US20160376570A1 - O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof - Google Patents

O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof Download PDF

Info

Publication number
US20160376570A1
US20160376570A1 US14/902,492 US201414902492A US2016376570A1 US 20160376570 A1 US20160376570 A1 US 20160376570A1 US 201414902492 A US201414902492 A US 201414902492A US 2016376570 A1 US2016376570 A1 US 2016376570A1
Authority
US
United States
Prior art keywords
gene
cell
filamentous fungal
protein
mutation
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/902,492
Other languages
English (en)
Inventor
Jari Natunen
Jukka Hiltunen
Anne Huuskonen
Markku Saloheimo
Christian Ostermeier
Benjamin Patrick Sommer
Ramon Wahl
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Glykos Finland Ltd
Original Assignee
Glykos Finland Ltd
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=48740957&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20160376570(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Glykos Finland Ltd filed Critical Glykos Finland Ltd
Publication of US20160376570A1 publication Critical patent/US20160376570A1/en
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GLYKOS FINLAND OY, NOVARTIS AG
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HILTUNEN, JUKKA, NATUNEN, JARI
Assigned to TEKNOLOGIAN TUTKIMUSKESKUS VTT reassignment TEKNOLOGIAN TUTKIMUSKESKUS VTT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HUUSKONEN, ANNE, SALOHEIMO, MARKKU
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TEKNOLOGIAN TUTKIMUSKESKUS VTT
Assigned to NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD., GLYKOS FINLAND OY reassignment NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GLYKOS FINLAND OY
Assigned to NOVARTIS PHARMA AG reassignment NOVARTIS PHARMA AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OSTERMEIER, CHRISTIAN, SOMMER, Benjamin Patrick, WAHL, Ramon
Assigned to GLYKOS FINLAND OY, NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD. reassignment GLYKOS FINLAND OY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS PHARMA AG
Assigned to NOVARTIS AG reassignment NOVARTIS AG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD.
Assigned to GLYKOS FINLAND OY, NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD. reassignment GLYKOS FINLAND OY NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS PHARMA AG
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: HILTUNEN, JUKKA, NATUNEN, JARI
Assigned to TEKNOLOGIAN TUTKIMUSKESKUS VTT reassignment TEKNOLOGIAN TUTKIMUSKESKUS VTT NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: HUUSKONEN, ANNE, SALOHEIMO, MARKKU
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: TEKNOLOGIAN TUTKIMUSKESKUS VTT
Assigned to NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD., GLYKOS FINLAND OY reassignment NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD. NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: GLYKOS FINLAND OY
Assigned to NOVARTIS PHARMA AG reassignment NOVARTIS PHARMA AG NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: OSTERMEIER, CHRISTIAN, SOMMER, Benjamin Patrick, WAHL, Ramon
Assigned to NOVARTIS AG reassignment NOVARTIS AG NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD.
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY NUNC PRO TUNC ASSIGNMENT (SEE DOCUMENT FOR DETAILS). Assignors: GLYKOS FINLAND OY, NOVARTIS AG
Assigned to GLYKOS FINLAND OY reassignment GLYKOS FINLAND OY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GLYKOS FINLAND OY, NOVARTIS AG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1048Glycosyltransferases (2.4)
    • C12N9/1051Hexosyltransferases (2.4.1)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/37Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from fungi
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y204/00Glycosyltransferases (2.4)
    • C12Y204/01Hexosyltransferases (2.4.1)
    • C12Y204/01109Dolichyl-phosphate-mannose-protein mannosyltransferase (2.4.1.109)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation

Definitions

  • the present disclosure relates to compositions and methods useful for the production of heterologous proteins in filamentous fungal cells.
  • Posttranslational modification of eukaryotic proteins, particularly therapeutic proteins such as immunoglobulins, is often necessary for proper protein folding and function. Because standard prokaryotic expression systems lack the proper machinery necessary for such modifications, alternative expression systems have to be used in production of these therapeutic proteins. Even where eukaryotic proteins do not have posttranslational modifications, prokaryotic expression systems often lack necessary chaperone proteins required for proper folding. Yeast and fungi are attractive options for expressing proteins as they can be easily grown at a large scale in simple media, which allows low production costs, and yeast and fungi have posttranslational machinery and chaperones that perform similar functions as found in mammalian cells.
  • yeasts like Pichia pastoris and Saccharomyces cerevisiae tend to hyper-mannosylate heterologously expressed biopharmaceuticals, thereby triggering adverse effects when applied to humans.
