<<

(19) TZZ _¥_T

(11) EP 2 516 637 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Date of publication and mention (51) Int Cl.: of the grant of the patent: C12N 9/10 (2006.01) C12P 19/38 (2006.01) 20.04.2016 Bulletin 2016/16 (86) International application number: (21) Application number: 10798096.3 PCT/EP2010/070581

(22) Date of filing: 22.12.2010 (87) International publication number: WO 2011/076894 (30.06.2011 Gazette 2011/26)

(54) THERMOSTABLE BIOCATALYST COMBINATION FOR NUCLEOSIDE SYNTHESIS Wärmebeständige Biokatalysatorkombination zur Nukleotidsynthese Combinaison de biocatalyseur thermostable pour synthèse de nucléoside

(84) Designated Contracting States: • TARAN S A ET AL: "Enzymatic AL AT BE BG CH CY CZ DE DK EE ES FI FR GB transglycosylation of natural and modified GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO nucleosides by immobilized thermostable PL PT RO RS SE SI SK SM TR nucleoside phosphorylases from Geobacillus stearothermophilus", RUSSIAN JOURNAL OF (30) Priority: 22.12.2009 EP 09382296 BIOORGANIC CHEMISTRY, vol. 35, no. 6, November 2009 (2009-11), pages 739-745, (43) Date of publication of application: XP002585194, 31.10.2012 Bulletin 2012/44 • TRELLES J A ET AL: "Screening of Catalytically Active Microorganisms for the Synthesis of (73) Proprietor: Plasmia Biotech, S.L. 6-Modified Purine Nucleosides", 08100 Mollet del Vallès (ES) BIOTECHNOLOGY LETTERS, KLUWER ACADEMIC PUBLISHERS, DO LNKD- (72) Inventors: DOI:10.1007/S10529-005-5628-7, vol. 27, no. 11, 1 • MONTILLA AREVALO, Rafael June 2005 (2005-06-01), pages 759-763, E-08100 Mollet del Vallès (ES) XP019230857, ISSN: 1573-6776 • DERONCELÉ THOMAS, Víctor, Manuel • DATABASE UniProt [Online] 1 November 1999 E-08100 Mollet del Vallès (ES) (1999-11-01), "SubName: Full=Uridine • LÓPEZ GÓMEZ, Cristina phosphorylase; EC=2.4.2. E-08100 Mollet del Vallès (ES) 3;", XP002585195, retrieved from EBI • ESTÉVEZ COMPANY, Carlos accession no. UNIPROT:Q9YA34 Database E-08100 Mollet del Vallès (ES) accession no. Q9YA34 & KAWARABAYASHI • CASTELLS BOLIART, Josep YUTAKA ET AL: "Complete genome sequence of E-08100 Mollet del Vallès (ES) an aerobic hyper-thermophilic crenarchaeon, aeropyrum pernix K1", DNA RESEARCH, (74) Representative: Oficina Ponti, SLP UNIVERSAL ACADEMY PRESS, JP LNKD- C. Consell de Cent, 322 DOI:10.1093/DNARES/6.2.83, vol. 6, no. 2, 1 08007 Barcelona (ES) January 1999 (1999-01-01) , pages 83-101, XP002957164, ISSN: 1340-2838 (56) References cited: WO-A1-95/16785 WO-A2-03/035012 US-A1- 2005 142 645

Note: Within nine months of the publication of the mention of the grant of the European patent in the European Patent Bulletin, any person may give notice to the European Patent Office of opposition to that patent, in accordance with the Implementing Regulations. Notice of opposition shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention). EP 2 516 637 B1

Printed by Jouve, 75001 PARIS (FR) (Cont. next page) EP 2 516 637 B1

• HAMAMOTO TOMOKI ET AL: "Cloning of purine • CACCIAPUOTI GIOVANNA ET AL: "Purification nucleoside phosphorylase II gene from Bacillus and characterization of extremely thermophilic stearothermophilus TH 6-2 and characterization and thermostable 5’-methylthioadenosine of its gene product", BIOSCIENCE phosphorylase from the archaeon Sulfolobus BIOTECHNOLOGY AND BIOCHEMISTRY, vol. 61, solfataricus: Purine nucleoside phosphorylase no. 2, 1997, pages 276-280, XP002097506, ISSN: activity and evidence for intersubunit disulfide 0916-8451 cited in the application bonds", JOURNAL OF BIOLOGICAL • HAMAMOTO TOMOKI ET AL: "Cloning and CHEMISTRY, vol. 269, no. 40, 1994, pages expression of purine nucleoside phosphorylase 24762-24769, XP002625003, ISSN: 0021-9258 I gene from Bacillus stearothermophilus TH 6-2", • CACCIAPUOTI GIOVANNA ET AL: "Extremely BIOSCIENCE BIOTECHNOLOGY AND thermophilic and thermostable BIOCHEMISTRY, vol. 61, no. 2, 1997, pages 5’-methylthioadenosine phosphorylase from the 272-275, XP002097508, ISSN: 0916-8451 cited in archaeon Sulfolobus solfataricus: Gene cloning the application and amino acid sequence determination", • CACCIAPUOTI GIOVANNA ET AL: "Purine EUROPEAN JOURNAL OF BIOCHEMISTRY, vol. nucleoside phosphorylases from 239, no. 3, 1996, pages 632-637, XP002625004, hyperthermophilic require a CXC motif ISSN: 0014-2956 for stability and folding.", THE FEBS JOURNAL • FRANCIS E JENNEY JR ET AL: "The impact of OCT2009 LNKD- PUBMED:19740110, vol. 276, no. extremophiles on structural genomics (and vice 20, October 2009 (2009-10), pages 5799-5805, versa)", EXTREMOPHILES ; LIFE UNDER XP002585196, ISSN: 1742-4658 EXTREME CONDITIONS, SPRINGER-VERLAG, • CACCIAPUOTI GIOVANNA ET AL: "A novel TO, vol. 12, no. 1, 13 June 2007 (2007-06-13), hyperthermostable 5 ’-deoxy-5 pages 39-50, XP019593097, ISSN: 1433-4909 ’-methylthioadenosine phosphorylase from the • CACCIAPUOTI G ET AL: "Heterologous archaeon Sulfolobus solfataricus", FEBS Expression of 5’-Methylthioadenosine JOURNAL, vol. 272, no. 8, April 2005 (2005-04), Phosphorylase from the ArchaeonSulfolobus pages 1886-1899, XP002585197, cited in the solfataricus:Characterization of the application Recombinant Protein and Involvement of • CACCIAPUOTI GIOVANNA ET AL: "Biochemical Disulfide Bonds in Thermophilicity and and structural characterization of Thermostability", PROTEIN EXPRESSION AND mammalian-like purine nucleoside PURIFICATION, ACADEMIC PRESS, SAN DIEGO, phosphorylase from the Archaeon Pyrococcus CA, vol. 16, no. 1, 1 June 1999 (1999-06-01), pages furiosus", FEBS JOURNAL, vol. 274, no. 10, May 125-135, XP004441691, ISSN: 1046-5928, DOI: 2007 (2007-05), pages 2482-2495, XP002585198, 10.1006/PREP.1999.1076 cited in the application • DATABASE UniProt [Online] 1 October 2001 (2001-10-01), "SubName: Full=Purine nucleoside phosporylase (DeoD); EC=2.4.2.1;", XP002625014, retrieved from EBI accession no. UNIPROT:Q97Y30 Database accession no. Q97Y30 -& SHE QUNXIN ET AL: "The complete genome of the crenarchaeon Sulfolobus solfataricus P2", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 98, no. 14, 3 July 2001 (2001-07-03), pages 7835-7840, XP002625002, ISSN: 0027-8424

2 EP 2 516 637 B1

Description

Field of the Invention

5 [0001] The invention belongs to the field of biotechnology.

Background of the invention

[0002] (Deoxy)nucleosides are glycosylamines consisting of a base like a purine or a pyrimidine bound to a ribose or 10 deoxyribose sugar, the latter being cyclic pentoses. Examples of these include cytidine, uridine, adenosine, guanosine, thymidine, and inosine. Nucleoside analogues are extensively used as antiviral and anticancer agents because of their ability to act as reverse transcriptase inhibitors or chain terminators in RNA or DNA synthesis [1]. [0003] Chemical synthesis of nucleoside analogues has been achieved stereoselectively but using expensive or pol- luting reagents [2] and involving multistage processes that can be time consuming. Biocatalytic procedures offer a good 15 alternative to the chemical synthesis of nucleosides because biocatalyzed reactions are regio- and stereoselective and allow the decrease of by-products content. Of particular interest within the biocatalytic procedures is the enzymatic transglycosylation between a sugar-donating nucleoside and an acceptor base by means of enzymes that catalyse the general reversible reactions [3] as depicted in Figures 1 and 2. [0004] Nucleoside phosphorylases are transferases widely distributed in mammalian cells and bacteria and play a 20 central role in the nucleoside metabolism salvage pathway. They have a dual functionality. On the one hand, they catalyse the reversible cleavage of the glycosidic bond of ribo- or deoxyribo nucleosides in the presence of inorganic phosphate in order to generate the base and ribose- or deoxyribose-1-phosphate. These enzymatic reactions employing the purine nucleoside phosphorylases and the pyrimidine nucleoside phosphorylases are shown in Figure 1. On the other hand, these enzymes catalyse phosphate-dependent pentose transfer between purine or pyrimidine bases and 25 nucleosides, i.e. transglycosylation reactions, to produce nucleosides with differing bases. Figure 2 shows an example of a one-pot synthesis using nucleoside phosphorylases. [0005] When the pyrimidine and purine nucleoside phosphorylases are used in combination, it is possible to transfer the sugar from a donor pyrimidine nucleoside to a purine or pyrimidine acceptor base as well as from a donor purine nucleoside to a pyrimidine or purine acceptor base, depending on the starting materials used [4]. As a consequence, 30 nucleoside phosphorylases from different sources, mainly bacterial, have been exploited as tools for the enzymatic synthesis of nucleoside analogues. [0006] In nature these enzymes have been described in various microbial strains, particularly in thermophilic bacteria (i.e. bacteria thriving at temperatures between 45 °C and 80 °C), which have been used as sources of nucleoside phosphorylases in numerous works for obtaining modified nucleosides by enzymatic transglycosylation. However, al- 35 though in these studies the target products yields were sufficiently high, the amount or ratio of the enzymatic activities necessary for transglycosylation was non-optimal [5]. They required either a considerable extension in the reaction time (up to several days) or an increase in the used bacterial biomass to reach the necessary transformation depth. [0007] Besides, when developing a transglycosylation process another problem arises: the difficult solubilization of large amounts of substrates and products, many of them poorly soluble in aqueous medium at room temperature. 40 Although this problem could be solved using higher temperatures, it requires enzymes sufficiently stable in these harder reaction conditions. [0008] The Archaea are a group of single-celled microorganisms that are one of the three domains of life; the others being Bacteria and Eukarya. They were formerly called Archaebacteria under the taxon Bacteria, but now are considered separate and distinct. The archaeal domain is currently divided into two major phyla, the Euryarchaeota and Crenar- 45 chaeota. The Euryarchaeota includes a mixture of methanogens, extreme halophiles, thermoacidophiles, and a few hyperthermophiles. By contrast, the includes only hyperthermophiles. Hyperthermophiles are those or- ganisms that thrive in extremely hot environments, from 60 °C upwards, optimally above 80 °C. [0009] Cacciapuoti et al. [6-8] describe two purine nucleoside phosphorylases (PNPases) from hyperthermophilic Archaea, in particular it discloses the enzymes 5’-deoxy-5’-methylthioadenosine phosphorylase II (SsMTAPII, EC 50 2.4.2.28) from Sulfolobus solfataricus, and purine nucleoside phosphorylase (PfPNP) fromPyrococcus furiosus. The Pyrococcus furiosus enzyme was firstly annotated as MTAPII but renamed to PNP as it is unable to cleave methylthio- adenosine. Sulfolobus solfataricus belongs to the Crenarchaeota, while Pyrococcus furiosus belongs to the Euryarchae- ota. The EC code above is the conventional enzyme nomenclature provided by the International Union of Biochemistry and Molecular Biology that classifies enzymes by the reactions they catalyse. 55 [0010] Most enzymes characterized from hyperthermophiles are optimally active at temperatures close to the host organism’s optimal growth temperature. When cloned and expressed in mesophilic hosts likeEscherichia coli, hyper- thermophilic enzymes usually retain their thermal properties. Sometimes the enzymes are optimally active at tempera- tures far above the host organism’s optimum growth temperature [9]. Other times enzymes have been described to be

