, S(O)20Rk , OS(O)Rk, OS(O)2Rk, OS(O)ORk, OS(O)2ORk, ORk, OtherkN(RkRL,+N(Rk)(RLRm, P(O)(ORk)(ORL), OP(O)(ORk)(ORL), SiRkRLRmWith(O)RkWith(O)ORkWith(O)N(RLRk, OC(O)Rk, OC(O)ORk, OC(O)N(RkRLN(RL)C(O)RkN(RL)C(O)ORkand N(RL)C(O)N(RmRkwhere Rk, RLand Rmindependently selected from H and optionally substituted C1-4of alkyl, C1-4heteroalkyl, C3-7cycloalkyl, C3-7geterotsiklicheskie, C4-12aryl, or C4-12heteroaryl, where two or more of Rk, RLand Rmoptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, and/or R5+ R5'and/or R6+ R6'and/or R7+ R7'independently represent =O, =S, or =NR12and R12selected from H and optionally substituted C1-6of alkyl, and/or R5'and R6'and/or R6'and R7'and/or R7'and R14'no, that means that a double bond is present between tomamae, which are attached to R5'and R6'and/or R6'and R7'and/or R7'and R14'accordingly, two or more of R , R5', R6, R6', R7, R7', R14and R14'optionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles; X1selected from O, S, and NR13where R13selected from H and optionally substituted C1-8of alkyl; X3selected from O, S, and NR15where R15selected from H and optionally substituted C1-8the alkyl or acyl, or-X3- represents-X3aand X3b-where X3ais connected by a carbon atom that is attached to X4and X3bconnected to the phenyl ring in ortho-position to R10where X3aindependently selected from H and optionally substituted C1-8the alkyl or acyl and X3bselected from the same group of substituents, and R8; X4selected from N and CR16where R16selected from H and optionally substituted C1-8the alkyl or acyl; X5selected from O, S, and NR17where R17selected from H and optionally substituted C1-8the alkyl or acyl; R8, R9, R10and R11each independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, RX, SRx, S(O)Rx, S(O)2Rx, S(O)ORx, S(O)2ORx, OS(O)Rx, OS(O 2Rx, OS(O)ORx, OS(O)2ORx, ORx, OtherXN(RxRy,+N(Rx)(RyRz, P(O)(ORx)(ORy), OP(O)(ORx)(ORy), SiRxRyRZWith(O)RxWith(O)ORxWith(O)N(RyRx, OC(O)Rx, OC(O)ORx, OC(O)N(RxRyN(Ry)C(O)RxN(Ry)C(O)ORxN(Ry)C(O)N(RzRxand water-soluble group, where Rx, Ryand RZindependently selected from H and optionally substituted C1-15of alkyl, C1-15heteroalkyl, C3-15cycloalkyl, C3-15geterotsiklicheskie, C4-15aryl, or C4-15heteroaryl, and one or more optional substituents in Rx, Ryand Rzoptionally present water-soluble group, and two or more of RXRyand Rzoptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, and at least one of R8, R9, R10and R11includes at least one water-soluble group, two or more of R8, R9, R10, R11or X3boptionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles; a and b independently wybir the Ute from 0 and 1; c is chosen from 0, 1 and 2; provided that a) at least one of the substituents R1, R2, R3, R3', R4, R4', R5, R5', R6, R6', R7, R7', R8, R9, R10, R11, R12, R13, R14, R15, R16and R17contains or is a fragment COOH, and if there is only one COOH fragment and the specified COOH fragment is contained in R8, R9, R10or R11there is at least another water-soluble group present in R8, R9, R10or R11and/or b) at least one water-soluble groups in R8, R9, R10and R11represents aliphatic fragment of a secondary amine, which is not anywhereman with aromatic fragment or a carbonyl group and/or c) R8choose from and/or d) R9is OMe and R8is OCH2CH2N(CH3)2. Compounds of formulas (VII) and (VIII) have a solubility that is higher than the solubility of the same compounds, formerly known. Conjugates of the compounds of formulas (VII) and (VIII) may therefore have improved properties compared with the conjugates of the same compounds, formerly known. Conjugates of formula (VII) and (VIII) can, for example, the R, to have high solubility, may demonstrate less ability to aggregation, and show fewer side effects as premature compounds of formula (VII) or (VIII) of such conjugates before such conjugate penetrates into micheneau cell, releases the connection, which is less efficiently penetrates into cells, including not miseriya cells, due to its high polarity compared with compounds having no water-soluble group. The optional presence of a carboxylic acid group or a secondary aliphatic amino group in the compounds of formula (VII) or (VIII), in addition, provides a convenient (optional) possibility to obtain conjugates. It should be noted that the assumed properties and options for the above compounds of formulas (I) and (II)also apply to compounds of formulas (VII) and (VIII), unless they involve structural parts of the compounds (I) and (II)that are not present in (VII) and (VIII), or in the context States otherwise. Therefore, it is necessary to understand that in this description and the claims, when referring to the compounds of formula (I) or (II)they include references to compounds of formula (VII) or (VIII), respectively, if only we are not talking about the structural parts of the compounds (I) and (II)that are not present in (VII) and (VIII), Il is in the context States otherwise. Similarly, if we are talking about structural part (fragment), the conjugate linker-agent or conjugates of formula (I) or (II)they include links to similar structural parts (fragments), the conjugated linker-agent or conjugates of formula (VII) or (VII), respectively, if only we are not talking about structural parts (I) and (II)that are not present in (VII) and (VIII) or in the context States otherwise. Similarly, if you are referring to compounds of formula (I'or (II'), it should be understood that they include references to the containing cyclopropyl analogs of compounds (VII) or (VIII), if only we are not talking about structural parts (I) and (II'), not present in the containing cyclopropyl analogues (VII) and (VIII) or in the context States otherwise. Conjugates and conjugates of linker-agent In another aspect, the present invention relates to conjugates of the compounds of formula (I) or (II)that can be transformed in vivo into compounds of formula (I) or (II) in one or more stages, respectively. These conjugates may favorably affect the pharmacokinetic properties and other characteristics of the compounds of formula (I) or (II). In one embodiment, the present invention relates to a conjugate comprising the compound of formula (I) or (II)conjugated with at least one prefragmented, i.e. a fragment that can be removed in vivo emission of compounds of formula(I) or (II). In another embodiment, the present invention relates to a conjugate comprising the compound of formula (I) or (II)conjugated with one prefragments. In another aspect, the present invention relates to the compound of formula (III): or its pharmaceutically acceptable salt, or MES, where V2or is absent, or represents a functional fragment; each L2independently absent or represents a linking group linking V2with L or V1or Y, if L is absent; each L is independently absent or represents a linking group linking the L2or V2if L2no, with one or more of V1and/or Y; each V1independently represents H or conditionally ottsepleny, or conditionally-transformed fragment, which can be split or transform, using a chemical, photochemical, physical, biological, or enzymatic process. each Y is independently absent or is self-destructing spacer elements of the system, which consists of 1 or more of the self-destructing spacers and connected with V1not necessarily L, and one or more Z; each of p and q is the number representing the degree of branching, and each independently represents a positive integer with the number; z represents a positive integer equal to or less than the total number of sites attach to Z in one or more of V1-Y fragments; each Z is independently a compound of formula (I) or (II)as above, where one or more of the X1, R6, R7, R8, R9, R10and R11may, optionally, additionally be substituted by the Deputy of the formula (V): where each V2'L2', L', V1', Y', Z', p', q' and z' have the above values for V2L2, L, V1, Y, Z, p, q, and z, and one or more of the substituents of formula (V) is independently connected to one or more of X1, R6, R7, R8, R9, R10and R11through Y' or V1'if Y' is absent, each Z is connected c Y or V1if Y is absent, or through the X1or via an atom in R6, R7, R8, R9, R10or R11; provided that at least one of the one or more of V1and one or more of V1'does not represent H. It should be understood from the formula (III)that L can be connected or V1and/or Y. If L is connected to Y, this means that both V1and L, as well as one or more of Z, is connected with the Y. If Y is absent, L must be connected to V1; L cannot be directly connected to Z. V 2(-L2-L(-V'-Y)p)q(Z)z-1and one or more of V2'(-L2'-L'(-V1'-Y')P')q'(Z')z'-1fragments connected with the compound of the formula (I) or (II)in the description called prefragments. The present invention also relates to the compound of formula (IV): or its pharmaceutically acceptable salt, or MES, where RM represents a reactive fragment and L, V1, Y, Z, p and z have the above values, except that L now connects RM with one or more of V1and/or Y, and V1, Y and Z may contain a protective group, and one or more of V2'-L2'fragments, not necessarily present in Z, as described above, may optionally and independently be replaced by RM', which is reactive fragment, and where, if there is more than one reactive fragment in (IV), reactive fragments of the same or different. These conjugates linker agent can or cannot be considered as intermediate compounds for the compounds of formula (III). RM-L(-V1-Y)p(Z)z-1and one or more of the RM'-L'(-V1'-Y')P'(Z')z'-1fragments connected with the compound of the formula (I) or (II) in this description, referred to as prefragmented. It should be understood that h is about considering the invention relates to enantiomerically pure and/or diastereoisomers pure compounds of formulae (III) and (IV) as well as enantiomeric and/or diastereoisomeric mixtures of compounds of formulas (III) and (IV). If one or more of Y and/or V1fragments in the compounds of formula (III) or (IV) contains sites attach to Z, which is not attached to Z, for example as a result of incomplete reaction joining in the synthesis process, consider that these sites attach attached instead to H, OH or tsepliaeva group. If all of these sites joining connected with Z, then z is equal to the number of these sites connection; otherwise, z is smaller. Compounds of the present invention may exist in the form of a mixture where each component of the mixture has a different z-values. For example, the compounds can exist as a mixture of two separate connections, one connection z=4, and the other connection z=3. In addition, for a specific value z of the compounds can exist as a mixture of isomers, since Z can be connected with different sets of sites joining in one or more of Y and/or V1the fragments. For clarity, if there is a link to the connection of one of the first fragments with other fragments in formulas (III) or (IV), usually referred to only those other specified fragments, which immediately follow the specified first fragment in formulas (III) or (IV). It should be understood that if oneof these other fragments missing, the first fragment is actually connected to the fragment, which is the first audience, unless otherwise indicated. For example, if you specify that "V1otscheplaut of Y", this phrase actually means "V1otscheplaut of Y, or Z, if Y is missing," and then it should read "V1otscheplaut of Z, if we are talking about the connection, in which Y is absent. In the compounds of formula (III) or (IV), compounds of formula (I) or (II) can be conjugated with prefragmented due to its water-soluble group. In this case, a water-soluble group may make a smaller contribution to the solubility of the compounds of formula (III) or (IV), but can again contribute to the water-solubility of Z after the removal of the specified prefragments. In this paper, wherever mentioned V2L2, L, V1, Y, Z, RM, p, q, or z, you need to understand that the same applies to each of the V2'L2', L', V1', Y', Z', RM', p', q' and z', respectively. V1fragment In the compounds of formula (III) or (IV) (V1the fragment may represent a group that is conventionally otseplena or transformed. In other words, it is designed to be transformed and/or tsepliaeva of Y under the action of a chemical, photochemical, physical, biological or fermentative processes after the CSOs, as flagged in some conditions. Such conditions can be, for example, the compounds of the present invention in the aquatic environment, which leads to hydrolysis of V1or the location of the connection of the present invention in an environment that contains an enzyme that recognizes and it V1or the location of the connection of the present invention in reducing conditions, which leads to the recovery and/or removal of V1or conversion of a compound of the present invention in contact with radiation, for example, under the action of UV radiation, which leads to transformation and/or cleavage, or heating compounds of the present invention, which leads to transformation and/or cleavage, or the location of the connection of the present invention in conditions of reduced pressure, which leads to transformation, for example, to heterocyclisation, and/or the elimination of, or the location of the connection of the present invention in conditions of high pressure or high pressure, which leads to transformation and/or cleavage. Such conditions may occur after administration of the compounds of the present invention to an animal, e.g. a mammal, such as man, these conditions may occur if the connection is localized, for example, in a particular organ, tissue, cell, sub-cellular target or microbial targets, for example, in the presence of the of current factors (for example, machinespecific enzymes or hypoxia) or external factors (for example, under the action of radiation, magnetic fields) or the above conditions can occur immediately after the injection (for example, under the action of ubiquitary enzymes). Usually the transformation V1directly or indirectly leads to the elimination of V1of Y. the Connection of the present invention may contain more than one V1the fragment on promiment (p and/or q>1). These V1fragments may or may not be the same or may require, or may not require the same conditions for transformation and/or cleavage. In one aspect of the present invention, the conjugate is used for targeting one or more of the fragments Z miseriya cells. In this case, V1the fragment may, for example, contain a substrate molecule that is cleaved by the enzyme present in proximity to Milenium cells or inside nesenevich cells, e.g. tumor cells. V1may, for example, contain a substrate that hatshepsuts under the action of an enzyme present in high quantities in the vicinity of or inside nesenevich cells compared with other parts of the body, or under the action of the enzyme, which is present only near or inside nesenevich cells. It is important to realize that if the specificity nesenevich cells is achieved only on the basis of selective transformation and/or cleavage of the specified V 1in misheneva the website, the conditions that cause banding, should preferably be at least to some extent, specific to nesenevich cells, whereas the presence of other machinespecific fragments in the compound of the present invention, for example in the V2fragment reduces or eliminates this requirement. For example, if V2causes selective internalization in miseriya cells, the enzyme is also present in other cells, can transform and/or split V1. In one embodiment, the transformation and/or cleavage V1is intracellular. In another embodiment, transformation and/or cleavage V1is extracellular. In one embodiment, V1contains di-, tri-, Tetra -, or Oligopeptide consisting of the amino acid sequence recognized a proteolytic enzyme, such as plasmin, a cathepsin, cathepsin B, prostatespecific antigen (PSA), a plasminogen activator of the urokinase-type (u-PA) or a member of the family of matrix metalloproteinases present in the vicinity or inside nesenevich cells, e.g. tumor cells. In one embodiment, V1represents the peptide. In another embodiment, V1represents a dipeptide. In another embodiment, V1is a Tripeptide. In another embodiment, V1is chatrapati is. In yet another variant V1is peptidomimetic. In another embodiment, V1contains β-glucuronide, which is recognized by β-glucuronidase present near or inside the tumor cell. In one embodiment, V1contains a substrate for the enzyme. In one embodiment, V1contains a substrate for extracellular enzyme. In another embodiment, V1contains a substrate for intracellular enzyme. In yet another variant V1contains a substrate for the lysosomal enzyme. In yet another variant V1contains a substrate for plasmin of semipretioase. In yet another variant V1contains a substrate for one or more of cathepsins, such as cathepsin B. In yet another variant V1contains a substrate for galactosidase. If V1otscheplaut extracellular, one or more of the Z fragments can be isolated extracellular. This may provide the advantage that the Z fragments are not only able to influence the cell (cell), directly surrounding the site of activation (for example, mistapasifika cells, but also cells that are at some distance from the site of activation (for example, misintegration cells) due to diffusion (the observer effect). The enzyme that it V1can also be transported near or inside nesenevich cells or mistaway the fabric, for example, proletarienne therapy using antibody directed enzyme (ADEPT), proletarienne therapy using a directed polymer-enzyme (PDEPT) or proletarienne therapy using-directed macromolecule enzyme (MDEPT), proletarienne therapy using directed by virus enzyme (VDEPT) or proletarienne therapy using directed enzyme gene (GDEPT). In one embodiment, the transformation and/or cleavage V1occurs due to the enzyme linked to the antibody. And again in another embodiment, V1contains a fragment, such as nitro(hetero)aromatic fragment that can be transformed and/or derived by a recovery in hypoxia or by restoring using nitroreductase. After nitrogroup reduction and cleavage of the resulting fragment removing the spacer elements of the system Y, if present, leads to the selection of one or more of the fragments Z. In one embodiment under consideration, the invention relates to a compound in which V1represents a monosaccharide, disaccharide or oligosaccharide from hexoses or pentoses or Leptos, which can also be included in the group deoxysaccharides or amino sugars and belong to the D-range or L-range and disaccharides or oligosaccharides od is nakova or different. Such V1usually cleaved by glycosidases. In another embodiment, V1has the formula C(ORa'Rb'CHRc'Rd'where Ra'is hydroxyamino group, optionally substituted C1-8alkyl group, which optionally forms a ring with R5, R5', R6, R6', R7, R7', R14or R14'or monosaccharide, disaccharide glycosides or oligosaccharide residue, optionally substituted methyl group, hydroxymethylene group and/or a radical of the formula-NRe'Rf'where Re'and Rf'the same or different and are selected from hydrogen, C1-6the alkyl or aminosidine group and where the hydroxy-group sharidny residues optionally protected; Rb', Rc'and Rd'independently selected from hydrogen, optionally substituted C1-8the alkyl or C1-8cycloalkyl, or a group of the formula-NRe'Rf'where Re'and Rf'have the above values. In one embodiment under consideration, the invention relates to a conjugate in which V1represents a dipeptide, Tripeptide, tetrapeptide or Oligopeptide fragment consisting of natural L amino acids, not natural D-amino acids or synthetic amino acids or peptidomimetics, or any combinations thereof. In another embodiment, rassmatrivaemaya refers to the connection, in which V1includes the Tripeptide. The Tripeptide may be linked via its C-end with Y. In one embodiment, the residue C-terminal amino acids of Tripeptide selected from arginine, citrulline and lysine, and the rest of the middle amino acid Tripeptide selected from alanine, valine, leucine, isoleucine, methionine, phenylalanine, cyclohexylglycine, tryptophan and Proline, N-terminal amino acid Tripeptide choose from any natural or not natural amino acids. In another embodiment under consideration, the invention relates to a compound in which V1includes dipeptide. The dipeptide can be linked via its C-end with Y. In one embodiment, the residue C-terminal amino acids of the dipeptide is selected from alanine, arginine, citrulline, and lysine and N-terminal amino acids of the dipeptide selected from any natural or not natural amino acids. In one embodiment, if the α-amino group of the N-terminal amino acids of V1not connected with L, the amino acid may be functionalized with a suitable blocking group, linked to the α-amino group or may be a natural amino acid, which prevents unwanted premature decomposition V1under the action of, for example, ubiquitary enzymes or ectopeptidases. In the next version V1choose from D-alanylhistidine, D-in illazilla, D-alanylhistidine, D-valentinevalentine, D-valilehtipainikkeen, D-halonitrobenzenes, albertinelli, vallillesia, alanylhistidine, valentinevalentine, valilehtipainikkeen, halonitrobenzenes, D-alanylhistidine, D-wallacestone, D-alanylhistidine, D-valentinevalentine, D-valilehtivalitsimen, D-halonitrobenzenes, alanylhistidine, wallacestone, alalalalalong, valentinevalentine, valilehtivalitsimen and halonitrobenzenes. In yet another variant V1choose from fenilalanina, valiligine, villania, D-phenylenedimaleimide, phenylenedimethylene, gliciltirosin, alanalysis, wallcarolina, N-metiletilchetone, phenylalkylamine, IsolatedStorage, triptorelin, tryptophansynthroid, fenilalkilamina, phenylalkylamine, glycolpolyvinylpyrrolidone, alalalalalalala, alalalalalalala, i.e. phenylalanyl-N9-tilarginine, i.e. phenylalanyl-N9-nitroarginine, lallison, laicization and i.e. phenylalanyl-O-benzotriazine. In the next version V1choose from fenilalanina, valiligine and wallcarolina. Therefore, in one variations which those present invention relates to a connection, in which V1contains a substrate that can be cleaved by proteolytic enzyme, plasmin, a cathepsin, cathepsin B, β-glucuronidase, galactosidase, prostatespecific antigen (PSA), a plasminogen activator of the urokinase-type (u-PA), a member of the family of matrix metalloproteinases, or an enzyme localized by direct enzymatic proletarienne therapy, such as ADEPT, VDEPT, MDEPT, GDEPT, or PDEPT, or if V1contains a fragment that can be split or transform, using a recovery in hypoxia or using recovery nitroreductases. In another aspect of the present invention, the conjugate of the present invention is used for (also) improvements (pharmacokinetic) properties Z. If there is no need for selective removal of prefragments at mesiniaga site, V1specified prefragments may be, for example, a group, or may contain a group which is cleaved ubiquitary enzymes, for example, a complex virasami that are present in the circulating or intracellular enzymes, such as protease and phosphatase, due to the pH-controlled intramolecular cyclization or by hydrolysis catalyzed by acid, base, or hydrolysis without catalyst, or V1may be, for example, or may contain the disulfide or to form a disulfide with neighboring fragment. Therefore V1may optionally together with the linking atom (atoms) L and/or Y, for example, to form carbonate, urethane, urea, ester, amide, imine, hydrazono, oximo, disulfide, acetamino or catalog group. This means that V1not necessarily together with the connecting atom (atoms) L and/or Y may, for example, also be-OC(O)-, -C(O)O-, -OC(O)O-, -OC(O)N(Rd)-, -N(Rd)C(O)-, -C(O)N(Rd)-, -N(Rd)C(O)O-, -N(Rd)C(O)N(Re)-, -C(O)-, -OC(Rd)(Re)-, -OC(Rd)(Re)O-, -OC(Rd)(Re)O-, -C(Rd)(Re)-, -S-, -S -,- C=,= -,- N=, =N-, -C=N-, -N=C-, -O-N=, =N-O-, -C=N-O-, -O-N=C-, -N(Rf)-N=, =N-N(Rf)-, -N(Rf)-N=C - or-C=N-N(Rf)-, where Rd, Reand Rfindependently selected from H and optionally substituted C1-10the alkyl or aryl, and two or more of Rd, Reand Rfconnected to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles. If V1or V1-Y represent preferment, or L is connected to Y and not with V1, V1it is possible, in this case to choose from, for example, Rg- [(RnO)P(O)]pp-, Rg-C(O)-, Rg-OC(O)- and Rg-N(Rn)C(O)-, where pp is chosen from 1-3, and each Rgand Rnindependently selected from H and optionally substituted C1-15 of alkyl, C1-15heteroalkyl, C1-15cycloalkyl, C1-15geterotsiklicheskie, C4-15aryl, or C4-15heteroaryl, Rgand Rnnot necessarily connected with education optionally substituted carbocycle or heterocycle. In one embodiment, V1choose from phosphono, phenylenecarbonyl, 4-(piperidino)piperidinylcarbonyl, piperazinylcarbonyl and 4-methylpiperidine. It should be noted that V1or in the form of mono-, di - or oligosaccharide fragments, or in the form of di-, tri-, Tetra - or oligopeptides, or in any other form, may contain a protective group. Compounds of the present invention, including such protected V1may not allocate any Z fragments, if they are placed in the conditions under which