AU740293B2 - Lipid splitting enzyme - Google Patents

Lipid splitting enzyme Download PDF

Info

Publication number
AU740293B2
AU740293B2 AU44675/99A AU4467599A AU740293B2 AU 740293 B2 AU740293 B2 AU 740293B2 AU 44675/99 A AU44675/99 A AU 44675/99A AU 4467599 A AU4467599 A AU 4467599A AU 740293 B2 AU740293 B2 AU 740293B2
Authority
AU
Australia
Prior art keywords
cells
conjugate
azt
active substance
pharmacologically active
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.)
Ceased
Application number
AU44675/99A
Other versions
AU4467599A (en
Inventor
Dieter Herrmann
Hans-Georg Opitz
Harald Zilch
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.)
Heidelberg Pharma Holding GmbH
Original Assignee
Roche Diagnostics GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU39278/95A external-priority patent/AU711367B2/en
Application filed by Roche Diagnostics GmbH filed Critical Roche Diagnostics GmbH
Priority to AU44675/99A priority Critical patent/AU740293B2/en
Publication of AU4467599A publication Critical patent/AU4467599A/en
Application granted granted Critical
Publication of AU740293B2 publication Critical patent/AU740293B2/en
Assigned to HEIDELBERG PHARMA HOLDING GMBH reassignment HEIDELBERG PHARMA HOLDING GMBH Alteration of Name(s) in Register under S187 Assignors: ROCHE DIAGNOSTICS GMBH
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/34Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase
    • C12Q1/44Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving hydrolase involving esterase

