WO2006058486A1 - Use of arginase in combination with 5fu and other compounds for treatment of human malignancies - Google Patents

Use of arginase in combination with 5fu and other compounds for treatment of human malignancies Download PDF

Info

Publication number
WO2006058486A1
WO2006058486A1 PCT/CN2005/002001 CN2005002001W WO2006058486A1 WO 2006058486 A1 WO2006058486 A1 WO 2006058486A1 CN 2005002001 W CN2005002001 W CN 2005002001W WO 2006058486 A1 WO2006058486 A1 WO 2006058486A1
Authority
WO
WIPO (PCT)
Prior art keywords
arginine
arginase
compound
agent
human
Prior art date
Application number
PCT/CN2005/002001
Other languages
French (fr)
Inventor
Ning Man Cheng
Yun Chung Leung
Wai Hung Lo
Original Assignee
Bio-Cancer Treatment International Limited
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 Bio-Cancer Treatment International Limited filed Critical Bio-Cancer Treatment International Limited
Publication of WO2006058486A1 publication Critical patent/WO2006058486A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/50Hydrolases (3) acting on carbon-nitrogen bonds, other than peptide bonds (3.5), e.g. asparaginase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/51Lyases (4)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention is related to pharmaceutical compositions for the treatment of human malignancies containing arginase.
  • Cancer remains one of the most difficult to treat human diseases.
  • certain forms of cancer such as liver cancer, there is no known effective drug.
  • Arginine degrading enzymes can be developed as drugs to treat cancer.
  • Arginase I (EC 3.5.3.1; L-arginine amidinohydrolase), is a key mammalian liver enzyme that catalyses the final step in urea formation in the Urea cycle, converting arginine into ornithine and urea.
  • PCT publication WO 2004/000349 discloses a pharmaceutical composition containing human recombinant arginase.
  • US patent application no. 10/757,843 discloses a different arginine degrading enzyme, arginine deiminase modified with polyethylene glycol, used for the treating of cancer, and the treating and/or inhibiting of metastasis.
  • 09/905,201 discloses a therapeutic composition and method for treatment of cancer comprising arginine decarboxylase of E. coli and modifications thereof. [0004] It is an object of the present invention to provide improved methods of treatment and compositions for the treatment of cancer.
  • one aspect of the present invention teaches a kit comprising at least one therapeutic dose of an arginine reducing compound and at least one therapeutic dose of an anti-neoplastic compound such as 5-fmorouracil (5FU), for the treatment of human malignancies.
  • the arginine reducing compound is an enzyme or a compound that is capable of degrading or removing arginine from the subject, so as to achieve a physiological arginine level of below 10 ⁇ M.
  • Some examples of arginine reducing compounds include, but are not limited to, arginine degrading enzymes such as arginase, human arginase I, arginine deiminase and arginine decarboxylase, modifications thereof or combinations thereof.
  • An antineoplastic compound may be, but is not limited to, an alkylating agent, antimetabolite agent, antimitotic agent, DNA production inhibiting agent or DNA repair inhibiting agent, etc.
  • a method for treating cancer in a human patient comprising reducing the physiological arginine levels in the patient to below 10 ⁇ M combined with a suitable anti-neoplastic or antimetabolite compound such as 5FU.
  • the reduction of physiological arginine levels in a human patient may be achieved through various treatments which include, but are not limited to, embolization, dialysis or administration of an arginine reducing compound.
  • a pharmaceutical composition comprising an arginine reducing compound and an anti-neoplastic compound.
  • an arginine reducing compound in combination with an anti-neoplastic compound for the manufacture of a medicament for the treatment of cancer.
  • the arginine reducing compound may be an arginine degrading enzyme.
  • Some examples of such arginine degrading enyzmes include, but are not limited to, arginase, arginine deiminase, arginine decarboxylase, or modifications and combinations thereof.
  • the arginase may also be human arginase I or pegylated human arginase.
  • the anti-neoplastic compound may, for example, be an alkylating agent, antimetabolic agent, antibiotic agent, DNA production inhibiting agent or DNA repair inhibiting agent, etc.
  • the anti-neoplastic compound may preferably be 5-fluorouracil.
  • Figure 1 shows plasmid map of pABlOl. This plasmid carries the gene encoding Arginase (arg) and only replicates in E. coli but not in B. subtilis.
  • Figures 2A, 2B and 2C show nucleotide sequence and its deduced amino acid sequence of the human Arginase I.
  • Figure 2A shows the nucleotide sequence (SEQ ID NO: 1) from EcoRVMunl to Xb al sites of plasmid pABlOl.
  • Nucleotide (nt) 1-6 EcoRllMunl site; nt 481-486, -35 region of promoter 1; nt 504-509, -10 region of promoter 1; nt 544-549, -35 region of promoter 2; nt 566-571, -10 region of promoter 2; nt 600-605, ribosome binding site; nt 614-616, start codon; nt 632- 637, Ndel site; nt 1601-1603, stop codon; nt 1997-2002, Xbal site.
  • Figure 2B shows the encoding nucleotide sequence (SEQ ID NO: 2) and its corresponding encoded amino acid sequence (SEQ ID NO: 3) of a modified human Arginase.
  • Nucleotide 614-1603 from Figure 2 A is an encoding region for the amino acid sequence of the modified Arginase.
  • the 6xHis (SEQ ID NO: 4) tag at the N-terminus is underlined. Translation stop codon is indicated by asterisk.
  • Figure 2C shows the encoding nucleotide sequence (SEQ ID NO: 8) and its corresponding encoded amino acid sequence (SEQ ID NO: 9) of the normal human Arginase I.
  • Figure 3 is a schematic drawing of the construction of a B. subtilis prophage allowing expression of Arginase.
  • Figure 4 shows the comparison of average tumor size for four groups of nude mice which have tumors induced by implantation with tumor cells.
  • the four groups are negative control, arginase (BCT) alone, arginase (BCT) and arginase (BCT) in combination with 5FU, an anti-neoplastic and antimetabolite compound.
  • Combined administration or “combined with administering” merely refers to a general period of time in which both arginase and an anti-neoplastic agent are administered to the human body for the treatment of human malignancies. It does not restrict the method of treatment to a simultaneous administration of the two types of compounds.
  • combined with merely refers to a general period of time in which the two steps of treatment for cancer are performed, this includes and is not limited to the possibility of simultaneous performance of the two steps.
  • the term “medicament” may refer to two different compounds applied at different times, as long as the two compounds belong to the same combination treatment.
  • pegylated Arginase refers to Arginase of present invention modified by pegylation to increase the stability of the enzyme and minimize immunoreactivity.
  • arginase I in both its modified and unmodified forms is the preferred arginase.
  • Pegylated arginase I may also be referred to as "BCT" and is used interchangeably in this application.
  • Human arginase I and other amino acid sequences as used herein include amino acid sequences that are substantially the same, meaning that they may have "slight and non-consequential sequence variations" from the actual sequences disclosed herein. Species with sequences that are substantially the same are considered to be equivalent to the disclosed sequences and as such are within the scope of the appended claims.
  • “slight and non-consequential sequence variations” means that the amino acid sequences are functionally equivalent to the sequences disclosed and/or claimed herein. Functionally equivalent sequences will function in substantially the same manner to produce substantially the same compositions as the amino acid compositions disclosed and claimed herein.
  • half-life refers to the time that would be required for the concentration of the Arginase in human plasma in vitro, to fall by half.
  • Arginase may be obtained from Ikemoto et al. (Ikemoto et al. Biochem J. 1990. 270: 697-703) or by the method disclosed in PCT publication WO 2004/000349. The arginase may also be produced using the methods described below.
  • CCAAACCATATGAGCGCCAAGTCCAGAACCATA-3' (SEQ ID NO: 5) and Arg2 (3'-CCAAACTCTAGAATCACATTTTTTGAATGACATGGACAC-S ⁇ (SEQ ID NO: 6), respectively, were purchased from Genset Singapore Biotechnology Pte Ltd. Both primers have the same melting temperature (Tm) of 72 degree C.