transforms and/or cleaved corresponding unprotected V1. However, if you remove the protective group in these compounds, they will select one or more of the Z fragments, if they are placed in the appropriate conditions. Connections, including such protected V1also fall in the scope of the present invention. Especially the above can be predicted for compounds of formula (IV). Suitable protective groups for functional groups, in particular amino acids, well known to chemists organikum, and they can be found, for example, in T.W. Greene, Protective Groups in Organc Synthesis, John Wiley & Sons, New York, 1981. Compounds of formulas (III) and (IV) ultimately created to highlight the compounds of formula (I) or (II)or compounds of formula (I') or (II'), after transformation and/or cleavage of one or more of V1and V1'the fragments. The emission of compounds of formula (I) or (II), compounds of formula (I') or (II'), or derivatives thereof, of the conjugate of the present invention through a different mechanism, however, is not excluded from the present invention. In another aspect of the present invention the compound of formula (III) represents an intermediate compound for producing compounds of formula (I) or (II) or other compounds of formula (III). In this case, for example, V2L2, L, and Y are absent, p, q, and z all represent 1, and V1the fragment can be a protective group. May exist and may not exist in one or more of V2'(-L2'-L'(-V1'-Y')p')q'(Z')z'-1fragments, in which V2'L2', L' and Y' may be absent or may be present, and p', q', and z', all may or may not represent 1. In one embodiment, the compound of formula (III) represents compounds of formula (I) or (II), which are attached to V1the fragment. In another embodiment, the compound of formula (III) is of formula (I) or (II), which are attached to V1fragment and V2'(-L2'-L'(-Vl'-Y')P')q'(Z') the fragment. In yet another embodiment, the compound of formula (III) represents compounds of formula (I) or (II), which are attached to V1fragment and V1'the fragment. In one embodiment, V1does not represent a protective group. In another embodiment, V2L2, L, and Y are absent, and p, q, and z all represent 1. In the next version V1is chemically a group to delete. Even the next version V1is chemically removed a group connected with Z by X1. In yet another embodiment, V1represents a benzyl group connected with Z by X1. In one embodiment, V1connected to L via more than one functional group on V1. In one embodiment, V1connected to L via a single functional group at V1. In one embodiment, V1connected to L via a functional group in the side chain of one of the natural or not natural amino acids V1. In another embodiment, N-terminal amino acid V1linked via its α-amino group with L. Self-destructing spacer elements system Y Self-destructing spacer elements system Y, if present, binds V1and optional L with one or more of the fragments Z. Self-destructing spacer elements system Y can be embedded in a conjugate of the present invention to, for example, is ussite properties Z or conjugate as a whole, to ensure appropriate acceding chemical compounds, and/or to create a space between the V1and Z. The compound of the present invention may contain more than one spacer elements system Y on prefragments. These fragments Y may be the same or different. After removal or transformation V1left side Y can be unlocked, which will lead to the eventual selection of one or more of the fragments Z. self-destructing spacer elements of the system can, for example, be those that are listed in WO 02/083180 and WO 2004/043493, which is included here for your reference in its entirety, as well as other self-destructing the spacers, known to specialists in this field. In one aspect the present invention relates to compounds in which Y is chosen from (W-)W(X-)X(A-)S (W-)w(X-)xC((A)s-)ror (W-)w(X-)xC(D((A)s-)d)ror (W-)w(X-)xC(D(E((A)s-)e)d)ror (W-)w(X-)xC(D(E(F((A)s-)f)e)d)r where W and X each represents 1,2+2n electronic cascade spacer (n≥1) single selection, and they may be the same or different; And is ω-amino aminocarbonyl cyclization the spacer, which forms a cyclic Ave is izvozna urea after cyclization; C, D, E and F, each is self-destructing the spacer or spacer elements system multiple selection, which after activation can best allocate r, d, e and f groups, respectively; s represents 0 or 1; r, d, e and f are numbers representing degree of branching; w and x are numbers representing degree of polymerization; r, d, e and f independently represent integers from 2 (inclusive) to 24 (inclusive); w and x independently represent integers from 0 (inclusive) to 5 (inclusive). The following aspect of the present invention self-destructing spacer elements of the system multiple selection C, D, E and F independently are selected from fragments of the formula: where B is selected from NR21, O and S; P is C(R22)(R23)Q-(W-)W(X-)X; Q is absent or is-O-CO-; W and X each represents 1,2+2n electronic cascade spacer (n≥1) single selection, and they may be the same or different; G, H, I, J, K, L, M, N and O independently selected from fragments of the formulas: G, J and M can also be selected from group P, and hydrogen, provided that if two of G, J and M are hydrogen, the remaining group should be or and at the same time to be conjugated with R21selected from H and optionally substituted C1-6of alkyl; R22, R23, R24and R25independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, RX, SRX, S(O)Rx, S(O)2Rx, S(O)ORx, S(O)2ORx, OS(O)Rx, OS(O)2Rx, OS(O)ORx, OS(O)2ORXORx, OtherXN(RXRX1,+N(Rx)(RxlRx2, P(O)(ORx)(ORx1), OP(O)(ORx)(ORx1), C(O)RxWith(O)ORxC(O)N(Rx1Rx, OC(O)Rx, OC(O)ORx, OC(O)N(RxRx1N(Rx1)C(O)RxN(Rx1)C(O)ORxand N(Rx1)C(O)N(Rx2Rxwhere RX, RX1and RX2independently selected from H and optionally substituted C1-6of alkyl, C1-6heteroalkyl, C3-20cycloalkyl, C3-20geterotsiklicheskie, C4-20aryl or C4-20heteroaryl, and RX, Rx1and RX2optionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, two or more of the for the of estately R 21, R22, R23, R24and R25not necessarily connected to each other to form one or more aliphatic or aromatic cyclic structures; g, h, i, j, k, l, m, n, o, h', g', k', j', n', m' are numbers representing degree of branching, and independently represent 0, 1 or 2 under conditions that if G = hydrogen or P, g, h, i, h', and g' are all 0; if J = hydrogen or P, j, k, 1, k'and j' are all 0; if M = hydrogen or P, m, n, o, and n'and m1all 0; if G, H, I, J, K, L, M, N, or O represent then g+g'=1, h+h'=1, i=1, j+j'=1, k+k'=1, l=1 , m+m'=1, n+n'=1 or o=1, respectively; if G, H, I, J, K, L, M, N, or O represent then g+g'=2, h+h'=2, i=2, j+j'=2, k+k'=2, l=2, m+m'=2, n+n'=2, o=1, respectively; if g'=0 and G is not hydrogen or P, then h, h', and i is equal to 0, and g>0; if g=0 and G is not hydrogen or P, then g'>0; if g'>0 and h'=0, then i=0 and h>0; if g'>0 and h=0, then h'>0 and i>0; if j'=0 and J is not hydrogen or P, then k, k', and l is 0, and j>0; if j=0 and J is not hydrogen or P, then j'>0; if j'>0 and k=0, then l=0 and k>0; if j'>0 and k=0, then k'>0 and l>0; if m'=0 and M is not hydrogen or P, then n, n', and o is 0 and m>0; if m=0 and M is not hydrogen or P, then m'>0; if m'>0 and n'=0, then o=0 and n>0; if m'>0 and n=0, then the n'> 0 and o>0; w and x represent the number of polymerization and independently are integers from 0 (inclusive) to 5 (inclusive). In accordance with the following variant of the present invention 1,2+2n electronic cascade spacers W and X are independently selected from fragments following formula: where Q' = -R30C=CR31-, S, O, NR31, -R31C=N-, or-N=CR31-; B = NR32, O, S; P = C(R28)(R29)Q; R26, R27A , B, and (T-)t(T')t'(T-)tP connected with CaCbCcand Cdso that B and (T-)t(T')t'(T-)tP is bonded to two adjacent carbon atoms or Caand Cd; Q is absent or is-O-CO-; t, t' and t" are numbers representing degree of polymerization, and independently are integers from 0 (inclusive) to 5 (inclusive); T, T' and T" are independently selected from fragments of the formulas: R26, R27, R28, R29, R30, R31, R32, R33and R34independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, RX, SRx, S(O)Rx, S(O)2Rx, S(O)ORx, S(O)2ORx, OS(O)Rx, OS(O)2Rx, OS(O)ORx, OS(O)2ORxORx , OtherXN(RXRX1,+N(RX)(RX1RX2, P(O)(ORx)(ORx1), OP(O)(ORx)(ORx1), C(O)RXWith(O)ORxWith(O)N(Rx1Rx, OC(O)Rx, OC(O)ORx, OC(O)N(RxRx1N(Rx1)C(O)RxN(Rx')C(O)ORxand N(Rx1)C(O)N(Rx2Rxwhere RX, RX1and Rx2independently selected from H and optionally substituted C1-6of alkyl, C1-6heteroalkyl, C3-20cycloalkyl, C3-20geterotsiklicheskie, C4-20aryl, or C4-20heteroaryl, Rx, Rx1and Rx2optionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles, two or more of the substituents R26, R27, R28, R29, R30, R31, R32, R33or R34not necessarily connected to each other to form one or more aliphatic or aromatic cyclic structures. In the above formulas, Q can be O-CO, but it can also be absent. For example, it was reported that the connection with benzylamino the relationship between self-destructing the spacer and the group that is cleaved, oxycarbonyl function of no is e (Q is absent), undergoes destruction11. In accordance with the following variant of the present invention ω-amino aminocarbonyl cyclization destroy the spacer is A fragment of the formula: where a represents the integer 0 or 1; b represents an integer 0 or 1; c represents an integer of 0 or 1, provided that a+b+c=2 or 3; R35and R36independently selected from H and optionally substituted C1-6of alkyl; R37, R38, R39, R40, R41and R42independently selected from H, OH, SH, NH2N3, NO2NO, CF3, CN, C(O)NH2With(O)H, C(O)OH, halogen, RX, SRx, S(O)Rx, S(O)2Rx, S(O)ORx, S(O)2ORx, OS(O)Rx, OS(O)2Rx, OS(O)ORx, OS(O)2ORxORx, OtherXN(Rx)Rx1,+N(RX)(Rx1Rx2, P(O)(ORx)(ORx1), OP(O)(ORx)(ORx1), C(O)RxWith(O)ORxWith(O)N(Rx1Rx, OC(O)Rx, OC(O)ORx, OC(O)N(RxRx1N(Rx1)C(O)RxN(Rx1)C(O)ORxand N(Rx1)C(O)N(Rx2Rxwhere RX, RX1and Rx2independently selected from H and optionally substituted C1-61-6heteroalkyl, C3-20cycloalkyl, C3-20geterotsiklicheskie, C4-20aryl, or C4-20heteroaryl, Rx, Rx1and Rx2optionally joined to form one or more optionally substituted aliphatic or aromatic carbocycles or heterocycles and R35, R36, R37, R38, R39, R40, R41and R42and do not necessarily represent the part of one or more optionally substituted aliphatic or aromatic cyclic structures, with two or more of the substituents R35, R36, R37, R38, R39, R40, R41or R42not necessarily connected to each other to form one or more aliphatic or aromatic carbocycles or heterocycles. In one embodiment, the present invention relates to the compound, where X1is O and Y is connected to X1through the ω-amino aminocarbonyl cyclization the spacer forming part Y. In one embodiment, the spacer elements of the system Y choose from In another embodiment, spacer elements, the system Y is In another embodiment, spacer elements, the system Y is Other examples of self-destructing spacers include, but are not on ranciato, the spacers, which can undergo cyclization, such as optionally substituted amides of 4-aminobutyric acid, suitably substituted bicyclo[2.2.1] bicyclo[2.2.2] ring system and amides of 2-aminophenylamino acid and trimethyl-lock" cyclization the spacers13. Glycine spacer, in which Listeria tsepliaeva group attached to the α-position, is another useful spacer for compounds of the present invention.14 In the conjugates of the present invention spacer elements system Y can be connected with more than one V1the fragment. In this case, the transformation and/or cleavage of one of these V1fragments can stimulate the secretion of one or more of the Z fragments. If V1fragments that are transformed or hatshepsuts in various conditions, connected with the same Y, the selection of one or more of the Z fragments can occur if the conjugate of the present invention is one of several different conditions. Alternatively, you can use the spacer elements of the system Y, which requires that it stimulated twice or even several times to self-destruct. An example of such self-destructing spacer is bichenovii the spacer.15If such a spacer is used in combination with different selectionmodelabel V 1fragment connected with the specified spacer, selectivity allocation Z can be increased as may be necessary coincidence of two different conditions, before an allocation will occur Z. The linking group L The linking group L associates one or more of V1and/or Y fragments with L2or RM. The synthesis may be more simple if L is connected with V1and the connection may be less prone to premature decomposition. Joining L to Y may have the advantage that transform and/or split V1will be easier. Another reason for joining L to Y can be, for example, that (part of) Y remains associated with L after removal of V1that prevents the release of reactive small molecules, or that the connection demonstrates superior (pharmacokinetic) properties of solubility and ability to aggregation. L can be a bond connecting V1or Y directly or L2or RM. In another aspect, however, L represents a linking group, which functionally connects or separates one or more of the fragments V'/Y and L2or RM fragment. In the compound of formula (IV), spatial separation can make reactive fragment RM more accessible for the reaction partner, for example, if a functional fragment connec is N. In the compound of formula (III), spatial separation can provide better availability V1since V2next, remove that, especially in the case of enzymatic cleavage or transformation V1may increase the speed with which the transformation and/or cleavage V1. The linking group L may be a water-soluble fragment, or may contain one or more of the water-soluble fragments, such that L has brought its contribution to the solubility of the compounds of formula (III) or (IV). L can also be a fragment or contain one or more of the fragments that reduce aggregation of the compounds of formula (III) or (IV), which may or may not be framenum/fragments, which(e) also increases (increase) the solubility of the compounds of formula (III) or (IV). The linking group L must contain appropriate functional groups at both of its ends, to provide selective connection of one or more of V1and/or Y fragments and L2or RM. In one aspect, L fragment represents a linear, branched or dendritic fragment, so it may not necessarily be connected with more than one V1and/or Y fragment. Branching can be placed through one or more of the cyclic structures or one or more of the atoms branching, which is there to be for example, carbon, nitrogen, silicon or phosphorus. The number of branches in L, which are connected with V1and/or Y will not necessarily be equal to the total number of branches, as in the reaction of joining V1/Y not all branching can join V1and/or Y fragments due to incomplete chemical transformations. This means that L can contain branching, which is not attached to V1or Y, but instead end, for example, a functional group, H, OH or tsepliaeva group. Therefore, if L is branched, the compounds of the present invention may exist in the form of a mixture where each component of the mixture has a different value of p. For example, the compound may exist as a mixture of two separate compounds, where one connection value p=2, and the other compounds value p=3. In addition, at a given value of p, the compound may exist as a mixture of isomers, since V1/Y can be associated with different sets of branches at L. In one embodiment, L represents the relationship. In another embodiment, L is a linear linker. In another embodiment, L is a linear linker, built in the cycloaddition reaction between a molecule containing an azide group, and a molecule containing an acetylene group. In another embodiment, L p is ecstasy branched linker. In another embodiment, L represents a dendritic linker. Dendritic structure can be, for example, is embedded in the cycloaddition reactions between molecules containing azide group, and molecules containing acetylene group. In one embodiment, p is 1. In other embodiments, p is 2 or 3, or 4, or 6, or 8, or 9. In another embodiment, L is represented by the formula: where X81and X82each independently represents O, NR85or S; each X83and X84independently represents O, NR86or S; each h, h, h and h independently represent 0 or 1; p is a number representing the degree of branching, and is an integer selected from 1 (included) to 128 (inclusive); p'" is a number representing the degree of branching, and is an integer chosen from 0 (inclusive) to 127 (inclusive); p"+p'"≤128; Each DD is independently represents H, OH or tsepliaeva group; R80is absent or represents either dendritic or branched or unbranched fragment, which is chosen from optionally substituted alkylene or polyalkylene, optionally substituted heteroalkyl or polyheteroarylenes, optionally substituted of arylene or polyarylene,optionally substituted of heteroaryl or polyheteroarylenes, optionally substituted cycloalkyl or polycyclohexylene, optionally substituted heterocyclization or polyheterocycles, -(CH2CH2O)v-, -alkylene-(CH2CH2O)v-, -(CH2CH2O)v-alkylene-, -alkylene-(CH2CH2O)v-alkylene-, -heteroalkyl-(CH2CH2O)v-, -(CH2CH2O)v-heteroalkyl-, -heteroalkyl-(CH2CH2O)v-alkylene-, -heteroalkyl-(CH2CH2O)v-heteroalkyl-, -alkylene-(CH2CH2O)v-heteroalkyl-, dendritic structure and oligopeptides, or any combination of two or more of the above; R85and R86independently selected from H and C1-8of alkyl; v is selected from 1 (included) to 500 (inclusive). For example, L may be selected from optionally substituted C1-10alkylene, C1-10alkilenkarbonatov, C1-12alkilenkarbonatov, C1-12carbonylation, C1-12carbonyldiimidazole and (CH2CH2O)v-carbonyl. In one embodiment, L is chosen from Reactive fragment RM and the linking group L2 Reactive fragment RM in the compound of formula (IV) is connected with the linking group L and capable of reacting with an appropriate functional group at actiongo partner. In one embodiment of the present invention reactive fragment RM designed so that it could react with the functional group of fragment V2that leads to the formation of compounds of formula (III). In this reaction a fragment of RM is transformed into a fragment of the L2. In another embodiment, the reactive fragment RM designed so that it could react with a complementary fragment in situ, for example, in vivo, resulting in a connection that may or may not be a compound of the formula (III). In one aspect of the present invention reactive fragment RM contains an electrophilic group that reacts with the nucleophilic group of the reaction partner, for example, V2for example, a thiol group, amino group or hydroxy-group. In another aspect of the present invention reactive fragment RM contains a nucleophilic group that reacts with the electrophilic group of the reaction partner, for example, V2for example, aldehyde group. In another aspect of the present invention reactive fragment RM contains a fragment of partner cycloaddition, for example, alkene, diene, 1,3-dipole or 1,3-dipolarophiles, which reacts with the corresponding complementary fragment partner cycloaddition, such as V2for example, Diana is, the alkene, 1,3-dipolarophiles or 1,3-dipole. In another aspect of the present invention reactive fragment RM contains a group which can be connected to the corresponding complementary group of the reaction partner, for example, V2in terms of using a metal catalyst, an enzyme or enzyme catalyst, for example, using a palladium catalyst. In one aspect of the present invention reactive fragment RM represents, without limitation, where X8selected from-Cl, -Br, -I, -F, -OH, -O-N-succinimide, -O-(4-nitrophenyl), -O-pentafluorophenyl, -O-tetrafluorophenyl, -O-C(O)-R50and-O-C(O)-OR50; X9selected from-Cl, -Br, -I, -O-masila, -O-trivella and-O-Totila; R50represents C1-10alkyl or aryl. In one embodiment, the fragment RM choose from that ensures its ability to react with the thiol group of the reaction partner, for example a fragment V2. In one embodiment, the fragment of the RM is that ensures its ability to react with the thiol group of the reaction partner, for example a fragment V2. In another embodiment, the fragment RM choose from it provides the ability is ü to react with the amino group, for example, primary or secondary amino group of the reaction partner, for example a fragment V2. In another embodiment, the fragment RM choose from that ensures its ability to react with the aldehyde group of the reaction partner, for example a fragment V2. The linking group L2in the compounds of the formula (III) represents the remainder of the RM after reactive fragment RM reacted with V2. The group then binds the fragment V2with L. the Group, which remains may be a connection. Usually, however, L2represents a linking group. If the compound of formula (III) is formed otherwise than through the compound of the formula (IV), L2should not represent the remainder of the RM, but may provide similar or the same parts and, in addition, may be selected from, for example, optionally substituted C1-6alkylene, C1-6heteroalkyl, C3-7cycloalkyl, C3-7heterocyclization, C5-10arylene and C5-10heteroaryl. In one embodiment, the fragment of L2represents the relationship. In another embodiment, the fragment of L2represents, without limitation, In the following embodiment, the fragment of L2is Fragment V2 V 2represents a functional fragment, which means that it adds additional functionality to the compound of the present invention. In one embodiment, V2presents a guide to the target fragment. In another embodiment, V2the fragment is a fragment, which improves the pharmacokinetic properties of the compounds of the present invention. In yet another variant V2the fragment is a fragment that causes the accumulation of the compounds of the present invention have mesiniaga site. In yet another variant V2the fragment is a fragment, which improves the solubility of the compounds of the present invention. In yet another variant V2the fragment is a fragment, which increases the hydrophobicity of the compounds of the present invention. In yet another variant V2the fragment is a fragment, which reduces transudate compounds of the present invention. In yet another variant V2the fragment is a fragment, which reduces the emission of compounds of the present invention. In yet another variant V2the fragment is a fragment, which reduces the immunogenicity of the compounds of the present invention. In yet another variant V2the fragment is a fragment, which increases the circulation of the compounds of the present invention. Another is Ariane V 2the fragment is a fragment, which increases the ability of the compounds of the present invention to overcome the biological barrier, for example, the membrane, the cell wall or the blood-brain barrier. In yet another variant V2the fragment is a fragment that enhances the ability to assimilate compounds of the present invention. In yet another variant V2the fragment is a fragment that causes the aggregation of the compounds of the present invention. In yet another variant V2the fragment is a fragment that reduces aggregation of the compounds of the present invention. In yet another variant V2the fragment is a fragment, which makes the compounds of the present invention to form micelles or liposomes. In yet another variant V2the fragment is a fragment that causes the complexing compounds of the present invention with other molecules, such as biomolecules. In yet another variant V2a polynucleotide fragment is a fragment that forms a complex with the complementary nucleotide sequence, such as RNA or DNA. In yet another variant V2the fragment is a fragment, which enables connection of the present invention to communicate, to associate, interact or affect Iwate complex with another fragment, for example, (functionalized) surface or solid carrier. In another embodiment, V2demonstrates two or more different functions. In one aspect of the present invention the fragment V2includes in its scope any fragment that binds or jet associated with, or forms a complex with the receptor, the receptor complex, antigen, or other receptive fragment associated with population under consideration nesenevich cells. V2can be any molecule that binds to a fragment of a population of cells, forms a complex with a fragment of a population of cells, or reacts with a fragment of a population of cells that can be therapeutically or otherwise biologically modified. Action V2the fragment is reduced to the delivery of one or more of the fragments Z to specific populations nesenevich cells that V2reacts with, or that V2bound. Such V2fragments include, but are not limited to, aptamers, full length antibodies and antibody fragments, lectins, biologically responsive modifiers, enzymes, vitamins, growth factors, steroids, nutrients, sugar, residues of oligosaccharides, hormones, and any derivatives thereof, or combinations thereof. After binding, the reaction of the Association or of complexable the Finance of the