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

-1-
AUSTRALIA
PATENTS ACT 1990 DIVISIONAL APPLICATION NAME OF APPLICANT(S): Roche Diagnostics GmbH r r ADDRESS FOR SERVICE: DAVIES COLLISON CAVE Patent Attorneys 1 Little Collins Street Melbourne, 3000.
INVENTION TITLE: Lipid Splitting Enzyme The following statement is a full description of this invention, including the best method of performing it known to us: Q:\OPER\TDO\ROCHE.DIV 24/8/99 r 1A LIPID SPLITTING ENZYME The lipid cleavage enzyme complex (lipid cleavage enzyme; LCE) and its analogues, the subject matter of the present invention, are membranous enzymes that have previously not been described which can for example be isolated from the cell membrane fractions of human peripheral blood leucocytes or human macrophages and which cleave conjugates of pharmacologically active substances that are bound to a lipid-like carrier molecule to release the pharmacologically active substance or a monophosphate thereof. The invention also concerns the use of these conjugates which serve as substrates of this enzyme complex for the production of pharmaceutical agents that contain these conjugates as a pharmaceutically active substance. The pharmaceutical agents are suitable for the specific release and accumulation of pharmacologically active substances in appropriate target cells. In addition the invention concerns in vitro test systems which contain this enzyme complex to screen for further substrates of this enzyme complex as well as test systems for finding LCE analogues. LCE is understood as the enzyme complex, isolated enzyme as well as possible isoenzymes.
Apart from the inadequate efficacy of the therapeutically active substances used, the therapy of malignant neoplasias (carcinomas, sarcomas, haemoblastoses, haematological neoplasias), inflammatory diseases or autoimmune diseases as well as diseases caused by viruses or retroviruses such as for example AIDS, ARC (AIDS related complex), cytomegaly, herpes or hepatitis, is often accompanied by their extreme side effects. This effect is due to inadequate in vivo selectivity or the 2limited therapeutic range of the pharmacologically active substances used. The advantageous pharmacological in vitro properties of the pharmacologically active substances can often not be transferred to the in vivo conditions.
Therefore for years attempts have been made to modify the chemical structure of pharmacologically active substances to provide new substances which have improved properties with regard to therapeutic range. Moreover new pharmaceutical forms of administration are often developed with the aim of transporting the active substance specifically to its site of action where it is intended to display its therapeutic action. It is intended in particular to avoid undesired interactions with healthy cells. In the case of tumour cells which have corresponding surface antigens, antibodies have for example been produced that recognize these special surface antigens arnd thus selectively bind to the cancer cell. The antibodies are modified with suitable toxins in such a way that the toxin is released after binding to the cancer cell and the cancer cell is killed.
Mnother alternative for improving the therapeutic range is to change the physical properties of the underlying active substance by slight modification of the pharmacologically active substance for example by producing acid or base addition salts or by preparing simple esters [for example fatty acid esters; Pharm.
Sdi. 79, 531 (1990) in such a way that the solubility or compatibility of the active substance is improved.
These slightly chemically modified compounds are often referred to as so-sailed "prodrugs" since on contact with body fluids or in the liver (first pass metabolism) they are almost immediately converted into the actual therapeutically active agent.
-3 The technical problem that f orms the basis of the present invention was to find a new target which occurs as specifically as possible on or in cells that are a target f or the administration of pharmacologically active substances. The target should interact with appropriate pharmaceutically active substances so that the active substances are transported as specifically as possible to these target cells, and can be recognized, bound and taken up by these. In this case the pharmaceutically active substance should be essentially :4,00,composed of two components the first component being responsible f or the recognition and interaction with the target (ligand-specific part) and the second component being the actual active substance (active substancespecific part) which only develops its activity after specific binding to the target molecule and the actual :active agent or its monophosphate has been cleaved off intracellularly. This should avoid the undesired release of the pharmacologically active substance in the body 6' fluids so that healthy cells are not adversely effected by the pharmacologically active agent and undesired side-effects are substantially avoided. In this connection the pharmaceutically active substance used for this purpose for the production of the pharmaceutical form should on the one hand serve as a ligand for the target and on the other hand contain the actual active pharmacological agent whereby this can also in particular be based on already known pharmaceutically active structures for which it is intended to significantly improve the therapeutic range in this manner.
It was now surprisingly found that LCE is suitable as a target, LCE being mainly located on or in malignant, activated or virus-infected cells in particular on human 4 peripheral blood leucocytes, macrophages, kidney, adrenal or ovarian cells, cells of the lymphatic system or the lymphoid organs or cells of the brain. In the following this enzyme complex and its analogues are also abbreviated as LCE. Surprisingly LCE does not exhibit a uniform statistical distribution over all organs but is observed primarily in the membranes of certain cells which come into consideration as the target for the administration of pharmacologically active substances. A relatively very low enzyme activity was found in cardiac, bone marrow and liver cells. The homogenate and membrane fractions of cells, organs or tissues used to isolate the LCE have different specific activities or affinities of the LCE, the specific activity or affinity being strongly increased in activated cells compared to non-activated cells. This can be demonstrated for human peripheral blood leucocytes, lymphocytes and granulocytes as well as for human and non-human and murine mononuclear cells such as e.g. monocytes/ macrophages.
Surprisingly a characterisitic of LCE is that it cleaves lipid-like compounds as the substrate between the lipid backbone and the linker structure of a physiologically active substance covalently bound to this bridge. Such substrates can be described by the general formula I L B D
(I)
in which L represents a lipid residue, B a bridge and D represents a pharmacologically active substance or B-D represents an active substance phosphonate. Surprisingly the pharmaceutically active substances of formula I have a larger therapeutic range compared to the 5 pharmacologically active free or unmodified substances D or Moreover they often improve their retention time in the body, the bioavailability or the membrane permeability which is often known to be a critical factor blood-brain barrier, cell membranes etc.) of the pharmacologically active substances. Substrates of formula I therefore serve as a carrier system (carrier) for the pharmacologically active substance.
The conjugates of formula I can be referred to as intracellular drug storage, drug targeting and drug delivery systems with regard to their function. Their effect is that the pharmacologically active substance or its prodrug form is released intracellularly after oral .administration, this release taking place advantageously not unspecifically in all cells, organs or tissues of the body but rather specifically in those cells that contain the LCE in the cell membrane or also partially intracellularly. However, it is particularly surprising that cleavage does not already take place during the transport of the substrate through the body fluids such as blood, serum or lymph fluid or through the liver but only on or in the respective target cells. In this manner the undesired excretion of the cleavage product by the kidney or cleavage of the substrate in the liver is avoided so that the major proportion of the active substance is transported to the respective target cells.
As already stated above, such cells are in particular pathophysiologically or physiologically activated cells which come into consideration as a target for the administration of pharmacologically active substances such as for example blood leucocytes, lymphocytes, macrophages and other cell populations of the immunological lymphatic system. These are in particular activated cells macrophages, granulocytes, lymphocytes, leucocytes, thrombocytes, monocytes etc.) 6 which play a pathological, physiological, pathophysiological or symptomatic role in the respective disease process.
LCE is an enzyme complex which has previously not been described. It is characterized in particular in that it cleaves the compound (3'-deoxy-3'-azidothymidine)-5'phosphoric acid-(3-dodecyl-mercapto-2-decyloxy)-propyl ester (abbreviated as AZT-DMDOPE in the following) as a substrate to form 3'-deoxy-3'-azidothymidinemonophosphate and (3-dodecylmercapto-2-decyloxy)propanol (DMDOP). A further preferred substrate of LCE is the compound (3'-deoxy-3'-fluorothyidine)-5'phosphoric acid-(3-dodecyl-mercapto-2-decyloxy)-propyl ester (FLT-DMDOPE) which is cleaved into 3'-deoxy-3'and DMDOP.
Alternatively (5-fluorouridine)-5 '-phosphoric acid-(3dodecylmercapto-2-decyloxy)-propylester (5-FU-DMDOPE) is also cleaved into (5-FU-MP) and DMDOP.
t* The preparations containing LCE that have been produced are free of phospholipase C. This has been demonstrated by the different cation dependency of the LCE activity as well as by phospholipase C-specific inhibitors which do not inhibit LCE.
An enzyme assay was established (example 6) to determine the turnover rate of AZT-DMDOPE by cell homogenates, membrane and cytosol fractions, preferably of various human cell types.
The test principle is based on a cleavage of the parent substance by the LCE into AZT-MP and the corresponding 7 thioether lipid part. 14 C]-AZT-DMDOPE and nonradioactively labelled AZT-DMDOPE were used for this. The thioether lipid metabolite DMDOP was extracted from the mixtures (example 7) and the amount of radioactively labelled substance was measured in a liquid scintillation analyzer. Since a defined amount of 14 C]-AZT-DMDOPE was used in the enzyme assay it was possible to determine the turnover rate of the enzymatic cleavage.
The enzyme is preferably isolated from human peripheral blood leucocytes which have previously been activated by PHA or other stimulants cytokines etc.) or from murine kidney cells. Preliminary investigations have yielded a molecular weight of about 120,000 160,000 determined by the SDS-PAGE method. Cells which contain this enzyme can be identified in a simple manner by admixing them under suitable test conditions with a solution of AZT-DMDOPE or FLT-DMDOPE and detecting the cleavage products AZT-monophosphate or FLT-monophosphate or DMDOP for example by thin layer chromatographic methods or, in the case of radioactively labelled samples by scintillatographic methods (see examples 4 In contrast to the biochemically related phospholipases C or D, the LCE is not inhibited by known inhibitors of phospholipase C or D nor is it activated by activators of phospholipase C or D. In contrast to phospholipase C the enzyme is activated by the substance D 609 (tricyclodecane-9-yl-xanthogenate;
C
11
H
15 0S 2
K)
whereas D 609 inhibits phospholipase C.
In its non-phosphorylated form intracellular AZT does not have an inhibitory effect on viral reverse transcriptase (RT) (Nakashima et al., 1986, Antimicrob.
Agents Chemother. 3Q, 933-937; Mitsuya et al., 1985 Proc. Natl. Acad. Sci. USA A2, 7096-7100). The 8 structural analogue of thymidine is converted by successive phosphorylation by the intracellular enzymes thymidine kinase, thymidilate kinase and pyrimidine nucleoside diphosphate kinase (Yarchoan et al., 1989, N.
Engl. J. Med. 321, 726-738; Toyoshima et al., 1991, Anal. Biochem. 196, 302-307) via AZT-MP and AZT-DP into the therapeutically active RT-inhibiting ATZ-TP. AZT- DMDOPE as a thioether lipid AZT conjugate was converted by intracellular enzymatic cleavage into AZT or directly into the already phosphorylated form AZT-MP. In example 8 the intracellular concentrations of phosphorylated and non-phosphorylated AZT are determined after incubation with equipotent concentrations of AZT-DMDOPE and AZT.
In addition the enzyme complex and its analogues do not exhibit a uniform statistical organ distribution in various species human, mouse, rat, dog, ape) but are only found in membranes of certain cells, organs and tissues which serve as target cells for cleavable conjugates of formula I. The natural substrates of these enzymes are still unknown. In cytosolic fractions the LCE activity is always below or just at the limit of detection. A very low activity is for example also found in bone marrow cells which indicates a very low or even a complete absence of bone marrow toxicity of the pharmaceutically active substances of formula I used as substrates of the LCES.
The LCE activity exhibits a linear protein and time dependency, a specific dependency on metal cations (inhibition by Ca 2 Zn 2 and Mn2+) and a classical Michaelis-Menten kinetics (substrate dependency) (example In addition to the higher activity of the enzyme complex in activated cells there is also a higher affinity of the LCE to its substrate under these 9 conditions.
The isolated LCE or LCE strongly enriched from membrane fractions can also be used to screen for new potentially cleavable substrates or for substrates which occur naturally. The substrates found in this manner can then be investigated more extensively with regard to their essential structural features which are necessary for the recognition of the substrate and binding to LCE.
Such identified structural properties can then be used to produce chemically modified substrates which contain these essential features as well as in addition suitable functional groups that are suitable for coupling to pharmacologically active substances.
s.
This also enables a screening for inhibitors or activators of the lipid cleavage enzyme.
The isolated or strongly enriched enzyme can be used diagnostically when for example increased or reduced lipid cleavage enzyme activities lead in vitro or in vivo to pathological changes or when corresponding diseases or disease symptoms are associated with these pathological changes.
The LCE can also be used to produce diagnostic agents which are used to check the cleavage of these substrates when the pharmaceutically active substances are administered to patients which results in a specific and individual adaptation of the therapy modalities in these patients (drug monitoring).
Recombinant LCE can be produced by well-established methods by determining the amino acid sequence of LCE or 10 LCE fragments and screening a human or other mammalian gene library with oligonucleotide probes that have been constructed appropriately. The found gene is then expressed by an appropriate vector in a prokaryotic or eukaryotic cell system. The recombinant LCE can then be purified by methods known in protein chemistry (see e.g.
Maniatis, Molecular Cloning).
A particular characteristic of the LCE is that it can cleave compounds of the general type I: L -B D (I) t Cleavage by LCE in which L represents a lipid moiety, B represents a bridge and D represents a pharmacologically active substance or B-D denotes an active substance phosphonate. The very specific cleavage takes place between the lipid moiety and phosphate residue. An unspecific cleavage of compounds of formula I at other functional groups in the molecule is not observed.
In this connection the lipid moiety L of the conjugate of formula I represents the following residue of formula II
RI-X--CH
R-YCH-)m (CH2)min which 11
R
1 is a straight-chain or branched, saturated or unsaturated alkyl chain with 1 30 carbon atoms which cab be optionally substituted once or several times by halogen, C 5
-C
7 cycloalkyl, phenyl, C 1 -Cg alkoxy, C 1 -Cg alkylmercapto, CI-Cg alkoxycarbonyl,
C
1
-C
6 alkylsulfinyl or CI-C 6 alkylsulfonyl groups
R
2 is hydrogen, a straight-chain or branched, saturated or unsaturated alkyl chain with 1 carbon atoms which can be optionally substituted nce or several times by halogen, C 5
-C
7 cycloalkyl, phenyl, C 1
-C
6 alkoxy, C 1
-C
6 alkylmercapto, Cl-C 6 alkoxycarbonyl or c 1
-C
6 alkylsulfonyl groups X represents a valency dash, oxygen, sulphur, aminocarbonyl, oxycarbonyl, carboxyamino, carbonyloxy, carbonylamido, amidocarbonyl, a sulfinyl or sulfonyl group Y is a valency dash, aminocarbonyl, oxycarbonyl, carboxyamino, carbonyloxy, carbonylamido, amidocarbonyl, an oxygen or sulphur atom and m represents an integer between 1 and
R
1 in the general formula II preferably denotes a straight-chain or branched Ce-C 15 alkyl group which can additionally be substituted by a c1-Cs alkoxy or a Cl-CS alkylmercapto group. R 1 in particular represents a nonyl, decyl, undecyl, dodecyl, tridecyl or tetradecyl group. Methoxy, ethoxy, butoxy and hexyloxy groups come preferably into consideration as C 1 -CG alkoxy substituents of R 1 If R 1 is substituted by a C 1
-C
6 alkylmercapto residue, then this is in particular 12 understood as a methylmercapto, ethylmercapto, propylmercapto, butylmercapto and hexylmercapto residue.
R
2 preferably denotes a straight-chain or branched C g
C
15 alkyl group which can additionally be substituted by a c 1 -c 6 alkoxy group or a C 1
-C
6 alkylmercapto group. R2 in particular represents an octyl, nonyl, decyl, undecyl, dodecyl, tridecyl or tetradecyl group. A methoxy, ethoxy, propoxy, butoxy and hexyloxy group preferably come into consideration as C 1
-C
6 alkoxy substituents of R2. If R2 is substituted by a C1-Cg alkylmercapto residue then this is in particular understood as a methylmercapto, ethylmercapto, butylmercapto and hexylmercapto residue.
X is preferably sulphur, sulfinyl or sulfonyl and Y is oxygen. The heteroatoms X and Y in the lipid moiety L can only be replaced in special cases by the carboxylic acid ester known from lecithin since otherwise a hydrolytic cleavage to form the corresponding -lysolecithin derivatives or glycerol esters with a corresponding more rapid elimination of the pharmacologically active substance would already occur in the serum or in the liver (first pass effect). The thioether lipids and ether lipids Y O,S) of this application do not exhibit this cleavage in the serum of various species including humans.
Compounds are also preferred in which X and Y represent a valency dash, R 2 is hydrogen and R 1 represents a C 1 alkyl chain which can optionally be substituted by
C-C
6 alkoxy or Ci-C 6 alkylmercapto.
m is preferably 1 or 2 and particularly preferably 1.
13 The bridge B represents a valency dash or is expressed by the formula III
(III)
in which n can be 1, 2 or 3 but is preferably 1 or 2 and especially 1, Z is either 0 or S, and A is either 0, S or a valency dash and preferably 0.
The lipid moiety L and the phosphate bridge B have the above-mentioned meaning in which L preferably represents a residue of formula II and B is preferably a phosphate bridge of formula III. A phosphate bridge of formula III is particularly preferred in which n 1 and a lipid moiety of formula II is particularly preferred in which
R
1 and R 2 represent an alkyl residue with 8 15 C atoms, X equals sulphur and Y is oxygen. Furthermore compounds are preferred in which B as a phosphonate is a constituent of the structure of the active substance in which case n 1 and A is a valency dash.
The term "pharmacologically active substance" (named D or B-D in the case of phosphonates in iormula I) in this application represents an active substance within the legal pharmaceutical sense. This active substance can be an active substance of a pharmaceutical agent that has already been introduced and licenced by the pharmaceutical authorities or an active substance which is currently being registered as a pharmaceutical agent.
The definition "pharmacologically active substance" also encompasses such derivatives of active substances that can be chemically modified by introducing one or several functional groups (for example such groups which enable D to be coupled to the lipid carrier moiety L such as 14 e.g. hydroxy or amino groups). The definition also encompasses prodrug forms that are tonmed from the active substance D which are also physiologically active. In particular pharmacologically active substances D come into consideration whose clinical development has been discontinued or not been started due to undesired side effects or which only have a very narrow dose-effect spectrum so that the administration of the therapeutically required amount would be associated with high risks or virtually impossible to get under control.