  • Primer Argl contains a Ndel restriction enzyme recognition site (underlined) and primer Arg2 contains a Xbal site (underlined). These two primers (final concentration 300 nM of each) were added to 5 ⁇ l of the human liver 5'- stretch plus cDNA library (Clontech, California, USA) in a 0.2-ml micro-tube.
  • DNA polymerase (2.6 units, 0.75 ⁇ l), the four deoxyribonucleotides (4 ⁇ l of each; final concentration 200 ⁇ M of each) and reaction buffer (5 ⁇ l) and dH 2 O (17.75 ⁇ l) were also added.
  • PCR was performed using the following conditions: pre-PCR (94 degree C, 5 min), 25 PCR cycles (94 degree C, 1 min; 57 degree C, 1 min; 72 degree C, 1 min), post-PCR (72 degree C, 7 min).
  • PCR product (5 ⁇ l) was analyzed on a 0.8% agarose gel and a single band of 1.4 kb was observed. This DNA fragment contains the gene encoding Arginase.
  • Plasmid pSGl 113 which is a derivative of plasmid pSG703 (Thornewell SJ et al. Gene. 1993. 133: 47-53), was isolated from the E. coli DH5 ⁇ clone carrying pSG1113 by using the Wizard Plus Minipreps DNA Purification System (Promega, Wisconsin, USA) following the manufacturer's instruction. This plasmid, which only replicates in E. coli but not in B. subtilis, was used as the vector for the sub-cloning of the Arginase gene.
  • the reaction mixture was subjected to agarose gel (0.8%) electrophoresis, and the 1.4 kb DNA fragment was recovered from the gel by using the Qiaex II Gel Extraction Kit (Qiagen, California, USA). Separately, the plasmid pSG1113 was treated with the same restriction endonucleases in the same way.
  • the reaction mixture was subjected to agarose gel (0.8%) electrophoresis, and a DNA fragment having a size of about 3.5 kb was recovered from the gel. This DNA fragment was joined by using T4 DNA ligase to the above 1.4 kb DNA fragment. The ligation mixture was used to transform E.
  • coli XLI-Blue using the conventional calcium method (Sambrook J et al. Molecular Cloning, A Laboratory Manual, second edition. Cold Spring Harbor Laboratory Press, New York. 1989) and plated on nutrient agar plate containing 100 ⁇ g/ml ampicillin. Colonies were screened for a plasmid with the appropriate insert by restriction analysis. The plasmid constructed was designated pABlOl (Fig. 1). ORI is the E. coli origin of replication and bla is the ampicillin resistant marker gene.
  • DNA sequencing was performed with primers Argl (SEQ ID NO: 5), Arg2 (SEQ ID NO: 6) and Arg6 (5'-CTCTGGCCATGCCAGGGTCCACCC-S') (SEQ ID NO: 7) to confirm the identity of the gene encoding Arginase (Fig. 2A, B, C).
  • subtilis lA304( ⁇ l05MU331) according to the known method (Anagnostopoulos C and Spizizen J. J Bacteriol. 1961. 81: 741-746).
  • the B. subtilis strain 1A304 ( ⁇ l05MU331) was obtained from J. Errington (Thornewell S et al. 1993. Gene. 133: 47-53). The strain was produced according to the publications by Thornewell, S. et al., 1993, Gene 133, 47-53 and by Baillie, L. W. J. et al., 1998, FEMS Microbiol. Letters 163, 43-47, which are incorporated herein in their entirety. Plasmid pABlOl (shown linearized in Fig.
  • the erythromycin resistance gene (ermC) was replaced by the Arginase gene (arg) by a double crossover event in a process of homologous recombination.
  • the 0.6 kb Munl-Ndel ⁇ lO5 phage DNA fragment (labeled as " ⁇ l05") and the cat gene provided the homologous sequences for the recombination.
  • the Arginase gene was targeted to the expression site in the prophage DNA of B. subtilis 1A304 ( ⁇ j>105MU331) and the Arginase gene was put under the control of the strong thermoinducible promoter (Leung YC and Errington J. Gene. 1995. 154: 1-6).
  • the fed-batch fermentation was carried out in a 2-liter fermentor at 37 degree C, pH 7.0 and dissolved oxygen 20% air saturation.
  • the feeding medium contained 200 g/L glucose, 2.5 g/L MgSO 4 «7H 2 O, 50 g/L tryptone, 7.5 g/L K 2 HPO 4 and 3.75 g/L KH 2 PO 4 .
  • the medium feeding rate was controlled with the pH-stat control strategy. In this strategy, the feeding rate was adjusted to compensate the pH increase caused by glucose depletion. This control strategy was first implemented when the glucose concentration decreased to a very low level at about 4.5-h fermentation time. If pH > 7.1, 4 mL of feeding medium was introduced into the fermentor.
  • a 5-ml HiTrap Chelating column (Pharmacia, New Jersey, USA) was equilibrated with 0.1 M NiCl 2 in dH 2 O, for 5 column volumes.
  • Fractions with arginase activity and high arginase purity were pooled (24 ml) and diluted ten times with start buffer [0.02 M sodium phosphate buffer (pH 7.4), 0.5 M NaCl]. This was loaded onto a second 5-ml HiTrap Chelating column (Pharmacia, New Jersey, USA), repeating the same procedure as above.
  • mPEG-SPA cat. no. 2M4MOH01, Nektar, USA
  • MW 5,000 5.82 g
  • the mixture was then dialyzed extensively by ultra-dialysis against 15 L of PBS buffer using the F50(S) capillary dialyser (Fresenius Medical Care, Bad Homburg, Germany) to remove all the unincorporated PEG.
  • the mPEG-SPA uses amino groups of lysines and the N- terminus of the protein as the site of modification.
  • the measured specific activity of the pegylated Arginase was as high as 592 1.U./mg.
  • EXAMPLE 5 1 A-LIFE DETERMINATION OF PEGYLATED ARGINASE IN VITRO USING THE METHOD IN HUMAN BLOOD PLASMA
  • Pegylated Arginase (305.6 ⁇ l) at a concentration of 1 mg/ml was added into human plasma (1 ml) and the final concentration of pegylated Arginase was 0.24 mg/ml. The mixture was divided into 20 aliquots in eppendorf tubes (65 ⁇ l mixture in each eppendorf tube) and then incubated at 37 0 C. A 1-2 ⁇ l portion of the mixture from each eppendorf tube was used to test the Arginase activity. In this example, the 1 A-MQ was determined to be approximately 3 days. It took about 3 days to reduce the relative activity from 100% to 50%.
  • arginase BCT was used in combination with 5FU.
  • Hep 3B cells obtained from the American Type Culture Collection (ATCC) were propagated through four passages according to the supplier's recommendations before 40 nude mice were implanted with a tumor of this cell line of at least 3mm 3 .
  • mice were divided randomly into four groups of 10 animals each. These are: Group 1, negative control (0.2 ml of 0.9% normal saline as negative control); Group 2, 250 IU of BCT; Group 3, combination of 250 IU arginase (BCT) and 10 mg/kg 5FU (5- fluorouracil) (Ebewe Arzneistoff Ges.m.b.H., Austria, Europe) ; and Group 4, 10 mg/kg 5FU.
  • the animals were treated by intraperitoneal injection of the compound(s) or normal saline once a week..
  • the implanted animals were observed once every two days for growth of the solid tumor in situ by digital caliper measurements to tumor size and weight.
  • the tumor size was the average of two perpendicular diameters and one diagonal diameter.
  • the tumor weight was taken to be the (length x width 2 )/2; assuming a specific gravity of 1.0 g/cm 3 .
  • tumour growth was measured for 71 days and is shown in Figure 4.
  • Statistical analysis was performed by SPSS 11.0 software (SPSS 5 Chicago, USA), based on Kaplan-Meier estimation and groups were compared by the log-rank test.
  • the blood samples of the patients may be taken daily throughout treatment for arginine levels, Arginase (BCT) activities, complete blood picture and full clotting profile. Renal and liver functions are taken at least every other day, or sooner if deemed necessary.
  • BCT Arginase
  • BCT On day 1 BCT is infused over 30 minutes at 2,000 IU per kg. Thereafter, BCT is infused weekly for 8-12 weeks. This may be continued if anti-tumour activity is observed. Twenty minutes before each BCT infusion, pre-medication with dipheneramine 10 mg iv. and hydrocortisone 100 mg iv. is given.
  • 5FU As for 5FU, it may administered at 125mg per meter square by short infusion every day from day 1 to day 5. Each 5FU infusion from day 1 to day 5 is preceded by administration of Folinic acid of 50mg per meter square. This same 5FU and Folinic acid treatment may be repeated every 4 weeks.
  • EXAMPLE 8 USING EMBOLIZATION AS A METHOD TO REDUCE PHYSIOLOGICAL ARGININE LEVELS IN COMBINATION WITH AN ANTI-NEOPLASTIC COMPOUND
  • Tumor embolization is a procedure used to reduce the vascularity of a tumor.
  • a particulate form such as microspheres, is administered via catheter, which is positioned in the tumor's arterial blood supply.
  • the particles are released from the catheter and carried by blood flow to the arterioles and capillary bed where they embolize and thus retards blood flow to the tumor (Microspheres and Regional Cancer Therapy, CRC Press, October 20, 1993).