  • O-mannosylation to Serine and Threonine residues includes in mammals GalNAc based oligosaccharides or GlcNAc/N-acetyllactosamine comprising O-linked mannose glycans. In fungi O-mannosylation occurs as hexose monomers or oligomers. In yeasts, there are typically several protein(/polypeptide) O-mannosyltransferases, which often function as complexes. Part of the knock-outs are harmful, at least for cell structures and stability and not all yeast knock-outs or combinations are tolerated (for a review, see Goto 2007 , Biosci. Biotechnol. Biochem. 71(6), 1415-1427).
  • WO/2010/034708 reports no significant level of O-mannosylation of recombinant hydrophobin Trichoderma protein when expressed in pmt1 knock-out of S. cerevisiae host cell. Such O-mannosylation appears to be artificial yeast glycosylation of the original non-mannosylated filamentous fungal protein.
  • WO/2010/128143 further reports single chain antibody-albumin fusion construct in yeast S. cerevisiae pmt1 and/or pmt4 knock-out strains.
  • Aspergillus species Aspergillus nidulans, Aspergillus fumigatus , and/or Aspergillus awamori ) are described in Goto et al, 2009 (Eukaryotic cell 2009, 8(10):1465); Mouyna et al, 2010 (Molecular Microbiology 2010, 76(5), 1205-1221); Zhou et al, 2007 (Eukaryotic cell 2007, 6(12):2260); Oka et al, 2004 (Microbiology 2004, 150, 1973-1982); Kriangkripipat et al, 2009; Fang et al, 2010 (Glycobiology, 2010, vol. 20 pp 542-552); and Oka et al, 2005 (Microbiology 2005, 151, 3657-3667).
  • filamentous fungal cells such as Trichoderma fungus cells
  • the present invention relates to improved methods for producing proteins with no or reduced O-mannosylation in filamentous fungal expression systems, and more specifically, glycoproteins, such as antibodies or related immunoglobulins or fusion proteins which may be O-mannosylated when produced in filamentous fungal expression systems.
  • the present invention is based in part on the surprising discovery that filamentous fungal cells, such as Trichoderma cells, can be genetically modified to reduce or suppress O-mannosylation activity, without adversely affecting viability and yield of produced glycoproteins.
  • the invention relates to a PMT-deficient filamentous fungal cell comprising
  • said PMT-deficient cell further expresses a heterologous protein containing serine and/or threonine residues.
  • the expressed heterologous protein with serine and/or threonine residues has reduced O-mannosylation due to said mutation in said PMT gene.
  • the O-mannosylation level of the heterologous protein expressed in a PMT-deficient cell of the invention is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% or at least 90% lower as compared to the O-mannosylation level of the heterologous protein when expressed in the parental filamentous fungal cell which does not have said second PMT-deficient mutation.
  • said second mutation that reduces endogenous O-mannosyltransferase activity is a deletion or a disruption of a PMT gene encoding an endogenous protein O-mannosyltransferase activity.
  • said second PMT-deficient mutation in a PMT gene may be a mutation (such as a deletion or disruption) in either:
  • said PMT-deficient cell has a third mutation that reduces or eliminates the level of expression of an ALG3 gene compared to the level of expression in a parental cell which does not have such third mutation.
  • said PMT-deficient cell further comprises a first polynucleotide encoding N-acetylglucosaminyltransferase I catalytic domain and a second polynucleotide encoding N-acetylglucosaminyltransferase II catalytic domain.
  • said PMT-deficient cell further comprises one or more polynucleotides encoding a polypeptide selected from the group consisting of:
  • said PMT-deficient cell further comprises one or more polynucleotides encoding a ⁇ 1,4 galactosyltransferase and/or a fucosyltransferase.
  • said PMT-deficient cell is a Trichoderma cell comprising at least a mutation that reduces or eliminates the protein O-mannosyltransferase activity of Trichoderma pmt1, and, optionally, further comprising mutations in at least one or more other PMT genes that reduces or eliminates the protein O-mannosyltransferase activity selected from the group consisting of pmt2 and pmt3.
  • the PMT deficient cells comprise mutations that reduce or eliminate the activity of at least two, or at least three endogenous proteases.
  • said cell may be a Trichoderma cell and may comprise mutations that reduce or eliminate the activity of
  • the filamentous fungal cell of the invention does not comprise a deletion or disruption of an endogenous gene encoding a chaperone protein.
  • said filamentous fungal cell of the invention expresses functional endogenous chaperone protein Protein Disulphide Isomerase (PDI).
  • PDI Protein Disulphide Isomerase
  • the invention relates to a method for producing a protein having reduced O-mannosylation, comprising:
  • said mutation in a PMT gene is a mutation, such as a deletion or disruption, in either:
  • said PMT-deficient cell is a Trichoderma reesei cell and said mutation is a deletion or a disruption of T. reesei PMT1 gene.