3 EP 2 516 637 B1

optimally active at 10 °C to 20 °C below the organism’s optimum growth temperature [10-11]. However, the Sulfolobus solfataricus 5’-methylthioadenosine phosphorylase (a hexameric enzyme containing six intersubunit disulfide bridges), when expressed in a mesophilic host, forms incorrect disulfide bridges and is less stable and less thermophilic than the native enzyme [12]. 5 [0011] The are a hyperthermophilic class of the Crenarchaeota. From the genomes sequenced and available for the Archaea Thermoprotei class, only three sequences for purine-nucleoside phosphorylase (EC 2.4.2.1) and only three sequences for uridine phosphorylase (EC 2.4.2.3), were found. These six proteins have been entered, respectively, in UniProtKB/TrEMBL with the accession numbers: A1RW90 (A1RW90_THEPD), for the hypothetical protein from Thermofilum pendens (strain Hrk 5); Q97Y30 (Q97Y30_SULSO), for the hypothetical protein from Sulfolobus 10 solfataricus; A3DME1 (A3DME1_STAMF), for the hypothetical protein fromStaphylothermus marinus (strain ATCC 43588 / DSM 3639 / F1); Q9YA34 (Q9YA34_AERPE), for the hypothetical protein fromAeropyrum pernix; A2BJ06 (A2BJ06_HYPBU) for the hypothetical protein from butylicus (strain DSM 5456 / JCM 9403); and D9PZN7 (D9PZN7_ACIS3) for the hypothetical protein fromAcidilobus saccharovorans (strain DSM 16705 / VKM B-2471 / 345-15). All these sequences were under the annotation status of unreviewed, which means that their presence in the 15 Archaea has only been verified by computer. [0012] Even though many genes can be successfully expressed inEscherichia coli at high yields, several proteins from hyperthermophiles are poorly or not at all expressed, partially due to the usage of rare codons. Indeed, and to the best of our knowledge, no party was yet successful in expressing any of the mentioned genes above. [0013] In view of the prejudices above, in view of the technical difficulties, the inventors unexpectedly were able to 20 prepare viable recombinant vectors and importantly, obtain recombinant phosphorylases that were optimally active at temperatures higher than 60 °C. The thermostable and chemically stable catalysts of the present invention are a purine nucleoside phosphorylase (PNPase, E.C. 2.4.2.1), and a uridine phosphorylase (UPase, E.C. 2.4.2.3), originating from the Archaea Thermoprotei class, wherein the PNPase is from Sulfolobus solfataricus (SEQ ID NO. 7) and the UPase is from Aeropyrum pernix (SEQ ID NO. 8). 25 [0014] In particular, it has been surprisingly found that the recombinant nucleoside phosphorylases derived from the hyperthermophilic Thermoprotei have unique structure-function properties like enhanced thermostability, high catalytic efficiency, and optimal enzymatic activities at temperatures near or above 100°C. These recombinant enzymes can advantageously be used for transglycosylation reactions, in the form of cell lysate and in the form of crude or purified extracts, for industrial production of natural and modified nucleoside analogues. They are in particular versatile since 30 they can catalyze transglycosylations in aqueous media, in organic solvents, at temperatures between 60 °C and 120°C, or in a combination of these parameters, allowing the preparation of many and diverse types of nucleosides at acceptable production yields, reaction times, and employing economical amounts of the enzymes. Importantly, the biocatalysts described in the present invention can be used for bioconversion reactions that require the presence of organic solvents, temperatures above 60°C, or both, in order to solubilize the substrates or the reaction products. These phosphorylases 35 are ideal in reactions with water-insoluble substrates. Another advantage of these phosphorylases resides in their organic solvent tolerance, and in that they can be reused for several reaction cycles. [0015] More advantageously, the invention offers a combination of Thermoprotei nucleoside phosphorylases that is useful for one-pot synthesis of nucleosides. The enzymes can be used to produce natural or analog nucleosides in a one-step (one-pot) or two-step synthetic methods. In the one-step synthesis, a pyrimidine nucleoside phosphorylase 40 and a purine nucleoside phosphorylase are used in the same batch in order to change the base linked to the sugar by another one of choice. In the two step, a pyrimidine nucleoside phosphorylase is used for the liberation of the sugar of a pyrimidine nucleoside, and then, the 1-phosphate-sugar is isolated and later on, in another vessel, a purine base is linked to the sugar using a purine nucleoside phosphorylase. [0016] Taran S A et al., "Enzymatic transglycosylation of natural and modified nucleosides by immobilized thermostable 45 nucleoside phosphorylases from Geobacillus stearothermophilus", Russian Journal of Bioorganic Chemistry, vol. 35, No. 6, 6 November 2009, pages 739-745, discloses enzymatic transglycosylation of natural and modified nucleosides by immobilized thermostable nucleoside phosphorylases from Geobacillus stearothermophilus: purine nucleoside phos- phorylase II (E. C. 2.4.2.1) and pyrimidine nucleoside phosphorylase (E. C. 2.4.2.2). [0017] Trelles J A et al., "Screening of Catalytically active microorganisms for the synthesis of 6-modified purine 50 nucleosides", Biotechnology Letters, Kluwer Academic Publishers, DO LNKD-DOI: 10.1007/S10529-005-5628-7, vol. 27, no. 11, 1 June 2005, pages 759-763 discloses a screening method to find transglycosylation activity in microorganisms for the synthesis of 6-modified purine nucleosides. [0018] Kawarabayashi Yutaka et al. "Complete genome sequence of an aerobic hyperthermophilic crenarchaeon, Aeropyrum pernix K1", DNA Research, Universal Academy Press, JP LNKD-DOI:10.1093/DNARES/6.2.83., vol 6, no. 55 2, 1 January 1999, pages 83-101, discloses a uridine phosphorylase from Aeropyrum pernix. [0019] Hamamoto Tomoki et al. "Cloning of purine nucleoside phosphorylase II gene from Bacillus stearothermophilus TH 6-2 and characterization of its gene product", Bioscience Biotechnology and Biochemistry, vol. 61, no. 2, 1997, pages 276-280, discloses the process of cloning and sequencing Pu-NPase II from TH 6-2 strain fromBacillus stearother-

4 EP 2 516 637 B1

mophilus. The Pu-NPase was purified and characterized. [0020] Hamamoto Tomoki et al. "Cloning and expression of purine nucleoside phosphorylase I gene from Bacillus stearothermophilus TH 6-2", Bioscience Biotechnology and Biochemistry, vol. 61, no. 2, 1997, pages 272-275, discloses the process of cloning, sequencing and expressing the Pu-NPase I gene from TH 6-2 strain from Bacillus stearother- 5 mophilus. The cloned gene was overexpressed in E. coli by using the trc promoter to produce an active enzyme in large quantitites. [0021] Cacciapuoti Giovanna et al. "Purine nucleoside phosphorylases from hyperthermophilic Archaea require a CXC motif for stability and folding", The Febs Journal Oct 2009 LNKDS-PUBMED: 19740110, vol. 276, no. 20, October 2009, pages 5799-5805, discloses the study of how the CXC motif improves both stability and folding in hyperthermophilic 10 proteins with disulfide bonds, in particular in the hyperthermophilic 5’-desoxy-5’-methyladenosine phosphorylase II from Sulfolobus solfataricus (SsMTAPII). [0022] Cacciapuoti Giovanna et al. "A novel hyperthermostable 5’-deoxy-5’-methylthioadenosine phosphorylase from the archaeon Sulfolobus solfataricus", Febs Journal, vol. 272, no. 8, April 2005, pages 1886-1899, discloses the hyper- thermostable 5’-desoxy-5’-methythioladenosine phosphorylase from the archaeon Sulfolobus solfataricus, which was 15 cloned and expressed in E. coli. [0023] Cacciapuoti Giovanna et al. "Biochemical and structural characterization of mammalian-like purine nucleoside phosphorylase from the Archaeon Pyrococcus furiosus", Febs Journal, vol. 274, no. 10, May 2007, pages 2482-2495, discloses the characterization of the first mammalian-like purine nucleoside phosphorylase from the hyperthermophilic achaeon Pyrococcus furiosus (PfPNP). The gene PF0853 encoding PfPNP was cloned and expressed inE. coli and 20 the recombinant protein was purified to homogeneity. [0024] She Quinxin et al. "The complete genome of the crenarchaeon Sulfolobus solfataricus P2", Proceedings of the National Academy of Sciences of the United States of America, vol. 98, no. 14, 3 July 2001, pages 7835-7840, discloses the sequence of the DeoD protein from Sulfolobus solfataricus whose coding sequence (nucleotides 1372685-1371861 of the genome of Sulfolobus solfataricus) corresponds to SEQ ID NO: 7 of the present invention. 25 Summary of the invention

[0025] The present invention relates to a recombinant expression vector as defined in the claims. [0026] In addition, the present invention relates to a transglycosylation method as defined in the claims. 30 Description of the figures

[0027]

35 Figure 1 shows an example of two enzymatic reactions catalyzed by nucleoside phosphorylases. The first reaction

on top is a phosphorolysis that takes place through an S N1-like mechanism via an oxonium-like intermediate to give α-ribose-1-phosphate. The second reaction occurs through an S N2 mechanism where phosphate is substituted by a base affording the β-nucleoside [13]. In the scheme, uridine nucleoside phosphorylase catalyzes the phosphorolytic cleavage of the C-N glycosidic bond of uridine resulting in ribose-1-phosphate and uracil. The purine nucleoside 40 phosphorylase (adenosine nucleoside phosphorylase) catalyzes the cleavage of the glycosidic bond, in the presence

of inorganic orthophosphate (Pi) as a second substrate, to generate the purine base and ribose(deoxyribose)-1- phosphate. For the natural substrates, the reactions are reversible.

