EP1294405A2 - Durch thimet oligopeptidase spaltbare prodrugs - Google Patents

Durch thimet oligopeptidase spaltbare prodrugs

Info

Publication number
EP1294405A2
EP1294405A2 EP01950291A EP01950291A EP1294405A2 EP 1294405 A2 EP1294405 A2 EP 1294405A2 EP 01950291 A EP01950291 A EP 01950291A EP 01950291 A EP01950291 A EP 01950291A EP 1294405 A2 EP1294405 A2 EP 1294405A2
Authority
EP
European Patent Office
Prior art keywords
seq
oligopeptide
therapeutic agent
leu
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP01950291A
Other languages
English (en)
French (fr)
Inventor
Matthew H. Nieder
Vincent Dubois
Sanjeev Gangwar
Thomas J. Lobl
Leslie B. Pickford
Andre Trouet
Geoffrey T. Yarranton
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
ER Squibb and Sons LLC
Original Assignee
Corixa Corp
Medarex LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Corixa Corp, Medarex LLC filed Critical Corixa Corp
Publication of EP1294405A2 publication Critical patent/EP1294405A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention is directed to new compounds useful as prodrugs.
  • Such prodrugs may be used for treatment of disease, especially tumors, in patients.
  • anthracyclines and vinca alkaloids are especially effective for the treatment of cancers.
  • these molecules are often characterized in vivo by an acute toxicity, especially a bone marrow and mucosal toxicity, as well as a chronic cardiac toxicity in the case of the anthracyclines and chronic neurological toxicity in the case of the vinca alkaloids.
  • methotrexate may be used for the treatment of inflammatory reactions, such as rheumatic diseases, but its high toxicity limits its applications.
  • Development of more specific and safer antitumor agents is desirable for greater effectiveness against tumor cells and a decrease in the number and severity of the side effects of these products (toxicity, destruction of non-tumor cells, etc.).
  • Prodrugs are molecules capable of being converted to drugs (active therapeutic compounds) in vivo by certain chemical or enzymatic modifications of their structure. For purposes of reducing toxicity, this conversion should be confined to the site of action or target tissue rather than the circulatory system or non-target tissue. Prodrugs are often characterized by a low stability in blood and serum, however, since blood and serum contain enzymes which degrade, or activate, the prodrugs before the prodrugs reach the desired sites within the patient's body.
  • a desirable class of prodrugs that overcomes such problems have been disclosed in Patent Cooperation Treaty International Publication No. WO 96/05863 and in U.S. Patent No. 5,962,216, both incorporated herein by reference. Further useful prodrug compounds and methods of making such prodrugs are desirable, however, as are methods of making the prodrugs.
  • a particular object of the invention is Prodrugs that display a high specificity of action, a reduced toxicity, and an improved stability in blood relative to prodrugs of similar structure (especially the closest structure) that have existed in the public domain are particularly desirable.
  • the compound of the invention is a prodrug form of a therapeutic agent, in which the therapeutic agent is linked directly or indirectly to an oligopeptide, which in turn, is linked to a stabilizing group.
  • the compound is cleavable by an enzyme associated with a target cell.
  • the present invention may be described as new prodrug compounds of a therapeutic agent, especially prodrugs comprising an antitumor therapeutic agent, displaying improved therapeutic properties relative to the products of the prior art, especially improved therapeutic properties in the treatment of cancerous tumors and/or in the treatment of inflammatory reactions such as rheumatic diseases.
  • Improved therapeutic properties include decreased toxicity and increased efficacy.
  • Particularly desired are prodrugs which display a high specificity of action, a reduced toxicity, an improved stability in the serum and blood, and which do not move into target cells until activated by a target cell associated enzyme.
  • Prodrug compounds of a marker enabling tumors to be characterized (diagnosis, progression of the tumor, assay of the factors secreted by tumor cells, etc.) are also contemplated.
  • the invention includes a diagnosis or assay kit employing a compound of the invention.
  • the present invention also relates to the pharmaceutical composition
  • the pharmaceutical composition comprising the compound according to the invention and optionally a pharmaceutically acceptable carrier, adjuvant, vehicle, or the like.
  • a method of decreasing toxicity and improving safety index by modifying a therapeutic agent to create a prodrug is disclosed.
  • Other aspects of the invention include a method of designing a prodrug for administration to a patient and a method for treating a patient by administering a therapeutic dose of the compound.
  • Several processes for creating a prodrug of the invention are also described.
  • Figs. 1A-1D are a table of abbreviations, names, and structures.
  • Fig. 2 is an exemplary scheme of cleavage of a prodrug of the invention in the extracellular vicinity of the target cell and within the target cell.
  • Fig. 3 illustrates a synthesis of Fmoc- ⁇ Ala-Leu- Ala-Leu, a typical intermediate of the invention.
  • Fig. 4 illustrates an "Fmoc-route" synthesis of Methyl-succinyl- ⁇ Ala-Leu-Ala-
  • Fig. 5 illustrates an "Fmoc route" synthesis of a salt form of Suc- ⁇ Ala-Leu- Ala- Leu -DOX, a typical compound of the invention.
  • Fig. 6 illustrates an "Ester route" synthesis of a salt form of Suc- ⁇ Ala-Leu- Ala- Leu -DOX, a typical compound of the invention.
  • Fig. 7 illustrates a synthesis of an amino-protected ⁇ Ala-Leu- Ala-Leu-DOX, a typical intermediate of the invention.
  • Fig. 8 illustrates an "Allyl ester route” synthesis of a salt form of Suc- ⁇ Ala-Leu- Ala-Leu-DOX, a typical compound of the invention.
  • Fig. 9 illustrates a "Resin route” synthesis of Suc- ⁇ Ala-Leu- Ala-Leu-DOX, a typical compound of the invention.
  • Figs. 10A-10C are a table of oligopeptides useful in the prodrug of the invention.
  • Fig. 11 is a graph of survival in a mouse xenograft model for animals given vehicle with or without drug.
  • Fig. 12 is a graph of survival in a mouse xenograft model comparing a doxorubicin prodrug and doxorubicin.
  • Fig. 13 is a graph of the activation and then inhibition of HeLa cell trouase by increasing concentrations of DTT.
  • Fig. 14 illustrates the removal of free therapeutic agent through the use of scavenging resin or beads.
  • Fig. 15 is a graph of tumor growth inhibition in a mouse xenograft model.
  • Fig. 16 illustrates a large scale synthesis of MeOSuc- ⁇ Ala-Leu- Ala-Leu-Dox, a typical intermediate of the invention.
  • Fig. 17 illustrates the NMR assignment for MeOSuc- ⁇ Ala-Leu-Ala-Leu-Dox, a typical compound of the invention.
  • Fig. 18 is a comparison of the effects of Suc-b Ala-Leu- Ala-Leu-Dox and doxorubicin compared with vehicle on the growth of MX-1 human breast tumors in female nude mice.
  • Aib Aminoisobutyric acid All - Allyl
  • Aloe Allyloxycarbonyl
  • Dox-HCL Hydrochloride salt of Doxorubicin
  • HATU O-(7-A-zabe ⁇ zotriazol-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate
  • HBTU 2-(lH-Benzotriazole-l-yl)l,l,3,3-tetramethyluronium hexafluorophosphate
  • MeOSuc Methyl hemisuccinate/Methyl hemisuccinyl
  • NMP N-methylpyrrolidine
  • Nva Norvaline
  • PAM resin 4-hydroxymethylphenylacetamidomethyl
  • Phg Phenylglycine
  • SD-MTD Single dose-Maximum Tolerated Dose
  • TCE trichloroethyl
  • Tic Tetrahydroisoquinoline-3-carboxylic acid
  • the invention includes compounds that may be described as prodrug forms of therapeutic agents. Each of these therapeutic agents is modified by linking directly or indirectly to an oligopeptide, which in turn, is linked to a stabilizing group.
  • the prodrug, and especially the oligopeptide portion of the prodrug, are cleavable by an enzyme associated with a target cell.
  • the enzyme is preferably a trouase and more preferably is Thimet ohgopeptidase.
  • the prodrug of the invention is a modified form of a therapeutic agent and comprises several portions, including: (1) a therapeutic agent,
  • the stabilizing group is directly linked to the oligopeptide at a first attachment site of the oligopeptide.
  • the oligopeptide is directly or indirectly linked to the therapeutic agent at a second attachment site of the oligopeptide. If the oligopeptide and the therapeutic agent are indirectly linked, then a linker group is present.
  • Direct linkage of two portions of the prodrug means a covalent bond exists between the two portions.
  • the stabilizing group and the oligopeptide are therefore directly linked via a covalent chemical bond at the first attachment site of the oligopeptide, typically the N-terminus of the oligopeptide.
  • the oligopeptide and the therapeutic agent are directly linked then they are covalently bound to one another at the second attachment site of the oligopeptide.
  • the second attachment site of the oligopeptide is typically the C-terminus of the oligopeptide, but may be elsewhere on the oligopeptide.
  • Indirect linkage of two portions of the prodrug means each of the two portions is covalently bound to a linker group.
  • the prodrug has indirect linkage of the oligopeptide to the therapeutic agent.
  • the oligopeptide is covalently bound to the linker group which, in turn, is covalently bound to the therapeutic agent.
  • the prodrug of the invention is cleavable within its oligopeptide portion.
  • the prodrug typically undergoes in vivo modification and an active portion, i.e., a transport- competent portion, of the prodrug enters the target cell.
  • a first cleavage within the oligopeptide portion of the prodrug may leave a transport-competent portion of the prodrug as one of the cleavage products.
  • further cleavage by one or more peptidases may be required to result in a portion of the prodrug that is capable of entering the cell.
  • the active or transport-competent portion of the prodrug has at least the therapeutic agent and is that part of the prodrug which can enter the target cell to exert a therapeutic effect directly or upon further conversion within the target cell.
  • the compound has an active portion, and the active portion is more capable of entering the target cell after cleavage by an enzyme associated with a target cell than prior to cleavage by the enzyme associated with a target cell.
  • the structures of the stabilizing group and oligopeptide are selected to limit clearance and metabolism of the prodrug by enzymes which may be present in blood or non-target tissue and are further selected to limit transport of the prodrug into cells.
  • the stabilizing group blocks degradation of the prodrug and may act in providing preferable charge or other physical characteristics of the prodrug.
  • the amino acid sequence of the oligopeptide is designed to ensure specific cleavage by an enzyme associated with a target cell, more specifically by a trouase enzyme, and even more specifically by Thimet ohgopeptidase ("TOP").
  • the target cells are usually tumor cells or cells participating in inflammatory reactions, especially those associated with rheumatic diseases, such as macrophages, neutrophils, and monocytes.
  • Modification of the therapeutic agent to a prodrug form reduces some of the side effects of the therapeutic agents. Modification of the therapeutic agent to a prodrug form further allows for administration of an increased dosage of the therapeutic agent in prodrug form to the patient relative to the dosage of the therapeutic agent in unconjugated form.
  • the therapeutic agent acts either directly on its specific intracellular action site or, after a modification under the action of intracellular peptidases, kills the target cell or blocks its proliferation. Since normal cells release little to no TOP in vivo, the compound according to the invention is maintained inactive and does not enter the normal cells or does so in a relatively minor amount. Although TOP is believed to be widely distributed in the body, it is typically present as an intracellular enzyme. Therefore it is not generally accessible to peptide prodrugs in the circulation, i the environment of the tumor, TOP is believed to be released from necrotic tissue.
  • the prodrug is administered to the patient, carried through the blood stream in a stable form, and when in the vicinity of a target cell, is acted upon by TOP. Since the enzyme activity is only minimally present within the extracellular vicinity of normal cells, the prodrug is maintained and its active portion (including the therapeutic agent) gains entry into the normal cells only minimally, at best. In the vicinity of tumor or other target cells, however, the presence of TOP in the local environment causes cleavage of the prodrug. Once the stabilizing group is removed, further amino acids can be removed by other peptidases in the vicinity of target cells.
  • the example shown in Fig. 2 depicts an N-capped tetrapeptide prodrug being cleaved extracellularly and gaining entry into the target cell.
  • the target cell may be further modified to provide therapeutic effect, such as by killing the target cell or blocking its proliferation.
  • the active portion of the prodrug may also enter the normal cells to some extent, the active portion is freed from the remainder of the prodrug primarily in the vicinity of target cells. Thus, toxicity to normal cells is minimized.
  • normal cells means non-target cells that would be encountered by the prodrug upon administration of the prodrug in the manner appropriate for its intended use. Since normal (i.e., non-target) cells liberate little or none of the target-cell enzyme(s), e.g., TOP, that are responsible for cleaving the bond that links the active portion (including the therapeutic agent) of the prodrug from the remainder of the prodrug in vivo, the compound of the invention is maintained inactive and does not enter the normal cells.
  • target-cell enzyme(s) e.g., TOP
  • the orientation of the prodrug may be reversed so that the stabilizing group is attached to the C-terminus of the oligopeptide and the therapeutic agent is directly or indirectly linked to the N-terminus of the oligopeptide.
  • the first attachment site of the oligopeptide may be the C-terminus of the oligopeptide and the second attachment site of the oligopeptide may be the N-terminus of the oligopeptide.
  • the linker group may optionally be present between the therapeutic agent and the oligopeptide.
  • the prodrugs of the invention are designed to take advantage of preferential activation through interaction with an enzyme associated with the target cell, at or near the site targeted within the body of the patient.