Surprisingly it was found that the therapeutic range of a pharmacologically active substance D or B-D in the case of phosphonates of the active substance is significantly improved when the substance is coupled to a lipid-like carrier molecule. The conjugate prepared in this manner serves as a new active substance for the production of pharmaceutical forms of administration.
Overall the coupling results in an increased activity of the pharmaceutically active substance D or B-D in vivo since, due to the drug-delivery transport system that is formed, the pharmacologically active substance is a. ~.:localized in target cells and thus the efficiency of the pharmacologically active substance is increased. This means that on the one hand the amount of pharmacologically active substance that has to be administered can be reduced or on the other hand that an increased pharmacological effect is achieved while retaining the same effective amount.
The chemical structure of the pharmacologically active substances D or B-D can in addition be modified in such a way that the substances are changed with regard to their physical or chemical properties and for example 15 have a higher or lower lipophilicity but have essentially the same properties as the unmodified substance D or B-D with regard to their therapeutic effect. In particular it is advantageous when the substance D is chemically modified by the introduction of functional groups in such a way that it can be coupled via a suitable bridge to the lipid moiety L.
This is for example achieved by the introduction of hydroxy groups which are coupled via the phosphate group B to the lipid.
The pharmacologically active substance D or B-D is a chemically or biologically based substance (antibody, peptide, protein, hormone, toxin etc.; INDEX NOMINUM, International Drug Directory, Medpharm) with a biological effect as well as derivatives thereof chemically modified by the introduction of a functional group a hydroxy group). A prerequisite is that the pharmacologically active substance or its prodrug form are activated by this endogeneous enzyme via the .cleavage of the liponucleotide by the lipid cleavage enzyme. In this connection pharmacologically active substances are preferred which after cleavage by the LCE act in vivo as an intermediate product as the pharmacologically active substance monophosphate and are further phosphorylated by cellular enzymes such as e.g.
nucleoside monophosphate to nucleoside triphosphate, or are cleaved to form a free pharmacologically active substance (see example 8).
Within the sense of the invention all pharmacologically active substances come into consideration which are effective in vitro but are toxic in vivo in the therapeutic range i.e. all substances with a narrow therapeutic range which have a chemical functional group 16 for a covalent linkage to phosphate. In addition those substances can also be used which, although at first containing no functional group in their pharmacologically active form, can have one introduced by chemical modification without a loss in the effect of the substance.
Those pharmacologically active substances are preferably used for conjugation with a lipid residue L which normally reach their active form after phosphorylation (such as in the case of nucleosides) or phosphonates of the active substance. The pharmacologically active substance phosphate is then released from the conjugate by enzymatic hydrolysis of the conjugate. The release of the phosphorylated substance is particularly important since this process can also take place in cells which do not normally have the necessary enzymes (kinases) to phosphorylate the pure pharmacologically active substance. The conjugated pharmacologically active substance that is released intracellularly or in the cell membrane by LCE can for example have a cytostatic, cytotoxic, antitumoral, antiviral, antiretroviral, immunosuppressive or immunostimulating effect.
Compounds that are suitable as pharmacologically active substances D which can be optionally converted into a derivative capable of coupling by introduction of a functional group which does not significantly influence its action which then for example slows tumour growth, is a substance which intercalates into DNA and/or RNA, inhibits topoisomerase I and II, is a tubulin inhibitor, is an alkylating agent, is a ribosome inactivating compound, is a tyrosine phosphokinase inhibitor, is a differentiation inducer, a hormone, hormone agonist or hormone antagonist, is a substance which changes 17 pleiotropic resistance to cytostatic agents, is a calmodulin inhibitor, is a protein kinase C inhibitor, is a P-glycoprotein inhibitor, is a modulator of mitochondrially bound hexokinase, is an inhibitor of Y-glutamylcysteine synthetase or glutathione-s transferase, is an inhibitor of superoxide dismutase, is an inhibitor of reverse transcriptase of HIV-1 and HIV-2 or inhibitors of hepatitis viruses A-E.
The pharmacologically active substance D or B-D can have an antiinflammatory, antirheumatic, antiphlogistic, S: analgetic or antipyretic action. It can in addition be an antiarrhythmic agent, a calcium antagonist, antihistamine drug, an inhibitor of phosphodiesterase or a sympathomimetic or parasympathomimetic.
All substances are suitable as the pharmacologically active substances D or B-D which have a short half-life, in particular also compounds with different organ, tissue or cell half-lives, a poor bioavailability i.e. a poor resorption, high liver cleavage or rapid elimination, poor membrane penetration cell membrane, blood-brain barrier), bone marrow toxicity or other limiting organ toxicities cardiotoxicities, liver toxicities, nephrotoxicities, neurotoxicities etc), whose active concentration in vivo is too low. In addition those substances are suitable which interact specifically with the cell nucleus of the target cells and interfere with the molecular process at the DNA or RNA level such as e.g. antisense oligonucleotides, DNA fragments and those which can be used for gene therapy.
Pharmacologically active substances D in formula I are for example: AZT (azidothymidine), FLT (fluorois thyinidine), 5-fluorouracil, 5-f luorouridine, 6-M4PR, fludarabin, cladribin, pentostatin, ara-C, ara-A, ara-G, ara-i, Acyclovir, Ganciclovir, doxorubicin, 4'-epidoxorubicin, 4 '-deoxy-doxorubicin, etoposide, daunomycin, idarubicin, epirubicin, mitoxantron, vincristine, vinbiastine, Taxol, coichicine, meiphalan, 3' -deoxy-2 -f luoro-adenosime, FdA, 5-ethinyluraci 1-9 -B-Darabinofuranaside, 5-prop inyluracil-9-B-D-arabinofuranoside, d4T, ddU, ddl, ddA, d2T, 2'-deoxy-2',2'difluorocytidine, 5-trifluoromethyl-2' -deoxyuridine, chloro-2' ,3 '-dideoxy-3 '-fluorouridine, 3 '-deoxy-3 fluoromyoinositol, neplanocin A, ribavirin, myoinositol, fialuridine, 3TC (Lamivudine), doxifluridine, Tegafur, hypericii, pseudohypericin, Usevir, Faiiciclovir, Penciclovir, Foscarnet, carvedilol, actinomycii A, A. bleomycin, daunorubicin, floxuridine, mithramycin, mitomycin C, mitoxantirone, streptozotocin, vindesin, netilmycin, amaikacin, gentamycin, streptomycin, kanamycin A, tobramycin, neomycin B, plicamycin, papamycin, aiphotericin B, vancomycin, idoxuridine, trifluridine, vidarabin as well as morphines, .prostaglandines, leukotrienes or cyclosporins. moreover terfenadin, dexamethasone, terbutalin, predniso lone, fenoterol, orciprenaline, salbutamol, isoprenalile, muscarine, bupranolol, oxyphenbutazone, oestrogen, salicylic acid, propranolol, ascorbic acid, spongiadiol, diclofenac, isospongiadiol, fi-ufenamic acid, digoxin, 4-3nethyl-aminophenaolQ, allopurinol, theophylline, epoprostenol, nifedipine, quinine, reserpine, methotrexate, chioroanbucil, spergjualine, ibuprof en, indomethacin, sulfasalazine, penicillinamine, chioroquine, azathioprile also come into consideration.
Pharmacologically active substances B-D in f ormnula I are for example PMEA and other acyclic nucleoside 19 phosphonate analogues, quinoline-phosphonic acids, MP- 101, fostedil, HAB-439, 2-amino-7-phosphonoheptanoic acid, fosfosal, fosphenytoin, atrinositol, cyplatate, difficidin, fosquidone, alafosfalin, foteumstine, etoposide, encyt/E, 4-oxo-S-phosphono-D-norvalin and other NMDA-antagonist phosphonates.
Preferred pharmacologically active substances are for example also peptides, proteins and oligonucleotides such as e.g. corticotropin, calcitonin, desmopressin, gonadotropin, goserelin, insulin, zypressin, betamelanotropin, alpha-melanotropin, muramyldipeptide, oxytocin, vasopressin, FK-506, octreotide or enalkiren.
0* The above-mentioned pharmacologically active substances and the conjugates to be prepared therefrom only represent examples and do not limit the inventive idea.
Compounds of formula I and the production thereof are for example described in the applications WO 92/03462, WO 93/16092, WO 93/16091, WO 94/03465, PCT/EP94/02123, DE 4402492, DE 4418690 as well as for example in WO 91/19726; EP 0 350 287; US 5,223,263; US 5,194,654; US 4,921,951; US 4,622,392; US 4,291,024; US 4,283,394.
In the case of antivirally effective nucleosides lipid derivatives (diacylglycerol nucleosides) and their use in liposomal form are described in EP 0 350 287 and US 5,223,263. An uptake of the substance preferably in the form of liposomes by cells of the reticuloendothelial system (RES) e.g. macrophages and monocytes should be possible.
It was possible to show by appropriate comparative experiments that the therapeutic effects in vivo of the 20 diacylglycerol conjugates known from EP 0 595 133 are inferior to the thioether or etherlipid conjugates from WO 92/03462. This is due to unspecific hydrolysis of the fatty acid esters which does not only take place at the site of action. In contrast the non-hydrolysable thioether and ether residues exhibit significant advantages since a substance with biological action or appropriate intermediate products are only released in the membranes of the target tissue or intracellularly by the special enzyme (LCE).
The conjugates of formula I exhibit significant advantages in comparison with the unconjugated .pharmacologically active substance D or B-D The specific carrier or L) covalently bound to the pharmacologically active substance improves the bioavailability of the poorly resorbed pharmacologically active substances, the tolerance of potentially toxic active molecules, the retention time of rapidly eliminated or metabolized pharmaceutical agents and the -membrane penetration of compounds with poor membrane permeability blood-brain, cells etc.). The enzymatic cleavage in vivo into carrier and pharmacologically active substance D (or substance derivative) or into carrier and pharmacologically active substance phosphate (D-monophosphate) or phosphonate B-D usually does not occur in the serum but only intracellularly. In addition the carrier moiety with its lecithin-like structure, which is essential for the claimed effect, improves the penetration or membrane permeability of the pharmacologically active substance and exhibits a depot effect in many cases. Moreover the gastrointestinal tolerance of the lipid conjugates L-B-D is considerably better than that of the pure pharmacologically active substances D. The lipid 21 conjugate also exhibits a better penetration through membrane structures during resorption and thus it is more able to overcome the resorption barriers. The same also applies to penetration e.g. of the blood-brain barrier by facilitated diffusion or possibly active transport. Conjugates of formula I with a lipid moiety as limited by formula II are particularly preferred which cleave off a substance with biological activity by enzymatic hydrolysis with the lipid cleavage enzyme which leads to less side effects in vivo than after administration of the pharmacologically active substance :alone.
in addition the in vivo distribution is improved by a better binding of the conjugate to plasma and tissue proteins. The conjugate is primarily oxidized by normal biotransformation from a thioether (X S) to a sulfoxide (X SO) which, however, due to the equipotent action of the sulfoxide in comparison to the thioether, 17 does not represent a disadvantage. The slow release of -the pharmacologically active substance from the conjugate ensures a low level of active substance that is, however, constant over a longer period of time and 27 thus improves the efficacy or avoids trjxic side-effects.
The released pharmacologically active substance in the fonm of a monophosphate no longer penetrates out of the cell due to its high hydrophilicity.
The total body, cell and the organ half-lives of the pharmacologically active substance are considerably extended by the conjugation due to the longer retention time of the conjugate in the organism. Due to the lack of LCE activity for cleavage in serum and in some organs, almost no or only slight bone marrow and organ toxicity can be observed. It is particularly 22 advantageous that the conjugates of formula I are specifically accumulated in various target organs, tissues or cells.
The compounds of formula I can be used as active substances for the production of pharmaceutical agents which can be used for all diseases in which a high level of pharmacologically active substance in cells, organs or tissues is required or is beneficial. An essential requirement for this transport system denoted "drugstorage-delivery-targeting" is that the cells which are to respond in accordance with the intended therapy have the lipid cleavage enzyme in the inner or outer cell membrane so that the active substance binds to the LCE in a first step and is subsequently transported through the cell membrane into the interior of the cell in the process of which the active substance is cleaved to form the physiologically active substance either essentially simultaneously with transport through the cell membrane or even later partially within the cell. Intracellular cleavage takes place especially in those cases in which the LCE is also located within the cell. Within the sense of the invention the cleavage of the active substance linked with the intracellular release of the pharmacologically active substance can also occur such that either the physiologically or pharmacologically active substance is formed directly in this process or an appropriate precursor of this substance (prodrug form). Suitable target cells are for example blood leucocytes (PBLs), monocytes, kidney cells or macrophages and cells of the immunological lymphatic system.
The effect of compounds of formula I is in particular immunomodulating, para/sympatholytic, /sympathomimetic, 23 centrally and/or peripherally muscle-relaxing; antihypertensive, antihypotensive, antiobstructive, analgetic, anti-phlogistic, antiemetic, antiinflammatory, antiallergic, antiasthmatic, antipyretic, antiulcerative, antacidic, antianginal, antiarrhythmic, antipsychotic, antidepressive, antiepileptic, anticonvulsive, antiparkinsonoid, antihistaminergic, antimuscarineric, antiserotoninergic, antigabaergic, antiadrenergic, anticholinergic, glycosidic, chronotropic, bathmotropic, dromotropic, inotropic, diuretic, antidiuretic, uricosuric, uricostatic, antihypolipidaemic, antifibrinogenic, antidiabetic, hypoglycaemic, antioestrogenic, antiandrogenic, antigestagenic, antiosteoporetic, thyreostatic, bonegrowth stimulating, narcotic, anaesthetic, antihypnotic, antiinfectious, antibiotic, antituberculostatic or haematopoetic or they represent a vitamin.
The advantageous effect of one of the compounds according to the invention can be increased by S combination with suitable pharmaceutical agents or combination of two different conjugates of the present application.
The effect of a cytostatic or cytotoxic conjugate of this application can for example be increased by combination with other cytostatic or cytotoxic compounds preferably when components with different mechanisms of action are used or a combination of a cytostatic or cytotoxic conjugate with an antiviral conjugate (synergism e.g. in the case of AIDS).
The conjugate combinations are especially suitable according to the invention in which one component has 24 cytostatic or cytotoxic potential and the other component overcomes for example multi drug resistance or a component inhibits reverse transcriptase of HIV and the other component inhibits the protease for example or both liponucleotides of the combination do not exhibit cross-resistance. Compounds of formula I and their pharmaceutical preparations can also be used for the production of pharmaceutical agents which are suitable for the treatment and prophylaxis of various diseases in combination with other pharmaceutical agents.
Alkali, alkaline-earth and ammonium salts of the phosphate group come above all into consideration as possible salts of the compounds of the general formula I. Lithium, sodium and potassium salts are preferred as the alkali salts. Magnesium and calcium salts come in particular into consideration as alkaline-earth salts.
Ammonium salts are understood according to the invention as salts which contain the ammonium ion that can be substituted up to four times by alkyl residues with 1-4 .carbon atoms and/or by aralkyl residues preferably by benzyl residues. In this case the substituents can be the same or different.
The compounds of the general formula I can contain basic groups in particular amino groups which can be converted using suitable inorganic or organic acids into acid addition salts. Acids which come for example into consideration are: hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, fumaric acid, succinic acid, tartaric acid, citric acid, lactic acid, maleic acid or methanesulfonic acid.
In addition to the compounds of the general formula I 25 and the specifications according to formula II and III this application also claims tautomers thereof and their physiologically tolerated salts of inorganic and organic acids or bases as well as processes for their production and pharmaceutical agents containing these compounds.
Since the compounds of the general formula I contain asymmetric carbon atoms all optically active forms and racemic mixtures of these compounds are also a subject matter of the present invention.
This application also concerns new liponucleotides. The synthesis of these conjugates is exemplified in examples 14-16. The LCE also cleaves 2-chloro-2'-deoxy-adenosine conjugates (cladribin conjugates) of formula V, 9-(B-Darabino-furanosyl)-2-fluoroadenine conjugates (fludarabine conjugates) of formula VI, 3-(2-deoxy-B-Derythro-pentofuranosyl)-3, 6, 7, 8-tetrahydroimidazo diazepin-8-ol conjugates (pentostatin conjugates) of formula VII. The said compounds can be -used therapeutically better than the corresponding nucleosides alone since they have a very good efficacy and a large therapeutic range and have the same advantages as the above-mentioned derivatives of formula I:
NH
2 -O N~ -N C
OH
26 N
KN
L-a -o N N F O.0 (Vi) 6H
HO
N
:NH
L 0 0 V II)
OH
Within the sense of the present invention the following active substances are used in particular as compounds of formula I for producing pharmaceutical agents: (3 1-deoxy-3 '-azidothymidine)-5' -phosphoric acid-(3dodecyl-mercapto-2-decyloxy) -propyl ester 2. -deoxy-3 '-azidothyrmidine) -5 '-phosphoric acid- (3undecyl-mercapto-2-undecyloxy) -propyl ester 3. (3 1 -deoxy-3 I -f luorothymidine) -phosphoric acid- (3-dodecyl-meapto-2 -decyloxy) -propyl ester 4. (3'1 -deoxy-3 I'-f luorothymidiine) -5 -phosphoric acid- (3 -undecyl-mercapto-2-uldecyloxy) -propyl ester 27 S. (21,3 -dideoxycytidine) -5 '-phosphoric acid- (3dodecyl-mercapto-2-decyloxy) -propyl ester 6. -dideoxyinosine) -5 '-phosphoric acid- (3dodecyl-znercapto-2-decyloxy) -propyl ester 7. (3 '-deoxythymidine) -phosphoric acid- (3-dodecylmercapto-2 -decyloxy) -propyl ester 8. (5 '-fluorouridine) -5 '-phosphoric acid- (3-dodecylmercapto-2-decyloxy) -propyl ester (6-mercaptopurine-9-J3-D-ribofuranoside) 0 0 0phosphoric acid- (3-dodecylmercapto-2-decyloxy) propyl ester 10. (5-trifluoromethyluridine) -phosphoric acid- (3dodecyl-mercapto-2-decyloxy) -propyl ester 11. (1-B-D-arabinofuranosyl-5-ethinyluracil) phosphoric acid- (3-dodecyl-mercapto-2-decyloxy) e: 0.propyl ester 12. (2 '-deoxy-5-propinyluridine) -phosphoric acid- (3dodecyl-mercapto-2-decyloxy) -propyl ester 13. (1-hydroxymethyl) ethoxy~methyllguanine) phosphoric acid- (3-dodecyl-mercapto-2-decyloxy) propyl ester 14. 2 '-L9-(ethoxylethyl) guanine] )phosphoric acid- (3dodecyl-mercapto-2-decyloxy) propyl ester 28 Example 1: Enzymatic cleavage of AZT-DMDOPE mg (3'-deoxy-3'-azidothymidine)-5'-phosphoric acid- (3-dodecyl-mercapto-2-decyloxy-propyl ester (AZT-DMDOPE) is suspended in 0.5 ml 0.1 M Tris buffer solution and incubated for 15 hours at 37°C after addition of the enzyme (0.5 mg phosphodiesterase and 0.1 mg phospholipase/nuclease). Subsequently the solution is examined by thin layer chromatography. After addition of 5 drops of saturated NaHCO 3 solution it was incubated for a further 6 hours at 37"C and subsequently the solution was examined by thin layer chromatography.
The solution was examined for the formation of possible cleavage products (AZT, AZT-monophosphate, DMDOP and lipid monophosphate) by thin layer chromatography using various eluting agents.
