EP2268319A2 - Precursors of lipid metabolism for the diagnosis and treatment of cancer - Google Patents

Precursors of lipid metabolism for the diagnosis and treatment of cancer

Info

Publication number
EP2268319A2
EP2268319A2 EP08737300A EP08737300A EP2268319A2 EP 2268319 A2 EP2268319 A2 EP 2268319A2 EP 08737300 A EP08737300 A EP 08737300A EP 08737300 A EP08737300 A EP 08737300A EP 2268319 A2 EP2268319 A2 EP 2268319A2
Authority
EP
European Patent Office
Prior art keywords
prodrug
cancer
treatment
residue
detectable label
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP08737300A
Other languages
German (de)
French (fr)
Inventor
Sven Norbert Reske
Boris Zlatopolskiy
Christoph Solbach
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.)
Universitaet Ulm
Original Assignee
Universitaet Ulm
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universitaet Ulm filed Critical Universitaet Ulm
Publication of EP2268319A2 publication Critical patent/EP2268319A2/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/0402Organic compounds carboxylic acid carriers, fatty acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds

Definitions

  • the present invention pertains to methods for the treatment and diagnosis of cancer employing prodrugs for targeting of lipid metabolism comprising either a detectable label or a therapeutic residue.
  • the invention is based on the finding that the present prodrugs are specifically enriched in tumor tissue, in which they may be detected by suitable imaging techniques.
  • a prodrug comprising a therapeutic residue such as a suitable radioactive residue, may be used for the treatment of cancer.
  • Cancer represents one of the leading causes of death, and if current trends continue, cancer is expected to be the leading cause within some years.
  • Lung and prostate cancer represent one of the most frequent cancer killers for men, while lung and breast cancer are the most frequent cancer killers for women.
  • Currently available cancer therapies include surgery, chemotherapy, hormonal therapy and/or radiation treatment to eradicate neoplastic cells in a patient.
  • advances in cancer therapy have led to employment of biological therapy and immunotherapy to treat cancer.
  • Surgery may be contraindicated due to the health of the patient or may be unacceptable to the patient. Additionally, surgery may not completely remove the neoplastic tissue.
  • Hormonal therapy is rarely given as a single agent and although it can be effective, is often used only to prevent or delay recurrence of cancer after other treatments have removed the majority of the cancer cells.
  • Biological and immuno therapies are limited in number and may produce side effects such as rashes or swellings, flu-like symptoms, including fever, chills and fatigue, digestive tract problems or allergic reactions. Despite the availability of a variety of chemotherapeutic agents, chemotherapy has many drawbacks.
  • chemotherapeutic agents are toxic, and chemotherapy causes significant, and often dangerous, side effects, such as severe nausea, bone marrow depression, and immunosuppression. Additionally, even with administration of combinations of chemotherapeutic agents, many tumor cells are resistant to or develop resistance to the chemotherapeutic agents. In fact, cells resistant to a particular chemotherapeutic agent often prove to be resistant to other drugs, even drugs that act by wholly unrelated mechanisms; the so called pleiotropic drug or multidrug resistance. Thus, many cancers prove refractory to standard chemotherapeutic treatment protocols because of drug resistance.
  • Known assays base for example on the detection of nucleic acids, proteins or other biomarkers released from tumor cells in body fluids.
  • WO2006128192 discloses for example the use of free circulating DNA as a marker for diagnosis, prognosis, and treatment of cancer.
  • the diagnostic and prognostic tumor biomarkers in use today are not adequate in early identification of cancer due to their low sensitivity. Accordingly, there is still a need to develop more robust assays and genotypic markers that can be related to functional tumor biology. There is also a need in developing improved methods in the treatment of cancer.
  • the present invention seeks to overcome the problems associated with prior art methods.
  • the present invention provides particular prodrugs for targeting of lipid metabolism as well as their use for diagnosis and/or the treatment of cancer.
  • the present prodrugs for targeting of lipid metabolism are selected from
  • R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue.
  • the present invention is based on the finding that the present prodrugs for targeting of lipid metabolism are enriched in tumor cells. Labeling said prodrugs for targeting of lipid metabolism with a detectable label renders in vivo or in vitro diagnosis possible, whereas marking with a therapeutic residue permits the treatment of cancer.
  • the present prodrugs for targeting of lipid metabolism are esterified to the respective Acyl-CoA derivatives with coenzyme A (CoA) and enriched in tumor cells by specific interaction with enzymes, such as racemases, of lipid metabolism of the tumor. It has been surprisingly found that the activity said enzymes is highly upregulated in tumor cells giving the possibility to quantitatively, selectively and sensitively detect tumors. Another advantage resides in the possibility to treat tumors with non-invasive techniques, i.e. by administering the present prodrugs for targeting of lipid metabolism exhibiting therapeutic residues, which are almost exclusively incorporated in tumor tissue. Another advantage of the use of the present prodrugs for targeting of lipid metabolism resides is that they may be easily adapted to the changing requirements in the clinical field since the prodrug may be easily provided with another detectable label or therapeutic residue.
  • Fig. 1 shows the synthesis of 11 -aminoundecylmalonic acid and of /?-[*I]-iodobenzoyl-l l- aminoundecylmalonic acid according to an embodiment of the present invention
  • Fig. 2 shows the synthesis of n C-dodecylmalonic acid according to an embodiment of the present invention
  • Fig. 3 shows the synthesis of 11-fluorododecylmalonic acid as well as 11-[ 18 F]- fluorododecylmalonic acid (FSU 01) according to an embodiment of the present invention
  • Fig. 4 shows the synthesis of 13-iodotridecen-12-ylmalonic acid as well as 13-[*Ij- iodotridecen-12-ylmalonic acid according to an embodiment of the present invention
  • Fig. 5 shows an uptake of FSU 01 in prostate carcinoma cells according to an embodiment of the present invention, wherein only a neglectable cellular retention of FSU 01 may be observed after 24 h incubation, suggesting that said compound is metabolically degraded and eliminated from the cell in a quantitative manner;
  • Fig. 6 shows the biodistribution of FSU 01 in the transgene TRAMP - prostate carcinoma - model of the mouse according to an embodiment of the present invention, wherein FSU 01 is fast and intensively enriched in the prostate carcinoma tissue of the primary tumor and in lymph nodes, wherein the remaining organs show only a minor and uniform uptake of FSU
  • FSU 01 with exception of the skeleton and liver and kidney as excretion organs, wherein the high activity of FSU 01 in the bladder is probably due to the degradation to low molecular excretion products and subsequent renal excretion, wherein further analysis has shown that partial enrichment of FSU 01 in lymph nodes and skeleton was due to the formation of metastasis.
  • a “prodrug” and a “prodrug for targeting of lipid metabolism”, i.e. a prodrug of lipid metabolism, as used herein pertains to a pharmacological substance, i.e. a free fatty acid (FFA), which is metabolized in vivo into an active metabolite.
  • FFA free fatty acid
  • Metabolisation is hereby performed inter alia by the action of racemases, i.e. enzymes which may catalyze the inversion around the asymmetric carbon atom in a substrate having one center of asymmetry. It will be, however, appreciated that the substrates of e.g. racemases do not necessarily exhibit a center of asymmetry.
  • the present inventors surprisingly found in cell culture experiments and in mouse model experiments a strong uptake of a radio labelled prodrug of lipid metabolism, such as 2-(l l- [ 18 F]fluoroundecyl)malonic acid (also termed [F18]-FSU 01), in tumor tissue giving raise not only to the detection and/or localisation of the tumor, but also permits treatment of the tumor by employing a suitable therapeutic residue instead of a therapeutic label.
  • a radio labelled prodrug of lipid metabolism such as 2-(l l- [ 18 F]fluoroundecyl)malonic acid (also termed [F18]-FSU 01)
  • detectable label refers to any atom or molecule that may be used to provide a detectable (preferably quantifiable) effect and that can be attached to the present prodrugs.
  • these labels comprise e.g. enzymes which produce a detectable signal, for example by colorimetry, fluorescence or luminescence, such as horseradish peroxidase, alkaline phosphatase, [beta]-galactosidase or glucose-6-phosphate dehydrogenase, chromophores, such as fluorescent, luminescent or dye compounds, groups with an electron density detectable by electron microscopy or by virtue of their electrical property, such as conductivity, amperometry, voltametry or impedance, detectable group, for example the molecules of which are sufficiently large to induce detectable modifications of their physical and/or chemical characteristics; this detection can be carried out by optical methods such as diffraction, surface plasmon resonance, surface variation or contact angle variation, or physical methods such as atomic force spectros
  • Indirect systems may be used as well, such as, for example, ligands capable of reacting with an anti-ligand.
  • the ligand/anti-ligand pairs are well known to those skilled in the art, which is the case, for example, of the following pairs: biotin/streptavidin, hapten/antibody, antigen/antibody, peptide/antibody, sugar/lectin, polynucleotide/sequence complementary to the polynucleotide.
  • the anti-ligand may be detected directly by the labels described above or can itself be detectable by another ligand/anti-ligand pair.
  • An imaging technique as used herein refers to any kind of apparatus or device, capable of producing a visual signal or an image upon detecting a detectable label.
  • the apparatus permits localization of the detectable label within a mammal.
  • These kinds of biological imaging include for example radiology.
  • Non-limiting examples include positron- emission-tomography (PET), which produces a three-dimensional image or map of functional processes in the body.
  • PET positron- emission-tomography
  • the system detects pairs of gamma rays emitted indirectly by a positron-emitting radioisotope, which is introduced into the body on a metabolically active molecule, i.e. a prodrug.
  • Images of metabolic activity in space are then reconstructed by computer analysis, which may be supported by a CT X-ray scan performed on the patient at the same time and in the same device in order to obtain a three dimensional image enabling localization of tissue in which the prodrug is enriched.
  • positron emission a proton is converted via the weak force to a neutron, a positron and a neutrino. Isotopes, which undergo this so called beta plus decay, and thereby emit positrons.
  • Suitable positron-emitting radionuclides for this purpose include 11 C, 13 N, 18 F, 62 Cu, 64 Cu, 68 Ga, 75 Br, 76 Br, 77 Br, 80m Br, 86 Y, 121 I, 124 I of which 11 C and 18 F may be preferred.
  • the present invention should not be assumed to be intended thereto; other radionuclides may be used.
  • the present prodrugs may be also labeled with technetium and rhenium isotopes using known chelating complexes. Methods for the generation of radionuclides as well as radio labeling of compounds are well known to the skilled person. US2007273308 and WO2007122488 pertain e.g.
  • PET is preferably coupled with a computer tomography (CT).
  • CT computer tomography
  • Such PET/CT devices enable quantitative detection and allocation of the signals detected to particular tissues, i.e. a localization of the radionuclides employed and hence of the prodrugs attached thereto.
  • Function and operation of PET/CT as well as devices are well known to the skilled person (cf. Reske, S.N., Der Onkologe, 13(8) (2007) pp.
  • MRT magnetic resonance tomography