  • embolization induces a leakage of hepatic arginase from the liver into the circulation and the hepatic arginase released into the systemic circulation, in such a way, rapidly depletes plasma arginine (Cheng PN, Leung YC, Lo WH, Tsui SM, Lam KC. Cancer Lett. 2005 Jun 16;224(l):67-80. Epub 2004 Dec 25.).
  • embolization effectively acts as a natural source of arginase that becomes released into the patient's blood stream after embolization i.e. it essentially acts as the administration of arginase to a patient, and is therefore expected to produce the same synergistic effect when administered in combination with an anti-neoplastic drug.
  • EXAMPLE 9 USING DIALYSIS AS A METHOD TO REDUCE PHYSIOLOGICAL ARGININE LEVELS IN COMBINATION WITH AN ANTI-NEOPLASTIC COMPOUND
  • Dialysis is a method used to remove waste materials and extra fluids from the blood. This method can also be used to remove certain amino acids from the blood (Gouyon JB, Desgres J, Mousson C. Pediatr Res. 1994 Mar; 35(3):357- 61). Therefore, dialysis can also be used to reduce the physiological levels of arginine in a human patient to below 10 ⁇ M, and is thus another implementation which may be used in combination with the administration of an anti-neoplastic drug.
  • arginine depletion is a new form of anti-neoplastic therapy that differs from the mode of operation of traditional anti-neoplastic drugs, such as, alkylating agents and mitotic inhibitors, etc.
  • Arginine deprivation results in the rapid and selective death of culture transformed and malignant cells (Scott L, Lamb J, Smith S, Wheatley DN. Br J Cancer. 2000 Sep;83(6):800-10). It has been suggested that loss of viability in the malignant phenotypes may be the result of the loss of control primarily at the key Gl checkpoint, which normally prevents cells from reinitiating DNA synthesis under adverse conditions.
  • nutrient depletion refers to the reduction of physiological arginine levels to below lO ⁇ M.
  • the reduction of physiological arginine levels may be achieved by different implementations.
  • one implementation of the present invention is the administration of arginine reducing compounds to the human patient.
  • arginine reducing compounds may be, but are not limited to, arginine degrading enzymes (such as arginase I, arginine decarboxylase, arginine deiminase, human, bovine and other animal arginase etc.).
  • Another implementation of the present invention for this purpose is the use of embolization or dialysis, or other methods of treatment to reduce arginine levels.
  • anti-neoplastic drugs are directed to damaging different parts of the cell e.g. the metabolic pathways or DNA synthesis/repair/transcription.
  • Families of anti-neoplastic drugs include, but are not limited to, alkylating agents such as chlorambucil, cyclophosphamide, thiotepa, and busulfan, mitotic inhibitors such as plant alkaloids (e.g. actinomycin D 5 mitomycin) and podophyllotoxins, and antibiotics (e.g. mitoxantrone, bleomycin), such as anthracyclines (e.g. doxorubicin). It is there part of the present invention that any one or combination of these drugs may be used as the cell-damaging agent referred to above.
  • 5FU is an example of an antimetabolite drug, and it is anticipated that other antimetabolite drugs, such as purine antagonists (e.g. 2- chlorodeoxyadenosine) and folate antagonists (e.g. methotrexate) would also provide synergistic effects.
  • 5FU is used in combination with administering arginase (BCT).
  • BCT arginase
  • 5FU is an analogue of uracil with a fluorine atom at the C-5 position in place of hydrogen. It rapidly enters the cell using the same facilitated transport mechanism as uracil.
  • 5FU is converted intracellularly to several active metabolites, which disrupt RNA synthesis and the action of the nucleotide synthetic enzyme thymidylate synthase (TS) (Leffingwell R, Rustum Y. Fluoropyrimidines in Cancer Therapy. Humana Press. January 1, 2003. p61-62).
  • TS thymidylate synthase
  • Example 6 the two compounds were administered at short time intervals, one immediately after another to facilitate experimental work.
  • the term "combinatorial” or “used in combination” refer to the administration of the two compounds, such as an arginine degrading enzyme (e.g. arginase or arginine deiminase) and an anti-neoplastic compound such as 5FU, either simultaneously or consecutively within a temporal interval wherein the compound first administered is still at a concentration to exert an effect with regards to the treatment target cells, tissues or organs.
  • the overlapping period of time in which the two compounds may be administered can be over a period of days or weeks, such as described in Example 7 above.
  • a compound such as 5FU may be administered topically.
  • the dosage of 5FU When used in human patients, the dosage of 5FU may be determined by a qualified health professional depending on the condition being treated, the size and overall health of the patient, as well as the particular regimen used. It is envisaged that when used in combination with an arginine degrading enzyme such as arginase (BCT) as taught by the present invention, the dosage of the anti-neoplastic compound such as 5FU can be significantly lower, thus also eliciting fewer and less severe side effects.
  • BCT arginase
  • Formulations of the pharmaceutical composition of the present invention can be used in the form of a solid, a solution, an emulsion, a dispersion, a micelle, a liposome, and the like, wherein the resulting formulation contains one or more of the modified arginine degrading enzyme such as human arginase in the practice of the present invention, as active ingredients, in a mixture with an organic or inorganic carrier or excipient suitable for enteral or parenteral applications.
  • the active ingredients may be the arginase, for example, with the usual non-toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, solutions, emulsions, suspensions, and any other form suitable for use in
  • I l manufacturing preparations in solid, semisolid, or liquid form.
  • auxiliary, stabilizing, thickening and coloring agents and perfumes may be used.
  • the active ingredients of one or more arginase are included in the pharmaceutical formulation in an amount sufficient to produce the desired effect upon the target process, condition or disease.
  • compositions containing the active ingredients contemplated herein may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Formulations intended for oral use may be prepared according to any method known in the art for the manufacture of pharmaceutical formulations.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract, thereby providing sustained action over a longer period. They may also be coated to form osmotic therapeutic tablets for controlled release.
  • formulations for oral use may be in the form of hard gelatin capsules wherein the active ingredients are mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate, kaolin, or the like. They may also be in the form of soft gelatin capsules wherein the active ingredients are mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil.
  • the pharmaceutical formulations may also be in the form of a sterile injectable solution or suspension. This suspension may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,4-butanediol.
  • Sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides, fatty acids (including oleic acid), naturally occurring vegetable oils like sesame oil, coconut oil, peanut oil, cottonseed oil, or synthetic fatty vehicles, like ethyl oleate, or the like. Buffers, dextrose solutions preservatives, antioxidants, and the like, can be incorporated or used as solute to dissolve the soluble enzyme as required.
  • the pharmaceutical formulations may also be an adjunct treatment together with other chemotherapeutic agents.
  • an arginase that has an amino acid sequence substantially the same as the sequence shown in SEQ ID No. 9 means that the sequence is at least 30% identical to that shown in SEQ ID No. 9 or that using the Arginase activity assay as described herein, there is no significant difference in the enzymatic activity between the enzyme of SEQ ID No. 9 and the one that is substantially similar.
  • the six histidines are provided for ease of purification, and the additional methionine group provided at the amino terminus thereof is to allow translation to be initiated.
  • SEQ ID NO. 1 (Fig. 2A) SEQ ID NO. 3: (Fig. 2B: amino acid sequence (SEQ ID NO:3)
  • SEQ ID NO. 5 5'-CCAAACCATATGAGCGCCAAGTCCAGAACCATA-S'