The following enzymes were used in the above-mentioned test: a) Phospholipase C from Bacillus cereus, 2000 U/0.5 ml, Boehringer Mannheim GmbH b) Phospholipase D, type I, from cabbage, 150 300 U/mg, Sigma c) Phosphodiesterase from calf spleen, 2 U/mg, Boehringer Mannheim GmbH d) Phosphodiesterase from snake venom, Boehringer Mannheim GmbH e) Nuclease from Staphylococcus aureus, 15,000 U/mg, Boehringer Mannheim GmbH f) LCE (lipid cleavage enzyme) 29 Result: The following Table shows that a weak fluorescence quenching at the level of the lipid phosphate is only found in the case of phospholipase D. In none of the cases a) e) used as reference enzymes was AZT or AZT monophosphate formation detected. Apart from the unchanged AZT-DMDOPE no additional spot was found in these experiments within the detection limit.
Enzyme Fluorescence quenching
TLC
Phospholipase C negative Phospholipase D weak Phosphodiesterase from negative calf spleen Phosphodiesterase from negative snake venom Nuclease from negative Styphylococcus aureus LCE positive *9 *o 5.
S
Oe* O
OS..
sample 2: Figure legends Fig. 1: This figure shows the activity of the LCE which was obtained from the membrane fraction of human PHA-stimulated peripheral blood leucocytes relative to the protein concentration. There is a linear relationship between the protein concentration and the amount of the cleavage product DMDOP formed.
30 Fig. 2: This figure shows the substrate kinetics (Michaelis-Menten kinetics) of the LCE which was obtained from the membrane fraction of human, PHA-stimulated peripheral blood leucocytes.
Fig. 3: This figure shows the substrate kinetics (Michaelis-Menten kinetics) of the LCE which was obtained from the membrane fraction of human, PHA-stimulated peripheral blood leucocytes [M] and from quiescent (non stimulated) peripheral blood leucocytes Fig. 4: This figure shows the dependence of the specific enzyme activity of the LCE which was obtained from the membrane fraction of human, PHAstimulated peripheral blood leucocytes on the calcium ion concentration.
9* Fig. 5: This figure shows the organ-specific and tissuespecific distribution of the LCE of cell membrane preparations in the untreated dog.
Fig. 6: Intracellular concentrations of AZT and AZT nucleotides in stimulated human PBL after 1, 3, 6 and 24 h incubation with: 0.03 pg AZT/ml or 1 Wg AZT-DMDOPE/ml (0) 0.3 ig AZT/ml or 10 ig AZT-DMDOPE/ml (0) (means, n 2 determinations) Fig. 7: Intracellular concentrations on AZT and AZT nucleotides in non-stimulated human PBL after 1, 3, 6 and 24 h incubation with: 0.03 Ig AZT/ml or 1 jg AZT-DMDOPE/ml
(D)
0.3 Mg AZT/ml or 10 Mg AZT-DMDOPE/ml (0) 31 (means, n 2 determinations) Fig. 8: Intracellular concentrations of AZT and AZT nucleotides in stimulated human PBL after 6 and 24 h incubation with 1 g AZT-DMDOPE/ml and pg AZT-DMDOPE/ml treatment with alkaline phosphatase O no treatment with alkaline phosphatase (means, n determinations) Fig. 9: Intracellular concentration of AZT and AZT nucleotides in P3X63-Ag8.653 cells after 6, 24 and 48 h incubation with: 0.03 pg AZT/ml or 1 pg AZT-DMDOPE/ml (0) 0.3 pg AZT/ml or 10 g AZT-DMDOPE/ml (0) (means, n 2 determinations) Fig.10: Intracellular concentration of AZT and AZT nucleotides in P3X63-Ag8.65 3 cells after 6 and 24 h incubation with 1 pg BM 21.1290 Na/ml and pg AZT-DMDOPE/ml U treatment with alkaline phosphatase no treatment with alkaline phosphatase S(means, n determinations) Fig.ll: Intracellular decay kinetics of AZT and AZT nucleotides in stimulated human PBL after 24 h incubation with 0.3 pg AZT/ml(A) or 10 pg AZT- DMDOPE/ml(B). Intracellular concentration of AZT and AZT nucleotides 1, 3, 6, 24 and 48 h after removing the AZT or ATZ-DMDOPE incubation solution (means, n 2 determinations) 32 Fig.12: Enzymatic cleavage of [1 4 C]-AZT-DMDOPE by membrane fractions (100 Mg protein/preparation) of stimulated human PBL. Thin layer chromatogram of the n-heptane phase after adsorption of the substrate to silica gel 60H and separation in the IBA system (2-propanol:n-butyl acetate:redistilled water, 10:6:4, v/v/v) using silica gel 60 as a stationary phase.
@0*
S
00 *5
S
@055 .5 eo.
0* ft S. *ft 50 *o 1 oeO f *n Fig.13: Specific activity [pmol mg- 1 min
I
of the LCE in cell homogenates, cytosol and membrane fractions of stimulated human PBL (mean SD, n 6 determinations) Fig.14: Cleavage of AZT-DMDOPE by the membrane fractions of stimulated human PBL in relation to the protein concentration (mean SD, n 3 determinations).
Fig.15: Cleavage of AZT-DMDOPE by the membrane fraction of human monocytes/macrophages (stimulated monocytes) in relation to the protein concentration (mean, n 2 determinations).
Fig.16: Specific activity of the LCE in the membrane fractions of stimulated and non-stimulated human PBL and of human monocytes and monocytes/macrophages (stimulated monocytes) (human PBL: mean SD, n 6 determinations) (human monocytes: mean SD, n 4 determinations.
Fig.17: Cleavage of AZT-DMDOPE by membrane fractions of stimulated human PBL in relation to the incubation time (mean SD, n 3 determinations) 33 Fig.18: Thin layer chromatogram of 1 1 4 C]-AZT-DMDOPE (A) and of 1 4 C]-AZT-DMDOPE after enzymatic cleavage by cell homogenates of 5 X 10 7 CEI4-SS cells (B) after an incubation of 6 h at 37 0 C. Extraction of the thioether lipids with diethyl ether 2 propanol vf v) and separation in the IBAE system (2-propanol :n-butyl acetate: redistilled water: glacial acetic acid, 3:5:1:1, v/v/v/v) using silica gel 60 as a stationary phase Examnle 3: influence of various en zyme inhibitors or activators on *9 the LCE activity Inhibitor/ Site of attack of LCE Influence on activator the inhibition/ membrane LCE activity activation fraction SQ 22536 adenylate-cyclase kidney (Balbfc) none 7-nitro- NO synthesis kidney (Balb/c) none indazole RHC-80267 DAG lipase kidney (Balb/c) none neomycini PLC, (PLD) kidney (Balb/c) none wortmannin PLD, (PLC) kidney (Balb/c) none acetyl- PLC kidney (Balb/c) none salicylic acid GTP-B-S Gp activator kidney (Balb/c) none D 609 PLC kidney (Balb/c) stimulation CEM-68 stimulation 34 Example 4: Subcellular fractionation a. Preparation of cell homocenates, membrane and cytosol fractions The cells cultured for the preparation of membrane and cytosol fractions were transferred to polypropylene tubes (50 ml) and sedimented for min at 1600 rpm in a Minifuge T at room temperature. In order to remove residues of the culture medium the cell sediment was washed three times with cold PBS and taken up in lysis buffer at a cell density of 1 5 x 10 6 cells/mi. The cell suspension was subsequently transferred to a glass homogenizer cooled on ice. The cells were mechanically disrupted by several-fold movement of a teflon piston during which the cell disruption was monitored optically in a reverse phase contrast microscope. The disrupted cells were subsequently centrifuged for 10 min at 1700 rpm and 4°C in a Minifuge T to remove the cell nuclei and the nondisrupted cells. The supernatant was carefully removed with a pipette and diluted to 10 (w/v) sucrose with 50 mM Tris buffer (pH The cell homogenate was divided into portions and stored at or used to isolate membrane and cytosol fractions. For this 3.2 ml of the cell homogenate was transferred to a thick-walled polycarbonate tube (3.2 ml) cooled on ice and centrifuged for 1 h at 4°C and 75,000 rpm in a Beckmann table-top ultracentrifuge with a TLA-100.4 rotor. The supernatants were carefully removed with a Combitip 35 pipette and the readily visible membrane sediments were each admixed with I ml 50 mM Tris buffer (pH 7.5)/10 sucrose. The membrane sediment was coarsely homogenized with the aid of a syringe ml) with a cannula (0.9 x 40 mm). The coarse homogenates were combined and finely homogenized using cannulae of a smaller diameter (0.8 x 40 mm and 0.45 x 25 mm). The membrane and cytosol fractions were divided into portions and stored at -700C.
Lys.is buffer 50 mM Tris pH sucrose Ag/ml APMSF In order to dissolve the sucrose the buffer has to be gently warmed while .stirring.
b. Isolation of the plasma membrane of stiulated huza n peripheral blood lymphocytes (PBL) (Record et al. (1985). Biochem. Biophys. Acta 8/9 1-9) Human PBL were isolated by centrifugation in an isotonic density gradient and stimulated for 72 h with PKA-M. The cells were washed three times with cold PBS to remove residues of the culture medium and, after determination of the cell number, were frozen in liquid N 2 and subsequently stored at 0 C. The cells were lysed by freezing and thawing three times in buffer I whereby the cell density was adjusted to 2.5 x 108 cells/ml. A mixture of 11 ml Percoll and 2.13 ml buffer 2 was placed in Ti6O centrifuge tubes (30 ml) which had previously 36 been adjusted to pH 9 with 70 A 1 2 N NaOH. For the isolation of the plasma membrane 4 ml of the homogenate was applied to the mixture and subsequently centrifuged for 10 min at 4°C and 39,000 rpm in a Ti60 rotor of a Beckmann ultracentrifuge L 5 50 with the brake switched off.
1 ml fractions were taken from the upper phase of the centrifuge tubes and diluted with 2 ml buffer 3. The solutions were subsequently transferred to TiS0 tubes (10.4 ml) and centrifuged for 45 min at 4 0 C and 45,000 rpm in a Beckmann ultracentrifuge L 5 50 with a Ti5O rotor to remove residues of the *separation medium. The supernatants were removed with a pipette, fractionated, frozen in liquid N 2 and stored at -70°C. The fractions were characterized by determining the protein content and the enzyme activity of alkaline phosphatase.
Buffer 1 100 mM KCI mM MgC2 x 6 1mM ATP 2 mM (4-amedinophenyl)-methane sulfonylfluoride (APMSF) 25 mM Tris pH 9.6 Buffer 2 400 mM KCl WM MgCI 2 X 400 mM Tris pH 9.6 Buffer 3. 100 mM KCI mM MgC1 2 x 6 mM Tris pH 7.42 37 Example B.
Production of substrate solutions for the LCE assay A mixture of [14C]-AZT-DMDOPE and non-radioactively labelled AZT-DMDOPE was used as a substrate in the LCE assay.
Starting with a 1 4 C]-AZT-DMDOPE stock solution, a solution with a specific activity of 83.27 kBq/ml was prepared by dilution with ethanol. The ethanolic solution could be stored for 2 months at 4°C under inert o In a separate mixture the non-radioactively labelled component of the substance mixture was prepared. For this a solution of AZT-DMDOPE at a concentration of I jmol/ml in ethanol was prepared and continuously sonicated at 30 energy release for 1 min in an ice/water bath with an ultrasonic probe.
4 4 4 In order to prepare the substrate mixture of radioactive and non-radioactive AZT-DMDOPE, 1.67 kBq of the radioactively labelled 14 C]-AZT-DMDOPE and 0.7 5 nmol of the non-labelled compound were transferred per mixture from these ethanolic solutions into a glass tube ml). The solvent was evaporated in a N 2 stream and AZT-DMDOPE was taken up in a suitable volume of redistilled water and continuously sonicated at 30 energy release for 1 min while cooling in an ice/water bath.
38 Example 6: Lipid cleavaqe enzyme (LCE) assay *~9
S
•eee e eeem r In order to examine the enzymatic cleavage relative to time and the protein concentration 5 nmol AZT-DMDOPE and 0.98 rnol 1 4 C]-AZT-DMDOPE (1.67 kBq) In a volume of 200 pl redistilled water were usually used in the LCE assay.
The pipetting scheme is given in the following table.
Table Pipetting scheme for the LCE assay to determine the turnover of AZT-DMDOPS Reaction mixture Control EGTA [20 mM] 50 Tris, pH: 8.0 [1 M] 50 [Ai/mixture] AZT-DMDOPE/[ 1C -AZT- DMDOPE [nmol/mixture] 0.10 5.98 0.10 5.98 [Al/mixture] 5 200 5 200 Protein [mg/mixture] 0.0125 0.2 [gl/mixture] x redistilled water ad 500 ad 500 [p1] In order to examine the enzymatic cleavage in relation to the substrate concentration, a substrate stock solution of 0.98 nmol C 1 4 C]-AZT-DMDOPE and 0.7 nmol AZTp1 redistilled water was used in the LCE assay. The various substrate concentrations were adjusted in the LCE assay by continuously doubling or halving the volume of the substrate solution. The 39 mixtures were placed in glass tubes (5 ml) and started by adding the substrate solution. The incubation was carried out at 37 0 C in a water bath while gently shaking.
Examuple 7: Extraction -o-f UDOP from an agueovs matrix The reaction mixtures of the LCE assays were taken from the water bath after an appropriate incubation period and the reaction was stopped by adding 750 g1 2-propanol.
After thorough mixing 700 A1 n-heptane heated to room temperature was added to the mixtures. The mixtures were again thoroughly mixed for 30 sec and centrifuged for min in a Minifuge T at 3200 rpm at room temperature to accelerate the phase separation. Subsequently 500 p1 of the upper phase (n-heptane) was removed and transferred *to new glass tubes (5 ml) containing 10 mg silica gel H and 200 Al. n-heptane. The mixtures were thoroughly mixed f or 30 sec and centrifuged f or 10,mi under the conditions mentioned above to sediment the silica gel.
Subsequently 500 Al was taken from the supernatant and admixed with 3 ml scintillation liquid. The radioactivity was measured for 4 min in a liquid scintillation analyzer. In order to determine the absolute value of radioactivity, 200 Al of the substrate solution was mixed with 3 ml scintillation liquid and measured under the same conditions in the liquid scintillation analyzer.
40 EBample 8: In vitro pharmakokinetic studies with AZT-DMDOPE and A2T a) Determination of the intracellular concentrations of AZT and/or AZT nucleotides in stimulated and non-stimulated human PBL after incubation with AZT- DMDOPE and AZT Human PBL were isolated from the buffy coat in order to determine the intracellular concentrations of AZT and/or AZT nucleotides after incubation with AZT-DMDOPE and AZT.
e For this the buffy coat of healthy donors is mixed and fractionated by centrifugation in an isotonic medium with a density of 1.077 g/ml. Since mononuclear blood cells (monocytes and lymphocytes) have a lower density than erythrocytes and polymorphonuclear granulocytes they form a ring at the border between the separation medium and the sample and can be removed with the aid of a pipette. Erythrocytes and polymorphonuclear granulocytes sediment through the medium as a result of their higher density and can thus be separated from the PBLs.
In order to stimulate cell proliferation phytohaemagglutinin A (mucoprotein) (PHA-M) the mitogenic lectin extract from the red scarlet runner (Phaseolus vulgaris) was used.
Phytohaemagglutinin is a family of 5 isolectins which are present as tetramers. The subunits are composed of a lymphocyte-reactive type L and an 41 erythrocyte-reactive type E. Type L has a high affinity to lymphocyte surface receptors and is thus responsible for the mitogenic properties. The lymphocytes were stimulated for 72 h at 37°C and
CO
2 in KPMI1 1640 complete medium III. After the stimulation was complete the cells were cultured for a further 24 h in RPMI 1640 complete medium IV to increase the cell number.
For the determination of the intracellular concentrations of AZT and AZT nucleotides the cells were transferred into RPMI 1640 complete medium II •0 at a density of l x 106 cells/ml in tissue culture nf flasks (50 ml/25 cm 2 and incubated for 1 to 24 h in the presence of 0.03 and 0.3 pg AZT/ml (lot and 1 or 10 Ag AZT-DMDOPE/ml.
The concentrations of AZT-DMDOPE and AZT in this case depend on the ICSO values for the antiretroviral activity which was determined for AZT and AZT-DMDOPE in HIV-1 infected human PEL.
The cultures were subsequently incubated for 1, 3, 2 6 and 24 h at 37°C and 5 CO 2 AZT and its ee nucleotides were subsequently extracted with 60 methanol. The extracted nucleotides were quantitatively dephosphorylated to AZT with alkaline phosphatase since only AZT but not the corresponding nucleotides can be detected with the aid of a radioimmunoassay.
Accordingly the concentrations of intracellular AZT that were determined with the aid of a radioimmunoassay were composed of that of the AZT and that of 42 the AZT nucleotides together. A separate quantification of the concentrations of AZT-MP, AZT-DP and AZT-TP was not carried out. Experimental observations on stimulated PBL were able to demonstrate a 40-fold higher concentration of AZT- MP compared to AZT-DP and AZT-TP (Arn&r et al., 1992, J. Biol. Chem. 267, 10968-10975).
Consequently it can be assumed that the nucleotide fraction is almost exclusively composed of AZT-MP.
In stimulated human PBL a maximum concentrations of 2.90 and 30.97 ng/10 6 cells was reached after an incubation of 6 and 3 h respectively when incubated 'with 0.03 and 0.3 gg AZT/ml respectively (Fig. 6).
The concentrations subsequently decreased as the incubation period increased to 2.32 and 19.88 ng/10 6 cells respectively. After incubation with 1 and 10 pg AZT-DMDOPE/ml there was a continuous increase in the intracellular level of AZT and its nucleotides over the entire incubation period. After 24 h incubation with AZT-DMDOPE and AZT the intracellular levels were identical. In further experiments it was possible to show that the intracellular concentrations of AZT and AZT nucleotides after incubation with AZT-DMDOPE for a period of 18 48 h was even higher than after incubation with an equipotent concentration of AZT.
In the case of non-stimulated human PBL the opposite relationships were found (Fig. Thus after incubation with AZT-DMDOPE the intracellular concentrations of AZT and AZT nucleotides were higher than after incubation with an equipotent concentration of AZT over the entire incubation period. After incubation with 1 and 10 pg AZT- 43 DMDOPE/ml maximum intracellular concentrations of AZT and AZT nucleotides of 0.62 and 4.74 ng/10 6 cells respectively were reached whereas after incubation with 0.03 and 0.3 pg AZT/ml concentrations of 0.10 and 0.47 ng/10 6 cells respectively were detected.
In order to separately quantify AZT and AZT nucleotides after incubation with BM 21.1290 Na all intracellular concentrations of AZT and AZT nucleotides were determined with and without treatment with alkaline phosphatase. These experiments were carried out on stimulated human
PBL.
For this cell suspensions with a density of 106 cells/ml containing 1 and 10 pg AZT-DMDOPE/ml were incubated for 6 and 24 h at 37°C and 5 CO 2 The maximum intracellular concentrations of free AZT after incubation with 1 and 10 Ag AZT-DMDOPE/ml were detected as 0.03 and 0.15 ng/10 6 cells respectively after 6 and 24 h respectively (Fig.
If after treatment with alkaline phosphatase the phosphorylated intracellular AZT nucleotides x were additionally measured then maximum concentrations of 3.85 and 6.41 ng/10 6 cells respectively were found after 24 h.
b. Determination of the intracellular concentrations of AZT andlor AZT nucleotides in P3X63AS8,653 cells after incubation with AZT-DMDOPE and AZT The determination of the intracellular concentrations of AZT and AZT nucleotides in 44 thymidine kinase deficient cells should yield more information about the intracellular cleavage of the test substance AZT-DMDOPE. Intracellular AZT cannot be converted into AZT-MP and thus into the therapeutically effective AZT-TP due to a lack of thymidine kinase (TK) in these cells. The determination of the intracellular concentrations of AZT and AZT nucleotides after incubation with AZT-DMDOPE could thus give a further information on the cleavage of the test substance.
For this 1 x 107 cells in RPMI 1640 complete medium .9 I containing 0.03 or 0.3 gg AZT/ml and 1 or 10 Ag .9 AZT-DMDOPE/ml were incubated for 6, 24 and 48 h in @9 tissue culture plates (60 x 15 mm). After extracting the AZT and AZT nucleotides and treatment with alkaline phosphatase the concentration of AZT was determined with the aid of the radioimmunoassay.
The graphical plot of the experimental results shows that the intracellular concentrations of AZT and AZT nucleotides were considerably higher with ~AZT-DMDOPE than after incubation with equipotent "-:concentrations of AZT. Thus after 6 h incubation with 1 and 10 Ag AZT-DMDOPE/ml maximum concentrations of AZT and AZT nucleotides of 0.55 and 4.40 ng/10 6 cells respectively were achieved (Fig. 9).
The maximum intracellular concentrations of AZT and AZT nucleotides after incubation with 0.03 and 0.3 AZT/ml were determined as 0.03 and 0.31 ng/10 6 cells respectively.
45 In order to quantify the proportion of phosphorylated AZT the intracellular concentrations of AZT and AZT nucleotides were compared before and after treatment of the cellular extracts with alkaline phosphatase. The substance level of phosphorylated AZT after incubation with 1 and Ag AZT-DMDOPE/ml yielded values in this case of 0.48 and 2.42 ng/10 6 cells respectively (Fig. If only AZT was measured then maximum concentrations of 0.05 and 1.10 ng/ml respectively were reached after 24 h. The differences in the intracellular concentrations of AZT nucleotides after incubation with AZT and AZT-DMDOPE can be explained by an intracellular cleavage of the test substance AZT-DMDOPE to AZT-MP.
c. Kinetics of the decrease of AZT and AZT nucleotides in stimulated human PBL after incubation with AZT- DMDOPE and AZT Stimulated human PBL were incubated for 24 h with 0.3 Aq AZT/ml or 10 &g AZT-DMDOPE/ml. Subsequently 1 x 10 7 cells were transferred into tissue culture flasks (50 ml/25 cm 2 containing RPMI 1640 complete medium I. After 1, 3, 6, 24 and 48 h AZT and AZT nucleotides were extracted from the cellular matrix. After treatment with alkaline phosphatase the concentration of AZT was determined with the aid of a radioimmunoassay.