Figure 2. Scheme of one-pot synthesis using nucleoside phosphorylase enzymes. 45 Figure 3 shows a genetic map of the initial expression vector pET102/D-TOPO® before cloning. Vector length of 6315 nucleotides. T7 promoter: bases 209-225; T7 promoter priming site: bases 209-228; lac operator (lacO): bases 228-252; ribosome binding site (RBS): bases 282-288; His-patch (HP) thioredoxin ORF: bases 298-627; TrxFus forward priming site: bases 607-624; EK recognition site: bases 643-657; TOPO ® recognition site 1: bases 670-674; 50 overhang: bases 675-678; TOPO® recognition site 2: bases 679-683; V5 epitope: bases 700-741; polyhistidine (6xHis) region: bases 751-768; T7 reverse priming site: bases 822-841; T7 transcription termination region: bases 783-911; bla promoter: bases 1407-1505; Ampicillin ( bla) resistance gene (ORF): bases 1506-2366; pBR322 origin: bases 2511-3184; ROP ORF: bases 3552-3743 (complementary strand); lacI ORF: bases 5055-6146 (complemen- tary strand). 55 Figure 4 depicts a "Doehlert Matrix" where five temperatures were combined with three pH values, resulting in seven combinations of temperature and pH.

5 EP 2 516 637 B1

Figure5. Contour plot showing the interactive effectof pH and temperatureon theUPase activity.Data was statistically analyzed with the Response Surface Methodology (RSM), using the "Minitab" software. The enzyme appears highly thermophilic; its activity increased sharply up to the maximal assayed temperature (100°C) and the activity displayed a distinct pH optimum around the neutrality (6.5-7.5), preferably 7.0. 5 Figure 6. Contour plot showing the interactive effect of pH and temperature on the PNPase activity. Data was statistically analyzed with the Response Surface Methodology (RSM), using the "Minitab" software. The enzyme appears highly thermophilic; its activity increased sharply up to the maximal assayed temperature (100°C) and the activity displayed a distinct pH optimum around the neutrality (6.5-7.0). 10 Figure 7 depicts the DNA sequence (SEQ ID NO. 7) of the coding region of the purine nucleoside phosphorylase (PNPase) of Sulfolobus solfataricus, a.k.a. deoD gene. GenBank accession number AE006766.

Figure 8 depicts the DNA sequence (SEQ ID NO. 8) of the coding region of the pyrimidine nucleoside phosphorylase 15 (UPase) of Aeropyrum pernix, a.k.a. udp gene. GenBank accession number NC000854.

Description of the invention

[0028] The present invention relates to a recombinant expression vectoras defined in the claims, being the rest of the 20 embodiments related thereto not part of the invention, but representing background art useful for understanding the invention. [0029] Aeropyrum pernix and Sulfolobus solfataricus are hyperthermophilic Archaea capable of growing at high tem- peratures, over 90°C. Archaea are organisms belonging to a third group of organisms distinct from eukaryotes and prokaryotes. They are considered to descend from primeval organisms, and are special organisms which have neither 25 evolved nor adapted to ordinary temperature environments. [0030] UPase and PNPase in their intracellular natural environment do not allow the synthesis of nucleoside or nucl- eoside analogues with high yield as desired at industrial level. To overcome this serious limitation, inventors have used recombinant DNA technology to design an expression vector comprising udp and deoD genes and appropriate elements to over-express nucleoside phosphorylases in selected hosts, like bacteria. The designed expression vector also facil- 30 itates the solubilization and the purification of the different phosphorylases. [0031] The vectors of the present invention comprise a nucleotide sequence encoding different nucleoside phospho- rylases and nucleotide sequences that allow said vector to be selectable and autonomously replicable in the host cell. [0032] The construction of the recombinant expression vector is carried out using conventional recombinant DNA technologies, i.e. procedures to join together DNA segments in a cell-free system. 35 [0033] The term "vector" refers to a DNA molecule originating from a virus, a plasmid, or the cell of a higher organism in which another DNA fragment of appropriate size can be integrated (cloned) without loss of the vector capacity for self-replication. Examples are plasmids, cosmids, and yeast artificial chromosomes. Vectors are often recombinant molecules containing DNA sequences from several sources. The term "expression vector" means a vector that further comprises the necessary control or regulatory sequences to allow transcription and translation of the cloned gene or 40 genes. Circular or linearized DNA vectors are useful for this invention. [0034] To allow the vector of the invention to be selectable and autonomously replicable in host cells, the selected vector must be compatible with the selected host cells. The nucleotide sequence which allows said vector to be selectable and autonomously replicable in Escherichia coli is the T7 promoter-encoding gene which permits the T7 RNA polymerase of the selected strain of Escherichia coli to bind to the promoter. The term "selectable" means that the vector remains 45 stable in the descendent bacteria. The selection is achieved by stringent medium conditions according to the introduction of an appropriate selectable marker gene in the vector whose expression allows one to identify cells that have been transformed with the vector. The selectable marker gene is often an antibiotic-resistant gene. Preferred selectable marker genes for this invention are kanamycin, tetracycline, carbenicillin and more preferably, ampicillin. [0035] The present invention further relates to the E. coli host cell as defined in the claims. 50 [0036] The term "host cell" refers to a cell transformed with the recombinant expression vector that comprises the PNPase and UPase nucleotide sequence. Another aspect of the recombinant DNA vector allows the host cell to produce nucleoside phosphorylases, and when medium conditions are suitable, said nucleoside phosphorylases catalyze the obtention of nucleosides. PNPase and UPase genes from Sulfolobus solfataricus and Aeropyrum pernix, respectively, were introduced in the DNA expression vector. Figure 7 and Figure 8 list nucleic acid and amino acid sequences relevant 55 to the invention, namely the nucleic acid sequence of Sulfolobus solfataricus deoD (SEQ ID NO. 7) and nucleic acid sequence of Aeropyrum pernix udp (SEQ ID NO. 8), respectively. [0037] Those skilled in the art will appropriately choose the expression system constituted by an initial vector and a host cell strain to maximize the production of nucleosides.

6 EP 2 516 637 B1

[0038] The Escherichia coli belongs to BL21 bacterial strain. Suitable expression vectors forEscherichia coli BL21 are for instance pET vectors, trcHis vectors and pUB vectors (all of them from Invitrogen), and pGEX vectors and GST vectors (from Amersham). Escherichia coli DH5 alfa bacterial strain in combination with pUC vectors and Escherichia coli F’ in combination with PSL vectors, PEZZ vectors or M13 vectors (all of them from Amersham) are also useful in 5 this invention. [0039] In one embodiment, a lysate is obtained by lysing the host cell as defined in the claims. [0040] The present invention further relates to a transglycosylation methodas defined in the claims, being the rest of the embodiments related thereto not part of the invention, but representing background art useful for understanding the invention. 10 [0041] The term "sugar-donating nucleoside" refers to a glycosylamine consisting of a nucleobase (often referred to as simply base) bound to a ribose or deoxyribose sugar via a beta-glycosidic linkage. Examples of "sugar-donating nucleosides" include, without being limited to, cytidine, uridine, adenosine, guanosine, thymidine and inosine, as well as those natural or modified nucleosides containing D-ribose or 2’-deoxyribose; nucleosides containing the ribose group modified in the 2’, 3’, and/or 5’ positions; and nucleosides in which the sugar is beta-D-arabinose, alpha-L-xylose, 3’- 15 deoxyribose, 3’,5’-dideoxyribose, 2’,3’-dideoxyribose, 5’-deoxyribose, 2’,5’-dideoxyribose, 2’-amino-2’-deoxyribose, 3’- amino-3’-deoxyribose, or 2’-fluoro-2’-deoxyribose. [0042] The term "acceptor base" refers to a nucleobase, nucleotide base, nitrogenous base, or simply base. In nature, bases are part of DNA or RNA. The primary nucleobases are cytosine, guanine, adenine (DNA and RNA), thymine (DNA) and uracil (RNA), abbreviated as C, G, A, T, and U, respectively. The term "acceptor base" in the present invention 20 is meant to comprise also modified and analog nucleobases. In DNA, the most common modified base is 5-methylcytidine (m5C). In RNA, there are many modified bases, including pseudouridine ( Ψ), dihydrouridine (D), inosine (I), ribothymidine (rT) and 7-methylguanosine (m7G). Hypoxanthine and xanthine are two of the many bases created through mutagen presence. Other examples of acceptor bases include natural or substituted pyrimidine and purine bases; purine bases substituted at one or more of the 1, 2, 6 positions; pyrimidine bases substituted at one or more of the 3, 5 positions; and 25 purine, 2-azapurine, 8-azapurine, 1-deazapurine (imidazopyridine), 3-deazapurine, 7-deazapurine, 2,6-diaminopurine, 5-fluorouracil,5-trifluoromethyluracil, trans-zeatin, 2-chloro-6-methylaminopurine,6-dimethylaminopurine, 6-mercaptop- urine. [0043] This transglycosylation method is useful for the preparation of nucleosides, nucleosides analogs, and particularly active pharmaceutical ingredients (API); comprising, containing, or consisting of nucleoside moieties, or analogs thereof. 30 Understanding API as any substance or mixture of substances intended to be used in the manufacture of drug (medicinal) product and that, when used in the production of a drug, becomes an active ingredient of the drug product. Such substances are intended to furnish pharmacological activity or other effect in the diagnosis, cure, mitigation, treatment, or prevention of disease or to affect the structure and function of the body. (Eudralex, Part II of volume 4 EU Guidelines to Good Manufacturing Practice). 35 [0044] The combination of uridine phosphorylase (UPase, E.C. 2.4.2.3) and purine nucleoside phosphorylase (PNP; E.C. 2.4.2.1) efficiently transfers a sugar moiety from a donor nucleoside to an acceptor base. [0045] When pyrimidine nucleosides are prepared departing from other pyrimidine nucleosides and pyrimidine bases as starting materials, then the use of the UPase alone is sufficient, but the use of both enzymes PNPase and UPase is preferred because the PNPase can also contribute to the phosphorolysis step. Conversely, when purine nucleosides 40 are prepared departing from other purine nucleosides and purine bases as starting materials, then the use of both PNPase and UPase is also preferred. On the other hand, the use of both enzymes PNPase and Upase is much more successful when the reaction is from a pyrimidine to a purine nucleoside, for instance from a uridine to a 2,6 diaminopurine riboside, when compared to the use of each type of enzyme per separate. [0046] The transglycosylation method uses a combination of the UPase and PNPase. The crude cell lysates or the 45 clarified crude enzyme solutions may be mixed in different proportions in order to obtain an optimized biocatalyst for a particular transglycosylation reaction. [0047] In the transglycosylation method of the present invention, the UPase of Aeropyrum pernix and the PNPase of Sulfolobus solfataricus are provided by a host cell as defined in the claims. [0048] In one embodiment, in the transglycosylation method of the present invention, the combination of UPase, and 50 PNPase are used in the form of a lysate. [0049] In one embodiment, the transglycosylation method comprises the steps of: (i) culturing the host cell in a suitable culture medium; (ii) overexpressing the UPase, PNPase, or both; (iii) optionally preparing a cell lysate; (iv) adding a sugar-donating nucleoside, an acceptor base, and phosphate ions, and (v) recovering nucleosides from the reaction mixture. 55 [0050] In a particular embodiment the Escherichia coli transformant comprising the vector may be grown in a culture medium comprising tryptone, yeast extract, sodium chloride and an antibiotic selected from the group consisting of kanamycin, tetracycline, carbenicillin, and ampicillin, preferably at 37°C, to an optical density between 0.5-0.8 at a wavelength of approximately 600 nm. The culture may be then added with isopropyl-beta-D-thiogalactopyranoside