  • Spacers or “linkers” are molecules that are characterized in that they have a first end attached to the prodrug and a second end attached to the detectable label or therapeutic residue. Thus, the spacer molecule separates the prodrug and the detectable label or therapeutic residue, but is attached to both.
  • the spacers may be synthesized directly on or may be attached as a whole to the prodrug. Bindings within the spacer may include carbon- carbon single bonds, carbon-carbon double bonds, carbon-nitrogen single bonds, or carbon- oxygen single bonds. In addition, the spacer may have side chains or other substitutions.
  • the detectable label or therapeutic residue may be reacted by suitable means to form for example preferably a covalent bound between the spacer and prodrug.
  • Suitable linkers include alkyl, alkenyl, alkynyl chains, aromatic, polyaromatic, and heteroaromatic rings any of which may be optionally substituted for example with one or more ether, thiooether, ester, amine, sulphonamide, or amide functionality, monomers and polymers comprising ethylene glycol, amino acid, or carbohydrate subunits.
  • the linkers may be chosen to provide good in vivo pharmacokinetics, such as favorable excretion characteristics of the prodrug upon conversion in the mammal. The use of linkers with different lipophilicities and or charge can significantly change the in vivo pharmacokinetics of the prodrug to suit the diagnostic and /or therapeutic needs.
  • Linkers including a polyethylene glycol moiety have been found to slow blood clearance which may be desirable in some circumstances.
  • Spacers may be also in form of chelators capable of forming a complex with the detectable label or therapeutic residue.
  • aliphatic or "aromatic” substituent refers to residues composed of carbon and hydrogen. Aromatic substituents include ring structures, such as benzene, whereas aliphatic compounds do not. The present aliphatic or aromatic substituents may exhibit in addition to the detectable label or therapeutic residue one or more other residues, such as a hydroxy or amino group.
  • lower alkyl refers either to linear alley 1 residues of 1 - 8 C atoms or to branched alkyl residues of 3 - 8 C atoms.
  • Lower alkenyl designates residues exhibiting different numbers of double bounds; the length of said residues is in case linear alkenyl residues 3 - 8 C and branched alkenyl residues from 4 - 8 C atoms, respectively.
  • “Lower alkynyl” is directed to residues exhibiting different numbers of tripple bounds; the length of said residues is 3 - 8 C.
  • Said residues may also have one or more substituents, such as heteroatoms.
  • n is used to designate the number of C-atoms of lower alkyl, alkenyl and alkynyl residues. It will be appreciated that lower alkenyl residues may also contain single bonds, whereas lower alkynyl residues may contain single and/or double bonds.
  • therapeutic residue refers to an atom, such as 32 P, " 7 Cu 5 89 Sr, 88 Y, 90 Y, 123 I, 125 I, 131 I, 153 Sm, 165 Dy; 169 Er, 177 Lu, 178 Ta 5 186 Re, 188 Re, 195m Pt, 211 At, 213 Bi, 225 Ac or a complex containing such an atom in a coordinated form. It will be appreciated that respective radioactive therapeutic residues may be generated and attached to the present prodrugs for targeting of lipid metabolism in the same manner as detectable labels.
  • the use of the present prodrugs with therapeutic residues in cancer treatment combines the advantage of target selectivity with that of being systemic, as with chemotherapy, and it may be used as part of a therapeutic strategy with curative intent or for disease control and palliation.
  • therapeutic residue and detectable label are identical permitting diagnosis and treatment of cancer simultaneously.
  • sample is meant to include any specimen or culture of biological material from a mammal.
  • Biological samples may be human from human origin, fluid, such as blood or urine, solid or tissue.
  • the sample may be used as such in an assay or may be subjected to a preliminary isolation step prior to performing the assay.
  • the samples may be for example taken in order to follow elimination of radionuclides from the body and in order to assure that complete elimination has occurred.
  • room temperature refers to a temperature between 21 and 23 0 C.
  • reduced pressure designates any pressure below atmospheric pressure. A reduced pressure is preferably below 0.1 bar.
  • saturated saline refers to a saturated aqueous NaCl solution.
  • R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue.
  • Free fatty acids represent the substrates of lipid metabolism, lipid conversion and lipid storage particularly in the liver and adipose tissue and energy production in heart and skeletal muscles. FFA are therefore incorporated by heart and liver in great amounts after intravenous injection and dependent from work load and hormonal regulation of the skeletal muscles. FFA further represent important substrates for biosynthesis of cellular membranes, modification of proteins, transcriptional regulation and intracellular signal transduction (Berk, P. D. (1996). "How do long-chain free fatty acids cross cell membranes" Proc Soc Exp Biol Med 212(1): 1 - 4).
  • Energy demand of the tumor and energy metabolism, of, for example, the prostate carcinoma is very low due to a slow growth with a tumor cell growth fraction of around 1% and a very low division rate of tumor cells in comparison to energy demand of the heart muscle or skeletal muscles under workload.
  • the contribution to lipid metabolism is insignificant in comparison to e.g. the liver and membrane biosynthesis of the slow growing tumor cells is notably reduced in comparison to fast proliferating tissue, such as hematopoietic bone marrow or mucosa of the small intestine.
  • tumors comprise inter alia prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancer, malignant lymphoma, neoplasia of the hematopoietic system and musculoskeletal tumors.
  • Characteristic for some of the faster growing tumors is also that they supply their energy demand by highly increased glucose consumption.
  • the cancer is characterized by increased consumption of substrates of tumoral lipid metabolism.
  • cancers/tumors include prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancers, malignant lymphomas, neoplasias of the hematopoietic system and musculoskeletal tumors.
  • PCa prostate carcinoma
  • the prodrug further comprises a linker arranged between said prodrug and said detectable label, in that one end of the linker or spacer is attached to the prodrug, whereas the other end is attached to the detectable label.
  • the linker is preferably selected in a manner to provide good in vivo pharmacokinetics, inter alia by enabling rapid uptake of the prodrug in tumor tissue.
  • said detectable label is a radioactive label.
  • a suitable radioactive label detection with PET preferably PET/CT
  • PET/CT is enabled thus facilitating the localization of tumors in healthy tissue.
  • the radioactive label may be for example of 11 C, 13 N, 18 F, 62 Cu, 64 Cu, 68 Ga, 75 Br, 76 Br, 77 Br, 8Om Br, 86 Y or 124 I.
  • radioactive halogens are employed. More preferably, 11 C and 18 F are used due to their minor influence on the interaction between enzyme and prodrug.
  • said imaging technique is selected from positron-emission- tomography (PET), and positron-emission-tomography/computer tomography (PET/CT).
  • PET/CT is preferred due to the improved possibilities of visualizing/indicating the position and dimensions of a tumor in healthy tissue.
  • Another advantage resides in that even metastasis comprising few tumor cells may be detected and localized.
  • the metastasis comprise preferably less than 10 5 tumor cells, more preferably less than 10 4 tumor cells and most preferably less than 10 2 tumor cells.
  • a prodrug of lipid metabolism wherein said prodrug is selected from
  • R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue.
  • Preferred prodrugs comprise malonic, alpha - methyl - substituted carboxylic acid derivatives with a single substituent, malonic acid monomethylester and malonic acid monoamide. It will be appreciated that the synthesis of appropriate prodrugs for targeting of lipid metabolism is well within the knowledge of the skilled person and that the skilled artisan is not restricted to a particular route of synthesis.
  • P-Iodbenzoyl-l l-aminodecylmalonic acid (standard) and 11-Aminoundecylmalonic acid (precursor) may be produced starting from DMUSM in three steps as outlined in figure 1.
  • the corresponding radio labelled malonic acid derivate may be obtained by acetylating e.g. 11 -aminodecylmalonic acid with p- I-benzene-N-hydroxysuccinate. It will be appreciated that other radio labels or therapeutic residues may be attached in a similar manner.
  • u C-dodecylmalonic acid may be produced (cf. fig. 2).
  • LDA lithium diisopropylamide
  • HMPT hexamethylphosphortriamide
  • the precursor is generated in situ.
  • 11 CO 2 followed by hydrolysis, the 11 C marked product is obtained.
  • the corresponding standard is obtained by hydro lysing of the dimethylester, which is obtained by alcylation of sodium dimethylmalonate with dodecylbromide.
  • 2-Methyl-13-(methylsulfonyloxy)methyltridecanoate serving as precursor for the preparation of 13-[ i8 F]fluoro-2-methyItridecanic acid, is synthesised starting from 2-methyldimethyl- malonate. In the same manner 13-fluoro-2-methyltridecanic acid may be obtained. Synthesis of 3-Hydroxy-(2S)-methylmethylpropionate is known in the state of the art.
  • a synthesis of 13-iodotridecen-12-ylmalonic acid as well as 13- [*I]-iodotridecen-12-ylmalonic acid are synthesized are exemplary outlined in fig.4.
  • Quality control of the compounds may be easily checked by high performance liquid chiOmatographie (HPLC) or in case of e.g. 18 F labelling or another labelling with a halogen by comparison of the retention times of the radio labelled products with those of the respective "cold" standard compounds.
  • HPLC high performance liquid chiOmatographie
  • the prodrug preferably comprises a linker arranged between said prodrug and said detectable label.
  • the linker may be selected in a manner bestowing the present prodrug advantageous properties, such as good in vivo pharmacokinetics.
  • the detectable label preferably comprises a chemoluminescent, fluorescent, bioluminescent, radioactive label or a label detectable by PET, CT or MRT.
  • chemoluminescent, fluorescent, bioluminescent or radioactive labels also in vitro approaches may be used.
  • In vitro assay for the diagnosis of cancer may for example based on interaction with enzymes, e.g. racemases, characteristic for the presence of a tumor from the body.
  • a method is envisaged comprising the steps of providing a sample from a mammal, optionally purifying the sample, adding a present prodrug exhibiting one of the above mentioned markers and detecting the degradation products caused by the activity of enzymes characteristic for the presence of a tumor. It will be appreciated that such methods are well known to the skilled person.
  • prodrug may be alpha - methyl - carboxylic or malonic acids and their derivatives.
  • the prodrug is 2-[l l-[ 18 F]fluoroundecyl]malonic acid (designated [F18]- FSU 01), which has been found to be incorporated in tumor tissue, such as prostate tumor tissue, fast and in high amounts.
  • the aliphatic substituent may be selected from C8 - C20 linear alkyl or C8 - C20 branched alkyl. Instead of an aliphatic substituent also an aromatic substituent may be employed. According to another embodiment of the present invention, the aliphatic substituent may be C9, CI l, C13, C15, C17 or C19 linear alkyl. According to a further embodiment of the present invention, the aliphatic substituent may be Cl 1 linear alkyl.
  • the detectable marker may be selected from the group consisting of 11 C, 13 N, 18 F, 32 P 5 35 S 5 64 Cu, 62 Cu, 67 Cu, 67 Ga, 68 Ga, 75 Br, 76 Br, 77 Br, S0m Br, S6 Y, 89 Sr, 88 Y 5 90 Y, 99m Tc, 111 In, 121 I, 123 I, 124 I, 125 I, 127 I, 131 I, 153 Sm, 165 Dy; 169 Er, 1 77 Lu, 178 Ta, 186 Re, ISS Re 295m Pt, 211 At, 213 Bi, 225 Ac.
  • the diagnostic residue may be 11 C, 18 F or 123 I.
  • the therapeutic residue may be preferably selected from the group consisting Of 32 P, 67 Cu, 89 Sr, 88 Y, 90 Y, 123 1, 125 1, 131 I, 153 Sm, 1 65 Dy; 169 Er, 177 Lu, 178 Ta, 186 Re, 188 Re, 195m Pt, 211 At, 213 Bi, 225 Ac.
  • the therapeutic residue may be 90 Y.
  • the detectable marker may be 18 F, 11 C or 123 I. According to an embodiment of the present invention, the detectable marker may be 1 8 F and 123 I.