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides a method for treating cancer in a human patient by reducing the physiological arginine levels to below 10μM in combination with administering an anti-neoplastic compound. The present invention also provides a pharmaceutical composition comprising an arginine reducing compound, such as pegylated human arginase I, and an anti-neoplastic compound, such as 5-fluorouracil, for the treatment of malignancies in a human patient.

Description

USE OF ARGINASE IN COMBINATION WITH 5FU AND OTHER COMPOUNDS FOR TREATMENT OF HUMAN MALIGNANCIES
FIELD OF INVENTION
[0001] The present invention is related to pharmaceutical compositions for the treatment of human malignancies containing arginase.
BACKGROUND OF INVENTION
[0002] Cancer remains one of the most difficult to treat human diseases. For certain forms of cancer, such as liver cancer, there is no known effective drug.
[0003] Arginine degrading enzymes can be developed as drugs to treat cancer. Arginase I (EC 3.5.3.1; L-arginine amidinohydrolase), is a key mammalian liver enzyme that catalyses the final step in urea formation in the Urea cycle, converting arginine into ornithine and urea. PCT publication WO 2004/000349 discloses a pharmaceutical composition containing human recombinant arginase. US patent application no. 10/757,843 discloses a different arginine degrading enzyme, arginine deiminase modified with polyethylene glycol, used for the treating of cancer, and the treating and/or inhibiting of metastasis. Lastly, US patent application no. 09/905,201 discloses a therapeutic composition and method for treatment of cancer comprising arginine decarboxylase of E. coli and modifications thereof. [0004] It is an object of the present invention to provide improved methods of treatment and compositions for the treatment of cancer.
SUMMARY OF INVENTION
[0005] Accordingly, one aspect of the present invention teaches a kit comprising at least one therapeutic dose of an arginine reducing compound and at least one therapeutic dose of an anti-neoplastic compound such as 5-fmorouracil (5FU), for the treatment of human malignancies. The arginine reducing compound is an enzyme or a compound that is capable of degrading or removing arginine from the subject, so as to achieve a physiological arginine level of below 10 μM. Some examples of arginine reducing compounds include, but are not limited to, arginine degrading enzymes such as arginase, human arginase I, arginine deiminase and arginine decarboxylase, modifications thereof or combinations thereof. An antineoplastic compound may be, but is not limited to, an alkylating agent, antimetabolite agent, antimitotic agent, DNA production inhibiting agent or DNA repair inhibiting agent, etc.
[0006] According to another aspect of the present invention there is provided a method for treating cancer in a human patient, the method comprising reducing the physiological arginine levels in the patient to below 10 μM combined with a suitable anti-neoplastic or antimetabolite compound such as 5FU. The reduction of physiological arginine levels in a human patient may be achieved through various treatments which include, but are not limited to, embolization, dialysis or administration of an arginine reducing compound. [0007] According to a further aspect of the present invention there is provided a pharmaceutical composition comprising an arginine reducing compound and an anti-neoplastic compound.
[0008] According to yet another aspect of the present invention there is provided a use of an arginine reducing compound in combination with an anti-neoplastic compound for the manufacture of a medicament for the treatment of cancer. The arginine reducing compound may be an arginine degrading enzyme. Some examples of such arginine degrading enyzmes include, but are not limited to, arginase, arginine deiminase, arginine decarboxylase, or modifications and combinations thereof. The arginase may also be human arginase I or pegylated human arginase. The anti-neoplastic compound may, for example, be an alkylating agent, antimetabolic agent, antibiotic agent, DNA production inhibiting agent or DNA repair inhibiting agent, etc. The anti-neoplastic compound may preferably be 5-fluorouracil.
BRIEF DESCRIPTION OF FIGURES [0009] Figure 1 shows plasmid map of pABlOl. This plasmid carries the gene encoding Arginase (arg) and only replicates in E. coli but not in B. subtilis.
[0010] Figures 2A, 2B and 2C show nucleotide sequence and its deduced amino acid sequence of the human Arginase I. Figure 2A shows the nucleotide sequence (SEQ ID NO: 1) from EcoRVMunl to Xb al sites of plasmid pABlOl. Nucleotide (nt) 1-6, EcoRllMunl site; nt 481-486, -35 region of promoter 1; nt 504-509, -10 region of promoter 1; nt 544-549, -35 region of promoter 2; nt 566-571, -10 region of promoter 2; nt 600-605, ribosome binding site; nt 614-616, start codon; nt 632- 637, Ndel site; nt 1601-1603, stop codon; nt 1997-2002, Xbal site.
[0011] Figure 2B shows the encoding nucleotide sequence (SEQ ID NO: 2) and its corresponding encoded amino acid sequence (SEQ ID NO: 3) of a modified human Arginase. Nucleotide 614-1603 from Figure 2 A is an encoding region for the amino acid sequence of the modified Arginase. The 6xHis (SEQ ID NO: 4) tag at the N-terminus is underlined. Translation stop codon is indicated by asterisk.
[0012] Figure 2C shows the encoding nucleotide sequence (SEQ ID NO: 8) and its corresponding encoded amino acid sequence (SEQ ID NO: 9) of the normal human Arginase I.
[0013] Figure 3 is a schematic drawing of the construction of a B. subtilis prophage allowing expression of Arginase.
[0014] Figure 4 shows the comparison of average tumor size for four groups of nude mice which have tumors induced by implantation with tumor cells. The four groups are negative control, arginase (BCT) alone, arginase (BCT) and arginase (BCT) in combination with 5FU, an anti-neoplastic and antimetabolite compound. DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
[0015] As used herein and in the claims, "comprising" means including the elements that follow but not excluding others. [0016] "Combined administration" or "combined with administering" merely refers to a general period of time in which both arginase and an anti-neoplastic agent are administered to the human body for the treatment of human malignancies. It does not restrict the method of treatment to a simultaneous administration of the two types of compounds. When in reference to treatments that do not require the administration of a compound (such as dialysis or embolization), "combined with" merely refers to a general period of time in which the two steps of treatment for cancer are performed, this includes and is not limited to the possibility of simultaneous performance of the two steps.
[0017] Similarly, the term "medicament" may refer to two different compounds applied at different times, as long as the two compounds belong to the same combination treatment.
[0018] As used herein, the term "pegylated Arginase" refers to Arginase of present invention modified by pegylation to increase the stability of the enzyme and minimize immunoreactivity. In particular, arginase I, in both its modified and unmodified forms is the preferred arginase. Pegylated arginase I may also be referred to as "BCT" and is used interchangeably in this application.
[0019] Human arginase I and other amino acid sequences as used herein include amino acid sequences that are substantially the same, meaning that they may have "slight and non-consequential sequence variations" from the actual sequences disclosed herein. Species with sequences that are substantially the same are considered to be equivalent to the disclosed sequences and as such are within the scope of the appended claims. In this regard, "slight and non-consequential sequence variations" means that the amino acid sequences are functionally equivalent to the sequences disclosed and/or claimed herein. Functionally equivalent sequences will function in substantially the same manner to produce substantially the same compositions as the amino acid compositions disclosed and claimed herein.
[0020] As used herein, the term "half-life" ('/.-life) refers to the time that would be required for the concentration of the Arginase in human plasma in vitro, to fall by half. [0021] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to describe and disclose specific information for which the reference was cited in connection with.
[0022] Arginase may be obtained from Ikemoto et al. (Ikemoto et al. Biochem J. 1990. 270: 697-703) or by the method disclosed in PCT publication WO 2004/000349. The arginase may also be produced using the methods described below.
[0023] All references cited above and in the following description are incorporated by reference herein. The practice of the invention is exemplified in the following non-limiting examples. The scope of the invention is defined solely by the appended claims, which are in no way limited by the content or scope of the examples. Examples 8 and 9 described below are alternative ways of practicing the present invention.