7 EP 2 516 637 B1

(IPTG) to a final concentration of 100 mg/l and the inductions may be done at 37°C between 6-12 h. Cells may be harvested by centrifugation at 4°C and cell pellets may be lysed by three freeze-thaw cycles. The recombinant host cells may be disrupted by standard techniques known to those skilled in the art. The resulting cell lysate may be directly used as biocatalyst or centrifuged to remove cell debris and to obtain a clarified crude enzyme solutions. As used herein, the 5 term "biocatalyst" refers to any biological entity capable of catalyzing the conversion of a substrate into a product, in this case the nucleoside biotransformation. [0051] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from natural or modified nucleosides containing D-ribose and 2’-deoxyribose; nucleosides containing the ribose group modified in the 2’, 3’ and/or 5’ positions; and nucleosides in which the sugar is beta-D-arabinose, alpha-L- 10 xylose, 3’-deoxyribose, 3’,5’-dideoxyribose, 2’,3’-dideoxyribose, 5’-deoxyribose, 2’,5’-dideoxyribose, 2’-amino-2’-deox- yribose, 3’-amino-3’-deoxyribose, 2’-fluoro-2’-deoxyribose. [0052] In one embodiment, in the transglycosylation method of the present invention, the acceptor base is selected from natural or substituted pyrimidine and purine bases; purine bases substituted at the 1, 2, 6 positions, or a combination thereof of the purine ring; pyrimidine bases substituted at the 3, 5 positions, or a combination thereof of the pyrimidine 15 ring; for instance purine, 2-azapurine, 8-azapurine, 1-deazapurine (imidazopyridine), 3-deazapurine, 7-deazapurine, 2,6-diaminopurine, 5-fluorouracil, 5-trifluoromethyluracil, trans-zeatin, 2-chloro-6-methylaminopurine, 6-dimethylami- nopurine, 6-mercaptopurine. [0053] In one embodiment, in the transglycosylation method of the present invention, the resulting nucleoside analogue is an active pharmaceutical ingredient (API) as known in the art. 20 [0054] In one embodiment, in the transglycosylation method of the present invention, said method is carried out between 60 and 100°C. [0055] In one embodiment, the transglycosylation method of the present invention is carried out in aqueous media, or in an aprotic polar co-solvent system. [0056] In one embodiment, the aprotic polar co-solvent system is selected from dimethylsulfoxide, tetrahydrofuran, 2- 25 methyltetrahydrofuran, dimethylformamide, or any combination thereof. [0057] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is 2,6-diaminopurine. [0058] In one embodiment, it is provided a one-pot enzymatic synthesis of nucleosides using uridine or 2’-deoxyuridine as donors of sugar moiety, because the recombinant UPase enzyme of the present invention is most specific for these 30 substrates. Nevertheless, this enzyme can be used with any donor of sugar moieties because it does not discriminate between uridine, 2’-deoxyuridine, and other pyrimidine nucleosides, as unfortunately occurs in many lower organisms, such as B. stearothermophilus [14]. [0059] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is 5-fluorouracil. 35 [0060] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is 5-trifluoromethyluracil. [0061] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is trans-zeatin. [0062] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside 40 is selected from uridine and 2’-deoxyuridine, and the acceptor base is 2-chloro-6-methylaminopurine. [0063] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is 6-dimethylaminopurine. [0064] In one embodiment, in the transglycosylation method of the present invention, the sugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is 6-mercaptopurine. 45 [0065] The term "thermostable nucleoside phosphorylase" refers to enzymes that are stable to heat, are heat resistant, and retain sufficient nucleoside phosphorylase activity to effect another reactions and does not become irreversibly denatured (inactivated) when subjected to elevated temperatures for the time necessary to effect the transglycosylation reactions. [0066] The purpose of the Examples given below is to illustrate the present invention without constituting a limitation 50 of the field of application thereof.

Examples

[0067] As stated above, the transglycosylation reactions according to the invention were carried out using UPase and 55 PNPase enzymesobtained from UPase- or PNPase-producing cells.Such cells are preferablycells ofgenetically modified Escherichia coli, capable of expressing considerable quantities of UPase or PNPase. The manner of obtaining such cells, the enzymes and their characteristics, as well as the production of some nucleoside analogues are given in the accompanying examples, which are set out below.

8 EP 2 516 637 B1

Example 1

Construction of the Escherichia coli deoD

5 [0068] The Purine Nucleoside Phosphorylase (PNPase) of Sulfolobus solfataricus sequence was found in GenBank, with the accession number AE006766. The gene was amplified by PCR, using 2 units of Platinum Pfx enzyme (Invitrogen),

1mM MgSO4 and 1x enzyme amplification buffer, 200 mM dNTPs and 0.3 mM of each primer, with the oligonucleotides 5’-caccgtgccatttttagaaaatggttcc-3’ (Sulfolobus solfataricus deoD forward; SEQ ID NO. 1) and 5’- aatcagttttaagaatcttaag- gtaat-3’ (Sulfolobus solfataricus deoD reverse; SEQ ID NO. 2) from the Sulfolobus solfataricus P2 [15]. The PCR reaction 10 was performed with an initial denaturation step at 94°C for 30 min, followed by 36 temperature cycles of a denaturation step at 94°C for 1 min, and an annealing/extension step at 60°C for 1.5 min and 68°C for 1 min. After the 36 cycles, the sample was subjected to 68°C for 10 min and finally at 4°C. PCR product was analyzed by agarose gel electrophoresis, and the DNA band was purified from the gel (S.N.A.P.™ UV-Free Gel Purification Kit, Invitrogen). The amplified fragment was cloned into the polylinker region of the pUC18 vector that carries the ampicillin resistance gene [17]. The cloned 15 region was completely sequenced and it was found to be completely identical to the data bank sequence.

Example 2

Construction of the Escherichia coli udp 20 [0069] The Pyrimidine Nucleoside Phosphorylase (UPase) of Aeropyrum pernix sequence was found in GenBank with the accession number NC_000854.2. The gene was amplified by PCR, using 2 units of Platinum Pfx enzyme (Invitrogen),

1mM MgSO4 and 1x enzyme amplification buffer, 200 mM dNTPs and 0.3 mM of each primer, with the oligonucleotides 5’- caccgtggcccgctacgttctcctc-3’ (Aeropyrum pernix udp forward; SEQ ID NO. 3) and 5’-gaattcctatgtgcgtctgcacgccagg- 25 3’ (Aeropyrum pernix reverse; SEQ ID NO. 4) from the Aeropyrum pernix K1 [16]. The PCR reaction was performed with an initial denaturation step at 94°C for 30 min, followed by 36 temperature cycles of a denaturation step at 94°C for 1 min, and an annealing/extension step at 60°C for 1.5 min and 68°C for 1 min. After the 36 cycles, the sample was subjected to 68°C for 10 min and finally at 4°C. PCR product was analyzed by agarose gel electrophoresis, and the DNA band was purified from the gel (S.N.A.P.™ UV-Free Gel Purification Kit, Invitrogen). The amplified fragment was 30 cloned into the polylinker region of the pUC18 vector that carries the ampicillin resistance gene [17]. The cloned region was completely sequenced and it was found to be completely identical to the data bank sequence.

Example 3

35 Cloning into pET102/D-TOPO® vector and cells transformation

[0070] The DNA fragments containing the Aeropyrum pernix udp gene sequence and Sulfolobus sulfactaricus deoD gene sequence were cloned into pET102/D-TOPO ® vector (pET102 Directional TOPO ® Expression Kit, Invitrogen) that comprises a pBR322ori for plasmid replication, ampicillin resistance gene, T7 promoter that permits binding of T7 RNA 40 polymerase, lac operator that permits inhibition of expression when isopropylthiogalactopyranoside (IPTG) is not present, a ribosome binding site for translation of RNA, a His-patch-thioredoxin for increasing the solubility of the fusion protein, and a polyhistidine tag (6xHis) for detection and purification of the fusion protein (Figure 3). The vector was introduced by thermal shock into Escherichia coli BL21 (pET102 Directional TOPO ® Expression Kit, Invitrogen), a commercial strain that is used for regulated expression of heterologous genes. It comprises the gene encoding the T7 RNA polymerase, 45 which makes the strain compatible with the use of pET vectors comprising T7 promoter for the overexpression of recombinant proteins with IPTG. [0071] The resulting recombinant vectors were analyzed by DNA restriction with 10 units of the enzyme HindIII. The positive clone was sequenced using the sequencing primers TrxFus forward 5’TTCCTCGACGCTAACCTG3’ (SEQ ID NO. 5) and T7 reverse 5’TAGTTATTGCTCAGGGGTGG3’ (SEQ ID NO. 6). 50 Example 4

Fermentation of the recombinant strains

55 [0072] The recombinant strains to which the present invention relates were cultivated separately in batch mode, at pH=7, in trypticase soy solid medium supplemented with ampicillin. One colony of the culture was passed into nutrient broth n° 2 (Oxoid) containing Lab-lemco powder 10 g/l, peptone 10 g/l and NaCl 5 g/l, supplemented with 200 mg/l ampicillin. It was incubated at 37°C with vigorous shaking (200 rpm). Escherichia coli strains harbouring the expression

9 EP 2 516 637 B1

plasmid were grown to Optical Density at 600 nm of 0.6, then IPTG (up to 100 mg/l) was added and the culturing was continued for an additional 8 h. When fermentation was complete, the culture medium was centrifuged, the cell pellet was washed in 30 mM-pH 7 phosphate buffer. The biomass obtained was stored at -20° C until it was brought into use.

5 Example 5

Partial UPase and PNPase purification (cell lysate preparation)

[0073] For a partial purification of the protein, cell paste, separated by centrifugation or by microfiltration from a culture 10 of the recombinant strain that expresses the enzyme UPase or PNPase, was disrupted by the addition of 160 mg lysozyme and one hour of incubation at 0°C followed by three ultra rapid freezing-thawing cycles (-80°C / 37°C) and a final step for reducing viscosity adding 1,000 units of deoxyribonuclease I.

Example 6 15 Determination of the enzymatic activity of the UPase enzyme

[0074] One hundred microliters of centrifuged cell lysate, comprising a suspension of UPase expressing cells, diluted 1:100 (volume/volume) in potassium phosphate buffer at pH 7.0, was added to 800 microliters of 75 mM uridine solution 20 in 100 mM phosphate buffer at pH 7.0, preincubated at 30°C. After 5 minutes, the phosphorolysis reaction was stopped by addition of 1 ml of 2N HCl. An aliquot of the reaction mixture was analyzed using a high performance liquid chroma- tograph (HPLC), equipped with a Kromasil 100-5C18 (Akzo Nobel) column, with a size of 250 x 4.6mm. The elution was carried out using a 4% methanol-water solution. The enzymatic activity of the cell lysate was expressed as units per ml (mmoles of uracil x min-1 x ml-1) and was calculated relative to a standard uracil solution eluted in the same conditions. 25 Approximately 590 units per ml of centrifuged cell lysate were recovered.