  • Rl and R2 may be independently selected from H, Cl - C8 linear alkyl, C2 - C8 branched alkyl, C3 - C8 linear alkenyl, C4 - C8 branched alkenyl, C3 - C8 linear alkynyl, or C4 - C8 branched alkynyl.
  • Rl and/or R2 may be H or Cl - C2 linear alkyl, since these residues may easily react with enzymes in tumor tissue due to a reduced steric hindrance. According to another embodiment of the present invention, Rl and/or R2 may be H.
  • a method for the treatment of cancer comprises the steps of providing a prodrug for targeting of lipid metabolism to a mammal, wherein said prodrug comprises a therapeutic residue, administering said prodrug to a mammal.
  • the prodrug may be selected from
  • R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue.
  • the neoplastic tissue i.e. the tumor tissue
  • the therapeutic residue may be selected in order to correspond to a suitable detectable label, in that upon administering of the present prodrug not only enrichment in the target tissue and elimination from the body, but also the activity of the therapeutic residue on the tumor tissue may be observed.
  • the pharmacokinetics may be further influenced by choice of a suitable linker.
  • said therapeutic residue may be selected from the group consisting Of 32 P, 67 Cu, 89 Sr, 88 Y, 90 Y 5 123 I, 125 I 5 131 I, 153 Sm, 165 Dy, 169 Er 5 177 Lu, 178 Ta 5 186 Re 5 188 Re, 195m Pt, 211 At 5 213 Bi, and 225 Ac, According to an embodiment of the present invention, the therapeutic residue may be identical with a detectable label.
  • prodrug may be alpha - methyl - carboxylic or malonic acids and their derivatives.
  • the aliphatic substituent may be selected from C4 - C25 linear alkyl or C4 - C25 branched alkyl.
  • an aromatic substituent may be employed.
  • the aliphatic substituent may be CI l 5 C13 or Cl 5 linear alkyl.
  • the aliphatic substituent may be CI l linear alkyl.
  • Rl and R2 may be independently selected from Cl - C8 linear alkyl, C3 - C8 branched alkyl, C3 - C8 linear alkenyl, C4 - C8 branched alkenyl, C3 - C8 linear alkynyl, and C4 - C8 branched alkynyl.
  • Rl and/or R2 may be H or Cl - C2 linear alkyl, since these residues may easily react with enzymes in tumor tissue due to a reduced steric hindrance.
  • Rl and/or R2 may be H.
  • a cancer to be treated is selected from prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancers, malignant lymphomas, neoplasias of the hematopoietic system and musculoskeletal tumors.
  • the prodrug is administered by intravenous, subcutaneous, intramuscular or intracavitary injection.
  • the prodrug is contained in a pharmaceutical composition.
  • Said composition may be in the form of a parenteral formulation since due to the lipophilic nature of said compound administration via injection is advisable.
  • other kind of formulations such as oral formulations, may be envisaged as well.
  • the methods of preparation may include the step of bringing the active compound into association with a carrier, which constitutes of one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into desired formulations.
  • compositions for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or a suspension in an aqueous or non- aqueous liquid; or as an oil-in- water or water-in-oil emulsion.
  • Such compositions may be prepared by any suitable method of pharmacy, which includes the step of bringing into association the active compound and a suitable carrier.
  • the compositions are prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture.
  • a tablet may be prepared by compressing or molding a power or granules containing the active compound, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, and/or surface active/dispensing agent(s).
  • Molded tablets may be prepared by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
  • a syrup may be made by adding the active compound to a concentrated aqueous solution of a sugar, for example sucrose to which may also be added any accessoiy ingredient(s).
  • compositions for oral administration may optionally include enteric coatings known in the art to prevent degradation of the compositions in the stomach and provide release of the drug in the small intestine.
  • compositions suitable for buccal or sub-lingual administration include lozenges comprising the active compound in a flavoured base, usually sucrose and acacia or tragacanth and pastilles comprising the compound in an inert base such as gelation and glycerin or sucrose and acacia.
  • compositions suitable for parenteral administration comprise sterile aqueous and nonaqueous injection solutions of the active compound, which preparations are preferably isotonic with the blood of the intended recipient.
  • compositions may contain anti-oxidants, buffers, bacteriostats and solutes, which render the compositions isotonic with the blood of the intended recipient.
  • Aqueous and non-aqueous sterile suspensions may include suspending agents and thickening agents.
  • the compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried or a lyophilized condition requiring only the addition of the sterile liquid carrier, for example, saline or water- for- injection immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • the use of the present prodrugs for targeting of lipid metabolism for the treatment and/or prevention of cancer is envisaged.
  • PCa-cell lines were used: LNCaP (human prostate carcinoma, DSMZ Cat.-No. ACC 256) and TRAMP (murine prostate adenocarcinoma, obtainable from LGC Cat. No. CRL-2730; Greenberg, N.M. et al., Proc Natl Acad Sci U S A, 92(8) (1995) pp. 3439-43).
  • PC3 DSMZ Cat.-No. ACC 465) was used as reference.
  • Human colon adenocarcinoma CX-I were obtained from DSMZ Cat.-No. ACC 129.
  • PC3 was cultivated/maintained in 50% Ham's F12 with 50% RPMI, 10% FKS, 1% penicillin + streptomycin, 1% L-glutamate.
  • LNCaP was cultivated/maintained in RPMI with 10% FKS, 1% penicillin + streptomycin, 1% L-glutamate, 1% sodium pyruvate, 1% not essential amino acids.
  • TRAMP cells were cultivated/maintained in DMEM (high glucose), 5% Nu-serum IV, 5 % FKS; 1% penicillin + streptomycin, 10 "s M dihydrotestosterone, 5 ⁇ g/ml insuline.
  • CX-I cells were cultivated/maintained in DMEM (high glucose), 10% FKS.
  • Radioactivity was measured according to manufactures instructions with a ⁇ -counter (Cobra 2, GR Healthcare / Packard Instruments, Palo Alto, USA).
  • THF was distilled under reduced pressure.
  • the residue was diluted with 1 M HCl (10 ml) and water (200 ml).
  • the resulting emulsion was extracted with pentane (100 ml).
  • the pentane solution was washed with 1 M HCl (50 ml), water (10 * 50 ml) and a saturated saline solution (50 ml) and afterwards dried over magnesium sulfate.
  • the target compound (0.43 g, 81%) was obtained as colourless liquid.
  • GE remote controlled synthesis unit of Nuclear Interface
  • [ 18 F]fluoride produced by the cyclotron was fixed on a conditioned (10 ml 1 M NaHCO 3 , 10 ml water) anion exchange cartridge (Sep-Pak light, Accell Plus QMA 5 Waters) and thereby separated from the [ 18 O]water-matrix.
  • [ ] S F]fluoride was eluted with 360 ⁇ L 0,066 M KOH and subsequently 20 mg Kryptofix 2.2.2. in 1 ml acetonitrile was added to a [ 18 F] fluoride solution.
  • the radionuclide was activated for the radiolabeling by means of subsequent azeotropic distillation for 6 min. under vacuum (approx. 6 mbar) until dryness at first 95 0 C and later 100°C.
  • the radiolabeling was conducted by addition of compound (9 mg, 23.65 ⁇ mol) obtained in example 4 in acetonitrile (1 ml) and heating at 85 0 C for 10 min.
  • Not converted [ 18 F] fluoride was removed from the neutralized and diluted reaction mixture by means of a Al 2 ⁇ 3 -carti ⁇ dge (Sep-Pak light, Alumina N 5 Waters) and purified by semipreparative HPLC (stationaiy phase: LiChrospher Select B 5 ⁇ , 250x10 mm, Chromatographic Service; mobile phase: CH 3 CN/H 2 O 60/40 v/v, 0,1 % TFA; flow: 5 ml/min; radioactivity detector and UV- detector (210 nm)) by means of an automated injection unit (flow-detector). The product fraction was isolated after approx. 7 min. by valve switching and was diluted with 50 ml water.
  • Quality control of the produced fluorine- 18 labelled fatty acids was performed by an analytical Radio-HPLC-system (stationary phase: LiChrospher Select B 5 ⁇ , 250x4,6 mm, Chromatographic Service; mobile CH 3 CN/H 2 O 50/50 v/v, 0,1 % TFA; flow: 2 ml/min; radio activity detector und UV-detector (210 nm)).
  • Product identification was performed by comparison of the retention times of the products with those of the respective "cold" standard compounds (the compounds obtained in example 3).
  • Tridecynyl- 12-01-1 (4.95 g, 29.71 mmol), Ph 3 P (11.69 g, 44.57 mmol) and imidazole (3.034 g, 44.57 mmol) were dissolved in THF (50 ml) under argon atmosphere and ice cooling. Iodine (10.406 g 5 41.0 mmol) in THF (20 ml) was added drop wise within 20 min. The reaction mixture was stirred for 2 hours at room temperature. Afterwards saturated Na 2 S 2 O 3 solution (3 ml) was added drop wise within 2 min. The reaction mixture was dried over magnesium sulfate. The obtained residue was extracted with pentane (20 x 50 ml).
  • the pentane solution was washed successively with 1 M KHSO 4 (50 ml), water (5 x 100 ml) and saturated saline (50 ml).
  • the target compound (3.25 g, 80%) was obtained as colourless oil, which crystallizes slowly as colourless solid.
  • the target compound was obtained as a mixture Z/E isomers (30:70), containing less than 5 mol% of the elimination product (tridecyn-12-ylmalonic acid) (0.175 g, > 89%) as colourless solid.
  • Trifluoroacetic acid/acetoniti ⁇ le 10/90 v/v (50 ⁇ l) was added.
  • the reaction mixture was diluted with acetonitrile/water (0.1 % TFA) 33/67 v/v (750 ⁇ l) and separated by a semipreparative HPLC(stationary phase: LiChrospher 60 Select B 5 ⁇ RP C-8; 250 x 10 mm, Chromatographic Service; mobile phase gradient: CH 3 CN with 0,1% TFA and 0.1% TFA 5 0.5 min. 50% CH 3 CN with 0.1% TFA 5 5 - 10 min. 50 - 80% CH 3 CN with 0.1% TFA 5 10 - 50 min.
  • Quality control of the produced radio iodine labelled fatty acid was carried out by analytical radio HPLC (stationary phase: LiChrospher Select B 5 ⁇ , 250x4,6 mm, Chromatographic Service; mobile phase: CH 3 CN/H 2 O 80/20 v/v; flow: 1 ml/min; radio activity detector and TJV-detector (220 nm)).
  • Product identification was carried out by comparison of the retention times of the radio labelled products with that of the respective "cold" standard compound. The radio chemical purity of the product was >95%.
  • PC3 cells serve hereby as androgen independent negative control, whereas LNCap and TRAMP (Greenberg, N.M. et al., Proc Natl Acad Sci U S A 5 92(8) (1995) p. 3439-43) cell lines are both androgen dependent, i.e. exhibiting an enzyme expression profile essentially corresponding to that of human prostate cancer cells in vivo.
  • the cells were incubated for 24 h in the respective media according to the instructions provided by the German Collection of Microorganisms and Cell Cultures.
  • Values at 1 h were determined by removing 100 ⁇ l from each well and adding 900 ⁇ l PBS (activity per well: x ⁇ 10 MBq).
  • Values at 20 h were determined by removing 200 ⁇ l from the original solution and adding directly to each well (200 ⁇ l per well).
  • Tissue/organ uptake of FSU 01 was calculated 30 and 60 min after intravenous application of FSU 01 in a caudal vein (cf. fig. 6).
  • a high uptake of [F-IS]FSU 01 in prostate primary tumors and lymph node metastasis of the prostate carcinoma may be derived therefrom.
  • the 18 F labeled alkylmalonic acid was enriched in the transgene TRAMP mouse model, for example, in prostate carcinoma and its metastasis in lymph nodes more than 10 -
  • control tissue such as periprostatic soft tissues, intestinal tissue, skeletal muscles or normal prostate tissue.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Epidemiology (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Radiology & Medical Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention pertains to methods for the treatment and diagnosis of cancer employing prodrugs for targeting of lipid metabolism comprising either a detectable label or a therapeutic residue. The invention is based on the finding that the present prodrugs are specifically enriched in tumor tissue, in which they may be detected by suitable imaging techniques. Alternatively, a prodrug comprising a therapeutic residue, such as a suitable radioactive residue, may be used for the treatment of cancer.