  • an enzyme associated with the target cell is trouase, described in greater detail in PCT/US99/30393, incorporated herein by reference.
  • Trouase is the type of enzyme that is thought to activate the prodrug at the target tissue.
  • Trouase is a class of endopeptidases which shows a remarkable degree of discrimination between leucine and isoleucine at the carboxyl side of the oligopeptide cleavage site.
  • a defining characteristic is that under appropriate assay conditions, a trouase readily cleaves Suc- ⁇ Ala-Leu-Ala-Leu-Dnr while it is at least twenty-fold less active with Suc- ⁇ Ala-Ile-Ala-Leu-Dnr.
  • TOP is a member of the trouase class of enzymes.
  • Target cells are believed to release trouase.
  • the enzyme is generated either by target cells or by normal cells that are associated with the target cells, such as stromal tissue, neutrophils, eosinophils, macrophages or B cells.
  • the target cell associated enzyme may be associated with or bound on (at least the active site) the outer cell surface, secreted, released, or present in some other manner in the extracellular vicinity of the target cell. So, for example, the trouase may be secreted or present in some other manner in the extracellular vicinity of the target cell.
  • the prodrug of the invention includes a therapeutic agent for the treatment of cancer and the target cell is a tumor cell.
  • the trouase may be secreted extracellularly by the tumor cell or it may be present extracellularly, e.g., because there is a fair amount of cell lysis associated with tumors generally.
  • Cell lysis is also associated with inflammatory tissue, another target site.
  • Trouase activity is low in human plasma, however. Trouase activity has been observed in carcinoma cell extracts and conditioned media from cultured carcinoma cells, red blood cells and various human tissues, especially kidney.
  • a partial purification scheme of trouase from HeLa (cervical carcinoma) cell homogenate ultracentrifugation (145,000xg 30 min) supernatant consists of four steps as follows:
  • TOP is an example of a trouase.
  • the trouase isolated from these sources is believed to be TOP.
  • Both structural and functional evidence indicate that a trouase found in carcinoma cells is Thimet ohgopeptidase or "TOP".
  • TOP, or EC 3.4.24.15 is a thiol-activated zinc metallopeptidase which catalyzes internal (endo) cleavage of various oligopeptides having 6 to 17 amino acids (Dando, et al., "Human thimet ohgopeptidase,” Biochem J 294:451-457 (1993)). It also is referred to as Pz-peptidase, coUagenase-like peptidase, kininase A, amyloidin protease, and metalloendopeptidase 24.15.
  • the enzyme has been isolated from chicken embryo (Morales, et al., "PZ-peptidase from chick embryos. Purification, properties, and action on collagen peptides," JBiol Chem 252:4855-4860 (1977)), chicken liver (Barrett, et al., “Chicken liver Pz-peptidase, a thiol-dependent metallo-endopeptidase," Biochem J271 :701-706 (1990)), rat testis (Orlowski, et al.,
  • the gene for this enzyme has been cloned and DNA sequence obtained from human brain (Dovey et al., WO92/07068), rat testis (Pierotti, et al., "Endopeptidase-24.15 in rat hypothalamic/pituitary/gonadal axis," Mol Cell Endocrinol 76:95-103 (1991)) and pig liver (Kato, et al., "Cloning, amino acid sequence and tissue distribution of porcine thimet ohgopeptidase. A comparison with soluble angiotensin-binding protein," EurJ Biochem 221:159-165 (1994)).
  • Trouase purified from HeLa cells shows sequence identity with human TOP based on the mass-to-charge ratio of tryptic fragments covering 33% of all residues distributed over the full length of the known human enzyme sequence, as seen in Example 12.
  • Immunoprecipitation using a specific anti-TOP antibody preparation with partially purified HeLa cell fraction (FI) and MCF-7/6 cell homogenate also indicates structural identity, as seen in Example 13.
  • the size, by SDS polyacrylamide gel electrophoresis, of the 74KD purified HeLa cell trouase is within the range reported for TOP.
  • a minor 63KD band, which co-purifies from HeLa cells, has not been previously reported and may be a proteolysis product of TOP formed during extraction.
  • the gel filtration estimated size of the native HeLa cell trouase is 68KD rather than the reported TOP size of 78KD; however, the difference may be explained by the inherent error of such native protein size estimation methods.
  • the isoelectric point of the purified carcinoma cell trouase is 5.2, as seen in Example 9, which is within the range reported for TOP.
  • TOP and human carcinoma cell trouase show the same substrate specificity with nine different experimental compounds, as seen in Example 6. This specificity includes the ability to cleave Suc- ⁇ Ala-Leu- Ala-Leu-Dox at approximately a twenty-fold faster rate than cleavage of Suc- ⁇ Ala-Ile- Ala-Leu-Dox.
  • the carcinoma cell trouase also has essentially the same pH optimum (see Example 11) and inhibitor profile (see Example 8) as TOP.
  • carcinoma cell trouase is inhibited by the metallopeptidase inhibitors EDTA and 1,10-phenanthroline but not serine, thiol, or acid proteinase inhibitors such as aminoethylbenzene-sufonate, E64, pepstatin, leupeptin, aprotinin, CA074, or fumagillin.
  • metallopeptidase inhibitors EDTA and 1,10-phenanthroline but not serine, thiol, or acid proteinase inhibitors such as aminoethylbenzene-sufonate, E64, pepstatin, leupeptin, aprotinin, CA074, or fumagillin.
  • EDTA- treated carcinoma cell trouase is reactivated by Co 2+ (50-100 ⁇ M) or Mn 2+ (50-1000 ⁇ M).
  • Co 2+ 50-100 ⁇ M
  • Mn 2+ 50-1000 ⁇ M
  • it is also possible to reactivate EDTA-deactivated chicken Barrett, et al., “Chicken liver Pz-peptidase, a thiol-dependent metallo-endopeptidase," Biochem J 271 :701-706 (1990)
  • rat Orlowski et al., "Endopeptidase 24.15 from rat testes.
  • TOP activity may be inhibited in oxygenated solutions (such as blood) and activated in mildly reducing (hypoxic) environments, as demonstrated by thiol activation of air-inactivated preparations (Shrimpton, et al., "Thiol activation of endopeptidase EC 3.4.24.15. A novel mechanism for the regulation of catalytic activity," JBiol Chem 272: 17395-17399 (1997)). Accordingly, it is a useful enzyme for a general approach to designing prodrugs that are to be activated in hypoxic environments such as tumor tissue.
  • CD10 (CALLA, neprilysin, neutral endopeptidase, EC 3.4.24.11) is an ohgopeptidase bound to the outer cell membrane of a number of cells including a limited number of cancer tumor types. Since it is also present in high concentrations in the brush boarder of the proximal kidney tubule, and at lower levels in some colon tissue and a number of immune system cells such as B-lymphocytes it may contribute to systemic activation of peptidyl prodrugs. This added systemic activation could lead to increased toxicity to normal tissues when compared to a peptidyl prodrug that is not a CD 10 substrate.
  • Su- ⁇ Ala-Leu- Ala-Leu-Dox is a substrate for CD 10 with cleavage occurring between Ala and Leu (AA2 and AA1) as shown in Example 17.
  • CD10 cleaves poorly when glycine or alanine is present in the PI' cleavage site (Pozgay et al, Biochemistry (1986)) [see the CD10 patent under substrate specificity] thus Sue ⁇ Ala-Leu-Ala-Gly- Dox and ⁇ Ala-Leu- Ala- Ala-Dox are expected to be poorly cleaved by CD 10.
  • Suc- ⁇ Ala-Leu-Ala-Gly-Dox (and presumably Suc- ⁇ Ala-Leu- Ala- Ala-Dox are well cleaved by TOP.
  • the preferred embodiment of this invention is a compound which is activated by TOP but not by CD 10 as exemplified by Suc- ⁇ Ala-Leu-Ala-Gly-Dox or Suc- ⁇ Ala-Leu-Ala- Ala-Dox.
  • the prodrug when treating non-CD 10 containing tumors the prodrug is not cleavable by the CD 10 enzyme.
  • the stabilizing group serves to protect the prodrug compound from cleavage in circulating blood when it is administered to the patient and allows the prodrug to reach the vicinity of the target cell relatively intact.
  • the stabilizing group typically protects the prodrug from cleavage by proteinases and peptidases present in blood, blood serum, and normal tissue.
  • the stabilizing group caps the N-terminus of the oligopeptide, and is therefore sometimes referred to as an N-cap or N-block, it serves to ward against peptidases to which the prodrug may otherwise be susceptible.
  • the stabilizing group is useful in the prodrug of the invention if it serves to protect the prodrug from degradation, i.e., cleavage, when tested by storage of the prodrug compound in human blood at 37°C for 2 hours and results in less than 20%, preferably less than 2%, cleavage of the prodrug by the enzymes present in the human blood under the given assay conditions.
  • the stabilizing group is either (1) other than an amino acid, or
  • an amino acid that is either (i) a non-genetically-encoded amino acid having four or more carbons or (ii) aspartic acid or glutamic acid attached to the N-terminus of the oligopeptide at the ⁇ -carboxyl group of aspartic acid or the ⁇ -carboxyl group of glutamic acid.
  • dicarboxylic (or a higher order carboxylic) acid or a pharmaceutically acceptable salt thereof may be used as a stabilizing group. Since chemical radicals having more than two carboxylic acids are also acceptable as part of the prodrug, the end group having dicarboxylic (or higher order carboxylic) acids is an exemplary N-cap.
  • the N-cap may thus be a monoamide derivative of a chemical radical containing two or more carboxylic acids where the amide is attached onto the amino terminus of the peptide and the remaining carboxylic acids are free and uncoupled.
  • the N-cap is preferably succinic acid, adipic acid, glutaric acid, or phthalic acid, with succinic acid being most preferred.
  • N-caps in the prodrug compound of the invention include diglycohc acid, fumaric acid, naphthalene dicarboxylic acid, pyroglutamic acid, acetic acid, 1- or 2- naphthylcarboxylic acid, 1,8- naphthyl dicarboxylic acid, aconitic acid, carboxycinnamic acid, triazole dicarboxylic acid, gluconic acid, 4-carboxyphenyl boronic acid, a (PEG) n -analog such as polyethylene glycolic acid, butane disulfonic acid, maleic acid, isonipecotic acid, and nipecotic acid.
  • PEG polyethylene glycolic acid
  • butane disulfonic acid maleic acid
  • isonipecotic acid and nipecotic acid.
  • the linked stabilizing group be negatively charged or neutral.
  • cytotoxic compounds inherently have low solubility.
  • Positively charged anthracyclines for example form aggregates at high concentration and these aggregates may induce intravenous coagulation when the aggregates are administered intravenously.
  • Trouase recognizes a specific set of peptide sequences.
  • a cytotoxic compound for example: doxorubicin
  • it results in a less soluble compound which may form large aggregates when injected intravenously as a concentrated bolus. Since most peptides have exposed, positively-charged amino termini at physiological pH, these aggregates may form a polypositively charged surface in vivo.
  • Oligopeptides are generally defined as polypeptides of short length, typically twenty amino acids or fewer.
  • An oligopeptide useful in the prodrug of the invention is at least four amino acids in length, however. At the upper end, oligopeptides of less than or equal to twelve amino acids are most useful, although an oligopeptide may have a chain length greater than twelve amino acids and fall within both the definition of the term as generally recognized in the scientific field and additionally within the scope of the invention.
  • the oligopeptide portion of the prodrug of the invention has four or more amino acids.
  • the oligopeptide portion of the prodrug of the invention has four to twelve amino acids, inclusive. Preferably, it has four or five amino acids.
  • the oligopeptide has a formula or sequence (AA) n -AA 4 -AA 3 -AA 2 -AA 1 , wherein: each AA independently represents an amino acid; n is an integer from 0 to 16; AA 4 represents a non-genetically-encoded amino acid;
  • AA 3 represents any amino acid
  • AA represents any amino acid
  • AA represents any amino acid. This corresponds to a position sequence P(n+2)...P2-P1-P1'-P2'.
  • the TOP is believed to cleave between the PI and PI' positions.
  • the oligopeptide is written in the conventional manner with the carboxyl-terminus (or C-terminus) at the right and the amino-terminus (or N-terminus) at the left.
  • AA 1 is the carboxyl-terminus.
  • amino acids are in the L configuration. Although any amino acids may be present in the oligopeptide portion of the prodrug, with the exception of AA 4 , which is a non-genetically-encoded amino acid serving a blocking function as described in further detail below, certain amino acids are preferred.
  • one of the following amino acids is most preferably present: ⁇ -Alanine, Thiazolidine-4-carboxylic acid, 2-Thienylalanine, 2- Naphthylalanine, D-Alanine, D-Leucine, D-Methionine, D-Phenylalanine, 3-Amino-3- phenylpropionic acid, ⁇ -Aminobutyric acid, and 3-amino-4,4-diphenylbutyric acid.
  • Tetrahydroisoquinoline-3-carboxylic acid 4-Aminomethylbenzoic acid, nipecotic acid, isonipecotic acid, or Aminoisobutyric acid are also preferred in the P2 position.
  • one of the following amino acids is most preferred: Leucine, Tyrosine, Phenylalanine, j-.-Cl-Phenylalanine, ⁇ -Nitrophenylalanine, Valine, Norleucine, Norvaline, Phenylglycme, Tryptophan, Tetrahydroisoquinoline-3-carboxylic acid, 3-Pyridylalanine, Alanine, Glycine, or Thienylalanine. Also preferred are Methionine, Valine, or Proline in the PI position.
  • AA 2 is most preferably selected from the following amino acids: Alanine, Leucine, Tyrosine, Glycine, Serine, 3-Pyridylalanine, 2-Thienylalanine, Norleucine, Homoserine, Homophenylalanine, p-Cl-phenylalanine, or p-
  • Nitrophenylalanine Also preferred in this position are Aminoisobutyric Acid, Threonine, and Phenylalanine.
  • one of the following amino acids is most preferably present: Leucine, Phenylalanine, Isoleucine, Alanine, Glycine, Tyrosine, 2- Naphthylalanine, Serine, p-Cl-phenylalanine, p-Nitrophenylalanine, l-Naphthylalanine, Threonine, Homoserine, Cyclohexylalanine, Thienylalanine, Homophenylalanine, or Norleucine. Also preferred is ⁇ -Alanine in the P2' position.
  • Oligopeptides especially useful in the prodrug of the invention include those shown in Figs. 10A-10D, particularly one the following: D-AlaThi ⁇ Ala ⁇ AlaLeuAlaLeu (SEQ ID NO: 1), Thi ⁇ Ala ⁇ AlaLeuAlaLeu (SEQ ID NO: 2), ⁇ Ala ⁇ AlaLeuAlaLeu (SEQ ID NO: 3), ⁇ AlaAlaAlalle (SEQ ID NO: 4), ⁇ AlaAlaAlaLeu (SEQ ID NO: 5), ⁇ AlaPheTyrLeu (SEQ ID NO: 6), ⁇ AlaPheThrPhe (SEQ ID NO: 7), ⁇ AlaPheGlylle (SEQ ID NO: 8), ⁇ AlaPheGlyLeu (SEQ ID NO: 9), ⁇ AlaPhePhePhe (SEQ ID NO: 10), ⁇ AlaPhePhelle (SEQ ID NO: 11), ⁇ AlaPhePheLeu (
  • the oligopeptide portion of the prodrug includes a blocking amino acid as AA 4 of the oligopeptide sequence, i.e. at position P2 of the position sequence, according to the numbering scheme described above.