compounds of the present invention may or may not be internalizable. If there is internalization, transformation and/or cleavage V1preferably occurs inside mistaway cells. Suitable reimmunization-able protein, polypeptide, or peptide fragments V2include, but are not limited to, transferrin, epidermal growth factor ("EGF"), bombezin, gastrin and its derivatives, especially those that contain tetrapeptide sequence Trp-Met-Asp-Phe-NH2the peptide secreting gastrin, growth factor derived from platelets, IL-2, IL-6, transforming growth factors ("TGF"), such as TGF-α and TGF-P, tumor growth factor, vaccinia growth factor ("VGF"), insulin and insulin-like growth factors I and II, lectins and apoprotein of low-density lipoprotein. Suitable fragments of polyclonal antibodies V2represent heterogeneous populations of antibody molecules. To obtain polyclonal antibodies of interest antigens can use various procedures well known to specialists. Suitable fragments of a monoclonal antibody V2are homogeneous populations of antibodies to a specific antigen (for example, the antigen of the cancer cells). Monoclonal antibody (mAb) to interest antigen can be obtained using any known in the art techniques that will ensure received the e molecules monoclonal antibodies. Suitable fragments of a monoclonal antibody V2include, but are not limited to, human monoclonal antibodies, gumanitarnye monoclonal antibodies or chimerical human-mouse (or other species) monoclonal antibodies. Human monoclonal antibodies can be obtained using any of the countless methods known in the art. V2the fragment may also be bespecifically antibody. Methods of obtaining bespecifically antibodies known to experts. V2the fragment can be functionally active fragment, derivative or analog of an antibody which immunospecificity binds to antigens on nesenevich cells, such as antigens of cancer cells. In this regard, the term "functionally active" means that the fragment, derivative or analog is able to detect the anti-anti-idiotypical antibodies that recognize the same antigen, which recognizes the antibody from which the fragment, derivative, or similar. Other suitable V2fragments include fragments of antibodies, including, but not limited to, F(ab')2fragments that contain variable area, constant area light chain and the CH1 domain of the heavy chain, which can be obtained using basenowy prewar molecules of antibodies and Fab fragments, which can be the to create, reducing the disulfide bridges of F(ab')2the fragments. Other suitable V2fragments are dimers heavy chains and light chains of the antibodies, or any minimal fragments, such as Fvs or single-chain antibody (SCA), domain antibodies, anticalin, officila, nanotesla, or any other molecule with the same, similar or comparable specificity as the antibody. Additionally, recombinant antibodies, such as chimerical and gumanitarnye monoclonal antibodies, containing both human and not human plots that can be generated using standard recombinant DNA techniques, are useful V2the fragments. Chimerical antibody is a molecule in which different parts obtained from different animal species, such as those that contain variable plot derived from murine monoclonal and human immunoglobulin constant plot. Gumanitarnye antibodies are antibody molecules derived from non-human species that contains one or more of the complementarity determining plots (CDR) from a non human species and skeletal portion of human immunoglobulin molecules. Completely human antibodies are particularly preferred as V2the fragments. Such antibodies m the tenderly, for example, be obtained using transgenic mice that fail to Express the endogenous genes of the heavy and light chains of immunoglobulins, but which can Express human genes of heavy and light chains. In other embodiments, V2the fragment is a hybrid protein antibody or functionally active fragment or derivative, for example, one in which the hybridized antibody through a covalent bond (e.g., a peptide bond)or N-Termini or C-Termini with the amino acid sequence of another protein (or portion thereof, preferably at least 10, 20 or 50 amino acid portion of the protein), which is not an antibody. Preferably, the antibody or its fragments were covalently linked to other proteins at the N end of the constant domain. Antibody V2fragments include analogs and derivatives that are modified, for example by covalent joining of any type of molecule as long as such covalent connection allows the antibody to retain its binding antigen immunospecificity. For example, but not limited to, derivatives and analogs of the antibodies include those that have been further modified, e.g., by glycosylation, acetylation, by treatment with polyethylene glycol, disulfide recovery, FOSFA the financing, amidation, obtain derivatives with known protective or blocking groups, proteolytic cleavage, linkage with other proteins, etc. in Addition, analog or derivative may contain one or more of the natural amino acids. Antibody V2fragments include antibodies containing the modification (e.g., substitution (for example, cysteine to serine), deletions, or additions in the amino acid residues that interact with Fc receptors. In particular, they include antibodies containing modifications in amino acid residues that are identified as involved in the interactions between the Fc domain and the FcRn receptor. Can also be entered modifications so that you can attach antibodies conjugated linker agent in a specific position on the antibody. In a specific embodiment, the antibody immunospecific against the cancer antigen or tumor, is used as V2fragment in accordance with the compounds, compositions and methods of the present invention. Antibodies immunospecificity against the antigen of the cancer cells, can be obtained commercially or obtained by methods known to experts in this field, for example, in the field of chemical synthesis or in the field of recombinant expression techniques. Nucleotide sequence, tiraumea antibodies immunospecificity against the antigen of the cancer cells, can be obtained, for example, from the GenBank database or similar database, from commercial or other sources, literary publications, or by routine cloning and sequencing. Examples of antibodies available for the treatment of cancer include, but are not limited to, HERCEPTIN (trastuzumab, Genentech, CA)which is gumanitarnym anti-HER2 monoclonal antibody for the treatment of patients with metastatic breast cancer; RITUXAN (rituximab; Genentech, CA)which is a chimerical anti-CD20 monoclonal antibody for the treatment of patients with non-Hodgkin's lymphoma; OvaRex (oregovomab; AltaRex Corporation, MA), which is a murine antibody for the treatment of ovarian cancer; Panorex (edrecolomab; Glaxo Wellcome, NC)which is an IgG2aantibody for the treatment of colorectal cancer; IMC-BEC2 (mitooma; ImClone Systems, NY), which is a murine IgG antibody for the treatment of lung cancer; 1MC-C225 (Erbitux; Imclone Systems, NY), which is a chimerical IgG antibody for the treatment of head and neck cancer; Vitaxin (MedImmune, MD), which is gumanitarnoe antibody for the treatment of sarcoma; Campath I/H (alemtuzumab, Leukosite, MA)which is gumanitarnoe IgG1antibody for the treatment of chronic lymphocytic leukemia (CLL); SGN-70 (Seattle Genetics WA), which is gumanitarnoe anti-CD70 antibody for the treatment of hematologic malignancies; Smart MI95 (Protein Design Labs, CA), which is gumanitarnoe IgG antibody for the treatment of acute myeloid leukemia (AML); LymphoCide (epratuzumab, Immunomedics, NJ)which is gumanitarnoe IgG antibody for the treatment of non-Jackinsky lymphoma; SGN-33 (Seattle Genetics, WA), which is gumanitarnoe anti-CD33 antibody for the treatment of acute myeloid leukemia; Smart ID 10 (Protein Design Labs, CA), which is gumanitarnoe antibody for the treatment of non-Jackinsky lymphoma; Oncolym (Techniclone, CA), which is a murine antibody for the treatment of not Jackinsky lymphoma; Allomune (BioTransplant, CA)which is gumanitarnoe anti-CD2 mAb for the treatment of Hodgkin's disease or for the treatment of non-Jackinsky lymphoma; anti-VEGF (Genentech, CA)which is gumanitarnoe antibody for the treatment of lung cancer or colorectal cancer; SGN-40 (Seattle Genetics, WA) which is gumanitarnoe anti-CD40 antibody for the treatment of multiple myeloma; SGN-30 (Seattle Genetics, WA), which is a chimerical anti-CD30 antibody for the treatment of Hodgkin's disease; CEAcide (Immunomedics, NJ)which is gumanitarnoe anti-CEA antibody for the treatment of colorectal cancer; IMC-1C11 (ImClone Systems, NJ), which represents the STI-KDR chimerical antibody for the treatment of colorectal cancer cancers of the lung and melanoma; and Cetuximab (ImClone Systems, NJ), which is an anti-EGFR chimerical antibody for the treatment of positive cancers epidermal growth factor. Other antibodies that can be used for the treatment of cancer include, but are not limited to, antibodies against the following antigens: CA125, CA15-3, CA19-9, L6, Lewis Y, Lewis X, alpha fetoprotein, CA 242, placental alkaline phosphatase, prostatespecific antigen, prostatic acid phosphatase, receptor, epidermal growth factors, HER2, EGFR, VEGF, MAGE-1, MAGE-2, MAGE-3, MAGE-4, the receptor for anti-transferrin, p97, MUC1-KLH, MUC18, PSMA, CTLA4, CEA, gpl00, MART1, PSA, IL-2 receptor, CD2, CD4, CD20, CD30, CD52, CD56, CD74, CD33, CD22, HLA-DR, HLA-DR10, human chorionic gonadotropin, CD38, CD40, CD70, mucin, P21, MPG, and Neu oncogenic product. Many other internalized and not internalized antibodies that are associated with associated with tumor antigens, can be used in the present invention, some of which are listed in the overview16. In some embodiments, the antibody is an anti-nuclear antibody, or an antibody that can bind to a receptor or receptor complex expressed on mistaway cell. The specified receptor or receptor complex can include a member of the immunoglobulin gene superfamily, integrin, a receptor of the chemokine, a member of the superfamily, Retz is Perov TNF, a cytokine receptor, a major histocompatibility protein, the complement control protein or lectin. In another specific embodiment, the antibody immunospecific against an antigen associated with an autoimmune disease, is used as V2the fragment in accordance with the compounds, compositions and methods of the present invention. In another specific embodiment, the antibody immunospecific against viral or microbial antigen is used as V2the fragment in accordance with the compounds, compositions and methods of the present invention. In the sense used here, the term "viral antigen" includes, but is not limited to, any viral peptide or polypeptide protein, which is able to induce an immune response. In the sense used here, the term "microbial antigen" includes, but is not limited to, any microbial peptide, polypeptide, protein, saccharide, polysaccharide, or lipid, which is able to induce an immune response. New antibodies continuously discover and create, and in the present invention is proposed that these new antibodies can also be included in the compounds of the present invention. V2may react with reactive fragment RM through, for example, the heteroatom y V2. Heteroatoms, which can prisutstvie the VAT on V 2include, without limitation, sulfur (in one embodiment of the sulfhydryl group), oxygen (in one embodiment of the carboxyl or hydroxyl group) and nitrogen (in one embodiment, a primary or secondary amino group). V2may also respond via, for example, the carbon atom (in one embodiment of the carbonyl group). These atoms may be present at V2in V2's natural state, such as the naturally occurring antibody, or can be entered in the V2as a result of chemical modification. Free sulfhydryl groups can be created in the antibodies or fragments of antibodies, restoring antibody (fragment) regenerating agent such as dithiothreitol (DTT) or Tris(2-carboxyethyl)phosphine (TCEP). In this case, you can obtain a modified antibody, which may contain from 1 to about 20 sulfhydryl groups, but is usually from about 1 and to about 9 sulfhydryl groups. Alternatively, V2may contain one or more of the hydrocarbon groups that can be chemically modified to contain one or more of the sulfhydryl groups. As another alternative, sulfhydryl groups can be created in the reaction of amino groups such as lysine fragments, V2using 2-aminothiols (reagent trout) or other reagent, g is nesiruosiu sulfhydryl. In one embodiment, V2the fragment is a fragment that binds the receptor. In another embodiment, V2fragment is an antibody or antibody fragment. In another embodiment, V2the fragment is a monoclonal antibody or fragment. In one embodiment, V2contains one or more of the sulfhydryl groups and V2reacts with one or more of the RM fragments of compounds of the formula (IV) via one or more of the sulfur atoms of these sulfhydryl groups with the formation of the compounds of formula (III). In yet another variant V2contains a disulfide bond, which can be chemically restored to sulfhydryl groups (two for each disulfide bond), which can then be subjected to interaction with one or more of the reactive fragments RM. In another embodiment, V2contains from about 1 to about 3 sulfhydryl groups, which can react with one or more of the reactive fragments RM. In another embodiment, V2contains from about 3 to about 5 sulfhydryl groups, which can react with one or more of the reactive fragments RM. In another embodiment, V2contains from about 7 to about 9 sulfhydryl groups, which can react with one or more of the reactive fragments RM. In another variant is NTE V 2may contain one or more of the hydrocarbon groups that can be chemically modified to obtain one or more sulfhydryl groups. V2reacts with RM fragments through one or more of the sulfur atoms of these sulfhydryl groups. In another embodiment, V2may contain one or more of the groups of lysine, which can be chemically modified to contain one or more of sulfhydryl groups, which can react with one or more of the reactive fragments RM. Reactive fragments, which can react with the sulfhydryl group include, but are not limited to, carbamoylated, allalone, α-halogenated, halogeometricum, vinylsulfonic, maleimide and 2-desulfonylation. In yet another variant V2may contain one or more of the hydrocarbon groups that can be oxidized to obtain one or more of the aldehyde groups. The corresponding aldehyde (aldehyde) can then react with one or more of the reactive fragments RM. Reactive fragments, which can react with a carbonyl group at V2include , but are not limited to, hydrazine, hydrazide, amine and hydroxylamine. In yet another variant V2may contain one or more of the amino is Rupp, for example, lysine residues that can react with one or more of the reactive fragments RM. Reactive fragments, which can react with amino group include, but are not limited to, carbamoylated, α-halogenoacetyl, allalone, aldehyde, isocyanate and isothiocyanate. The conjugate of formula (III) can exist in the form of a mixture where each component of the mixture has a different q values. For example, the compound may exist as a mixture of two separate connections, one connection, in which q=3 and other compounds in which q=4. Analyzing the compound of formula (III), it is clear that q can be (rounded) average number of L2-L(-V1-Y-)p(Z)z/qunits V2the fragment. In addition, for a given q, the compound may exist as a mixture of isomers, as q L2-L(-V1-Y-)p(Z)z/qthe fragments can be connected with different sets of functional groups at V2. It should be noted that the number Z of the fragments in each unit may be equal to z/q, only if all units are the same and/or contain the same number of Z slices. In one embodiment, V2fragment connected with L2through the sulfur atom. In another embodiment, V2fragment connected with L2through the sulfur atom, and q takes values from about 1 d is about 20. In another embodiment, V2fragment connected with L2through the sulfur atom, and q takes values from about 1 to about 9. In another embodiment, V2fragment connected with L2through the sulfur atom, and q takes values from about 1 to about 3. In another embodiment, V2fragment connected with L2through the sulfur atom, and q is about 2. In another embodiment, V2fragment connected with L2through the sulfur atom, and q takes values from about 3 to about 5. In another embodiment, V2fragment connected with L2through the sulfur atom, and q is equal to about 4. In another embodiment, V2fragment connected with L2through the sulfur atom, and q takes values from about 7 to about 9. In another embodiment, V2fragment connected with L2through the sulfur atom, and q is equal to about 8. In one embodiment, the compound of formula (III) exists as a mixture of individual compounds. In one embodiment, the compound of formula (III) exists as a mixture of individual compounds, where q for the three compounds is 1, 2, and 3, respectively. In one embodiment, the compound of formula (III) exists as a mixture of individual compounds, where q for the three compounds is 3, 4, and 5, respectively. In one embodiment, the compound of formula (III) exists as a mixture of individual compounds, where q for the three compounds is 5, 6 and 7, respectively. In odnawiane compound of formula (III) exists as a mixture of individual compounds, where q for the three compounds is 7, 8 and 9, respectively. In another embodiment, V2fragment connected with L2through the nitrogen atom. In yet another variant V2fragment connected with L2through the carbon atom. In another aspect of the present invention V2the fragment includes any fragment that causes the accumulation of the compounds of the present invention have mesiniaga site or in proximity to it through a mechanism that differs from the binding or reaction of the Association or complexation with a receptor, antigen or other receptive fragment, related to this Milenium site, for example, population nesenevich cells. One way to achieve this is through the use of, for example, a large macromolecules, such as V2the fragment, which is directed to fabric solid tumors due to the effects of increased permeability and retention (EPR). Ringsdorf reports about the use of polymers for the direction of anticancer agents to tumors.17Due to the specified EPR effect, macromolecules passively accumulate in solid tumors as a result of unorganized pathology angiogenic tumor vasculature with its discontinuous endothelium, leading to hyperproliferate for large macromolecules, and the lack of effective swollen the left lymphatic drainage. V2the fragment can be, for example, branched or non-branched polymer, such as, for example, poly[N-(2-hydroxypropyl)methacrylamide] (HPMA), poly(2-hydroxyethylmethacrylate) (HEMA), polyglutamine acid or poly-L-glutamic acid (PG), carboxymethylchitin (CMDex), Polyacetal, chitosan, polypeptide, oligoethyleneglycols or polyethylene glycol (PEG), or a copolymer, such as HPMA copolymer, copolymer of HPMA-methacrylic acid, a copolymer of HEMA-methacrylic acid, CMDex copolymer, a copolymer of β-cyclodextrin, the PEG copolymer or copolymer of poly(lactic and glycolic) acid18. In the specified document as a polymer, and the copolymer referred to as a polymer. The polymer can be connected to the L2through any suitable functional group that can be located with one or both ends of the polymer, which means that q is the conjugate takes on the values 1 and 2, or alternatively, the functional group can (also) be placed on the groups suspensions of the polymer so that L is connected (also) to the polymer through these groups, pendants, and q values are typically in the range of from 1 to about 1000. Optionally, the polymer may also contain additional guide to the target group, which may contact or reactive to be associated with or form a complex with receptive slice in the example, the antibody or derivative of the antibody bound to the polymer or through suspended group or end group, so that is improved targeting mechaniy site. Alternatively, V2the fragment can be a dendrimer or protein, or fragment of a protein, such as albumin, which has no property of a target, except for its ability to accumulate in mesiniaga customers due to their size or molecular weight. In one embodiment, V2the fragment represents a polymer. In another embodiment, V2the fragment represents the polymer, and q takes values from 1 to about 1000. In other embodiments, V2the fragment represents the polymer, and q takes values from 1 to about 500, or 400, or 300, or 200, or 100, or less than 100. In another embodiment, V2the fragment represents the polymer, and q takes values from 1 to 2. In a specific embodiment, V2the fragment represents oligoethyleneglycols or polyethylene glycol or derivatives thereof. In another embodiment, V2the fragment represents a dendrimer, a protein or protein fragment. In another embodiment, V2the fragment is a fragment that is capable of transporting the conjugate through a biological barrier, for example, cell membrane, or with pre-linking, Association or complexation with the recipe for the rum or receptor complex, or without it. In one embodiment, V2the fragment is Tat peptide, or derivative, or fragment, or its equivalent, or its fragment, which has similar properties transmembrane delivery. In another embodiment, V2the fragment is a protein or protein fragment, antibody, or antibody fragment, receptor-binding or peptide fragment of the vector, or polymeric or dendritic fragment, or any combination of them, which are attached to the Tat peptide or its derivative, fragment, or analog, or fragment, which has similar properties transmembrane delivery. Thus, in one aspect of the present invention the fragment V2presents a guide to the target fragment and, for example, selected from the group consisting of a protein or protein fragment, or antibody, or antibody fragment, receptor-binding or peptide fragment of the vector, and polymeric or dendritic fragment, or any combination thereof. In another aspect of the present invention V2the fragment is a fragment, which improves the pharmacokinetic properties of the conjugate of the present invention. For example, the fragment V2can be chosen in such a way as to increase the solubility of the conjugate (more). This can be achieved by choosing this V2that is hydrof the d fragment. Alternatively, V2the fragment can be used, for example, to increase the residence time of the compounds in circulation, to reduce extravasation and selection, to reduce aggregation and/or to reduce the immunogenicity of the connection. This can, for example, be achieved by choosing this V2that would be a glycol or oligoarticular, or their derivatives. If the fragment V2represents the fragment, which improves the pharmacokinetic properties of the compounds of the present invention, V1represents a fragment that can be derived or transformed specificeski and are not present V1'and V2'fragments, the connection itself is improving (pharmacokinetic) properties of one or more of Z slices. In one embodiment, V2represents the fragment, which improves the pharmacokinetic properties, and V1represents a fragment that can be derived or transformed in a specific manner. In another embodiment, V2is oligoethyleneglycols or polyethylene glycol or its derivative, and V1represents a fragment that can be derived or transformed in a specific manner. In one embodiment, V2represents the fragment, which improves the pharmacokinetic properties, and V1represents a fragment, which can be from EPLAN or transformed specificeski. In another embodiment, V2is oligoethyleneglycols or polyethylene glycol or its derivative, and V1represents a fragment that can be derived or transformed specificeski. In another embodiment, V2is oligoethyleneglycols or polyethylene glycol or its derivative, and V1represents a fragment that can be derived ubiquitary enzymes. In another embodiment, V2is oligoethyleneglycols or polyethylene glycol or its derivative, and V1represents a hydrolyzable fragment. In one aspect of the present invention V2a fragment represented by formula (VI): where V2*L2*, L*, V1*, Y*, p*, q* and z* are the above values for V2L2, L, V1, Y, p, q, and z, as defined in the description, except that Y* is connected with L2. It should be noted that z* is in fact equal to q. If the compound of formula (III) contains V2a fragment represented by formula (VI), then one or more of the L2fragments connected with Y*. It should be understood that in this document, each time, as mentioned V2L2, L, V1, Y, p, q, or z, the same can be applied to each of the V2*L2*, L*, V1*, Y*, p*, q* or z*, respectively. Using V2 fragment of the formula (VI) in the conjugate of formula (III) implies that the two conditionally otsepleniya or conditionally-transformed fragment may be present in the same prefragmented, and therefore two separate removal/transformation may be required for complete removal of prefragments. The specified requirement that was