Example 7

Determination of the enzymatic activity of the PNPase enzyme 30 [0075] One hundred microliters of centrifuged cell lysate, comprising a suspension of PNPase expressing cells, diluted 1:100 (volume/volume) in potassium phosphate buffer at pH 7.0, was added to 800 microliters of 60 mM inosine solution in 100 mM phosphate buffer at pH 7.0, preincubated at 30°C. Exactly 10 minutes later, the phosphorolysis reaction was stopped by addition of 1 ml of 2N HCl. An aliquot of the reaction mixture was analyzed using a high performance liquid 35 chromatograph (HPLC), equipped with a Kromasil 100-5C18 (Akzo Nobel) column, with a size of 250 x 4.6mm. The elution was carried out using a 4% methanol-water solution. The enzymatic activity of the cell lysate was expressed as units per ml (mmoles of hypoxanthine x min-1 x ml-1) and was calculated relative to a standard hypoxanthine solution eluted in the same conditions. Approximately 310 units per ml of centrifuged cell lysate were recovered.

40 Example 8

Determination of transglycosylation catalytic activity

[0076] For the determination of transglycosylation catalytic activity the mixture of cell lysates containing both UPase 45 and PNPase were prepared by mixing the lysates so as to have UPase: PNPase enzymatic-activity ratio of about 1:1, determined as in Examples 6 and 7. Transglycosylation reaction was carried out at analytical scale in the following conditions: 250 ml of cell lysates (equivalent to 14 units of each UPase and PNPase enzymatic activities) was added to 10 ml of a solution having the following composition: 4 mM 1- β-D-ribofuranosyluracil (uridine nucleoside), 4 mM adenine base, 30 mM potassium phosphate buffer pH 7, thermostatically controlled at 60°C. After 1.5 hours at 60°C, the reaction 50 was stopped by diluting the mixture 1:5 and cooling in ice. The percentage of bioconversion of adenine base to 9- β-D- ribofuranosyladenine (adenosine nucleoside) was determined by analyzing an aliquot of the reaction mixture by high performance liquid chromatography (HPLC) with the use of Kromasil 100-5C18 (Akzo Nobel) column of a size of 250 x 4.6mm and eluted with a 4% methanol-water solution. The transglycosylation catalytic activity was expressed as units · ml -1 (mmoles ofAra-A formed in 1.5 hours · ml -1 of mixture of cell lysates) or in units · g -1 of moist resin (mmoles of 55 D-ribofuranosyladenine formed in 1.5 hours · ml -1 of cell lysate) and was calculated relative to a standard D-ribofuran- osyladenine solution eluted by HPLC in the same conditions. Under these conditions, about 55 percent of adenosine nucleoside was formed (approximately 9 units per ml of cell lysate).

10 EP 2 516 637 B1

Example 9

Effect of temperature and pH on the nucleoside phosphorylases activities

5 [0077] The influence of pH and temperature on the performance of the nucleoside phosphorylases was investigated by using a "design of experiments method". Different temperatures were combined with different pH values to ascertain the conditions that resulted in maximum enzymatic activities. An experimental domain was defined between 80°C-100°C and pH 5.5-8.5. As shown in Figure 4, five temperatures were combined with three pH values according to the "Doehlert Matrix", resulting in seven combinations of temperature and pH. The substrates were incubated in the selected reaction 10 solution and temperature without enzyme, and then the enzyme was added and incubated at selected conditions. The samples were processed as described above for the determination of the enzymatic activity of the PNPase or UPase enzymes. Activity values were expressed as a percentage of the corresponding maximal values found (100%). The results are shown in Figures 5 and 6, respectively.

15 Example 10

Thermostability

[0078] Thermostability of the enzymes was assayed at 80°C. Aliquots of the enzyme were prepared, between 100 20 and 200 microliters of a suspension of UPase or/and PNPase expressing cells (cell lysate), diluted 1:100 or 1:1000 volume/volume in potassium phosphate buffer at pH 7.0-7.2. The biocatalysts were incubated at 80°C for diferent times. After heating, the solutions were immediately kept on ice and the measuring of the nucleoside phosphorylases activities was performed as described in examples 6 and 7. No loss of nucleoside phosphorylases activities was observed after 10 h at 80°C. 25 Solvent effects on nucleoside phosphorylase activity

[0079] Organic co-solvents are commonly used in reactions and isolated enzymes must be able to survive under conditions of relatively high concentrations of co-solvent. Experiments were run in the presence of organic solvents such 30 as methanol (protic polar solvent) and dimethylsulfoxide (aprotic polar solvent). The nucleoside phosphorylase enzymes of the present invention were resistant to organic solvents, exhibing activity in a buffer solution containing 5 and 10% by volume of the organic solvent.

Table 1. Stability of PNPase enzyme against some organic solvents. 35 Organic solvents Relative activity (%)a DMSO MeOH 5% (v/v) 94 95

40 10% (v/v) 115 86 aThe relative activity of cell lysate comprising PNPase enzyme was determined using the standard assay described in example 7 at two concentrations of organic solvents and its relative activity was compared with the activity of the cell lysate comprising PNPase enzyme but no organic solvent. DMSO: dimethylsulfoxide; MeOH: methanol.

45

Table 2. Stability of UPase enzyme against some organic solvents. Organic solvents Relative activity (%)a DMSO MeOH 50 5% (v/v) 91 151 10% (v/v) 70 206 bThe relative activity of cell lysate comprising UPase enzyme was measured using the standard assay described in 55 example 6 at two concentrations of organic solvents and its relative activity was compared with the activity of the cell lysate comprising UPase enzyme but no organic solvent.DMSO: dimethylsulfoxide; MeOH: methanol

11 EP 2 516 637 B1

Example 11

General bioconversion procedures

5 [0080] One-pot transglycosylation reactions were performed at selected reaction temperatures and pHs, in reaction vessels with gentle stirring. Catalytic transglycosylation activity of cell lysate mixture used was about 12 units per ml (as determined in Example 8). Nucleosides were prepared using a mixture of the cell lysate prepared to obtain a PNPase:UP- ase activity ratio of 1:1. Substrates were: 10 1) natural 2’-deoxyribonucleosides or ribonucleosides, which act as 2’-deoxyribose-1-phosphate or ribose-1-phos- phate donors; 2) a secondary natural or synthetic purine or pyrimidine base, which is linked to the position 1 of the sugar.

15 [0081] At the end of the reactions, the reaction mixture was centrifuged and then filtered using an Amicon Ultrafiltration device (YM-3 membrane) and the products were separated, according to the protocol provided by the manufacturer. The biocatalysts can be recycled for consecutive reactions (typically between 3-5 times) by addition of newly prepared reaction mixtures. The filtered solutions were monitorized by high performance liquid chromatography (HPLC) in the same conditions described above. Bioconversion yields were calculated based on the initial concentration of base 20 analogue.

Example 12

Preparation of 2,6-Diaminopurine nucleosides 25 [0082] The preparation of purine nucleosides is more difficult because of the low solubility of these bases in aqueous medium. In the present invention, the inventors used the enzyme competitive properties to solve this problem. The strategy used by the inventors encompassed the use of organic co-solvents and/or high temperatures to increase the solubility of the substrates. 30 12.1 Preparation of 2,6-Diaminopurine nucleosides using co-solvents

[0083] Assays were performed in 5 and 10% of aprotic dipolar co-solvents, at 60°C, twelve units/ml cell lysate were added to 150 ml of a solution kept thermostatically at 60 °C, and having the following composition of substrate solutions: 35 1. 15 mM Uridine/2’-Deoxyuridine, 2. 5 mM 2,6-Diaminopurine, and 3. 30 mM potassium phosphate buffer, pH 7.

40 [0084] After 5 hours, the reaction mixture was filtered by centrifugation at 20003 g for 30 min, at 4°C, through an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. In table 3, the production yields of 2,6-Diaminopurine nucleosides prepared in the presence of co-solvents is shown. The resulting 2,6-Diaminopurine nucleosides (2,6-Diaminopurine riboside or 2,6-Diaminopurine deoxyriboside) were analyzed by HPLC. In these reactions, the products were obtained in high yields (over 80%). 45 Table 3. Production yields of 2,6-Diaminopurine nucleosides using different % co-solvent at 60°C. THF: Tetrahydrofuran; DMSO: dimethylsulfoxide Co-solvent Substrate Analog base Product Yield (%)

50 10% DMSO Uridine 2,6-Diaminopurine 2,6-Diaminopurine riboside 97 10% DMSO 2’-Deoxyuridine 2,6-Diaminopurine 2,6-Diaminopurine 96 deoxyriboside 5% THF Uridine 2,6-Diaminopurine 2,6-Diaminopurine riboside 84 55 5% THF 2’-Deoxyuridine 2,6-Diaminopurine 2,6-Diaminopurine 90 deoxyriboside

12 EP 2 516 637 B1

12.2. Preparation of 2,6-Diaminopurine nucleosides without the use of co-solvents

[0085] The preparation of 2,6-Diaminopurine nucleosides without co-solvents could only be carried out at temperatures above 80°C due to limited water solubility of this purinic base. Assays were performed at different temperatures (80, 90 5 and 100°C). Twelve units/ml cell lysate were added to 150 ml of a solution kept thermostatically at the selected temper- ature, and having the following composition of substrate solutions:

1. 15 mM Uridine/2’-Deoxyuridine, 2. 5 mM 2,6-Diaminopurine, and 10 3. 30 mM potassium phosphate buffer, pH 7.

[0086] After 5 hours, the reaction mixture was filtered by centrifugation at 20003 g for 30 min, at 4°C, through an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. In table 3, the production yields of 2,6-Diaminopurine nucleosides prepared in the presence of co-solvents is shown. 15 The resulting 2,6-Diaminopurine nucleosides (2,6-Diaminopurine riboside or 2,6-Diaminopurine deoxyriboside) were analyzed by HPLC. In table 4, the production yields of 2,6-Diaminopurine nucleosides prepared without organic solvents are shown. The bioconversion yield of the reaction was around 80% in all temperatures assayed.

Table 4. Production yields of 2,6-Diaminopurine nucleosides at different temperatures without use organic co-solvents. 20 Temperature (°C) 80 90 100 Biotransformation (%) 79.3 79.6 81.1

Example 13 25 Preparation of other nucleosides

[0087] Transglycosylation reactions were carried out using various acceptor bases. The reactions were carried out at 80°C for various periods of time with 10% of DMSO as co-solvent for purinic base and without co-solvent use for 30 pyrimidinic aceptor bases. The percentage of bioconversion was calculated relative to the initial concentration of acceptor base and was determined by HPLC analysis of the reaction mixture.