  • the blocking amino acid is a non-genetically- encoded amino acid.
  • the function of the blocking amino acid at position P2 is to maintain selectivity for cleavage of the prodrug by TOP and inhibit cleavage of the oligopeptide by, or at least avoid providing a cleavage site for, other enzymes in that portion of the oligopeptide most closely linked (directly linked or indirectly linked) to the therapeutic agent portion of the prodrug compound. More particularly, by placing a blocking amino acid at position P2, undesired cleavage within the peptide linkages of the four amino acids of the oligopeptide sequence AA 4 -AA 3 -AA 2 -AA 1 and position sequence P2-P1- Pl'-P2' is reduced.
  • trouase cleaves between the PI and PI' positions of the oligopeptide. Since it is known that blood and normal cells are associated with a variety of peptidases, placing a blocking amino acid at position P2 serves to protect the oligopeptide portion of the prodrug in vivo until the prodrug is in the vicinity of the target cell. Specifically, by placing a blocking amino acid at position P2, it is believed that the oligopeptide is protected from undesired cleavage between P2 and PI . Without the blocking amino acid, the prodrug might be vulnerable to both exopeptidases and endopeptidases present in blood and normal tissue, both classes of enzymes which might otherwise degrade the prodrug before it reaches its target. Example 14 below illustrates this important feature of the prodrug.
  • AA 3 represents any amino acid
  • AA 2 represents any amino acid
  • AA 1 represents any amino acid
  • the oligopeptide may be linked to a therapeutic agent and/or a stabilizing group when testing for cleavability by TOP.
  • Therapeutic agents that are particularly advantageous to modify to a prodrug form according to the invention are those with a narrow therapeutic window.
  • a drug or therapeutic agent with a narrow therapeutic window is one in which the dose at which toxicity is evident, by general medical standards, is very close to the dose at which efficacy is evident.
  • the therapeutic agent conjugated to the stabilizing group and oligopeptide and, optionally, the linker group to form the prodrug of the invention may be useful for treatment of cancer, inflammatory disease, or some other medical condition.
  • the therapeutic agent is selected from the following classes of compounds:
  • Alkylating Agents Antiproliferative agents, Tubulin Binding agents, Ninca Alkaloids, Enediynes, Podophyllotoxins or Podophyllotoxin derivatives, the Pteridine family of drugs, Taxanes, Anthracyclines, Dolastatins, Topoiosomerase inhibitors, Mytansinoids, and Platinum coordination complex chemotherapeutic agents.
  • the therapeutic agent is advantageously selected from the following compounds or a derivative or analog thereof: Doxorubicin, Daunorubicin, Vinblastine, Vincristine, Calicheamicin, Etoposide, Etoposide phosphate, CC-1065, Duocarmycin, KW-2189, Methotrexate, Methopterin, Aminopterin, Dichloromethotrexate, Docetaxel, Paclitaxel, Epithiolone, Combretastatin, Combretastatin A Phosphate, Dolastatin 10,
  • Fluorouracil 6-Mercaptopurine, Fludarabine, Tamoxifen, Cytosine arabinoside,
  • derivative is intended a compound that results from reacting the named compound with another chemical moiety, and includes a pharmaceutically acceptable salt, acid, base or ester of the named compound.
  • analog is intended a compound having similar structural and functional properties, such as biological activities, to the named compound.
  • Linker structures are dictated by the required functionality. Examples of potential linker chemistries are hydrazide, ester, ether, and sulphydryl.
  • Amino caproic acid is an example of a bifunctional linker group. When amino caproic acid is used in the linker group, it is not counted as an amino acid in the numbering scheme of the oligopeptide.
  • the optionally present linker group is not cleavable by TOP, i.e. it is not cleavable by TOP under physiological conditions
  • An especially useful embodiment is a compound that is cleavable by a trouase but resistant to cleavage by CD 10 or other systemic or blood enzymes.
  • a method of designing a prodrug is another aspect of the invention and entails initially identifying an oligopeptide as described above. Then the oligopeptide is linked at a first attachment site of the oligopeptide to a stabilizing group that hinders cleavage of the oligopeptide by enzymes present in whole blood, and directly or indirectly linked to a therapeutic agent at a second attachment site of the oligopeptide.
  • the linkage of the oligopeptide to the therapeutic agent and the stabilizing group may be performed in any order or concurrently.
  • the resulting conjugate is tested for cleavability by TOP.
  • the resulting conjugate may also be tested for stability in whole blood. Test compounds stable in whole blood are selected.
  • the first attachment site is usually the N-terminus of the oligopeptide but may be the C-terminus of the oligopeptide or another part of the oligopeptide.
  • the second attachment site is usually the C-terminus of the oligopeptide, but may be the N-terminus of the oligopeptide or another part of the oligopeptide.
  • a prodrug designed by such a method is also part of the invention.
  • the invention includes a method for decreasing toxicity of a therapeutic agent that is intended for administration to a patient.
  • a modified, prodrug form of the therapeutic agent is formed by directly or indirectly linking the therapeutic agent to an oligopeptide cleavable by a trouase, or more specifically, cleavable by TOP.
  • the oligopeptide is also linked to a stabilizing group.
  • the prodrug thus formed provides for decreased toxicity of the therapeutic agent when administered to the patient.
  • the modification of the therapeutic agent in this manner also allows for administration of an increased dosage of the therapeutic agent to the patient relative to the dosage of the therapeutic agent in unconjugated form.
  • the invention also includes a pharmaceutical composition
  • a pharmaceutical composition comprising a compound, particularly a prodrug compound, according to the invention and, optionally, a pharmaceutically acceptable carrier, for example an adjuvant or vehicle, or the like.
  • the invention also relates to the use of the pharmaceutical composition for the preparation of a medicinal product intended for the treatment of a medical condition.
  • the pharmaceutical composition may, for example, be administered to the patient parenterally, especially intravenously, intramuscularly, or intraperitoneally.
  • compositions of the invention for parenteral administration comprise sterile, aqueous or nonaqueous solutions, suspensions, or emulsions.
  • a pharmaceutically acceptable solvent or vehicle propylene glycol, polyethylene glycol, injectable organic esters, for example ethyl oleate, or cyclodextrins may be employed.
  • Isotonic saline may be part of the pharmaceutical composition.
  • These compositions can also comprise wetting, emulsifying and/or dispersing agents.
  • the sterilization may be carried out in several ways, for example using a bacteriological filter, by incorporating sterilizing agents in the composition or by irradiation. They may also be prepared in the form of sterile solid compositions which may be dissolved at the time of use in sterile water or any other sterile injectable medium.
  • the pharmaceutical composition may also comprise adjuvants which are well known in the art (e.g., vitamin C, antioxidant agents, etc.) and capable of being used in combination with the compound of the invention in order to improve and prolong the treatment of the medical condition for which they are administered.
  • adjuvants which are well known in the art (e.g., vitamin C, antioxidant agents, etc.) and capable of being used in combination with the compound of the invention in order to improve and prolong the treatment of the medical condition for which they are administered.
  • Doses for administration to a patient of the compounds according to the invention are generally at least the usual doses of the therapeutic agents known in the field, described in Brace A. Chabner and Jerry M. Collins, Cancer Chemotherapy, Lippincott Ed., ISBN 0-397-50900-6 (1990) or they may be adjusted, within the judgment of the treating physician, to accommodate the superior effectiveness of the prodrug formulations or the particular circumstances of the patient being treated.
  • the doses administered hence vary in accordance with the therapeutic agent used for the preparation of the compound according to the invention.
  • a method for the therapeutic treatment of a medical condition that involves administering, especially parenterally or intravenously, to the patient a therapeutically effective dose of the pharmaceutical composition is also within the scope of the invention.
  • a method for treating a patient includes administering to the patient a therapeutically effective amount of a compound comprising:
  • AA 4 represents a non-genetically-encoded amino acid, AA represents any amino acid, AA represents any amino acid, and AA 1 represents any amino acid, (3) a stabilizing group, and
  • a linker group not cleavable by TOP wherein the oligopeptide is directly linked to the stabilizing group at a first attachment site of the oligopeptide and the oligopeptide is directly linked to the therapeutic agent or indirectly linked through the linker group to the therapeutic agent at a second attachment site of the oligopeptide, wherein the stabilizing group hinders cleavage of the compound by enzymes present in whole blood, and wherein the compound is cleavable by an enzyme associated with the target cell.
  • the enzyme associated with the target cell is preferably a trouase and, more preferably, TOP.
  • the prodrug compound is useful for the treatment of many medical conditions including cancer, neoplastic diseases, tumors, inflammatory diseases, and infectious diseases. Examples of preferred diseases are breast cancer, colorectal cancer, liver cancer, lung cancer, prostate cancer, ovarian cancer, brain cancer, and pancreatic cancer.
  • the prodrug compound of the invention is also useful in addressing the problem of multi-drug resistant target cells.
  • MDR multi- drug resistance
  • doxorubicin and other adriamycin analogs There are various underlying mechanisms of action of MDR that generally involve changes in expression or activity of a range of transport-associated cell membrane proteins. These include the P-glycoprotein pump, which actively transports doxorubicin or other therapeutic agent out of cells.
  • the effective dose to kill tumor cells increases until it approaches the overall toxic dose.
  • otherwise very effective chemotherapeutics are no longer useful as drugs, due to unacceptable side- effect levels and lethality.
  • a method of treating resistance to a therapeutic agent in a patient in need of such treatment comprises: administering to the patient a therapeutically effective amount of a compound comprising:
  • a therapeutic agent capable of entering a target cell (2) an oligopeptide of the formula (AA) n -AA 4 -AA 3 -AA 2 -AA 1 , wherein: each AA independently represents an amino acid, n is an integer from 0 to 16,
  • AA 4 represents a non-genetically-encoded amino acid, AA represents any amino acid,
  • AA 2 represents any amino acid
  • AA 1 represents any amino acid
  • a linker group not cleavable by TOP wherein the oligopeptide is directly linked to the stabilizing group at a first attachment site of the oligopeptide and the oligopeptide is directly linked to the therapeutic agent or indirectly linked through the linker group to the therapeutic agent at a second attachment site of the oligopeptide, wherein the stabilizing group hinders cleavage of the compound by enzymes present in whole blood, and wherein the compound is cleavable by TOP.
  • the prodrug compound can be administered to animals or humans in intravenous doses ranging from 0.05 mg/kg/dose/day to 300 mg/kg/dose/day. It can also be administered via intravenous drip or other slow infusion method.
  • human patients are the usual recipients of the prodrug of the invention, although veterinary usage is also contemplated.
  • an article of manufacture such as a kit, for diagnosis or assay is also within the scope of the invention.
  • Such an article of manufacture would preferably utilize a compound as described above, except that a marker, such as coumarin is conjugated to the oligopeptide and stabilizing group instead of a therapeutic agent.
  • a marker intends any moiety that can be conjugated to the oligopeptide and is readily detectable by any method known in the art.
  • At least one reagent useful in the detection of the marker is typically included as part of the kit.
  • the article of manufacture would include the following: (1) a compound comprising: (a) a marker,
  • each AA independently represents an amino acid
  • n is an integer from 0 to 16
  • AA 4 represents a non-genetically-encoded amino acid
  • AA 3 represents any amino acid
  • AA 2 represents any amino acid
  • AA 1 represents any amino acid
  • a linker group not cleavable by TOP wherein the oligopeptide is directly linked to the stabilizing group at a first attachment site of the oligopeptide and the oligopeptide is directly linked to the marker or indirectly linked through the linker group to the marker at a second attachment site of the oligopeptide, wherein the stabilizing group hinders cleavage of the compound by enzymes present in whole blood, and wherein the compound is cleavable by TOP, and (2) optionally at least one reagent useful in the detection of said marker.
  • the article of manufacture may be used, for example, with patient samples to diagnose tumors or to identify patients susceptible to treatment by prodrug therapy.
  • Oligopeptide General Method for the synthesis of peptides
  • the peptide, or oligopeptide, sequences in the prodrug conjugates of this invention may be synthesized by the solid phase peptide synthesis (using either Boc or Fmoc chemistry) methods or by solution phase synthesis.
  • Boc and Fmoc methods are widely used and are described in the following references: Merrifield, J. A. Chem. Soc, 88:2149 (1963); Bodanszky and Bodanszky, Ebe Practice of Peptide Synthesis, Springer- Verlag, Berlin, 7-161 (1994); Stewart, Solid Phase Peptide Synthesis, Pierce Chemical, Rockford, (1984).
  • a peptide of desired length and sequence is synthesized through the stepwise addition of amino acids to a growing chain which is linked to a solid resin.
  • useful Fmoc compatible resins include, but are not limited to, Wang resin, HMPA-PEGA resin, Rink acid resin, or a hydroxyethyl-photolinker resin.