satisfied with two different conditions, before they are allocated one or more of Z, can favorably affect the properties of the conjugate. For example, this can improve the guide, the effectiveness of the conjugate. Two acts of transformation/removal can occur in a variety of extracellular/intracellular locations. In this case, you can use the fragment, which must be removed during the second cleavage or, as a consequence, the second transformation, to provide transportation Z from the first extracellular or intracellular position in the second extracellular or intracellular position. It should be clear that V2the fragment of the formula (VI) can be used not only in the conjugates of the compounds of formula (I) or (II), but it can be used in a similar conjugates with other therapeutic agents, diagnostic fragments, etc. It should be understood that the functional fragment V2can have multiple joint functional properties. For example, V2m what may be a fragment, which improves the pharmacokinetic properties of the compounds of the present invention and at the same time is the pilot to the target fragment, or contains the specified fragment. The conjugates of the present invention can contain one or more of prefragments. These prefragmented may be the same or different. The presence of two or more prefragmented can favorably affect the properties of the conjugate. For example, it can increase the solubility and/or to guide increase the efficiency of the conjugate. In one embodiment, if there are two or more of prefragments, these prefragmented differ from each other. Two or more of different prefragmented may have different functions and can be removed under different conditions and in different extracellular/intracellular provisions. In one embodiment, the present one promiment associated with Z. In another embodiment, there is one promiment associated with Z in X1. In another embodiment, there are two promiment associated with Z. In another embodiment, there are two promiment associated with Z, one of which is connected through the X1. In yet another embodiment, there are three prefragments associated with Z. In yet another embodiment, there are three prefragments associated with Z, one of which is coupled through the X 1. In one embodiment, the compound of formula (III) represented by the compound of the formula (III-1) or (III-2): In another embodiment, the compound of formula (III) represented by the compound of the formula (III-3) or (III-4): where Y' is connected to the atom, which is part of R8, R9, R10or R11. In one embodiment, p is an integer from 1 (included) to 128 (inclusive). In another embodiment, q represents an integer from 1 (included) to 1000 (inclusive). In other embodiments, p is an integer from 1 (included) to 64 (inclusive), or 32 (inclusive), or 16 (inclusive), or 8 (inclusive), or 4 (inclusive), or 2 (inclusive). In other embodiments, q is an integer from 1 (included) to 500 (inclusive), or 400 (inclusive), or 300 (inclusive), or 200 (inclusive), or 100 (inclusive), or 16 (inclusive), or 8 (inclusive), or 6 (inclusive), or 4 (inclusive) or 2 (inclusive). In one embodiment, if more than 1 promiment connected to the first Z and in one of prefragmented there is more than one site join for Z fragments, then the other of these prefragmented connected with a first Z contains one site of accession to the Z fragment. In one VA is iante compound of formula (III) is represented as In one embodiment, the p in the compound of formula (IIIa) represented by 1. In another embodiment, the compound of formula (IIIa) p presents 1 and z is equal to q. In another embodiment, the compound of formula (IIIa) is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R1ais chlorine (Cl) or bromine (Br), (AA)aachoose from wallcarolina, valiligine, fenilalanina, albertinelli and D-alanylhistidine, ss represents 1 or 2, R2aand R5brepresent methyl and H, respectively, or H and methyl, respectively, R8dchoose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy, R9dselected from H and methoxy, 'LL choose from qq takes values from 1 to about 20, rr and rr'each, independently accepts values from 1 to about 4, and Ab represents the antibody or fragment or derivative. In one embodiment, the compound of formula (III) is represented as In one embodiment, the p* in the compound of formula (IIIa*) is 1. In another embodiment, the compound of formula (IIIa*) p* is 1, and z* is q*. In another embodiment, the compound of formula (III) is represented as In one embodiment, the p in the compound of formula (IIIb) is 1. In one embodiment, the compound of formula (III) is represented as In one embodiment, the p* in the compound of formula (IIIb*) is 1. In another embodiment, the compound of formula (IIIb*) p* is 1, and z* is q*. In another embodiment, V1in the compound of formula (IIIb*) is ottsepleny enzyme substrate. In yet another variant V1can be derived intracellular enzyme. In another embodiment, V1is optionally substituted dialkylaminoalkyl group, where the two alkyl groups may be the same or different, and may not necessarily be connected to each other with formation of an optionally substituted carbocycle or heterocycle. In yet another variant V1is piperazinylcarbonyl. In another embodiment, the compound of formula (IIIb*) is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R1ais chlorine (Cl) or bromine (r), (AA)aachoose from wallcarolina, valiligine, fenilalanina, albertinelli, and D-alanylhistidine, ss represents 1 or 2, R2aand R5brepresent methyl and H, respectively, or H and methyl, respectively, R8dchoose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino, (piperidine-4-yl)methoxy, 2-(N-methyl-N-(carboxymethyl)amino)ethoxy, and 2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy, R9dselected from H and methoxy, 'LL choose from qq takes values from 1 to about 20, rr and rr' are each independently takes values in the interval from 1 to about 4, and Ab represents the antibody or fragment or derivative. In another embodiment, the compound of formula (III) is represented as In yet another embodiment, the compound of formula (III) is represented as In one embodiment, the compound of formula (IIId) is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R1ais chlorine (Cl) or bromine (Br), (AA)aachoose from wallcarolina, valiligine, f is nylalanine, albertinelli, and D-alanylhistidine, ss represents 1 or 2, R2aand R5brepresent methyl and H, respectively, or H and methyl, respectively, R8echoose from 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino and (piperidine-4-yl)methoxy, and in which the carbonyl connected with R8eattached to the nitrogen atom in R8e, R9dselected from H and methoxy, 'LL choose from qq' takes values from 1 to about 20, rr and rr1each, independently takes values in the interval from 1 to about 4, and Ab represents the antibody or fragment or derivative. In one embodiment under consideration, the invention relates to the compound of formula (IIIdl) or (IIId2): where V1represents a monosaccharide, disaccharide or oligosaccharide from hexoses or pentoses, or heptose, which can also be included in the group deoxysaccharides or amino sugars and belong to the D-range or L-range and disaccharides or oligosaccharides are identical or different, or where V1has the formula-C(ORa'Rb'CHRc'Rd'where Ra', Rb', Rc'and Rd'have the above values. In one embodiment, the compound of the present invention is represented as or is th (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers, where R8cchoose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy and 2-(N,N-dimethylamino)acetylamino, and R9cselected from H and OCH3. In a more specific embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another more specific embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another more specific embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another more specific embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another more specific embodiment, the compound of the present invention is represented as <>or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In one embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention predstavlenie or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R,10S) isomer, its (1R,10R) - isomer, its (1S,10S) isomer, or a mixture of two or more of these isomers. In another embodiment, the compound of the present invention is represented as or (1R) isomer or a mixture of two isomers. Synthesis, in vitro cytotoxicity and biological evaluation of the compounds of the present invention As disclosed in more detail hereinafter, the compounds of formula (I), (II) and (III), as well as the compounds of formula (IV), can be conveniently obtained by the method, which is partially similar to the methods for producing the compounds disclosed, for example, in WO 01/83448, DE 4415463, WO 2004/043493 and WO 02/083180. In one embodiment, compounds of formula (I) or (II) used to produce the compounds of formula (III). In another embodiment, compounds of formula (I) or (II) used to produce the compounds of formula (IV). In another embodiment, the connection fo the formula (IV) is used to produce the compounds of formula (III). In another embodiment, the compound of formula (III), where V1represents a protective group, is used to produce other compounds of formula (III), where V1represents the in vivo remove the fragment. Some linking DNA fragments containing a water-soluble group, were obtained with good yields. Figure 4 revealed the synthesis of DNA binding compounds 13, containing 5-(2-(morpholine-4-yl)ethoxy) group. 4-NITROPHENOL 9 alkylate 4-(2-chloroethyl)morpholine, receiving the connection 10. In the nitrogroup reduction to obtain compound (11) and subsequent indole synthesis, Fischer receive indole 12. After saponification eventually get a connection 13 with 37% of the total output. Figure 5 revealed the synthesis of DNA binding compounds 19. On the basis of aldehyde 14 in the alkylation get the connection 15, and the subsequent andolina condensation results azide 16. After ring closure get indole 17, which amyraut to connection 18. Substitution of chloride with dimethylamine results in connection 19 with 50% of full output. Figure 6 revealed the synthesis of DNA binding compounds 27, based on utilisedictated (20). By methylation get a connection 21, followed by reduction of the ester group to obtain compound 22. It is attached to 1-chloro-4-nitrobenzene (23), receiving the connection one of the nitro group gives compound 25. Indole synthesis, Fischer leads to the connection 26; subsequent saponification gives compound 27 with 40% of the total output. Obtaining DNA binding connection 30 is presented on Fig.7. On the basis of a complex ester 28, restoration nitro and the subsequent addition of N,N-dimethylglycine network connection 29. Saponification of the ester group yields the connection 30 with 68% yield. On Fig presents obtaining DNA binding compounds 41 and its use to obtain the Boc-protected DNA binding connections 37 connecting DNA connection 38, and tert-butyl-protected DNA binding connection 39. Figure 2 presents partial removal of the protective groups double-protected DNA alkylating compounds 1, then attach the amino group with DNA binding compounds 2, 13, 19, 27, 30, 37, 38 and 39, respectively. This results in secure compounds 3a-h, a protective group which is removed, receiving agents 4a-h. Figure 9 presents a synthesis agent 45 of the connection 44 and DNA binding of compound 2 in accordance with a scheme similar to the scheme for the agents 4a-h. The connection 44 receive similarly, to obtain compounds 110using as a starting material o-tolualdehyde instead of benzaldehyde. Similarly, figure 10 shows the synthesis of agent 33. On the basis of the connection 31, the cha is in part the removal of the protective groups, and then attach to the DNA binding compound 2, get the connection 32. After removal of the protective groups receive agent 33. Figure 3 presents a synthesis of several conjugates galactose. Based on alkylating fragments with 5 partially removed protective groups, attaching O-(2,3,4,6-Tetra-O-acetyl-α-D-galactopyranosyl)trichloroacetimidate (6), with the subsequent removal of the protective groups and attach binding DNA fragment have protected conjugates 7a-e. Synthesis of conjugates 7b and 7d requires additional stages of recovery using PtO2·H2O/H2. The final removal of the protective groups results in conjugates galactose (1S,10R)-8a-e/(1R,10S)-8a'. Similarly, figure 11 shows the synthesis of the conjugate 36. On the basis of the connection 34, the connection of the connection 6, the removal of the protective groups and joining DNA binding 2, receive connections (1S,10R)-35/(1R,l0S)-35', which in turn connections 36/36'using methanolysis. On Fig presents the synthesis of conjugates of linker-agent 47a-47f. On the basis of agent 4a, the activated phenolic hydroxyl group, joining Boc-protected N,N'-dimethylethylenediamine or piperazine and removal of the protective groups of an amino group receive connections 46a and 46b. As a result of the accession of activated steam-nitrophenylarsonic linker structures 57a, 57b and 58 receive connections 47a-47d and protect the seal connection 47e-47f. Connection 47e-47f receive the subsequent removal of the protective groups, catalyzed by palladium. The activated linker of the structure is in accordance with the scheme presented on Fig. In short, the reactions proceed in connection 51a and citrulline or connection 51b and Boc-protected lysine give compound 52a or connection 52b, respectively. Attaching a pair of aminobenzamide alcohol to the compound 52a network connection 53, which in turn connections 57a and 57b, removing the protective group by piperidine, carrying out the reaction of the joining connections 55 and 56, respectively, and activating the hydroxyl group of bis(para-nitrophenyl)carbonate. Replacement of the Boc-group on the Aloc group 52b, and then attach a pair of aminobenzamide alcohol gives compound 54, which in turn connection 58 is similar to the transformation connection 53 at 57b. Connection and 59b 60 obtained from compounds 57b and 58, attaching a pair of aminobenzoyl alcohol and activating the hydroxyl group. Connection 59a receive 5 stages of connection 53. On Fig disclosed the synthesis of conjugates of linker-agent 48c and 48d of 47c and 47d, respectively, using catalyzed by Cu(I) reaction containing maleimide acetylene. On Fig disclosed the synthesis of conjugates of linker-agent 50a-50c, based on the connection 49. The connection 49 is produced from compound 5, introducing first methylpiperazine ragment same way, presented at Fig, and then attach to the DNA binding compound 37 in accordance with the scheme presented in figure 2. Removing the protective group from compounds 49 and then attach to the activated steam-nitrophenylarsonic linker structures 59a, 59b and 60, leads to the formation of compounds 50a-50b and secure connection 50c. Connection 50c obtained after subsequent removal of the protective groups, catalyzed by palladium. Connection 47e, 47f, 50b and 50c can be converted into compounds similar to the compound 48c using catalyzed by Cu(I) cycloaddition to acetylene 40. The conjugates of (1S,10R)-8a and (1R,10S)-8a' are tested in in vitro tests for cytotoxicity against cell lines of lung carcinoma A549 (Fig). Mixture of diastereomers (1S,10R)-8a and (1R,10S)-8a' shows IR50against A549 cells 7900 nm in the absence of the enzyme β-D-galactosidase, whereas IR50in the presence of a specified enzyme is 1.2 nm. This means that the ratio of cytotoxicity, IR50in the absence of enzyme, divided by IR50in the presence of the enzyme, is 6583. Conjugate (1S,10R)-8a shows IR50in the absence of enzyme 3600, whereas IR50reduced to 0.75 nm in the presence of the enzyme β-D-galactosidase, and the ratio of cytotoxicity takes a value of 4800. Conjugates 8b-e test in the same cell line, and b is lo shows their ratio of cytotoxicity similar to or significantly higher than the ratios reported previously (see reference 10), and agents 4b-e show nanomolar or subnanomolar activity. The compound (+)-8a evaluated in orthotopic model of breast cancer SCID mice, using the concept of an ADEPT. Female mice of strain SCID inoculant cells 1×106MDA-MB-231 (estrogen-independent cell line of human cancer cells of the breast), suspended in 25 μl of sterile PBS in the mammary fad pat 4thcomplex breast cancer. Tumors give rise, and ADEPT, therapy is started after tumor volume reaches detectable sizes. Mice treated in accordance with the treatment plan presented on Fig, and spend two loop monoclonal anti-human urokinase plasminogen activator receptor (uPAR) antibody conjugated with β-galactosidase, uPAR*β-Gal in PBS, followed by three injections of the conjugate of (+)-8a in 1% solution of DMSO/NaCl. Control groups injected or antibody-enzyme and 1% solution of DMSO/NaCl (antibody-enzyme one group), PBS, and conjugate (conjugate one group), or PBS and 1% solution of DMSO/NaCl (control group). After one cycle of treatment, average volume of primary tumors in ADEPT-treated mice reaches 0,23±0,07 cm3while cf is dni volume of primary tumors in the treated carrier mice is a 0.27±0.10 cm 3. After two cycles of treatment, it was found that the average amount of the amount of 0.33±0,13 cm3and 0.52±0,29 cm3for ADEPT-treated group and control group, respectively (Fig). Inhibitory effect for the concept ADEPT at the rate of tumor growth is slightly more pronounced after the second cycle of therapy. Increased inhibitory effect on tumor growth was observed in mice with relatively small tumors at start of therapy (data not shown). In addition, tumors, treated by the method ADEPT therapy, show a more necrotic tissue within the tumor than tissue tumors in mice treated with media. Processing or separately uPAR*β-Gal, or only one conjugate did not demonstrate any inhibitory effect on tumor growth compared with control media. ADEPT well tolerated by all animals. The General condition of the animals on the day of the killing was similar to the state of mice that were injected only media. Blood parameters in mice subjected to treatment did not differ from the parameters of the control animals. Equivalent in vivo experiment is carried out in a model in syngeneic mice using cell lymphoma A20 and anti-CD19 monoclonal antibody. In accordance with the same procedure, which is disclosed above, lymphoma, subjected ADEPT therapy, n who has demonstrirovali inhibition of growth of tumors. Application, the methods and compositions In one aspect the present invention relates to the use of compounds of formula (I) or (II) for obtaining the compounds of formula (III). In another aspect, the present invention relates to the use of compounds of formula (IV) to obtain the compounds of formula (III). In another aspect the present invention relates to the use of compounds of formula (I) or (II) for obtaining the compounds of formula (IV). In another aspect the present invention relates to the use of compounds of formula (III), where V1represents a protective group, to obtain other compounds of formula (III), where V1represents the in vivo remove the fragment. In another aspect under consideration, the invention relates to the use of any of the above defined compounds for preparing pharmaceutical compositions for the treatment of in need of this mammal. In one embodiment under consideration, the invention relates to the use of any of the above defined compounds for preparing pharmaceutical compositions for treating tumors in a mammal. Considering the invention also relates to any particular above compound as a medicinal product or an active component, or the active substance in the medicinal product. The following aspect to consider is finding relates to a method for producing a pharmaceutical composition, contains the connection defined above, to obtain a solid or liquid compositions for administration orally, topically or by injection. This method or process at least includes a step of mixing the compound with a pharmaceutically acceptable carrier. In one embodiment, the compound of the present invention used for the treatment of diseases characterized by unwanted proliferation. In another embodiment, the compound of the present invention used for the treatment of diseases characterized by unwanted cell proliferation. In another embodiment, the compound of the present invention is used to treat tumors. In yet another embodiment, the compound of the present invention is used to treat inflammatory diseases. In yet another embodiment, the compound of the present invention used for the treatment of autoimmune diseases. In yet another embodiment, the compound of the present invention is used to treat bacterial or microbial infection. In the following embodiment, the present invention relates to a method of treatment of a mammal, a disease which is characterized by unwanted (cell) proliferation, the compound of the present invention. In another embodiment, the present invention relates to a method of treating tumors in a mammal the compound of this izaberete the Oia. In yet another embodiment, the present invention relates to a method of treating inflammatory disease in a mammal the compound of the present invention. In yet another embodiment, the present invention relates to a method of treating autoimmune disease in a mammal the compound of the present invention. In yet another embodiment, the present invention relates to a method of treating a bacterial or microbial infection compound of the present invention. In the next version considering the invention relates to a method of treatment in need thereof of a mammal, with the specified method comprises administration to a mammal a pharmaceutical composition comprising the compound of the present invention, in a therapeutically effective dose. In one embodiment under consideration, the invention relates to a method of treating or preventing a tumor in a mammal, and the method includes the administration to a mammal a pharmaceutical composition comprising the compound of the present invention, in a therapeutically effective dose. In another embodiment under consideration, the invention relates to a method of treating or preventing an inflammatory disease in a mammal, and the method includes the administration to a mammal a pharmaceutical composition comprising a compound of the crust is asego of the invention, in a therapeutically effective dose. In another embodiment under consideration, the invention relates to a method of treating or preventing autoimmune disease in a mammal, and the method includes the administration to a mammal a pharmaceutical composition comprising the compound of the present invention in a therapeutically effective dose. In another embodiment under consideration, the invention relates to a method of treating or preventing a bacterial or microbial infection in a mammal, and the method includes the administration to a mammal a pharmaceutical composition comprising the compound of the present invention in a therapeutically effective dose. Considering the invention also relates to pharmaceutical compositions comprising the compounds of the present invention, as defined above. The compound of the present invention can be introduced in purified form together with a pharmaceutical carrier as a pharmaceutical composition. The preferred form depends on the intended route of administration and therapeutic application. The pharmaceutical carrier can be any compatible, non-toxic substance suitable for delivery of the compounds of the present invention to the patient. Pharmaceutically acceptable carriers well known in the art and include, for example aqueous solutions, such as (sterile) water or physiologically superyoung saline or media, such as glycols, glycerol, oils such as olive oil, or organic esters for injection, alcohol, fats, waxes, and inert solids. Pharmaceutically acceptable carriers may further contain physiologically acceptable compounds that act, for example, as stabilizers and / or increase the absorption of the compounds of the present invention. Such physiologically acceptable compounds include, for example, hydrocarbons, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients. Professionals should be aware that the choice of pharmaceutically acceptable carrier, including a physiologically acceptable compound, depends, for example, from the method of administration of the composition. Pharmaceutically acceptable adjuvants, buferiruemoi agents, dispersing agents, etc. may also be included in pharmaceutical compositions. For oral administration the active ingredient can be introduced in solid dose forms, such as capsules, tablets and powders, or in liquid dose forms, such as elixirs, syrups and suspensions. Active ingredient (s) may be enclosed in elastinovye capsules together with inactive ingredients and powdered carriers, such as glucose, lactose, sucrose, mannitol, starch, cellulose or cellulose derivatives, magnesium stearate, stearic acid, Nachrichten, talc, magnesium carbonate, etc. are Examples of additional inactive ingredients, which can be added to achieve the desired color, taste, stability, bufferarray ability, dispersion or other known desirable characteristics include red iron oxide, silica gel, nutriceuticals, titanium dioxide, edible white ink, etc. Similar diluents can be used to obtain CT. As tablets and capsules can be prepared in the form of products with delayed allocation to achieve constant release of the drug in a matter of hours. Molded tablets may be coated with sugar or film-coated to mask any unpleasant taste and protect the tablet from atmospheric influences, or they can be with gastric coated for selective disintegration in the gastrointestinal tract. Liquid dose forms for oral administration may contain coloring and flavoring agents to improve reception by the patient. Compounds of the present invention it is preferable, however, to enter parenteral. Preparations of the compounds of the present invention for parenteral administration is be sterile. Sterilization is easily accomplished by filtration through sterile filtration membranes, optionally before or after lyophilization and recovery. Parenteral route of administration of the compounds of the present invention is carried out in accordance with, for example, injection or infusion by intravenous, intraperitoneally, intramuscularly, intraarterially or in the affected area. Compounds of the present invention can be entered continuously by infusion or by bolus injection. A typical composition for intravenous infusion should contain up to 100 to 500 ml of sterile 0.9% NaCl or 5% glucose optionally supplemented with a 20% solution of albumin, and from 1 mg to 10 g of compound of the present invention, depending on the specific type compounds of the present invention and the desired mode of administration doses. Methods of making compositions for parenteral administration are well known in the art and are disclosed in more detail in various sources, including, for example, Remington''s Pharmaceutical Science19. Considering the invention is illustrated further by the following examples. These examples are only for illustrative purposes and in no way limit the scope of the present invention. EXAMPLES Example 1 rac-{(1,10)-Anti-5-benzyloxy-3-[(5-(2-(N,N-dimethylamino)ethoxy)indol-2-yl)carbonyl]-1-(10-chloroethyl)-1,2-is ihydro-3H-benzo[e]indol} (rac-3a) Racemic compound 1 (100 mg, 228 μmol) suspended in 4M HCl/EtOAc and stirred at room temperature for 3 hours. The reaction mixture was concentrated and dried in vacuum for 1 hour. The residue is dissolved in DMF (10 ml). The solution is cooled to 0ºC and added EDC·HCl (131 mg, 684 μmol, 3.0 equiv.) and compound 2 (82,0 mg, 288 μmol, 1.3 equiv.). The reaction mixture was stirred at room temperature for 2 hours, diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3and extracted with ethyl acetate (3×100 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2C12/MeOH=10:1) receive a racemic compound 3 in the form of a solid light brown color (81 mg, 143 μmol, 63%). 