  • said PMT-deficient cell is a PMT-deficient cell of the invention as described above.
  • said polynucleotide encoding a protein is a recombinant polynucleotide encoding a heterologous protein.
  • said heterologous protein may be a mammalian protein selected from the group consisting of
  • said polynucleotide encoding said protein further comprises a polynucleotide encoding CBH1 catalytic domain and linker as a carrier protein and/or cbh1 promoter.
  • said polynucleotide encodes a protein with serine or threonine, which may be O-mannosylated in a PMT functional parental strain, and further comprising at least one N-glycan.
  • the invention also relates to a method for producing an antibody having reduced 0-mannosylation, comprising:
  • said PMT-deficient cell is a Trichoderma reesei cell and said mutation is a deletion or a disruption of T. reesei PMT1 gene.
  • At least 70%, 80%, 90%, 95%, or 100% of the produced antibody is not O-mannosylated.
  • the invention also relates to the protein composition or antibody composition obtainable or obtained by the methods of the invention as described above.
  • at least 70%, 80%, 90%, 95%, or 100% of the antibodies as obtained or obtainable the methods of the invention are not O-mannosylated.
  • such protein e.g. a glycoprotein
  • antibody composition with reduced O-mannosylation comprises, as a major glycoform, either,
  • the composition when the core of the glycan consists of Man3, then the composition essentially lacks Man5 glycoforms.
  • less than 0.1%, 0.01%, 0.001% or 0% of the N-glycans and/or O-glycans of the protein composition comprises Neu5Gc and/or Gal ⁇ structure.
  • less than 0.1%, 0.01%, 0.001% or 0% of the N-glycans and/or O-glycans of the antibody composition comprises Neu5Gc and/or Gal ⁇ structure.
  • less than 0.1%, 0.01%, 0.001%, or 0% of the N-glycan of the glycoprotein composition comprises core fucose structures. In an embodiment that may be combined with the preceding embodiments, less than 0.1%, 0.01%, 0.001%, or 0% of the N-glycan of the antibody composition comprises core fucose structures.
  • less than 0.1%, 0.01%, 0.001%, or 0% of N-glycan of the glycoprotein composition comprises terminal galactose epitopes Gal ⁇ 3/4GlcNAc. In an embodiment that may be combined with the preceding embodiments, less than 0.1%, 0.01%, 0.001%, or 0% of the N-glycan of the antibody composition comprises terminal galactose epitopes Gal ⁇ 3/4GlcNAc.
  • less than 1.0%, 0.5%, 0.1%, 0.01%, 0.001%, or 0% of the glycoprotein composition comprises glycation structures. In an embodiment that may be combined with the preceding embodiments, less than 1.0%, 0.5%, 0.1%, 0.01%, 0.001%, or 0% of the antibody composition comprises glycation structures.
  • the glycoprotein composition such as an antibody is devoid of one, two, three, four, five, or six of the structures selected from the group of Neu5Gc, terminal Gal ⁇ 3Gal ⁇ 4GlcNAc, terminal Gal ⁇ 4GlcNAc, terminal Gal ⁇ 3GlcNAc, core linked fucose and glycation structures.
  • the invention also relates to a method of reducing O-mannosylation level of a recombinant glycoprotein composition produced in a filamentous fungal cell, for example, Trichoderma cell, typically, Trichoderma reesei , said method consisting of using a filamentous fungal cell having a mutation in a PMT gene wherein said PMT gene is either:
  • FIG. 1 depicts results for Southern analyses of Trichoderma reesei pmt1 deletion strains expressing antibody MAB01.
  • A) A 5.7 kb signal is expected from parental strains M124 and M304 with pmt1 ORF probe after SpeI+XbaI digestion. No signal is expected from pure pmt1 deletion strains.
  • B) A 3.5 kb signal is expected for pmt1 5′flank probe from deletion strains after SpeI+AscI digestion.
  • C) A 1.7 kb signal is expected for pmt1 3′flank probe from deletion strains after AscI+XbaI digestions.
  • FIG. 2 depicts a spectra of light chain of flask cultured parental T. reesei strain M317 (pyr4 ⁇ of M304) (A) and ⁇ pmt1 disruptant clone 26-8A (B), day 7.
  • FIG. 3 depicts results for Western analyses of Trichoderma reesei pmt1 deletion strain M403 from fed-batch fermentation.
  • Upper panel MAB01 light chain
  • lower panel MAB01 heavy chain. 0.1 ⁇ l of supernatant was loaded on each lane.