The graphic representation of the experimental results shows that after incubating the cells with AZT the intracellular concentrations of AZT and AZT nucleotides decrease rapidly and reach a constant 46 value of 0.40 ng/10 6 cells after 6 h. In contrast in the incubation with an equipotent concentration of AZT-DMDOPE the AZT and AZT nucleotides decrease less rapidly and after 3 h reach a substantially higher concentration of 1.80 ng/10 6 cells which remains constant over 48 h (Fig. 11). Since nonphosphorylated AZT was removed from the cell by washing several times with PBS, the intracellular concentrations are mainly attributable to phosphorylated AZT nucleotides. Thus an incubation with AZT-DMDOPE provides the cell with a higher concentration of phosphorylated AZT compared to an equipotent concentration of AZT. These large differences in the cellular concentrations of AZT nucleotides after incubation with AZT-DMDOPE and AZT can be explained by a direct intracellular cleavage of the thioether-lipid-AZT conjugate AZT- DMDOPE to AZT-MP and the corresponding thioether lipid moeity DMDOP.
SSramle 9
S
Characterization of the AZT-DMDOPE cleavage enzyme/ enzyme system (LCE)
OS
0S Investigations on the enzymatic cleavage of AZT-DMDOPE by cell homogenates of human PBL and CEM-SS cells showed that AZT-DMDOPE is metabolized to DMDOP and AZT-MP. The characterization of the LCE was the object of the following investigations. Thus the dependency of the enzymatic cleavage of AZT-DMDOPE on the protein concentration, the incubation period and divalent metal cations was examined. The dependency of the apparent Michaelis-Menten parameters KM and vmax on various 47 substrate concentrations is determined in enzyme kinetic experiments.
For these experiments an enzyme assay is established in which AZT-DMDOPE and 14 C]-AZT-DMDOPE were used as the substrate (example In protein and time dependent measurements 5 nmol AZT-DMDOPE and 0.98 nmol (1.67 kBq) 14 C]-AZT-DMDOPE were used per mixture. In the case of substrate-dependent conversions a stock solution of 0.98 nmol 14 C]-AZT-DMDOPE and 0.7 nmol AZT-DMDOPE/80 Al was used. The various substrate concentrations were subsequently adjusted by continuously doubling or halving the volume of the substrate solution.
Cell homogenates, cytosol and membrane fractions of known protein concentrations from various human cells can for example be used as the enzyme source.
a. Isolation and quantification of DMDOP after enzymatic cleavage of AZT-DMPOPE The quantification of the enzymatic cleavage of AZT-DMDOPE was carried out by two-step extraction of the metabolite DMDOP with n-heptane and subsequent adsorption of residues of the substrate AZT-DMDOPE on silica gel 60 H.
For this the reaction mixtures were admixed with 2propanol and n-heptane in the process of which the DMDOP which is less polar than the substrate accumulates in the n-heptane phase. The n-heptane phase was then admixed with silica gel 60H which adsorbs residues of the substrate AZT-DMDOPE. The n-heptane phase was afterwards separated from the 48 silica gel by centrifugation, transferred to 3 ml aqua luma and the amount of radioactively labelled DMDOP contained therein was determined over a period of 5 min in a liquid scintillation analyzer.
From these results it was possible to calculate the percentage conversion of the substrate AZT-DMDOPE to DMDOP and the specific activity of the LCE. In this case the specific activity is defined as the amount of DMDOP which is formed per minute per 1 mg protein of the cell preparations used.
In order to check the selective extraction of DMDOP from the reaction mixture, optimization experiments were carried out with membrane fractions of stimulated human PBL. In this case the protein concentration was 100 ag/mixture. Mixtures without protein were tested in parallel. After the extraction the n-heptane phase was evaporated in an
N
2 stream and the residues were taken up in a mixture of methanol and ethyl acetate v/v).
The analysis by thin layer chromatography was carried out in the IBA system. The reaction value -in this case only allowed the parent substance 14 C]-AZT-DMDOPE to be detected. In the reaction mixture a substance peak was detected at the end of the mobile solvent front which could be identified unequivocally as the metabolite 1 4 C)-DMDOPE on the basis of its Rf value (Fig. 12). The peak which runs immediately in front of the DMDOP substance peak cannot be allocated to either of the known compounds. It is presumably a compound which is formed by oxidation of the sulphur in the thioether lipid moeity of DMDOP. Hence an extraction with nheptane enables the enzymatic cleavage product 49 DMDOP to be isolated from the enzyme assay. A simple and rapid quantification of the metabolite DMDOP released from AZT-DMDOPE was thus ensured.
b. Cleavage of AZT-DMDOPE by cell homogenates, membrane and cytosol fractions of stimulated human
PBL
In a first series of experiments the enzymatic cleavage of AZT-DMDOPE by cell homogenates and by .".cytosol and membrane fractions of stimulated human PBL was examined.
After determining the protein concentration in cell homogenates, cytosol and membrane fractions with the aid of the bicinchoninic acid (BCS) test 0.025 0.20 mg protein/mixture were used. The reaction mixtures were incubated for 2 h at 370C, subsequently the product DMDOP was isolated from the mixtures by extraction with n-heptane and the specific activity of the LCE was determined (Fig.
13).
The AZT-DMDOPE-cleaving activity in cell homogenates and in cytosol fractions of 6.48 0.38 (n 6) and 1.65 0.40 pmol mg- 1 min- 1 (n 6) was 1.59-fold and 6.3-fold smaller than the specific activity in the membrane fractions which was 14.23 0.70 (n 6) pmol mg- 1 min- 1 Apparently there is an enrichment of the LCE when the membrane fractions are isolated. Based on these results membrane fractions were used in the subsequent experiments for determining the enzymatic parameters of the LCE.
50 The membrane fraction which was obtained by mechanically disrupting the cells and subsequent ultracentrifugation of the cell homogenate is a mixture of fragments of the plasma membrane and nuclear membrane as well as membranes of the cell organelles.
It was possible to differentiate the various membrane fragments by ultracentrifugation of the cell homogenate in Percoll density gradients followed by a subsequent characterization by the plasma membrane marker alkaline phosphatase.
*fo.
For the isolation of the plasma membranes 2 ml of a cell suspension of stimulated human PBL with a cell density of 2.5 x 1 0 8 cells/ml was disrupted by freezing and thawing three times and fractionated by centrifugation in a Percoll density gradient. fractions (1 ml) were taken from the centrifugate, residues of the separating medium were removed and the activity of alkaline phosphatase in the individual fractions was measured at 305 nm using p-nitrophenylphosphate as the substrate.
The highest activity of alkaline phosphatase was determined in fraction 8. The absorbance of the remaining fractions was much less. This indicates an enrichment of the plasma membranes in fraction 8. The specific LCE activity was determined in each fraction after determining the protein concentration. For this the protein concentration was adjusted to 0.05 mg/mixture and the reaction mixtures were incubated for 2 h at 37"C. The highest specific LCE activity of 4.40 pmol mg- 1 51 min 1 was determined in this case in fraction 8.
In all other fractions lower specific activities of 1.10 1.90 pmol mg 1 l min 1 were measured (Fig.
4.32). The highest specific activity of LCE was thus determined in the fraction which at the same time had the highest activity of alkaline phosphatase. Hence this finding shows that LCE occurs in the fraction with the highest amount of fragments of the plasma membrane.
c. Dependency of the specific LCE activity on the protein concentration In further investigations it was intended to determine the dependency of the turnover of AZT- DMDOPE on increasing protein concentrations of the cell preparations.
S For this membrane fractions of stimulated and nonstimulated human PBL and of human blood monocytes were used. The conversion of AZT-DMDOPE to DMDOP by the membrane fractions of stimulated human PBL exhibited a linear behaviour when using 0.025 0.2 mg protein/mixture (Fig. 14). The specific LCE activity was 14.23 0.7 pmol mg min- 1 (n 6) (Fig. 16). When AZT-DMDOPE was converted by proteins of the membrane fraction of non-stimulated human PBL there was a linear relationship only in the concentration range up to 0.05 mg protein/mixture. At higher protein concentrations there was no longer a linearity. The specific LCE activity which was calculated from the turnovers in the linear range was 1.45 0.32 pmol mg
I
min" 1 52 (n 6).
In order to determine the specific LCE activity in human blood monocytes these were isolated in hypertonic density gradients. Monocytes have a density of 1.068 g/ml on average which is somewhat lower than that of lymphocytes with 1.070 g/ml.
This difference in the density is however, very small so that it is not possible to separate these blood cells in an isotonic gradient.
0. Under hypertonic conditions lymphocytes lose water more rapidly than monocytes resulting in an increase in their density. Hence it is possible in a hypertonic separating medium to isolate monocytes from whole blood or leucocyte rich plasma. It is also possible to isolate human blood monocytes from buffy coat. For this mononuclear cells are separated under isotonic conditions in a density gradient and monocytes are subsequently separated in tissue culture flasks (175 cm 2 /800 ml) on the basis of their adherence.
"With the aid of flow cytofluorometric analysis it was possible to detect a stimulation of these cells by adherence after determination of size and granularity as well as after specific antibody staining. As a result of this stimulation the monocytes which were isolated by adherence to the bottom of tissue culture bottles were denoted monocytes/macrophages (stimulated monocytes) in the following.
53 In the enzymatic turnover of AZT-DMDOPE by the membrane fraction of human monocytes which had been stimulated during the course of their isolation by adherence, a linear dependency was found up to a concentration of 0.1 mg protein/mixture (Fig. The specific activity was 8.8 0.26 pmol mg- 1 min- 1 (n 4) (Fig. 16). When the monocytes were isolated directly in a hypertonic density gradient then there was a linear behaviour of the enzymatic cleavage only up to a protein concentration of 0.025 mg protein/mixture. At higher protein concentrations the substrate turnover was almost constant. The specific activity was calculated from the linear range analogously to the turnover with the membrane fractions of non-stimulated human PBL and was 3.2 0.60 pmol mg 1 min- 1 (n 4).
o In summary it was established that higher specific LCE activities were observed when AZT-DMDOPE was converted by enzymes of the membrane fractions of stimulated human PBL and monocytes than when AZT- DMDOPE was converted by membrane fractions of nonstimulated cells (Fig. 16).
d. Dependency of the specific LCE activity on the incubation period The enzymatic cleavage of the experimental substance AZT-DMDOPE in relation to the incubation period was carried out with membrane fractions of stimulated human PBL as the enzyme source. The reactions mixtures were incubated for 0.5, 1, 2, 3 and 6 h at 37*C and subsequently the amount of the metabolite DMDOP was determined. After plotting the 54 experimental results on a graph it was possible to show that the conversion of AZT-DMDOPE to DMDOP is linear over the entire period of incubation of 6 h (Fig. 17).
e. Dependency of the specific LCE activity on divalent metal cations The dependency of the enzymatic cleavage of AZT- DMDOPE by LCE on divalent metal cations was examined with membrane fractions of stimulated human PBL as the enzyme source.
The divalent metal cations were used at a concentration of 2 mM in the LCE assay. The protein concentration was fixed at 0.068 mg/mixture.
An experiment with EGTA was carried out in parallel. EGTA is a specific complexing agent for Ca 2 which is essential for the activity of phospholipase C. After an incubation time of 2 h at 37"C the conversion of the parent substance was measured and the specific activity of the LCE was determined (following table).
55 Table:. Dependence of the cleavage of AZT-DMDOFEB by membrane fractions of stimulated human PBL on EGTA and divalent metal cations (mean± SD, n 3 determinations) Ef fector DMDOP Specific activity Inhibition [2.3M] Lpmol]3 (pmol mg-i mn-i] Ilk] NOTA 84.96 ±11.18 10.41 ±1.37 M0 Cad1 2 50.50 2.13 6.19 ±0.26 41.0 xqc1 2 84.36 ±17.02 10.34 ±2.08 0 2nC1 2 0 0 100.0 Mind1 2 4.7S 0.63 0.58 0.08 94 1 The highest specific activity achieved when using EGTA and MgCl 2 Was 10.41 1.37 (n 3) and 10.34± 2.08 pmol mg- 1 min- 1 (n respectively. Thus it was not possible to detect any inhibition by MgC1 2 within the limits of the measurement accuracy. When Cad1 2 was used in the enzyme assay a specific activity of 6.19 0.26 pmol mg- 1 inr 1 (n 3) was determined. In comparison with the mixture containing ECTA this means an inhibition of the LCE activity of 41.0 0.25 (n In the case of ZnCl 2 no DMDOP was detectable within the limits of the measurement accuracy. ZnC1 2 and lcgCl 2 led to a total inhibition of the conversion of AZT-DMDOPE to DMDOP and AZT-MP.
56 f. Determination of the AR9&Liwnt_ Michaelis -Menten paramleters KMan aVpx In order to deter-mine the apparent Michael is-Menten parameters for the LCE the dependence of the enzymatic cleavage of AZT-DMflOPE on increasing substrate concentrations was examined.
Table: Apparent Michael is-Menton par-ameters KM and vm,-L of the LCE in stimulated and non-stimulated human PilL and in human monocytes and monocytes/ macropha ges (stimulated monocytes) [pmol mg inl-i[m nlon-timulated PDL 2.03 ±0.19 5.51 0.99 stimulated human PBL 15.29 ±0.37 2.26 0.16 human monocytes 1.61 ±0.37 12.056 4.13 human 0.09 ±0.44 4.63 ±0.45 monocytes /macrophages (stimulated monocytes) For this membrane fractions of stimulated and nonstimulated human PBL as well as human lnonocytes and monocytes/macrophageg (stimulated monocytes) were used. The reaction mixtures contained a constant protein amount of 0.068 mg/mixture and were incubated for 2 h at 37 0 C. Subsequently the amount of the metabolite DMDOP was determined.
57 Example Experimental animals In order to determine the pharmakokinetic parameters of the experimental substance AZT-DMDOPE, in vivo experiments were carried out on female Balb/c mice (Charles River Wiga, Sulzfeld; Bormholtgard, Ry (Denmark); Iffa Credo, L'Abresle (France)). The animal deliveries were examined before the experiment started for virus antibodies (mice hepatitis virus, reo virus, paro virus). In the experiments only animals were used which had a negative antibody titre.
Keeping the experimental animals i The animals were kept in fully air-conditioned animal cages at a room temperature of 22 24°C, a relative air humidity of 50 70 and a day-night rhythm of 12 hours. Laminar flow boxes ensured that the air in the animal cage was exchanged 15 20 times per hour. The animals were administered a standard diet (Ssniff, Soest) and water ad libitum via a drinking bottle.
Example 11: Cell culture methods a. Cryopreservation of cell lines For the cryopreservation, cells were adjusted to a cell density of 5 x 10 6 cells/ml in RPMI 1640 medium and sedimented for 10 min at 1600 rpm in a 58 Minifuge T. Subsequently the cell sediment was taken up in an equal volume of cryopreservation medium. After resuspension of the cell sediment 1 ml of the cell suspension was transferred in each case to cryo-tubes (1.8 ml) and frozen for 24 h at The cryo tubes were transferred to a thermocontainer containing liquid N 2 for the final storage.
Cryopreservation 60 RPMI 1640 medium medium 0.05 mM 2-mercaptoethanol 100 U/ml penicillin 100 Ag/ml streptomycin foetal calf serum
DMSO
b. Type culturinq and growth conditions Culturing of CEM-SS cells The suspension cell line CEH-SS was cultured in RPMI 1640 complete medium I. For this 1 x 10 7 cells •were transferred to a tissue culture flask (83 cm 2 /260 ml) containing 50 ml medium. Subsequently ~the culture was incubated for 3 days at 370C and
CO
2 For the cell passage the cells were removed from the tissue culture flask with a sterile pipette and sedimented for 10 min at 1600 S" rpm in a Minifuge T. After resuspending the cells in culture medium and determining the cell number by eosin staining in a Neubauer counting chamber, 1 x 10 7 cells were used for passage into 50 ml fresh medium.
59 Culture of P3X63Acf8.653 gells P3X63Ag8.653 cellS Were cultured as a mfonolayer in RPM'I 1640 complete medium 1. For this 5 x 106 cells were transferred to a tissue culture flask (83 cm 2 /260 ml) containing 40 ml medium. The cell culture was incubated for 5 days at 37 0 C and 5 C0 2 until the bottom of the tissue culture flask was covered with a confluent, cell monolayer. For the cell passage the adherent cells were mechanically detached from the bottom of the flask using a cell scraper and subsequently sedimented by centrifugation for 10 min at 1600 rpm in a Minifuge T. After resuspendinig the cells in culture medium the cell number was determined by eosin staining in a Neubauer counting chamber and 5 x 106 cells were ~.used for passage into 40 ml fresh medium P. eterminini the cell c-ount Determination of the cell count by eosin stainingT in a Neubauer counting chamber (Lindl and Bauer.
1989, "Zell und Gewebekultur". p. 75-90, Gustav Fisher Verla-Stuttgart, New YorI For the determination of the cell count in suspension and monolayer cultures a cell concentration of 0.1 1 X 106 cells/ml was adjusted in PBS and a portion of this cell suspension was admixed with 20 gl eosin.
Subsequently the cell suspension was carefully admixed with a pipette, incubated for 2 min at room temperature and transferred to a Neubauer counting chamber. The number of cells was determined 60 immediately after the incubation period in a reverse phase contrast microscope whereby the cells were counted within four large squares. In this process vital cells were not stained in contrast to avital cells. Weakly stained cells were considered to be avital.
The number of vital cells/ml was derived from the number of non-stained cells of the four large squares multiplied by the chamber factor of 10 4 and the dilution factor of the cell suspension in the eosin solution.
Determination of the cell number by electronic counting in a Coulter Counter (Lindl and Bauer.
1989) The measurement of the cell number in a Coulter counter is based on the cells flowing between two platinum electrodes. If a cell passes through the opening of the two electrodes then the resistance of the current flowing through the electrodes changes proportionally to the size of the cell.
This generates a voltage impulse which is recorded thus enables the cells to be quantified.
In order to determine the number of cells 25 Al of the cell suspension was transferred to 3 ml Iso- Osmol a carrier liquid for the Coulter counter and injected into the apparatus 61 in this type of cell counting the total cell number of the cell suspension is determined. it is not possible to differentiate between vital and avital cells.
d. Detachment of adherent cells by trvvsinlEDTA Adherent cells could be detached enzymatically f rom.
the bottom of the respective culture vessel by treatment with trypsin/EDTA. For this the culture supernatant was carefully decanted and the cells were washed twice with prewarmed (379C) culture medium. In order to detach the cells from the bottom of the culture vessel these were incubated f or 5 min with a trypsinfEDTA solution (1 x) at room temperature. The volume of trypsin/EDTA solution was selected in this case such that the bottom of the respective culture vessel was covered with 2 3 mm of the enzymatic solution. At the end of the incubation period the cells were detached from the bottom by carefully shaking the flask, removed from the culture vessel and admixed with an X. equal volume of RPMI 1640 complete medium I.
Afterwards the cells were sedimenv-ad for 10 min at 1600 rpm in a Kinifuge T. The supernatant was discarded, the cell sediment was resuspended in cold PBS and again centrifuged under the same conditions. The cells were then resuspended in culture medium or buffer depending on their further use.
62 e. Isolation of mononuclear cells frpom hman_blod Isolation of human PBL from buffv coat by centrifugation in isotonic density gradients 1968. Scand. J._Clin. Lab. Invest. 21 (SuLpl 97). 77-89: BvSum 1976. Scand. J. Clin. Lab.
Invest. 5 (Suppl. 5-15) In order to isolate human PBL the buffy coat of healthy donors was mixed and diluted with RPMI 1640 complete medium II in a volume ratio of 1:2. 20 ml cold lymphocyte separation medium was placed in polypropylene tubes (50 ml) and carefully covered with a layer of 15 ml of the diluted buffy coats.