13.1 Preparation of Trifluoromethyluracil nucleosides

35 [0088] One ml catalyst (cell lysate) with transglycosylation activity of about 12 units/ml cell lysate were added to 150 ml of a solution kept thermostatically between 80°C, and having the following composition:

- 7.5mM uridine/2’-Deoxyuridine, - 2.5mM Trifluoromethyluracil, and 40 - 20 mM potassium phosphate buffer, pH 7.

[0089] After 3 hour at 80 °C, the reaction mixture was filtered by centrifugation at 2000 3g for 30 min, at 4°C, through an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. The bioconversion yield of the reaction was higher than 60%. The resulting trifluoromethyluracil nucleosides (trifluor- 45 omethyluridine or 2’-deoxytrifluoromethyluridine) were analyzed by HPLC.

13.2 Preparation of 5-Fluoruracil nucleosides

[0090] 5-Fluoruracil nucleosides were prepared as previously described above. One ml catalyst (cell lysate) with 50 transglycosylation activity of about 12 units/ml cell lysate was added to 150 ml of a solution kept thermostatically at 80 °C and having the following composition:

- 7.5mM uridine/2’-Deoxyuridine, - 2.5mM 5-fluorouracil, and 55 - 30 mM potassium phosphate buffer, pH 7.

[0091] After 3 hour at 80 °C, the reaction mixture was filtered by centrifugation at 2000 3g for 30 min, at 4°C, through

13 EP 2 516 637 B1

an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. The bioconversion yield of the reaction was higher than 50%. The resulting 5-fluoruracil nucleosides (5-fluorouridine and 5-fluoro-2’-deoxyuridine) were analyzed by HPLC.

5 13.3 Preparation of trans-Zeatin nucleosides

[0092] trans-Zeatin nucleosides were prepared as described above. One ml catalyst (cell lysate) with transglycosylation activity of about 12 units/ml cell lysate was added to 150 ml of a solution kept thermostatically at 80 °C and having the following composition: 10 - 7.5mM Uridine/2’-Deoxyuridine, - 2.5mM trans-Zeatin, and - 30 mM potassium phosphate buffer, pH 7.

15 [0093] The assays were performed in 10% (v/v) DMSO as co-solvent using the conditions described above. After 5 hours at 80°C, the reaction mixture was filtered by centrifugation at 2000 3g for 30 min, at 4°C, through an Amicon ultra- 4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. The biocon- version yield of the reaction was higher than 80%. The resultingtrans- zeatin nucleosides (trans-zeatin riboside and trans-zeatin deoxyriboside) were analyzed by HPLC. 20 13.4 Preparation process for 2-Chloro-6-methylaminopurine nucleosides

[0094] 2-Chloro-6-methylaminopurine nucleosides were prepared as described before. One ml catalyst (cell lysate) with transglycosylation activity of about 12 units/ml cell lysate were added to 150 ml of a solution kept thermostatically 25 at 80 °C and having the following composition:

- 15mM Uridine/2’-Deoxyuridine, - 5mM 2-Chloro-6-Methylaminopurine, and - 30 mM potassium phosphate buffer, pH 7. 30 [0095] The assays were performed in 10% (v/v) DMSO as co-solvent using the conditions described above. After 5 hours at 80 °C, the reaction mixture was filtered by centrifugation at 20003 g for 30 min, at 4°C, through an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. The bioconversion yield of the reaction was higher than 80%. The resulting 2-chloro-6-methylaminopurine nucleosides (2- 35 chloro-6-methylaminopurine riboside and 2-chloro-6-methylaminopurine deoxyriboside) were analyzed by HPLC.

13.5 Preparation of 6-Dimethylaminopurine nucleosides

[0096] 6-Dimethylaminopurine nucleosides were prepared as described above. One ml catalyst (cell lysate) with trans- 40 glycosylation activity of about 12 units/ml cell lysate was added to 150 ml of a solution kept thermostatically at 80 °C and having the following composition:

- 15mM Uridine/2’-Deoxyuridine, - 5mM 6-Dimethylaminopurine, and 45 - 30 mM potassium phosphate buffer, pH 7.

[0097] The assays were performed in 10% (v/v) DMSO as co-solvent using the conditions described above. After 5 hours at 80 °C, the reaction mixture was filtered by centrifugation at 20003 g for 30 min, at 4°C, through an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. The 50 bioconversion yield of the reaction was higher than 80%. The resulting 6-dimethylaminopurine nucleosides (6-dimeth- ylaminopurine riboside and 6-dimethylaminopurine deoxyriboside) were analyzed by HPLC.

13.6 Preparation of 6-Mercaptopurine nucleosides

55 [0098] 6-Mercaptopurine nucleosides were prepared as described above. One ml catalyst (cell lysate) with transgly- cosylation activity of about 12 units/ml cell lysate was added to 150 ml of a solution kept thermostatically at 80 °C and having the following composition:

14 EP 2 516 637 B1

- 15mM Uridine/2’-Deoxyuridine, - 5mM 6-Mercaptopurine, and - 30 mM potassium phosphate buffer, pH 7.

5 [0099] The assays were performed in 10% (v/v) of DMSO as co-solvent using the conditions described above. After 5 hours at 80°C, the reaction mixture was filtered by centrifugation at 2000 3g for 30 min, at 4°C, through an Amicon ultra-4 Centrifugal Filter Devices (Millipore, Bedford, MA) with a 3000-Da cut-off, and the filtrate was recovered. The bioconversion yield of the reaction was higher than 50%. The resulting 6-mercaptopurine nucleosides (6-mercaptopurine riboside and 6-mercaptopurine deoxyriboside) were analyzed by HPLC. 10 References

[0100]

15 1. Van Rompay A, Johansson M, & Karlsson A (2003) Substrate specificity and phosphorylation of antiviral and anticancer nucleoside analogues by human deoxyribonucleoside kinases and ribonucleoside kinases. (Translated from eng) Pharmacol Ther 100(2):119-139 (in eng). 2. Yu X, Li G, Qi X, & Deng Y (2005) Stereoselective synthesis of 9-beta-d-arabianofuranosyl guanine and 2-amino- 9-(beta-d-arabianofuranosyl)purine. (Translated from eng) Bioorg Med Chem Lett 15(3):683-685 (in eng). 20 3. Hutchinson, Trends Biotechnol. 8, 348-353, 1990 4. EP1141328, Keryos Spa 5. Taran SA, Verevkina KN, Esikova TZ, Feofanov SA, & Miroshnikov AI (2008) [Synthesis of 2-chloro-2’-deoxya- denosine by microbiological transglycosylation using a recombinant Escherichia coli strain]. (Translated from rus) Prikl Biokhim Mikrobiol 44(2):181-186 (in rus). 25 6. Cacciapuoti G, et al. (2009) Purine nucleoside phosphorylases from hyperthermophilic Archaea require a CXC motif for stability and folding. FEBS J 276(20):5799-5805 7. Cacciapuoti G, et al. (2005) A novel hyperthermostable 5’-deoxy-5’-methylthioadenosine phosphorylase from the archaeon Sulfolobus solfataricus. (Translated from eng) FEBS J 272(8):1886-1899 (in eng). 8. Cacciapuoti G, et al. (2007) Biochemical and structural characterization of mammalian-like purine nucleoside 30 phosphorylase from the Archaeon Pyrococcus furiosus. (Translated from eng) FEBS J 274(10):2482-2495 (in eng). 9. Brown, S.H. and Kelly, R.M. (1993) Characterization of Amylolytic Enzymes, Having Both alpha-1,4 and alpha- 1,6 Hydrolytic Activity, from the Thermophilic Archaea Pyrococcus furiosus and Thermococcus litoralis. Appl Environ Microbiol 59, 2614-2621. 10. Fujiwara, S., Lee, S.G., Haruki, M., Kanaya, S., Takagi, M. and Imanaka, T. (1996) Unusual enzyme character- 35 istics of aspartyl-tRNA synthetase from hyperthermophilic archaeon Pyrococcus sp. KOD1. FEBS Lett 394, 66-70. 11.Purcarea, C., Simon, V., Prieur, D.and Hervé, G. (1996) Purificationand characterization of carbamoyl-phosphate synthetase from the deep-sea hyperthermophilic archaebacterium Pyrococcus abyssi. Eur J Biochem 236, 189-199. 12. Cacciapuoti, G., Fusco, S., Caiazzo, N., Zappia, V. and Porcelli, M. (1999) Heterologous expression of 5’- methylthioadenosine phosphorylase from the archaeon Sulfolobus solfataricus: characterization of the recombinant 40 protein and involvement of disulfide bonds in thermophilicity and thermostability. Protein Expr Purif 16, 125-135. 13. Bzowska A, Kulikowska E, & Shugar D (2000) Purine nucleoside phosphorylases: properties, functions, and clinical aspects. (Translated from eng) Pharmacol Ther 88(3):349-425 (in eng). 14. Saunders, P.P., Wilson, B.A. and Saunders, G.F. (1969) Purification and comparative properties of a pyrimidine nucleoside phosphorylase from Bacillus stearothermophilus. J Biol Chem 244, 3691-3697. 45 15. She Q, et al. (2001) The complete genome of the crenarchaeon Sulfolobus solfataricus P2. (Translated from eng) Proc Natl Acad Sci U S A 98(14):7835-7840 (in eng). 16. Kawarabayasi Y, et al. (1999) Complete genome sequence of an aerobic hyperthermophilic crenarchaeon, Aeropyrum pernix K1. (Translated from eng) DNA Res 6(2):83-101, 145-152 (in eng). 17. Yanisch-Perron C, et al. (1985) Improved M13 phage cloning vectors and host strains: nucleotide sequences 50 of the M13mp18 and pUC19 vectors. Gene. 33(1):103-19.

SEQUENCE LISTING

[0101] 55 <110> INSTITUT UNIV. DE CIENCIA I TECNOLOGIA, S.A.

<120> THERMOSTABLE BIOCATALYST COMBINATION FOR NUCLEOSIDE SYNTHESIS

15 EP 2 516 637 B1

<130> P25190EP00

<140> <141> 5 <150> EP093822963 <151> 2009-12-22 <160> 8

10 <170> PatentIn version 3.3

<210> 1 <211> 28 <212> DNA 15 <213> artificial

<220> <223> primer related to sulfolobus solfataricus deoD forward

20 <400> 1 caccgtgcca tttttagaaa atggttcc 28

30 <210> 2 <211> 27 25 <212> DNA <213> artificial

<220> <223> primer related to sulfolobus solfataricus deoD reverse 30 <400> 2 aatcagtttt aagaatctta aggtaat 27

<210> 3 35 <211> 25 <212> DNA <213> artificial

<220> 40 <223> primer related to Aeropyrum pernix udp forward

<400> 3 caccgtggcc cgctacgttc tcctc 25

45 <210> 4 <211> 28 <212> DNA <213> artificial

50 <220> <223> primer related to Aeropyrum pernix udp reverse

<400> 4 gaattcctat gtgcgtctgc acgccagg 28 55 <210> 5 <211> 18 <212> DNA

16 EP 2 516 637 B1

<213> artificial

<220> <223> primer related to TrxFus forward 5 <400> 5 ttcctcgacg ctaacctg 18

<210> 6 10 <211> 20 <212> DNA <213> Artificial

<220> 15 <223> primer related to T7 reverse

<400> 6 tagttattgc tcaggggtgg 20

20 <210> 7 <211> 825 <212> DNA <213> Sulfolobus solfataricus

25 <400> 7

30

35

40

45

<210> 8 <211> 765 50 <212> DNA <213> Aeropyrum pernix

<400> 8

55

17 EP 2 516 637 B1

5

10

15

20

Claims

25 1. A recombinant expression vector comprising: the sequence encoding a purine nucleoside phosphorylase (PNPase, E.C. 2.4.2.1) with the sequence encoding a uridine phosphorylase (UPase, E.C. 2.4.2.3); each of the sequences operably linked to one or more control sequences that direct the production of said phosphorylases in a suitable expressionhost; said sequences originating fromthe Archaea Thermoprotei class, characterized in that the PNPase is from Sulfolobus solfataricus (SEQ ID NO. 7) and the UPase is from Aeropyrum pernix (SEQ ID NO. 8) and in 30 that said suitable expression host is the BL21 bacterial strain of E. coli; and further in that said vector comprises a T7 promoter that permits the binding of T7 RNA polymerase, a lac operator that permits inhibition of expression when isopropylthiogalactopyranoside (IPTG) is not present and a His-patch-thioredoxin.