  • FIG. 4 depicts a spectrum of light chain of fermenter cultured T. reesei strain M403 (pmt1 deletion strain of MAB01 antibody producing strain, clone 26-8A), day 7.
  • FIG. 5 depicts a phylogeny of PMTs of selected filamentous fungi.
  • FIG. 6 depicts a partial sequence alignment of the results of the PMT BLAST searches.
  • an “expression system” or a “host cell” refers to the cell that is genetically modified to enable the transcription, translation and proper folding of a polypeptide or a protein of interest, typically of mammalian protein.
  • polynucleotide or “oligonucleotide” or “nucleic acid” as used herein typically refers to a polymer of at least two nucleotides joined together by a phosphodiester bond and may consist of either ribonucleotides or deoxynucleotides or their derivatives that can be introduced into a host cell for genetic modification of such host cell.
  • a polynucleotide may encode a coding sequence of a protein, and/or comprise control or regulatory sequences of a coding sequence of a protein, such as enhancer or promoter sequences or terminator.
  • a polynucleotide may for example comprise native coding sequence of a gene or their fragments, or variant sequences that have been optimized for optimal gene expression in a specific host cell (for example to take into account codon bias).
  • the term, “optimized” with reference to a polynucleotide means that a polynucleotide has been altered to encode an amino acid sequence using codons that are preferred in the production cell or organism, for example, a filamentous fungal cell such as a Trichoderma cell.
  • Heterologous nucleotide sequences that are transfected in a host cell are typically optimized to retain completely or as much as possible the amino acid sequence originally encoded by the original (not optimized) nucleotide sequence.
  • the optimized sequences herein have been engineered to have codons that are preferred in the corresponding production cell or organism, for example the filamentous fungal cell.
  • the amino acid sequences encoded by optimized nucleotide sequences may also be referred to as optimized.
  • a “peptide” or a “polypeptide” is an amino acid sequence including a plurality of consecutive polymerized amino acid residues.
  • the peptide or polypeptide may include modified amino acid residues, naturally occurring amino acid residues not encoded by a codon, and non-naturally occurring amino acid residues.
  • a “protein” may refer to a peptide or a polypeptide or a combination of more than one peptide or polypeptide assembled together by covalent or non-covalent bonds. Unless specified, the term “protein” may encompass one or more amino acid sequences with their post-translation modifications, and in particular with either O-mannosylation or N-glycan modifications.
  • glycoprotein refers to a protein which comprises at least one N-linked glycan attached to at least one asparagine residue of a protein, or at least one mannose attached to at least one serine or threonine resulting in O-mannosylation.
  • O-mannosylation or “O-mannosyltransferase activity” are used herein to refer to the covalent linkage of at least one mannose to one specific amino acid via one oxygen (typically from serine or threonine).
  • O-mannosyltransferase protein typically adds mannose to hydroxyl groups such as hydroxyl of serine or threonine residues.
  • O-mannosyltransferase activity may refer to the specificity of O-mannosyltransferase activity of fungal PMT gene encoding enzymes, and more specifically with the same specificity of T. reesei PMT1.
  • glycan refers to an oligosaccharide chain that can be linked to a carrier such as an amino acid, peptide, polypeptide, lipid or a reducing end conjugate.
  • the invention relates to N-linked glycans (“N-glycan”) conjugated to a polypeptide N-glycosylation site such as -Asn-Xaa-Ser/Thr- by N-linkage to side-chain amide nitrogen of asparagine residue (Asn), where Xaa is any amino acid residue except Pro.
  • the invention may further relate to glycans as part of dolichol-phospho-oligosaccharide (Dol-P-P-OS) precursor lipid structures, which are precursors of N-linked glycans in the endoplasmic reticulum of eukaryotic cells.
  • the precursor oligosaccharides are linked from their reducing end to two phosphate residues on the dolichol lipid.
  • ⁇ 3-mannosyltransferase Alg3 modifies the Dol-P-P-oligosaccharide precursor of N-glycans.
  • the glycan structures described herein are terminal glycan structures, where the non-reducing residues are not modified by other monosaccharide residue or residues.
  • glycolipid and carbohydrate nomenclature is essentially according to recommendations by the IUPAC-IUB Commission on Biochemical Nomenclature (e.g. Carbohydrate Res. 1998, 312, 167; Carbohydrate Res. 1997, 297, 1; Eur. J. Biochem. 1998, 257, 29).