Description

Precursors of lipid metabolism for the diagnosis and treatment of cancer
The present invention pertains to methods for the treatment and diagnosis of cancer employing prodrugs for targeting of lipid metabolism comprising either a detectable label or a therapeutic residue. The invention is based on the finding that the present prodrugs are specifically enriched in tumor tissue, in which they may be detected by suitable imaging techniques. Alternatively, a prodrug comprising a therapeutic residue, such as a suitable radioactive residue, may be used for the treatment of cancer.
Cancer represents one of the leading causes of death, and if current trends continue, cancer is expected to be the leading cause within some years. Lung and prostate cancer represent one of the most frequent cancer killers for men, while lung and breast cancer are the most frequent cancer killers for women. Currently available cancer therapies include surgery, chemotherapy, hormonal therapy and/or radiation treatment to eradicate neoplastic cells in a patient. Recently, advances in cancer therapy have led to employment of biological therapy and immunotherapy to treat cancer.
AU known cancer treatments pose significant drawbacks for the patient. Surgery, for example, may be contraindicated due to the health of the patient or may be unacceptable to the patient. Additionally, surgery may not completely remove the neoplastic tissue. Hormonal therapy is rarely given as a single agent and although it can be effective, is often used only to prevent or delay recurrence of cancer after other treatments have removed the majority of the cancer cells. Biological and immuno therapies are limited in number and may produce side effects such as rashes or swellings, flu-like symptoms, including fever, chills and fatigue, digestive tract problems or allergic reactions. Despite the availability of a variety of chemotherapeutic agents, chemotherapy has many drawbacks. Almost all chemotherapeutic agents are toxic, and chemotherapy causes significant, and often dangerous, side effects, such as severe nausea, bone marrow depression, and immunosuppression. Additionally, even with administration of combinations of chemotherapeutic agents, many tumor cells are resistant to or develop resistance to the chemotherapeutic agents. In fact, cells resistant to a particular chemotherapeutic agent often prove to be resistant to other drugs, even drugs that act by wholly unrelated mechanisms; the so called pleiotropic drug or multidrug resistance. Thus, many cancers prove refractory to standard chemotherapeutic treatment protocols because of drug resistance.
Accordingly, there remains a significant and unmet need for additional cancer therapies as all current treatments have significant disadvantages. Further, it is uncommon for a particular treatment to be effective to treat every instance of a given cancer.
Currently available methods for the diagnosis of cancer exhibit similar drawbacks. Known assays base for example on the detection of nucleic acids, proteins or other biomarkers released from tumor cells in body fluids. WO2006128192 discloses for example the use of free circulating DNA as a marker for diagnosis, prognosis, and treatment of cancer.
The diagnostic and prognostic tumor biomarkers in use today are not adequate in early identification of cancer due to their low sensitivity. Accordingly, there is still a need to develop more robust assays and genotypic markers that can be related to functional tumor biology. There is also a need in developing improved methods in the treatment of cancer.
The present invention seeks to overcome the problems associated with prior art methods.
The present invention provides particular prodrugs for targeting of lipid metabolism as well as their use for diagnosis and/or the treatment of cancer. The present prodrugs for targeting of lipid metabolism are selected from
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alkyl, alkenyl or alkynyl, n = 1 - 8.
The present invention is based on the finding that the present prodrugs for targeting of lipid metabolism are enriched in tumor cells. Labeling said prodrugs for targeting of lipid metabolism with a detectable label renders in vivo or in vitro diagnosis possible, whereas marking with a therapeutic residue permits the treatment of cancer.
Without wishing to be bound by any theory, it is presently assumed that the present prodrugs for targeting of lipid metabolism are esterified to the respective Acyl-CoA derivatives with coenzyme A (CoA) and enriched in tumor cells by specific interaction with enzymes, such as racemases, of lipid metabolism of the tumor. It has been surprisingly found that the activity said enzymes is highly upregulated in tumor cells giving the possibility to quantitatively, selectively and sensitively detect tumors. Another advantage resides in the possibility to treat tumors with non-invasive techniques, i.e. by administering the present prodrugs for targeting of lipid metabolism exhibiting therapeutic residues, which are almost exclusively incorporated in tumor tissue. Another advantage of the use of the present prodrugs for targeting of lipid metabolism resides is that they may be easily adapted to the changing requirements in the clinical field since the prodrug may be easily provided with another detectable label or therapeutic residue.
In the figures
Fig. 1 shows the synthesis of 11 -aminoundecylmalonic acid and of /?-[*I]-iodobenzoyl-l l- aminoundecylmalonic acid according to an embodiment of the present invention;
Fig. 2 shows the synthesis of nC-dodecylmalonic acid according to an embodiment of the present invention;
Fig. 3 shows the synthesis of 11-fluorododecylmalonic acid as well as 11-[18F]- fluorododecylmalonic acid (FSU 01) according to an embodiment of the present invention;
Fig. 4 shows the synthesis of 13-iodotridecen-12-ylmalonic acid as well as 13-[*Ij- iodotridecen-12-ylmalonic acid according to an embodiment of the present invention;
Fig. 5 shows an uptake of FSU 01 in prostate carcinoma cells according to an embodiment of the present invention, wherein only a neglectable cellular retention of FSU 01 may be observed after 24 h incubation, suggesting that said compound is metabolically degraded and eliminated from the cell in a quantitative manner; and
Fig. 6 shows the biodistribution of FSU 01 in the transgene TRAMP - prostate carcinoma - model of the mouse according to an embodiment of the present invention, wherein FSU 01 is fast and intensively enriched in the prostate carcinoma tissue of the primary tumor and in lymph nodes, wherein the remaining organs show only a minor and uniform uptake of FSU
01, with exception of the skeleton and liver and kidney as excretion organs, wherein the high activity of FSU 01 in the bladder is probably due to the degradation to low molecular excretion products and subsequent renal excretion, wherein further analysis has shown that partial enrichment of FSU 01 in lymph nodes and skeleton was due to the formation of metastasis.
Definitions:
A "prodrug" and a "prodrug for targeting of lipid metabolism", i.e. a prodrug of lipid metabolism, as used herein pertains to a pharmacological substance, i.e. a free fatty acid (FFA), which is metabolized in vivo into an active metabolite. Metabolisation is hereby performed inter alia by the action of racemases, i.e. enzymes which may catalyze the inversion around the asymmetric carbon atom in a substrate having one center of asymmetry. It will be, however, appreciated that the substrates of e.g. racemases do not necessarily exhibit a center of asymmetry. Fatty acid utilization in normal prostate tissue and in prostate carcinoma is in comparison to FFA utilization in liver (Iozzo, P. et al., Eur J Nucl Med MoI Imaging, 30(8) (2003), pp. 1160-4) and heart muscles (Reske, S. N. et al., Am J Physiol Imaging 1(4) (1986), pp. 214-229) is comparatively low (Toghrol, F. et al., Toghrol, 25(6) (1980), pp. 371-6;del Hoyo, N. et al., Biosci Rep, 10(1) (1990), pp. 105-12). The present inventors surprisingly found in cell culture experiments and in mouse model experiments a strong uptake of a radio labelled prodrug of lipid metabolism, such as 2-(l l- [18F]fluoroundecyl)malonic acid (also termed [F18]-FSU 01), in tumor tissue giving raise not only to the detection and/or localisation of the tumor, but also permits treatment of the tumor by employing a suitable therapeutic residue instead of a therapeutic label.
The expression "detectable label" refers to any atom or molecule that may be used to provide a detectable (preferably quantifiable) effect and that can be attached to the present prodrugs. A non-limiting list of these labels comprise e.g. enzymes which produce a detectable signal, for example by colorimetry, fluorescence or luminescence, such as horseradish peroxidase, alkaline phosphatase, [beta]-galactosidase or glucose-6-phosphate dehydrogenase, chromophores, such as fluorescent, luminescent or dye compounds, groups with an electron density detectable by electron microscopy or by virtue of their electrical property, such as conductivity, amperometry, voltametry or impedance, detectable group, for example the molecules of which are sufficiently large to induce detectable modifications of their physical and/or chemical characteristics; this detection can be carried out by optical methods such as diffraction, surface plasmon resonance, surface variation or contact angle variation, or physical methods such as atomic force spectroscopy or the tunnel effect, radioactive molecules such as 11C, 13N, 18F5 32P5 35S1 64Cu, 62Cu, 67Cu, 67Ga, 68Ga, 75Br, 76Br, 77Br,80mBr, 86Y, 89SrZ8Y, 90Y, 99mTc, 111In, 1211, 123I, 124I5 127I, 125I, 131I, 153Sm, 165Dy; 169Er, 177Lu, 178Ta, 186Re, 188Re 195mPt, 211At, 213Bi, 225Ac. Indirect systems may be used as well, such as, for example, ligands capable of reacting with an anti-ligand. The ligand/anti-ligand pairs are well known to those skilled in the art, which is the case, for example, of the following pairs: biotin/streptavidin, hapten/antibody, antigen/antibody, peptide/antibody, sugar/lectin, polynucleotide/sequence complementary to the polynucleotide. The anti-ligand may be detected directly by the labels described above or can itself be detectable by another ligand/anti-ligand pair.
An imaging technique as used herein refers to any kind of apparatus or device, capable of producing a visual signal or an image upon detecting a detectable label. Preferably the apparatus permits localization of the detectable label within a mammal. These kinds of biological imaging include for example radiology. Non-limiting examples include positron- emission-tomography (PET), which produces a three-dimensional image or map of functional processes in the body. The system detects pairs of gamma rays emitted indirectly by a positron-emitting radioisotope, which is introduced into the body on a metabolically active molecule, i.e. a prodrug. Images of metabolic activity in space are then reconstructed by computer analysis, which may be supported by a CT X-ray scan performed on the patient at the same time and in the same device in order to obtain a three dimensional image enabling localization of tissue in which the prodrug is enriched. In positron emission a proton is converted via the weak force to a neutron, a positron and a neutrino. Isotopes, which undergo this so called beta plus decay, and thereby emit positrons. Suitable positron-emitting radionuclides for this purpose include 11C, 13N, 18F, 62Cu, 64Cu, 68Ga, 75Br, 76Br, 77Br,80mBr, 86Y, 121I, 124I of which 11C and 18F may be preferred. The present invention should not be assumed to be intended thereto; other radionuclides may be used. The present prodrugs may be also labeled with technetium and rhenium isotopes using known chelating complexes. Methods for the generation of radionuclides as well as radio labeling of compounds are well known to the skilled person. US2007273308 and WO2007122488 pertain e.g. to the production of radionuclides, the contents of which are incorporated herein by way of reference. Radio labeling is for example outlined in WO 2007148089 and WO 2007148083, the contents of which are incorporated herein by way of reference. PET is preferably coupled with a computer tomography (CT). Such PET/CT devices enable quantitative detection and allocation of the signals detected to particular tissues, i.e. a localization of the radionuclides employed and hence of the prodrugs attached thereto. Function and operation of PET/CT as well as devices are well known to the skilled person (cf. Reske, S.N., Der Onkologe, 13(8) (2007) pp. 677-690; Reske, S.N et al., Urologe A, 45(10) (2006), pp. 1240-1250; Reske, S.N. et al., Urologe A, 45(6) (2006) pp. 707-714; Schδder, H. and S.M. Larson, Semin Nucl Med, 34(4) (2004) pp. 274-292; Morris, MJ. and H.I., Scher Eur. J. Nucl. Med. MoI. Imaging, 34(2) (2007) pp. 181-4). Other suitable techniques comprise single photon emission computer tomography (SPECT) and techniques based on magnetic resonance tomography (MRT) wherein the quantum mechanical magnetic properties of an atom's nucleus is detected. Useable contrast agents for MRT may involve attachment of Gadolinium, C- 13, F- 19 or ferrous species to the prodrugs for targeting of lipid metabolism for enhancing MR - contrast in the targeted tissue.
"Spacers" or "linkers" are molecules that are characterized in that they have a first end attached to the prodrug and a second end attached to the detectable label or therapeutic residue. Thus, the spacer molecule separates the prodrug and the detectable label or therapeutic residue, but is attached to both. The spacers may be synthesized directly on or may be attached as a whole to the prodrug. Bindings within the spacer may include carbon- carbon single bonds, carbon-carbon double bonds, carbon-nitrogen single bonds, or carbon- oxygen single bonds. In addition, the spacer may have side chains or other substitutions. The detectable label or therapeutic residue may be reacted by suitable means to form for example preferably a covalent bound between the spacer and prodrug. Suitable linkers include alkyl, alkenyl, alkynyl chains, aromatic, polyaromatic, and heteroaromatic rings any of which may be optionally substituted for example with one or more ether, thiooether, ester, amine, sulphonamide, or amide functionality, monomers and polymers comprising ethylene glycol, amino acid, or carbohydrate subunits. The linkers may be chosen to provide good in vivo pharmacokinetics, such as favorable excretion characteristics of the prodrug upon conversion in the mammal. The use of linkers with different lipophilicities and or charge can significantly change the in vivo pharmacokinetics of the prodrug to suit the diagnostic and /or therapeutic needs. Linkers including a polyethylene glycol moiety have been found to slow blood clearance which may be desirable in some circumstances. Spacers may be also in form of chelators capable of forming a complex with the detectable label or therapeutic residue.