  • the C-terminus of the peptide chain is covalently linked to a polymeric resin and protected ⁇ -amino acids were added in a stepwise manner with a coupling reagent.
  • a preferred ⁇ -amino protecting group is the Fmoc group, which is stable to coupling conditions and can readily be removed under mild alkaline conditions.
  • the reaction solvents are preferably, but not limited to, DMF, NMP, DCM, MeOH, and EtOH.
  • Examples of coupling agents are: DCC, DIC, HATU, HBTU.
  • Cleavage of the N-terminal protecting group is accomplished in 10 - 100% piperidine in DMF at 0 - 40°C, with ambient temperature being preferred.
  • the final Fmoc protecting group is removed using the above N-terminal cleavage procedure.
  • the remaining peptide on resin is cleaved from the resin along with any acid sensitive side chain protecting groups by treating the resin under acidic conditions.
  • an acidic cleavage condition is a mixture of trifluroacetic acid (TFA) in dichloromethane. If the hydroxyethyl-photolinker resin is used, the appropriate wavelength for inducing cleavage is ⁇ 365 nm ultraviolet light.
  • TFA trifluroacetic acid
  • N-terminus derivatized peptides are conveniently accomplished on solid phase.
  • the terminal Fmoc is removed while the peptide is still on the solid support.
  • the N-cap of choice is coupled next using standard peptide coupling conditions onto the N-terminus of the peptide.
  • the peptide is cleaved from the resin using the procedure described above.
  • the Merrifield resin or PAM resin is useful.
  • the amino acids are coupled to the growing chain on solid phase by successive additions of coupling agent activated Boc-protected amino acids.
  • Examples of coupling agents are: DCC, DIC, HATU, HBTU.
  • the reaction solvents may be DMF, DCM, MeOH, and NMP.
  • Cleavage of the Boc protecting group is accomplished in 10 - 100% TFA in DCM at 0 - 40°C, with ambient temperature being preferred.
  • the N-terminus protecting group (usually Boc) is removed as described above.
  • the peptide is removed from the resin using liquid HF or trifluoromethane sulfonic acid in dichloromethane.
  • the prodrug peptide intermediate may be made via a solution phase synthesis, utilizing either Boc or Fmoc chemistry.
  • the C-terminal Leu tetrapeptide is generally used as an example, but it will be understood that similar reactions may be performed with other C-terminal tetrapeptides, as well.
  • the peptide can be built up by the stepwise assembly in analogy to the solid phase method (in the N-terminal direction or in the C-terminal direction) or through the coupling of two suitably protected dipeptides or a tripeptide with a single amino acid.
  • One method of solution phase synthesis is a stepwise building up of the prodrug peptide intermediate using Fmoc chemistry, shown in Fig. 4.
  • the C-terminus must be protected to reduce the formation of side products.
  • the C-terminal R group in Fig. 4 is Me, tBu, benzyl or TCE. (Note when the N-cap is methyl succinyl the C-terminus R group cannot be Methyl.)
  • DMF is given as the solvent, other solvents such as DMSO, CH 3 CN, or NMP (or mixtures thereof) may be substituted therefor.
  • Pyridine, Et 3 N or other bases may be substituted for piperidine in deprotecting the growing peptide chain protected amino terminus.
  • HBTU is given in the diagram above as the activating agent
  • other activating agents such as DCC, DIC, DCC + HOBt, OSu, activated esters, azide, or triphenyl phosphoryl azide may be used.
  • the protected peptide acid chloride or acid bromide may be used to couple directly to the amino acid or peptide fragment.
  • the N- terminus deprotected and the C-terminus protected peptide is ready to accept the desired N-cap.
  • the N- cap When constructing the N-capped oligopeptide by solution phase synthesis, the N- cap needs to be synthesized by a slightly modified procedure (Fig. 4). First the C- terminus of the Fmoc oligopeptide needs to be protected with an acid labile or hydrogenation sensitive protecting group compatible with the selective deprotection of the C-terminus over the N-cap. Then the Fmoc protecting group needs to be removed from the oligopeptide to reveal the N-terminus. With the N-terminus deprotected and the C-terminus protected, the oligopeptide is reacted with the activated hemiester of the desired N-cap.
  • the N-cap can be activated using methods for activating amino acids such as DCC or HATU in base and an appropriate solvent.
  • the coupling may also be done via methyl hemisuccinyl chloride (or other acid halide) (Fig. 4) using an inert solvent in the presence of an organic or inorganic base, such as DEEA, triethylamine or Cs 2 CO 3 .
  • an organic or inorganic base such as DEEA, triethylamine or Cs 2 CO 3 .
  • One example of such a synthesis can be by reacting methyl-hemisuccinate and ⁇ Ala-Leu- Ala-Leu benzyl ester.
  • the coupling method can be any one of the methods generally used in the art (see for example: Bodanszky, M., The Practice of Peptide Synthesis, Springer Verlag, 185 (1984); Bodanszky, M., Principles of Peptide Synthesis, Springer Verlag, 159 (1984).
  • the benzyl group then can be removed by catalytic hydrogenation providing the desired N-cap methyl-succinyl form of ⁇ Ala-Leu- Ala-Leu.
  • suitable, selectively removable C-terminal protecting groups can be, but are not limited to, tBu, alkoxy-methyl and TCE. Other methods of accomplishing this step are described in the literature.
  • the N-cap form of the oligopeptide-therapeutic agent described in this invention can be synthesized by coupling an Fmoc form (which means Fmoc is attached to the N- terminus of the oligopeptide) of the oligopeptide with daunorubicin, doxorubicin, or any appropriate therapeutic agent using any of the standard activating reagents used in peptide synthesis (Fig. 5).
  • the solvent may be toluene, ethyl acetate, DMF, DMSO,
  • the activating agent may be selected from one of the following: PyBOP, HBTU, HATU, EDC, DIC, DCC, DCC+HOBT, OSu activated esters, azide, or triphenylphosphorylazide.
  • HBTU or HATU is the preferred activating agent.
  • the acid chloride or the acid bromide of the protected peptide can also be used for this coupling reaction.
  • the base can be selected from inorganic bases such as CsCO 3) Na- or K 2 CO 3 , or organic bases, such as TEA, DIEA, DBU, DBN, DBO, pyridine, substituted pyridines, N-methyl-morpholine etc., preferably TEA, or DIEA.
  • the reaction can be carried out at temperatures between -15 °C and 50 °C, advantageously between -10 °C and 10 °C.
  • the reaction time is between 5-90 minutes and is advantageously 20-40 minutes.
  • the product is isolated by pouring the reaction mixture into water and filtering the precipitate formed.
  • the crude product can be further purified by recrystallization from DCM, THF, ethyl acetate, or acetonitrile, preferably from dichloromethane or acetonitrile.
  • the isolated Fmoc form of the oligopeptide therapeutic agent conjugate is then deprotected over 2-90 minutes, preferably 3-8 minutes, using a ten- to hundred- fold excess of base at a temperature between -10 °C and 50 °C. Ideally, 5-60 equivalents of the base are preferred.
  • Piperidine is the preferred base to deprotect Fmoc groups.
  • the deprotected amino terminus of the oligopeptide therapeutic agent conjugate is acylated by a diacid anhydride as an activated hemi-ester to give the final N-cap form of the oligopeptide-therapeutic agent.
  • the final prodrug can be similarly prepared from the protected N- cap form of the oligopeptide such as a methyl-hemi ester form of succinyl-N-cap oligopeptide and conjugated to a therapeutic agent. This method is illustrated in Fig. 6.
  • the protected N-Cap-oligopeptide therapeutic agent is now deprotected by methods compatible to the stability of the therapeutic agent. For example, anthracyclines may be protected with a methyl group and deprotected with an esterase. For other therapeutic agents, benzyl protecting groups and catalytic hydrogenation may be chosen to deprotect.
  • Conversion to the salt form of the negatively charged N-cap oligopeptide therapeutic agent is carried out with a solvent selected from the following group: alcohol (including methanol, ethanol, or isopropanol), water, acetonitrile, tetrahydrofuran, diglyme or other polar solvents.
  • the sodium source is one molar equivalent of NaHCO 3 , NaOH, Na 2 CO 3 , NaOAc, NaOCH 3 (in general sodium alkoxide), or NaH.
  • An ion exchange column charged with Na + (such as strong or weak ion exchangers) is also useful for this last step of making the salt form of the N-cap oligopeptide therapeutic agent when appropriate.
  • Sodium is described as an example only.
  • the prodrug may be converted to a pharmaceutically acceptable salt form to improve solubility of the prodrug.
  • the N-cap-oligopeptide therapeutic agent is neutralized with a pharmaceutically acceptable salt, e.g., NaHCO 3 , Na 2 CO 3 , NaOH tris(hydroxymethyl) aminomethane, KHCO 3 , K 2 CO 3 , CaCO 3 , NH 4 OH, CH 3 NH 2 ,
  • prodrug (CH 3 ) 2 NH, (CH 3 ) 3 N, acetyltriethylammonium.
  • the preferred salt form of prodrug is sodium and the preferred neutralizing salt is NaHCO 3 .
  • anthracycline type molecules including doxorubicin and daunorubicin form gels in organic solvents in very low concentrations (Matzanke, et al., Eur. J. Biochem. 207:747-55 (1992); Chaires, et al., Biochemistry 21 :3927-32 (1982); Hayakawa, et al., Chem. Pharm. Bull. 39:1282-6 (1991). This maybe a considerable obstacle to getting high yields of clean product when making peptide anthracycline conjugates. The gel formation contributes to the formation of undesirable side reactions.
  • Enzymes can promote the hydrolysis without destroying the substrate or the product. Enzymes suitable for this reaction can be esterases, or lipases and can be in their natural, water soluble forms or immobilized by cross coupling, or attachment to commercially available solid support materials. Of the soluble enzymes evaluated, Candida Antarctica "B" lipase (Amis Biologies) is especially useful. An example of an enzyme immobilized by cross coupling is ChiroCLEC-PCTM (Altus Biologies).
  • Candida Antarctica "B” lipase can be immobilized by reaction with NHS activated SepharoseTM 4 Fast Flow (American Pharmacia Biotech).
  • the pH of the reaction mixture during the hydrolysis is carefully controlled and maintained by a pH-stat between 5.5 and 7.5, advantageously between 5.7 and 6.5, via controlled addition of NaHCO solution.
  • the product is isolated by lyophilization of the filtered reaction mixture.
  • the immobilized enzymes remain on the filter cake and can be reused if desired.
  • the prodrug can also be prepared via coupling an allyl-hemiester or alkyl- hemiester form of the N-cap oligopeptide with a therapeutic agent and then liberating the free acid from the conjugate.
  • Fig. 8 illustrates this process with Succinyl- ⁇ -Ala-Leu- Ala-Leu and doxorubicin.
  • the coupling of allyl-succinyl- ⁇ -Ala-Leu- Ala-Leu with doxorubicin can be carried out via any one of the oligopeptide conjugation methods.
  • Allyl-succinyl- ⁇ Ala-Leu-Ala-Leu-doxorabicin can also be synthesized by reacting allyl hemisuccinate, which was prepared via known methods (Casimir, etal., Eet. Eett. 36:3409 (1995)), with ⁇ Ala-Leu-Ala-Leu-doxorubicin similarly as coupling of the protected tetrapeptide precursors to doxorubicin was described in the previous methods, shown in Fig. 5.
  • Suitable inert solvents are THF, dichloromethane, ethyl acetate, toluene, preferably THF from which the acid form of the product precipitates as the reaction progresses.
  • the isolated acid is converted to its sodium salt as described earlier. Reaction times vary between 10-180 minutes, advantageously 10-60 minutes, at temperatures between 0-60 °C, preferably 15-30 °C.
  • Removal of the allyl or alkyl group can be done with Pd (0), or Ni(0), advantageously Pd(0) promoted transfer of the allyl or alkyl group to acceptor molecules, as it is well known in the art and documented in the professional literature (Genet, et al., Eet. Eett. 50:497 (1994); Bricout, et al. Eet. Eett. 54:1073 (1998), Genet, et al. Synlett 680 (1993); Waldmann, et al, Bioorg. Med. Chem. 7:749 (1998); Shaphiro, et al., Eet. Eett. 35:5421 (1994)).
  • the amount of catalyst can be 0.5-25 % mol to the substrate.
  • the prodrug may also be synthesized via the method shown in Fig. 7.
  • This approach utilizes an R'-oligopeptide, where R' is trityl or substituted trityl.
  • the coupling of R' -oligopeptide with a therapeutic agent can be carried out via any one of the methods described earlier for conjugation of a protected oligopeptide with a therapeutic agent at 30-120 minutes at 0-20°C.
  • trityl or substituted trityl group can be achieved under acidic conditions to give the positively charged prodrug.
  • This positively charged prodrug is N- capped as illustrated in Fig. 4 and described earlier.
  • the trityl deprotection can be accomplished with acetic acid, formic acid and dilute hydrochloric acid.
  • the prodrug can be converted into succinyl or glutaryl ⁇ Ala-Leu- Ala-Leu therapeutic agent by reacting with succinic anhydride.
  • Succinyl or glutaryl ⁇ Ala-Leu- Ala-Leu therapeutic agent can be converted to any pharmaceutically acceptable salt.
  • the solvent for coupling step DMF, DMSO, CH 3 CN, NMP, or any other suitable solvent is known in the art.
  • the prodrug compound of the present invention can be synthesized by using solid phase chemistry via "step wise” inverse (from the N-terminal to the C-terminal) direction methods.
  • One way is to use resins to immobilize a succinyl-hemi ester, for example succinyl-mono-benzyl ester or -allyl ester.
  • resins could be selected are “Wang Resins” (Wang, J. Am. Chem. Soc. 95:1328 (1973); Zhang, et al. Eet. Eett. 37:5457(1996)), “Rink Resins” (Rink, Eet. Eett. 28:3787 (1987)), "Trityl-, or substituted- trityl Resins” (Chen, et.al., J Am. Chem. Soc.
  • the molecule is then liberated from the resin by using mildly acidic conditions to form a free prodrug, such as free Suc- ⁇ Ala-Leu- Ala- Leu-Dox.
  • a free prodrug such as free Suc- ⁇ Ala-Leu- Ala- Leu-Dox.
  • This methodology is represented on the scheme of Fig. 9.
  • Another version of phase synthesis utilizes immobilized succinyl oligopeptide. This is then C-terminally deprotected, followed by the coupling step to doxorubicin or other therapeutic agent, and finally liberated from the resin as represented on the scheme of Fig. 9.
  • the acid form of the prodrug molecule may then be converted finally into its sodium salt as described above.
  • the prodrug compound can be synthesized using a simple and efficient three-step process of the invention.
  • the first step involves the coupling of an alkyl-ester protected oligopeptide fragment to a therapeutic agent.
  • a preferred embodiment of the first step involves the coupling of MeOSuc- ⁇ Ala-Leu- Ala-Leu-OH with doxorubicin (FIG. 16) using HATU as a coupling agent to give MeOSuc- ⁇ Ala-Leu- Ala-Leu-Dox.