EXAMPLE 1: CONSTRUCTION OF THE RECOMBINANT STRAIN LLClOl
(a) Isolation of the gene encoding human Arginase I
[0024] The gene sequence of human Arginase I was published in 1987 (Haraguchi Y. Proc Natl Acad Sci. 1987. 84: 412-415) and primers designed therefrom. Polymerase chain reaction (PCR) was performed to isolate the gene encoding a human Arginase using the Expand High Fidelity PCR System Kit (Roche, Indianapolis, USA). Primers Argl (5'-
CCAAACCATATGAGCGCCAAGTCCAGAACCATA-3') (SEQ ID NO: 5) and Arg2 (3'-CCAAACTCTAGAATCACATTTTTTGAATGACATGGACAC-S^ (SEQ ID NO: 6), respectively, were purchased from Genset Singapore Biotechnology Pte Ltd. Both primers have the same melting temperature (Tm) of 72 degree C. Primer Argl contains a Ndel restriction enzyme recognition site (underlined) and primer Arg2 contains a Xbal site (underlined). These two primers (final concentration 300 nM of each) were added to 5 μl of the human liver 5'- stretch plus cDNA library (Clontech, California, USA) in a 0.2-ml micro-tube. DNA polymerase (2.6 units, 0.75 μl), the four deoxyribonucleotides (4 μl of each; final concentration 200 μM of each) and reaction buffer (5 μl) and dH2O (17.75 μl) were also added. PCR was performed using the following conditions: pre-PCR (94 degree C, 5 min), 25 PCR cycles (94 degree C, 1 min; 57 degree C, 1 min; 72 degree C, 1 min), post-PCR (72 degree C, 7 min). PCR product (5 μl) was analyzed on a 0.8% agarose gel and a single band of 1.4 kb was observed. This DNA fragment contains the gene encoding Arginase.
(b) Isolation of plasmid pSG1113 [0025] Plasmid pSGl 113, which is a derivative of plasmid pSG703 (Thornewell SJ et al. Gene. 1993. 133: 47-53), was isolated from the E. coli DH5α clone carrying pSG1113 by using the Wizard Plus Minipreps DNA Purification System (Promega, Wisconsin, USA) following the manufacturer's instruction. This plasmid, which only replicates in E. coli but not in B. subtilis, was used as the vector for the sub-cloning of the Arginase gene.
(c) Sub-cloning the 1.4 kb PCR product into plasmid pSG1113 to form plasmid p ABlOl [0026] The PCR product, prepared using the above protocol, was treated with restriction endonucleases Ndel and Xbal (Promega, Wisconsin, USA) in a reaction medium composed of 6 mM Tris-HCl (pH 7.9), 6 mM MgCl2, 150 raM NaCl, 1 mM DTT at 37 degree C for 1.5 h. After completion of the treatment, the reaction mixture was subjected to agarose gel (0.8%) electrophoresis, and the 1.4 kb DNA fragment was recovered from the gel by using the Qiaex II Gel Extraction Kit (Qiagen, California, USA). Separately, the plasmid pSG1113 was treated with the same restriction endonucleases in the same way. After completion of the treatment, the reaction mixture was subjected to agarose gel (0.8%) electrophoresis, and a DNA fragment having a size of about 3.5 kb was recovered from the gel. This DNA fragment was joined by using T4 DNA ligase to the above 1.4 kb DNA fragment. The ligation mixture was used to transform E. coli XLI-Blue using the conventional calcium method (Sambrook J et al. Molecular Cloning, A Laboratory Manual, second edition. Cold Spring Harbor Laboratory Press, New York. 1989) and plated on nutrient agar plate containing 100 μg/ml ampicillin. Colonies were screened for a plasmid with the appropriate insert by restriction analysis. The plasmid constructed was designated pABlOl (Fig. 1). ORI is the E. coli origin of replication and bla is the ampicillin resistant marker gene. DNA sequencing was performed with primers Argl (SEQ ID NO: 5), Arg2 (SEQ ID NO: 6) and Arg6 (5'-CTCTGGCCATGCCAGGGTCCACCC-S') (SEQ ID NO: 7) to confirm the identity of the gene encoding Arginase (Fig. 2A, B, C).
(d) Construction of the novel recombinant B. subtilis prophage strain LLClOl [0027] The plasmid pABlOl was extracted and purified from the clone carrying the pABlOl by using the Wizard Plus Minipreps DNA Purification System (Promega, Wisconsin, USA). In the plasmid pABlOl (Fig. 1), the Arginase gene (arg) was flanked by the 0.6 kb Munl-Ndel <j>105 phage DNA fragment (labeled as "φlO5") and the cat gene (Fig. 1 and Fig. 3). This plasmid DNA (Ig) was used to transform competent B. subtilis lA304(φl05MU331) according to the known method (Anagnostopoulos C and Spizizen J. J Bacteriol. 1961. 81: 741-746). The B. subtilis strain 1A304 (φl05MU331) was obtained from J. Errington (Thornewell S et al. 1993. Gene. 133: 47-53). The strain was produced according to the publications by Thornewell, S. et al., 1993, Gene 133, 47-53 and by Baillie, L. W. J. et al., 1998, FEMS Microbiol. Letters 163, 43-47, which are incorporated herein in their entirety. Plasmid pABlOl (shown linearized in Fig. 3) was transformed into the B. subtilis strain 1A304 (φl05MU331) with selection for the CmR marker, and the transformants were screened for an Ers phenotype. Such transformants should have arisen from a double-crossover event, as shown in Fig. 3, placing transcription of the Arginase gene (arg) under the control of the strong phage promoter (Leung and Erington. Gene. 1995. 154: 1-6). The thick lines represent the prophage genome, broken lines the B. subtilis chromosome, and thin lines plasmid DNA. The genes are shown in Fig. 3 as shaded arrows pointing in the direction of transcription and translation. Regions of homology are bounded by broken vertical lines and homologous recombination events by 'X'. [0028] Fifty-two chloramphenicol resistant (CmR) colonies were obtained from plating 600 μl of the transformed cells on an agar plate containing chloramphenicol (5g/ml). Ten of these colonies were selected randomly and streaked onto an agar plate containing erythromycin (20 μg/ml) and one of these colonies did not grow, indicating that it was erythromycin sensitive (Ers). This chloramphenicol resistant but erythromycin sensitive colony was thus isolated and named as LLClOl. In the chromosome of this newly constructed prophage strain, the erythromycin resistance gene (ermC) was replaced by the Arginase gene (arg) by a double crossover event in a process of homologous recombination. The 0.6 kb Munl-Ndel φlO5 phage DNA fragment (labeled as "φl05") and the cat gene provided the homologous sequences for the recombination. In this way, the Arginase gene was targeted to the expression site in the prophage DNA of B. subtilis 1A304 (<j>105MU331) and the Arginase gene was put under the control of the strong thermoinducible promoter (Leung YC and Errington J. Gene. 1995. 154: 1-6).
EXAMPLE 2: FERMENTATION OF B. SUBTILIS LLClOl CELLS
[0029] The fed-batch fermentation was carried out in a 2-liter fermentor at 37 degree C, pH 7.0 and dissolved oxygen 20% air saturation. The feeding medium contained 200 g/L glucose, 2.5 g/L MgSO4«7H2O, 50 g/L tryptone, 7.5 g/L K2HPO4 and 3.75 g/L KH2PO4. The medium feeding rate was controlled with the pH-stat control strategy. In this strategy, the feeding rate was adjusted to compensate the pH increase caused by glucose depletion. This control strategy was first implemented when the glucose concentration decreased to a very low level at about 4.5-h fermentation time. If pH > 7.1, 4 mL of feeding medium was introduced into the fermentor. Immediately after the addition of glucose, the pH value would decrease below 7.1 rapidly. After approximate 10 min, when the glucose added was completely consumed by the bacterial cells, the pH value would increase to a value greater than 7.1, indicating that another 4 mL of feeding medium was due to be added into the fermentor. Heat shock was performed at 5-6 h when the culture density (OD6oonm) was between 12.0 and 13.0. During the heat shock, the temperature of the fermentor was increased from 37 degree C to 50 degree C and then cooled immediately to 37 degree C. The complete heating and cooling cycle took about 0.5 h. Cells were harvested for separation and purification of Arginase at 3 h and 6 h after heat shock. In this example, the aforementioned strain produced active human Arginase in an amount of at least about 162 mg per L of the fermentation medium at 6 h after heat shock.
EXAMPLE 3: PURIFICATION OF ARGINASE AT 6 H AFTER HEAT SHOCK AFTER FED-BATCH FERMENTATION AT LOW CELL DENSITY
[0030] Fed-batch fermentation was performed as described in Example 2. The cell culture (650 ml) collected at 6 h after heat shock at OD 12.8 was centrifuged at 5,000 rpm for 20 min at 4 degree C to pellet the cells. The wet weight of the cells was 24 g. The culture supernatant liquor was discarded and the cell pellet was stored at -80°C. The cells are stable at this temperature for a few days. To extract intracellular proteins, the cell pellet was resuspended in 140 ml solubilization buffer [50 niM Tris-HCl (pH 7.4), 0.1 M NaCl5 5 rnM MnSO4, lysozyme (75 μg/ml)]. After incubation at 30 degree C for 15 min, the mixture was sonicated for eight times, each time lasted for 10 s (the total time was 80 s), at 2 min intervals using the Soniprep 150 Apparatus (MSE, Osaka, Japan). About 500 units of deoxyribonuclease I (Catalog No. D 4527, Sigma, Missouri, USA) was added and the mixture was incubated at 37 degree C for 10 min to digest the chromosomal DNA. After centrifugation at 10,000 rpm for 20 min at 4 degree C, the supernatant, containing the crude protein extract, was assayed for the presence of the Arginase activity and analyzed by SDS-PAGE (Laemmli. Nature. 1970. 227: 680-685).