The fractionation was carried out by centrifugation for 30 min at room temperature and 1600 rpm (acceleration 4/brake 4) in a Minifuge T. The *readily visible band between the sample and separation medium was isolated with the aid of a Combitip pipette. The collected fractions were sedimented for 10 min at room temperature and 1600 rpm to remove the separation medium. The supernatant was decanted and the cells were washed three times with RPMI 1640 complete medium II or cold PBS depending on their use.
Stimulation and culture of human PBL After isolation of human PBL by means of centrifugation in isotonic density gradients it was possible to set up peripheral lymphocyte cultures.
Normally blood cells die relatively rapidly in culture. However, lymphocytes can be kept in culture for several generations. In order to 63 achieve a proliferation of these cells they were stimulated with the mitogen PHA-M, the lectin extract from the red scarlet runner (Phaseolus vulgaris) For the stimulation the isolated cells were adjusted to a cell density of 1 x 106 cells/ml in RPMI 1640 complete medium II in tissue culture flasks (175 cm 2 /800 ml) and incubated for 72 h at 37°C and 5 CO 2 In order to avoid the cell density becoming too high, the cell suspension was diluted after 24 h with culture medium in a volume ratio of 1:1. After completion of the stimulation period the mitogen PHA-M was removed by washing three times with RPMI 1640 complete medium II. For the cell expansion the stimulated lymphocytes were adjusted to a cell density of 5 7 x 106 cells/ml in RPMI 1640 complete medium IV and cultured for 24 hours.
Preparation of leucocyte-rich plasma from whole blood (Byum, 1968) Venous fresh blood from healthy donors was admixed with sterile 10 EDTA solution in a volume ratio of 49:1 to prevent coagulation. In order to achieve a rapid mixing of these components small volumes of 8 9 ml blood were collected from the donor in polystyrene tubes (14 ml). In order to prepare leucocyte-rich plasma, EDTA-blood and dextran 75 were mixed in a volume ratio of 10;1.
The two components were thoroughly mixed and the erythrocytes were sedimented for a period of 60 min at room temperature. Subsequently the almost clear 64 supernatant, the leucocyte-rich plasma, was carefully removed and used to isolate monocytes.
Isolation of human monocytes from leucocyte-rich plasma by centrifugation in a hypertonic density gradient (Bbvum. 1983, Scan. J. Clin. Lab. Invest.
17. 429-436) It was possible to isolate non-stimulated monocytes from whole blood or leucocyte-rich plasma in a hypertonic solution. For this 3 ml of the hypertonic separation medium NycoPrep 1.068 was placed in polystyrene tubes (14 ml) and carefully covered with a layer of 6 ml leucocyte-rich plasma.
The tubes were closed and centrifuged for 15 min and 600 x g (acceleration 4/brake 4) in a Minifuge e T. After centrifugation the clear plasma phase was .999 S. removed and discarded up to 5 mm above the broad diffuse band. The remaining plasma phase and half o of the broad diffuse band was removed and the cells were sedimented by centrifugation in a Minifuge T for 7 min under the same conditions. The cell sediment was subsequently resuspended and washed twice in 6 ml wash solution to remove residues of the separation medium.
Nycoprep 1.068 13 Nycodenz 0.58 NaCl 5 (mM) Tricine/NaOH pH 7.4 density 1.068 0.001 g/ml osmolarity 335 5 mOsm 65 Wash solution 0.9 NadC 0.13 EDTA 1 BSA (fraction V) Isolation of human monocytes from buffv coat by adherence (Andreesen et 1983. J. Immun. Methods 56. 295-304) Mononuclear cells were isolated from the buffy coat by centrifugation in isotonic density gradients and washed three times with serum-free RPMI 1640 medium. In order to isolate the monocytes the cells were adjusted to a cell density of 5 x 106 cells/ml in tissue culture flasks (175 cm 2 /800 ml) containing 20 ml RPMI 1650 complete medium II and incubated for 45 min at 37°C and 5 CO 2 Nonadherent cells were removed by decanting the medium after the incubation was completed. The cell layer at the bottom of the tissue culture flask was o. ~subsequently washed twice with pre-heated (37°C) serum-free RPMI 1640 medium to remove residues of suspension cells. The cell layer was admixed with 30 ml RPMI 1640 complete medium V and incubated for a further 24 h under the same conditions. The adherent monocytes were detached by treatment with trypsin/EDTA.
9*S
CC
9 66 Example 12: imnmunological methods a. Flow cytpfluorometric analysis of cell subpopulations from human blood by direct and sequential antibody staining For the flow cytofluorometric analysis the cells to be characterized were adjusted in FACS-PBS to a cell density of 1 x 107 cells/mi. The antibody stock solutions were diluted with FACS-PBS in a volume ratio of 1:50. For the direct and sequential antibody staining 50 1l of the cell suspension (1 x 107 cells/ml) was placed in the wells of a microtitre plate with a conical bottom.
9S be eq. 0 6.
C.
V C
O*
C r Sn.
)00 Antibodies for the flow cvtofluorometric analysis of mnnonuclear human blood cells en.
a 0* S Sep.
0* S. *0 8b
C
Antibodies Specificity Fluorescent Antibody dye staining anti-Leu-12 Leu-12 (CD19) PE direct (CD19) B lymphocytes anti-Leu-5b Leu-5 (CD2) FITC direct (CD2) T lymphocytes anti-monocyte gp 55 sequential (CD14) monocytes macrophages anti-leu-2a cytotoxic T FITC direct (CD8) lymphocytes anti-leu-3a helper T PE direct (CD4) lymphocytes 67 Subsequently the cell suspension was admixed with an equal volume of the antibody which in the case of a direct antibody staining is coupled to a fluorescent dye. The mixtures were incubated for min at 4 0 C and subsequently centrifuged at 1400 rpm and room temperature for 2 min in a Minifuge T to sediment the cells.
The supernatants were removed and the cell sediment was resuspended in 200 g1 FACS-PBS and centrifuged again as described above to remove residues of nonbound antibody. The supernatant was removed and the cell sediment was resuspended in 100 p1 FACS-PBS in the case of a direct antibody staining. In the case of the sequential antibody staining the cell sediment was taken up in 100 pl of a solution of the goat-anti mouse Ig-R-PE (3 pg/ml) antibody elabelled with phycoerythrin after completion of the centrifugation of the cell sediment and incubated for 30 min at 4°C. Residues of the non-bound antibody were removed as described by washing with 200 p1 FACS-PBS and the cell sediment was resuspended in 100 p1 FACS-PBS for the flow •cytofluorometric cell analysis.
0 b. Determination of the cell vitality by pronidium iodide staining in the flow cytofluorometric analyzer oooo
S.
The number of vital and avital cells can be determined by flow cytofluorometric analysis after staining the cells with the fluorescent dye propidiumn iodide. This dye is only taken up by avital cells. For this the cell suspension is 68 adjusted in PBS to a concentration of 0.5 1 x 106 cells/ml. 180 p] is taken from this cell suspension and admixed with 20 p1 of a propidium iodide solution (5 pg/ml). After an incubation period of min 100 Al of the solution was injected into the cell analyzer. The percentage distribution of the vital and avital cells could be determined by a statistical analysis in the Lysis II program of the cell analyzer.
c. Ouatitative determination of AZT in cell extracts by means of a 1 25 1-AZT radioimmunoassay A commercial 12 5I-AZT RIA test kit was used for the quantitative determination of AZT. Buffers and solutions were prepared according to the instructions of the manufacturer and used in the test kit. The reaction mixtures were pipetted according to the scheme (Tab.) shown below, carefully mixed and incubated for 2 h at room temperature. Subsequently 500 1l of the goat antirabbit antibody was added to precipitate the complex of 12 5I-AZT and the AZT antibody (from rabbits). The mixtures were mixed and incubated for a further 30 min at room temperature.
69 Pipetting schem~e of tho AZrdomruosa fir-the guantitative dentel non-binding zero standard sample standard standard zero standard 300 200 [All standard (4l] -200sample [Al] 200 1 25 1-AZT [pl) 100 100 100 100 AZT Antibody 100 100 100 [All The mixtures were subsequently centrifuged at 1000 x g for 20 min to sediment the precipitation complex. After decanting the supernatant the radioactivity of the sediment was measured for sec in a liquid scintillation analyzer. In order to determine the absolute value, the radioactivity of 100 pl 1 2 SI-AZT was determined under the same conditions after an incubation of 2 h at room temperature. The concentrations of AZT in the biological matrix were determined with the aid of a calibration curve which was established with standards from the test kit with a defined substance concentration.
70 .KmIMle 13: Characerization of the intracellular enzymatic cleavage of BM 21.1290 Na The results of the in vitro pharmakokinetic studies with AZT-DMDOPE and AZT in stimulated and non-stimulated human PBL as well as in thymidine kinase deficient P3X63Ag8.653 cells indicate an intracellular enzymatic cleavage of AZT-DMDOPE with a direct release of AZT-MP and of the corresponding thioether lipid moeity DMDOP.
It is intended to verify this intracellular enzymatic S"cleavage demonstrated in further experiments at a subcellular level by a direct detection of the Corresponding metabolites. Hence for an unequivocal characterization of the cleavage it was necessary to identify the previously known metabolites which can be formed from the parent substance AZT-DMDOPE. These include the substances DMDOP, AZT and AZT-MP as well as substances which are formed by oxidation of the sulphur -in the thioether lipid moiety of the parent substance
AZT-DMDOPE.
a. Development Qf methods for the determination of the Rf values of AZT-DMDQPE and its potential It was possible to identify the potential metabolites of AZT-DMDOPE with the aid of TLC. For this the non-radioactively labelled pure substances were analysed with the aid of various separation systems and subsequently the Rf values were determined.
71 For this the substances were dissolved at a concentration of 4 mg/ml in a mixture of ethyl acetate and methanol 5 Al of these solutions was transferred with the aid of a microcapillary to the stationary phase and analysed.
Thioether lipids such as AZT-DMDOPE, DMDOP as well as those which are formed by oxidation of the sulphur could be labelled with the aid of a reagent containing iodine which stains the thioether lipid moiety of these compounds. AZT and AZT-MP could only be made visible on the TLC plates with a fluorescence indicator on the basis of their absorbance in the ultraviolet range at 254 nm. AZT- DMDOPE, the substance with a thioether lipid moiety and a chromophoric group could be detected with both types of detection.
Three separation systems were established for the thin layer chromatographic analysis of the substances which differed with regard to their stationary and mobile phases.
In the first separation system, the IBA system, using silica gel 60 as the stationary phase the parent substance AZT-DMDOPE could be identified in addition to DMDOP. A mixture of 2-propanol:n-butyl acetate: redistilled water (10:6:4 v/v/v) was used as the mobile phase.
This separation system was developed further by using 2-propanol:n-butyl acetate redistilled water: glacial acetic acid v/v/v/v) as the 72 mobile phase (IBAE system). The substances AZT- DMDOPE and DMDOP could be separated in this case on silica gel 60 TLC plates as described for the IBA separation system. In addition AZT and AZT-MP could be separated with a fluorescence indicator in UV light at 254 nm and using a stationary phase of silica gel 60. Due to the AZT moiety it was also possible to detect the parent substance AZT-DMDOPE as well as its oxidation products.
After the substances were visualized on the TLC plate the Rf values were determined.
A further separation system was available to differentiate the substance AZT-DMDOPE from DMDOP.
For this a mixture of n-heptane and ethyl acetate v/v) was used as the mobile phase and silica 60 as the stationary phase. The detection was carried out with the reagent containing iodine already described above by staining the thioether lipid moiety of the substances.
In comparison to the IBA and IBAE system a considerably lower Rf value was determined for the substance DMDOP with this separation system. The reason for this is a unpolar mobile phase compared to the IBA and IBAE system consisting of n-heptane and ethyl acetate. The detection limit of the substances after thin layer chromatographic analysis and detection with an iodine-containing reagent was determined to be 0.1 gg in all separation systems.
73 in suimmary it can be stated that with the aid of the described thin layer chromatographic separation systems it is possible to unequivocally characterize all substances via their Rf values which come into consideration according to the present state of knowledge as potential metabolites of AZT-DMDOPE.
Rf values of AZT-DMDOPE and DNDOP after separation in the IBA system (2-propanolsn' butyl aaetate:redistilled water, 10:6:4.
v/v/v) with silica gel 60 as the stationary phase (meansISD, ni 6 determinations) Test su~bstance Rf value AZT-DXDOPE 0.68 5.2 X 0- DMDOP 0.93±0.00 74 a.
a. TbeRf values of A2T-DMDOPE, DMDQPp AZT and A.ZT- MP after separation in the IBAR system (2propanol: n-butyl acetate: rodistilled water:glacial acetic acid, 3:5:1:1v v/v/v/v) with silica gel 60 and 60 P 254 as the stationary phase (means t SD, n =6 det erminat ions) Test substance Rf value AZT-DIIDOPE 0.57 I X 0- DEDOP 0.99 0.00 AZT 0.85 ±0000 k2T-NP 0.14 ±0.00 b. Enzymatic cleavage of AZT-DMDOPE by call homoggenates of stimulat-ed anid non-stimulated human PEL and CM-SS cells The cleavage of AZT-DMDOPE was exazined with the aid of an enzyme assay using 1 4 C]-AZT-DMDOPE and AZT-DMDOPE as the substrate. Firstly cell homogenates of CEM-SS cells and human PBL were used as the enzyme source.
r- 75 9 9 *999 9.
9 9 9 9.
9 *999 Table Rf values of AZT-DMDOPE and DEDOP after thin layer chromatographic separation in the HE system (n-heptane;ethyl acetate, 4:1, v/v) with silica gel 60 as the stationary phase (means±SD, a 6 determinations) Test substance Rf value AST-DMDOPE 0.0 0.00 DMDOP 0.36 0.00 The latter were homogenized directly after their isolation after stimulation with PHA-M as well as in the non-stimulated state and used in the enzyme assay.
In order to prepare the cell homogenates, cell suspensions were mechanically disrupted in a glass homogenizer at a density of 5 x 10 7 cells/ml 50 mM Tris (pH 7.4) The cell disruption in this case was optically monitored in a reverse phase contrast microscope.
76 Table Rf values of the substances 1 -4 (Pig. 18) after 1, 3, 6 and 24 h incubation and enzymatic cleavage of 14 C]-AZT-DMDOPE by cell homogenates of 5 x L07 stimulated and non-stimulated human PBL and CEM-SB cells.
Separation in the IBA/IBAE system with silica gel 60 as the stationary phase (means±SD, n 4 determinations) o
S*
.o* o o e g oooo 0 Rf value Rf value Rf value Rf value substance I substance 2 Substance 3 Substance 4 CEM-SB 0.41 0.56 0.86 0.96±0.0 ±2.5 X 10- 2 ±2.5 x 10- 2 ±5.8 x 10- 3 stimulated 0.65 0.86 0.94±0.0 human PBL ±9.5 x 10- 3 +5.8 x 10- 3 non-stimulated 0.65 0.92 human PBL 1.7 x 10- 2 ±1.2 x 10- 2 Firstly 1 ml of this cell homogenate was used in the enzyme assay without prior determination of the protein concentration. 0.98 nmol 14
C]-AZT-DMDOPE
(1.67 kBq) was used as the substrate.
In order to ensure a substrate saturation a further nmol of the non-radioactively labelled compound was added. [1 4 Cj-AZT-DMDOPE carries the radioactive label in the thioether lipid moiety and thus enables an unequivocal identification of the potential metabolite 14
C]-DMDOP.
77 The mixtures were incubated for 1, 3, 6 and 24 h in a water bath at 37"C. A mixture without addition of cell homogenate was also carried out as a reference. Subsequently the cleavage products were extracted with diethyl ether:2-propanol v/v) and analysed by thin layer chromatography in the IBA and IBAE system (Fig.18). After the substances were separated the TLC plate was measured for min in a radio TLC analyzer to detect the radioactively labelled substances. It was finally possible to unequivocally identify the metabolites by determining the Rf values.
o Table Rf values of the substances 1 and 2 (Fig. 18) after 1, 3, 6 and 24 h incubation of [14C]-BM 21.1290 Na. Separation in the IBA/IBAE system with silica gel 60 as the stationary phase (meanstOD, n 4 determinations) a Rf value Rf value Substance 1 Substance 2 CEM-SS 0.415.7 x 10- 3 0.56±5.8 X 10- 3 a.
stimulated 0.65±9.6 x 10 3 human PBL non-stimulated 0.65±1.9 x 10- 3 human PBL The thin layer chromatographic analysis of the enzymatic cleavage products of AZT-DMDOPE by cell homogenates of stimulated and non-stimulated human 78 PBL was carried out by using the IBA separation system. In the subsequent experiments with the CEM- SS cell homogenate the analysis of the cleavage products was carried out in the IBAE separation system. Since the Rf values of the pure substances have been determined for both separation systems it was possible to directly compare the results to identify the substances after enzymatic cleavage of
AZT-DMDOPE.
After enzymatic cleavage of [14C]-AZT-DMDOPE by cell homogenates of stimulated and non-stimulated human PBL three unknown substances were detected in the TLC chromatogram. With Rf values of 0.65 X 10-2 (n 4) and 0.65 1.7 x 10-2 (n 4) for stimulated and non-stimulated human PBL the .fl.
substance peak 2 could be unequivocally assigned to the parent substance 14 C]-AZT-DMDOPE. The Rf -values of substance peak 4 of 0.94 0.00 (n 4) and 0.92 1.2 X 10-2 (n 4) were identical to the values of DMDOP which were determined by TLC analysis of the pure substances. Substance peak 3 having an Rf value of 0.86 5.8 x 10 3 (n 4) could not be allocated to any of the known 4substances. The results were confirmed by analysis of the enzymatic cleavage of [1 4 C]-AZT-DMDOPE by CEM-SS cell homogenates. Substance peaks 2 and 4 having Rf values of 0.56 2.5 x 10-2 (n 4) and 0.96 0.00 (n 4) could be unequivocally identified as AZT-DMDOPE and DMDOP respectively whereas substance peak 1 having an Rf value of 0.41 2.5 x 10 2 (n 4) could be assigned to an oxidation product. It was not possible to assign substance peak 3 even in the IBAE system after comparison of the Rf values (Fig. 18). Furthermore 79 it was possible to show that after incubation of 14 C]-AZT-DMDOPE only the parent substance was detected when the cell homogenate was not added (Fig. 18).
The thin layer chromatographic analysis of the reaction mixtures therefore proves that the substance DMDOP is released from the parent substance.
xample 14: 9- (B-D-Arabinofuranosyl) acid-(3-dodecylmercapto-2-decyloxy)-propyl ester (fludarabine conjugate) 34.8 g (0.07 mol) phosphoric acid (3-dodecylmercapto-2decyloxy)-propyl ester was dissolved in 130 ml absolute pyridine, admixed with 15 g methanesulfonic acid chloride under nitrogen and stirred for 3 hours at room temperature.
Then 20 g fludarabine was carefully added and the solution was stirred for a further 48 hours at room temperature. Fludarabine was synthesized analogously to J. Heterocyclic Chem. 16, 157 (1979).
After hydrolysis of the reaction mixture by adding 30 ml 1 M triethylammonium bicarbonate solution and stirring for 1 hour, the pyridine was removed in a vacuum and the residue was partitioned between 200 ml t-butylmethyl ether (MTB) and 150 ml water, the organic phase was separated and evaporated in a rotary evaporator.
80 The residue was purified chromatographically on RP-18 with methanol/0.02 M acetate buffer pH 4 8/2 as the eluant.
The fractions containing product were concentrated down to the water portion, extracted with MTB and the MTB phase was adjusted with sodium methylate solution against Friscolyt to pH 7.
After the solvent was removed by evaporation the residue was suspended in acetone, the amorphous precipitate was suction filtered and dried.
Yield: 23.7 g (43 Amorph. Rf 0.45 (TLC-mobile solvent: isopropanol/butyl acetate/water/ammonia 50/30/15/5).
*8 Example S. I 2-Chloro-2 -deo dadenosine-5' -phosphoric acid-(3dodecylmercapto-2-decvloxy)-propvl ester (cladribine conjiuate) go.
S
The cladribine conjugate was prepared in a 35 yield analogously to example 14 using 4.2 g phosphoric acid- (3-dodecylmercapto-2-decyloxy)-propyl ester, 3 g methanesulfonic acid, 100 ml pyridine and 2 g cladribine. Rf 0.41 (mobile solvent as in example 14).
Cladribine was prepared analogously to J. Am. Chem. Soc.
106, 6379 (1984).
81 r3- (2-Deoxy-B-D-erythro-pentgfuranosyl)1 67.9 r1.3ldiaLzeyin-S-olJ phosphoric acid- (3-dodecvlmercapto-2 -decvloxy-,triQpvl ester (1pentostatin coniugate) The pentostatin conjugate was prepared in a 27 yield analogously to example 14 using 4.2 g phosphoric acid- (3 -dodecylinercapto-2-decyloxy) -propyl ester, 3 g methanesulfonic acid, 100 ml pyridine and 2.1 g pentostatin. Rf 0.52 (mobile solvent as in example 14). Pentostatin was prepared analogously to J. Org.
Chem. 47, 3457 (1982) and J. Am. Chem. Soc. 101, 6127 (1979).
.:..*Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggest ion that that prior art forms part of the common general knowledge in Australia.