  • the focus of this step is on the purity and the yield of the methyl ester, since it was found that the hydrolysis step did not have an impact on purity.
  • the second step is the hydrolysis of the alkyl-ester group by an enzyme (esterase), which directly gives the prodrug compound in good yield with a final purity of at least 90%.
  • the second step may be the hydrolysis of the methyl ester group in MeOSuc- ⁇ Ala-Leu- Ala-Leu-Dox by an enzyme (CLEC CAB, crosslinked Candida Antartica "B” Lipase), which directly gives the sodium salt of Suc- ⁇ Ala-Leu- Ala-Leu-Dox in quantitative yields with high purity.
  • the final step is to isolate the product after the hydrolysis step. Since most therapeutic agents are toxic substances, it is preferable to add an extra step to eliminate any free therapeutic agent from the coupled product.
  • the focus of final step is to isolate the final product. For example, Sue- ⁇ Ala-Leu- Ala-Leu-Dox, after the hydrolysis step. This is simply achieved by filtering the reaction mixture from the hydrolysis step using 0.2 micron filter and then lyophilizing the filtrate to yield Sue- ⁇ Ala-Leu- Ala-Leu- Dox.Na.
  • Unconjugated therapeutic agent may be present late in the process of making the prodrug.
  • Unconjugated therapeutic agent may be present late in the process of making the prodrug.
  • the reaction did not proceed completely. There was about 2-4% of residual doxorubicin remaining in the coupled product.
  • Initial attempts to remove doxorubicin completely from the product by acidic washes did not result in complete removal.
  • the complete removal of doxorubicin is crucial. The complete removal of the free therapeutic agent was effected by the process outlined in Example 41 and Fig.
  • the invention includes a method of making a compound comprising:
  • AA 4 represents a non-genetically-encoded amino acid
  • AA 3 represents any amino acid
  • AA 2 represents any amino acid
  • AA 1 represents any amino acid
  • a method of making a compound comprises the following steps:
  • AA 4 represents a non-genetically-encoded amino acid
  • AA 3 represents any amino acid
  • AA 2 represents any amino acid
  • AA 1 represents any amino acid
  • a compound of the invention may also be made via the following steps:
  • AA 3 represents any amino acid
  • AA 2 represents any amino acid
  • AA 1 represents any amino acid
  • trityl-protected oligopeptide of the formula trityl-(AA) n -AA 4 -AA 3 -AA 2 -AA 1 , wherein: each AA independently represents an amino acid, n is an integer from 0 to 16,
  • AA 4 represents a non-genetically-encoded amino acid, AA represents any amino acid, AA represents any amino acid, and AA 1 represents any amino acid, (2) coupling the trityl-protected oligopeptide to a therapeutic agent by activating the trityl-protected oligopeptide with an activating agent in the presence of a therapeutic agent, thereby making a trityl-protected oligopeptide-therapeutic agent conjugate,
  • Another possible step in connection with any of these methods is removing uncoupled therapeutic agent by use of scavenging resin or beads. Further, the compound may be neutralized with a pharmaceutically acceptable salt if desired.
  • the invention includes a method of removing free therapeutic agent comprising:
  • step (1) coupling an optionally protected stabilizing group-oligopeptide conjugate with the free therapeutic agent, (2) contacting the reactants of step (1) with a polymeric resin to bind free therapeutic agent remaining after step (1) and to form a therapeutic agent-polymeric resin complex, and
  • the polymeric resin may be polystyrene methylisocyanate or polystyrene sulfonyl chloride.
  • Compounds of the invention include the prodrugs, Suc- ⁇ Ala-Leu- Ala-Leu-Dox, Suc- ⁇ Ala-Leu- Ala-Leu-Dnr, and Gl- ⁇ Ala-Leu- Ala-Leu-Dox.
  • Methyl-succinyl- ⁇ Ala-Leu-Ala-Leu-OBn Methyl-succinyl- ⁇ Ala-Leu-Ala-Leu
  • DMEM:F12 (1:1) 50 mg/L bovine serum albumin, ITS-X (10 mg/L insulin, 5.5 mg/L transferrin, 6.7 ⁇ g/L Na selenite, 2 mg/L ethanolamine), and Lipid Concentrate (Gibco #21900-030).
  • 100 mL of cells were harvested by centrifugation at 4°C 10,000xg, for 20 min and decanting the supernatant. The pellet was resuspended in 2 mL phosphate buffered saline (Gibco) and centrifuged at 18, OOOxg for 10 min.
  • the cells After decanting the supernatant, the cells (approximately 300 ⁇ L wet) were homogenized by grinding in 1.7 mL 10 mM pH 7.2 HEPES buffer (sodium salt). The homogenate was centrifuged at 18,000xg at 4°C for 5 min and the supernatant was aliquoted and stored at ⁇ -20°C for subsequent use in the compound screen.
  • MCF 7/6 cells were grown to confluence in DMEM/F12 (1:1) medium containing 10 % fetal bovine serum, 0.05% (w/v) L-glutamine, 250 IU/mL penicillin, and 100 ⁇ g/mL streptomycin. Cells were then washed twice with phosphate buffered saline and incubated 24 hr at 5% CO 2 , 37°C, in DMEM/F12 (1:1), 0.02% BSA, ITS-X (10 mg/L insulin, 5.5 mg/L transferrin, 6.7 ⁇ g/L Na selenite, 2 mg/L ethanolamine).
  • the conditioned media was then decanted and, using a stirred cell apparatus with a YM10 (10,000 MW cutoff) ultrafiltration membrane(Millipore), exchanged once with 10 mM HEPES buffer, pH 7.2 and concentrated twenty-fold. This solution was stored in aliquots at -20°C for use in the compound screen.
  • YM10 10,000 MW cutoff ultrafiltration membrane(Millipore)
  • the lysis solution contained 0.02% w/v Triton X-100, 0.04% w/v sodium azide, and a cocktail of protease inhibitors (2 tablets/ 50 mL CompleteTM, EDTA-free tablets, Roche Molecular Biochemicals).
  • the cell homogenate was centrifuged 30 minutes at 4°C at 5000xg and the pellet was homogenized in a second 108 mL of lysis solution using a Dounce homogenizer and centrifuged as before.
  • the column was then washed with 250 ml of the equilibration buffer at a flow rate of 1 mL/minute. Proteins were eluted in a NaCl linear concentration gradient (0-0.5 M in the equilibration buffer, total volume of the gradient was 1000 ml) at a flow rate of 3 ml/minute. Two-minute fractions were collected and used for enzyme activity determination using ⁇ Ala-Leu- Ala-Leu-Dox as the substrate. Its transformation into Ala-Leu-Dox was quantified by reverse phase high performance liquid chromatography utilizing fluorescence detection of the anthracycline moiety.
  • fractions containing the highest activity levels were pooled (fractions #43-46; ⁇ 0.13 M NaCl), supplemented with protease inhibitors (CompleteTM, EDTA-free tablets, Roche Molecular Biochemicals), and stored as aliquots at -80°C.
  • HeLa cell fraction 1 was prepared from 50 billion HeLa cells as described in the Example 3 except that 6 runs with a load of about 350 mg of proteins each were performed and 50 ⁇ M CoCl 2 was added to the equilibration and elution buffers.
  • the FI fraction was concentrated by ultrafiltration (30 KD MWCO) and incubated in the presence of 1.25% EDTA for 2 hours at 4°C.
  • EDTA was removed on a desalting column (PD10) equilibrated and eluted with equilibration buffer (20 mM phosphate, 0.01% Triton-XlOO, 0.02% NaN 3 , 0.5 M NaCl, pH 7.2). About twenty mg of protein corresponding to the FI fraction were then loaded onto a 12 x 150 mm Chelating-
  • Sepharose (Amersham Pharmacia Biotech) column previously treated successively with 250 mL 5% EDTA, 250 ml water, 250 mL 0.1 M CoCl 2 , 250 mL water and 250 mL of the equilibration buffer. After sample adsorption, the column was washed with 150 mL of the equilibration buffer and eluted with a 600 mL 0-0.2 M imidazole gradient. All steps were carried out with a flow rate of 0.1 ml min. Forty-minute fractions were collected.
  • the activity-containing fractions ( ⁇ 1 mg of protein) were pooled, concentrated by ultrafiltration and diluted (1:1) with electrophoresis sample buffer (0.12 M Tris-HCl, 5% glycerol, 0.01%) bromophenol blue, pH 6.8).
  • electrophoresis sample buffer (0.12 M Tris-HCl, 5% glycerol, 0.01%) bromophenol blue, pH 6.8.
  • This sample was fractionated by preparative native polyacrylamide gel electrophoresis.
  • a Model 491 PrepCell BioRad was used with a 37 x 120 mm, 7% T, 2.6% C resolving gel buffered with 0.37 M Tris-HCl, pH 8.8, and a 37 x 5 mm, 4% T, 2.6% C concentrating gel buffered with 0.12 M Tris-HCl, pH 6.8.
  • the electrode buffer was 25 mM Tris, 192 mM glycine, pH 8.3, and the elution buffer 100 mM triethanolamine, 0.01% Triton X-l 00, 50 ⁇ M CoCl 2 , pH 7.2. After 30 minutes at 30 mA, separation was performed for about 24 hours at 40 mA. Twelve-minute fractions were collected with an elution flow rate of 0.4 ml/min.
  • Activity containing fractions (-150 ⁇ g of protein) were pooled, concentrated by ultrafiltration and the sample was applied to a gel filtration HPLC column (TosoHaas TSK G3000SW X , 7.8 x 600 mm) equilibrated and eluted at 0.3 mL/min with 50 mM, pH 7.0 phosphate buffer containing 0.2M K 2 SO 4 . Fractions of 0.5 min were collected. The activity-containing fractions were stored at -80°C.
  • prodrug to free toxin occurs via a series of enzyme catalyzed cleavage reactions.
  • the prodrug, Suc- ⁇ Ala-Leu- Ala-Leu-Dox is converted to Leu-Dox in extracts of carcinoma cells or carcinoma cell conditioned media in two steps catalyzed by at least two enzymes.
  • Initial endopeptidase cleavage occurs between the AA 3 (PI) and AA 2 (PI') amino acids to yield Ala-Leu-Dox.
  • exopeptidase removes alanine to give leucyl-doxorubicin which is known to be taken up into cells where the active toxin, doxorubicin, is released.
  • leucyl-doxorubicin which is known to be taken up into cells where the active toxin, doxorubicin, is released.
  • a good candidate for a prodrug with improved therapeutic index is activated by cancer cells but relatively stable in whole human blood.
  • Three different preparations of carcinoma were used to screen various N-capped peptidyl-toxins. These three preparations were as follows:
  • HeLa (cervical carcinoma) cell extract anion exchange fraction pool Compounds which could be hydrolyzed to a single amino acid toxin conjugate (i.e., AA 1 -(optional linker)-therapeutic agent) were further tested for stability in whole human blood.
  • the whole blood was collected using commercial acid buffered citrate whole blood collection tubes (Becton Dickinson). Test compounds were incubated for 2 hr at 37°C at a concentration of 12.5 ⁇ g/mL with the three different preparations of carcinoma enzyme and with whole blood. Following incubation, three volumes of acetonitrile were added to stop the reaction and remove protein from the mixture.
  • the sample was centrifuged at 18,000g for 5 minutes and 100 ⁇ L of supernatant was mixed with 300 ⁇ L of water prior to analysis by HPLC.
  • 50 ⁇ L of sample was injected on a 4.6 x 50 mm 2 ⁇ TSK
  • the oligopeptide having SEQ 3D NO: 1 was tested with Aminomethylbenzoic acid as a stabilizing group and Daunorubicm as the therapeutic agent.
  • the oligopeptide having SEQ ID NO: 35 was also tested with Diglycohc acid and Malic acid as stabilizing groups and Daunorubicin as the therapeutic agent.
  • the oligopeptide having SEQ ID NO: 38 was also tested with a number of additional stabilizing groups and therapeutic agents.
  • test compounds of the oligopeptide of SEQ ID NO: 38 included 1- Admantenecarbonyl- ⁇ Ala-Leu-Ala-Leu-Dnr, Diphenyl-Acetyl- ⁇ Ala-Leu-Ala-Leu-Dnr, Maleic- ⁇ Ala-Leu-Ala-Leu-Dox, 4-Morpholinecarbonyl- ⁇ Ala-Leu-Ala-Leu-Dnr, PEG- ⁇ Ala-Leu-Ala-Leu-Dox, 2-Furoyl- ⁇ Ala-Leu-Ala-Leu-Dnr, Acetyl- ⁇ Ala-Leu- Ala-Leu- Dnr, Diglycolic- ⁇ Ala-Leu-Ala-Leu-Dox, and Napth- ⁇ Ala-Leu-Ala-Leu-Dox.
  • results for carcinoma enzyme cleavage were the same for a partially purified fraction from HeLa cells, MFC 7/6 cell homogenate, or MCF 7/6 conditioned media.
  • Rates of Hydrolysis For comparison of rates of hydrolysis for different prodrugs or measurement of trouase and TOP activity after immunoprecipitation, enzyme test solution (as prepared in Examples 1-4 above) was incubated with 10 ⁇ g/mL substrate in pH 7.2 10 mM HEPES with 100 ⁇ M MnCl 2 at 37°C for up to 2 hr. The reaction was stopped by adding three volumes of acetonitrile. Precipitated protein was removed by centrifugation and the supernatant was diluted into three volumes of water before HPLC analysis, as described in Example 5 above. The fraction of substrate hydrolyzed was calculated by dividing peak areas for products by total peak area for substrate and products.
  • Substrate specificity of partially purified HeLa cell trouase was essentially identical to that of recombinant rat TOP (rRTOP) produced in E coli according to the method of Glucksman and Roberts. (Glucksman and Roberts, "Strategies for characterizing, cloning, and expressing soluble endopeptidases," Methods in Neurosciences, 23: 296-316 (1995))
  • rRTOP recombinant rat TOP
  • carcinoma cell trouase and TOP have nearly identical substrate specificity.
  • TOP is inactivated by exposure to metal chelating agents such as EDTA and 1,10 phenanthroline (Barrett, et al., "Thimet ohgopeptidase and ohgopeptidase M or neurolysin," Methods Enzymol 248: 529-556 (1995)).
  • metal chelating agents such as EDTA and 1,10 phenanthroline
  • the peptide compound N-fl-tRSJ-carboxypropyl-Ala-Ala-Phe- ⁇ - aminobenzoate (Cpp-AAF- ⁇ AB)] is a more selective and sensitive inhibitor (Knight and Barrett, "Structure/function relationships in the inhibition of thimet ohgopeptidase by carboxyphenylpropyl-peptides," EEES Eett 294: 183-186 (1991)).
  • Cpp-AAF-/ ⁇ AB also inhibits the closely related metallopeptidase neurolysin, only neurolysin activity is inhibited by 5 mM of the dipeptide Pro-He (Serizawa, et al., "Characterization of a mitochondrial metallopeptidase reveals neurolysin as a homologue of thimet ohgopeptidase," JBiol Chem 270:2092-2098 (1995)).
  • trouase activity was inhibited 9-fold by 1 mM ⁇ DTA and 2 mM 1,10 phenanthroline while inhibitors of non-metallopeptidases were not effective.
  • activity was not inhibited by 50 ⁇ M aminoethylbenzenesulfonylfluoride, 4 ⁇ g/mL aprotinin (both inhibit serine peptidase), 20 ⁇ M ⁇ -64 (inhibits cysteine peptidase), 1.5 ⁇ M pepstatin (inhibits aspartate peptidase), 20 ⁇ M leupeptin (inhibits serine and cysteine peptidase), or 1 ⁇ M CA-074 (inhibits cathepsin B).
  • aprotinin both inhibit serine peptidase
  • 20 ⁇ M ⁇ -64 inhibits cysteine peptidase
  • 1.5 ⁇ M pepstatin inhibits aspartate peptidase
  • 20 ⁇ M leupeptin inhibits serine and cysteine peptidase
  • CA-074 inhibits cathepsin B
  • Barrett and Brown (“Chicken liver Pz-peptidase, a thiol-dependent metalloendopeptidase," Biochem J, 271 :701-706 (1990)) used purified chicken TOP to measure reactivation after dialysis against EDTA. At 50 ⁇ M concentration, Zn 2+ completely restored activity. Other divalent cations at the same concentration partially restored activity in the following order of effectiveness: Mn >Ca >Co >Cd . Other divalent cations such as Cu 2+ had no effect. An excess of Zn 2+ (> 100 ⁇ M) was inhibitory. In reconstitution experiments with EDTA treated MCF-7/6 cell homogenate, activity was completely restored with 50 ⁇ M Co 2+ or Mn 2+ , but not Zn 2+ or Cu 2+ .