[0031] A 5-ml HiTrap Chelating column (Pharmacia, New Jersey, USA) was equilibrated with 0.1 M NiCl2 in dH2O, for 5 column volumes. The crude protein extract (140 ml) was loaded onto the column. Elution was performed with a linear gradient (0-100%) at a flow rate of 5 ml/min for 15 column volumes under the following conditions: Buffer A = start buffer [0.02 M sodium phosphate buffer (pH 7.4), 0.5 M NaCl]; Buffer B = start buffer containing 0.5 M imidazole. Fractions with arginase activity and high arginase purity (fractions 13 -24) were pooled (24 ml) and diluted ten times with start buffer [0.02 M sodium phosphate buffer (pH 7.4), 0.5 M NaCl]. This was loaded onto a second 5-ml HiTrap Chelating column (Pharmacia, New Jersey, USA), repeating the same procedure as above. Fractions 12-24 containing high protein levels as measured by protein gel were pooled as the arginase fractions and salt was removed using a 50-ml HiPrep 26/10 desalting column (Pharmacia, New Jersey, USA) with the following conditions: flow rate = 10 ml/min, buffer = 10 mM Tris-HCl (pH 7.4) and length of elution = 1.5 column volume. The protein concentration was measured by the method of Bradford (Bradford M. Anal Biochem. 1976. 72: 248-254).
[0032] In this example, a total of 85.73 mg of Arginase was purified from 650 ml cell culture. The yield of purified Arginase was estimated to be 132 mg/1 cell culture or 3.57 mg/g wet cell weight.
EXAMPLE 4: PREPARATION OF HIGHLY ACTIVE PEGYLATED ARGINASE
[0033] The purified Arginase as prepared using the methods described above was used for pegylation. Using the previous examples to illustrate the method, an arginase sample with specific activity = 518 I.U./mg was dissolved in PBS buffer before carrying out pegylation.
[0034] The mPEG-SPA (cat. no. 2M4MOH01, Nektar, USA) , MW 5,000 (5.82 g) was added into 555 ml of the purified Arginase (813.64 mg, 1.466 mg/ml) solution slowly in a 1 L beaker and then stirred for 2 h 40 min at room temperature (mole ratio of Arginase: mPEG-SPA = 1 : 50). The mixture was then dialyzed extensively by ultra-dialysis against 15 L of PBS buffer using the F50(S) capillary dialyser (Fresenius Medical Care, Bad Homburg, Germany) to remove all the unincorporated PEG. The mPEG-SPA uses amino groups of lysines and the N- terminus of the protein as the site of modification. In this example, the measured specific activity of the pegylated Arginase was as high as 592 1.U./mg. EXAMPLE 5: 1A-LIFE DETERMINATION OF PEGYLATED ARGINASE IN VITRO USING THE METHOD IN HUMAN BLOOD PLASMA
[0035] Purified Arginase (1 mg) was dissolved in 1 ml of 125 niM borate buffer solution (pH 8.3) on ice. Activated PEG (mPEG-SPA, MW 5,000) (7.14 mg) was added into the protein solution slowly at a mole ratio of Arginase: PEG = 1 : 50. The mixture was stirred on ice for 2.5 h.
[0036] Pegylated Arginase (305.6 μl) at a concentration of 1 mg/ml was added into human plasma (1 ml) and the final concentration of pegylated Arginase was 0.24 mg/ml. The mixture was divided into 20 aliquots in eppendorf tubes (65 μl mixture in each eppendorf tube) and then incubated at 370C. A 1-2 μl portion of the mixture from each eppendorf tube was used to test the Arginase activity. In this example, the 1A-MQ was determined to be approximately 3 days. It took about 3 days to reduce the relative activity from 100% to 50%.
EXAMPLE 6: THE RESPONSE OF TUMOR SIZE TO COMBINATORIAL ADMINISTRATION OF ARGINASE (BCT) AND 5FU IN MICE
[0037] In this example, arginase (BCT) was used in combination with 5FU. Hep 3B cells obtained from the American Type Culture Collection (ATCC) were propagated through four passages according to the supplier's recommendations before 40 nude mice were implanted with a tumor of this cell line of at least 3mm3.
[0038] When the tumors reached an average diameter of 5mm, the mice were divided randomly into four groups of 10 animals each. These are: Group 1, negative control (0.2 ml of 0.9% normal saline as negative control); Group 2, 250 IU of BCT; Group 3, combination of 250 IU arginase (BCT) and 10 mg/kg 5FU (5- fluorouracil) (Ebewe Arzneimittel Ges.m.b.H., Austria, Europe) ; and Group 4, 10 mg/kg 5FU. The animals were treated by intraperitoneal injection of the compound(s) or normal saline once a week..
[0039] The implanted animals were observed once every two days for growth of the solid tumor in situ by digital caliper measurements to tumor size and weight. The tumor size was the average of two perpendicular diameters and one diagonal diameter. The tumor weight was taken to be the (length x width2)/2; assuming a specific gravity of 1.0 g/cm3.
[0040] The tumour growth was measured for 71 days and is shown in Figure 4. Statistical analysis was performed by SPSS 11.0 software (SPSS5 Chicago, USA), based on Kaplan-Meier estimation and groups were compared by the log-rank test.
[0041] It may be seen from Figure 4 that the combinatorial administration of arginase (BCT) and 5FU yields unexpected synergistic results when compared to either compound on its own. Furthermore, the doses of arginase (BCT) used were lower than what is required to see efficacy when BCT is used alone. EXAMPLE 7: TREATMENT PROTOCOL OF PATIENTS USING EXOGENOUSLYADMINISTERED ARGINASE (BCT)
[0042] When BCT is administered into a patient, the blood samples of the patients may be taken daily throughout treatment for arginine levels, Arginase (BCT) activities, complete blood picture and full clotting profile. Renal and liver functions are taken at least every other day, or sooner if deemed necessary.
[0043] Vital signs (BP, Pulse, Respiratory rate, Oximeter reading) are taken every 15 minutes for 1 hour after commencement of Arginase infusion then hourly until stable. Thereafter, vital signs are taken at the discretion of the treating physician.
[0044] On day 1 BCT is infused over 30 minutes at 2,000 IU per kg. Thereafter, BCT is infused weekly for 8-12 weeks. This may be continued if anti-tumour activity is observed. Twenty minutes before each BCT infusion, pre-medication with dipheneramine 10 mg iv. and hydrocortisone 100 mg iv. is given. [0045] As for 5FU, it may administered at 125mg per meter square by short infusion every day from day 1 to day 5. Each 5FU infusion from day 1 to day 5 is preceded by administration of Folinic acid of 50mg per meter square. This same 5FU and Folinic acid treatment may be repeated every 4 weeks.
EXAMPLE 8: USING EMBOLIZATION AS A METHOD TO REDUCE PHYSIOLOGICAL ARGININE LEVELS IN COMBINATION WITH AN ANTI-NEOPLASTIC COMPOUND
[0046] Tumor embolization is a procedure used to reduce the vascularity of a tumor. A particulate form, such as microspheres, is administered via catheter, which is positioned in the tumor's arterial blood supply. The particles are released from the catheter and carried by blood flow to the arterioles and capillary bed where they embolize and thus retards blood flow to the tumor (Microspheres and Regional Cancer Therapy, CRC Press, October 20, 1993). It is known that embolization induces a leakage of hepatic arginase from the liver into the circulation and the hepatic arginase released into the systemic circulation, in such a way, rapidly depletes plasma arginine (Cheng PN, Leung YC, Lo WH, Tsui SM, Lam KC. Cancer Lett. 2005 Jun 16;224(l):67-80. Epub 2004 Dec 25.).
[0047] Thus, embolization effectively acts as a natural source of arginase that becomes released into the patient's blood stream after embolization i.e. it essentially acts as the administration of arginase to a patient, and is therefore expected to produce the same synergistic effect when administered in combination with an anti-neoplastic drug.
EXAMPLE 9: USING DIALYSIS AS A METHOD TO REDUCE PHYSIOLOGICAL ARGININE LEVELS IN COMBINATION WITH AN ANTI-NEOPLASTIC COMPOUND
[0048] Dialysis is a method used to remove waste materials and extra fluids from the blood. This method can also be used to remove certain amino acids from the blood (Gouyon JB, Desgres J, Mousson C. Pediatr Res. 1994 Mar; 35(3):357- 61). Therefore, dialysis can also be used to reduce the physiological levels of arginine in a human patient to below 10μM, and is thus another implementation which may be used in combination with the administration of an anti-neoplastic drug.
[0049] While the present invention has been described using the aforementioned examples, it is clear that other combinations of drugs, may also have the same synergistic effect. An important aspect of the present invention is the recognition that arginine depletion is a new form of anti-neoplastic therapy that differs from the mode of operation of traditional anti-neoplastic drugs, such as, alkylating agents and mitotic inhibitors, etc.