The term "aliphatic" or "aromatic" substituent refers to residues composed of carbon and hydrogen. Aromatic substituents include ring structures, such as benzene, whereas aliphatic compounds do not. The present aliphatic or aromatic substituents may exhibit in addition to the detectable label or therapeutic residue one or more other residues, such as a hydroxy or amino group.
The term "lower alkyl" as used herein refers either to linear alley 1 residues of 1 - 8 C atoms or to branched alkyl residues of 3 - 8 C atoms. "Lower alkenyl" designates residues exhibiting different numbers of double bounds; the length of said residues is in case linear alkenyl residues 3 - 8 C and branched alkenyl residues from 4 - 8 C atoms, respectively. "Lower alkynyl" is directed to residues exhibiting different numbers of tripple bounds; the length of said residues is 3 - 8 C. Said residues may also have one or more substituents, such as heteroatoms. The letter "n" is used to designate the number of C-atoms of lower alkyl, alkenyl and alkynyl residues. It will be appreciated that lower alkenyl residues may also contain single bonds, whereas lower alkynyl residues may contain single and/or double bonds.
The term "therapeutic residue" as used herein refers to an atom, such as 32P, "7Cu5 89Sr,88Y, 90Y, 123I, 125I, 131I, 153Sm, 165Dy; 169Er, 177Lu, 178Ta5 186Re, 188Re, 195mPt, 211At, 213Bi, 225Ac or a complex containing such an atom in a coordinated form. It will be appreciated that respective radioactive therapeutic residues may be generated and attached to the present prodrugs for targeting of lipid metabolism in the same manner as detectable labels. The use of the present prodrugs with therapeutic residues in cancer treatment combines the advantage of target selectivity with that of being systemic, as with chemotherapy, and it may be used as part of a therapeutic strategy with curative intent or for disease control and palliation. Preferably, therapeutic residue and detectable label are identical permitting diagnosis and treatment of cancer simultaneously.
The expression "sample" is meant to include any specimen or culture of biological material from a mammal. Biological samples may be human from human origin, fluid, such as blood or urine, solid or tissue. The sample may be used as such in an assay or may be subjected to a preliminary isolation step prior to performing the assay. The samples may be for example taken in order to follow elimination of radionuclides from the body and in order to assure that complete elimination has occurred.
The term "room temperature" refers to a temperature between 21 and 230C. The expression "reduced pressure" designates any pressure below atmospheric pressure. A reduced pressure is preferably below 0.1 bar. The wording "saturated saline" refers to a saturated aqueous NaCl solution. According to an embodiment of the present invention a method for the diagnosis of cancer in vivo is provided. Said method comprises the step of providing a prodrug for targeting of lipid metabolism to a mammal, wherein the prodrug comprising a detectable label, followed by detecting and/or localizing the label by an imaging technique. The prodrug is selected from
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alkyl, alkenyl or alkynyl, n = 1 - 8. This highly selective enrichment in tumor cells may be used in diagnosis or treatment of said tumors by employing detectable labels or therapeutic residues attached to the present prodrugs.
Free fatty acids represent the substrates of lipid metabolism, lipid conversion and lipid storage particularly in the liver and adipose tissue and energy production in heart and skeletal muscles. FFA are therefore incorporated by heart and liver in great amounts after intravenous injection and dependent from work load and hormonal regulation of the skeletal muscles. FFA further represent important substrates for biosynthesis of cellular membranes, modification of proteins, transcriptional regulation and intracellular signal transduction (Berk, P. D. (1996). "How do long-chain free fatty acids cross cell membranes" Proc Soc Exp Biol Med 212(1): 1 - 4). Energy demand of the tumor and energy metabolism, of, for example, the prostate carcinoma, is very low due to a slow growth with a tumor cell growth fraction of around 1% and a very low division rate of tumor cells in comparison to energy demand of the heart muscle or skeletal muscles under workload. The contribution to lipid metabolism is insignificant in comparison to e.g. the liver and membrane biosynthesis of the slow growing tumor cells is notably reduced in comparison to fast proliferating tissue, such as hematopoietic bone marrow or mucosa of the small intestine.
The fast and intensive uptake of substrates of fatty acid metabolism in tumor cells, of e.g. the prostate cancer, is therefore unexpected and surprising. Examples of such tumors comprise inter alia prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancer, malignant lymphoma, neoplasia of the hematopoietic system and musculoskeletal tumors. Characteristic for some of the faster growing tumors is also that they supply their energy demand by highly increased glucose consumption. (Reske, S. N. and J. Kotzerke, Eur J Nucl Med 28 (11) (2001), pp. 1707-23; Czernin, J. and M.-E. Phelps, Annu Rev Med, 53 (2001), pp. 89-112; Gatenby, R. A. and R. J. Gillies, Nature Cancer 4, November (2004), pp. 891-899).
According to another embodiment of the present invention the cancer is characterized by increased consumption of substrates of tumoral lipid metabolism. Not limiting examples of such cancers/tumors include prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancers, malignant lymphomas, neoplasias of the hematopoietic system and musculoskeletal tumors.
Preferably, a method for the diagnosis of prostate carcinoma (PCa) is provided. It has been found that by employing the present prodrugs for targeting of lipid metabolism the differentiation from benign hyperplasia and focal chronic prostatitis is significantly facilitated.
According to an embodiment the prodrug further comprises a linker arranged between said prodrug and said detectable label, in that one end of the linker or spacer is attached to the prodrug, whereas the other end is attached to the detectable label. The linker is preferably selected in a manner to provide good in vivo pharmacokinetics, inter alia by enabling rapid uptake of the prodrug in tumor tissue.
According to yet another embodiment said detectable label is a radioactive label. By using a suitable radioactive label detection with PET, preferably PET/CT, is enabled thus facilitating the localization of tumors in healthy tissue. Another advantage resides in that even new formed metastasis of few cells may be localized due to the high sensitivity of PET. The radioactive label may be for example of 11C, 13N, 18F, 62Cu, 64Cu, 68Ga, 75Br, 76Br, 77Br, 8OmBr, 86Y or 124I. Preferably, radioactive halogens are employed. More preferably, 11C and 18F are used due to their minor influence on the interaction between enzyme and prodrug.
According to an embodiment said imaging technique is selected from positron-emission- tomography (PET), and positron-emission-tomography/computer tomography (PET/CT). PET/CT is preferred due to the improved possibilities of visualizing/indicating the position and dimensions of a tumor in healthy tissue. Another advantage resides in that even metastasis comprising few tumor cells may be detected and localized. The metastasis comprise preferably less than 105 tumor cells, more preferably less than 104 tumor cells and most preferably less than 102 tumor cells.
According to a preferred embodiment of the present invention a prodrug of lipid metabolism, wherein said prodrug is selected from
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alkyl, alkenyl or alkynyl, n = 1 - 8. Preferred prodrugs comprise malonic, alpha - methyl - substituted carboxylic acid derivatives with a single substituent, malonic acid monomethylester and malonic acid monoamide. It will be appreciated that the synthesis of appropriate prodrugs for targeting of lipid metabolism is well within the knowledge of the skilled person and that the skilled artisan is not restricted to a particular route of synthesis.
P-Iodbenzoyl-l l-aminodecylmalonic acid (standard) and 11-Aminoundecylmalonic acid (precursor) may be produced starting from DMUSM in three steps as outlined in figure 1. The corresponding radio labelled malonic acid derivate may be obtained by acetylating e.g. 11 -aminodecylmalonic acid with p- I-benzene-N-hydroxysuccinate. It will be appreciated that other radio labels or therapeutic residues may be attached in a similar manner.
Starting from tetradecanic acid uC-dodecylmalonic acid may be produced (cf. fig. 2). By addition of lithium diisopropylamide (LDA) in presence of hexamethylphosphortriamide (HMPT) the precursor is generated in situ. By converting with 11CO2, followed by hydrolysis, the 11C marked product is obtained. The corresponding standard is obtained by hydro lysing of the dimethylester, which is obtained by alcylation of sodium dimethylmalonate with dodecylbromide.
2-Methyl-13-(methylsulfonyloxy)methyltridecanoate, serving as precursor for the preparation of 13-[i8F]fluoro-2-methyItridecanic acid, is synthesised starting from 2-methyldimethyl- malonate. In the same manner 13-fluoro-2-methyltridecanic acid may be obtained. Synthesis of 3-Hydroxy-(2S)-methylmethylpropionate is known in the state of the art.
According to an embodiment, a synthesis of 13-iodotridecen-12-ylmalonic acid as well as 13- [*I]-iodotridecen-12-ylmalonic acid are synthesized are exemplary outlined in fig.4.
Quality control of the compounds may be easily checked by high performance liquid chiOmatographie (HPLC) or in case of e.g. 18F labelling or another labelling with a halogen by comparison of the retention times of the radio labelled products with those of the respective "cold" standard compounds.
The prodrug preferably comprises a linker arranged between said prodrug and said detectable label. The linker may be selected in a manner bestowing the present prodrug advantageous properties, such as good in vivo pharmacokinetics.
The detectable label preferably comprises a chemoluminescent, fluorescent, bioluminescent, radioactive label or a label detectable by PET, CT or MRT. It will be appreciated that also in case of chemoluminescent, fluorescent, bioluminescent or radioactive labels also in vitro approaches may be used. In vitro assay for the diagnosis of cancer may for example based on interaction with enzymes, e.g. racemases, characteristic for the presence of a tumor from the body. Accordingly, a method is envisaged comprising the steps of providing a sample from a mammal, optionally purifying the sample, adding a present prodrug exhibiting one of the above mentioned markers and detecting the degradation products caused by the activity of enzymes characteristic for the presence of a tumor. It will be appreciated that such methods are well known to the skilled person.
According to an embodiment of the present invention, prodrug may be alpha - methyl - carboxylic or malonic acids and their derivatives. According to another embodiment of the present invention, the prodrug is 2-[l l-[18F]fluoroundecyl]malonic acid (designated [F18]- FSU 01), which has been found to be incorporated in tumor tissue, such as prostate tumor tissue, fast and in high amounts.
According to an embodiment of the present invention, the aliphatic substituent may be selected from C8 - C20 linear alkyl or C8 - C20 branched alkyl. Instead of an aliphatic substituent also an aromatic substituent may be employed. According to another embodiment of the present invention, the aliphatic substituent may be C9, CI l, C13, C15, C17 or C19 linear alkyl. According to a further embodiment of the present invention, the aliphatic substituent may be Cl 1 linear alkyl.
According to an embodiment of the present invention, the detectable marker may be selected from the group consisting of 11C, 13N, 18F, 32P5 35S5 64Cu, 62Cu, 67Cu, 67Ga, 68Ga, 75Br, 76Br, 77Br,S0mBr, S6Y, 89Sr, 88Y5 90Y, 99mTc, 111In, 121I, 123I, 124I, 125I, 127I, 131I, 153Sm, 165Dy; 169Er, 177Lu, 178Ta, 186Re, ISSRe 295mPt, 211At, 213Bi, 225Ac. According to another embodiment of the present invention, the diagnostic residue may be 11C, 18F or 123I.
According to an embodiment of the present invention, the therapeutic residue may be preferably selected from the group consisting Of 32P, 67Cu, 89Sr, 88Y, 90Y, 1231, 1251, 131I, 153Sm, 165Dy; 169Er, 177Lu, 178Ta, 186Re, 188Re, 195mPt, 211At, 213Bi, 225Ac. According to another embodiment of the present invention, the therapeutic residue may be 90Y.
According to an embodiment of the present invention, the detectable marker may be 18F, 11C or 123I. According to an embodiment of the present invention, the detectable marker may be 18F and 123I. Rl and R2 may be independently selected from H, Cl - C8 linear alkyl, C2 - C8 branched alkyl, C3 - C8 linear alkenyl, C4 - C8 branched alkenyl, C3 - C8 linear alkynyl, or C4 - C8 branched alkynyl. According to an embodiment of the present invention, Rl and/or R2 may be H or Cl - C2 linear alkyl, since these residues may easily react with enzymes in tumor tissue due to a reduced steric hindrance. According to another embodiment of the present invention, Rl and/or R2 may be H.
According to still another embodiment of the present invention a method for the treatment of cancer is provided. The method comprises the steps of providing a prodrug for targeting of lipid metabolism to a mammal, wherein said prodrug comprises a therapeutic residue, administering said prodrug to a mammal. The prodrug may be selected from
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alky L alkenyl or alkynyl, n = 1 - 8.
Since the neoplastic tissue, i.e. the tumor tissue, exhibits a considerable higher uptake of the present prodrugs, said tissue is subjected to the radiation originating from the therapeutic residue. It will be appreciated that the therapeutic residue may be selected in order to correspond to a suitable detectable label, in that upon administering of the present prodrug not only enrichment in the target tissue and elimination from the body, but also the activity of the therapeutic residue on the tumor tissue may be observed.
In addition to the good in vivo pharmacokinetics of the present prodrugs, i.e. fast and selective enrichment in tumor tissue and fast degradation and elimination from the body, the pharmacokinetics may be further influenced by choice of a suitable linker.
According to an embodiment, said therapeutic residue may be selected from the group consisting Of 32P, 67Cu, 89Sr, 88Y, 90Y5 123I, 125I5 131I, 153Sm, 165Dy, 169Er5 177Lu, 178Ta5 186Re5 188Re, 195mPt, 211At5 213Bi, and 225Ac, According to an embodiment of the present invention, the therapeutic residue may be identical with a detectable label.
According to an embodiment of the present invention, prodrug may be alpha - methyl - carboxylic or malonic acids and their derivatives.