  • MCF-7/6 cell homogenate trouase had an approximate molecular weight of 68 KD based on the retention volume of active gel filtration chromatographic fractions.
  • the MCF-7/6 cell homogenate and protein molecular weight standards were fractionated on a Superose S12, 10 x 300 column (Amersham-Pharmacia Biotech).
  • Purified trouase from HeLa cells was separated by SDS polyacrylamide gel electrophoresis (PAGE) into two protein bands corresponding to 74 and 63KD using methods described in example 11. Bands of about 74 and 63KD were also observed in SDS PAGE western immunoblots of HeLa cell FI stained with anti-thimet ohgopeptidase antibody.
  • the 74KD band was also observed in SDS PAGE western blots of crude homogenates of MCF-7/6, MDS-MD-231 , and EA hy926 cells. These results agree with the molecular weight of human, rat and porcine TOP which has been deduced from the DNA sequence as 78KD and reported between 74-80KD in various SDS PAGE determinations (Barrett, et al., "Thimet ohgopeptidase and ohgopeptidase M or neurolysin," Methods Enzymol 248: 529-556 (1995)).
  • MCF-7/6 conditioned medium was pre-incubated for 30 minutes at room temperature with dithiothreitol (DTT) at indicated concentrations. Then 10 ⁇ g/mL Suc- ⁇ Ala-Leu-Ala-Leu-Dox was added and incubated at 37°C. Hydrolysis products were extracted and analyzed on a Luna C18-3 ⁇ , 4.6 x 100 mm column (Phenomenex) as above. Residual Suc- ⁇ Ala-Leu- Ala-Leu-Dox substrate was extracted using pH 3.0 citrate buffer rather than borate and N-succinyl doxorubicin as internal standard and analyzed on the Luna Cl 8-3 ⁇ column as above.
  • DTT dithiothreitol
  • TOP is activated by low levels of thiol reducing agents such as 50 ⁇ M dithiothreitol (DTT) or 1 mM mercaptoethanol but inhibited at high concentrations such as 5 mM DTT (Orlowski, et al. "Endopeptidase 24.15 from rat testes.
  • DTT dithiothreitol
  • Oxypeptidase 24.15 from rat testes.
  • MFC-7/6 cell homogenate as prepared in Example 1 above, was incubated with ⁇ - Ala-Leu- Ala-Leu-Cou at 37°C in 100 mM triethanolamine buffered at various pH levels.
  • the amount of Leu-Cou was determined by treatment of the reaction product with leucine aminopeptidase and measurement of the resulting aminomethylcoumarin concentration in a spectrofluorometer.
  • TOP has a pH optimum of 7.8 for 10 minute assays with quenched fluorescent substrates (Barrett, 1995).
  • Assay with the quenched fluorescent substrate Mcc-Pro-Leu-Gly-Pro-D-Lys(DNP) was as described in Barrett, et al.
  • the trypsin digested protein from the 74KD band was analyzed by MALDI-tof (Matrix assisted laser desorbtion ionization - time of flight) mass spectrometry. Mass spectra of the tryptic digests were acquired on a Biflex (Bruker) MALDI-tof mass spectrometer equipped with delayed extraction operated in the reflector mode.
  • each digest in 25 mM ammonium bicarboante was deposited directly on the sample probe in a dry thin layer ⁇ -cyano-4-hydroxy-cinnamic acid (CCA) matrix mixed with nitrocellulose (4:3 v:v saturated CCA : 5 ng/mL nitrocellulose in 1:1 isopropanol : acetone).
  • CCA ⁇ -cyano-4-hydroxy-cinnamic acid
  • nitrocellulose 4:3 v:v saturated CCA : 5 ng/mL nitrocellulose in 1:1 isopropanol : acetone.
  • the samples were washed with 0.1% TFA before analysis.
  • the peptide mass fingerprint obtained for each digest was matched to predicted digest patterns from known protein sequences using MS-FIT (http://prospector.ucsf.edu/ucsfhtml3.2/msfit.htm).
  • Electrospray ionization (ESI) quadrupole time of flight (Q-tof) tandem mass spectrometry was performed using a Q-tof instrument (Micromass) with a Z-Spray ion source working in the nanospray mode. About 3-5 ⁇ L of purified sample was introduced into a sample needle (PROTANA Inc., Odense, DK) to run MS and MS/MS experiments. The average capillary potential was 1000 V and the sample cone was set to 50 V. Human B was used to calibrate the instrument in the MS/MS mode. MS/MS spectra were transformed using MaxEnt3 and sequences were determined using PepSeq (Micromass BioLynx).
  • HEPES mM hydroxyethylpiperazine
  • Promyelocytic leukemia cells HL-60 were cultured in RPMI media containing 10% heat inactivated fetal calf serum (FCS). On the day of the study, the cells were collected, washed and resuspended at a concentration of 0.5 x 10 cells/ml in RPMI containing 10% FCS. 100 ⁇ l/well of cell suspension was added to 96 well plates. Serial dilutions (3-fold increments) of doxorubicin or test compounds were made and 100 ⁇ l of compounds were added per well. Finally, 10 ⁇ l of a 100 ⁇ Ci/ml 3 H-thymidine was added per well and the plates were incubated for 24 hours.
  • FCS heat inactivated fetal calf serum
  • Doxorubicin exhibits potent cytotoxic activity with an IC 50 of 0.075 ⁇ M in HL60 cells.
  • the prodrug Sue- ⁇ Ala-Leu-Ala-Leu-Dox has poor cellular uptake and an IC 50 greater than 50 ⁇ M in the HL-60 proliferation assay.
  • leucyl-doxorubicin was the intermediate formed after proteolytic cleavage of the prodrug Suc- ⁇ Ala-Leu- Ala-Leu-Dox. Therefore, leucyl-doxorubicin was tested and shown to have an IC 50 of 0.222 ⁇ M.
  • Two groups of ICR normal female mice were administered a single IN bolus dose with approximately 100 ⁇ mol/Kg of Sue- ⁇ Ala-Leu-Ala-Leu-Dox or 10 ⁇ mol/Kg of doxorubicin (Dox).
  • Plasma was obtained from three individual animals in each group at 5 minutes, 1, 2, 4, or 6 hr.
  • ⁇ -aminoacyl- doxorubicin L-Dox
  • L-Dox was the major metabolite over the first two hr while the dipeptidyl-conjugate AL-Dox was a more minor product that formed at about the same time as L-Dox.
  • Doxorubicin appeared later with the plasma concentration decreasing more slowly over time than the other metabolites as expected from the current and previously measured doxorubicin pharmacokinetic profiles (Nan der Nijgh, et al.
  • Suc- ⁇ Ala-Leu- Ala-Leu-Dox was dosed at approximatley twice its single dose (SD) and repeat dose (RD) MTD while doxorubicin was dosed at approximately 25% of its SD MTD, but at a dose which is equivalent to the RD MTD.
  • SD single dose
  • RD repeat dose
  • the rapid clearance of non-cleaved Suc- ⁇ Ala-Leu- Ala-Leu-Dox from plasma apparently results in higher relative tolerability of the Suc- ⁇ Ala-Leu- Ala-Leu-Dox compound compared with doxorubicin.
  • prodrug is advantageous in repeat-dose treatments, such as those used in chemotherapy, as it allows doseing to effecting levels of doxorubicin exposure while safely clearing excess drug, shown by the relative difference in MTD.
  • Suc- ⁇ Ala-Leu- Ala-Leu-Dox has proven to be efficacious in inhibiting the growth of human tumors in several nude mouse xenograft models, including the estrogen- dependent MCF-7/6 mammary tumor and the adriamycin-resistant colorectal carcinomas CXF280/10 and LS-174T.
  • the drug was safe and well-tolerated under repeat-dose levels and frequencies of dosing that demonstrated anti-tumor efficacy. Some dose-dependent body weight loss was observed, hi supporting studies, kidney toxicity and myelosuppression were not observed at doses of up to 106.8 mg/kg of Suc- ⁇ Ala-Leu- Ala-Leu-Dox.
  • Sue- ⁇ Ala-Leu-Ala-Leu-Dox an exemplary tetrapeptide prodrug of the invention, is well tolerated in mice.
  • SD-MTD Maximum Tolerated Dose
  • groups of five normal ICR mice were administered intravenous bolus doses of Suc- ⁇ Ala-Leu-Ala-Leu-Dox. The mice were observed daily for 49 days and body weights measured twice weekly. Dose levels tested were 0, 50, 75 or 100 mg/kg, equivalent to 0, 28, 42 or 56 mg/kg of doxorubicin, respectively. There was no acute toxicity, within 24 hours, at any dose level. Dose and time dependent signs of toxicity were observed during the study.
  • Toxicity including partial hind-end paralysis and significant body weight loss (>20% of their initial weight) was observed in the 75 and 100 mg/kg dose groups.
  • mortality was observed in 40% of the 75 mg/kg dose group.
  • the SD-MTD for Suc- ⁇ Ala-Leu-Ala-Leu-Dox was determined to be 50 mg/kg (equivalent to 28 mg/kg of doxorubicin). This dose was very well tolerated and no adverse effects were observed. Therefore, the SD-MTD was approximately 1.8-fold higher on a molar basis than the SD-MTD for doxorubicin alone (16 mg/kg). See Table 7. This is an approximate SD- MTD determination based on a range of doses at 14 mg/kg doxorubicin equivalents increments over the range tested.
  • Suc- ⁇ Ala-Leu-Ala-Leu-Dox at well-tolerated doses of 49 (Group 3), 57 (Group 4), and 64 mg/kg (Group5), showed superior efficacy compared to doxorubicin at 3.0 mg/kg (Group 2) and saline (Group 1) in inhibiting the rapidly-growing adriamycin-resistant LS-174T tumor (Fig. 5), and extending survival of tumor-bearing mice (Fig. 12).
  • MX-1 tumors which are moderately doxorubicin-sensitive, human breast carcinoma xenografts, were implanted subcutaneously (s.c.) and mice were weighed and tumors were measured (by caliper) at least once a week prior to start of dosing (Day 0), then twice a week during the study. Immediately before the start of dosing (Study Day - 2 to Day 0), mice were randomized to various groups based on the weight of the tumors. Mice were euthanized after the tumors reached a cutoff weight of 1.5 g (cancer endpoint). Studies were terminated at Day 60. Table 8. MX-1 Study Summary
  • TGI % tumor growth inhibition over control at Day 18 MDS: Mean day of survival
  • TGD Tumor growth delay over control
  • the tumor exhibited relatively homogeneous growth characteristics, reaching the cancer endpoint ranging from Day 15 - 24.
  • Doxorubicin was effective in significantly prolonging the survival of tumor-bearing mice (Table 8).
  • a dose response was established for Sue- ⁇ Ala-Leu-Ala-Leu-Dox both in inhibiting MX- 1 tumor growth and prolonging mouse survival (Fig. 18 andTable 8).
  • Sue- ⁇ Ala-Leu- Ala-Leu-Dox at 71 mg/kg was significantly better than doxorubicin in inhibiting tumor growth and extending mouse survival (Table 8).
  • Prodrugs are Useful in Evasion of Multi-Drug Resistance Mechanisms.
  • Suc- ⁇ Ala-Leu-Ala-Leu-Dox has been shown to be much more active than free doxorubicin on MDR human cell lines implanted into mice in xenograft models.
  • Doxorubicin delivered to the tumor in a modified form, specifically in prodrug form as Suc- ⁇ Ala-Leu-Ala-Leu-Dox shows activity in slowing tumor growth resulting in significant extension of survival in the dose group.
  • Table 9 and Figure 15 show that with Suc- ⁇ Ala-Leu-Ala-Leu-Dox there is dose- dependent increase in survival in the MDR human colorectal carcinoma LS174-T.
  • 5 LS 174T is a very aggressive and rapidly growing tumor that exhibits heterogeneous cell morphology with a necrotic center. It is very resistant to conventional chemotherapeutics, and there are always tumors in some animals that become so well established within a few days that they rapidly outgrow attempts to inhibit tumor growth, thus the animals reach the tumor endpoint despite treatment.
  • Doxorubicin alone is 0 completely inactive in this model, producing no effects on tumor growth or on survival.
  • TGI % tumor growth inhibition over control at Day 18 MDS: Mean day of survival
  • TGD Tumor growth delay over control
  • Table 9 provides a summary of the effects of Suc- ⁇ Ala-Leu- Ala- Leu-Dox at three dose levels as compared to doxorubicin in the LS174T colorectal carcinoma xenograft in nude mice (Q7Dx5).
  • Parameters measured include the calculated Mean Day of Survival (MDS) determined by termination due to tumors reaching the predetermined cutoff size of 1500 mg (tumor death), number of Long Term Survivors (LTS), and tolerability of the dose regimen, by number of mice exhibiting toxic death (> 20% body weight loss).
  • MDS Mean Day of Survival
  • LTS Long Term Survivors
  • the number of LTS at day 60 was zero in all groups. No toxic deaths were observed in any of the groups.
  • Suc- ⁇ Ala-Leu-Ala-Leu-Dox at 57 mg/kg significantly inhibited LS174T tumor growth (Fig. 15 and Table 9). All three dose regimens of Suc- ⁇ Ala-Leu-Ala-Leu-Dox and doxorubicin were very well tolerated, with no termination due to toxic endpoints. In addition, Suc- ⁇ Ala-Leu-Ala-Leu-Dox, dosed at three well-tolerated dose- levels (Q7dx5), but not doxorubicin, showed a dose dependent inhibitory effect on median tumor weight (Figure 15).
  • Figure 15 shows effects of Suc- ⁇ Ala-Leu- Ala-Leu-Dox compared with doxorubicin on tumor growth of LS174T tumor colorectal carcinoma xenografts in nude mice and vehicle-control.
  • Group D was statistically significantly different from the vehicle control group at Day 19 (p ⁇ 0.05).
  • Table 10 shows that 81 % of the doxorubicin passed through the 3,000 MWCO filter while only 5 % of the conjugate, ⁇ Ala-Leu-Ala-Leu- Dox passed through the 3,000 MWCO filter.
  • the data also show that the 50,000 MWCO unit retains over 40% of the ⁇ Ala-Leu-Ala-Leu-Dox.
  • These data demonstrate that a significant percentage of ⁇ Ala-Leu-Ala-Leu-Dox aggregates were larger that 50 kD (>50 molecules/aggregate). Thus, ⁇ Ala-Leu-Ala-Leu-Dox may aggregate under some conditions.
  • Analytical HPLC analyses were performed on a Waters 2690 using a C-8 column (3.5 ⁇ m, 4.6 x 150mm ID, flow rate lmL/min) eluting with a gradient of solvent A (80% 20mM ammonium formate and 20% acetonitrile) and solvent B (20% 20mM ammonium formate and 80% acetonitrile) and the data was processed at ⁇ 254 nm using the Waters Millennium system.
  • Analytical HPLC gradient started with 100% of solvent A to 100% of solvent B over a period of 30 minutes (linear).
  • Preparative purification of crude products was achieved using a Waters Delta Prep 4000 system using a C-4 column (15 ⁇ m, 40 x 100mm ID, flow rate 30 mL/min) eluting with a gradient of solvent A (H 2 O), and solvent B (MeOH).
  • the preparatory HPLC gradient started with 80% of solvent A and goes to 100 % of solvent B over a period of 70 minutes (linear).
  • the data was processed at ⁇ 254 nm using the Waters Millennium System.
  • Analytical HPLC was accomplished on a Hewlett Packard instrument using a TSK superODS column (TosoHaas); solvent A (TFA 0.1% in water); solvent B (TFA 0.1%) in acetonitrile); gradient: 30 to 36% of B in 2 minutes, 36 to 41% of B in 10 minutes, 41 to 90% of B in 3 minutes, 5 minutes at 90% B, detection wavelength ⁇ 254 nm.
  • Doxorubicin and Daunorubicin were supplied by Meiji (Japan), Pd(PPh 3 ) 4 by Strem chem (Newburyport, MA), PEG by Shearwater( Huntsville, Alabama), solvents, HATU by Aldrich (Milwaukee, WI); all resins and amino acids were supplied by ABI (Foster City, CA), Novabiochem (San Diego, CA), Advanced ChemTech (Louisville, KY), Peptide International (Louisville, KY), or SynPep (Dublin, CA).