1H-NMR (300 MHz, CDCl3): δ=1,65 (d, J=6.8 Hz, 3H, 11-CH3), 2,37 (c, 6H, NMe2), 2,78 (t, J=5.8 Hz, 2H, 2"-H2), 3,96-a 4.03 (m, 1H, 1-H), of 4.12 (t, J=5.8 Hz, 2H, 1"-H2), 4,51-of 4.66 (m, 2H, 2-Ha, 10-H), 4,88 (mwith, 1H, 2-Hb), from 5.29 (mC2H, OCH2Ph), 7,06 (d, J=2.3 Hz, 1H, 3'-H), 7,07 (DD, J=8,5, 2.4 Hz, 1H, 6'-H), to 7.15 (d, J=2.4 Hz, 1H, 4'-H), 7,31-7,46, 7,50-7,57 (m, 8H, 7-H, 7'-H, 8-H, 5×Ph-H), 7,71 (d, J=8,3 Hz, 1H, 9-H), 8,17 (Cuser., 1H, 4-H), at 8.36 (d, J=8.5 Hz, 1H, 6-H), 9,51 (Cuser., 1H, NH);13C-NMR (75 MHz, CDCl3): δ=24,0 (11-CH3), 46,0 (NMe2), 47,6 C-1), of 53.7 (C-2), 58,5 (C-2'), was 59.9 (C-10), of 66.6 (C-1'), 70,3 (OCH2Ph), was 98.2 (C-4), also, 103.5 (C-4'), 105,9 (C-3'), 112,7 (C-7'), 117,2 (C-6', C-5a), 122,6, 123,7, 123,7, 123,9 (C-6, C-7, C-9, C-9b), 127,5, 127,6, 128,0, 128,2, 128,6 (C-3a', C-8,5×Bn-CH), 130,0, 130, 8mm, 131,3 (C-2', C-7a', C-9a), 136,8 (Bn-C), 142,4 (C-3a), other 153.9 (C-5'), 155,6 (C-5), of 160.4 (C=O); MC (EI, 70 eV): m/z (%) = 567,4 (1) [M]+, 531,5 (5) [M-Cl]+; C34H34ClN3O3(568,11): calculated 567,2289 found 567,2289 (EI-HRMS). Example 2 rac-{(1,10)-Anti-1-(10-chloroethyl)-5-hydroxy-3-[(5-(2-(N,N-dimethylamino)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indole hydrochloride} (rac-4a) Racemic compound 3a (200 mg, 352 μmol) dissolved in a mixture of 4M HCl/ethyl acetate (15 ml) and stirred for 2 hours at room temperature. The solution concentrate. The residue is dried in vacuum for 1 hour and then suspended in THF (15 ml). Then added at room temperature with 10% palladium on activated carbon (75 mg) and monitorial (25% aqueous solution, 0.75 ml). The reaction mixture is stirred for 2 hours at 40ºC and filtered through celite, which is washed thoroughly with methanol (200 ml). The obtained filtrate was concentrated in vacuo and the residue purified using column chromatography (CH2Cl2/MeOH = 5:1, 1% conc. HCl)to give rac-4a powder form of green-yellow (146 mg, 284 μmol 81%). 1H-NMR (300 MHz, DMSO-d6): δ=1,62 (d, J=6,7 Hz, 3H, 11-CH3), 2,84 (c, 6H, NMe2), 3,51 (t, J=5.0 Hz, 2H, 2"-H2), 4,16 (mwith, 1H, 1-H), 4,39 (t,J=5.0 Hz, 2H, 1"-H2), 4,57 (mwith, 1H, 2-Ha), 4,66-4,80 (m, 2H, 2-Hb, 10-H), of 6.99 (DD, J=8,9, and 2.4 Hz, 1H, 6'-H), 7,16 (d, J=1.6 Hz, 1H, 3'-H), 7,25 (d, J=2.4 Hz, 1H, 4'-H), 7,35 (t, J=7,6 Hz, 1H, 7-H), 7,44 (d, J=8,9 Hz, 1H, 7'-H), to 7.50 (t, J=7,6 Hz, 1H, 8-H), 7,88 (d, J=8,4 Hz, 1H, 9-H), of 7.96 (Cuser., 1H, 4-H), 8,13 (d, J=8,4 Hz, 1H, 6-H), accounted for 10.39 (c, 1H, OH), 10,87 (Cuser., 1H, NH+), 11,64 (Cuser., 1H, NH);13C-NMR (75 MHz, DMSO-d6): δ=23,4 (11-CH3), and 42.7 (NMe2), with 45.8 (C-1), to 52.1 (C-2), of which 55.4 (C-2'), is 61.5 (C-10), 63,0 (C-1'), 100,3 (C-4), 104,0 (C-4'), of 105.2 (C-3'), of 113.2 (C-7'), 115,5, 115, 8mm (C-6', C-5a), 122,1, 122,8, 122,9, 123,1 (C-6, C-7, C-9, C-9b), 126,9, 127,4 (C-3a', C-8), 129,8, 131,3, 131,9 (C-2', C-7a', C-9a), to 142.1 (C-3a), to 152.0 (C-5'), 153,8 (C-5), 159,8 (C=O); MC (ESI): m/z (%) = 478,4 (100) [M-Cl+H]+; C27H29Cl2N3O3(514,44): calculated 478,1892 found 478,1892, [M-Cl]+(ESI-HRMS). Example 3 [(1,10)-Anti-1-(10-chloroethyl)-3-[(5-(2-(N,N-dimethylamino)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-2,3,4,6-Tetra-O-acetyl-β-D galactopyranoside ((+)-7a/(-)-7a') A solution of rac-5 (381 mg, 1.10 mmol) in dry CH2C12(50 ml) and molecular sieves 4Ǻ (1.5 g) is stirred for 30 minutes at room temperature. After adding O-(2,3,4,6-Tetra-O-acetyl-α-D-galactopyranosyl)trichloroacetimidate (6) (564 mg, 1.15 mmol, 1.05 equiv.), the resulting mixture was cooled down to-10ºC, and slowly add a solution BF3·OEt2(69,0 µl, 548 mmol, 0.5 equiv.) in dry CH2C12(5.5 ml). After 3 hours at-10ºC add additional portion BF3·OEt (of 0.42 ml, 3,29 mmol, 3.0 equiv.) in dry CH2C12(5.0 ml) and the resulting mixture was warmed to room temperature. After 5 hours the resulting solution is separated from the molecular sieves, molecular sieves washed thoroughly with CH2C12(2×20 ml)and the combined organic portion was concentrated in vacuo. The residue is dried in vacuum for 1 hour and then dissolved in dry DMF (60 ml). The solution is cooled to 0ºC, and added EDC·HCl (630 mg, 3,29 mmol, 3.0 equiv.) and compound 2 (468 mg, of 1.64 mmol, 1.5 equiv.). After 20 hours at room temperature, the reaction mixture was diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3(50 ml)and then extracted with ethyl acetate (4×100 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2C12/MeOH = 10:1) get a mixture of diastereomers 7a and 7a' in the form of a solid yellow (475 mg, 588 mmol, 54%). MC (ESI): m/z (%) = 1637 (52) [2M+Na]+, 830,3 (66) [M+Na]+, 808,3 (100) [M+H]+. Example 4 [(1,10)-Anti-1-(10-chloroethyl)-3-[(5-(2-(N,N-dimethylamino)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-β-D galactopyranoside ((+)-8a/(-)-8a') To a solution of 7a/7a' (473 mg, 585 μmol) in dry MeOH (20 ml) was added NaOMe in methanol (0,22 approximately 5 ml,4M solution of 1.17 mmol, 2,0 ek is Yves.). The reaction mixture was stirred at room temperature for 2 hours and diluted with water (5 ml). Add ion-exchange resin (Amberlite-IR 120) up until the pH of the solution does not reach the neutral values. The solution was concentrated in vacuo and the residue purified using column chromatography (CH2Cl2/MeOH =1:1)to give a mixture of 8a and 8a' in the form of a solid pale yellow color (307 mg, 480 μmol, 82%). 1H-NMR (600 MHz, DMSO-d6): δ=1,65 (d, J=6,7 Hz, 3H, 11-CH3), 2,24 (c, 6H, NMe2), to 2.66 (t, J=5.7 Hz, 2H, 2"-H2), 3,41-to 3.73 (m, 4H, 2"'-H*, 4"'-H*, 6"'-H2in ), 3.75-3,86 (m, 2H, 3"'-H*, 5"'-H*), 4,07 (t, J=5.7 Hz, 2H, 1"-H2), 4,21-to 4.28 (m, 1H, 1-H), 4,49-of 4.67 (m, 3H, 2×OH, 2-Ha), 4,68-5,00 (m, 4H, 1"'-H, 2-Hb, 10-H, OH), and 5.30 (Cuser., 1H, OH), 6,92 (DD, J=8,8, 2.4 Hz, 1H, 6'-H), 7,13-7,20 (m, 2H, 3'-H, 4'-H), 7,38 was 7.45 (m, 2H, 7-H, 7'-H), EUR 7.57 (mwith, 1H, 8-H), of 7.96 (mwith, 1H, 9-H), 8,23 (Cuser., 1H, 4-H), 8,32 is 8.38 (m, 1H, 6-H), are 11.62 (Cuser., 1H, NH);13C-NMR (150 MHz, DMSO-d6): δ=23,4 (11-CH3), 45,5 (NMe2), 45,9/46,0 (C-1), 52,1 (2 signals)(C-2), of 57.8 (C-2'), 59,7/a 59.8 (C-6"'), 61,3 (2 signals)(C-10), 66,3 (C-1"), 67,6/67,7, 70,5/70,6, 73,2 (2 signal), and 75.2 (2 signals)(C-2"', C-3"', C-4"', C-5"'), 101,9 (C-4), 102,1/to 102.3 (C-1"'), 103,3 (2 signals)(C-4'), 105,4 (C-3'), of 113.2 (C-7'), 115,9 (2 signals)(C-6'), 118,9 (2 signals)(C-5a), 122,90, 123,0, 123,3/123,4, 123,6/123,6 (C-6, C-7, C-9, C-9b), RUB 127.3 (C-8), of 127.5 (C-3a'), 129,5 (2 signals), 130,9, 131,7 (2 signals)(C-2', C-7a', C-9a), 141,9/142,0 (C-3a), 153,0 (2 signals)(C-5'), 153,6 (2 signals)(C-5), 160,1/160,2 (C=O); MC (ESI): m/z (%)= 13010 (100) [2M+2H] +, 662,3 (82) [M+Na]+, 640,3 (76) [M+H]+; C33H38C1N3O8(640,12): calculated 640,2420 found 640,2420, [M+H]+(ESI-HRMS). Both diastereoisomer receive separately in accordance with the same procedures on the basis of (+)-5 and (-)-5, respectively. Example 5 4-[2-(4-Nitrophenoxy)ethyl]morpholine (10) A solution of KOH (3.3 g, 58 mmol) in EtOH (15 ml) is added dropwise to a solution of 4-NITROPHENOL (9) (7.0 g, 50 mmol) in EtOH (10 ml). After stirring for 30 minutes at room temperature salt is collected by filtration, washed with cold EtOH and dried in vacuum, obtaining the potassium salt of compound (9) in the form of a solid yellow color (of 7.1 g, 40 mmol). The obtained Sol (7,1 g, 40 mmol) dissolved in toluene (100 ml)add a solution of 4-(2-chloroethyl)of the research (6.5 g, 43 mmol) in toluene (100 ml)and the resulting mixture is refluxed for 24 hours. After cooling to room temperature the precipitate are filtered, thoroughly washed with toluene, and the obtained filtrate was concentrated in vacuo, receiving the connection 10 in the form of a solid light-yellow color (7.2 g, 57% of the total output), which is used in the next stage without further purification. 1H NMR (200 MHz, CDCl3): δ=2,54-of 2.64 (m, 4H, 3-H2, 5-H2), 2,84 (t, J=5.5 Hz, 2H, 1'-H2), 3,69-of 3.78 (m, 4H, 2-H2A 6-H2), 4,20 (t, J=5.5 Hz, 2H, 2'-H2), 6,92-7,01 (m, 2H, 2"H, 6"-H), 8,16 is 8.25 (m, 2H, 3"-H, 5"-H) ppm;13C NMR (50,3 MHz, CDCl3): δ=54,1 (C-3, C-5), 57,3 (C-1'), 66,7 (C-2'), 66,9 (C-2, C-6), of 114.5 (C-2', C-6"), 125,9 (C-3, C-5"), 141,6 (C-4"), of 163.7 (C-1') ppm; MC (70 eV, EI): m/z (%) = 252 (8) [M]+, 100 (100) [M-CH2OC6H4-NO2]+. Example 6 4-(2-(Morpholine-4-yl)ethoxy)phenylamine (11) A solution of compound 10 (6,00 g of 23.8 mmol) in 95% EtOH (40 ml) hydronaut over Pd/C (10%, 350 mg) at 58 psi H2for 1 hour at room temperature. The resulting solid product is removed by filtration through celite, which is washed thoroughly with EtOH (100 ml) and MeOH (50 ml); the resulting filtrate was concentrated in vacuo, receiving the connection 11 (5.29 g, quantitative) as a reddish brown oil, which was used in the next stage without further purification. 1H NMR (300 MHz, CDCl3): δ=2.57 m (mwith, 4H, 3"-H2, 5"-H2), was 2.76 (t, J=5.8 Hz, 2H, 2'-H2), 3,40 (Cuser., 2H, NH2), to 3.73 (mwith, 4H, 2"-H2, 6"-H2), a 4.03 (t, J=5.8 Hz, 2H, 1'-H2), 6,59 of 6.66 (m, 2H, 2-H, 6-H), of 6.71-of 6.78 (m, 2H, 3-H, 5-H) ppm;13C NMR (50,3 MHz, CDCl3): δ=54,0 (C-3, C-5"), 57,7 (C-2'), to 66.3 (C-1'), to 66.8 (C-2', C-6"), by 115.7 (C-3, C-5), to 116.2 (C-2, C-6), 140,2 (C-1), to 151.7 (C-4) ppm; MC (70 eV, EI): m/z (%) = 222 (17) [M]+, 100 (100) [M-CH2OC6H4-NH2]+. Example 7 Ethyl ester of 5-(2-(morpholine-4-yl)ethoxy)-1H-indole-2-carboxylic acid (12) Stir a solution of 4-(2-(morpholine-4-yl)ethoxy)phenylamine (11 (4,00 g, 18.0 mmol) in water (38 ml) and concentrated HCl (12 ml) is treated dropwise at 0ºC solution NaNO2(1,36 g of 19.8 mmol) in water (3.8 ml)and the resulting mixture is stirred for 30 minutes at 0ºC (solution A). Ethyl 2-methylacetoacetate (2,75 ml of 18.9 mmol) is added dropwise to a suspension of NaOAc (15.3 g) in EtOH (29 ml) at 0ºC. After stirring for 30 minutes at this temperature, add ice (18 g) (solution B). Then solution A is added to solution B, moving the cannula at 0ºC, the mixture was left to warm to room temperature and stirred for another 2.5 hours. Then the reaction mixture was alkalinized by slowly adding saturated aqueous solution of Na2CO3at 0ºC, and extracted with CH2Cl2(3×200 ml). The combined organic layers washed with water (300 ml), dried (MgSO4) and the solvent is removed in vacuum. The residue is then dissolved in absolute EtOH (15 ml), treated with freshly prepared saturated solution of HCl in absolute EtOH (15 ml) and refluxed for 40 minutes. After cooling to room temperature the solvent is removed under reduced pressure and the residue is divided between water (50 ml) and CH2Cl2(100 ml). The aqueous layer was alkalinized using a saturated aqueous solution of Na2CO3and extracted with CH2Cl2(3×100 ml). The combined organic layers are washed with the salt solution (200 ml), dried (MgS04) and concentrated in vacuo. As a result of crystallization of iPr2O and processing by means of column chromatography (CH2Cl2/MeOH, 20:1) residue. After evaporation of the mother liquor get a connection 12 (4,07 g, 71% total yield) as a solid yellow color. 1H NMR (300 MHz, CDCl3): δ=1,41 (t, J=7,1 Hz, 3H, OCH2CH3), 2,61 (mwith, 4H, 3"-H2, 5"-H2), 2,84 (t, J=5.7 Hz, 2H, 2'-H2), 3,76 (mwith, 4H, 2"-H2, 6"-H2), is 4.15 (t, J=5.7 Hz, 2H, 1'-H2), and 4.40 (q, J=7,1 Hz, 2H, OCH2CH3), of 6.99 (DD, J=9,0, 2.4 Hz, 1H, 6-H), was 7.08 (d, J=2.4 Hz, 1H, 4-H), 7,13 (d, J=1.3 Hz, 1H, 3-H), 7,30 (d, J=9.0 Hz, 1H, 7-H), 9,13 (Cuser., 1H, NH) ppm;13C NMR (50,3 MHz, CDCl3): δ=14.4V (OCH2CH3), 54,1 (C-3, C-5"), 57,7 (C-2'), 60,9 (OCH2CH3), 66,2 (C-1'), 66,9 (C-2', C-6"), for 103.7 (C-4), 108,1 (C-3), 112,7 (C-7), 117,3 (C-6), 127,7, to 127.9 (C-2, C-3a), 132,3 (C-7a), 153,7 (C-5), 162,0 (C=O) ppm; MC (70 eV, EI): m/z (%) = 318 (14) [M]+, 100 (100) [CH2N(CH2)4On]+; C17H22N2O4(318,37): calculated C 64,13, N 6,97; found C 63,85, N 7,12. Example 8 Hydrochloride 5-(2-(morpholine-4-yl)ethoxy)-1H-indole-2-carboxylic acid (13) A suspension of ester compounds 12 (1,02 g, 3,20 mmol) in MeOH (8 ml) is treated with NaOH solution (150 mg, 3.75 mmol) in water (4 ml) and refluxed for 3 hours. After cooling to room temperature the pH of the solution was adjusted to pH 6 using 1M HCl, and actuarial removed under reduced pressure. The residue is dissolved in MeOH, is added dropwise 1M HCl and the resulting precipitate is collected by filtration, receiving the connection 13 (727 mg, 70%) as a solid brown color. Purification of the residue obtained upon evaporation of the mother liquor using a column filtration on silica gel (MeOH, 1% conc. HCl), gives the second connection portion 13 (232 mg, 22%). 1H NMR (300 MHz, DMSO-d6): δ=3,07-3,61 (m, 6H, 2'-H2, 3"-H2, 5"-H2), 3,76-4,06 (m, 4H, 2"-H2, 6"-H2), 4,43 (t, J=5.0 Hz, 2H, 1'-H2), 6,98 (DD, J=8,9, and 2.3 Hz, 1H, 6-H), 7,01 (d, J=1.8 Hz, 1H, 3-H), 7,20 (d, J=2.3 Hz, 1H, 4-H), 7,37 (d, J=8,9 Hz, 1H, 7-H), is 11.39 (Cuser., 1H, NH+), 11,65 (c, 1H, NH), 12,85 (Cuser., 1H, CO2H) ppm;13C NMR (50,3 MHz, DMSO-d6): δ=51,6 (C-3, C-5"), 54,9 (C-2'), of 62.8 (C-1'), to 63.1 (C-2', C-6"), to 103.9 (C-4), 106,9 (C-3), 113,5 (C-7), 115,9 (C-6), 127,0 (C-2), 129,0 (C-3a), 133,0 (C-7a), to 152.0 (C-5), 162,6 (C=O) ppm; MC (70 eV, EI): m/z (%) = 290 (6) [M-Cl]+, 100 (100) [CH2N(CH2)4On]+; C15H19ClN2O4(326,78): calculated C 55,13, N 5,86; found C 54,96, N 5,81. Example 9 (+)-{(1,10)-Anti-5-benzyloxy-3-[(5-(2-(morpholine-4-yl)ethoxy)indol-2-yl)carbonyl]-1-(10-chloroethyl)-1,2-dihydro-3H-Benz[e]indol} ((+)-3b) (+)-1 (150 mg, 342 μmol) suspended in a mixture of 4M HCl/ethyl acetate (14 ml) and stirred at room temperature for 3 hours. The resulting mixture was then concentrated and dried in vacuum for 1 hour. The residue is dissolved in DMF (10 ml), the solution is Hledat to 0ºC, and added EDC·HCl (197 mg, 1,03 mmol, 3.0 equiv.) and the connection 13 (146 mg, 445 mmol, 1.3 equiv.). After 21 hours at room temperature, add ethyl acetate (50 ml), water (50 ml) and saturated aqueous solution of NaHCO3(50 ml)and the resulting mixture is extracted with ethyl acetate (4×50 ml). The combined organic layers are dried brine (4×100 ml), dried over MgSO4filter and concentrate. As a result of processing using column chromatography (CH2C12/MeOH = 30:1) receive (+)-3b in the form of a solid pale brown (156 mg, 256 mmol, 75%). 1H-NMR (300 MHz, CDCl3): δ=1,62 (d, J=6,7 Hz, 3H, 11-CH3), 2,61 (mwith, 4H, 3"'-H2, 5"'-H2), and 2.83 (t, J=5.8 Hz, 2H, 2"-H2), 3,76 (mwith, 4H, 2"'-H2, 6"'-H2), 3,89-3,98 (m, 1H, 1-H)to 4.15 (t, J=5.8 Hz, 2H, 1"-H2), 4,47-br4.61 (m, 2H, 2-Ha, 10-H), 4,85 (mwith, 1H, 2-Hbin ), 5.25 (mwith, 2H, OCH2Ph), 6,94-7,02 (m, 2H, 3'-H, 6'-H), 7,12 (d, J=2.3 Hz, 1H, 4'-H), 7.29 trend-EUR 7.57 (m, 8H, 7-H, 7'-H, 8-H, 5×Ph-H), to 7.68 (d, J=8,2 Hz, 1H, 9-H), 8,19 (Cuser., 1H, 4-H), to 8.34 (d, J=8,2 Hz, 1H, 6-H), 9,88 (Cuser., 1H, NH);13C-NMR (75 MHz, CDCl3): δ=24,0 (11-CH3), and 47.5 (C-1), 53,6 (C-2), 54,1 (C-3"', C-5"'), of 57.8 (C-2'), was 59.9 (C-10), to 66.3 (C-1'), 66,9 (C-2"', C-6"'), 70,3 (OCH2Ph), was 98.2 (C-4), for 103.7 (C-4'), or 105.8 (C-3'), of 112.8 (C-7'), 117,0, 117,2 (C-6', C-5a), 122,6 (C-9), 123,7, 123,7, 123,9 (C-6, C-7, C-9b), 127,4, 127,6, 127,9, 128,1, 128,5 (C-3a', C-8, 5×Bn-CH), 130,0, 130, 8mm, 131, 5mm (C-2', C-7a', C-9a), 136,7 (Bn-C), 142,4 (C-3a), 153,7 (C-5'), 155,6 (C-5), 160,5 (C=O); MC (EI, 70 eV): m/z (%) = 609,2 (1) [M] +, 573,1 (3) [M-Cl-H]+, 100,0 (100) [CH2(CH2)4On]+. C36H36CIN3O4(610,14): calculated 609,2394 found 609,2394 (EI-HRMS). Example 10 (+)-{(1,10)-Anti-1-(10-chloroethyl)-5-hydroxy-3-[(5-(2-(morpholine-4-yl)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic} ((+)-4b) A solution of (+)-3b (80 mg, 131 μmol) in a mixture of 4M HCl/ethyl acetate (10 ml) was stirred at room temperature for 2 hours. The resulting mixture was then concentrated and dried in vacuum for 1 hour. The residue is suspended in THF (8 ml), add 10% palladium on charcoal (28 mg) and monitorial (25% aqueous solution of 0.28 ml). The reaction mixture is stirred at 40 for 1 hour and then filtered through celite, which is washed with methanol (150 ml). The obtained filtrate is concentrated, and the residue purified using column chromatography (CH2C12/MeOH = 5:1, with 0.1% conc. HCl)to give (+)-4b in the form of a solid yellow (67 mg, 120 μmol, 92%). 1H-NMR (300 MHz, DMSO-d6): δ=1,62 (d, J=6.5 Hz, 3H, 11-CH3), 3,14-3,63 (m, 6H, 2"-H2, 3"'-H2, 5"'-H2), 3,83-a 4.03 (m, 4H, 2"'-H2, 6"'-H2), 4,12-is 4.21 (m, 1H, 1-H), to 4.41 with 4.64 (m, 3H, 1"-H2, 2-Ha), with 4.64-4,82 (m, 2H, 2-Hb, 10-H), 7,00 (DD, J=9,0, 1.8 Hz, 1H, 6'-H), 7,16 (Cuser., 1H, 3'-H), 7,25 (mwith, 1H, 4'-H), 7,35 (mwith, 1H, 7-H), 7,41-of 7.55 (m, 2H, 7'-H, 8-H), 7,88 (d, J=8,4 Hz, 1H, 9-H), 7,98 (Cuser., 1H, 4-H), to 8.14 (d, J=8,3 Hz, 1H, 6-H), 10,42(c, 1H, OH), 11,65 (c, 1H, NH), 11,93 (Cuser., 1H, NH+);13C-NMR (75 MHz, DMSO-d6): δ=23,4 (11-CH3), TO 45.9 (C-1), 51,6 (C-3"', C-5"'), OF 52.1 (C-2), 54,9 (C-2'), IS 61.5 (C-10), OF 62.8 (C-1'), 63,0 (C-2"', C-6"'), TO 100.4 (C-4), 104,0 (C-4'), OF 105.2 (C-3'), OF 113.2 (C-7'), 115,6, 115,9 (C-6', C-5a), 122,2 (C-9b), 122,8, 122,9, 123,1 (C-6, C-7, C-9), 126,9 (C-8), 127,4 (C-3a'), 129,8, 131,3, 131,9 (C-2', C-7a', C-9a), to 142.1 (C-3a), up 151.9 (C-5'), other 153.9 (C-5), 159,8 (C=O); MC (ESI): m/z (%) = 520,1 (100) [M-Cl]+; C29H31Cl2N3O4(556,48): calculated 520,2003 found 520,1998, [M-C1]+(ESI-HRMS). Example 11 [(+)-(1,10)-Anti-1-(10-chloroethyl)-3-[(5-(2-(morpholine-4-yl)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-2,3,4,6-Tetra-O-acetyl-β-D galactopyranoside ((+)-7b) A solution of (+)-5 (142 mg, 408 μmol) in dry CH2C12(18 ml) and molecular sieves 4Ǻ (0.8 g) is stirred for 30 minutes at room temperature. After adding O-(2,3,4,6-Tetra-O-acetyl-β-D-galactopyranosyl)trichloroacetimidate (6) (207 mg, 0,420 mmol, of 1.03 equiv.), the resulting mixture was cooled down to-10ºC and slowly add a solution BF3·OEt2(26 μl, 205 mmol, 0.5 equiv.) in dry CH2C12(2.0 ml). After 4 hours at-10ºC add additional portion BF3·OEt2(0,155 ml, 1,22 mmol, 3.0 equiv.) in dry CH2C12(1.9 ml)and the resulting mixture is heated to room temperature. After 5 hours the solution is separated from the molecular sieves, molecular sieves washed thoroughly with CH2Cl2(2×10 ml) and the volume of the United organic portion was concentrated in vacuo. The residue is dried in vacuum for 1 hour and then dissolved in dry DMF (19 ml). The solution is cooled to 0ºC, and added EDC·HCl (235 mg, of 1.23 mmol, 3.0 equiv.) and compound 13 (200 mg, 0,612 mmol, 1.5 equiv.). After 20 hours at room temperature, the reaction mixture was diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3(50 ml) and then extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2Cl2/MeOH = 10:1) receive N-oxide (+)-7b in the form of a solid pale brown (142 mg, 164 μmol, 40%). To a solution of N-oxide (67 mg, 77 μmol) in dry ethanol (15 ml) add PtO2·H2O (6 mg, 21 mmol, 0.3 equiv.). Then after the reaction mixture was bubbled hydrogen for 5 hours. The reaction mixture was filtered through celite and the celite is washed thoroughly with methanol (50 ml). The obtained filtrate was concentrated in vacuo and the residue purified using column chromatography (CH2Cl2/MeOH = 10:1)to give (+)-7b in the form of a solid yellow (53 mg, 62 μmol, 81%). C43H48ClN3O13(850,31): calculated 850,2954 found 850,2948, [M+H]+(ESI-HRMS). Example 12 [(+)-(1,10)-Anti-1-(10-chloroethyl)-3-[(5-(2-(morpholine-4-yl)ethoxy)in the ol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-β-D galactopyranoside ((+)-8b) To a solution of compound 7a (51 mg, 60 μmol) in dry MeOH (6 ml) was added NaOMe in methanol (22 μl to about 5,4M solution, 120 μmol, 2.0 equiv.). The reaction mixture was stirred at room temperature for 20 minutes and diluted with water (2 ml) and methanol (2 ml). Add ion-exchange resin (Amberlite-IR 120) up until the pH of the solution does not reach the neutral values. The resin is removed and washed with methanol (10 ml). The combined organic portion was concentrated in vacuo and the residue purified using column chromatography (gradient: CH2Cl2/MeOH = 10:1 // CH2Cl2/MeOH = 4:1). The obtained solid is washed with pentane (4×15 ml)to give compound 8b in the form of a solid yellow (37 mg, 54 μmol, 90%). 1H-NMR (600 MHz, DMSO-d6): δ=1,65 (d, J=6.6 Hz, 3H, 11-CH3), 2,48-2,52 (m, 4H, 3"'-H2, 5"'-H2), by 2.73 (t, J=5.8 Hz, 2H, 2"-H2), 3,45-3,70 (m, 8H, 2"'-H2, 6"'-H2, 3""-H, 5""-H, 6""-H2), 3,76-of 3.85 (m, 2H, 2""-H, 4""-H), 4,11 (t, J=5.8 Hz, 2H, 1"-H2), 4,25 (mwith, 1H, 1-H), to 4.52 with 4.65 (m, 3H, 2-Ha, 2×OH), 4,72-rate 4.79 (m, 1H, 2-Hb), 4,79-4,88 (m, 2H, 10-H, OH), 4.92 in (mwith, 1H, 1"'-H), and 5.30 (Cuser., 1H, OH), 6,92 (DD, J=8,9, and 2.4 Hz, 1H, 6'-H), 7,14-7,20 (m, 2H, 3'-H, 4'-H), 7,40 (d, J=8,8 Hz, 1H, 7'-H), 7,43 (mwith, 1H, 7-H), EUR 7.57 (mwith, 1H, 8-H), of 7.96 (d, J=8,4 Hz, 1H, 9-H), by 8.22 (Cuser., 1H, 4-H), at 8.36 (d, J=8.5 Hz, 1H, 6-H), 11,63 (c, 1H, NH);13C-NMR (150 MHz, DMSO-d6): δ=23,4 (11-CH3), 46,0 (C-1), TO 52.1 (C-2), 53,6 (C-"', C-5"'), 57,1 (C-2'), 59,5 (C-6""), WITH 61.3 (C-10), 65,9 (C-1'), 66,2 (C-2"', C-6"'), OF 67.5 (C-4""), 70,6 (C-2'"), AND 73.2 (C-3""), A 75.1 (C-5""), 101,9 (C-4), TO 102.3 (C-1""), 103,4 (C-4'), 105,4 (C-3'), of 113.2 (C-7'), 115,9 (C-6'), 118,9 (C-5a), 122,9, 123,0, 123,4, 123,7 (C-6, C-7, C-9, C-9b), RUB 127.3, of 127.5 (C-3a', C-8), 129,5, 130,9, 131,7 (C-2', C-7a', C-9a), 142,0 (C-3a), 152,9 (C-5'), of 153.6 (C-5), 160,1 (C=O); MC (ESI): m/z (%) = 1385,1 (13) [2M+Na]+, 704,3 (100) [M+Na]+, 682,3 (32) [M+H]+; C35H40ClN3O9(682,16): calculated 682,2531 found 682,2526, [M+H]+(ESI-HRMS). Example 13 3-(2-Chloroethoxy)-4-methoxybenzaldehyde (15) A mixture of 3-hydroxy-4-methoxybenzaldehyde (14) (10.0 g, to 65.7 mmol), K2CO3(to 45.4 g, 329 mmol), 1,2-dichloroethane (104 ml, 1,31 mol) and DMF (300 ml) was stirred at 65-70ºC for 16 hours. After cooling, 1,2-dichloroethane is removed under reduced pressure, the remaining suspension was poured on ice and the resulting mixture was extracted with Et2O (3×250 ml) and EtOAc (4×250 ml). The combined organic layers washed with water (4×400 ml) and brine (2×400 ml), dried (MgSO4) and the solvent is removed in vacuum. As a result of crystallization from a mixture of EtOAc/hexane get the connection 15 (11.9 g, 84%) as colorless needles. 