According to an embodiment of the present invention, the aliphatic substituent may be selected from C4 - C25 linear alkyl or C4 - C25 branched alkyl. Instead of an aliphatic substituent also an aromatic substituent may be employed. According to another embodiment of the present invention the aliphatic substituent may be CI l5 C13 or Cl 5 linear alkyl. According to a further embodiment of the present invention the aliphatic substituent may be CI l linear alkyl.
Rl and R2 may be independently selected from Cl - C8 linear alkyl, C3 - C8 branched alkyl, C3 - C8 linear alkenyl, C4 - C8 branched alkenyl, C3 - C8 linear alkynyl, and C4 - C8 branched alkynyl. According to another embodiment of the present invention Rl and/or R2 may be H or Cl - C2 linear alkyl, since these residues may easily react with enzymes in tumor tissue due to a reduced steric hindrance. According to a further embodiment of the present invention Rl and/or R2 may be H.
According to yet another embodiment, a cancer to be treated is selected from prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancers, malignant lymphomas, neoplasias of the hematopoietic system and musculoskeletal tumors.
According to still another embodiment, the prodrug is administered by intravenous, subcutaneous, intramuscular or intracavitary injection.
According to yet another preferred embodiment the prodrug is contained in a pharmaceutical composition.
Said composition may be in the form of a parenteral formulation since due to the lipophilic nature of said compound administration via injection is advisable. However, other kind of formulations, such as oral formulations, may be envisaged as well. The methods of preparation may include the step of bringing the active compound into association with a carrier, which constitutes of one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into desired formulations.
Suitable compositions for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or a suspension in an aqueous or non- aqueous liquid; or as an oil-in- water or water-in-oil emulsion. Such compositions may be prepared by any suitable method of pharmacy, which includes the step of bringing into association the active compound and a suitable carrier. In general, the compositions are prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture. For example, a tablet may be prepared by compressing or molding a power or granules containing the active compound, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, and/or surface active/dispensing agent(s). Molded tablets may be prepared by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder. A syrup may be made by adding the active compound to a concentrated aqueous solution of a sugar, for example sucrose to which may also be added any accessoiy ingredient(s). Such accessory ingredient(s) may include flavourings, suitable preservatives, an agent to retard crystallization of the sugar, and an agent to increase the solubility of any other ingredient, such as a polyhydric alcohol, for example glycerol or sorbitol. Compositions for oral administration may optionally include enteric coatings known in the art to prevent degradation of the compositions in the stomach and provide release of the drug in the small intestine. Compositions suitable for buccal or sub-lingual administration include lozenges comprising the active compound in a flavoured base, usually sucrose and acacia or tragacanth and pastilles comprising the compound in an inert base such as gelation and glycerin or sucrose and acacia. Compositions suitable for parenteral administration comprise sterile aqueous and nonaqueous injection solutions of the active compound, which preparations are preferably isotonic with the blood of the intended recipient.
These preparations may contain anti-oxidants, buffers, bacteriostats and solutes, which render the compositions isotonic with the blood of the intended recipient. Aqueous and non-aqueous sterile suspensions may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried or a lyophilized condition requiring only the addition of the sterile liquid carrier, for example, saline or water- for- injection immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
According to still another embodiment the use of the present prodrugs for targeting of lipid metabolism for the treatment and/or prevention of cancer is envisaged.
It is to be understood that the above description is intended to be illustrative only and not restrictive. Many embodiments will be apparent to those of skill in the art upon reviewing the above description. By way of example, the invention has been described preliminary with reference to diagnosis of [FlS]-FSU 01. It should be clear that all kinds of suitable therapeutic residues and detectable labels may be synthesized and attached to the present prodrugs for targeting of lipid metabolism. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the appended claims, along with the full scope of equivalents to which such claims are entitled.
Examples
The following PCa-cell lines were used: LNCaP (human prostate carcinoma, DSMZ Cat.-No. ACC 256) and TRAMP (murine prostate adenocarcinoma, obtainable from LGC Cat. No. CRL-2730; Greenberg, N.M. et al., Proc Natl Acad Sci U S A, 92(8) (1995) pp. 3439-43). PC3 (DSMZ Cat.-No. ACC 465) was used as reference. Human colon adenocarcinoma CX-I were obtained from DSMZ Cat.-No. ACC 129.
PC3 was cultivated/maintained in 50% Ham's F12 with 50% RPMI, 10% FKS, 1% penicillin + streptomycin, 1% L-glutamate. LNCaP was cultivated/maintained in RPMI with 10% FKS, 1% penicillin + streptomycin, 1% L-glutamate, 1% sodium pyruvate, 1% not essential amino acids. TRAMP cells were cultivated/maintained in DMEM (high glucose), 5% Nu-serum IV, 5 % FKS; 1% penicillin + streptomycin, 10"s M dihydrotestosterone, 5 μg/ml insuline. CX-I cells were cultivated/maintained in DMEM (high glucose), 10% FKS.
For analysis of standard and radiolabeled compounds a Dionex HPLC System with UV- detector (210 nm) and radio detector has been employed. Stationary phase: LiChrospher Select B 5μ, 250x4.6mm, Chromatographic Service; mobile phase: CH3CN/H2O 50/50 v/v (0.1% TFA); flow: 2 ml/min.
[18F]-Synthesis has been performed according to manufactures instructions with a remote controlled synthesis unit by Nuclear Interface (GE) with semi preparative Radio-HPLC- System (stationary phase: LiChrospher Select B 5μ, 250x10 mm, Chromatographic Service; mobile phase: CH3CN/H2O 60/40 v/v (0.1% TFA); flow: 5 ml/min.
Radioactivity was measured according to manufactures instructions with a γ-counter (Cobra 2, GR Healthcare / Packard Instruments, Palo Alto, USA).
Example 1
Synthesis of 1 -bromo- 11 -fluoroundecane:
11-Bromoundecanol-l (4.1 g, 16.32 mmol) was added (teflon bottle) to DAST (5 ml, 6.1 g, 37.84 mmol) under Ar-atmosphere. The mixture was stirred for 10 min. at room temperature and afterwards heated to 50-55 C and stirred for other 6 hours. After cooling to room temperature, the mixture is diluted with ice water (200 ml) and neutralised (pH=7 ) with sodium bicarbonate. The obtained emulsion was extracted with pentane (2 x 100 ml). The pentane solution was washed with water (3 "< 50 ml) and saturated saline (50 ml) and afterwards dried over magnesium sulfate. Following filtration, the solvent was distilled under reduced pressure. The residue was purified by column chromatography on silica gel (mobile phase: pentane). The target compound (1.95 g, 47%) was obtained as colourless liquid. Rf = 0.38 (pentane). 1H-NMR(CDCl3, 500 MHz): δ = 1.20-1.35 (m, 10 H), 1.35-1.46 (m, 4 H), 1.67-1.74 (m, 2 H), 1.84 (dt, J = 14.9, 6.7 Hz), 3.39 (t, J = 6.9 Hz), 4.42 (dt, J = 47.8, 6.7 Hz); 13C-NMR(CDCl3, 125.77 MHz): δ = 25.1 (d, J = 5.5 Hz), 28.15, 28.7, 29.2, 29.38, 29.39, 29.44, 30.4 (d, J = 19.3 Hz), 32.8, 34.0, 84.2 (d, J = 164.0 Hz). MS (EI): m/z = 254, 252 (0.5) [M+], 173.2 (6) [CnH22F+], 151, 149 (10) [C5H10Br+], 135, 137 (100) [C4H8Br+], 117 (7) [C7H14F+], 97 (20) [C7H13 +], 83 (10), 71 (15), 69 (34) [C5H9 +], 55 (38) [C4H7 +].
Example 2 Synthesis of dimethyl- 11-fluoroundecylmalonate:
To a solution of compound 1 (1 g, 3.95 mrnol) in butanone-2 (20 ml) sodium iodide (0.62 g, 4.15 mmol) was added. The reaction mixture was heated for 16 h under reflux. The reaction mixture was allowed to cool to room temperature and diluted with water (100 ml) and 10% Na2S2O3-solution (40 ml). The resulting emulsion was extracted with pentane (2 x 100 ml). The pentane solution was washed with water (5 x 50 ml) and saturated saline (50 ml) and afterwards dried over magnesium sulfate. Following filtration, the solvent was distilled under reduced pressure. 1-Iodo-l l-fluoroundecane (1.1 g, 93%) was obtained as colourless liquid. NaH (0.119 g, 2.97 mmol; 60%ige suspension in paraffin oil) was suspended in tetrahydrofurane (THF) (10 ml) under argon atmosphere and stirred at room temperature. Dimethylmalonate (0.342 ml, 0.394 g, 2.97 mmol) was added drop wise within 3 min. (exotherm reaction, gas evolution) and stirred afterwards for 5 min. Following this, first 1- iodo-11-fluoroundecane (0.51 g, 1.70 mmol) and afterwards DMSO (10 ml) were added. The reaction mixture was stirred for 15 hours. THF was distilled under reduced pressure. The residue was diluted with 1 M HCl (10 ml) and water (200 ml). The resulting emulsion was extracted with pentane (100 ml). The pentane solution was washed with 1 M HCl (50 ml), water (10 * 50 ml) and a saturated saline solution (50 ml) and afterwards dried over magnesium sulfate. The solvent was distilled after filtration and the residue was purified by column chromatography on silica gel (mobile phase: EtOAc.hexane = 1 :10). The target compound (0.43 g, 81%) was obtained as colourless liquid. Rf = 0.29 (EtOAc :hexane = 1 :10). 1H-NMR(CDCl3, 500 MHz): δ = 1.15-1.35 (m, 16 H), 1.67-1.74 (m, 2 H), 1.87 (dd, J = 7.3, 7.3 Hz, 2 H), 3.34 (t, J = 7.3 Hz, 1 H), 3.72 (s, 6 H), 4.42 (dt, J = 47.6, 6.3 Hz; 2 H); 13C-NMR(CDCl3, 125.77 MHz): δ = 25.1 (d, J = 5.5 Hz), 27.3, 28.83 (x 2), 29.15, 29.20, 29.3, 29.4 (x 2), 30.4 (d, J = 19.2 Hz), 51.7, 52.4, 84.2 (d, J = 164.0 Hz), 170.0. MS (ESI): positive mode m/z = 631.4 ([2M + Na]+), 327.2 ([M + Na]+), 305.2 ([M + Na]+); ESI HRMS: calculated for C16H29FO4Na+: 327.19421; found: 327.19410; calculated for C16H30FO4 +: 305.21226; found: 305.21222.
Example 3:
Synthesis of 11-fluoroundecylmalonic acid:
To a solution of compound 2 (0.35 g, 1.21 mmol) in methanol (5 ml) 10 M NaOH (1.3 ml) and water (1.3 ml) were added. The reaction mixture was stirred for 4 hours at 50°C. After cooling to room temperature the mixture was diluted with water (25 ml). The resulting aqueous solution was underlayed with hexane (3 x 20 ml). The hexane phase was discarded after phase separation. The aqueous phase was acidified to pH 1 with 1 M HCl. The resulting suspension was extracted with ether (100 ml). The ether solution was washed with 1 M HCl (2 x 10 ml), water (3 x 20 ml) and saturated saline (2 x 20 ml) and afterwards dried over magnesium sulfate. The solvent was distilled under reduced pressure after filtration. The target compound (0.43 g, 81%) was obtained as colourless solid after recrystallization of the residue from ether/pentane. Rf = 0.20 [EtOAc±exane = 1 :3 (3% AcOH)]. 1H- NMR[(CD3)2SO, 500 MHz]: δ = 1.25-1.44 (m, 16 H), 1.55-1.75 (m, 2 H)5 1.68 (dd, J = 7.0, 7.0 Hz, 2 H), 3.16 (t, J = 7.2 Hz, 2 H)5 4.40 (dt, J = 47.6, 6.2 Hz), 12.50-12.90 (br, 1 H); 13C- NMR[(CD3)2SO, 125.77 MHz]: δ = 24.6 (d, J = 5.5 Hz), 26.8, 28.4, 28.6, 28.7, 28.8, 28.86, 28.88 (x 2), 28.89, 29.8 (d, J = 19.2 Hz)5 51.6, 84.2 (d, J = 162.3 Hz), 171.0. MS (ESI): positive Mode m/z = 299.2 ([M + Na]+), 277.2 ([M + Na]+); ESI HRMS: calculated for C14H25FO4Na+: 299.16291; found: 299.16293; calculated for C14H26FO4 +: 277.18096; found: 277.18104.
Example 4:
Synthesis of dimethyl- 11-mesyloxyundecylmalonate:
Sodium methylate (0.848 g, 15.55 mmol) was dissolved in methanol (20 ml) and heated for 15 min. under reflux. Within 10 min. dimethylmalonate (1.78 ml, 2.054 g, 15.55 mmol) was added drop wise to the receiver. After 15 min. I l-bromoundecanol-1 (3.516 g, 13.99 mmol) dissolved in methanol (10 ml) was included drop wise within 15 min. The reaction mixture was heated for 20 hours under reflux. After cooling to room temperature and concentration under reduced pressure, the obtained residue was dissolved in ether (100 ml) and 1 M HCl (40 ml). The ether solution was washed with 1 M HCl (40 ml), water (3 * 50 ml) and saturated saline (2 x 20 ml) and afterwards dried over magnesium sulfate. The solvent was distilled under reduced pressure after filtration. The residue was purified by column chromatography on silica gel (mobile phase: EtOAc:hexane = 1:3, Rf = 0.15). Dimethyl-11- hydroxyundecylmalonate (1.81 g, 43%) was obtained as colourless oil. Dimethyl-11- hydroxyundecylmalonate (1.55 g, 5.13 mmol was dissolved in dichloromethane (20 ml) and triethylamine (0.67 ml, 0.928 g, 6.67 mmol) under ice cooling, stirring and argon atmosphere. Within 5 min. mesylchloride (0.52 ml, 0.763 g) was added drop wise and the mixture was stirred for 3 hours under ice cooling. The temperature of the reaction mixture was increased to room temperature by adding of water (50 ml) and ether (100 ml). The obtained organic layer was washed with water (2 x 30 ml), 1 M KHSO4 (2 x 30 ml), water (2 x 30 ml) and saturated saline (50 ml) and afterwards dried over magnesium sulfate. After filtration of the dried organic phase through a pad of silica gel (2 cm), distillation of the solvent was performed under reduced pressure. Toluene (50 ml) was three times distilled from the residue. The target compound (1.92 g, 98%) was obtained as yellowish oil, which crystallizes slowly as solid. Rf = 0.17 (EtOAc:hexane = 1 :3). 1H-NMR(CDCl3, 500 MHz): δ = 1.20-1.34 (m, 14 H)5 1.34-1.45 (m, 2 H), 1.72 (dt, J = 15.0, 7.2 Hz, 2 H), 1.87 (dd, J = 7.2 Hz, 2 H), 2.98 (s, 1 H), 3.33 (t, J = 7.5 Hz5 1 H)5 3.71 (s, 6 H)5 4.20 (t, J = 6.5 Hz; 2 H); 13C- NMR(CDCl3, 125.77 MHz): δ = 25.4, 27.3, 28.8, 29.0, 29.09, 29.12, 29.2, 29.3, 29.4 (x 2), 37.3, 51.7, 52.4, 70.1, 169.9. MS (ESI): positive mode m/z = 783.4 ([2M + Na]+), 403.2 ([M + Na]+), 381.2 ([M + Na]+); ESI HRMS: calculated for C17H32O7SK+: 419.15003; found: 419.15000; calculated for C17H32O7SNa+: 403.17610; found: 403.17618; calculated for C17H36O7SN+: 398.22070; found: 398.22074; calculated for C17H33O7S+: 381.19415; found: 381.19419.
Example 5
Synthesis of 2-(l l-[18F]fluoiOundecyl)malonic acid ([F18]-FSU 01):