  • Gal galactose
  • Glc glucose
  • GlcNAc N-acetylglucosamine
  • GalNAc N-acetylgalactosamine
  • Man Mannose
  • Neu5Ac Neu5Ac are of the D-configuration, Fuc of the L-configuration, and all the monosaccharide units in the pyranose form (D-Galp, D-Glcp, D-GlcpNAc, D-GalpNAc, D-Manp, L-Fucp, D-Neup5Ac).
  • the amine group is as defined for natural galactose and glucosamines on the 2-position of GalNAc or GlcNAc.
  • Glycosidic linkages are shown partly in shorter and partly in longer nomenclature, the linkages of the sialic acid SA/Neu5X-residues ⁇ 3 and ⁇ 6 mean the same as ⁇ 2-3 and ⁇ 2-6, respectively, and for hexose monosaccharide residues ⁇ 1-3, ⁇ 1-6, ⁇ 1-2, ⁇ 1-3, ⁇ 1-4, and ⁇ 1-6 can be shortened as ⁇ 3, ⁇ 6, ⁇ 2, ⁇ 3, ⁇ 4, and ⁇ 6, respectively.
  • Lactosamine refers to type II N-acetyllactosamine, Gal ⁇ 4GlcNAc, and/or type I N-acetyllactosamine.
  • Gal ⁇ 3GlcNAc and sialic acid refer to N-acetylneuraminic acid (Neu5Ac), N-glycolylneuraminic acid (Neu5Gc), or any other natural sialic acid including derivatives of Neu5X.
  • Sialic acid is referred to as NeuNX or Neu5X, where preferably X is Ac or Gc. Occasionally Neu5Ac/Gc/X may be referred to as NeuNAc/NeuNGc/NeuNX.
  • N-glycans found in mammalian glycoprotein
  • the sugars typically constituting N-glycans found in mammalian glycoprotein include, without limitation, N-acetylglucosamine (abbreviated hereafter as “GlcNAc”), mannose (abbreviated hereafter as “Man”), glucose (abbreviated hereafter as “Glc”), galactose (abbreviated hereafter as “Gal”), and sialic acid (abbreviated hereafter as “Neu5Ac”).
  • GlcNAc N-acetylglucosamine
  • Man mannose
  • Glc mannose
  • Glc glucose
  • Gal galactose
  • Neu5Ac sialic acid
  • N-glycans share a common pentasaccharide referred to as the “core” structure Man 3 GlcNAc 2 (Man ⁇ 6(Man ⁇ 3)Man ⁇ 4GlcNA ⁇ 4GlcNAc, referred to as Man3).
  • Man3 glycan or its derivative Man ⁇ 6(GlcNAc ⁇ 2Man ⁇ 3)Man ⁇ 4GlcNA ⁇ 4GlcNAc is the major glycoform.
  • the N-glycan or the core of N-glycan may be represented as Man 3 GlcNAc 2 (Fuc).
  • the major N-glycan is Man ⁇ 3[Man ⁇ 6(Man ⁇ 3)Man ⁇ 6]Man ⁇ 4GlcNA ⁇ 4GlcNAc (Man5).
  • Preferred hybrid type N-glycans comprise GlcNAc ⁇ 2Man ⁇ 3[Man ⁇ 6(Man ⁇ 3)Man ⁇ 6]Man ⁇ 4GlcNA ⁇ 4GlcNAc (“GlcNAcMan5”), or b4-galactosylated derivatives thereof Gal ⁇ 4GlcNAcMan3, G1, G2, or GalGlcNAcMan5 glycoform.
  • a “complex N-glycan” refers to a N-glycan which has at least one GlcNAc residue, optionally by GlcNAc ⁇ 2-residue, on terminal 1,3 mannose arm of the core structure and at least one GlcNAc residue, optionally by GlcNAc ⁇ 2-residue, on terminal 1,6 mannose arm of the core structure.
  • Such complex N-glycans include, without limitation, GlcNAc 2 Man 3 GlcNAc 2 (also referred as G0 glycoform), Gal 1 GlcNAc 2 Man 3 GlcNAc 2 (also referred as G1 glycoform), and Gal 2 GlcNAc 2 Man 3 GlcNAc 2 (also referred as G2 glycoform), and their core fucosylated glycoforms FG0, FG1 and FG2, respectively GlcNAc 2 Man 3 GlcNAc 2 (Fuc), Gal 1 GlcNAc 2 Man 3 GlcNAc 2 (Fuc), and Gal 2 GlcNAc 2 Man 3 GlcNAc 2 (Fuc).
  • the filamentous fungal cell may have increased or reduced levels of activity of various endogenous enzymes. A reduced level of activity may be provided by inhibiting the activity of the endogenous enzyme with an inhibitor, an antibody, or the like.