1H NMR (200 MHz, CDCl3): δ=3,88 (t, J=6,1 Hz, 2H, 2'-H2), 3,97 (c, 3H, OMe), 4,35 (t, J=6,1 Hz, 2H, 1'-H2), 7,01 (d, J=8,1 Hz, 1H, 5-H), the 7.43 (d, J=1.8 Hz, 1H, 2-H), 7,51 (DD, J=8,1, 1.8 Hz, 1H, 6-H), 9,86 (c, 1H, CHO) ppm;13C NMR (50,3 MHz, CDCl3): δ=41,5 (C-2'), 56,2 (OMe), for 68.9 (C-1'), 111,0, 111,3 (C-2, C-5), 127,4 (C-6), 129, 9mm (C-1), 148,1 (C-3), 54,9 (C-4), 190,7 (CHO) ppm; MC (70 eV, EI): m/z (%) = 214 (100) [M]+, 151 (57) [M-(CH2)2Cl]+; C10H11ClO3(214,65): calculated C 55,96, N 5,17; found C 56,04, N equal to 4.97. Example 14 Methyl ester 2-azido-3-[3-(2-chloroethoxy)-4-methoxyphenyl]acrylic acid (16) NaN3(22,2 g, 341 mmol) is added slowly to a solution of METHYLCHLOROSILANE (20,0 ml, 227 mmol) in DMSO (100 ml). After stirring at room temperature for 24 hours, add water (150 ml)and the resulting mixture was extracted with Et2O (3×100 ml). The combined organic fractions are dried (MgSO4) and concentrated in vacuo to 50 ml and Then add a solution of aldehyde 15 (lower than the 5.37 g, 25,0 mmol) in MeOH (50 ml)and the resulting mixture is cooled to-30ºC. Then the reaction mixture is treated with a mixture of 5,4M NaOMe/MeOH (35,0 ml, 189 mmol) for 30 minutes at-30ºC, heated to 0ºC and diluted with MeOH (50 ml). After stirring for 16 hours at 0ºC, add water (200 ml) and the resulting mixture was extracted with CH2Cl2(3×200 ml). The combined organic fractions washed with brine (200 ml), dried (MgSO4) and the solvent is removed in vacuum, obtaining the connection 16 (6,84 g, 88% out of 15) in the form of a solid pale yellow color, which is used in the next stage without further purification. 1H NMR (300 MHz, CDCl3): δ=a-3.84 (t, J=6,1 Hz, 2H, 2"-H2), a 3.87, 3,88 (2×c, 6H, OMe, CO2Me), the 4.29 (t, J=6,1 Hz, 2H, 1"-H2 ), 6,83 (c, 1H, 1'-H), 6.87 in (d, J=8.5 Hz, 1H, 5-H), was 7.36 (DD, J=8,5, and 2.1 Hz, 1H, 6-H), 7,53 (d, J=2.1 Hz, 1H, 2-H) ppm;13C NMR (50,3 MHz, CDCl3): δ=41,6 (C-2'), 52,8 (CO2Me)56,0 (OMe)and 69.3 (C-1'), 111,5 (C-2), to 116.2 (C-5), 123,4 (C-2'), 125,4, 126,0, 126,2 (C-1, C-1', C-6), 147,1, 150,9 (C-3, C-4), 164,1 (C=O) ppm Example 15 Methyl ester 5-(2-chloroethoxy)-6-methoxy-1H-indole-2-carboxylic acid (17) A solution of compound 16 (6.81 in) to 21.8 mmol) in toluene (200 ml) is refluxed for 4 hours. After cooling to room temperature the reaction mixture was concentrated in vacuo. The resulting precipitates are filtered and dried in vacuum, obtaining the connection 17 (4,48 g, 72%) as a solid pale yellow color. 1H NMR (300 MHz, DMSO-d6): δ=3,82, 3,84 (2×c, 6H, OMe, CO2Me)3,93 (t, J=5.3 Hz, 2H, 2'-H2), is 4.21 (t, J=5.3 Hz, 2H, 1'-H2), 6,92 (c, 1H, 7-H), 7,02 (d, J=1.5 Hz, 1H, 3-H), 7,15 (c, 1H, 4-H), are 11.62 (Cuser., 1H, NH) ppm -13C NMR (50,3 MHz, DMSO-d6): δ=42,9 (C-2'), 51,3 (CO2Me), is 55.5 (OMe), 69,4 (C-1'), or 94.8 (C-7), is 105.6 (C-4), 107,9 (C-3), 119,7 (C-3a), and 125.4 (C-2), 133,2 (C-7a), 144,1 (C-5), 149,8 (C-6), 161,4 (C=O) ppm MC (70 tV, EI): m/z (%) = 283(100) [M]+, 220 (50) [M-(CH2)2Cl]+- C13H14ClNO4(283,71): calculated C 55,04, N equal to 4.97, found C 54,86, N is 5.06. Example 16 5-(2-Chloroethoxy)-6-methoxy-1H-indole-2-carboxylic acid (18) A suspension of ester 17 (2.00 g, 7.05 mmol), Cs2CO3(3,45 g, 10.6 mmol), 95% EtOH (40 ml) and water (20 ml) is boiled with the reverse was built in the ICOM for 8 hours. After cooling to room temperature the solvent is removed in vacuum, the residue is treated with water (50 ml) and the resulting solution was acidified with 2M HCl. The resulting precipitates are filtered, washed with water (100 ml) and dried in vacuum, obtaining the connection 18 (1.80 g, 95%) as a solid beige color. 1H NMR (300 MHz, DMSO-d6): δ=3,81 (c, 3H, OMe), 3,93 (t, J=5.3 Hz, 2H, 2'-H2), is 4.21 (t, J=5.3 Hz, 2H, 1'-H2), 6,91 (c, 1H, 7-H), of 6.96 (d, J=2.1 Hz, 1H, 3-H), 7,15 (c, 1H, 4-H), 11,44 (Cuser., 1H, NH), 12,57 (Cuser., 1H, CO2H) ppm;13C NMR (50,3 MHz, DMSO-d6): δ=43,0 (C-2'), at 55.6 (OMe), 69,5 (C-1'), 94,9 (C-7), or 105.8 (C-4), 107,5 (C-3), to 119.8 (C-3a), to 126.8 (C-2), 133,0 (C-7a), 143,9 (C-5), of 149.5 (C-6), 162,4 (C=O) ppm; MC (70 eV, EI): m/z (%) = 269 (100) [M]+, 206 (58) [M-(CH2)2Cl]+; C12H12ClNO4(269,68): calculated C 53,44, N 4,49, found C 53,54, N 4,29. Example 17 Hydrochloride 5-(2-(N,N-dimethylamino)ethoxy)-6-methoxy-lH-indole-2-carboxylic acid (19) The mixture of acid 18 (300 mg, 1.11 mmol), 40% aqueous solution of Me2NH (2,81 ml of 22.2 mmol), Na2CO3(295 mg, 2,78 mmol) and water (20 ml) was stirred at 100ºC for 1.5 hours. After cooling to room temperature the solvent is removed in vacuum, the residue is dissolved in water (15 ml) and the resulting solution was acidified using 2M HCl. Then the solution is evaporated to dryness and the resulting crude product was then purified using column chromatography on silicagel is e (MeOH/CH 2Cl2, 10:1, 1% conc. HCl), getting a solid green color, which is dissolved in water. Insoluble silica gel was removed by filtration, the water is removed under reduced pressure and the residue is dried in vacuum, obtaining the connection 19 (343 mg, 98%) as a solid gray-green color. 1H NMR (300 MHz, DMSO-d6): δ=2,87 (c, 6H, NMe2), to 3.49 (t, J=5.0 Hz, 2H, 2'-H2), 3,82 (c, 3H, OMe), 4,34 (t, J=5.0 Hz, 2H, 1'-H2), 6,94 (c, 1H, 7-H), 6,97 (d, J=1.8 Hz, 1H, 3-H), 7.23 percent (c, 1H, 4-H), 10,99 (Cuser., 1H, NH+), 11,50 (Cuser.1H, NH), 12,60 (Cuser., 1H, CO2H) ppm;13C NMR (50,3 MHz, DMSO-d6): δ=42,7 (NMe2), 55,2 (C-2'), at 55.6 (OMe), and 64.5 (C-1'), or 94.8 (C-7), to 106.2 (C-4), 107,5 (C-3), 119,7 (C-3a), 127,0 (C-2), 133,2 (C-7a), 143,4 (C-5), 149,4 (C-6), 162,4 (C=O) ppm; MC (70 eV, EI): m/z (%) = 278 (7) [M-HCl]+, 58 (100) [CH2NMe2]+; C14H19C1N2O4(314,76): calculated 278,1267 [M-HCl]+found 278,1267. Example 18 (+)-{(1,10)-Anti-5-benzyloxy-3-[(5-(2-(N,N-dimethylamino)ethoxy)-6-methoxyindol-2-yl)carbonyl]-1-(10-chloroethyl)-1,2-dihydro-3H-Benz[e]indol} ((+)-3c) (+)-1 (150 mg, 342 μmol) suspended in a mixture of 4M HCl/EtOAc and stirred at room temperature for 3 hours. The reaction mixture was concentrated and dried in vacuum for 1.5 hours. The residue is dissolved in DMF (10 ml). The solution is cooled to 0ºC and added EDC·HCl (197 mg, 1,03 mmol, 3.0 equiv.) and the connection 19 (140 mg, 445 mmol, 1.3 equiv.). The reaction mixture was paramesh what happens at room temperature for 20 hours, dilute with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3(50 ml) and extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2C12/MeOH = 10:1) get a connection (+)-3c in the form of a solid pale brown (134 mg, 224 mmol, 66%). 1H-NMR (300 MHz, CDCl3): δ=1,60 (d, J=6,7 Hz, 3H, 11-CH3), 2,35 (c, 6H, NMe2), 2,80 (t, J=6,1 Hz, 2H, 2"-H2), or 3.28 (c, 3H, OCH3), 3,89-3,98 (m, 1H, 1-H), 4.09 to (t, J=6,1 Hz, 2H, 1"-H2), 4,48 with 4.65 (m, 2H, 2-Ha, 10-H), 4,78-4,88 (m, 1H, 2-Hb), 5,26 (mwith, 2H, OCH2Ph)of 6.71 (c, 1H, 7'-H), 6,98 (d, J=1.5 Hz, 1H, 3'-H), 7,07 (c, 1H, 4'-H), 7,22-7,43, 7,45-7,56 (2×m, 7H, 7-H, 8-H, 5×Ph-H), to 7.67 (d, J=8,2 Hz, 1H, 9-H), 8,35 (d, J=8,3 Hz, 1H, 6-H), 8,39 (Cuser., 1H, 4-H), is 10.68 (Cuser., 1H, NH);13C-NMR (75 MHz, CDCl3): δ=23,8 (11-CH3), 45,8 (NMe2), with 47.4 (C-1), 53,5 (C-2), 55,2 (OCH3), 58,0 (C-2'), 59,7 (C-10), of 67.4 (C-1'), 70,4 (OCH2Ph), for 93.9 (C-7'), to 98.5 (C-4), to 104.8 (C-4'), 106,5 (C-3'), 117,3 (C-5a), of 120.5 (C-3a'), 122,4 (C-9), 123,5, 123,6, 123,9 (C-6, C-7, C-9b), 127,5 (2 signals), 127,9, 128,5, (C-8, 5×Bn-CH), 128,7, 129, 9mm, 132,3 (C-2', C-7a', C-9a), 136,6 (Bn-C), 142,7 (C-3a), 145,0 (C-5'), of 150.4 (C-6'), 155,5 (C-5), 160,5 (C=O); MCS (ESI): m/z (%) = 1217,0 (19) [2M+Na]+, 598,2 (100) [M+H]+; C35H36ClN3O4(598,13): calculated 598,2473 found 598,2467, [M+H]+(ESI-HRMS). Example 19 (+)-{(1,10)-Anti-1-(10-chlorid the l)-5-hydroxy-3-[(5-(2-(N,N-dimethylamino)ethoxy)-6-methoxyindol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic} ((+)-4c) The compound (+)-3c (80 mg, 134 μmol) dissolved in a mixture of 4M HCl/ethyl acetate (15 ml) and stirred for 2 hours at room temperature. The solution concentrate. The residue is dried in vacuum for 1 hour and then suspended in THF (8 ml). Then add 10% palladium on charcoal (29 mg) and monitorial (25% aqueous solution of 0.29 ml) at room temperature. The reaction mixture is stirred for 20 minutes at 40ºC and filtered through celite, which is washed thoroughly with methanol (150 ml). The obtained filtrate was concentrated in vacuo, and the residue purified using column chromatography (CH2Cl2/MeOH = 5:1, with 0.1% conc. HCl)to give compound (+)-4c in the form of a powder green-yellow (63 mg, 116 μmol, 86%). 1H-NMR (300 MHz, DMSO-d6): δ=1,63 (d, J=6.6 Hz, 3H, 11-CH3), 2,89 (c, 6H, NMe2), 3,51 (mwith, 2H, 2"-H2), 3,84 (c, 3H, OCH3), 4,11-4,20 (mwith, 1H, 1-H), 4,39 (mwith, 2H, 1"-H2), 4,50-4,82 (m, 3H, 2-H2, 10-H),? 7.04 baby mortality (c, 1H, 7'-H), 7,14 (d, J=1.5 Hz, 1H, 3'-H), 7,30 (c, 1H, 4'-H), 7,34 (mwith, 1H, 7-H), 7,50 (mwith, 1H, 8-H), 7,87 (d, J=8,2 Hz, 1H, 9-H), 7,99 (c, 1H, 4-H), 8,13 (d, J=8,2 Hz, 1H, 6-H), the 10.40 (c, 1H, OH), 11,18 (Cuser., 1H, NH+), 11,51 (Cuser., 1H, NH);13C-NMR (75 MHz, DMSO-d6): δ=23,4 (11-CH3), 42,8 (NMe2), to 45.9 (C-1), to 52.1 (C-2), at 55.3 (C-2'), 55,6 (OCH3), is 61.5 (C-10), a 64.6 (C-1'), to 94.7 (C-7'), to 100.4 (C-4), 105,9 (C-3'), is 106.6 (C-4'), 115,6 (C-5a), 120,1 (C-3a'), 122,1 (C-9b), the 122.7, 122,8 (C-7, C-9), 123,1 (C-6), 126,9 (C-8), 129,4, 129,8 (C-2',C-9a), 132,3 (C-7a'), 142,3 (C-3a), 143,4 (C-5'), 149,3 (C-6'), 153,8 (C-5), 159,7 (C=O); MCS (ESI): m/z (%) = 508,2 (100) [M-Cl]+; C28H31Cl2N3O4(544,47): calculated 508,2003 found 508,1998, [M-C1]+(ESI-HRMS). Example 20 [(+)-(1S,10R)-1-(10-Chloroethyl)-3-[(5-(2-(N,N-dimethylamino)ethoxy)-6-methoxyindol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-2,3,4,6-Tetra-O-acetyl-β-D galactopyranoside ((+)-7c) A solution of (+)-5 (106 mg, 305 μmol) in dry CH2Cl2(14 ml) and molecular sieves 4Ǻ (0.8 g) is stirred for 30 minutes at room temperature. After adding O-(2,3,4,6-Tetra-O-acetyl-α-D-galactopyranosyl)trichloroacetimidate (6) (155 mg, 314 μmol, of 1.03 equiv.) the resulting mixture was cooled down to-10ºC and slowly add a solution BF3·OEt2(19 μl, 152 μmol, 0.5 equiv.) in dry CH2Cl2(1.5 ml). After 4 hours at-10ºC add additional portion BF3·OEt2(116 μl, 914 mmol, 3.0 equiv.) in dry CH2Cl2(1,4 ml)and the resulting mixture is heated to room temperature. After 5 hours the resulting solution is separated from the molecular sieves, molecular sieves washed thoroughly with CH2Cl2(2×10 ml)and the combined organic portion was concentrated in vacuo. The residue is dried in vacuum for 1 hour and then dissolved in dry DMF (14 ml). The solution is cooled to 0ºC, and added EDC·HCl (175 mg, 914 mmol, 3.0 equiv.) and the connection 19 (144 mg, 457 mmol, 1.5 equiv.). After 2 hours at room temperature, the reaction mixture was diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3(50 ml) and then extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. After processing by means of column chromatography (CH2Cl2/MeOH = 10:1) get a connection (+)-7c in the form of a solid pale brown (128 mg, 153 μmol, 50%). C42H48ClN3O13(838,30): calculated 838,2957 found 838,2948, [M+H]+(ESI-HRMS). Example 21 [(+)-(1S,10R)-1-(10-Chloroethyl)-3-[(5-(2-(N,N-dimethylamino)ethoxy)-6-methoxyindol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-β-D galactopyranoside ((+)-8c) To a solution of compound (+)-7c (125 mg, 149 μmol) in dry MeOH (7 ml) was added NaOMe in methanol (55 μl to about 5,4M solution, 298 mmol, 2.0 equiv.). The reaction mixture was stirred at room temperature for 30 minutes and diluted with methanol (9 ml) and water (5 ml). Ion-exchange resin (Amberlite-IR 120) add up until the pH of the solution does not reach the neutral values. The solution was concentrated in vacuo, and the residue purified using column chromatography (CH2Cl2/MeOH = 1:1)to give compound (+)-8c in the form of a solid, pale yellow (86 mg, 128 μmol, 86%). 1H-NMR (600 MHz, DMSO-d6): δ=1,66 (d, J=6,7 Hz, 3H, 11-CH3in ), 2.25 (c, 6H, NMe2), to 2.66 (t, J=6.0 Hz, 2H, 2"-H2), and 3.7 (m with, 1H, 3"'-H), 3,53-3,59 (m, 2H, 5"'-H, 6"'-Ha), to 3.67 (mwith, 1-H, 6"'-Hb), of 3.77-a 3.83 (m, 5H, 2"'-H, 4"'-H, OCH3), of 4.05 (t, J=6.0 Hz, 2H, 1"-H2), 4,24 (mwith, 1H, 1-H), 4,54-of 4.77 (m, 4H, 2×OH, 2-H2), 4,82 (mwith, 1H, 10-H), 4,85-4,99 (m, 2H, 1"'-H, OH), 5,32 (Cuser., 1H, OH), 6,98 (c, 1H, 7'-H), 7,14 (Cuser., 1H, 3'-H), 7,18 (c, 1H, 4'-H), 7,42 (t, J=7.7 Hz, 1H, 7-H), 7,56 (t, J=7,6 Hz, 1H, 8-H), 7,95 (d, J=8.6 Hz, 1H, 9-H), 8,23 (Cuser., 1H, 4-H), 8,35 (d, J=8.6 Hz, 1H, 6-H), 11,48 (c, 1H, NH);13C-NMR (150 MHz, DMSO-d6): δ=23,4 (11-CH3), 45,6 (NMe2), 46,0 (C-1), with 52.0 (C-2), 55,6 (OCH3), OF 57.8 (C-2'), TO 59.6 (C-6"'), WITH 61.3 (C-10), AND 67.2 (C-1'), TO 67.5 (C-4"'), 70,6 (C-2"'), 73,3 (C-3"'), AND 75.2 (C-5"'), TO 94.7 (C-7'), 101,9 (C-4), TO 102.3 (C-1"'), 104,7 (C-4'), 106,1 (C-3'), 118,6 (C-5a), to 120.3 (C-3a'), 122,8 (C-9), 122,9 (C-9b), 123,4 (C-6), to 123.5 (C-7), to 127.2 (C-8), 128,9, of 129.5 (C-2', C-9a), 131,8 (C-7a'), 142,2 (C-3a), 144,6 (C-5'), 149,4 (C-6'), of 153.6 (C-5), 160,1 (C=O); MC (ESI): m/z (%) = 670,3 (100) [M+H]+; C34H40ClN3O9(670,15): calculated 670,2531. found 670,2526, [M+H]+(ESI-HRMS). Example 22 Ethyl ester of 1-methylpiperidin-4-carboxylic acid (21) Ethyl isonipecotate (20) (5,00 g of 31.8 mmol) dissolved in a mixture of ice-glacial acetic acid (3.80 g, of 63.6 mmol) and water (11 ml). Then add 37% aqueous formaldehyde solution (2,85 ml of 38.2 mmol) and the reaction mixture hydronaut over Pd/C (10%, 338 mg) at 58 psi H2within 3.5 hours at room temperature. The hard part is removed by filtration through celite, which is washed thoroughly with water (50 ml)and the resulting filter is t is brought to pH 11, using 1 M NaOH while cooling. The resulting solution was extracted with Et2O (5×100 ml), the combined organic fractions are dried (MgSO4) and the solvent is removed under reduced pressure, obtaining the connection 21 (5,44 g, quantitative) as a colourless liquid, which is used in the next stage without further purification. 1H NMR (300 MHz, CDCl3): δ=1,25 (t, J=7,1 Hz, 3H, OCH2CH3), 1,69-to 2.06 (m, 6H, 2-Hax, 3-H2, 5-H2A 6-Hax), 2,18 of-2.32 (m, 1H, 4-H), and 2.27 (c, 3H, NMe), 2,75-2,87 (m, 2H, 2-HeqA 6-Heq), 4,13 (kV, J=7,1 Hz, 2H, OCH2CH3) ppm -13C NMR (75.5 MHz, CDCl3): δ=14,1 (OCH2CH3), 28,2 (C-3, C-5), 40,5 (C-4), 46,3 (NMe), 54,9 (C-2, C-6), 60,2 (OCH2CH3), 175,0 (C=O) ppm MC (70 eV, EI): m/z (%)= 171 (31) [M]+, 142 (59) [M-CH2CH3]+, 126 (40) [M-OCH2CH3]+, 98 (56) [M-CO2Et]+. -C9H17NO2(171,24). Example 23 (1 Methylpiperidin-4-yl)methanol (22) A solution of ester 21 (10.1 g, or 59.0 mmol) in Et2O (40 ml) is added dropwise to a suspension of LiAlH4(2,46 g, 64,9 mmol) in Et2O (200 ml) at 0ºC. Then the reaction mixture was left to warm to room temperature and then stirred for 4 hours at this temperature. Slowly add water (10 ml) and stirring is continued for a further 30 minutes the Resulting white precipitate are filtered and thoroughly washed with Et2O(250 ml). After removal of the solvent under reduced pressure, the resulting oil is purified by distillation (boiling point 108ºC, 14 mbar)to give alcohol 22 (6,40 g, 84%) as a colourless oil. - Rf= 0,53 (CH2Cl2/MeOH, 5:1, 5% of the NEt3, PMA: dark blue). 1H NMR (300 MHz, CDCl3): δ=1.28 (in DQC, J=12,2, and 3.7 Hz, 2H, 3-Hax, 5-Hax), 1,38-of 1.55 (m, 1H, 4-H), 1,68-to 1.79 (m, 2H, 3-Heq, 5-Heq), of 1.93 (dt, J=11.8 in, and 2.3 Hz, 2H, 2-HaxA 6-Hax), and 2.26 (c, 3H, NMe), 2,82 of 2.92 (m, 2H, 2-HeqA 6-Heq), 3,12 (Cuser., 1H, OH), of 3.46 (d, J=6,4 Hz, 2H, CH2OH) ppm -13C NMR (75.5 MHz, CDCl3): δ=28,8 (C-3, C-5), of 37.9 (C-4), 46,3 (NMe), is 55.5 (C-2, C-6), of 67.4 (CH2OH) ppm MC (70 eV, EI): m/z (%) = 129 (55) [M]+, 128 (100) [M-H]+, 112 (10) [M-OH]+, 98 (21) [M-CH2OH]+. - C7H15NO (129,20). Example 24 1-Methyl-4-(4-nitrophenoxy)piperidine (24) A mixture of 1-chloro-4-nitrobenzene (23) (2.37 g, 15.0 mmol), alcohol 22 (1,94 g, 15.0 mmol) and DMSO (25 ml) is treated with portions of NaH (60% in mineral oil, 660 mg, 16.5 mmol) at 40ºC. The resulting mixture was stirred at 70ºC for 3 hours, poured into water (150 ml) and extracted with Et2O (5×100 ml). The combined organic fractions washed with water (250 ml) and brine (250 ml), dried (MgSO4) and the solvent is removed in vacuum. The obtained solid product is recrystallized from Et2O, when receiving the connection 24 (3.12 g, 83%) as yellow needles. 1H NMR (300 MHz,CDCl 3): δ=1,36-of 1.56 (m, 2H, 3-Hax, 5-Hax), 1,75 is 1.91 (m, 3H, 3-Heq, 4-H, 5-Heq), to 1.98 (dt, J=11,9, 1.9 Hz, 2H, 2-HaxA 6-Hax), 2,30 (c, 3H, NMe), 2,85 are 2.98 (m, 2H, 2-HeqA 6-Heq), 3,90 (d, J=5.8 Hz, 2H, OCH2), 6,94 (mwith, 2H, 2'-H, 6'-H), 8,19 (mwith, 2H, 3'-H, 5'-H) ppm -13C NMR (50,3 MHz, CDCl3): δ=28,9 (C-3, C-5), and 35.1 (C-4), 46,4 (NMe), at 55.3 (C-2, C-6), 73,3 (OCH2), of 114.3 (C-2', C-6'), for 125.8 (C-3', C-5'), 141,3 (C-4'), 164,1 (C-1') ppm- MC (70 eV, EI): m/z (%) = 250 (79) [M]+, 249 (100) [M-H]+. - C13H18N2O3(250,29): calculated C 62,38, N of 7.25; found C 62,25, N 7,40. Example 25 4-((1-Methylpiperidin-4-yl)methoxy)phenylamine (25) A solution of l-methyl-4-(4-nitrophenoxy)piperidine (24) (1.0 g, 4.0 mmol), MeOH (20 ml)and concentrated HCl (1 ml) hydronaut over Pd/C (10%, 100 mg) at 58 psi H2for 1 hour at room temperature. Then the hard part is removed by filtration through celite, which is washed thoroughly with MeOH (200 ml), and the obtained filtrate was concentrated in vacuo. The residue is dissolved in water (50 ml) and the resulting solution is brought to pH 10 with saturated aqueous solution of NaHCO3and 2M NaOH. The resulting mixture was extracted with CH2C12(3×100 ml), dried (MgSO4) and the solvent is removed in vacuum. In the purification using column chromatography (CH2Cl2/MeOH, 10:1, 2% NEt3)get the connection 25 (0,72 g, 82%) as a solid reddish brown color. 1H NMR(300 MHz, CDCl3): δ=1,31 is 1.48 (m, 2H, 3'-Hax, 5'-Hax), 1,66-1,89 (m, 3H, 3'-Heq, 4'-H, 5'-Heq), a 1.96 (dt, J=11,9, and 2.3 Hz, 2H, 2'-Hax, 6'-Hax), 2,28 (c, 3H, NMe), 2,84-of 2.93 (m, 2H, 2'-Heq, 6'-Heq), 3,41 (Cuser., 2H, NH2), to 3.73 (d, J=6,4 Hz, 2H, OCH2), 6,60 is 6.67 (m, 2H, 3-H, 5-H), 6,70-6,76 (m, 2H, 2-H, 6-H) ppm -13C NMR (75.5 MHz, CDCl3): δ=29,1 (C-3', C-5'), 35,3 (C-4'), 46,4 (NMe), a 55.4 (C-2', C-6'), 73,2 (OCH2), 115,4 (C-3, C-5), 116,3 (C-2, C-6), 139,8 (C-1), 152,2 (C-4) ppm MC (70 eV, EI): m/z (%) = 220 (3) [M]+, 112 (100) [C7H14N]+. -C13H20N2O (220,31): calculated C 70,87, H 9.15; found C 70,50, N 8,94. Example 26 Ethyl ester of 5-((1-methylpiperidin-4-yl)methoxy)-1H-indole-2-carboxylic acid (26) To a stirred solution of 4-((1-methylpiperidin-4-yl)methoxy)phenylamine (25) (2.00 g, the remaining 9.08 mmol) in water (19 ml) and concentrated HCl (6 ml) is added dropwise a solution of NaNO2(689 mg, of 9.99 mmol) in water (2 ml) at 0ºC, and stirring is continued for 50 minutes at 0ºC (solution A). Ethyl 2-methylacetoacetate (1.39 ml, at 9.53 mmol) is added dropwise to a stirred suspension NaOAc (7,8 g) in EtOH (15 ml) at 0ºC, and stirring is continued for 30 minutes at this temperature; then add ice (9 g) (solution B). Solution A is added to solution B, moving the cannula at 0ºC, and the resulting mixture left to warm to room temperature. After 2.5 hours, the reaction mixture was alkalinized by slowly adding saturated the initial aqueous solution of Na 2CO3at 0ºC and extracted with CH2Cl2(3×100 ml). The combined organic layers washed with water (200 ml), dried (MgSO4) and the solvent is removed in vacuum. The residue is then dissolved in absolute EtOH (10 ml), treated with freshly prepared saturated solution of HCl in absolute EtOH (10 ml) and refluxed for 50 minutes After cooling to room temperature the solvent is removed under reduced pressure and the residue is divided between water (50 ml) and CH2Cl2(100 ml). The aqueous layer was alkalinized using a saturated aqueous solution of Na2CO3and extracted with CH2Cl2(3×100 ml). The combined organic layers washed with brine (200 ml), dried (MgSO4) and concentrated in vacuo. Purification via crystallization from iPr2O and column chromatography (CH2Cl2/MeOH, 30:1, 2% NEt3) of the residue obtained after evaporation of the mother liquor, results in compounds 26 (of 2.06 g, 72% total yield) as a solid yellow color 1H NMR (300 MHz, CDCl3): δ=1,36-and 1.54 (m, 5H, OCH2CH3, 3'-Hax, 5'-Hax), 1,73 e 2.06 (m, 5H, 2'-Hax, 3'-Heq, 4'-H, 5'-Heq, 6'-Hax), 2,30 (c, 3H, NMe), 2,87-of 2.97 (m, 2H, 2'-Heq, 6'-Heq), 3,83 (d, J=6.2 Hz, 2H, ArOCH2), and 4.40 (q, J=7.2 Hz, 2H, OCH2CH3), 6,98 (DD, J=8,9, and 2.4 Hz, 1H, 6-H), 7,05 (d, J=2.0 Hz, 1H, 3-H), and 7.1 (m with, 1H, 4-H), 7,30 (d, J=8,9 Hz, 1H, 7-H), 9,11 (Cuser., 1H, NH) ppm -13C NMR (50,3 MHz, CDCl3): δ=14.4V (OCH2CH3), 29,2 (C-3', C-5'), 35,3 (C-4'), 46,4 (NMe), is 55.5 (C-2', C-6'), 60,9 (OCH2CH3), 73,2 (ArOCH2), 103,4 (C-4), 108,1 (C-3), 112,7 (C-7), 117,2 (C-6), 127,8 (C-2, C-3a), 132,2 (C-7a), 154,1 (C-5), 162,0 (C=O) ppm MC (70 eV, EI): m/z (%) = 316 (6) [M]+, 112 (100) [C7H14N]+. - C18H24N2O3(316,39): calculated C 68,33, N of 7.65; found C 68,03, N 7,84. Example 27 Hydrochloride 5-((1-methylpiperidin-4-yl)methoxy)-lH-indole-2-carboxylic acid (27) A suspension of ester 26 (1,00 g, and 3.16 mmol) in MeOH (8 ml) is treated with NaOH solution (155 mg, 3.88 mmol) in water (4 ml) and refluxed for 3.5 hours. After cooling to room temperature the solution was adjusted to pH 6 using 2M HCl, and the solvent is removed under reduced pressure. The residue is dissolved in MeOH, 2M HCl is added dropwise and the resulting precipitate is collected by filtration, receiving the connection 27 (712 mg, 69%) as a solid brown color. The residue obtained after evaporation of the mother liquor, purified using column chromatography (CH2Cl2/MeOH, 6:1, 1% conc. HCl), receiving the second portion of the connection 27 (226 mg, 22%). 1H NMR (300 MHz, DMSO-d6): δ=1.56 to 1.77 in (m, 2H, 3'-Hax, 5'-Hax), 1,89-of 2.09 (m, 3H, 3'-Heq, 4'-H, 5'-Heq), 2,69 (c, 3H, NMe), 2,87 was 3.05 (m, 2H, 2'-Hax, 6'-Hax) 3,24-of 3.48 (m,2H, 2'-Heq, 6'-Heq), 3,79-to 3.92 (m, 2H, ArOCH2), of 6.90 (DD, J=9,0, 2.2 Hz, 1H, 6-H), 6,98 (d, J=1.7 Hz, 1H, 3-H), 7,12 (d, J=2.2 Hz, 1H, 4-H), 7,34 (d, J=9.0 Hz, 1H, 7-H), 10,84 (Cuser., 1H, NH+), 11,59 (c, 1H, NH), 12,74 (Cuser., 1H, CO2H) ppm-13C NMR (75.5 MHz, DMSO-d6): δ=25,8 (C-3', C-5'), is 32.8 (C-4'), 42,5 (NMe), of 52.8 (C-2', C-6'), 71,6 (OCH2), 103,4 (C-4), 106,8 (C-3), 113,3 (C-7), 116,0 (C-6), 127,1 (C-2), 128,7 (C-3a), to 132.6 (C-7a), 152,9 (C-5), are 162.5 (C=O) ppm MC (70 eV, EI): m/z (%) = 288 (14) [M-HCl]+, 112 (100) [C7H14N]+. - C16H21ClN2O3(324,80): calculated 288,1474 [M-HCl]+; found 288,1474. Example 28 (+)-{(1,10)-Anti-5-benzyloxy-3-[(5-((1-methylpiperidin-4-yl)methoxy)indol-2-yl)carbonyl]-1-(10-chloroethyl)-1,2-dihydro-3H-Benz[e]indol}((+)-3d) Connection (+)-1 (150 mg, 342 μmol) suspended in 4M HCl/EtOAc and stirred at room temperature for 3.5 hours. The reaction mixture was concentrated and dried in vacuum for 1 hour. The residue is dissolved in DMF (10 ml). The solution is cooled to 0ºC and added EDC·HCl (197 mg, 1,03 mmol, 3.0 equiv.) and the connection 27 (144 mg, 445 mmol, 1.3 equiv.). The reaction mixture was stirred at room temperature for 23 hours, diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3(50 ml) and extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. In the result of the processing by means of column chromatography (CH 2Cl2/MeOH = 10:1) receive (+)-3d in the form of a cream solid color (158 mg, 260 μmol, 76%). 