[18F]fluoride was produced by 18O(p, n)18F nuclear reaction by proton irradiation of enriched [18O] water (2 ml, RWE5 degree of enrichment >95%) using a PETtrace cyclotron (Ep = 16,5 MeV, GE). For radiosynthesis a remote controlled synthesis unit of Nuclear Interface (GE) was employed. [18F]fluoride produced by the cyclotron was fixed on a conditioned (10 ml 1 M NaHCO3, 10 ml water) anion exchange cartridge (Sep-Pak light, Accell Plus QMA5 Waters) and thereby separated from the [18O]water-matrix. [] SF]fluoride was eluted with 360 μL 0,066 M KOH and subsequently 20 mg Kryptofix 2.2.2. in 1 ml acetonitrile was added to a [18F] fluoride solution. The radionuclide was activated for the radiolabeling by means of subsequent azeotropic distillation for 6 min. under vacuum (approx. 6 mbar) until dryness at first 950C and later 100°C. The radiolabeling was conducted by addition of compound (9 mg, 23.65 μmol) obtained in example 4 in acetonitrile (1 ml) and heating at 850C for 10 min. Product was obtained afterwards by hydrolysis of the intermediate dimethyl ester with 400 μl 1 M NaOH for 10 min. at 7O0C. The reaction matrix was cooled to 300C and 2 ml HPLC- eluent (acetonitrile/water 60/40 v/v) and 300 μL acidic acid were added. Not converted [18F] fluoride was removed from the neutralized and diluted reaction mixture by means of a Al2θ3-cartiϊdge (Sep-Pak light, Alumina N5 Waters) and purified by semipreparative HPLC (stationaiy phase: LiChrospher Select B 5μ, 250x10 mm, Chromatographic Service; mobile phase: CH3CN/H2O 60/40 v/v, 0,1 % TFA; flow: 5 ml/min; radioactivity detector and UV- detector (210 nm)) by means of an automated injection unit (flow-detector). The product fraction was isolated after approx. 7 min. by valve switching and was diluted with 50 ml water. Afterwards solid phase extraction was carried out by means of a C- 18 cartridge (Sep- Pak light, C- 18, Waters). Elution was performed with 1 ml EtOH, followed by 10 ml isotonic NaCl-solution. Starting from approx. 45 GBq [18F]fluoride within 60 min. approx. 5 - 8 GBq product were obtained with a radiochemical purity > 95% Quality control
Quality control of the produced fluorine- 18 labelled fatty acids was performed by an analytical Radio-HPLC-system (stationary phase: LiChrospher Select B 5μ, 250x4,6 mm, Chromatographic Service; mobile CH3CN/H2O 50/50 v/v, 0,1 % TFA; flow: 2 ml/min; radio activity detector und UV-detector (210 nm)). Product identification was performed by comparison of the retention times of the products with those of the respective "cold" standard compounds (the compounds obtained in example 3).
Example 6
Synthesis of dimethyl-2-tridec- 12-ynylmalonate:
Tridecynyl- 12-01-1 (4.95 g, 29.71 mmol), Ph3P (11.69 g, 44.57 mmol) and imidazole (3.034 g, 44.57 mmol) were dissolved in THF (50 ml) under argon atmosphere and ice cooling. Iodine (10.406 g5 41.0 mmol) in THF (20 ml) was added drop wise within 20 min. The reaction mixture was stirred for 2 hours at room temperature. Afterwards saturated Na2S2O3 solution (3 ml) was added drop wise within 2 min. The reaction mixture was dried over magnesium sulfate. The obtained residue was extracted with pentane (20 x 50 ml). The solvent was distilled under reduced pressure and the residue was purified by column chromatography on silica gel (mobile phase: petrolether 40/60). 13-Iodotridecyn-l (6.4 g, 93%) was obtained as colourless liquid. Rf = 0.24 (petrolether 40/60). 1H-NMR(CDCl3, 500 MHz): δ = 1.23-1.33 (m, 11 H), 1.34-1.41 (m, 3 H), 1.49-1.55 (m, 2H), 1.81 (dt, J = 14.5, 7.2 Hz, 2 H) 1.92 (t, J = 2.7 Hz, 1 H), 2.17 (dt, J = 1.8, 7.2 Hz, 2 H), 3.18 (t, J = 7.0 Hz, 2 H). NaH (0.92 g, 22.86 mmol; 60% suspension in paraffin oil) was suspended in tetrahydrofurane (50 ml) under argon atmosphere and stirred at room temperature. Within 3 min. dimethylmalonate (2.62 ml, 3.02 g, 22.86 mmol) was added drop wise (exotherm reaction, gas evolution). The reaction mixture was stirred for 5 min. Afterwards, 13- iodotridecyn- 1 (4.0 g, 13.06 mmol) and DMSO (50 ml) were added successively and the reaction mixture was stirred for another 20 hours. THF was distilled under reduced pressure. The residue was diluted with 1 M KHSO4 (25 ml) and water (500 ml). The obtained emulsion was extracted with pentane (400 ml). The pentane solution was washed successively with 1 M KHSO4 (50 ml), water (5 x 100 ml) and saturated saline (50 ml). The organic phase was dried over magnesium sulfate. After filtration of the dried organic phase, the solvent was distilled under reduced pressure. The residue was purified by column chromatography on silica gel (mobile phase: petrolether 40/60:aceton = 10:1). The target compound (3.25 g, 80%) was obtained as colourless oil, which crystallizes slowly as colourless solid. Rf = 0.34 (petrolether 40/60:aceton = 10:1). 1H-NMR(CDCl3, 500 MHz): δ = 1.12-1.33 (m, 14 H), 1.33-1.42 (m, 2 H), 1.45-1.55 (m, 2 H), 1.87 (dd, J = 7.3, 7.3 Hz, 2 H)5 1.91 (t, J = 2.7 Hz, 1 H), 2.16 (dt, J = 2.7, 7.1 Hz5 2 H), 3.34 (t, J = 7.8 Hz, 1 H), 3.72 (s, 6 H); 13C-NMR(CDCl3, 125.77 MHz): δ = 18.4, 27.3, 28.5, 28.7, 28.8, 29.1, 29.2, 29.3, 29.4 (x 2), 29.5, 51.7, 52.4, 68.0, 84.8, 169.9.
Example 7
Synthesis of dimethyl- 13-tributylstannyltridec- 12-enylmalonate: Compound 6 (1.0 g, 3.22 mmol), tributylstannan (1.06 ml, 1.17g, 4.02 mmol) and AIBN (20 mg)were dissolved in toluene (10 ml) under argon atmosphere. The solution was carefully degassed and then stirred for 2 hours at 8O0C and for 1 hour at 1050C. After cooling the reaction mixture to room temperature, the solvent was distilled under reduced pressure. The residue was purified by column chromatography on silica gel (0.1% CaO) (mobile phase: petrolether 40/60:aceton = 12:1). The target compound (0.95 g, 49%) was obtained as a mixture of E/Z isomers (70:30) as colourless oil. Rf = 0.14 (petrolether 40/60 : aceton = 12:1). 1H-NMR(CDCl3, 500 MHz): δ = 0.75-0.90 (m, 14 H), 1.20-1.40 (m, 25 H), 1.45-1.55 (m, 6 H), 1.87 (dd, J = 7.3, 7.3 Hz, 2 H), 2.01 (dt, J = 6.8, 6.8 Hz, 0.6 H), 2.11 (dt, J - 6.0, 6.0 Hz, 1.4 H), 3.34 (t J = 7.8 Hz, 1 H), 3.72 (s, 6 H), 5.35-6.05 (m, 1.4 H), 6.44-6.65 (m, 0.6 H); 13C-NMR(CDCl3, 125.77 MHz): δ = 9.4, 10.25; 13.7, 27.26, 27.33, 27.35, 28.87, 28.94, 29.1, 29.2, 29.3, 29.5, 29.60, 29.62, 29.9; 37.2, 37.9; 51.7, 52.4; 126.9, 127.6; 149.3, 149.9; 170.0.
Example 8:
Synthesis of dimethyl- 13-iodtridec- 12-enylmalonate:
To a solution of compound 7 (0.3 g, 0.50 mmol) in dichloromethane (5 ml) iodine (0.216g, 0.5 mmol) in a solution of dichloromethane (3 ml) was added drop wise. The reaction mixture was stirred for 10 minutes at room temperature. Thereafter, a saturated solution of Na2S2O3 (0.5 ml) was added and the reaction mixture was stirred for further 5 min. at room temperature. After dilution of the reaction mixture with dichloromethane, (30 ml) 10% KF solution (30 ml) was added and was intensively stirred for 1 hour at room temperature. The aqueous phase was discarded and the organic phase was dried over magnesium sulfate. After filtration through a pad of silica gel (1 cm) the solvent was distilled under reduced pressure and the residue was purified by column chromatography on silica gel (mobile phase: petrolether 40/60:acetone = 10:1). The target compound (0.212 g, 100%) was obtained as E/Z- isomer mixture (70:30). It was obtained as colourless oil. Rf = 0.20 (petrolether 40/60:aceton = 10:1). 1H-NMR(CDCl3, 500 MHz): δ = 1.15-1.33 (m, 16 H), 1.33-1.40 (m, 2 H), 1.87 (dd, J = 6.9, 6.9 Hz5 2 H)5 2.03 (dt, J = 7.2, 7.0 Hz, 1.4 H), 2.11 (dt, J = 2.3, 1.7 Hz, 0.6 H), 3.34 (t, J = 7.6 Hz, 1 H), 3.72 (s, 6 H), 5.96 (d, J = 14.3 Hz, 0.7 H), 6.13-6.19 (m, 0.6 H), 6.49 (dt, J = 14.3, 7.2 Hz, 0.7 H); 13C-NMR(CDCl3, 125.77 MHz): δ = 27.3, 27.9, 28.3,
28.8, 28.9, 29.2, 29.26, 29.31, 29.47, 29.51; 34.7, 36.0; 51.7, 52.4; 74.2, 82.1; 141.5, 146.8; 170.0.
Example 9:
Synthesis of 13-iodotridec-12-enylmalonic acid: Compound 8 (0.2 g, 0.50 mmol) was dissolved in ethanol (10 ml) and 1 M NaOH (2 ml) was added and refluxed for 15 minutes. After cooling to room temperature, the reaction mixture was diluted with 1 M KHSO4 (5 ml) and extracted with ether (70 ml). The organic phase was separated and consecutively washed with 1 M KHSO4 (20 ml), water (3 x 20 ml) and saturated saline (2 >- 20 ml). After drying the organic phase over magnesium sulfate, the solvent was distilled by reduced pressure. The remaining residue was recrystallized from ether/pentane. Thereby, the target compound was obtained as a mixture Z/E isomers (30:70), containing less than 5 mol% of the elimination product (tridecyn-12-ylmalonic acid) (0.175 g, > 89%) as colourless solid. Rf = 0.38 [EtOAc:hexane = 3: 1 (3% AcOH)]. 1H-NMR(CDCl3, 500 MHz): δ = 1.0-1.45 (m, 18 H), 1.92 (dd, J = 7.2, 7.2 Hz, 2 H), 2.03 (dd, J = 7.2, 7.2 Hz, 1.4 H), 2.10- 2.25 (m, 0.6 H), 3.42 (t, J = 7.4 Hz, 1 H), 5.96 (d, J = 14.4 Hz, 0.7 H), 6.13- 6.21 (m, 0.6 H), 6.49 (dt, J = 14.4, 7.2 Hz, 0.7 H); 13C-NMR(CDCl3, 125.77 MHz): δ - 27.2,
27.9, 28.3, 28.7, 28.9, 29.1, 29.2, 29.3, 29.47, 29.51 ; 34.7, 36.0: 51.6; 74.2, 82.1; 141.5, 146.8; 175.1.
Example 10:
Synthesis of radioiodinated 13-iodotridec-12-enylmalonic acid:
Na*I (5 μL; solution in 0.05 M NaOH) was added to a solution of compound 7 (20 μl, 9,31 mg/ml in methanol) and acetic acid (1 μl) in ethanol (20 μl) . Afterwards chloroamine Tsolution (4 mg/ml in 66 % methanol) was added and the mixture was stirred for 3 min. The reaction was quenched by adding saturated solution of methionine (10 μl). The protected intermediate was obtained in > 94% yield. The reaction mixture was diluted with ethanol to total volume of 200 μl. 1 M NaOH (50 μl) was added and the reaction mixture was heated for 15 min at 80°C and thereafter cooled to ambient temperature. Trifluoroacetic acid/acetonitiϊle 10/90 v/v (50 μl) was added. The reaction mixture was diluted with acetonitrile/water (0.1 % TFA) 33/67 v/v (750 μl) and separated by a semipreparative HPLC(stationary phase: LiChrospher 60 Select B 5μ RP C-8; 250 x 10 mm, Chromatographic Service; mobile phase gradient: CH3CN with 0,1% TFA and 0.1% TFA5 0.5 min. 50% CH3CN with 0.1% TFA5 5 - 10 min. 50 - 80% CH3CN with 0.1% TFA5 10 - 50 min. 80% CH3CN with 0.1% TFA; flow: 4 ml/min; radio detector and UV-detector (220 nm)). The product containing fraction was collected via valve control,, diluted with water in a ratio of 1 :10 and concentrated via solid phase extraction (Sep-Pak light C- 18, Waters). After washing of the cartridge with 10 ml water, the product was eluted with 0.5 - 1 ml ethanol. The radiochemical yield ranged between 25 - 35 % with respect to the applied radioiodine.
Quality control
Quality control of the produced radio iodine labelled fatty acid was carried out by analytical radio HPLC (stationary phase: LiChrospher Select B 5μ, 250x4,6 mm, Chromatographic Service; mobile phase: CH3CN/H2O 80/20 v/v; flow: 1 ml/min; radio activity detector and TJV-detector (220 nm)). Product identification was carried out by comparison of the retention times of the radio labelled products with that of the respective "cold" standard compound. The radio chemical purity of the product was >95%.
Example 11
Uptake of [F18]-FSU 01 in LNCaP, PC3 and TRAMP cells. PC3 cells serve hereby as androgen independent negative control, whereas LNCap and TRAMP (Greenberg, N.M. et al., Proc Natl Acad Sci U S A5 92(8) (1995) p. 3439-43) cell lines are both androgen dependent, i.e. exhibiting an enzyme expression profile essentially corresponding to that of human prostate cancer cells in vivo.
5* 1O5 cells of each kind (LNCaP, PC3 und TRAMP cells) were plated in 12 well plates, respectively.
The cells were incubated for 24 h in the respective media according to the instructions provided by the German Collection of Microorganisms and Cell Cultures.
Addition of [F18]-FSU 01 in dilution series in respective medium (20 h-value~ 200 MBq /well -> final volume 200 μl on 2000 μl, 1 h- value ~ 10 MBq /well -> 100 μl on 2000 μl medium, 4 h- value ~ 25 MBq /well -> 100 μl on 2000 μl medium.
Incubation of the cells for Ih, 4h and 2Oh at 370C. The medium was removed (tube 1: unbound FSU 01), followed by 2 times gentle rinsing of the cells with PBS (2x 1 ml) (tube 1: unbound FSU 01). Cells were resuspended in 1 ml NaOH and transferred in tube 2 (bound FSU 01). The wells were washed with 1 ml HCL (tube 3: activity sticked to the inner wall)
Determination by γ-counter:
1 h: washing step 10 μl taken from 4.1 ml (2.1 ml medium + 2 ml PBS) 4 h: washing step 10 μl taken from 4.1 ml (2.1 ml medium + 2 ml PBS) 20 h: washing step 10 μl taken from 4.2 ml (2.2 ml medium + 2 ml PBS) cell lysate 1ml (I ml NaOH) HCl washing step (1 ml HCL)
Activity (FSU 01) has been added at 1 and 20 h.
Values at 1 h were determined by removing 100 μl from each well and adding 900 μl PBS (activity per well: x ~ 10 MBq).
Values at 4 h were determined by removing 250 μl from each well and adding 750 μl PBS (activity per well: x - 25 MBq).
Values at 20 h were determined by removing 200 μl from the original solution and adding directly to each well (200 μl per well).
Cellular uptake if FSU 01 in % dosage per 500000 cells has been calculated after 2 h and 24 h incubation (cf. fig. 4). High enrichment of [F-18]FSU 01 in LNCaP and TRAMP carcinoma cells could be detected.
Example 12
Biodistribution of [F-18]-FSU 01 in TRAMP-mice
Determination of [F- 18] - FSLI 01 biodistribution with the γ-counter, immuno histology, at 30 and 60 min. after intravenous application.
Number of the animals: 10 SV40 Tg+ animals, 2 SV40 Tg' animals Approx. 200 μl [F- 18] FSU 01 in isotonic NaCl were applied, (activity of the full and empty syringe as well as of the mouse has been measured in closable plastic bags). The mouse was sacrificed according to the appointments of the German ethic committee and the weight was determined. Tissue and organs (blood, heart, lungs, liver, spleen, kidney, large and small intestines, stomach, muscles, vertebral body, bones, genital tract, caudal) were removed. The weight of empty γ-counter tubes was determined, the tissue/organ included and the weight of the tubes with content determined. The caudal was additionally measured in the activimeter and the tubes were measured in the γ-counter.
Application scheme:
01 :00 Application mouse 3 (30 min for bio distribution, 25 MBq )
01:30 removal of organs of mouse 3 for bio distribution
01:45 Application mouse 4 (60 min for bio distribution, 25 MBq) 02:00 Application mouse 5 (30 min for bio distribution, 25 MBq)
02:30 removal of organs of mouse 5 for bio distribution
02:45 removal of organs of mouse 4 for bio distribution
03:00 Application mouse 6 (60 min for bio distribution, 25 MBq)
03:15 Application mouse 7 (30 min for bio distribution, 25 MBq) 03:45 removal of organs of mouse 7 for bio distribution
04:00 removal of organs of mouse 6 for bio distribution
04:15 Application mouse 8 (60 min for bio distribution, 25 MBq)
04:30 Application mouse 9 (30 min for bio distribution, 25 MBq)
05:00 removal of organs of mouse 9 for bio distribution 05:15 removal of organs of mouse 8 for bio distribution
05:30 Application mouse 10 (60 min for bio distribution, 25 MBq)
06:30 removal of organs of mouse 10 for bio distribution.
Tissue/organ uptake of FSU 01 was calculated 30 and 60 min after intravenous application of FSU 01 in a caudal vein (cf. fig. 6). A high uptake of [F-IS]FSU 01 in prostate primary tumors and lymph node metastasis of the prostate carcinoma may be derived therefrom. In particular, the 18F labeled alkylmalonic acid was enriched in the transgene TRAMP mouse model, for example, in prostate carcinoma and its metastasis in lymph nodes more than 10 -
20 fold in comparison to control tissue, such as periprostatic soft tissues, intestinal tissue, skeletal muscles or normal prostate tissue.