  • the filamentous fungal cell is genetically modified in ways to increase or reduce activity of various endogenous enzymes.
  • “Genetically modified” refers to any recombinant DNA or RNA method used to create a prokaryotic or eukaryotic host cell that expresses a polypeptide at elevated levels, at lowered levels, or in a mutated form. In other words, the host cell has been transfected, transformed, or transduced with a recombinant polynucleotide molecule, and thereby been altered so as to cause the cell to alter expression of a desired protein.
  • Genes which result in a decrease or deficiency in gene expression, in the function of the gene, or in the function of the gene product (i.e., the protein encoded by the gene) can be referred to as inactivation (complete or partial), knock-out, deletion, disruption, interruption, blockage, silencing, or down-regulation, or attenuation of expression of a gene.
  • a genetic modification in a gene which results in a decrease in the function of the protein encoded by such gene can be the result of a complete deletion of the gene (i.e., the gene does not exist, and therefore the protein does not exist), a mutation in the gene which results in incomplete (disruption) or no translation of the protein (e.g., the protein is not expressed), or a mutation in the gene which decreases or abolishes the natural function of the protein (e.g., a protein is expressed which has decreased or no enzymatic activity or action).
  • reference to decreasing the action of proteins discussed herein generally refers to any genetic modification in the host cell in question, which results in decreased expression and/or functionality (biological activity) of the proteins and includes decreased activity of the proteins (e.g., decreased catalysis), increased inhibition or degradation of the proteins as well as a reduction or elimination of expression of the proteins.
  • the action or activity of a protein can be decreased by blocking or reducing the production of the protein, reducing protein action, or inhibiting the action of the protein. Combinations of some of these modifications are also possible. Blocking or reducing the production of a protein can include placing the gene encoding the protein under the control of a promoter that requires the presence of an inducing compound in the growth medium.
  • Blocking or reducing the action of a protein could also include using an excision technology approach similar to that described in U.S. Pat. No. 4,743,546.
  • the gene encoding the protein of interest is cloned between specific genetic sequences that allow specific, controlled excision of the gene from the genome. Excision could be prompted by, for example, a shift in the cultivation temperature of the culture, as in U.S. Pat. No. 4,743,546, or by some other physical or nutritional signal.
  • an increase or a decrease in a given characteristic of a mutant or modified protein is made with reference to the same characteristic of a parent (i.e., normal, not modified) protein that is derived from the same organism (from the same source or parent sequence), which is measured or established under the same or equivalent conditions.
  • a characteristic of a genetically modified host cell e.g., expression and/or biological activity of a protein, or production of a product
  • a wild-type host cell of the same species and preferably the same strain, under the same or equivalent conditions.
  • Such conditions include the assay or culture conditions (e.g., medium components, temperature, pH, etc.) under which the activity of the protein (e.g., expression or biological activity) or other characteristic of the host cell is measured, as well as the type of assay used, the host cell that is evaluated, etc.
  • equivalent conditions are conditions (e.g., culture conditions) which are similar, but not necessarily identical (e.g., some conservative changes in conditions can be tolerated), and which do not substantially change the effect on cell growth or enzyme expression or biological activity as compared to a comparison made under the same conditions.
  • a genetically modified host cell that has a genetic modification that increases or decreases (reduces) the activity of a given protein (e.g., an O-mannosyltransferase or protease) has an increase or decrease, respectively, in the activity or action (e.g., expression, production and/or biological activity) of the protein, as compared to the activity of the protein in a parent host cell (which does not have such genetic modification), of at least about 5%, and more preferably at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55 60%, 65%, 70%, 75 80%, 85 90%, 95%, or any percentage, in whole integers between 5% and 100% (e.g., 6%, 7%, 8%, etc.).
  • a given protein e.g., an O-mannosyltransferase or protease
  • a genetically modified host cell that has a genetic modification that increases or decreases (reduces) the activity of a given protein (e.g., an O-mannosyltransferase or protease) has an increase or decrease, respectively, in the activity or action (e.g., expression, production and/or biological activity) of the protein, as compared to the activity of the wild-type protein in a parent host cell, of at least about 2-fold, and more preferably at least about 5-fold, 10-fold, 20-fold, 30-fold, 40-fold, 50-fold, 75-fold, 100-fold, 125-fold, 150-fold, or any whole integer increment starting from at least about 2-fold (e.g., 3-fold, 4-fold, 5-fold, 6-fold, etc.).
  • a given protein e.g., an O-mannosyltransferase or protease
  • nucleic acid or amino acid sequences refers to two or more sequences or subsequences that are the same.