Claims (23)

1. Use of a covalent conjugate of a lipid derivative and a pharmacologically active substance wherein the conjugate is an active substance of formula L-B-D and L represents the lipid moiety, B is a valency dash, a phosphate bridge or a thiophospate bridge and D represents a pharmacologically active substance or B-D is an active substance phosphonate, wherein said conjugate is tested in vitro with a membranous enzyme, for cleavage into parts L and B-D, which enzyme is a lipid cleavage enzyme (LCE) complex free ofphospolipase C activity, wherein said enzyme cleaves the conjugate AZT-DMDOPE [(3'-deoxy-3'-azidothymidine)-5'-phosphoric acid-(3- dodecyl-mercapto-2-decyloxy)-propyl ester] into AZT-MP [3'-deoxy-3'-azido- thymidine-monophosphate] and DMDOP [(3-dodecyl-mercapto-2-decyloxy)-propanol] or the conjugate FLT-DMDOPE [(3'-deoxy-3'-fluorothymidine)-5'-phosphoric acid-(3- dodecylmercapto-2-decyloxy)-propyl ester] into FLT-MP [3'-deoxy-3'-fluoro-thymidine- 5 '-monophospate] and DMDOP or the conjugate 5-FU-DMDOPE phosphoric acid-( 3 -dodecylmercapto-2-decyloxy)-propyl ester] into 5-FU-MP :fluorouridine monophosphate] and DMDOP, to produce a pharmaceutical agent for the targeted release of the residue B-D in suitable target cells which have been tested to o* contain said LCE-complex to transport the conjugate or the residue -B-D through the cell membrane of these target cells resulting in a specific intracellular accumulation of the active residue -B-D.
2. Use as claimed in claim 1, wherein the covalent conjugate is cleaved essentially on or in the membrane or in the respective target cells. P:\OPER\TDO\ROCHE.DIV -24/8199 83
3. Use as claimed in claim 1, wherein the pharmaceutical agent reduces the side effects compared to the administration of the free residue -B-D or D not bound to a lipid.
4. Use as claimed in claim 1 or 2, wherein there is no significant cleavage of the conjugate in the plasma or the liver. Use as claimed in claim 1 or 2, wherein the target cells are human leucocytes, monocytes, macrophages, immunological cells, tumour cells or cells of the lymphatic system, of the kidney, spleen or brain. ooo Use as claimed in one of the claims 1-5, wherein the pharmaceutical agent is capable of increasing the intracellular concentration of the residue -B-D or D in the target cells by at least 10% compared to the administration of free -B-D or D not bound to a lipid residue. Use as claimed in claim 6, wherein the concentration of the active substance in the target cells is increased by at least Use as claimed in one of the claims 1-7, wherein the pharmaceutical agent is administered in a dosage unit which has a lower equivalent content of the conjugated substance -B-D or D compared to the form of administration containing the free non-conjugated substance.
9. Use as claimed in claim 8, wherein the equivalent content of the conjugated substance -B-D or D is less than 50% compared to the form of administration containing the free non-conjugated substance. Use as claimed in one of the claims 1-8 for the production of pharmaceutical agents for reducing organ toxicities of pharmacologically active substances. P:\OPER\TDO\ROCHE.DIV 24/8/99 -84-
11. Use as claimed in one of the claims 1-9 for the production of pharmaceutically agents for reducing the bone marrow toxicity of pharmacologically active substances.
12. Use as claimed in one of the claims 1-9 for the production of pharmaceutical agents for reducing the cardiotoxicity, nephrotoxicity, haemotoxicity, hepatoxicity or neurotoxicity of pharmacologically active substances.
13. Use as claimed in one of the claims 1-9 for the production of pharmaceutical agents for the targeted transport of pharmacologically active substances through the blood- brain barrier.
14. Use as claimed in claim 13, wherein the pharmacologically active substance accumulates in cells of the brain.
15. Use as claimed in claim 13 or 14, wherein the pharmacologically active substance is a therapeutic agent that can be administered orally.
16. Use of a covalent conjugate L-B-D as claimed in one of the claims 1-15, wherein L represents a residue of formula II R 1 -X--CH 2 I 2 (II) (CH2) m in which R' is a straight-chain or branched, saturated or unsaturated alkyl chain with 1-30 carbon atoms which can be optionally substituted once or several times by halogen, C 5 -C 7 cycloaklyl, phenyl, C 1 alkoxy, Ci-C 6 alkylmercapto,Cl-C 6 alkoxycarbonyl, C,-C 6 alkylsulfiyl or alkylsulfonyl groups, P:\OPER\TDO\ROCHE.DIV 24/8/99 R 2 is hydrogen, a straight-chain or branched, saturated or unsaturated alkyl chain with 1-20 carbon atoms which can be optionally substituted once or several times by halogen, C 5 -C 7 cycloalkyl, phenyl, C 1 -C 6 alkoxy, C 1 -C 6 alkylmercapto, C 1 -C 6 alkoxycarbonyl or CI-C 6 alkylsulfonyl groups X represents a valency dash, oxygen, sulphur, aminocarbonyl, oxycarbonyl, carboxyamino, carbonyloxy, a sulfinyl or sulfonyl group Y is a valency dash, aminocarbonyl, oxycarbonyl, carbonylamino, carbonyloxy, an oxygen or sulphur atom and m represents an integer between 1 and
17. Use of a covalent conjugate L-B-D as claimed in claim 16, wherein R' denotes a straight-chain or branched C 8 -CI 5 alkyl group which can be substituted by a Ci-C, alkoxy or a CI-C 6 alkylmercapto group and in particular represents a nonyl, decyl, undecyl, dodectyl, tridecyl or tetradecyl group.
18. Use of a covalent conjugate L-B-D as claimed in one of the claims 17 or 19, wherein R 2 denotes a straight-chain or branched C 8 -C 15 alkyl group which can be substituted by a C,-C 6 alkoxy group or a C-C 6 alkylmercapto group and in particular represents an octyl, nonyl, decyl, undecyl, dodecyl, tridecyl or tetradecyl group.
19. Use of a covalent conjugate L-B-D as claimed in one of the claims 16-18, wherein X denotes a sulphur atom, a sulfinyl or sulfonyl group. Use of a covalent conjugate L-B-D as claimed in one of the claims 16-19, wherein Y denotes an oxygen atom.
21. Use of a covalent conjugate L-B-D as claimed in one of the claims 16-20, wherein m denotes the numbers 1 or 2. P:\OPER\TDO\ROCHE.DIV 24/8/99 -86-
22. Use of a covalent conjugate L-B-D as claimed in one of the claims 16-20, wherein m equals 1, X is S, Y is O, R' is a dodecyl residue and R 2 represents a decyl residue.
23. Use of a covalent conjugate L-B-D as claimed in claim 16, wherein X and Y represent a valency dash, R 2 is hydrogen and R' represents a C 1 -C 30 alkyl chain which can be optionally substituted by C,-C 6 alkylmercapto.
24. Use of a covalent conjugate L-B-D as claimed in one of the claims 16-23, wherein :B represents a phosphate bridge of formula III (III), in which n denotes the numbers 1, 2 or 3 and Z is O or S, and A denotes either O, S or a valency dash. Use of a covalent conjugate L-B-D as claimed in one of the claims 16-24, wherein D or -B-D represents a pharmacologically active substance selected from the group of antiviral, antiretroviral, cytotoxic, cytostatic, antitumoral, immunosuppressive or immunostimulatory substances.
26. Use according to any of claims 1-25 wherein said LCE is obtainable from leucocytes, monocytes, tumour cells, kidney cells, lymphocytes, cells of the immuno/lymphatic system or macrophages.
27. Use according to any one of claims 1-26 wherein in the case of conjugates of the type L-B-D in which L represents a lipid-like residue, B represents a phosphate bridge or a thiophosphate bridge and D denotes a pharmacologically active substance or B-D represents an active substance phosphonate the enzyme induces a cleavage of the covalent bond between the lipid moiety L and the residue -B-D. I:\OPERVEH\RES CLMS 200[\AUG 200I\207955 CLKIS.DOC- 619/01 -87-
28. Use according to any one of claims 1-27 wherein the activity of the LCE during cleavage is at least 2-fold higher in activated immune cells, human leucocytes, monocytes, tumour cells, lymphocytes, kidney cells, cells of the immuno/lymphatic system or macrophages compared to non-activated cells.
29. Use according to any one of claims 1-28 wherein the activity of the LCE is inhibited by Ca 2 or Mn 2 Process for the production of pharmaceutical agents containing a conjugate L-B-D for the targeted release of substances -B-D or D in target cells, wherein these target cells have been tested to contain the lipid cleavage enzyme, to transport the conjugate or the residue through the cell membrane of these target cells resulting in a specific intracellular accumulation of the active residue B-D, which enzyme is a lipid cleavage enzyme (LCE) complex free of phospolipase C activity, wherein it cleaves the conjugate AZT-DMDOPE [(3'-deoxy-3'-azidothymidine)-5'- phosphoric acid-(3-dodecyl-mercapto-2-decyloxy)-propyl ester] into AZT-MP deoxy-3'-azido-thymidine-monophosphate] and DMDOP [(3-dodecyl-mercapto-2- decyloxy)-propanol] or the conjugate FLT-DMDOPE [(3'-deoxy-3 phosphoric acid-(3-dodecylmercapto-2-decyloxy)-propyl ester] into FLT-MP [3'-deoxy- and DMDOP or the conjugate [5-fluoridine-5'-phosphoric acid-(3-dodecylmercapto-2-decyloxy)-propyl ester] into FU-MP [5-fluorouridine monophosphate] and DMDOP, and wherein the conjugate is an active substance of the formula L-B-D and L represents the lipid moiety, B is a valency dash, a phosphate bridge or a thiophosphate bridge and D represents a pharmacologically active substance or B-D is an active substance phosphonate comprising the steps a) selecting a pharmacologically active substance b) producing a covalent conjugate of the pharmacologically active substance and a Slipid-like carrier molecule P:\OPERJEH\RES CLMS 200]\AUG 2001\2207955 CLMS.DOC 7/9/01
88- c) testing said conjugate in vitro for cleavage by said LCE complex d) and producing a pharmaceutical form of administration which contains the conjugate produced according to b) as well as further pharmaceutical auxiliary or carrier substances. 31. Method as claimed in claim 30, wherein D or B-D comprises the following individual compounds of the following substance groups: antibody; peptide; hormone; toxin; a substance that intercalates in DNA or RNA; a tubulin inhibitor; an alkylating agent; a ribosome inactivating substance; a tyrosine phosphokinase inhibitor; a differentiation inducer; hormone agonist; hormone antagonist; a substance which changes the pleitropic resistance towards cytostatic agents; a calmodulin inhibitor; a protein kinase C inhibitor; a P-glycoprotein inhibitor; a modulator .of mitochondrially bound hexokinase; an inhibitor of y-glutamylcysteins synthetase; a glutathione-S-transferase inhibitor; a superoxide dismutase inhibitor; an inhibitor of the hepatitis viruses A-E; antiinflammatory agent; anti-rheumatic agent; anti-phlogistic agent; analgetic agent; antipyretic agent; anti-arrhythmic agent; calcium antagonist; antihistaminic agent; inhibitor of phosphodiesterase; symphathomimetic agent; parasympathomimetic agent; antisense oligonucleotides or active substance o phosphonates; prodrugs thereof or derivatives capable of coupling. 32. Method according to claim 30 or 31 wherein said LCE is obtainable from leucocytes, monocytes, tumour cells, kidney cells, lymphocytes, cells of the immuno/lymphatic system or macrophages. 33. Method according to any one of claims 30-32 wherein in the case of conjugates of the type L-B-D in which L represents a lipid-like residue, B represents a phosphate bridge or a thiophosphate bridge and D denotes a pharmacologically active substance or B-D represents an active substance phosphonate the enzyme induces a cleavage of the covalent bond between the lipid moiety L and the residue -B-D. P:\OPERTEH\2207955 CLMS.DOC 2/5/01 -89- 34. Method according to any one of claims 30-33 wherein the activity of the LCE during cleavage is at least 2-fold higher in activated immune cells, human leucocytes, monocytes, tumour cells, lymphocytes, kidney cells, cells of the immuno/lymphatic system or macrophages compared to non-activated cells. Method according to any one of claims 30-34 wherein the activity of the LCE is 2+ inhibited by Ca 2 or Mn2+ 36. Lipid conjugate of formula I of type L-B-D selected from the group of the following substances: 2-fluoro-9-(b-D-arabinofuranosyl)adenine-5'-pphophoric acid-(3-dodecylmercapto-2- decyloxypropyl) ester, 2-chloro-2'-deoxyadenosine-5'-phosphoric acid-(3- dodecylmercapto-2-decyloxypropyl) ester, 3 2 -deoxy-b-D-erythropentofuranoxyl)- 3, 6 7 ,8-tetrahydro-imidazo-[4,5-d][1,3]-diazepin-8-ol-5'-phosphoric acid-(3- dodecylmercapto-2-decyloxy-propyl) ester. S37. A method for the targeted release of the residue -B-D in suitable target cells said method comprising administering a covalent conjugate of a lipid derivative and a pharmacologically active substance wherein the lipid moiety of the covalent conjugate targets the conjugate to the target cells, tissues or organs and transports the conjugate or the residue -B-D through the cell membrane of these target cells, the conjugate being cleaved by a membranous enzyme, which enzyme is a lipid cleavage enzyme (LCE) complex free of phospolipase C activity, wherein it cleaves the conjugate AZT-DMDOPE [(3'-deoxy-3'- 3 -dodecyl-mercapto.2-decyloxy)-propy ester] into AZT-MP [3'-deoxy-3'-azido-thymidine-monophosphate] and DMDOP [(3-dodecyl- mercapto-2-decyloxy)-propanol] or the conjugate FLT-DMDOPE [(3'-deoxy-3'- fluorothymidine)-5'-phosphoricacid-(3-dodecylmercapto-2-decyloxy)-propy ester] into FLT-MP [3'-deoxy-3'-fluoro-thymidine-5'-monophospate] and DMDOP or the L-M ?den~l-lar-hyiin-.maoh1.' DW O h P:\OPERUEH\2207955 CLMS.DOC 2/5/01 conjugate 5-FU-DMDOPE [5-fluoridine-5'-phosphoric acid-(3-dodecylmercapto-2- decyloxy)-propyl ester] into 5-FU-MP [5-fluorouridine monophosphate] and DMDOP, resulting in a specific intracellular accumulation of the residue -B-D wherein the conjugate is an active substance of formula -B-D wherein the conjugate is an active substance of formula L-B-D and L represents the lipid moiety, B is the valency dash, a phosphate bridge or a thiophosphate bridge and D represents a pharmacologically active substance or B-D is an active substance phosphonate. 38. The method according to claim 37, wherein the covalent conjugate is cleaved essentially on or in the membrane or in the respective target cells. 39. The method according to claim 37, wherein the pharmaceutical agent reduces the side effects compared to the administration of the free residue -B-D or D not bound to a lipid. i 40. The method according to claim 37 or 38, wherein there is no significant cleavage of the conjugate in the plasma or in the liver. 41. The method according to claim 37 or 38, wherein the target cells are human leucocytes, monocytes, macrophages, immunological cells, tumour cells or cells of the lymphatic system, of the kidney, spleen or brain. 42. The method according to one of the claims 37-41, wherein the pharmaceutical agent is capable of increasing the intracellular concentration of the residue -B-D or D in the target cells by at least 10% compared to the administration of free -B-D or D not bound to a lipid residue. 43. The method according to claim 42, wherein the concentration of the active substance in the target cells is increased by at least P:\OPER\JEH\2207955 CLMS.DOC 2/5/01 -91- 44. The method according to one of the claims 37-43, wherein the pharmaceutical agent is administered in a dosage unit which has a lower equivalent content of the conjugated substance -B-D or D compared to the form of administration containing the free non- conjugated substance. The method according to claim 44, wherein the equivalent content of the conjugated substance -B-D or D is less than 50% compared to the form of administration containing the free non-conjugated substance. 46. The method according to one of the claims 37-44 wherein the organ toxicities of pharmacologically active substances are reduced. 47. The method according to one of the claims 37-45 wherein the bone marrow toxicity of pharmacologically active substances is reduced. S- 48. The method according to one of the claims 37-44 wherein the cardiotoxicity, nephrotoxicity, haemotoxicity, hepatoxicty or neurotoxicty of pharmacologically active substances is reduced. 49. The method according to one of the claims 37-44 wherein the targeted release targeted transport of pharmacologically active substances through the blood-brain barrier. The method according to claim 49, wherein the pharmacologically active substance S* accumulates in cells of the brain. o 51. The method according to claim 49 or 50, wherein the pharmacologically active substance is a therapeutic agent that can be administered orally. 52. The method according to any of claims 37-51 wherein L represents a residue of formula P:\OPERUEH\2207955 CLMS.DOC- 2/5/01 -92- R 1 -X--CH 2 R2-Y H I -(II) (CH 2 )m_ in which R' is a straight-chain or branched, saturated or unsaturated alkyl chain with 1-30 carbon atoms which can be optionally substituted once or several times by halogen, Cs-C 7 cycloalkyl, phenyl, CI-C 6 alkoxy, C 1 -Cs alkylmercapto, CI-C 6 alkoxycarbonyl, Ci-C 6 alkylsulfinyl or CI-C 6 alkylsulfonyl groups, R2 is hydrogen, a straight-chain or branched, saturated or unsaturated alkyl chain with 1-20 carbon atoms which can be optionally substituted once or several times by halogen, C 5 C7 cycloalkyl, phenyl, C-C 6 alkoxy, C 1 -C 6 alkylmercapto, Ci-C 6 alkoxycarbonyl or C 1 C 6 alkylsulfonyl groups X represents a valency dash, oxygen, sulphur, aminocarbonyl, oxycarbonyl, carboxyamino, carbonyloxy, a sulfinyl or sulfonyl group Y is a valency dash, aminocarbonyl, oxycarbonyl, carbonylamino, carbonyloxy, an oxygen or sulphur atom and m represents an integer between 1 and 53. The method according to claim 52 wherein R 1 denotes a straight-chain or branched Cs- C 15 is alkyl group which can be substituted by a CI-C 6 alkoxy or a C 1 -C 6 alkylmercapto group and in particular represents a nonyl, decyl, undecyl, dodecyl, tridecyl or tetradecyl group. PA\OPERUEH\2207955 CLMS.DOC. 2/5/01 -93- 54. The method according to one of claims 53 and 55, wherein R 2 denotes a straight-chain or branched Cs-Cis alkyl group which can be substituted by a CI-C 6 alkoxy group or a Ci-C 6 alkylmercapto group and in particular represents an octyl, nonyl, decyl, undecyl, dodecyl, tridecyl or tetradecyl group. The method according to one of claims 52-54, wherein X denotes a sulphur atom, a sulfinyl or sulfonyl group. 56. The method according to one of claims 52-54, wherein Y denotes an oxygen atom. 57. The method according to one of claims 52-56, wherein m denotes the numbers 1 or 2. 58. The method according to one of claims 52-56, wherein m equals 1, X is S, Y is O, R 1 is a dodecyl residue and R 2 represents a decyl residue. 59. The method according to claim 52, wherein X and Y represent a valency dash, R 2 is Shydrogen and R' represents a Ci-C 3 0 alkyl chain which can be optionally substituted by Ci-C 6 aklylmercapto. 60. The method according to one of claims 52-59, wherein B represents a phosphate bridge of formula III (III), *o in which n denotes the numbers 1,2 or 3 and Z is O or S, and A denotes either O,S or a valency dash. 61. The method according to one of claims 52-59, wherein D or -B-D represents a pharmacologically active substance selected from the group of antiviral, antiretroviral, P:\OPERUEH\2207955 CLMS.DOC- -94- cytotoxic, cytostatic, antitumoral, immunosuppressive or immunostimulatory substances. 62. Method according to any one of claims 37-61 wherein said LCE is obtainable from leucocytes, monocytes, tumour cells, kidney cells, lymphocytes, cells of the immuno/lymphatic system or macrophages. 63. Method according to any one of claims 37-62 wherein in the case of conjugates of the type L-B-D in which L represents a lipid-like residue, B represents a phosphate bridge or a thiophosphate bridge and D denotes a pharmacologically active substance or B-D represents an active substance phosphonate the enzyme induces a cleavage of the covalent bond between the lipid moiety L and the residue -B-D. 64. Method according to any one of claims 37-63 wherein the activity of the LCE during cleavage is at least 2-fold higher in activated immune cells, human leucocytes, monocytes, tumour cells, lymphocytes, kidney cells, cells of the immuno/lymphatic system or macrophages compared to non-activated cells. 65. Method according to any one of claims 37-64 wherein the activity of the LCE is inhibited by Ca 2 or Mn 2 66. Process for the production of pharmaceutical agents containing a conjugate L-B-D for the targeted release of substances -B-D or D in target cells, wherein these target cells contain the lipid cleavage enzyme (LCE), which LCE complex is free of phospolipase C activity, wherein it cleaves the conjugate AZT-DMDOPE '-deoxy-3 '-azidothymidine)-5 '-phosphoric acid-(3-dodecyl- mercapto-2-decyloxy)-propyl ester] into AZT-MP [3'-deoxy-3'-azido-thymidine- monophosphate] and DMDOP [(3-dodecyl-mercapto-2-decyloxy)-propanol] or the Sconjugate FLT-DMDOPE [(3'-deoxy-3'-fluorothymidine)-5'-phosphoric acid-(3- Sdodecylmercapto-2-decyloxy)-propyl ester] into FLT-MP [3'-deoxy-3'-fluoro- P P:\OPERVEH\2207955 CLMS.DOC 2/5/01 and DMDOP or the conjugate 5-FU-DMDOPE acid-(3-dodecylmercapto-2-decyloxy)-propyl ester] into MP [5-fluorouridine monophosphate] and DMDOP, wherein the conjugate is an active substance of the formula L-B-D and L represents the lipid moiety, B is a valency dash, a phosphate bridge or a thiophosphate bridge and D represents a pharmacologically active substance or B-D is an active substance phosphonate comprising the steps a) selecting a pharmacologically active substance, b) producing a covalent conjugate of the pharmacologically active substance and a lipid-like carrier molecule, c) and producing a pharmaceutical form of administration which contains the conjugate produced according to b) as well as further pharmaceutical auxiliary or carrier substances. S67. Method as claimed in claim 66, wherein D or B-D comprises the following individual compounds of the following substance groups: *o antibody; peptide; hormone; toxin; a substance that intercalates in DNA or RNA; a tubulin inhibitor; an aklylating agent; a ribosome inactivating substance; a tyrosine phosphokinase inhibitor; a differentiation inducer; hormone agonist; hormone antagonist; a substance which changes the pleitropic resistance towards cytostatic agents; a calmodulin inhibitor; a protein kinase C inhibitor; a P-glycoprotein inhibitor; a modulator of mitochondrially bound hexokinase; an inhibitor of y-glutamylcysteine synthetase; a glutathione-S-transferase inhibitor; a superoxide dismutase inhibitor; an inhibitor of the hepatitis viruses A-E; anti-inflammatory agent; anti-rheumatic agent; anti-phlogistic agent; analgetic agent; antipyretic agent; anti-arrhythmic agent; calcium antagonist; antihistaminic agent; inhibitor of phosphodiesterase; symphathomimetic agent; parasympathomimetic agent; antisense oligonucleotides or active substances Sphosphonates; prodrugs thereof or derivatives capable of coupling. P:\OPERJEH\2207955 CLMS.DOC- 2/5101 -96- 68. Method according to claim 66 or 67 wherein said LCE is obtainable from leucocytes, monocytes, tumour cells, kidney cells, lymphocytes, cells of the immuno/lymphatic system or macrophages. 69. Method according to any one of claims 66-68 wherein in the case of conjugates of the type L-B-D in which L represents a lipid-like residue, B represents a phosphate bridge or a thiophosphate bridge and D denotes a pharmacologically active substance or B-D represents an active substance phosphonate the enzyme induces a cleavage of the covalent bond between the lipid moiety L and the residue -B-D. Method according to any one of claims 66-69 wherein the activity of the LCE during cleavage is at least 2-fold higher in activated immune cells, human leucocytes, monocytes, tumour cells, lymphocytes, kidney cells, cells of the immuno/lymphatic system or macrophages compared to non-activated cells. 71. Method according to any one of claims 66-70 wherein the activity of the LCE is inhibited by Ca 2 or Mn 2 72. A use according to any one of claims 1 to 29, a process according to any one of claims 30 or 66, a method according to any one of claims 31 to 35, 37 to 65 or 67 to 71 and a lipid conjugate according to claim 36 substantially as hereinbefore described with reference to the Figures and/or Examples. DATED this 2 nd day of MAY, 2001 ROCHE DIAGNOSTICS GMBH by their Patent Attorneys DAVIES COLLISON CAVE
AU44675/99A 1994-11-12 1999-08-24 Lipid splitting enzyme Ceased AU740293B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU44675/99A AU740293B2 (en) 1994-11-12 1999-08-24 Lipid splitting enzyme