  • ABI Fester City, CA
  • Novabiochem San Diego, CA
  • Advanced ChemTech Konville, KY
  • Peptide International Louisville, KY
  • SynPep Dublin, CA
  • the Fmoc form of ⁇ Ala-Leu-Ala-Leu (24.34 g, 0.04 mol) was added into a round bottom flask with DMF (350 mL) and a magnetic stirrer. After the tetrapeptide was dissolved, benzyl bromide (4.76 mL, 0.04 mol), followed by cesium carbonate (13.04 g, 0.04 mol), was added to the solution with stirring. The reaction mixture was stirred at room temperature for 1.5 hrs. Then, the reaction mixture was slowly poured into a flask with 450 mL of iced water. A large amount of white solid precipitated out which was collected by suction filtration.
  • methyl hemisuccinate (3.19 g, 24.2 mmol) was dissolved in anhydrous DMF (50 mL).
  • DIEA (4.22 mL, 24.2 mmol) followed by HBTU (9.17 g, 24.2 mmol) were added into the solution.
  • the mixture was stirred at room temperature for 45 minutes.
  • a solution of ⁇ Ala-Leu-Ala-Leu benzyl ester (crude, containing 10.14 g, 21.3 mmol) in anhydrous DMF (150 mL). The mixture was continually stirred at room temperature for 2.5 hrs.
  • Methyl succinyl-N-cap form of ⁇ Ala-Leu-Ala-Leu benzyl ester (1.0 g, 86% purity; 1.46 mmol) was added into an Erlenmeyer flask with 100 mL of methanol. The solution was cloudy after being stirred for a few minutes. 50 mL of methanol was added, but the solution was still not clear. The solution was transferred into a hydrogenation reaction vessel. To this vessel, Pd-C (90 mg, 10% wet, 50% water; 0.042 mmol) was added. After hydrogenation for 2 hours at room temperature, the reaction was stopped and the catalyst was filtered. A white solid (0.77 g, 78%) was yielded after removal of solvents. MS m/z calcd. for C 2 H 4 oN 4 O 8 501.2, found 500.3.
  • This molecule was prepared according the procedure of Casimir, J. R., et.al. Tet Lett. 36(19):3409, (1995). 10.07g (0.1 mol) succinic anhydride and 5.808g (0.1 mol) allyl-alcohol were refluxed in 100 mL toluene for 6 hours. The reaction mixture was concentrated under reduced pressure. 15.5g; 98%. The resulting material was pure enough to use in subsequent reactions. The purity and identity of the semi-solid product was confirmed by 1HNMR and 13 CNMR, by LC/MS.
  • N-Cap-AUylhemisuccinyl form of ⁇ Ala-Leu-Ala- Leu (lg, 1.9 mmol) and doxorubicin (l.lg, 1.9 mmol) were dissolved in anhydrous DMF (50 ml). After the mixture was stirred for 5 minutes, DIEA (0.66 ml, 3.8 mmol) followed by HATU (0.76g,' 1.9 mmol) was added into the solution the mixture was stirred at room temperature for 2 hours. DMF was removed by a rotary evaporator and the residue was taken up in 4.0 ml 1 :1 DCM: MeOH.
  • Fmoc form of ⁇ Ala-Leu-Ala-Leu was synthesized using solid-phase approach with standard Fmoc chemistry.
  • a typical synthesis used Wang's alkoxy resin (0.60 mrnol/gm loading).
  • Fmoc-protected amino acids were used for solid-phase peptide synthesis.
  • 3 equivalents of amino acid were preactivated with HBTU as the activating agent for 5 minutes before being added to the resin together with 2 equivalents of DIEA.
  • the coupling reaction was carried out for 2 h and then washed with DMF (25 mL x 3) and DCM (25 L x 3).
  • Fmoc form of Thi-Tyr-Gly-Leu was synthesized using solid-phase approach with standard Fmoc chemistry and Wang's alkoxy resin (0.60 mmol/gm loading). Fmoc- protected amino acids and Fmoc-Thi-OH were used for solid-phase peptide synthesis.
  • 3 equivalent of amino acid was preactivated with HBTU as the activating agent for 5 minutes before being added to the resin together with 2 equivalent of DIEA.
  • the coupling reaction was carried out for 2 h and then washed with DMF (25 mL x 3) and DCM (25 mL x 3). The coupling reaction was repeated using 2 equivalent of amino acid using similar conditions.
  • the reaction progress was monitored using the ninhydrin test and if the ninhydrin test indicated incomplete reaction after 2 h then the coupling step was repeated for a third time. Deprotection was accomplished using 20% piperidine in DMF for 15-20 minutes. The coupling step was repeated with the next amino acid until the desired peptide was assembled on resin. The final cleavage of peptide from the resin was accomplished by treating the resin with a solution of 95%TFA and 5% water. After stirring the reaction mixture for 2h at RT, the resin was filtered under reduced pressure and washed twice with TFA. Filtrates were combined and adding 400 mL of cold ether precipitated the peptide.
  • the peptide was filtered under reduced pressure and dried to yield Fmoc form of Thi-Tyr-Gly-Leu (88% HPLC purity by method A). Crude Fmoc form of Thi-Tyr-Gly-Leu was used for the next step without any further purification.
  • Daunorubicin.HCl (185 mg, 0.329 mmol) and Fmoc form of ⁇ Ala-Leu-Ala-Leu (200 mg, 0.329mmol) were dissolved at room temperature in anhydrous DMF (15 mL).
  • DIEA (0.115 mL, 0.658 mmol) was added in one portion and the reaction mixture was stirred for 15 minutes at room temperature.
  • the reaction mixture was cooled to 0°C using an ice bath and 138 mg (0.362 mmol) of HATU was added slowly over 10 minutes. The reaction mixture was stirred for another 90 minutes at room temperature. Ice cold water (200 mL) was added to the reaction mixture which resulted in the formation of a red precipitate.
  • Daunorubicin.HCl (90 mg, 0.16 mmol) and Fmoc form of Thi-Tyr-Gly-Leu (120 mg, 0.16 mmol) were dissolved at room temperature in anhydrous DMF (15 mL).
  • DIEA (0.56 mL, 0.16 mmol) was added in one portion and the reaction mixture was stirred for 15 minutes at room temperature.
  • the reaction mixture was cooled to 0°C using an ice bath and 61 mg (0.16 mmol) of HATU was added slowly over 10 minutes. The reaction mixture was stirred for another 90 minutes at room temperature. Ice cold water (150 mL) was added to the reaction mixture which resulted in the formation of a red precipitate.
  • reaction mixture volume was then reduced to « 100 mL by rotary evaporation and then diluted with 125 mL chloroform.
  • 1400 mL diethyl ether was quickly added resulting in formation of a red precipitate.
  • This precipitate was isolated on a medium glass frit and triturated with 5 X 200 mL diethyl ether to yield material of 89.13% HPLC purity.
  • the precipitate was washed again with 1 x 20 mL diethyl ether and air dried to yield 3.62 g Suc- ⁇ Ala-Leu- Ala-Leu-Dox (81% physical yield, 88.2% HPLC purity).
  • the sodium salt was formed by addition of 0.025 M NaOH (4 mL, 0.10 mmol) dropwise to the suspension until complete dissolution of the solid. This solution was then lyophilized to give the sodium salt of Gl- ⁇ Ala-Leu- Ala-Leu-Dox in 97% yield with an HPLC purity of 87% by method D.
  • N-cap methyl hemisuccinyl form of ⁇ Ala-Leu- Ala-Leu (0.25 g, 0.5 mmol) and doxorubicin (0.29 g, 0.5 mmol) were dissolved in anhydrous DMF (20 mL). After the mixture was stirred for 5 minutes, DIEA (0.17 mL, 1.0 mmol) followed by HBTU (0.19 g, 0.5 mmol) was added into the solution. The mixture was stirred at room temperature for 4 hrs. DMF was removed by a rotary evaporator and the residue was taken up in 4.0 mL 1:1 methylenechloride:methanol.
  • MeOSuc- ⁇ -Ala-Leu- Ala-Leu-Dox MeOSuc- ⁇ -Ala-Leu-Ala-Leu-Dox (200 mg, 0.194 mmol), DIEA (0.068 mL,
  • the CLEC-PC cake was washed with 3 X 10 mL methanol and the methanol washes were combined with the filtered reaction mixture.
  • the filtered reaction mixture plus methanol washes were then concentrated to a red gum on a rotary evaporator equipped with a high vacuum pump and a 30° C water bath.
  • the red gum was then suspended in 50 mL deionized water at room temperature and rapidly stirred via mechanical stirrer. To this suspension a solution of 77.8 mg sodium bicarbonate
  • the precipitate was isolated by Celite 521 filtration, subsequent rinsing of the Celite pad with methanol, filtration of the methanol solution through a 10- 20 ⁇ M fritted glass filter and rotary evaporation of the filtered solution to yield 7.31 g of gummy red product.
  • This product was converted to the sodium salt by dissolution in 70 mL 76 mM NaHCO 3 (0.95 eq.) and lyophilized to yield 7.30 g, 66.1% physical yield sodium salt of succinyl-N-cap form of ⁇ Ala-Leu-Ala-Leu-Dox therapeutic agent, 84.5% pure by HPLC.
  • the flask was fitted with an overhead stirrer, and a pH-stat set to keep the pH of the reaction mixture between 5.9-6.2 by controlling a syringe pump.
  • the syringe pump was charged 0.1 M NaHCO 3 .
  • Progress of the reaction was followed by HPLC. After 6 days the immobilized enzym e was filtered off and the liquid phase was lyophilized. The dry solids were then suspended in about 11 mL dry THF and filtered off. 42.66g, 98.34% physical yield, 93.43%(254nm), 94.43%(480nm) pure by HPLC by method B.
  • PS-isocyanate beads 146.4 g PS-isocyanate beads (240 mmol; supplied by Argonaut Lab, San Carlos, CA) were dissolved in 1.5 L of anhydrous DMF and allowed to swell for 5-10 minutes at room temperature. The swelled beads were filtered through a glass-fritted funnel and washed with additional 500 mL of anhydrous DMF. 115g MeOSuc- ⁇ Ala-Leu- Ala-Leu- Dox (112 mmol) was dissolved in 1000 mL of anhydrous DMF and 2.1 mL DIEA (12 mmol) was added followed by the swelled PS-isocyanate beads.
  • the reaction mixture was stirred at room temperature and was monitored using HPLC till the amount of doxorubicin peak was less than 0.1%. It takes anywhere from 2-12 h depending upon the size of the batch.
  • the reaction mixture was filtered through a coarse sintered glass funnel to remove the beads.
  • a brine solution (at least 30% w/v) of 1.1 kg NaCl in 3.5 L water was prepared and cooled to 0° C. The filtered reaction mixture was then slowly added to the cooled brine solution with vigorous stirring over 45 minutes. The color of the solution remained red, a blue solution would have indicated that the pH needed adjustment immediately to between 5.8-6.0 by adding acetic acid.
  • the red precipitate was filtered through a medium sintered glass funnel, washed with water and dried under vacuum pressure over P 2 O 5 to yield MeOSuc- ⁇ Ala-Leu- Ala-Leu-Dox free of any residual doxorubicin.
  • MeOSuc- ⁇ Ala-Leu-Ala-Leu-Dox was dissolved in 1 L MeOH and the methanol solution was then slowly added to 14L of cooled ethyl ether with vigorous stirring over 60 minutes. The red precipitate was filtered through a medium sintered glass funnel, washed with ether (1 L) and dried under vacuum pressure to yield 110 g MeOSuc- ⁇ Ala- Leu-Ala-Leu-Dox. The purity was determined to be 96.5% by HPLC, as described in Example 44. MS m/z calcd. for C 50 H 67 N 5 O ⁇ 8 1025, found 1048 (M + +Na).
  • Example 51 Enzymatic hydrolysis of MeOSuc- ⁇ Ala-Leu- Ala-Leu-Dox to yield Suc- ⁇ Ala-Leu- Ala-Leu-Dox
  • the CLEC-CAB (Candida Antartica "B” Lipase) enzyme was purchased (from Altus Biologies., Boston, MA) in solution form, where the concentration of the enzyme is defined by the weight of dry enzyme per milliliter of solution.
  • the crude enzyme suspension was shaken for few minutes to obtain a homogenous solution. 504 mL (328 mmol) of this homogenous solution was aliquoted into a flask. 2.5 L of deionized water was added and the slurry was stirred for 10 minutes using a magnetic stirrer.
  • the enzyme solution was filtered using a coarse glass fritted funnel, without taking the enzyme to dryness. The enzyme was transferred back into a flask. The enzyme is suspended in water and filtered three more times.
  • the enzyme cake was resuspended into 550 mL of deionized water and transferred into aRB flask.
  • 109g MeOSuc- ⁇ Ala-Leu- Ala-Leu-Dox (106 mmol) was added and the reaction mixture was stirred at room temperature (25°C).
  • the pH of the reaction mixture was maintained between 5.8 and 6.1 by a pH-stat equipped with a syringe pump charged with 1 N NaHCO 3 solution. Progress of the reaction was followed with periodic HPLC monitoring, as described in Example 44. After 24 hours, the reaction seems to be 94% complete, as determined by HPLC.
  • the prodrug compound Suc- ⁇ Ala-Leu-Ala-Leu-Dox, was fully characterized by mass spectrum analysis, FTIR, NMR.
  • the HMQC spectrum shows that there are three protonated aromatic carbons at 120.5, 120.3 and 137.2 ppm; the aromatic protons did not show any long-range C-H coupling, nor any coupling between adjacent protons.
  • the aromatic protons signals are very broad, indicating a short T relaxation time, which explains the lack of any observed coupling. Given this lack of coupling, it is not possible to assign these three sites uniquely, and are collectively assigned to (7), (8) and (9).
  • the two non-protonated aromatic carbons at 157.2 and 156.1 ppm have chemical shifts consistent with (10) and (11), i.e., aromatic carbons attached to oxygen. No long- range coupling is observed.
  • the 13 C NMR signals at 112.3, and 112.0 ppm are consistent with aromatic carbons ortho to oxygen substitution, and are assigned to (12) and (13).
  • the C NMR signal at 121.3 also shows this effect, so is assigned to (14).
  • the remaining three non- protonated aromatic carbons are assigned to the last three carbons in the region, (15), (16) and (17).
  • the carbon at 102.3 ppm has a chemical shift consistent with a carbon bound to two separate oxygen, so must be (24). This has long-range C-H coupling to the proton at 1.74 ppm, which is assigned to (25). This proton is coupled to the carbon at 30.6 ppm. Of the carbons in the C-O region (80 to 60 ppm), only one is protonated, at 77.4 ppm, so must be (26). This has no long-range C-H coupling, to either the proton or the carbon. There are three carbons not yet assigned in the 80 to 60 ppm region, all methines attached to oxygen.
  • the remaining four methyls are in the isopropyl region, and one is at 1.25/1.34 ppm, and must correspond to the last remaining methyl, (32).
  • the protons of this methyl show long-range coupling to only one carbon, at 51.3 ppm, which must be (33).
  • the protons of (33) overlap severely with other protons and can not be used for any long- range correlations.
  • the remaining four methyls must all arise from the isopropyl methyls, collectively labeled (34).
  • the protons of (34) show long-range coupling between the paired methyls, and to the carbons at 25.9/25.8 and 41.6/41.7 ppm; the methines are assigned (35)/(36) and the methylenes (37)/(38). All of these protons overlap at 1.5 to 1.8 ppm, but show long-range coupling to the methines at 54.7/53.5 ppm, which must be the ones adjacent to the amides, and are assigned (39)/(40).
EP01950291A 2000-06-14 2001-06-11 Durch thimet oligopeptidase spaltbare prodrugs Withdrawn EP1294405A2 (de)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US21188700P 2000-06-14 2000-06-14
US211887P 2000-06-14
US29044801P 2001-05-11 2001-05-11
US290448P 2001-05-11
PCT/US2001/018903 WO2001095945A2 (en) 2000-06-14 2001-06-11 Prodrug compounds cleavable by thimet oligopeptidase