  • Two sequences are “substantially identical” if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 29% identity, optionally 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identity over a specified region, or, when not specified, over the entire sequence), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
  • the identity exists over a region that is at least about 50 nucleotides (or 10 amino acids) in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides (or 20, 50, 200, or more amino acids) in length.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • a “comparison window,” as used herein, includes reference to a segment of any one of the number of contiguous positions including, but not limited to from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well known in the art.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1981), by the homology alignment algorithm of Needleman and Wunsch (1970) J Mol Biol 48(3):443-453, by the search for similarity method of Pearson and Lipman (1988) Proc Natl Acad Sci USA 85(8):2444-2448, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection [see, e.g., Brent et al., (2003) Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (Ringbou Ed)].
  • BLAST and BLAST 2.0 algorithms Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1997) Nucleic Acids Res 25(17):3389-3402 and Altschul et al. (1990) J. Mol Biol 215(3)-403-410, respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul, (1993) Proc Natl Acad Sci USA 90(12):5873-5877).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.
  • “Functional variant” or “functional homologous gene” as used herein refers to a coding sequence or a protein having sequence similarity with a reference sequence, typically, at least 30%, 40%, 50%, 60%, 70%, 80%, 90% or 95% identity with the reference coding sequence or protein, and retaining substantially the same function as said reference coding sequence or protein.
  • a functional variant may retain the same function but with reduced or increased activity.
  • Functional variants include natural variants, for example, homologs from different species or artificial variants, resulting from the introduction of a mutation in the coding sequence. Functional variant may be a variant with only conservatively modified mutations.
  • Constantly modified mutations include individual substitutions, deletions or additions to an encoded amino acid sequence which result in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the disclosure.
  • the following eight groups contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Glycine (G); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W); 7) Serine (S), Threonine (T); and 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins (1984)).
  • filamentous fungal cells include cells from all filamentous forms of the subdivision Eumycota and Oomycota (as defined by Hawksworth et al., In, Ainsworth and Bisby's Dictionary of The Fungi, 8th edition, 1995, CAB International, University Press, Cambridge, UK). Filamentous fungal cells are generally characterized by a mycelial wall composed of chitin, cellulose, glucan, chitosan, mannan, and other complex polysaccharides. Vegetative growth is by hyphal elongation and carbon catabolism is obligately aerobic. In contrast, vegetative growth by yeasts such as Saccharomyces cerevisiae is by budding of a unicellular thallus and carbon catabolism may be fermentative.
  • the filamentous fungal cell is not adversely affected by the transduction of the necessary nucleic acid sequences, the subsequent expression of the proteins (e.g., mammalian proteins), or the resulting intermediates.
  • the proteins e.g., mammalian proteins
  • General methods to disrupt genes of and cultivate filamentous fungal cells are disclosed, for example, for Penicillium , in Kopke et al. (2010) Appl Environ Microbiol. 76(14):4664-74. doi: 10.1128/AEM.00670-10, for Aspergillus , in Maruyama and Kitamoto (2011), Methods in Molecular Biology, vol. 765, D0110.1007/978-1-61779-197-0_27; for Neurospora, in Collopy et al.
  • filamentous fungal cells include, without limitation, cells from an Acremonium, Aspergillus, Fusarium, Humicola, Mucor, Myceliophthora, Neurospora, Penicillium, Scytalidium, Thielavia, Tolypocladium , or Trichoderma/Hypocrea strain.
  • the filamentous fungal cell is from a Trichoderma sp., Acremonium, Aspergillus, Aureobasidium, Cryptococcus, Chrysosporium, Chrysosporium lucknowense, Filibasidium, Fusarium, Gibberella, Magnaporthe, Mucor, Myceliophthora, Myrothecium, Neocallimastix, Neurospora, Paecilomyces, Penicillium, Piromyces, Schizophyllum, Talaromyces, Thermoascus, Thielavia , or Tolypocladium strain.
  • the filamentous fungal cell is a Myceliophthora or Chrysosporium, Neurospora or Trichoderma strain.
  • Aspergillus fungal cells of the present disclosure may include, without limitation, Aspergillus aculeatus, Aspergillus awamori, Aspergillus clavatus, Aspergillus flavus, Aspergillus foetidus, Aspergillus fumigatus, Aspergillus japonicus, Aspergillus nidulans, Aspergillus niger, Aspergillus oryzae , or Aspergillus terreus.
  • Neurospora fungal cells of the present disclosure may include, without limitation, Neurospora crassa.
  • Myceliophthora fungal cells of the present disclosure may include, without limitation, Myceliophthora thermophila.