1H-NMR (300 MHz, DMSO-d6): δ=1.32 to a rate of 1.51 (m, 2H, 3"'-Hax, 5"'-Hax), and 1.63 (d, J=6,7 Hz, 3H, 11-CH3), 1,71-of 1.88 (m, 3H, 3"'-Heq, 4"'-H, 5"'-Heq), 2,07-2,22 (m, 2H, 2"'-Hax, 6"'-Hax), 2,30 (c, 3H, NCH3), 2,85-a 3.01 (m, 2H, 2"'-Heq, 6"'-Heq), 3,85 (d, J=5.7 Hz, 2H, 1"-H2), 4,16-4,27 (m, 1H, 1-H), 4,56-a 4.86 (m, 3H, 2-H2, 10-H), and 5.30 (mwith, 2H, OCH2Ph), 6,93 (d, J=8,9, and 2.3 Hz, 1H, 6'-H), 7,13-7,20 (m, 2H, 3'-H, 4'-H), 7,32-7,49, 7,51-7,62 (2×m, 8H, 7-H, 7'-H, 8-H, 5×Ph-H), of 7.96 (d, J=8,3 Hz, 1H, 9-H), 8,12 (Cuser., 1H, 4-H), 8,23 (d, J=8,4 Hz, 1H, 6-H), are 11.62 (Cuser., 1H, NH);13C-NMR (75 MHz, DMSO-d6): δ=23,3 (11-CH3), while 27.8 (C-3"', C-5"'), 34,2 (C-4"'), 45,1 (NCH3), to 45.9 (C-1), with 52.0 (C-2), 54,3 (C-2"', C-6"'), with 61.3 (C-10), 69,6 (OCH2Ph)72,3 (C-1'), to 98.4 (C-4), 103,4 (C-4'), or 105.3 (C-3'), 113,1 (C-7'), 115, 8mm (C-6'), 117,5 (C-5a), 122,6 (2 signals), 123,0, 123,7 (C-6, C-7, C-9, C-9b), 127,3, 127,4, 127,5, 127,8, 128,4 (C-3a', C-8,5×Bn-CH), of 129.6, 130,9, humidity 131.6 (C-2', C-7a', C-9a), 136,8 (Bn-C), to 142.1 (C-3a), 153,1 (C-5'), to 154.2 (C-5), 160,1 (C=O); MC (EI, 70 eV): m/z (%) = 607,0 (4) [M]+, 571,0 (23) [M-Cl-H]+; C37H38ClN3O3(608,17): calculated 608,2680 found 608,2675, [M+H]+(ESI-HRMS). Example 29 (+)-{(1,10)-Anti-1-(10-chloroethyl)-5-hydroxy-3-[(5-((1-methylpiperidin-4-yl)methoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic} ((+)-4d) The compound (+)-3d (90 mg, 148 μmol) dissolved in a mixture of 4M HCl/ethyl acetate (15 ml) and stirred for 2 is aces at room temperature. The solution concentrate. The residue is dried in vacuum for 1 hour and then suspended in THF (9 ml). Then added at room temperature with 10% palladium on activated carbon (32 mg) and monitorial (25% aqueous solution of 0.32 ml). The reaction mixture is stirred for 75 minutes at 40ºC and filtered through celite, which is washed thoroughly with methanol (150 ml). The obtained filtrate was concentrated in vacuo and the residue purified using column chromatography (CH2Cl2/MeOH = 5:l of 0.1% conc. HCl)to give (+)-4d in the form of a solid yellow (67 mg, 121 μmol, 82%). 1H-NMR (300 MHz, DMSO-d6): δ=1,60 (d, J=6.5 Hz, 3H, 11-CH3), 1,64-to 1.82 (m, 2H, 3"'-Hax, 5"'-Hax), 1,87-of 2.09 (m, 3H, 3"'-Heq, 4"'-H, 5"'-Heq), to 2.67 (c, 3H, NCH3), 2,85 totaling 3.04 (m, 2H, 2"'-Hax, 6"'-Hax), 3,26 is-3.45 (m, 2H, 2"'-Heq, 6"'-Heq), 3,85 (mwith, 2H, 1"-H2), 4,14 (mwith, 1H, 1-H), 4,47-4,79 (m, 3H, 2-H2, 10-H)6,91 (mwith, 1H, 6'-H), 7,11, 7,16 (2×suser., 2H, 3'-H, 4'-H), 7,28-7,44 (m, 2H, 7-H, 7'-H), of 7.48 (mwith, 1H, 8-H), 7,86 (d, J=8,3 Hz, 1H, 9-H), of 7.96 (Cuser., 1H, 4-H), to 8.12 (d, J=8,3 Hz, 1H, 6-H), 10,43 (c, 1H, OH), 10,97 (Cuser., 1H, NH+), 11,59 (c, 1H, NH);13C-NMR (75 MHz, DMSO-de): δ=23,4 (11-CH3), and 25.8 (C-3"', C-5"'), is 32.8 (C-4"'), 42,5 (NCH3), with 45.8 (C-1), to 52.1 (C-2), of 52.8 (C-2"', C-6"'), is 61.5 (C-10), of 71.7 (C-1'), to 100.4 (C-4), 103,6, of 105.2 (C-3', C-4'), 113,1 (C-7'), 115,6, 115, 8mm (C-6', C-5a), 122,2, 122,8, 122,9, 123,1 (C-6, C-7, C-9, C-9b), 126,9 (C-8), of 127.5 (C-3a'), 129,8, RB 131.1, 131,7 (C-2', C-7a', C-a), to 142.1 (C-3a), 152,9 (C-5'), other 153.9 (C-5), 159,8 (C=O); MC (ESI): m/z (%) = 518,0 (100) [M-Cl]+; C30H33Cl2N3O3(554,51): calculated 518,2210 found 518,2205, [M-Cl]+(ESI-HRMS). Example 30 [(+)-(1,10)-Anti-1-(10-chloroethyl)-3-[(5-((1-methylpiperidin-4-yl)methoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-2,3,4,6-Tetra-O-acetyl-β-D galactopyranoside ((+)-7d) A solution of (+)-5 (134 mg, 385 μmol) in dry CH2Cl2(17 ml) and molecular sieves 4Ǻ (0.8 g) is stirred for 30 minutes at room temperature. After adding O-(2,3,4,6-Tetra-O-acetyl-α-D-galactopyranosyl)trichloroacetimidate (6) (196 mg, 398 mmol, of 1.03 equiv.), the resulting mixture was cooled down to-10ºC and slowly add a solution BF3·OEt2(25 µl, 197 mmol, 0.5 equiv.) in dry CH2C12(1.9 ml). After 4 hours at-10ºC add additional portion BF3·OEt2(0,146 ml, 1.15 mmol, 3.0 equiv.) in dry CH2Cl2(1.8 ml)and the resulting mixture is heated to room temperature. After 5 hours the solution is separated from the molecular sieves, molecular sieves washed thoroughly with CH2Cl2(2×10 ml)and the combined organic portion was concentrated in vacuo. The residue is dried in vacuum for 1 hour and then dissolved in dry DMF (18 ml). The solution is cooled to 0ºC, and added EDC·HCl (222 mg, of 1.16 mmol, 3.0 equiv.) and the connection 27 (188 mg, 0,579 mmol, 1.5 equiv.). After 22 hours at room temperature the e reaction mixture was diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaHCO3(50 ml) and then extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (gradient: CH2Cl2/MeOH = 10:1 // CH2Cl2/MeOH = 5:1) receive (+)-7d (65 mg, 77 μmol, 20%) and its N-oxide (110 mg, 127 μmol, 33%), both in the form of a solid pale brown color. To a solution of N-oxide (40 mg, 46 μmol) in dry ethanol (6 ml), add PtO2·H2O (6 mg, 21 μmol, 0.5 equiv.). Then after the reaction mixture was bubbled hydrogen for 16 hours. The reaction mixture was filtered through celite, and the celite is washed thoroughly with methanol (50 ml) and ethanol (100 ml). The obtained filtrate was concentrated in vacuo, and the residue purified using column chromatography (CH2Cl2/MeOH = 10:1)to give (+)-7d in a solid yellow color (24 mg, 28 μmol, 61%). C44H50ClN3O12(848,33): calculated 848,3161 found 848,3156, [M+H]+(ESI-HRMS). Example 31 [(+)-(1,10)-Anti-1-(10-chloroethyl)-3-[(5-((1-methylpiperidin-4-yl)methoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-β-D galactopyranoside ((+)-8d) To a solution of compound 7d (74 mg, 87 μmol) in dry MeOH (6 ml) was added NaOMe in methanol (32 μl, approximately a 5.4 M solution, 173 μmol, 2,0 e is VIV.). The reaction mixture was stirred at room temperature for 30 minutes and diluted with water (2 ml) and methanol (2 ml). Ion-exchange resin (Amberlite-IR 120) add up until the pH of the solution does not take a neutral value. The resin is removed and washed with methanol (10 ml). The combined organic portion was concentrated in vacuo, and the residue purified using column chromatography (gradient: CH2Cl2/MeOH = 5:1 // CH2C12/MeOH = 7:1, 0.5% of the NEt3// CH2Cl2/MeOH = 5:1, 0.5% of the NEt3). The obtained solid product was washed with pentane (5×15 ml)to give compound 8d in the form of a solid, pale yellow (53 mg, 78 μmol, 90%). 1H-NMR (600 MHz, DMSO-d6, 100ºC): δ=of 1.40-1.50 (m, 2H, 3"'-Hax, 5"'-Hax), of 1.65 (d, J=6,7 Hz, 3H, 11-CH3), 1,77 is 1.86 (m, 3H, 3"'-Heq, 4"'-H, 5"'-Heq), and 2.14 (mwith, 2H, 2"'-Hax, 6"'-Hax), to 2.29 (c, 3H, NCH3), 2,87-of 2.93 (m, 2H, 2"'-Heq, 6"'-Heq)and 3.15 (Cuser.H2O, 4×OH), 3,47-3,51, 3,52-3,56, 3,60-3,64, 3,68-3,73 (4×m, 4H, 3""-H, 5""-H, 6""-H2), 3,81-to 3.92 (m, 4H, 1"-H2, 2""-H, 4""-H), 4,22 (mwith, 1H, 1-H), 4.63 to-to 4.73 (m, 2H, 2-H2), to 4.81 (mwith, 1H, 10-H), to 4.92 (d, J=7,6 Hz, 1H, 1""-H), 6,93 (mwith, 1H, 6'-H), 7,09, 7,18 (2×suser., 2H, 3'-H, 4'-H), 7,40 was 7.45 (m, 2H, 7-H, 7'-H), 7,56 (mwith, 1H, 8-H), to 7.93 (d, J=8,4 Hz, 1H, 9-H), 8,17 (Cuser., 1H, 4-H), of 8.37 (d, J=8.6 Hz, 1H, 6-H), 11,31 (c, 1H, NH);13C-NMR (150 MHz, DMSO-d6): δ=23,4 (11-CH3), 28,1 (C-3 is', C-5"'), 34,5 (C-4"'), 45,5 (NCH3), 46,0 (C-1), TO 52.1 (C-2), 54,5 (C-2"', C-6"'), TO 59.6 (C-6""), WITH 61.3 (C-10), OF 67.5 (C-4""), 70,6 (C-2'"), FROM 72.5 (C-1'), 73,3 (C-3""), AND 75.2 (C-5""), 101,9 (C-4), TO 102.3 (C-1""), 103,4 (C-4'), 105,4 (C-3'), of 113.2 (C-7'), 115, 8mm (C-6'), 118,9 (C-5a), 122,9, 123,0, 123,4, 123,7 (C-6, C-7, C-9, C-9b), RUB 127.3, of 127.5 (C-3a', C-8), 129,5, 130,9, 131,7 (C-2', C-7a'C-9a), 142,0 (C-3a), 153,2 (C-5'), of 153.6 (C-5), 160,2 (C=O); MS (ESI): m/z (%) = 702,3 (12) [M+Na]+, 680,3 (100) [M+H]+; C36H42C1N3O8(680,19): calculated 680,2739 found 680,2733, [M+H]+(ESI-HRMS). Example 32 Ethyl ester of 5-(2-(N,N-dimethylamino)acetylamino)-lH-indole-2-carboxylic acid (29) A solution of ethyl ester of 5-nitro-1H-indole-2-carboxylic acid (28) (750 mg, 3,20 mmol) in EtOAc (125 ml) hydronaut over Pd/C (10%, 300 mg) at 60 psi H2for 5 hours at room temperature. Then the hard part is removed by filtration through celite, which is washed thoroughly with CH2Cl2(400 ml) and MeOH (400 ml), and the obtained filtrate was concentrated in vacuo. The residue is dissolved in DMF (30 ml) and the solution cooled to 0ºC. Added EDC·HCl (1.84 g, 9,60 mmol) and the hydrochloride of N,N-dimethylglycine (670 mg, 4,80 mmol)and the reaction mixture left to warm to room temperature. After stirring for 21 hours at this temperature the solution was diluted with EtOAc (100 ml) and water (100 ml). The resulting mixture was adjusted to pH 9 with a saturated aqueous solution of NaHCO3and extracted with EtOAc (4×150 ml). The combined organic fractions washed the Ute water (4×200 ml) and brine (300 ml), dried (MgSO4) and the solvent is removed in vacuum. In the purification using column chromatography (CH2Cl2/MeOH, 10:1) to obtain compound 29 (626 mg, 68% of the total yield) as a solid pale brown color. 1H NMR (200 MHz, CDCl3): δ=1,42 (t, J=7,1 Hz, 3H, OCH2CH3), 2.40 a (c, 6H, NMe2), 3,11 (c, 2H, 1'-H2), to 4.41 (q, J=7,1 Hz, 2H, OCH2CH3), 7,18 (d, J=1.8 Hz, 1H, 3-H), was 7.36 (d, J=8,8 Hz, 1H, 7-H), the 7.43 (DD, J=8,8, 1.8 Hz, 1H, 6-H), 8,04 (Cuser., 1H, 4-H), 9,04 (Cuser., 1H, NH), 9,13 (Cuser., 1H, NH) ppm;13C NMR (50,3 MHz, CDCl3): δ=14,3 (OCH2CH3), 46,0 (NMe2), 61,0 (OCH2CH3), at 63.6 (C-1'), to 108.5 (C-3), to 112.2, of 112.8 (C-4, C-7), 119,2 (C-6), 127,5, 128,2 (C-2, C-3a), RB 131.1 (C-5), 134,1 (C-7a), 162,0 (NHCO), 168,6 (CO2Et) ppm; MC (70 eV, EI): m/z (%) = 289 (25) [M]+, 58 (100) [CH2NMe2]+; C15H19N3O3(289,33): calculated 289,1426 found 289,1426. Example 33 Hydrochloride 5-(2-(N,N-dimethylamino)acetylamino)-1H-indole-2-carboxylic acid (30) A mixture of ester 29 (0,200 g, 0,691 mmol), THF (6 ml), MeOH (2 ml)and water (2 ml) is treated with LiOH·H2O (0.035 g, 0,830 mmol) and stirred at 60ºC for 4 hours. After that, the solvent is removed in vacuum and the residue is dissolved in water. The resulting solution was acidified with 2M HCl, then water is evaporated under reduced pressure and the residue is placed in a mixture of acetone/EtOH (1:1). The remaining precipitate (LiCl) allocate filtration and the resulting filtrate concentrated under reduced pressure. In the purification of the crude product using column chromatography (CH2Cl2/MeOH, 5:1, 0.5% conc. HCl) are acid 30 (0,205 g, quantitative) as a solid pale yellow color . 1H NMR (300 MHz, DMSO-d6): δ=2,89 (c, 6H, NMe2), 4,16 (c, 2H, 1'-H2), 7,07 (d, J=2.1 Hz, 1H, 3-H), 7,38-7,46 (m, 2H, 6-H, 7-H), 8,01 (Cuser., 1H, 4-H), 10,11 (Cuser., 1H, NH"'), 10,80 (Cuser., 1H, NH), 11,74 (Cuser., 1H, NH), 12,90 (Cuser., 1H, CO2H) ppm;13C NMR (50,3 MHz, DMSO-d6): δ=43,1 (NMe2), of 57.8 (C-1'), 107,3 (C-3), 112,1, 112,6 (C-4, C-7), 118,4 (C-6), 126,6, to 129.2 (C-2, C-3a), 130,9 (C-5), RUR 134.4 (C-7a), 162,4, 162,5 (2×C=O) ppm MC (70 eV, EI): m/z (%) = 261 (7) [M-HCl]+, 58 (100) [C3H8N]+; C13H16ClN3C3(297,74): calculated 261,1113 [M-HCl]+found 261,1113. Example 34 (+)-{(1,10)-anti-5-benzyloxy-3-[(5-(2-(N,N-dimethylamino)acetylamino)indol-2-yl)carbonyl]-1-(10-chloroethyl)-1,2-dihydro-3H-Benz[e]indol} ((+)-3e) Connection (+)-1 (150 mg, 342 μmol) suspended in 4M HCl/EtOAc (14 ml) and stirred at room temperature for 3.5 hours. The reaction mixture was concentrated and dried in vacuum for 1 hour. The residue is dissolved in DMF (10 ml). The solution is cooled to 0ºC and added EDC·HCl (197 mg, 1,03 mmol, 3.0 equiv.) and the connection 30 (133 mg, 445 mmol, 1.3 equiv.). The reaction mixture was stirred at room temperature for 24 hours, diluted with ethyl acetate (50 ml), water (50 ml) and Ishenim aqueous solution of NaHCO 3(50 ml), and extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2Cl2/MeOH = 30:1) receive (+)-3e foam pale green (155 mg, 267 μmol, 78%). 1H-NMR (300 MHz, CDCl3): δ=1,63 (d, J=6.8 Hz, 3H, 11-CH3), 2,39 (c, 6H, NMe2), 3,11 (c, 2H, 1"-H2), 3,88-was 4.02 (m, 1H, 1-H), 4,47-4,63 (m, 2H, 2-Ha, 10-H), 4,84 (DD, J=10,8, 1.5 Hz, 1H, 2-Hb), 5,24 (mwith, 2H, OCH3Ph), 7,06 (d, J=1.4 Hz, 1H, 3'-H), 7,20 (DD, J=8,8, 1.9 Hz, 1H, 6'-H), 7,28-7,56 (m, 8H, 7-H, 7'-H, 8-H, 5×Ph-H), to 7.68 (d, J=8,2 Hz, 1H, 9-H), 8,13-of 8.28 (m, 2H, 4-H, 4'-H), 8,35 (d, J=8,3 Hz, 1H, 6-H), 9,11 (c, 1H, NH), there is a 10.03 (c, 1H, NH);13C-NMR (75 MHz, CDCl3): δ=23,9 (11-CH3), 46,0 (NMe2), with 47.4 (C-l), 53,5 (C-2), was 59.9 (C-10), at 63.6 (C-1'), 70,3 (OCH2Ph), was 98.2 (C-4), of 106.4 (C-3'), 12,1 (C-7'), 112,6 (C-4'), 117,3 (C-5a), of 118.8 (C-6'), 122,5 (C-9), 123,7, 123,8, 123,9 (C-6, C-7, C-9b), 127,4, 127,6, 127,9, 128,0, 128,5 (C-3a'), C-8, 5×Bn-CH), 129, 9mm (C-5'), 131,1, 131,2, 133,2 (C-2', C-7a', C-9a), 136,7 (Bn-C), 142,3 (C-3a), 155,5 (C-5), of 160.4 (C=O), 168,6 (C=O); MC(EI, 70 eV): m/z (%) = 580,0 (10) [M]+, 544,0 (7) [M-Cl-N]+; C34H33ClN4O3(581,10): calculated 580,2319 found 581,2314, [M+H]+(ESI-HRMS). Example 35 (+)-{(1,10)-Anti-1-(10-chloroethyl)-5-hydroxy-3-[(5-(2-(2-(N,N-dimethylamino)acetylamino)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic} ((+)-4e) The compound (+)-3e (80 mg, 138 μmol) dissolved in a mixture of 4M HCl/ethyl acetate 10 ml) and stirred for 2 hours at room temperature. The solution concentrate. The residue is dried in vacuum for 1 hour and then suspended in THF (8 ml). Then add 10% palladium on charcoal (30 mg) and monitorial (25% aqueous solution of 0.30 ml) at room temperature. The reaction mixture is stirred for 40 minutes at 40ºC and filtered through celite, which is washed thoroughly with methanol (150 ml). The obtained filtrate was concentrated in vacuo and the residue purified using column chromatography (MeOH, 0.1% conc. HCl). The resulting product is then dissolved in a small amount of CH2Cl2/MeOH, 0.1% conc. HCl. The solution is filtered and the resulting filtrate is concentrated and receiving (+)-4e in a solid green color (78 mg, 152 μmol, 86%). 1H-NMR (300 MHz, DMSO-d6): δ=1,63 (d, J=6.3 Hz, 3H, 11-CH3), 2,90 (c, 6H, NMe2), 4,11-of 4.25 (m, 3H, 1-H, 1"-H2), 4,53 with 4.64 (m, 1H, 2-Ha)and 4.65-to 4.81 (m, 2H, 2-Hb, 10-H), 7,21-of 7.55 (m, 5H, 3'-H, 6'-H, 7-H, 7'-H, 8-H), 7,88 (d, J=8,3 Hz, 1H, 9-H), 7,99 (Cuser., 1H, 4-H), 8.07-a 8,19 (m, 2H, 4'-H, 6-H), 10.30 a.m. (user., 1H, OH), 10,43, 11,03, 11,73 (3×c, 3H, 2×NH, NH+);13C-NMR (75 MHz, DMSO-d6): δ=23,3 (11-CH3), 43,1 (NMe2), with 45.8 (C-1), to 52.1 (C-2), 57,9 (C-1'), is 61.5 (C-10), 100,3 (C-4), is 105.6 (C-3'), 112,1, 112,3 (C-4', C-7'), 115,9 (C-5a), 118,0 (C-6'), 122,2, 122,9 (2 signal), 123,1 (C-6, C-7, C-9, C-9b), 126,9, 127,0 (C-3a', C-8), 129,7, 130,8, 131,4, 133,3 (C-2', C-5', C-7a', C-9a), to 142.1 (C-3a), 153,8 (C-5), 159,7 (C=O), 162,4 (C=O); MC (ESI): m/z (%) = 981,0 (11) [2M+H]+, 491,1 (100) [M-Cl]+; C27H28Cl N4O3(527,44): calculated 491,1850 found 491,1844, [M-Cl]+(ESI-HRMS). Example 36 [(+)-(1S,10R)-1-(10-Chloroethyl)-3-[(5-(2-(N,N-dimethylamino)acetylamino)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-2,3,4,6-Tetra-O-acetyl-β-D galactopyranoside ((+)-7e) A solution of (+)-5 (137 mg, 394 μmol) in dry CH2Cl2(17 ml) and molecular sieves 4Ǻ (0.8 g) is stirred for 30 minutes at room temperature. After adding O-(2,3,4,6-Tetra-O-acetyl-α-D-galactopyranosyl)trichloroacetimidate (6) (200 mg, 406 mmol, of 1.03 equiv.), the resulting mixture was cooled down to-10ºC and slowly add a solution BF3·OEt2(25 µl, 197 mmol, 0.5 equiv.) in dry CH2Cl2(1.9 ml). After 4 hours at-10ºC add additional portion BF3·OEt2(150 μl, 1.18 mmol, 3.0 equiv.) in dry CH2Cl2(1.8 ml)and the resulting mixture is heated to room temperature. After 5 hours the solution is separated from the molecular sieves, molecular sieves washed thoroughly with CH2Cl2(2×10 ml) and the combined organic portion was concentrated in vacuo. The residue is dried in vacuum for 1 hour and then dissolved in dry DMF (18 ml). The solution is cooled to 0ºC, and added EDC·HCl (227 mg, 1.18 mmol, 3.0 equiv.) and the connection 30 (176 mg, 591 mmol, 1.5 equiv.). After 21 hours at room temperature, the reaction mixture was diluted with ethyl acetate (50 ml), water (50 ml) and saturated aqueous NaCO 3(50 ml)and then extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2Cl2/MeOH = 25:1) receive (+)-7e in the form of a solid pale brown (155 mg, 189 μmol, 48%). C41H45ClN4O12(821,27): calculated 821,2801 found 821,2795. [M+H]+(ESI-HRMS). Example 37 [(+)-(1S,10R)-1-(10-Chloroethyl)-3-[(5-(2-(N,N-dimethylamino)acetylamino)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-β-D galactopyranoside ((+)-8) To a solution of (+)-7e (142 mg, 173 μmol) in dry MeOH (6 ml) was added NaOMe in methanol (64 μl to about 5,4M solution, 346 μmol, 2.0 equiv.). The reaction mixture was stirred at room temperature for 30 minutes and diluted with methanol (2 ml) and water (2 ml). Ion-exchange resin (Amberlite-IR 120) add up until the pH of the solution does not take a neutral value. The resin is separated from the solution and washed with methanol (10 ml). The combined organic fraction was concentrated in vacuo and the residue purified using column chromatography (gradient: CH2Cl2/MeOH = 7:1 // CH2Cl2/MeOH = 3:1). The crude product was washed with pentane (5×15 ml), giving (+)-8e in the form of a solid yellow (93 mg, 142 μmol, 82%). 1H-NMR (600 MHz, DMSO-d6): δ=1,65 (d, J=67 Hz, 3H, 11-CH3), 2,31 (c, 6H, NMe2), is 3.08 (c, 2H, 1"-H2), 3,45-to 3.52 (m, 1H, 3"'-H), 3,53-3,61 (m, 2H, 5"'-H, 6"'-Ha), 3,64-3,71 (m, 1H, 6"'-Hb), of 3.77-of 3.85 (m, 2H, 2"'-H, 4"'-H), 4,25 (mwith, 1H, 1-H), 4,55-of 4.66 (m, 3H, 2-Ha, 2×OH), 4,72-,of 4.90 (m, 3H, 2-Hb, 10-H, OH), is 4.93 (mwith, 1H, 1"'-H), 5,31 (Cuser., 1H, OH), 7,25 (Cuser., 1H, 3'-H), 7,37-7,46 (m, 3H, 6'-H, 7'-H, 7-H), EUR 7.57 (mwith, 1H, 8-H), of 7.96 (d, J=8,4 Hz, 1H, 9-H), 8,13 (Cuser., 1H, 4'-H), 8,24 (Cuser., 1H, 4-H), at 8.36 (d, J=8.6 Hz, 1H, 6-H), 9,60 (c, 1H, NH), 11,70 (c, 1H, NH);13C-NMR (150 MHz, DMSO-d6): δ=23,4 (11-CH3), 45,3 (NMe2), 46,0 (C-1), TO 52.1 (C-2), TO 59.6 (C-6"'), TO 61.4 (C-10), 63,2 (C-1'), TO 67.5 (C-4"'), 70,6 (C-2"'), 73,3 (C-3"'), AND 75.2 (C-5"'), 101,9 (C-4), TO 102.3 (C-1"'), OR 105.8 (C-3'), 112,1 (C-4'), to 112.2 (C-7'), 118,6 (C-6'), 119,0 (C-5a), 122,9, 123,1, 123,4, 123,7 (C-6, C-7, C-9, C-9b), 127,1 (C-3a'), RUB 127.3 (C-8), 129,5, 131,1, 131,5, 133,3 (C-2', C-5', C-7a', C-9a), 142,0 (C-3a), of 153.6 (C-5), 160,1 (C=O), 168,2 (C=O); MC (ESI): m/z (%) = 653,2 (100) [M+H]+; C33H37ClN4O8(653,12): calculated 653,2378 found 653,2373, [M+H]+(ESI-HRMS). Example 38 5-Benzyloxy-3-[(5-(2-(N,N-dimethylamino)acetylamino)indol-2-yl)carbonyl]-1-(10-chloromethyl)-1,2-dihydro-3H-Benz[e]indol} (rac-32) The compound rac-31 (300 mg, 708 mmol) is suspended in a mixture of 4M HCl/EtOAc (30 ml) and stirred at room temperature for 3 hours. The reaction mixture was concentrated and dried in vacuum for 2 hours. The residue is dissolved in DMF (25 ml). The solution is cooled to 0ºC and added EDC·HCl (407 mg, 2,12 mmol, 3.0 equiv.) and compound 2 (302 mg, 1.06 a MMO is ü, 1.5 equiv.). The reaction mixture was stirred at room temperature for 2.5 hours, diluted with ethyl acetate (100 ml), water (100 ml) and saturated aqueous NaHCO3(100 ml) and extracted with ethyl acetate (3×200 ml). The combined organic layers washed with brine (4×200 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2Cl2/MeOH = 10:1) to obtain rac-32 in the form of a solid yellow-brown (229 mg, 413 mmol, 58%). 1H-NMR (300 MHz, CDCl3): δ=2,37 (c, 6H, NMe2), 2,78 (t, J=5.7 Hz, 2H, 2"-H2), 3,44 (t, J=10,9 Hz, 1H, 10-Ha), of 3.95 (DD, J=11,2, 2,9 Hz, 1H, 10-Hb), Android 4.04-to 4.15 (m, 3H, 1-H, 1"-H2), 4,58-of 4.67 (m, 1H, 2-Ha), 4,78 (DD, J=11.0 in, and 1.6 Hz, 1H, 2-Hb) a 5.25 (mwith, 2H, OCH2Ph), 6,97-7,03 (m, 2H, 3'-H, 6'-H), 7,11 (d, J=2.3 Hz, 1H, 4'-H), 7,28-7,43, 7,46-7,57 (2×m, 8H, 7-H, 7'-H, 8-H, 5×Ph-H), of 7.69 (d, J=8,3 Hz, 1H, 9-H), 8,19 (Cuser., 1H, 4-H), to 8.34 (d, J=8,3 Hz, 1H, 6-H), 9,75 (Cuser., 1H, NH);13C-NMR (50 MHz, CDCl3): δ=43,1 (C-1), 45,8 (NMe2), 46,0 (C-10), 55,2 (C-2), 58,3 (C-2'), to 66.4 (C-1'), 70,3 (OCH2Ph)98,3 (C-4), also, 103.5 (C-4'), 106,1 (C-3'), 112,7 (C-7'*), 116,4 (C-5a), 117,3 (C-6'*), 122,1, 123,6, 123,7, 124,1 (C-6, C-7, C-9, C-9b), 127,6, 127,8, 128,0, 128,2, 128,5 (C-3a', C-8,5×Bn-CH), 129,7, 130,5, to 131.4 (C-2', C-7a', C-9a), 136,7 (Bn-C), to 142.1 (C-3a), 153,8 (C-5'), 155,8 (C-5), 160,7 (C=O); MC (EI,70 eV): m/z (%)= 553,3 (33) [M]+, 517,3 (7) [M-Cl]+; C33H32ClN3O3(554,08): calculated 553,2132 found 553,2132 (EI-HRMS). When the EP 39 1-(10-Chloromethyl)-5-hydroxy-3-[(5-(2-(2-(N,N-dimethylamino)acetylamino)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic} (rac-33) The compound rac-32 (100 mg, 180 μmol) dissolved in a mixture of 4M HCl/ethyl acetate (10 ml) and stirred for 2 hours at room temperature. The solution concentrate. The residue is dried in vacuum for 1 hour and then suspended in THF (15 ml). Then add 10% palladium on charcoal (38 mg) and monitorial (25% aqueous solution of 0.38 ml) at room temperature. The reaction mixture is stirred for 4 hours at 40ºC and filtered through celite, which is washed thoroughly with methanol (300 ml). The obtained filtrate was concentrated in vacuo, and the residue purified using column chromatography (CH2Cl2/MeOH = 5:1, with 0.1% conc. HCl)to give rac-33 in the form of a solid yellow (73 mg, 146 μmol, 81%). 1H-NMR (300 MHz, MeOH-d4): δ=2,98 (c, 6H, NMe2), 3,51-the 3.65 (m, 3H, 2"-H2, 10-Ha), 3,93 (DD, J=11,2, 3.0 Hz, 1H, 10-Hb), a 4.03-4,13 (m, 1H, 1-H), 4,32 (t, J=4,8 Hz, 2H, 1"-H2), to 4.52-of 4.66 (m, 2H, 2-H2), 6,99-7,06 (m, 2H, 3'-H, 6'-H), 7,24 (d, J=2.4 Hz, 1H, 4'-H), 7,33 (t, J=7,6 Hz, 1H, 7-H), the 7.43 (d, J=8,9 Hz, 1H, 7'-H), 7,49 (t, J=7,6 Hz, 1H, 8-H), 7,72 (d, J=8,4 Hz, 1H, 9-H), 7,81 (Cuser., 1H, 4-H), 8,18 (d, J=8,4 Hz, 1H, 6-H);13C-NMR (75 MHz, MeOH-d4): δ=43,5 (C-1)and 43.9 (NMe2), and 47.5 (C-10), with 56.8 (C-2), of 57.8 (C-2'), or 63.7 (C-1'), 101,4 (C-4), of 105.2 (C-4'), 107,2 (C-3'), 114,2 (C-7'), 117,1, 117,2 (C-5a, C-6), to 123.5 (C-9), 124,3, 124,5, 124,6 (C-6, C-7, C-9a), of 128.6(C-8), to 129.2, 131, 5mm, 132,5, 133,9 (C-2, C-3a', C-7a, C-9b), 143,1 (C-3a), 153,8 (C-5'), 155,8 (C-5), 162,6 (CO); MC (ESI): m/z (%) = 928,9 (35) [2M-2Cl]+, 464,2 (100) [M-Cl]+; C26H27Cl2N3O3(500,42): calculated 464,1741 found 464,1736, [M-Cl]+(ESI-HRMS). Example 40 [1-(10-Chloromethyl)-3-[(5-(2-(N,N-dimethylamino)acetylamino)indol-2-yl)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-2,3,4,6-Tetra-O-acetyl-β-D galactopyranoside (35/35') A solution of rac-34 (136 mg, 407 μmol) in dry CH2Cl2(18 ml) and molecular sieves 4Ǻ (0.8 g) is stirred for 30 minutes at room temperature. After adding O-(2,3,4,6-Tetra-O-acetyl-α-D-galactopyranosyl)trichloroacetimidate (6) (211 mg, 428 mmol, 1.05 equiv.), the resulting mixture was cooled down to-10ºC and slowly add a solution BF3·OEt2(26 μl, 205 mmol, 0.5 equiv.) in dry CH2Cl2(2.1 ml). After 4 hours at-10ºC add additional portion BF3·OEt2(155 μl, 1,22 mmol, 3.0 equiv.) in dry CH2Cl2(1.8 ml)and the resulting mixture is heated to room temperature. After 5 hours the solution is separated from the molecular sieves, molecular sieves washed thoroughly with CH2Cl2(2×10 ml) and the combined organic portion was concentrated in vacuo. The residue is dried in vacuum for 1 hour and then dissolved in dry DMF (19 ml). The solution is cooled to 0ºC, and added EDC.HCl (234 mg, 1,22 mmol, 3.0 equiv.) 2 (174 mg, 611 mmol, 1.5 equiv.). After 20 hours is at room temperature, the reaction mixture was diluted with ethyl acetate (25 ml), water (25 ml), and saturated aqueous NaHCO3(25 ml)and then extracted with ethyl acetate (4×50 ml). The combined organic layers washed with brine (4×100 ml), dried over MgSO4, filtered and concentrated in vacuo. As a result of processing using column chromatography (CH2Cl2/MeOH = 10:1) get a mixture of both diastereoisomers 35 and 35' in the form of a solid yellow color (173 mg, 218 μmol, 54%). (-)-35: C40H44ClN3O12(794,24): calculated 794,2692 found 794,2686, [M+H]+(ESI-HRMS). Example 41 [1-(10-Chloromethyl)-3-[(5-(2-(N,N-dimethylamino)acetylamino)carbonyl]-1,2-dihydro-3H-Benz[e]indol-5-yl]-β-D galactopyranoside (36/36') To a solution of 35/35' (170 mg, 214 μmol) in dry MeOH (9 ml) was added NaOMe in methanol (79 μl to about 5,4M solution, 428 μmol, 2.0 equiv.). The reaction mixture was stirred at room temperature for 2 hours and diluted with methanol (2 ml) and water (2 ml). Ion-exchange resin (Amberlite-IR 120) add up until the pH of the solution does not reach the neutral values. The resin is separated from the solution and washed with methanol (10 ml). The combined organic fraction was concentrated in vacuo and the residue purified using column chromatography (CH2Cl2/MeOH = 1:1)to give diastereoisomer 36/36' in the form of a solid pale yellow color is the one (94 mg, 150 μmol, 70%). 