2. The E. coli host cell comprising the recombinant expression vector according to claim 1. 35 3. A lysate obtained by lysing the host cell according to claim 2.

4. A transglycosylation method between a nucleobase bound to a ribose or deoxyribose sugar via a beta-glycosidic linkage and a nucleobase or modified nucleobase in the presence of phosphate ions, characterised in that said 40 method comprises the use of a purine nucleoside phosphorylase (PNPase) of Sulfolobus solfataricus (SEQ ID No. 7) with a uridine phosphorylase (UPase) of Aeropyrum pernix (SEQ ID No. 8), characterised in that said phospho- rylases are produced by the recombinant expression vector according to claim 1.

5. The transglycosylation method according to claim 4, wherein the UPase ofAeropyrum pernix and the PNPase of 45 Sulfolobus solfataricus are provided by a host cell according to any one of claims 2-3.

6. The transglycosylation method according to any one of claims 4-5, wherein the obtained products are active phar- maceutical ingredients (API) or intermediates thereof.

50 7. The transglycosylation method according to any one of claims 4-6, wherein the nucleobase bound to a ribose or deoxyribose sugar via a beta-glycosidic linkage is selected from natural or modified nucleosides containing D-ribose or 2’-deoxyribose; nucleosides containing the ribose group modified in the 2’, 3’, and/or 5’ positions; and nucleosides in which the sugar is beta-D-arabinose, alpha-L-xylose, 3’-deoxyribose, 3’,5’-dideoxyribose, 2’,3’-dideoxyribose, 5’- deoxyribose, 2’,5’-dideoxyribose, 2’-amino-2’-deoxyribose, 3’-amino-3’-deoxyribose, or 2’-fluoro-2’-deoxyubose. 55 8. The transglycosylation method according to any one of claims 4-7, wherein the nucleobase or modified nucleobase is selected from natural or substituted pyrimidine and purine bases; purine bases substituted at one or more of the 1, 2, 6 positions; pyrimidine bases substituted at one or more of the 3, 5 positions; and purine, 2-azapurine, 8-

18 EP 2 516 637 B1

azapurine, 1-deazapurine (imidazopyridine), 3-deazapurine, 7-deazapurine, 2,6-diaminopurine, 5-fluorouracil, 5- trifluoromethyluracil, trans-zeatin, 2-chloro-6-methylaminopurine, 6-dimethylaminopurine, 6-mercaptopurine.

9. The transglycosylation method according to any one of claims 4-8, wherein said method is carried out between 60 5 and 100 °C.

10. The transglycosylation method according to any one of claims 4-9, wherein said method is carried out in aqueous media, or in an aprotic polar co-solvent system.

10 11. The transglycosylation method according to claim 10, wherein the aprotic polar co-solvent system is selected from dimethylsulfoxide, tetrahydrofuran, 2-methyltetrahydrofuran, and dimethylformamide.

12. The transglycosylation method accordingto anyone of claims4-11,wherein thesugar-donating nucleoside is selected from uridine and 2’-deoxyuridine, and the acceptor base is selected from 2,6-diaminopurine, 5-fluorouracil, 5-trif- 15 luoromethyluracil, trans-zeatin, 2-chloro-6-methylaminopurine, 6-dimethylaminopurine, and 6-mercaptopurine.

Patentansprüche

20 1. Rekombinater Expressionvektor, umfassend: die eine Purin-Nukleosid-Phosphorylase (PNPase, E.C. 2.4.2.1) ko- dierende Sequenz mit der Sequenz, die eine Uridin-Phosphorylase (UPase, E.C. 2.4.2.3) kodiert; wobei jede der Sequenzen mit ein oder mehreren Kontrollsequenzen in funktionaler Weise verbunden ist, welche die Produktion der Phosphorylasen in einem geeigneten Expressionswirt steuern; die Sequenzen aus der Klasse der Archaea Thermoprotei stammen, dadurch gekennzeichnet, dass die PNPase aus Sulfolobus solfactorius (SEQ ID NO. 7) 25 und die UPase aus Aeropyrum pernix (SEQ ID NO. 8) stammt und dadurch gekennzeichnet, dass der geeignete Expressionswirt der BL21-Bakterienstamm von E. coli ist; und weiterhin dadurch gekennzeichnet, dass der Vektor einen T7-Promotor umfasst, der die Bindung der T7 RNA-Polymerase ermöglicht, einen lac-Operator, der die Inhi- bierung der Expression ermöglicht, wenn Isopropylthiogalactopyranosid (IPTG) nicht vorliegt und ein His-patch- thioredoxin. 30 2. E.-coli-Wirtszelle, umfassend den rekombinanten Expressionsvektor nach Anspruch 1.

3. Lysat, erhalten durch Lysieren der Wirtszelle nach Anspruch 2.

35 4. Transglykosylierungsverfahren zwischen einer Nukleobase, die an einen Ribose- oder Desoxyribosezucker über eine Beta-glykosidische Bindung gebunden ist, und einer Nukleobase oder einer modifizierte Nukleobase in Ge- genwart von Phosphationen, dadurch gekennzeichnet, dass das Verfahren die Verwendung einer Purin-Nukle- osid-Phosphorylase (PNPase) aus Sulfolobus solfactorius (SEQ ID NO. 7) mit einer Uridin-Phosphorylase (UPase) aus Aeropyrum pernix (SEQ ID NO. 8) umfasst, dadurch gekennzeichnet, dass die Phosphorylasen durch den 40 rekombinanten Expressionsvektor nach Anspruch 1 produziert werden.

5. Transglykosylierungsverfahren nach Anspruch 4, wobei die UPase vonAeropyrum pernix und die PNPase von Sulfolobus solfactorius durch eine Wirtszelle nach einem der Ansprüche 2-3 bereitgestellt werden.

45 6. Transglykosylierungsverfahren nach einem der Ansprüche 4-5, wobei die erhaltenen Produkte aktive pharmazeu- tische Inhaltsstoffe (API) oder Zwischenprodukte hiervon sind.

7. Transglykosylierungsverfahrennach einemder Ansprüche4-6, wobei die aneinen Ribose- oder Desoxyribosezucker über eine Beta-glykosidische Bindung gebundene Nukleobase ausgewählt wird aus natürlichen oder modifizierten 50 Nukleosiden, die D-Ribose oder 2-Desoxyribose enthalten; Nukleoside, welche die Ribosegruppe, modifiziert in den 2’-, 3’-, und/oder 5’-Positionen enthalten; und Nukleoside, in welchem der Zucker eine Beta-D-Arabinose, Alpha- L-Xylose, 3’-Desoxyribose, 3’,5’-Didesoxyribose, 2’,3’-Didesoxyribose, 5’-Desoxyribose, 2’,5’-Didesoxyribose, 2’- Amino-2’-desoxyribose, 3’-Amino-3’-desoxyribose, or 2’-Fluoro-2’-desoxyribose ist.

55 8. Transglykosylierungsverfahren nach einem der Ansprüche 4-7, wobei die Nukleobase oder die modifizierte Nukle- obase ausgewählt wird aus natürlichen oder substituierten Pyrimidin- und Purinbasen; Purinbasen, die an einem odermehreren der1-, 2-, 6-Positionen substituiert sind;Pyrimidinbasen, die an einoder mehreren der 3-, 5-Positionen substituiert sind; und Purin, 2-Azapurin, 8-Azapurin, 1-Deazapurin (Imidazopyridin), 3-Deazapurin, 7-Deazapurin,

19 EP 2 516 637 B1

2,6-Diaminopurin, 5-Fluoruracil, 5-Trifluormethyluracil, trans-Zeatin, 2-Chlor-6-methylaminopurin, 6-Diemethylami- nopurin, 6-Mercaptopurin.

9. Transglykosylierungsverfahren nach einem der Ansprüche 4-8, wobei das Verfahren zwischen 60 und 100 °C 5 ausgeführt wird.

10. Transglykosylierungsverfahren nach einem der Ansprüche 4-9, wobei das Verfahren in wässrigen Medien oder in einem aprotischen polaren Co-Lösungsmittelsystem durchgeführt wird.

10 11. Transglykosylierungsverfahren nach Anspruch 10, wobei das aprotische polare Co-Lösungsmittel ausgewählt wird aus Dimethylsulfoxid, Tetrahydrofuran, 2-Methyltetrahydrofuran, und Dimethylformamid.

12. Transglykosylierungsverfahren nach einem der Ansprüche 4-11, wobei das Zucker abgebende Nukleosid ausge- wählt wird aus Uridin und 2’-Desoxyuridin, und die Akzeptorbase ausgewählt wird aus 2,6-Diaminopurin, 5-Fluoru- 15 racil, 5-Trifluormethyluracil, trans-Zeatin,2-Chlor-6-methylaminopurin, 6-Dimethylaminopurin, und 6-Mercaptopurin.

Revendications

20 1. Vecteur d’expression recombinant comprenant : la séquence codant pour une purine nucléoside phosphorylase (PNPase, E.C. 2.4.2.1) avec la séquence codant pour une uridine phosphorylase (UPase, E.C. 2.4.2.3) ; chacune des séquences étant en liaison fonctionnelle avec une ou plusieurs séquences de contrôle qui dirigent la production desdites phosphorylases dans un hôte d’expression approprié ; lesdites séquences provenant de la classe Archaea Thermoprotei, caractérisé en ce que le la PNPase est issue de Sulfolobus solfataricus (SEQ ID NO: 7) et l’UPase 25 est issue d’Aeropyrum pernix (SEQ ID NO: 8) et en ce que ledit hôte d’expression approprié est la souche bactérienne BL21 d’E. coli ; et en outreen ce que ledit vecteur comprend un promoteur T7 qui permet la liaison de l’ARN polymérase T7,un opérateur lacqui permet une inhibition de l’expression lorsque de l’isopropylthiogalactopyranoside (IPTG) n’est pas présent et une thiorédoxine-marqueur His.

30 2. Cellule hôte d’E. coli comprenant le vecteur d’expression recombinant selon la revendication 1.