Claims

Claims
1. A method for the diagnosis of cancer in vivo, comprising: providing a prodrug of lipid metabolism to a mammal, said prodrug comprising a detectable label detecting and/or localizing the label by an imaging technique, wherein said prodrug is selected from
CO2H CO2H CO2H
R^ JLXO2H R^ λXONH2 R^ λXONHR1
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alkyl, alkenyl or alkynyl, n = 1 - 8.
2. The method according to claim 1, wherein the cancer is selected from prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancers, malignant lymphomas, neoplasias of the hematopoietic system and musculoskeletal tumors.
4. The method according to claim 1, wherein said prodrug further comprises a linker arranged between said prodrug and said detectable label.
5. The method according to claim 1, wherein said detectable label is a radioactive label or a label detectable by CT or MRT.
6. The method according to claim 1, wherein said imaging technique is selected from positron-emissions-tomography (PET), and positron-emissions-tomography/computer tomography (PET/CT).
7. A prodrug of lipid metabolism, wherein said prodrug is selected from
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alkyl, alkenyl or alkynyl, n = 1 - 8.
8. The prodrug according to claim 7, wherein said prodrug further comprises a linker arranged between said prodrug and said detectable label or said therapeutic residue.
9. The prodrug according to claim 7, wherein said detectable label comprises a chemoluminescent, fluorescent, bioluminescent, radioactive label or a label detectable by CT or MRT.
10. The prodrug according to claim 7, which selected from alpha-methyl-carboxcylic acid.
11. The prodrug according to claim 7, wherein the aliphatic substituent is selected from C4 - C25 linear alkyl, C4 - Cl 5 branched alkyl or C4 - C30 alicyclic.
12. The prodrug according to claim 7, wherein the detectable marker is selected from the group consisting of 11C5 13N, 18F, 32P9 35S5 64Cu, 62Cu5 67Cu, 67Ga, 68Ga, 75Br5 76Br5
77Br5 8001Br5 86Y5 89Sr5 88Y5 90Y5 99mTc5 111In5 121I5 123I, 124I5 125I5 127I5 131I, 153Sm5 165Dy; 169Er5 177Lu5 178Ta5 186Re5 188Re 195mPt5 211At5 213Bi, and 225Ac.
13. The prodrug according to claim 7, wherein the therapeutic residue is selected from the group consisting Of 32P5 67Cu, 89Sr, 88Y5 90Y, 123I5 125I5 131I5 153Sm, 165Dy; 169Er, 177Lu,
178Ta5 186Re, 188Re5 195mPt, 211At5 213Bi5 and 225Ac.
14. The prodrug according to claim 7, wherein Rl and R2 are selected from H5 Cl - C8 linear alkyl, C3 - C8 branched alkyl, C3 - C8 linear alkenyl, C4 - C8 branched alkenyl, C3 - C8 linear alkynyl or C4 - C8 branched alkynyl.
15. A method for the treatment of cancer, comprising: providing a prodrug for targeting of lipid metabolism to a mammal, said prodrug comprising a therapeutic residue administering said prodrug to a mammal, wherein said prodrug is selected from
CO?H CO2H CO2H
R^ JLXO2H R^ JLXONH2 R^ JLXONHR1
wherein R represents an aliphatic, aromatic, heterocyclic, aralkylic, heteroaralkylic, or alicyclic substituent comprising a detectable label or a therapeutic residue. Rl and R2 are independently selected from lower alkyl, alkenyl or alkynyl, n = 1 - 8.
16. The method for the treatment of cancer according to claim 15, wherein said prodrug further comprises a linker arranged between said prodrug and said therapeutic residue.
17. The method for the treatment of cancer according to claim 15, wherein said therapeutic residue is selected from the group consisting of 32P, 67Cu, 89Sr, 88Y, 90Y, 123I, 125I, 131I, 153Sm, 165Dy; 169Er, 177Lu, 178Ta, 186Re, 188Re, 195mPt, 211At5 213Bi, and
225Ac.
18. The method for the treatment of cancer according to claim 15, wherein said therapeutic residue is 32P, 67Ga, 89Sr, 90Y, 111In5 123I, 125I5 131I, 153Sm5 165Dy5 169Er5 177Lu5 178Ta5 186Re5 19SmPt, 211At5 212At5 213Bi5 or 225Ac.
19. The method for the treatment of cancer according to claim 155 wherein said prodrug is alpha-methyl-carboxylic acid.
20. The method for the treatment of cancer according to claim 15, wherein the aliphatic substituent is selected from C4 - C25 linear alkyl, C4 - C25 branched alkyl or C4 - C30 alicyclic.
21. The method for the treatment of cancer according to claim 15, wherein Rl and R2 are selected from Cl - C8 linear alkyl, C3 - C8 branched alkyl, C3 - C8 linear alkenyl, C4
- C8 branched alkenyl, C3 - C8 alkynyl, or C4 - C8 branched alkynyl.
22. The method for the treatment of cancer according to claim 15, wherein the cancer is selected from prostate carcinoma, colorectal carcinoma, breast cancer, lung tumors, tumors of the male or female genitourinary system, malignant melanoma, head and neck cancers, malignant lymphomas, neoplasias of the hematopoietic system and musculoskeletal tumors.
23. The method for the treatment of cancer according to claim 15, wherein the prodrug is administered by intravenous, subcutaneous, intramuscular or intracavitary injection.
24. The method for the treatment of cancer according to claim 15, wherein the prodrug is contained in a pharmaceutical composition.
25. The use of a prodrug of lipid metabolism according to claim 7 for the treatment and/or prevention of cancer.
26. The use of a prodrug of lipid metabolism according to claim 7 for the diagnosis of cancer.
EP08737300A 2008-03-07 2008-03-07 Precursors of lipid metabolism for the diagnosis and treatment of cancer Withdrawn EP2268319A2 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2008/000542 WO2009109798A2 (en) 2008-03-07 2008-03-07 Precursors of lipid metabolism for the diagnosis and treatment of cancer

Publications (1)

Publication Number Publication Date
EP2268319A2 true EP2268319A2 (en) 2011-01-05

Family

ID=41056404

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08737300A Withdrawn EP2268319A2 (en) 2008-03-07 2008-03-07 Precursors of lipid metabolism for the diagnosis and treatment of cancer

Country Status (2)

Country Link
EP (1) EP2268319A2 (en)
WO (1) WO2009109798A2 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE102010010666A1 (en) 2010-03-01 2011-09-01 Sven Reske Use of a fatty acid for the diagnostic and/or therapeutic treatment of a cancer e.g. genitourinary cancer, including prostate cancer, colorectal cancer, breast cancer, lung cancer, malignant melanoma, and head and neck carcinoma
WO2013150534A1 (en) * 2012-04-03 2013-10-10 Aposense Ltd. Novel targeting agents for diagnostic and therapeutic indications
JP2019156796A (en) * 2018-03-15 2019-09-19 国立大学法人大阪大学 Method for labeling radioisotope to boron cluster lipid and method for introducing labeled boron cluster lipid into virus particle

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6592847B1 (en) * 1998-05-14 2003-07-15 The General Hospital Corporation Intramolecularly-quenched near infrared flourescent probes
US7270799B2 (en) * 2004-01-15 2007-09-18 Nst Neurosurvival Technologies Ltd. Perturbed membrane-binding compounds and methods of using the same
AU2006251937A1 (en) 2005-05-27 2006-11-30 John Wayne Cancer Institute Use of free circulating DNA for diagnosis, prognosis, and treatment of cancer
DE102005061560A1 (en) 2005-12-22 2007-07-05 Siemens Ag Making radioactive isotopes for positron-emission tomography, employs accelerator designed to accelerate at least two different projectiles, especially protons and deuterons
WO2007122488A2 (en) 2006-04-20 2007-11-01 Hammersmith Imanet Limited Radiofluorinated compounds and their preparation
EP2029179A2 (en) 2006-06-21 2009-03-04 Hammersmith Imanet, Ltd Radiolabelling methods
WO2007148083A2 (en) 2006-06-21 2007-12-27 Ge Healthcare As Stabilisation of radiopharmaceutical precursors
JP5297379B2 (en) * 2006-09-19 2013-09-25 メタボロン、インコーポレイテッド Biomarker for prostate cancer and method of using the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2009109798A2 *

Also Published As

Publication number Publication date
WO2009109798A2 (en) 2009-09-11
WO2009109798A3 (en) 2010-10-28

Similar Documents

Publication Publication Date Title
Xu et al. 99mTc-labeling and evaluation of a HYNIC modified small-molecular inhibitor of prostate-specific membrane antigen
Shetty et al. Synthesis of novel 68Ga-labeled amino acid derivatives for positron emission tomography of cancer cells
KR101686264B1 (en) Labeled molecular imaging agents, methods of making and methods of use
CN115010629B (en) Prostate specific membrane antigen inhibitor, nuclide marker, preparation method and application
AU2011234320A1 (en) Lipophilic cationic probe for PET- imaging
CN112175025A (en) Glucose derivative containing benzene ring and application thereof
CN107522673B (en) 1,2,4, 5-tetrazine compound for bioorthogonal reaction and preparation method and application thereof
WO2009109798A2 (en) Precursors of lipid metabolism for the diagnosis and treatment of cancer
US20150297759A1 (en) Novel fluorine-18 labeled rhodamine derivatives for imaging with positron emission tomography
Mukai et al. Design of Ga–DOTA-based bifunctional radiopharmaceuticals: Two functional moieties can be conjugated to radiogallium–DOTA without reducing the complex stability
Kong et al. Synthesis of 99mTc-EC-AMT as an imaging probe for amino acid transporter systems in breast cancer
WO2023130587A1 (en) Water-soluble methylbenzene ether derivative, positron nuclide probe and nuclide marker, and preparation methods therefor and uses thereof
CN113583066B (en) Mannose derivative and application thereof
Mamat et al. Preparation of 4-halobenzoate-containing phosphane-based building blocks for labeling reactions using the traceless Staudinger ligation
CN113368264B (en) Radiolabeled cinnamycin, preparation method and application thereof
Chen et al. Synthesis and evaluation of technetium-99m-labeled pH (low) insertion peptide variant 7 for early diagnosis of MDA-MB-231 triple-negative breast cancer by targeting the tumor microenvironment
Kong et al. Development of Tyrosine‐Based Radiotracer 99mTc‐N4‐Tyrosine for Breast Cancer Imaging
CN106084004B (en) 18F click-labeled transferrin receptor targeting polypeptide T7 as well as preparation method and application thereof
Pulido et al. 4-N-Alkanoyl and 4-N-alkyl gemcitabine analogues with NOTA chelators for 68-gallium labelling
Wu et al. Synthesis and evaluation of an N-[18F] fluorodeoxyglycosyl amino acid for PET imaging of tumor metabolism
CN111407900A (en) Estrogen receptor targeted radioactive tracer, preparation method and application
TWI394587B (en) A radiolabeled nucleoside analogue, a method for preparing the same and the use thereof
US20130211066A1 (en) Compounds useful in imaging and therapy
CN109438265B (en) Compound with affinity with brown adipose tissue and preparation method and application thereof
CN103796685A (en) 3H- or 18F- labeled agents for imaging cystine/glutamate antiporter and oxidative stress

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20101006

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20141010

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

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

18D Application deemed to be withdrawn

Effective date: 20150915