[0050] Arginine deprivation results in the rapid and selective death of culture transformed and malignant cells (Scott L, Lamb J, Smith S, Wheatley DN. Br J Cancer. 2000 Sep;83(6):800-10). It has been suggested that loss of viability in the malignant phenotypes may be the result of the loss of control primarily at the key Gl checkpoint, which normally prevents cells from reinitiating DNA synthesis under adverse conditions.
[0051] Therefore, whereas normal cells move into a quiescent state (G0) during arginine deprivation, malignant cells which lack Gl checkpoint efficacy continue uncontrolled cell cycle advancement. The inventors of the present invention were able to recognize that uncontrolled cell cycle advancement of malignant cells under nutrient depletion in combination with cellular damage caused by an antineoplastic drug creates a significant synergistic cell killing.
[0052] In this description, nutrient depletion refers to the reduction of physiological arginine levels to below lOμM. The reduction of physiological arginine levels may be achieved by different implementations. For this purpose, one implementation of the present invention is the administration of arginine reducing compounds to the human patient. Such arginine reducing compounds may be, but are not limited to, arginine degrading enzymes (such as arginase I, arginine decarboxylase, arginine deiminase, human, bovine and other animal arginase etc.).
Another implementation of the present invention for this purpose is the use of embolization or dialysis, or other methods of treatment to reduce arginine levels.
[0053] Conventional anti-neoplastic drugs are directed to damaging different parts of the cell e.g. the metabolic pathways or DNA synthesis/repair/transcription. Families of anti-neoplastic drugs include, but are not limited to, alkylating agents such as chlorambucil, cyclophosphamide, thiotepa, and busulfan, mitotic inhibitors such as plant alkaloids (e.g. actinomycin D5 mitomycin) and podophyllotoxins, and antibiotics (e.g. mitoxantrone, bleomycin), such as anthracyclines (e.g. doxorubicin). It is there part of the present invention that any one or combination of these drugs may be used as the cell-damaging agent referred to above.
[0054] In this description, 5FU is an example of an antimetabolite drug, and it is anticipated that other antimetabolite drugs, such as purine antagonists (e.g. 2- chlorodeoxyadenosine) and folate antagonists (e.g. methotrexate) would also provide synergistic effects. [0055] In the most preferred embodiment of the present invention, 5FU is used in combination with administering arginase (BCT). 5FU is an analogue of uracil with a fluorine atom at the C-5 position in place of hydrogen. It rapidly enters the cell using the same facilitated transport mechanism as uracil. 5FU is converted intracellularly to several active metabolites, which disrupt RNA synthesis and the action of the nucleotide synthetic enzyme thymidylate synthase (TS) (Leffingwell R, Rustum Y. Fluoropyrimidines in Cancer Therapy. Humana Press. January 1, 2003. p61-62).
[0056] In Example 6 described above, the two compounds were administered at short time intervals, one immediately after another to facilitate experimental work. As used in this present invention, the term "combinatorial" or "used in combination" refer to the administration of the two compounds, such as an arginine degrading enzyme (e.g. arginase or arginine deiminase) and an anti-neoplastic compound such as 5FU, either simultaneously or consecutively within a temporal interval wherein the compound first administered is still at a concentration to exert an effect with regards to the treatment target cells, tissues or organs. The overlapping period of time in which the two compounds may be administered can be over a period of days or weeks, such as described in Example 7 above.
[0057] It is also not necessary that the routes of administration for the two compounds be the same. A compound such as 5FU may be administered topically.
When used in human patients, the dosage of 5FU may be determined by a qualified health professional depending on the condition being treated, the size and overall health of the patient, as well as the particular regimen used. It is envisaged that when used in combination with an arginine degrading enzyme such as arginase (BCT) as taught by the present invention, the dosage of the anti-neoplastic compound such as 5FU can be significantly lower, thus also eliciting fewer and less severe side effects.
[0058] It must be noted that as used herein and in the appended claims, the singular forms "a," "and," and "the" include plural references unless the context clearly dictates otherwise. Thus, for example, reference to "a pharmaceutical preparation" includes mixtures of different preparations and reference to "the method of treatment" includes reference to equivalent steps and methods known to those skilled in the art, and so forth.
[0059] The invention having been fully described, modifications within its scope will be apparent to those of ordinary skill in the art. All such modifications are within the scope of the invention.
[0060] Formulations of the pharmaceutical composition of the present invention can be used in the form of a solid, a solution, an emulsion, a dispersion, a micelle, a liposome, and the like, wherein the resulting formulation contains one or more of the modified arginine degrading enzyme such as human arginase in the practice of the present invention, as active ingredients, in a mixture with an organic or inorganic carrier or excipient suitable for enteral or parenteral applications. The active ingredients may be the arginase, for example, with the usual non-toxic, pharmaceutically acceptable carriers for tablets, pellets, capsules, suppositories, solutions, emulsions, suspensions, and any other form suitable for use in
I l manufacturing preparations, in solid, semisolid, or liquid form. In addition auxiliary, stabilizing, thickening and coloring agents and perfumes may be used. The active ingredients of one or more arginase are included in the pharmaceutical formulation in an amount sufficient to produce the desired effect upon the target process, condition or disease.
[0061] Pharmaceutical formulations containing the active ingredients contemplated herein may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Formulations intended for oral use may be prepared according to any method known in the art for the manufacture of pharmaceutical formulations. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract, thereby providing sustained action over a longer period. They may also be coated to form osmotic therapeutic tablets for controlled release. [0062] Li some cases, formulations for oral use may be in the form of hard gelatin capsules wherein the active ingredients are mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate, kaolin, or the like. They may also be in the form of soft gelatin capsules wherein the active ingredients are mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. [0063] The pharmaceutical formulations may also be in the form of a sterile injectable solution or suspension. This suspension may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,4-butanediol. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides, fatty acids (including oleic acid), naturally occurring vegetable oils like sesame oil, coconut oil, peanut oil, cottonseed oil, or synthetic fatty vehicles, like ethyl oleate, or the like. Buffers, dextrose solutions preservatives, antioxidants, and the like, can be incorporated or used as solute to dissolve the soluble enzyme as required.
[0064] The pharmaceutical formulations may also be an adjunct treatment together with other chemotherapeutic agents.
[0065] In the claims, an arginase that has an amino acid sequence substantially the same as the sequence shown in SEQ ID No. 9 (amino acid sequence of normal human arginase I) means that the sequence is at least 30% identical to that shown in SEQ ID No. 9 or that using the Arginase activity assay as described herein, there is no significant difference in the enzymatic activity between the enzyme of SEQ ID No. 9 and the one that is substantially similar. The six histidines are provided for ease of purification, and the additional methionine group provided at the amino terminus thereof is to allow translation to be initiated. It is clear to one skilled in the art that other forms of purification may also be used, and therefore a "substantially similar" arginase does not need to have any homology with the MHHHHHH sequence of SEQ ID No. 3. In some bacterial strains there may be at least 40% homology with SEQ. SEQ ID No. 9. Some mammalian arginase may be 70% homology with SEQ ID No. 9.
[0066] SEQUENCE LISTING
SEQ ID NO. 1: (Fig. 2A) SEQ ID NO. 3: (Fig. 2B: amino acid sequence (SEQ ID NO:3)
SEQ ID NO. 5: 5'-CCAAACCATATGAGCGCCAAGTCCAGAACCATA-S'
(Arginase I) SEQ ID NO. 6: 5'-CCAAACTCTAGAATCACATTTTTTGAATGACATGGACAC-S'
(Arginase II) SEQ ID NO. 7: 5'-CTCTGGCCATGCCAGGGTCCACCC-S' (Arg 6)
SEQ ID NO. 8 & 9: (Figure 2C: nucleotide sequence (SEQ ID NO. 8); and amino acid sequence (SEQ ID NO. 9))