3. Lysat obtenu par la lyse de la cellule hôte selon la revendication 2.

4. Procédé de transglycosylation entre une nucléobase liée à un sucre ribose ou désoxyribose via une liaison bêta- 35 glycosidique et une nucléobase ou une nucléobase modifiée en présence d’ions phosphate, caractérisé en ce que ledit procédé comprend l’utilisation d’une purine nucléoside phosphorylase (PNPase) deSulfolobus solfataricus (SEQ ID NO: 7) avec une uridine phosphorylase (UPase) d’Aeropyrum pernix (SEQ ID NO: 8), caractérisé en ce que lesdites phosphorylases sont produites par le vecteur d’expression recombinant selon la revendication 1.

40 5. Procédé de transglycosylation selon la revendication 4, dans lequel l’UPase d’Aeropyrum pernix et la PNPase de Sulfolobus solfataricus sont fournies par une cellule hôte selon l’une quelconque des revendications 2 à 3.

6. Procédé de transglycosylation selon l’une quelconque des revendications 4 à 5, dans lequel les produits obtenus sont des principes actifs pharmaceutiques (API) ou des intermédiaires de ceux-ci. 45 7. Procédé de transglycosylation selon l’une quelconque des revendications 4 à 6, dans lequel la nucléobase liée à un sucre ribose ou désoxyribose via une liaison bêta-glycosidique est sélectionnée parmi des nucléosides naturels ou modifiés contenant du D-ribose ou du 2’-désoxyribose ; des nucléosides contenant le groupe ribose modifié aux positions 2’, 3’, et/ou 5’ ; et des nucléosides dans lesquels le sucre est le bêta-D-arabinose, l’alpha-L-xylose, le 3’- 50 désoxyribose, le 3’,5’-désoxyribose, le 2’,3’-didésoxyribose, le 5’-désoxyribose, le 2’,5’-didésoxyribose, le 2’-amino- 2’-désoxyribose, le 3’-amino-3’-désoxyribose, ou le 2’-fluoro-2’-désoxyribose.

8. Procédé de transglycosylation selon l’une quelconque des revendications 4 à 7, dans lequel la nucléobase ou la nucléobase modifiée est sélectionnée parmi des bases pyrimidine et purine naturelles ou substituées ; des bases 55 purine substituées à une ou plusieurs des positions 1, 2, 6 ; des bases pyrimidine substituées à une ou plusieurs des positions 3, 5 ; et la purine, la 2-azapurine, la 8-azapurine, la 1-déazapurine (imidazopyridine), la 3-déazapurine, la 7-dézapurine, la 2,6-diaminopurine, le 5-fluorouracile, le 5-trifluorométhyluracile, la trans-zéatine, la 2-chloro-6- méthylaminopurine, la 6-diméthylaminopurine, la 6-mercaptopurine.

20 EP 2 516 637 B1

9. Procédé de transglycosylation selon l’une quelconque des revendications 4 à 8, où ledit procédé est réalisé entre 60 et 100 °C.

10. Procédé de transglycosylation selon l’une quelconque des revendications 4 à 9, où ledit procédé est réalisé dans 5 un milieu aqueux, ou dans un système de co-solvant polaire aprotique.

11. Procédé de transglycosylation selon la revendication 10, dans lequel le système de co-solvant polaire aprotique est sélectionné parmi le diméthylsulfoxyde, le tétrahydrofurane, le 2-méthyltétrahydrofurane, et le diméthylforma- mide. 10 12. Procédé de transglycosylation selon l’une quelconque des revendications 4 à 11, où le nucléoside donneur de sucre est sélectionné parmi l’uridine et la 2’-désoxyuridine, et la base accepteur est sélectionnée parmi la 2,6-diamino- purine, le 5-fluorouracile, le 5-trifluorométhyluracile, la trans-zéatine, la 2-chloro-6-méthylaminopurine, la 6-dimé- thylaminopurine, et la 6-mercaptopurine. 15

20

25

30

35

40

45

50

55

21 EP 2 516 637 B1

22 EP 2 516 637 B1

23 EP 2 516 637 B1

24 EP 2 516 637 B1

25 EP 2 516 637 B1

26 EP 2 516 637 B1

27 EP 2 516 637 B1

REFERENCES CITED IN THE DESCRIPTION

This list of references cited by the applicant is for the reader’s convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description

• EP 1141328 A [0100] • EP 093822963 A [0101]

Non-patent literature cited in the description

• TARAN S A et al. Enzymatic transglycosylation of • SHE QUINXIN et al. The complete genome of the natural and modified nucleosides by immobilized crenarchaeon Sulfolobus solfataricus P2. Proceed- thermostable nucleoside phosphorylases from Geo- ings of the National Academy of Sciences of the Unit- bacillus stearothermophilus. Russian Journal of ed States of America, 03 July 2001, vol. 98 (14), Bioorganic Chemistry, 06 November 2009, vol. 35 7835-7840 [0024] (6), 739-745 [0016] • VAN ROMPAY A ; JOHANSSON M ; KARLSSON • Screening of Catalytically active microorganisms for A. Substrate specificity and phosphorylation of anti- the synthesis of 6-modified purine nucleosides. viral and anticancer nucleoside analogues by human TRELLES J A et al. Biotechnology Letters. Kluwer deoxyribonucleoside kinases and ribonucleoside ki- Academic Publishers, 01 June 2005, vol. 27, 759-763 nases. (Translated from eng). Pharmacol Ther, 2003, [0017] vol. 100 (2), 119-139 [0100] • Completegenome sequence of an aerobic hyperther- •YUX; LI G ; QI X ; DENG Y. Stereoselective syn- mophilic crenarchaeon, Aeropyrum pernix K1.KA- thesis of 9-beta-d-arabianofuranosyl guanine and WARABAYASHI YUTAKA et al. DNA Research. 2-amino-9-(beta-d-arabianofuranosyl)purine. Bioorg Universal Academy Press, 01 January 1999, vol. 6, Med Chem Lett, 2005, vol. 15 (3), 683-685 [0100] 83-101 [0018] • HUTCHINSON. Trends Biotechnol., 1990, vol. 8, • HAMAMOTO TOMOKI et al. Cloning of purine nucl- 348-353 [0100] eoside phosphorylase II gene from Bacillus stearo- • TARAN SA ; VEREVKINA KN ; ESIKOVA TZ ; FE- thermophilus TH 6-2 and characterization of its gene OFANOV SA ; MIROSHNIKOV AI. Synthesis of product. Bioscience Biotechnology and Biochemis- 2-chloro-2’-deoxyadenosine by microbiological try, 1997, vol. 61 (2), 276-280 [0019] transglycosylation using a recombinant Escherichia • HAMAMOTO TOMOKI et al. Cloning and expression colistrain]. (Translated from rus). PriklBiokhim Mikro- of purine nucleoside phosphorylase I gene from Ba- biol, 2008, vol. 44 (2), 181-186 [0100] cillus stearothermophilus TH 6-2.Bioscience Bio- • CACCIAPUOTI G et al. Purine nucleoside phospho- technology and Biochemistry, 1997, vol. 61 (2), rylases from hyperthermophilic Archaea require a 272-275 [0020] CXC motif for stability and folding. FEBS J, 2009, vol. • CACCIAPUOTI GIOVANNA et al. Purine nucleoside 276 (20), 5799-5805 [0100] phosphorylases from hyperthermophilic Archaea re- • CACCIAPUOTI G et al. A novel hyperthermostable quire a CXC motif for stability and folding. The Febs 5’-deoxy-5’-methylthioadenosine phosphorylase Journal Oct 2009 LNKDS-PUBMED: 19740110, Oc- from the archaeon Sulfolobus solfataricus. FEBS J, tober 2009, vol. 276 (20), 5799-5805 [0021] 2005, vol. 272 (8), 1886-1899 [0100] • CACCIAPUOTI GIOVANNA et al. A novel hyperther- • CACCIAPUOTI G et al. Biochemical and structural mostable 5’-deoxy-5’-methylthioadenosine phos- characterization of mammalian-like purine nucleo- phorylase from the archaeon Sulfolobus solfataricus. side phosphorylase from the Archaeon Pyrococcus Febs Journal, April 2005, vol. 272 (8), 1886-1899 furiosus. FEBS J, 2007, vol. 274 (10), 2482-2495 [0022] [0100] • CACCIAPUOTI GIOVANNA et al. Biochemical and • BROWN, S.H. ; KELLY, R.M. Characterization of structural characterization of mammalian-like purine Amylolytic Enzymes, Having Both alpha-1,4 and al- nucleoside phosphorylase from the Archaeon Pyro- pha-1,6 Hydrolytic Activity, from the Thermophilic Ar- coccus furiosus. Febs Journal, May 2007, vol. 274 chaea Pyrococcus furiosus and Thermococcus lito- (10), 2482-2495 [0023] ralis. Appl Environ Microbiol, 1993, vol. 59, 2614-2621 [0100]

28 EP 2 516 637 B1

• FUJIWARA, S. ; LEE, S.G.; HARUKI, M.; KA- • BZOWSKA A ; KULIKOWSKA E ; SHUGAR D. Pu- NAYA, S. ; TAKAGI, M. ; IMANAKA, T. Unusual en- rine nucleoside phosphorylases: properties, func- zyme characteristics of aspartyl-tRNA synthetase tions, and clinical aspects. Pharmacol Ther, 2000, from hyperthermophilic archaeon Pyrococcus sp. vol. 88 (3), 349-425 [0100] KOD1. FEBS Lett, 1996, vol. 394, 66-70 [0100] • SAUNDERS, P.P. ; WILSON, B.A. ; SAUNDERS, • PURCAREA,C. ; SIMON, V. ; PRIEUR, D. ; HERVÉ, G.F. Purification and comparative properties of a py- G. Purification and characterization of car- rimidine nucleoside phosphorylase from Bacillus bamoyl-phosphate synthetase from the deep-sea stearothermophilus. J Biol Chem, 1969, vol. 244, hyperthermophilic archaebacterium Pyrococcus ab- 3691-3697 [0100] yssi. Eur J Biochem, 1996, vol. 236, 189-199 [0100] •SHEal.etQ The complete genome of the crenar- • CACCIAPUOTI, G. ; FUSCO, S. ; CAIAZZO, N. ; chaeon Sulfolobus solfataricus P2. Proc Natl Acad ZAPPIA, V. ; PORCELLI, M. Heterologous expres- Sci U S A, 2001, vol. 98 (14), 7835-7840 [0100] sion of 5’-methylthioadenosine phosphorylase from • KAWARABAYASI Y et al. Complete genome se- the archaeon Sulfolobus solfataricus: characteriza- quence of an aerobic hyperthermophilic crenarchae- tion of the recombinant protein and involvement of on, Aeropyrum pernix K1. DNA Res, 1999, vol. 6 (2), disulfide bonds in thermophilicity and thermostability. 83-101, 145-152 [0100] Protein Expr Purif, 1999, vol. 16, 125-135 [0100] • YANISCH-PERRON C et al. Improved M13 phage cloning vectors and host strains: nucleotide sequenc- es of the M13mp18 and pUC19 vectors. Gene, 1985, vol. 33 (1), 103-19 [0100]

29