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
DE4440472 1994-11-12
DE9518278 1995-05-18
AU39278/95A AU711367B2 (en) 1994-11-12 1995-11-09 Lipid-splitting enzyme
AU44675/99A AU740293B2 (en) 1994-11-12 1999-08-24 Lipid splitting enzyme

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU39278/95A Division AU711367B2 (en) 1994-11-12 1995-11-09 Lipid-splitting enzyme

Publications (2)

Publication Number Publication Date
AU4467599A AU4467599A (en) 1999-11-25
AU740293B2 true AU740293B2 (en) 2001-11-01

Family

ID=3726291

Family Applications (1)

Application Number Title Priority Date Filing Date
AU44675/99A Ceased AU740293B2 (en) 1994-11-12 1999-08-24 Lipid splitting enzyme

Country Status (1)

Country Link
AU (1) AU740293B2 (en)

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HOESTETLER AT AL. J BIOL. CHEM. 1990 265(11):6112-6117 *
PANTADOSI ET AL J MED CHEM 1991 34(4):1408-1414 *

Also Published As

Publication number Publication date
AU4467599A (en) 1999-11-25

Similar Documents

Publication Publication Date Title
US5599796A (en) Treatment of urogenital cancer with boron neutron capture therapy
US6180766B1 (en) Nucleosides and oligonucleotides containing boron clusters
Meier et al. cyclo Sal-Pronucleotides of 2 ‘, 3 ‘-Dideoxyadenosine and 2 ‘, 3 ‘-Dideoxy-2 ‘, 3 ‘-didehydroadenosine: Synthesis and Antiviral Evaluation of a Highly Efficient Nucleotide Delivery System
Skladanowski et al. Inhibition of DNA topoisomerase II by imidazoacridinones, new antineoplastic agents with strong activity against solid tumors.
ES2714324T3 (en) A method to identify or produce an aptamer
Rios Morales et al. Diastereoselective Synthesis of cycloSaligenyl‐Nucleosyl‐Phosphotriesters
AU711367B2 (en) Lipid-splitting enzyme
Nees Coronary flow increases induced by adenosine and adenine nucleotides are mediated by the coronary endothelium: a new principle of the regulation of coronary flow
Meyer Jr et al. 2'-O-Acyl-6-thioinosine cyclic 3', 5'-phosphates as prodrugs of thioinosinic acid
AU740293B2 (en) Lipid splitting enzyme
US7176011B2 (en) Lipid cleavage enzyme
Panagia et al. Subcellular localization of phosphatidylethanolamine N-methylation activity in rat heart
Manfredini et al. 5 ‘-Phosphoramidates and 5 ‘-Diphosphates of 2 ‘-O-Allyl-β-d-arabinofuranosyl-uracil,-cytosine, and-adenine: Inhibition of Ribonucleotide Reductase
EP0407816A2 (en) Base modified nucleosides
Creasey Biochemical effects of the vinca alkaloids—IV: Studies with Vinleurosine
Gupta Species specific differences in the toxicity of mithramycin, chromomycin A3, and olivomycin towards cultured mammalian cells
KR20180119758A (en) White blood cell specific aptamer and the use thereof
KR20010029544A (en) Natural antitumor or antiviral substances and use of the same
Barral et al. Synthesis and antiviral activity of boranophosphonate isosteres of AZT and d4T monophosphates
US20070009889A1 (en) Nucleoside derivative, modifed oligonucleotide, method for their syntesis and applicantions thereof
DE19518278A1 (en) Lipid-cleaving enzyme
Balducci et al. NAD-dependent inhibition of the NAD-glycohydrolase activity in A549 cells
Coetzee et al. The effect of several antitumor agents on 3H-TTP incorporation in host liver and hepatoma nuclei
Pegoraro et al. The synthesis of phospholipids in the nucleus and nuclear membrane of synchronized HeLa cells
Goswami Studies on the synthesis of liver proteins with particular reference to prothrombin

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
PC Assignment registered

Owner name: HEIDELBERG PHARMA HOLDING GMBH

Free format text: FORMER OWNER WAS: ROCHE DIAGNOSTICS GMBH