1H-NMR (600 MHz, DMSO-d6): δ=2,24 (c, 6H, NMe2), to 2.66 (t, J=5,9 Hz, 2H, 2"-H2), 3,35-3,86 (m, 6H, 2"'-H, 3"'-H, 4"'-H, 5"'-H, 6"'-H2), 3,89-3,95 (m, 1H, 10-Ha), was 4.02-4.09 to (m, 3H, 1"-H2, 10-Hb), 4,25-4,32 (m, 1H, 1-H), 4.53-in-4,63 (m, 3H, 2-H2, OH), 4,71 is equal to 4.97 (m, 3H, 1"'-H, 2×OH), 5,27 and 5.36 (m, 1H, OH), 6,91 (DD, J=8,9, and 2.3 Hz, 1H, 6'-H), 7,06-7,11 (m, 1H, 3'-H), 7,17-7,19 (m, 1H, 4'-H), 7,38 was 7.45 (m, 2H, 7-H, 7'-H), EUR 7.57 (mwith, 1H, 8-H), to $ 7.91 (d, J=8.6 Hz, 1H, 9-H), 7,98-of 8.28 (m, 1H, 4-H), 8,33, 8,35 (2×d, J=8.6 Hz, 1H, 6-H), are 11.62, 11,64 (2×suser., 1H, NH);13C-NMR (150 MHz, DMSO-d6): δ=41,0/41,2 (C-1), 45,5 (NMe2), and 47.5 (C-10), 54,9/55,0 (C-2), of 57.8 (C-2'), 59,6/59,7 (C-6"'), 66,3 (C-1"), 67,5/67,7, 70,4/70,5, 73,2 (2 signal), and 75.2 (C-2"', C-3"', C-4"', C-5"'), to 101.8 (C-4), 102,1/to 102.3 (C-1"'), 103,2/103,3 (C-4'), 105,2/or 105.3 (C-3'), 113,1/of 113.2 (C-7'), 115, 8mm/115,9 (C-6'), 117,9/to 118.0 (C-5a), 122,70 (C-9), 122,9/123,0 (C-9b), 123,3/123,4 (C-6), 123,7/123,8 (C-7), of 127.5 (C-3a', C-8), 129,5 (2 signals) (C-9a), 130,9 (2 signals), 131, 6mm/131,7 (C-2', C-7a'), to 142.1 (C-3a), 153,0 (2 signals) (C-5'), 153,7 (2 signals) (C-5), 160,3 (2 signals) (C=O); MC (ESI): m/z (%)= 1252,0 (11) [2M+2H]+, 626,4 (100) [M+H]+; C32H36ClN3O8(626,10): calculated 626,2269 found 626,2264, [M+H]+(ESI-HRMS). Example 42 (1S,10R)-1-(10-Chloroethyl)-5-hydroxy-3-[(5-(2-(N-methylamino)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic (4f) Compound 3f (12 mg, and 18.3 mmol) dissolved in THF (2 ml). Then add 10% palladium on activated carbon (4 mg) and monitorial (25% aqueous solution, 40 μl) at room temperature. The reaction is mesh stirred for 2 hours at 45ºC and filtered through celite, which is thoroughly washed with methanol (3×5 ml). The obtained filtrate was concentrated in vacuo. The residue is dissolved in 4n HCl in ethyl acetate (10 ml). The reaction mixture was stirred at room temperature for 1 hour, after which the reaction mixture is concentrated. The ethyl acetate added to the residue and the suspension concentrate. The residue is purified using column chromatography (CH2Cl2/MeOH = 5:1, with 0.1% conc. HCl)to give compound 4f (5.6 mg, and 11.2 mmol, 61%) as a solid pale green color. 1H-NMR (400 MHz, CDCl3/CD3OD): δ=1,65 (d, 3H, 10-CH3), 2,81 (c, 3H, N-CH3), of 3.45 (t, 2H, 2"-H), a 4.03 (m, 1H, 1-H), to 4.38 (t, 2H, 1"-H), 4,59 with 4.65 (m, 2H, 10-H, 2-H), 4,79 (d, 1H, 2-H), 7,07 (d, 1H, 6'-H), 7,09 (c, 1H, 3'-H), 7,24 (c, 1H, 4'-H), 7,38 (m, 1H, 7-H), 7,47 (d, 1H, 7'-H), 7,52 (t, 1H, 8-H), to 7.75 (d, 1H, 9-H), 7,79 (users, 1H, 4-H), 8,23 (d, 1H, 6-H). Example 43 (1S,10R)-1-(10-Chloroethyl)-5-hydroxy-3-[(5-(2-(N-methyl-N-(carboxymethyl)amino)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic (4g) The 4g connection is obtained from the 3g connection in accordance with the procedure described for the conversion of compound 3f to 4f. A 4g connection get in a solid pale green color with a 50% output. 1H-NMR (400 MHz, DMSO-d6): δ=1.61 of (m, 3H, 10-CH3), 2,93 (c, 3H, NCH3), 3,14-3,18 (m, 2H, OCH2CH2N), 3,61 (c, 2H, CH2CO2H), 3,82 (m, 1H, 1-H), 4,17-4,74 (m, 5H, OCH2CH2N, 2×2-H, 10-H), 6,93-to 8.12 (m, 9H, 6'-H, 3'-H, 4'-H, 7'H, 7-H, 8-H, 9-H, 4-H, 6-H), 10,42 (c, 1H, NH), 11,67 (c, 1H, OH); MC (ESI): m/z = 522 [M+H]+, 1043 [2M+H]+. Example 44 (1S,10R)-1-(10-Chloroethyl)-5-hydroxy-3-[(5-(2-(N-methyl-N-(2-methoxy-2-oxoethyl)amino)ethoxy)indol-2-yl)carbonyl]-1,2-dihydro-3H-benzo[e]indolepropionic (4h) Compound 4h produced from compound 3h in accordance with the procedure described for the conversion of compound 3a in 4a except that the lower stage of acidification with HCl in ethyl acetate. Compound 4h receive in the form of a solid pale green color with 87% yield. 1H-NMR (300 MHz, CDCl3/CD3OD): δ=1.57 in (d, 3H, 10-CH3), to 2.67 (c, 3H, NCH3), is 3.21 (t, 2H, OCH2CH2N)3,65 (c, 2H, CH2CO2Me), 3,70 (c, 3H, OCH3), 3,82 (m, 1H, 1-H)to 4.15 (t, 2H, OCH2CH2N)to 4.38 (m, 1H, 2-H), 4,48 (m, 1H, 10-H), 4,70 (m, 1H, 2-H), 6,85-of 6.90 (m, 2H, 6'-H, 3'-H), 7,06 (d, 1H, 4'-H), 7,27-7,38 (m, 2H, 7'-H, 7-H), 7,46 (m, 1H, 8-H), 7,60 (d, 1H, 9-H), 7,89 (users, 1H, 4-H), of 8.28 (d, 1H, 6-H), of 10.21 (c, 1H, NH); MC (ESI): m/z = 536 [M+H]+, 1093 [2M+Na]+. Example 45 (1S)-1-Chloromethyl-5-hydroxy-3-[(5-(2-(2-(N,N-dimethylamino)ethoxy)indol-2-yl)carbonyl]-9-methyl-1,2-dihydro-3H-benzo[e]indolepropionic (45) The connection 45 is produced from compound 44 in accordance with the procedure described for the conversion of compounds 4a of the connection 1. The connection 45 get in a solid pale green color 4-stage with a yield of 20%. 1H-NMR (300 MHz, CD3OD): δ=2,78 (c, 3H, 9-CH3), 2,98 (c, 6, N(CH3)2)at 3.25 (t, 1H, 2-H), to 3.58 (t, 2H, 2"-H), 3,68 (d, 1H, 2-H), 4,25 (DD, 1H, 1-H)to 4.33 (t, 2H, 1"-H), 4,50 (t, 1H, 10-H), with 4.64 (d, 1H, 10-H), 7,00-7,08 and 7,18-7,31 (2×m, 5H, 7-H, 3'-H, 4'-H, 6'-H, 7'-H), the 7.43 (d, 1H, 8-H), 7,85 (c, 1H, 4-H), 8,11 (d, 1H, 6-H); MC (ESI): m/z = 478 [M+H]+. Example 46 Compound 46a The compound (+)-4a (36 mg, 71 mmol) suspended in acetonitrile (10 ml) and the resulting mixture is cooled to 0ºC. Add pair-nitrophenylphosphate (28.4 mg, 0,141 mmol) and DIPEA (58,3 μl, 0,353 mmol)and the reaction mixture warmed to room temperature. After 1 hour the reaction mixture is concentrated to dryness. The residue is washed with toluene (2×5 ml) and dried in vacuum. The residue is dissolved in acetonitrile (10 ml) and add N-Boc-N,N'-dimethylethylenediamine (133 mg, 0,706 mmol). After 1 hour the reaction mixture was concentrated in vacuo. The residue is dissolved in dichloromethane; the solution is washed with water, dried over Na2SO4filter and concentrate. The crude residue is dissolved in TFA. After 30 minutes the reaction mixture was concentrated in vacuo, to give crude compound 46a quantitatively in the form of not-quite-white solid. Compound 46b: Connection 46b is produced from compound (+)-4a in accordance with the method for obtaining compounds 46a, except that N-Boc-piperazine is used instead of N-Boc-N,N'-dimethylethylenediamine. Compound 46b receive quantitatively in the form of not-quite-white solid. Example 47 A common way to obtain 47a-47f of the 46a/46b A solution of crude compound 46a or 46b (71 mmol) in DMF (1 ml) cooled to 0ºC. Add activated steam-nitrophenylarsonic linker 57a, 57b, 58 (0,142 mmol) and DIPEA (29 μl, 0,177 mmol)and the reaction temperature is then raised to room temperature. After 1 hour the reaction mixture is concentrated to dryness. The residue is purified using column chromatography (CH2Cl2/MeOH = 5:1)to give the product as not quite white solid. Connection 47e and 47f get out of their Aloe-protected analogues, dissolving in dichloromethane and adding Pd(PPh3)4(0.1 equiv.) and morpholine (10 equiv.). After 1 hour the reaction mixture was concentrated, the residue is dissolved in dichloromethane, and the solution is acidified (pH 3) triperoxonane acid. The solution is concentrated and the residue purified using preparative HPLC (acetonitrile/water solution ammonitrate), and receiving the product in a solid white color. Compound 47a: 38% yield;1H-NMR (400 MHz, CDCl3/CD3OD): δ=0,94 (d, J=6.4 Hz, 6H, Hval), 1,26-135 (m, 2H, 2×Hcapr), 1,49-of 1.74 (m, 10H, 10-CH3, 7×Hcit/Hcapr), to 1.87 (m, 1H, Hcit), to 2.06 (m, 1H, Hval), and 2.26 (t, J=7,6 Hz, 2H, CH2C(O)), 2.49 USD (c, 6H, N(CH3)2), 2,90-up 3.22 (m, 10H, NCH2,cit, 2×NCH3, OCH2CH2N), 3.46 in-a 3.83 (m, 6H, NCH2CH2N, CH2,capr), 4.09 to (m, 1H, 1-H), 4,15-is 4.21 (m, 3H, OCH 2CH2N, α-Hval), 4,53-4,69 (m, 3H, 2a-H, 10-H, α-Hcit), a 4.86 (m, 1H, 2b-H), 5,06-5,14 (m, 2H, CH2OC(O)), 6,72 (c, 2H, CH=CH), 7,00-7,06 (m, 2H, 6'-H, 3'-H), 7,17 (m, 1H, 4'-H), 7.23 percent-to 7.61 and 7,79-to 7.93 (m, 9H, 7'-H, 7-H, 8-H, 6-H, 9-H, 4×HAr,PABA), by 8.22 (users, 1H, 4-H). Connection 47b: 72% yield;1H-NMR (400 MHz, CDCl3/CD3OD, TFA salt): δ=0,94 (d, J=6.8 Hz, 3H, CH3,val), of 0.95 (d, J=6.8 Hz, 3H, CH3,val), of 1.28 to 1.34 (m, 2H, 2×Hcapr), a 1.50-1.77 in (m, 10H, 10-CH3, 7×Hcit/Hcapr), 1,89 (m, 1H, Hcit), was 2.05 (m, 1H, Hvalin ), 2.25 (t, J=7,6 Hz, 2H, CH2C(O)), 2,98 (c, 6H, N(CH3)2), 3,09 is 3.25 (m, 2H, NCH2,cit), a 3.50 (t, J=7,6 Hz, 2H, CH2,capr), of 3.54 (m, 2H, OCH2CH2N), 3,57-3,71 (m, 6H, Hpiperazine), 3,78-3,91 (m, 2H, Hpiperazine), of 4.05 (m, 1H, 1-H), 4,16 (d, J=7,6 Hz, 1H, α-Hval), and 4.40 (m, 2H, OCH2CH2N), 4,54 with 4.65 (m, 3H, 2a-H, 10-H, α-Hcit), is 4.85 (DD, 1H, 2b-H), 5,13 (c, 2H, CH2OC(O)), 6,70 (c, 2H, CH=CH), 6,98 (DD, J=2.4 Hz, J=9,2 Hz, 1H, 6'-H), 7,03 (c, 1H, 3'-H), 7,17 (d, J=2.4 Hz, 1H, 4'-H), 7,34 (d, 2H, HAr,PABA), 7,41 (d, J=9,2 Hz, 1H, 7'-H), was 7.45 (t, J=7.2 Hz, 1H, 7-H), 7,52 (t, J=7.2 Hz, 1H, 8-H), to 7.61 (d, J=8,4 Hz, 2H, HAr,PABA), to 7.77 (d, J=8,4 Hz, 1H, 9-H), 7,87 (d, J=8.0 Hz, 1H, 6-H), 8,25 (users, 1H, 4-H). Connection 47c: 64% yield;1H-NMR (400 MHz, CDCl3/CD3OD): δ=0,94 (d, J=6,7 Hz, 3H, CH3,val), and 0.98 (d, J=6,7 Hz, 3H, CH3,val), 1,48 is 1.60 (m, 2H, Hcit), 1,65-of 1.78 (m, 3H, 10-CH3Hcit), 1,89 (m, 1H, Hcit), 2,10 (m, 1H, Hval), 2,65 (c, 6H, N(CH3)2), 2,95 is 3.40 (m, 12H, 2×H3CNC(O), 2×HcitCH2N3CH2NMe2 ), 3,50-4,10 (m, 10H, CH2OCH2, NCH2CH2N, α-Hval, 1-H), 4,18-4,30 (m, 4H, OCH2CH2N, CH2CH2OC(O)), 4,53-and 4.68 (m, 3H, 2a-H, 10-H, α-Hcit), 4,80 (m, 1H, 2b-H), of 5.05-by 5.18 (m, 2H, CH2OC(O)), 6,94? 7.04 baby mortality (m, 2H, 6'-H, 3'-H), 7,16 (users, 1H, 4'-H), 7,20-to 7.95 (m, 9H, 4×HAr,PABA, 7-H, 7-H, 8-H, 9-H, 6-H), 8,23 (users, 1H, 4-H). Connection 47d: 85% yield;1H-NMR (400 MHz, CDCl3/CD3OD, TFA salt): δ=0,95 (d, J=6.8 Hz, 3H, CH3,val), 0,99 (d, J=6.8 Hz, 3H, CH3,val), 1,50-to 1.67 (m, 2H, Hcit), was 1.69 (d, J=6.9 Hz, 3H, 10-CH3), is 1.73 (m, 1H, Hcit), with 1.92 (m, 1H, Hcit), a 2.12 (m, 1H, Hval), 2,94 (c, 6H, N(CH3)2), 3,10-of 3.28 (m, 2H, Hcit), 3,40 (t, J=5.0 Hz, 2H, CH2N3), 3,50 (ushort, 2H, OCH2CH2N), 3,56-3,74 (m, 10H, 6×HpiperazineCH2OCH2), 3,80-of 3.94 (m, 2H, Hpiperazine), to 4.01 (m, 1H, 1-H), 4,11 (m, 1H, a-Hval), 4,24 (m, 2H, CH2CH2OC(O)), 4,36 (t, J=4.9 Hz, 2H, OCH2CH2N), 4,54-4,72 (m, 3H, 2a-H, 10-H, α-Hcit), a 4.86 (d, 1H, 2b-H), 5,15 (c, 2H, CH2OC(O)), 7,03 (DD, J=2.4 Hz, J=8,9 Hz, 1H, 6'-H), 7,07 (d, J=0.7 Hz, 1H, 3'-H), 7,21 (d, J=2.2 Hz, 1H, 4'-H), was 7.36 (d, J=8.6 Hz, 2H, HAr,PABA), 7,46 (d, J=8,9 Hz, 1H, 7'-H), of 7.48 (t, 1H, 7-H), 7,58 (t, J=6,8 Hz, 1H, 8-H), a 7.62 (d, J=8.7 Hz, 2H, HAr,PABA), 7,83 (d, J=8,4 Hz, 1H, 9-H), 7,88 (d, J=8.7 Hz, 1H, 6-H), 8,23 (users, 1H, 4-H). Connection 47e: 63% yield; MC (ESI): m/z = 587 [M+H]2+, 1173 [M+H]+, 1195 [M+Na]+. Connection 47f: 84% yield;1H-NMR (300 MHz, CDCl3/CD3OD): δ=of 1.33 to 1.48 (m, 2H, Hlys), 1,60-to 1.79 (m, 6H, 3×Hlys, 10-CH3), to 1.87 (m, 1H, H lys), 2,53 (c, 6H, N(CH3)2), 2,83 was 3.05 (m, 5H, CH2NH2, OCH2CH2N, Hphe), of 3.13 (DD, J=6.0 Hz, J=14.1 Hz, 1H, Hphe), to 3.35 (m, 1H, CH2N3), 3,52 of 3.75 (m, 10H, 6×HpiperazineCH2OCH2in ), 3.75-3,98 (users, 2H, Hpiperazine), was 4.02 (userd, 1H, 1-H), 4.09 to to 4.23 (m, 4H, CH2CH2OC(O)OCH2CH2N)to 4.41 (t, 1H, α-Hphe), 4,47 (DD, 1H, α-Hlys), to 4.52-and 4.68 (m, 2H, 2a-H, 10-H), 4,82 (d, J=10.5 Hz, 1H, 2b-H), 5,15 (c, 2H, CH2OC(O)), of 6.99 (DD, 1H, 6'-H), 7,01 (users, 1H, 3'-H), to 7.15 (m, 1H, 4'-H), 7,20 (users, 1H, Hphenyl), was 7.36 (d, 2H, HAr,PABA), 7,44 (t, J=7.7 Hz, 1H, 7-H), 7,52 (t, J=7.5 Hz, 1H, 8-H), 7,60 (d, J=8.5 Hz, 2H, HAr,PABA), of 7.75 (d, J=8.0 Hz, 1H, 9-H), 7,86 (d, J=7.9 Hz, 1H, 6-H), 8,28 (users, 1H, 4-H). Example 48 General method of preparing compounds 48c-48d from 47c-47d: a Solution of compound 47c or 47d (25 mmol), sodium ascorbate (3.0 mg, 15 μmol), compound 40 (10.1 mg, 38 µmol), and CuSO4(1.9 mg, 7.5 μmol) in a mixture of THF/water was stirred at room temperature. Over the course of the reaction is monitored by HPLC. After completion of the reaction, the reaction mixture was diluted with a mixture of THF/water, and the product purified using preparative HPLC (acetonitrile/water solution ammonitrate), and receiving the product in the form of not quite white solid after freeze-drying. Connection 48c: 31% yield;1H-NMR (400 MHz, CDCl3/CD3OD, TFA salt): δ=0,98 (m, 6H, Hval), 1,26-135 (m, 2H, 2×Hcapr), 1,49-to 1.61 (m, 2H, Hcit), of 1.62 and 1.75 (m, 4H, 10-CH3 Hcit), 1,89 (m, 1H, Hcit)by 2.13 (m, 1H, Hval), 2,93-of 3.25 (m, 13H, N(CH3)2, 2×NCH3CH2,cit), 3,50 -4,88 (m, 30H, 2×OCH2CH2O, 3×OCH2CH2N, NCH2CH2N, HNCH2α-Hvalα-Hcit, 2a-H, 2b-H, 1-H, 10-H), 5,02-5,20 (m, 2H, C6H4CH2OC(O)), 6,74 (c, 2H, HC=CH), 6,94 is 8.25 (m, 14H, 6'-H, 3'-H, 4'-H, 7-H, 8-H, 7'-H, 4×HAr,PABA, 9-H, 6-H, Htriazole, 4-H). Connection 48d: 48% yield;1H-NMR (300 MHz, CDCl3/CD3OD): δ=0,92 (d, J=6.9 Hz, 3H, Hval), of 0.95 (d, J=6.8 Hz, 3H, Hval), 1,46 is 1.58 (m, 2H, Hcit), of 1.64 (d, J=6.8 Hz, 3H, 10-CH3), to 1.70 (m, 1H, Hcit), to 1.87 (m, 1H, Hcit), is 2.09 (m, 1H, Hval), 2,50 (users, 6H, N(CH3)2), 2,97 (users, 2H, CH2NMe2), to 3.09 (m, 1H, Hcit), 3,18 (m, 1H, Hcit), 3,52-3,72 and of 3.78-3.90 and 3.95 to and 4.04-4,24 and 4.35 and 4,48-4,70 (m, 33H, 2×OCH2CH2O, 2×NCH2CH2O, OCH2CH2NMe2, 8×Hpiperazineα-Hvalα-Hcit, 2a-H, 1-H, 10-H, HNCH2), 4,82 (d, 1H, 2b-H), 5,11 (c, 2H, C6H4CH2OC(O)), 6,69 (c, 2H, HC=CH), of 6.96? 7.04 baby mortality (m, 2H, 6'-H, 3'-H), 7,15 (users, 1H, 4'-H), 7,32 (d, J=8.5 Hz, 2H, HAr,PABA), 7,37 was 7.45 (m, 2H, 7'-H, 7-H), 7,53 (t, 1H, 8-H), EUR 7.57 (d, J=8,4 Hz, 2H, HAr,PABA), 7,78-a 7.85 (m, 3H, 6-H, 9-H, Htriazole), 8,21 (users, 1H, 4-H). Example 49 General method of preparing compounds 50a-50c of the connection 49 A solution of compound 49 (78 mg, 0.11 mmol) in 4n HCl in ethyl acetate (15 ml) is stirred for 1 hour at room temperature. The reaction mixture was concentrated ostatok suspended in ethyl acetate (15 ml). The resulting mixture is concentrated and the residue is dried in vacuum for 1 hour. The residue is dissolved in DMF (2 ml) and the solution cooled to 0ºC. Add activated steam-nitrophenylarsonic linker 59a, 59b, or 60 (0.12 mmol) and triethylamine (38 μl, 0,272 mmol) and the reaction mixture was then warmed to room temperature. After 1.5 hours the reaction mixture was concentrated in vacuo and the crude product is purified using column chromatography (CH2Cl2/MeOH = 9:1)to give the product as not quite white solid. Connection 50c receive from his Aloc-protected analogue in accordance with the method used to produce compounds 47e and 47f. Compound 50a: 44% yield;1H-NMR (400 MHz, CDCl3/CD3OD, TFA salt): δ=0,92 (d, J=6,7 Hz, 3H, Hval), of 0.93 (d, J=6,7 Hz, 3H, Hval), 1,24-134 (m, 2H, 2×Hcapr), 1,48 is 1.75 (m, 10H, 10-CH3, 7×Hcit/Hcapr), to 1.86 (m, 1H, Hcit), to 2.06 (m, 1H, Hvai)of 2.23 (t, J=7.4 Hz, 2H, CH2C(O)), 2.91 in (c, 1H, NCH3), 3,06 (c, 3H, H3CNC(O)), is 3.08-3,20 (m, 2H, NCH2,cit), 3,24-of 3.42 (m, 4H, Hpiperazine), 3,47 (t, 2H, CH2,capr), of 3.60-4.25 in (m, 10H, 1-H, OCH2CH2N OCH2CH2N, α-Hval, 4×Hpiperazine), 4,49-of 4.67 (m, 3H, 2a-H, 10-H, α-Hcit), a 4.83 (m, 1H, 2b-H), of 5.05-5,13 (m, 4H, 2×CH2OC(O)), 6,68 (c, 2H, CH=CH), 6,88-to 7.59 (m, 14H, 6'-H, 3'-H, 4'-H, 7'-H, 7-H, 8-H, 8×HAr,PABA), 7,79 (d, J=8,2 Hz, 1H, 9-H), a 7.85 (d, J=8.0 Hz, 1H, 6-H), 8,25 (users, 1H, 4-H). Connection 50b: 74% yield;1H-NMR (30 MHz, CDCl3/CD3OD): δ=0,93 (d, J=6.8 Hz, 3H, Hval), of 0.97 (d, J=6.8 Hz, 3H, Hval), 1,49-to 1.61 (m, 2H, Hcit), and 1.63 (d, J=6,7 Hz, 3H, 10-CH3), 1,72 (m, 1H, Hcit), 1,90 (m, 1H, Hcit), 2,11 (m, 1H, Hval), 2,41 (c, 3H, N(CH3)), 2,53-to 2.67 (m, 4H, Hpiperazine), of 3.07 (c, 3H, H3CNC(O)), to 3.09 (m, 1H, Hcit), 3,21 (m, 1H, Hcit), to 3.36 (m, 2H, CH2N3), 3,59-and 3.72 (m, 8H, CH2OCH2, OCH2CH2N, 2×Hpiperazine), 3,90 (m, 2H,Hpiperazine), of 3.97 (m, 1H, 1-H), was 4.02 (d, J=6.2 Hz, 1H, α-Hval), 4,08-4,27 (m, 4H, CH2CH2OC(O)OCH2CH2N), 4,50-to 4.62 (m, 3H, 2a-H, 10-H, α-Hcit), and 4.75 (DD, 1H, 2b-H), 5,07-5,13 (m, 4H, 2×CH2OC(O)), to 6.88 (m, 1H, 6'-H), 6,97 (m, 1H, 3'-H), 7,06 and 7,11 (2×users, 1H, 4'-H), 7,25-7,38 and 7,40 -7,61 (2×m, 11H, 7-H, 8-H, 7'-H, 8×HAr,PABA), 7,73 (d, J=8,3 Hz, 1-H, 9-H), 7,86 (d, J=8,1 Hz, 1H, 6-H), 8,28 (users, 1H, 4-H). Example 50 Conjugation conjugate linker-agent 47a with Herceptin antibody Herceptin antibody (40 mg) is treated with 1.5 molar equivalents of dithiothreitol (DTT) of 0.025 M sodium borate pH 8, 0,025M NaCl, 1 mm DTPA for 2 hours at 37 degrees C. Excess DTT is removed using a column of Sephadex G-25 (0,025M sodium borate pH 8, 0,025M NaCl, 1 mm DTPA). Determination of thiol reagent of Allman shows that there are approximately three tirinya group on Herceptin. Compound 47a (1,13 mg, 1.1 equiv./SH group) is dissolved in about 100 μl DMSO and added dropwise to the solution recovered Herceptin in the reaction buffer (0,025M sodium borate pH 8, 0,025M NaCl, 1 mm DTPA). The floor is i.i.d. mixture incubated for 30 minutes at 4ºC. Excess compound 47a removed using gel filtration (G-25, PBS). The protein concentration and the content of drug was determined by spectral analysis at 280 and 320 nm, respectively. It was found that an average of 2.5 units of drugs conjugated to Herceptin. Using FPLC with the exception of the size found no traces of aggregation of the conjugate antibody-drug. Example 51 In vitro cytotoxicity of compounds (+)-8a-e and (+)-4a-e Adhesive cell line A549 sown in the triple instance 6 multilaunch tablets at concentrations of 102, 103, 104and 105cells per well. The culture medium is removed after 24 hours and cells washed in incubation media Ultraculture (UC, not containing serum special environment provided BioWhittalcer Europe, Verviers, Belgium). Then carry out incubation with the compounds (+)-8a-e and (+)-4a-e in Ultraculture medium in various concentrations for 24 hours. All substances used as freshly prepared solutions in DMSO (Merck, Darmstadt, Germany)diluted in incubation medium to a final concentration of 1% DMSO in the wells. After 24 hours exposure to the test substances were removed and the cells washed with fresh medium. The cultivation is carried out at 37 degrees C and 7.5% CO2on the air for 12 days. The medium is removed, and the clones dried and stained with methylene blue of Laffer (Merck, Darmstadt, Germany. Then they counted macroscopically. Value IR50based on the relative rate of formation of clones, which is determined in accordance with the following formula: relative rate of formation of the clones [%]=100×(number of clones counted after exposure)/(number of clones, as defined in the control). The release of drugs from their glycosidic prodrugs is achieved by adding 4,0 IU·ml-1β-D-galactosidase (EC 3.2.1.23, Grade X, the supplier Germany Sigma, Deisenhofen, Germany) to the cells during incubation with substances. Value IR50in the table below.
Example 52 In vivo evaluation of compounds (+)-8a in an orthotopic model of breast cancer in SCID mice, using the concept of ADEPT Before implantation, tumor CL is the current female mice of strain SCID anaesthetize intraperitoneal injection of 75 mg/kg cetomimidae with 15 mg/kg of xylazine. 1×106MDA-MB-231 cells (estrogen-independent human cell line breast cancer) are suspended in 25 μl of sterile superyoung phosphate saline (PBS) and then implanted in the mammary fad pat 4thcomplex breast cancer. After implantation, mice daily examine, determining the loss of weight, General condition and education of the tumor. On day 21 the animals randomly distributed in groups of 5-12 mice (control, only the antibody-enzyme only a prodrug and ADEPT therapy). On 22 and 30 day mice injected PBS (control group and the group is only a prodrug) or 50 μg monoclonal anti-human receptor activator urokinase plasminogen (uPAR) antibody conjugated with β-galactosidase uPAR*β-Gal in PBS (groups only the antibody-enzyme ADEPT therapy). On days 24, 26, 28, 32, 34 and 36 to mice intravenously injected with 1% DMSO/NaCl solution (control group, only the antibody-enzyme) or 104 μg (+)-8a in 1% DMSO/NaCl solution (only group a prodrug, ADEPT therapy). Mice killed on day 38, and the tumor excised, weighed, placed in superyoung phosphate 4% formalin for 16 hours at room temperature and conclude in paraffin. Get the tissue sections (2.5 μm), stained with hematoxylin and eosin (H & E), and examined using a conventional microscope. For immunohistochemistry, sections 2.5 microns fixed formalin and prisoners in paraffin tissue is placed on the glass, the wax is removed with xylene and rehydration in dilutions ethanol/water. Slides for Ki67 staining pretreated by boiling in a microwave oven (5×5 min in citrate buffer (pH 6.0) for the recovery of the antigen. Endogenous peroxidase inactivating the processing of 3% hydrogen peroxide for 10 minutes. Then add Tris-superyoung 10% solution of rabbit serum (Dako, Hamburg, Germany, No. X 0902) to block nonspecific binding of the protein, and the slide incubated with mouse anti-human Ki67 (Dianova, Hamburg, Germany, No. dia 505) for 60 minutes (2 μg/ml) followed by exposure biotinylating rabbit anti-mouse F(ab')2slice (Dako, No. E 0413, 16,8 µg/ml) for 30 minutes. Then the slices incubated with avidin-peroxidase solution (Dako, No. P 364), stained with aminoacylation (solution AEC substrate, Sigma), washed in water, and finally carry out contrast staining with hematoxylin. The average number of Ki67 positive nuclei counted in the three most cellular fields in tumors containing subjectively higher density of mitotic figures, and determine the ratio of the total number of nuclei stained H & E in the same areas. Tumor size was defined as a specific time points during the experiment treatment (days 21, 29 and 38) with flat panel volumetric prototype CT scanner is (fpVCT GE Global Research, Niskayuna NY, US). Anaesthetize mice pairs izoflurana at a concentration of 0.8-1% during the session to retrieve images, center on fpVCT the axis of rotation of the stand, and placed perpendicular to the z-axis of the system in such a way as to allow scanning of the entire body of the mouse for one turn. Contrast medium, 150 μl of Isovist® 300 (Schering, Berlin, Germany), administered intravenously for 20 seconds to scan. All aggregate results were obtained on the same scheme: 1000 images per rotation, 8 seconds, the time of rotation, 360 used detector rows, 80 kVA expansion, and 100 mA. For the reconstruction of images using a modified Feldkamp algorithm, receiving a set of bulk isotopic results with high resolution (512×512 matrix, a resolution of about 200 μm) References 1. Boger, D.L.; Johnson, D.S.; Wrasidlo, W. Bioorg. Med. Chem. Lett. 1994, 4, 631-636. 2. McGovren, J.P., Clarke, G.L., Pratt, E.A., DeKoning, T.F. J. Antibiot. 1984, 37, 63-70. 3. Carter, P.; Smith, L.; Ryan, M. Endocr.-Relat. Cancer 2004, 11, 659-687. 4. Bagshawe, K.D. Drug Dev. Res. 1995, 34, 220-230. 5. Melton, R.; Connors, T.; Knox, R.J. S.T.P. Pharma Sciences, 1999, 13 to 33. 6. Huber, B.E.; Richards, C.A.; Krenitsky, T.A. Proc. Natl. Acad. Sci. USA, 1991, 88, 8039-8043. 7. Bagshawe, K.D.; Springer, C.J.; Searle, F.; Antoniw, P.; Sharma, S.K.; Melton, R.G.; Sherwood, R.F. Br. J. Cancer, 1988, 58, 700-703. 8. Duncan, R. Nat. Rev. Drug Discov. 2003, 2, 347-360. 9. Tietze, L.F.; Lieb, M.; Herzig, T.; Haunert, F.; Schuberth, I. Bioorg. Med. Chem. 2001, 9, 1929-1939. 10. Tietze, L.F., Herzig, T.; Fecher, A.; Haunert, F., Schuberth, I. ChemBioChem 2001, 758-765. 11. Toki, B.E.; Cerveny, C.G.; Wahl, A.F.; Senter, P.D. . Org. Chem., 2002, 67, 1866-1872. 12. See for some recently disclosed cyclization spacers for example WO 2005/079398, WO 2005/105154, and WO 2006/012527. 13. Greenwald, R.B., Choe, Y.H., McGuire, J., Conover, C.D. Adv. Drug Delivery Rev. 2003, 55, 217-250. 14. Kingsbury, W.D.; Boehm; J.C.; Mehta, R.J.; Grappel, S.F.; Gilvarg, C. J. Med. Chem., 1984, 27, 1447-1451. 15. Greenwald, R.B.; Zhao, H.; Yang, K.; Reddy, P.; Martinez, A. J. Med. Chem. 2004, 47, 726-734. 16. (a) Franke, A.E.; Sievers, E.L.; and Scheinberg, D.A. Cancer Biother. Radiopharm. 2000, 15, 459-476. (b) Murray, J.L. Semin. Oncol. 2000, 27, 2564-2570 (c) Breitling, F., and Dubel, S., Recombinant Antibodies, John Wiley and Sons, New York, 1998. 17. Ringsdorf, H. J. Polym. Sci., Polym. Symp. 1975, 51, 135-153. 18. Elvira, C; Gallardo, A.; San Roman, J.; Cifuentes, A. Molecules 2005, 10, 114-125. 19. Remington''s Pharmaceutical Science (15th ed., Mack Publishing, Easton, PA, 1980) (incorporated by reference in its entirety for all purposes). 1. The compound of formula (I) 2. The compound according to claim 1, where R2represents C1-8alkyl. 3. The compound according to claim 2, where R2represents methyl. 4. The compound according to claim 1, where R5is not hydrogen. 5. The compound according to claim 1, where R8or R9choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino and (piperidine-4-yl)methoxy. 6. The compound according to claim 5, where R8choose from 2-(morpholine-4-yl)ethoxy, (1 methylpiperidin-4-yl)methoxy, 2-(N,N-dimethylamino)ethoxy, 2-(N,N-dimethylamino)acetylamino, 2-(methylamino)ethoxy, 2-(methylamino)acetylamino, 2-aminoethoxy, 2-aminoethylamino and (piperidine-4-yl)methoxy. 7. The compound according to claim 1, where R9is methoxy and R10and R11each represents hydrogen. 8. Connect the tion according to claim 1, which is 9. The compound according to claim 1, which is 10. The compound of formula (IIIa) 11. The connection of claim 10, where V1represents a dipeptide, Tripeptide, tetrapeptide or Oligopeptide fragment consisting of natural L amino acids, not natural D-amino acids, or synthetic amino acids or peptidomimetics, or any combinations thereof. 12. The connection of claim 10, which is a 13. The connection of claim 10, which is a 14. The compound of formula (IV) 15. The use of compounds according to claims 1, 10 or 14 for the preparation of pharmaceutical compositions for treating tumors in a mammal. 16. Pharmaceutical composition having antitumor activity containing a compound according to claims 1, 10 or 14 and a pharmaceutically acceptable carrier.
|
© 2013-2015 Russian business network RussianPatents.com - Special Russian commercial information project for world wide. Foreign filing in English. |