Publications (1)

Publication Number Publication Date
EP1294405A2 true EP1294405A2 (de) 2003-03-26

Family

ID=26906558

Family Applications (1)

Application Number Title Priority Date Filing Date
EP01950291A Withdrawn EP1294405A2 (de) 2000-06-14 2001-06-11 Durch thimet oligopeptidase spaltbare prodrugs

Country Status (5)

Country Link
EP (1) EP1294405A2 (de)
JP (1) JP2004510703A (de)
AU (2) AU7130101A (de)
CA (1) CA2411660C (de)
WO (1) WO2001095945A2 (de)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20030033007A (ko) 2001-05-31 2003-04-26 코울터 파머수티컬, 인코포레이티드 세포독소, 약물전구체, 링커 및 이에 유용한 안정화제
CA2450316A1 (en) 2001-06-11 2002-12-19 Medarex, Inc. Cd10-activated prodrug compounds
FR2858936A1 (fr) * 2003-08-22 2005-02-25 Diatos Potentialisation de l'activation de prodrogues de haut poids moleculaire
BRPI0717902A2 (pt) 2006-12-01 2013-10-29 Medarex Inc "anticorpo monoclonal humano isolado, composição, conjugado anticorpo-molécula parceria, imunoconjugado, molécula de ácido nucléico isolada, vetor de expressão, célula hospedeira, método para prepar um anticorpo anti-cd22, método para inibir o desenvolvimento de uma célula tumoral que expressa cd22 e método para tratar uma doença inflamatória ou autoimuneem um indivíduo"
CL2007003622A1 (es) 2006-12-13 2009-08-07 Medarex Inc Anticuerpo monoclonal humano anti-cd19; composicion que lo comprende; y metodo de inhibicion del crecimiento de celulas tumorales.
MX2009006277A (es) 2006-12-14 2009-07-24 Medarex Inc Anticuerpos humanos que se enlazan a cd70 y usos de los mismos.
DE102008007381A1 (de) * 2008-02-01 2009-08-13 Dritte Patentportfolio Beteiligungsgesellschaft Mbh & Co.Kg Amidine und Guanidine und deren Derivate zur Behandlung von Krankheiten
US9662308B2 (en) 2008-02-01 2017-05-30 Dritte Patentportfolio Beteiligungsgesellschaft Mbh & Co. Kg Orally bioavailable pentamidine prodrugs for the treatment of diseases
US10087221B2 (en) 2013-03-21 2018-10-02 Sanofi-Aventis Deutschland Gmbh Synthesis of hydantoin containing peptide products
AU2014234314B2 (en) 2013-03-21 2018-02-15 Sanofi-Aventis Deutschland Gmbh Synthesis of cyclic imide containing peptide products
CA3203072A1 (en) 2020-12-22 2022-06-30 Andrea CASAZZA Compounds comprising a tetrapeptidic moiety
WO2022167664A1 (en) 2021-02-07 2022-08-11 Cobiores Nv Compounds comprising a tetrapeptidic moiety

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI0769967T1 (sl) * 1994-08-19 2008-06-30 Wallone Region Konjugati vsebujoäśi protitumorna sredstva in njihova uporaba
AU773420B2 (en) * 1998-12-11 2004-05-27 Medarex, Inc. Prodrug compounds and process for preparation thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0195945A2 *

Also Published As

Publication number Publication date
WO2001095945A2 (en) 2001-12-20
WO2001095945A3 (en) 2002-08-15
CA2411660C (en) 2011-03-01
AU7130101A (en) 2001-12-24
JP2004510703A (ja) 2004-04-08
AU2001271301B2 (en) 2006-08-31
CA2411660A1 (en) 2001-12-20

Similar Documents

Publication Publication Date Title
US8034787B2 (en) Enzyme-cleavable prodrug compounds
EP1144011B1 (de) Prodrugs und verfahren zu deren herstellung
US7214663B2 (en) Tripeptide prodrug compounds
US7696313B2 (en) Prodrug compounds with isoleucine
AU2001275525A1 (en) Tripeptide prodrug compounds
US6897034B2 (en) CD10-activated prodrug compounds
AU2001266853A1 (en) Prodrug compounds with an oligopeptide having an isoleucine residue
AU2002316539A1 (en) CD10-activated prodrug compounds
AU2001271301B2 (en) Prodrug compounds cleavable by thimet oligopeptidase
AU2001271301A1 (en) Prodrug compounds cleavable by thimet oligopeptidase

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20030113

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE TR

AX Request for extension of the european patent

Extension state: AL LT LV MK RO SI

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MEDAREX, INC.

RIN1 Information on inventor provided before grant (corrected)

Inventor name: YARRANTON, GEOFFREY T.

Inventor name: TROUET, ANDRE

Inventor name: PICKFORD, LESLIE B.

Inventor name: LOBL, THOMAS J.

Inventor name: GANGWAR, SANJEEV

Inventor name: DUBOIS, VINCENT

Inventor name: NIEDER, MATTHEW H.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20130430