Claims

1. A method of treating cancer in a human patient, the method comprising reducing the physiological arginine levels in said patient to below lOμM combined with administering an anti-neoplastic compound.
2. The method according to claim 1 wherein said method of reducing the physiological arginine levels in said patient comprises embolization, dialysis, or administering an arginine reducing compound.
3. The method according to claim 2 wherein said arginine reducing compound is an arginine degrading enzyme.
4. The method according to claim 3 wherein said arginine degrading enzyme is arginase, arginine deiminase, arginine decarboxylase, modifications thereof or combinations thereof.
5. The method according to claim 4 wherein said arginase is pegylated human arginase I.
6. The method according to claim 1 wherein said anti-neoplastic compound is an alkylating agent, antimetabolite agent, antimitotic agent, DNA production inhibiting agent or DNA repair inhibiting agent.
7. The method according to claim 1 wherein said anti-neoplastic compound is 5-fluorouracil.
8. The method according to claim 1 wherein said arginine reducing step is achieved by administration of pegylated human arginase I and 5- fluorouracil to a patient.
9. A kit for the treatment of human malignancies comprising at least one therapeutic dose of an arginine reducing compound and at least one therapeutic dose of an anti-neoplastic compound.
10. The kit according to claim 9 wherein said arginine reducing compound is an arginine degrading enzyme.
11. The kit according to claim 10 wherein said arginine degrading enzyme is arginase, arginine deiminase, arginine decarboxylase, modifications thereof or combinations thereof.
12. The kit according to claim 11 wherein said arginase is human arginase I.
13. The kit according to claim 11 wherein said arginase is pegylated human arginase.
14. The kit according to claim 9 wherein said anti-neoplastic compound is an alkylating agent, antimetabolite agent, antibiotic agent, DNA production inhibiting agent or DNA repair inhibiting agent.
15. The kit according to claim 9 wherein said anti-neoplastic compound is 5- fluorouracil.
16. A pharmaceutical composition comprising an arginine reducing compound and an anti-neoplastic compound.
17. The pharmaceutical composition according to claim 16 wherein said arginine reducing compound is an arginine degrading enzyme.
18. The pharmaceutical composition according to claim 17 wherein said arginine degrading enzyme is arginase, arginine deiminase, arginine decarboxylase, or modifications and combinations thereof.
19. The pharmaceutical composition according to claim 18 wherein said arginase is human arginase I.
20. The pharmaceutical composition according to claim 18 wherein said arginase is pegylated human arginase.
21. The pharmaceutical composition according to claim 16 wherein said antineoplastic compound is an alkylating agent, antimetabolite agent, antibiotic agent, DNA production inhibiting agent or DNA repair inhibiting agent.
22. The pharmaceutical composition according to claim 16 wherein said antineoplastic compound is 5-fluorouracil.
23. The pharmaceutical composition according to claim 16 wherein said pharmaceutical composition is a therapeutic dose for treating cancer.
24. Use of an arginine reducing compound in combination with an antineoplastic compound for the manufacture of a medicament for the treatment of cancer.
25. The use according to claim 24, wherein said arginine reducing compound is an arginine degrading enzyme.
26. The use according to claim 25, wherein said arginine degrading enzyme is arginase, arginine deiminase, arginine decarboxylase, or modifications and combinations thereof wherein said arginine reducing compound is an arginine degrading enzyme.
27. The use according to claim 26, wherein said arginase is human arginase I.
28. The use according to claim 26, wherein said arginase is pegylated human arginase.
29. The use according to claim 24, wherein said anti-neoplastic compound is an alkylating agent, antimetabolite agent, antibiotic agent, DNA production inhibiting agent or DNA repair inhibiting agent.
30. The use according to claim 24, wherein said anti-neoplastic compound is 5- fluorouracil.
PCT/CN2005/002001 2004-12-03 2005-11-25 Use of arginase in combination with 5fu and other compounds for treatment of human malignancies WO2006058486A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US63339804P 2004-12-03 2004-12-03
US60/633,398 2004-12-03

Publications (1)

Publication Number Publication Date
WO2006058486A1 true WO2006058486A1 (en) 2006-06-08

Family

ID=36564754

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2005/002001 WO2006058486A1 (en) 2004-12-03 2005-11-25 Use of arginase in combination with 5fu and other compounds for treatment of human malignancies

Country Status (1)

Country Link
WO (1) WO2006058486A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2102231A2 (en) * 2006-12-12 2009-09-23 Bio-Cancer Treatment International Limited Improved expression system for recombinant human arginase i
WO2010124547A1 (en) * 2009-03-26 2010-11-04 The Hong Kong Polytechnic University Site-directed pegylation of arginases and the use thereof as anti-cancer and anti-viral agents
WO2014001956A3 (en) * 2012-06-25 2014-02-20 Ning Man Cheng Combinational use of pegylated recombinant human arginase with chemotherapeutic/target therapeutic drug in cancer treatment
EP2654776A4 (en) * 2010-12-21 2016-05-11 Bio Cancer Treatment Int Ltd Use of pegylated recombinant human arginase for treatment of leukemia

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003063780A2 (en) * 2002-01-25 2003-08-07 Cancer Treatments International Therapeutic composition for treatment of cancer by arginine depletion
WO2003078578A2 (en) * 2002-03-12 2003-09-25 Lsu Medical Center Modulation of the immune response through the manipulation of arginine levels
WO2004000349A1 (en) * 2002-06-20 2003-12-31 Bio-Cancer Treatment International Limited Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation
CA2431080A1 (en) * 2003-06-02 2004-12-02 Catherine Adele O'brien Enhancement of anticancer immunity through inhibition of arginase
WO2005068633A1 (en) * 2004-01-20 2005-07-28 Korea Research Institute Of Bioscience And Biotechnology Differentiation regulating agent containing gene which regulating differentiation from stem cells into natural killer cells as effective ingredient

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003063780A2 (en) * 2002-01-25 2003-08-07 Cancer Treatments International Therapeutic composition for treatment of cancer by arginine depletion
WO2003078578A2 (en) * 2002-03-12 2003-09-25 Lsu Medical Center Modulation of the immune response through the manipulation of arginine levels
WO2004000349A1 (en) * 2002-06-20 2003-12-31 Bio-Cancer Treatment International Limited Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation
CA2431080A1 (en) * 2003-06-02 2004-12-02 Catherine Adele O'brien Enhancement of anticancer immunity through inhibition of arginase
WO2005068633A1 (en) * 2004-01-20 2005-07-28 Korea Research Institute Of Bioscience And Biotechnology Differentiation regulating agent containing gene which regulating differentiation from stem cells into natural killer cells as effective ingredient

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2102231A2 (en) * 2006-12-12 2009-09-23 Bio-Cancer Treatment International Limited Improved expression system for recombinant human arginase i
EP2102231A4 (en) * 2006-12-12 2010-03-31 Bio Cancer Treatment Int Ltd Improved expression system for recombinant human arginase i
WO2010124547A1 (en) * 2009-03-26 2010-11-04 The Hong Kong Polytechnic University Site-directed pegylation of arginases and the use thereof as anti-cancer and anti-viral agents
US8507245B2 (en) 2009-03-26 2013-08-13 The Hong Kong Polytechnic University Site-directed pegylation of arginases and the use thereof as anti-cancer and anti-viral agents
EP2654776A4 (en) * 2010-12-21 2016-05-11 Bio Cancer Treatment Int Ltd Use of pegylated recombinant human arginase for treatment of leukemia
WO2014001956A3 (en) * 2012-06-25 2014-02-20 Ning Man Cheng Combinational use of pegylated recombinant human arginase with chemotherapeutic/target therapeutic drug in cancer treatment

Similar Documents

Publication Publication Date Title
US8679810B2 (en) Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation
JP6993970B2 (en) Bacteria engineered to reduce hyperphenylalanineemia
ES2266515T3 (en) LACTOCOCCUS strain with self-containment.
US10238695B2 (en) Use of microorganisms for reducing the level of trimethylamine in a human body cavity, in particular for the treatment of trimethylaminuria or of bacterial vaginosis and the prevention of cardiovascular diseases
CZ2003848A3 (en) Treatment method of disorders connected with bcl-2 by making use of antisense bcl-2 oligomers
JP2018535678A5 (en)
WO2006058486A1 (en) Use of arginase in combination with 5fu and other compounds for treatment of human malignancies
EP2468861B1 (en) Method of constructing gene transport support
AU2003250371B2 (en) Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation
EP0483113B1 (en) A method of producing an enzymatically active polypeptide analog of human Cu/Zn SOD
US10183062B2 (en) Treatment for cancer
US20080292609A1 (en) Use of Arginase in Combination with 5FU and Other Compounds for Treatment of Human Malignancies
JP2002501032A (en) Acute intermittent porphyria (AIP) and other methods of treating porphyria
TWI331531B (en) Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation
KR20050024361A (en) Pharmaceutical preparation and method of treatment of human malignancies with arginine deprivation
JP2001512683A (en) High expression module containing two or more tandem copies of the sequence encoding methioninase
Wellner et al. L-Threonine Deaminase as a Possible Antitumor Agent¹
KR20240054373A (en) Salmonella engineered for avirulent colonization of tumors
AU2022210461A1 (en) Cd27-extracellular domain car to target cd70-positive tumors
CN115916959A (en) Genetically engineered live bacteria and construction method thereof
US20040014191A1 (en) Nitroreductase enzymes
Dachs The effect of metronidazole on Bacteroides fragilis and Escherichia coli
WO1999025386A1 (en) delta-O6-METHYLGUANINE-DNA METHYLTRANSFERASE GENE TRANSFER FOR O6-BENZYLGUANINE AND (N,N&#39;-BIS(2-CHLOROETHYL)-N-NITROSOUREA) RESISTANCE

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KN KP KR KZ LC LK LR LS LT LU LV LY MA MD MG MK MN MW MX MZ NA NG NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 69(1) EPC (EPO F. 1205A DATED 04.09.2007)

122 Ep: pct application non-entry in european phase

Ref document number: 05813861

Country of ref document: EP

Kind code of ref document: A1

WWW Wipo information: withdrawn in national office

Ref document number: 5813861

Country of ref document: EP