  • the filamentous fungal cell is a Trichoderma fungal cell.
  • Trichoderma fungal cells of the present disclosure may be derived from a wild-type Trichoderma strain or a mutant thereof.
  • suitable Trichoderma fungal cells include, without limitation, Trichoderma harzianum, Trichoderma koningii, Trichoderma longibrachiatum, Trichoderma reesei, Trichoderma atroviride, Trichoderma virens, Trichoderma viride ; and alternative sexual form thereof (i.e., Hypocrea ).
  • the filamentous fungal cell is a Trichoderma reesei , and for example, strains derived from ATCC 13631 (QM 6a), ATCC 24449 (radiation mutant 207 of QM 6a), ATCC 26921 (QM 9414; mutant of ATCC 24449), VTT-D-00775 (Selinheimo et al., FEBS J., 2006, 273: 4322-4335), Rut-C30 (ATCC 56765), RL-P37 (NRRL 15709) or T. harzianum isolate T3 (Wolffhechel, H., 1989).
  • the invention described herein relates to a PMT deficient filamentous fungal cell, for example selected from Trichoderma, Neurospora, Myceliophthora or a Chrysosporium cells, such as Trichoderma reesei fungal cell, comprising:
  • protease activity enables to increase substantially the production of heterologous mammalian protein.
  • such proteases found in filamentous fungal cells that express a heterologous protein normally catalyse significant degradation of the expressed recombinant protein.
  • the stability of the expressed protein is increased, resulting in an increased level of production of the protein, and in some circumstances, improved quality of the produced protein (e.g., full-length instead of degraded).
  • proteases include, without limitation, aspartic proteases, trypsin-like serine proteases, subtilisin proteases, glutamic proteases, and sedolisin proteases. Such proteases may be identified and isolated from filamentous fungal cells and tested to determine whether reduction in their activity affects the production of a recombinant polypeptide from the filamentous fungal cell. Methods for identifying and isolating proteases are well known in the art, and include, without limitation, affinity chromatography, zymogram assays, and gel electrophoresis.
  • An identified protease may then be tested by deleting the gene encoding the identified protease from a filamentous fungal cell that expresses a recombinant polypeptide, such a heterologous or mammalian polypeptide, and determining whether the deletion results in a decrease in total protease activity of the cell, and an increase in the level of production of the expressed recombinant polypeptide.
  • Methods for deleting genes, measuring total protease activity, and measuring levels of produced protein are well known in the art and include the methods described herein.
  • Aspartic proteases are enzymes that use an aspartate residue for hydrolysis of the peptide bonds in polypeptides and proteins. Typically, aspartic proteases contain two highly-conserved aspartate residues in their active site which are optimally active at acidic pH. Aspartic proteases from eukaryotic organisms such as Trichoderma fungi include pepsins, cathepsins, and renins. Such aspartic proteases have a two-domain structure, which is thought to arise from ancestral gene duplication. Consistent with such a duplication event, the overall fold of each domain is similar, though the sequences of the two domains have begun to diverge. Each domain contributes one of the catalytic aspartate residues.
  • the active site is in a cleft formed by the two domains of the aspartic proteases.
  • Eukaryotic aspartic proteases further include conserved disulfide bridges, which can assist in identification of the polypeptides as being aspartic acid proteases.
  • Trichoderma fungal cells pep1 (tre74156); pep2 (tre53961); pep3 (tre121133); pep4 (tre77579), pep5 (tre81004), and pep7 (tre58669), pep8 (tre122076), pep11 (121306), and pep12 (tre119876).
  • Suitable aspartic proteases include, without limitation, Trichoderma reesei pep1 (SEQ ID NO: 22), Trichoderma reesei pep2 (SEQ ID NO: 18), Trichoderma reesei pep3 (SEQ ID NO: 19); Trichoderma reesei pep4 (SEQ ID NO: 20), Trichoderma reesei pep5 (SEQ ID NO: 21) and Trichoderma reesei pep7 (SEQ ID NO:23), Trichoderma reesei EGR48424 pep8 (SEQ ID NO:134), Trichoderma reesei EGR49498 pep11 (SEQ ID NO:135), Trichoderma reesei EGR52517 pep12 (SEQ ID NO:35), and homologs thereof.
  • Trypsin-like serine proteases are enzymes with substrate specificity similar to that of trypsin. Trypsin-like serine proteases use a serine residue for hydrolysis of the peptide bonds in polypeptides and proteins. Typically, trypsin-like serine proteases cleave peptide bonds following a positively-charged amino acid residue. Trypsin-like serine proteases from eukaryotic organisms such as Trichoderma fungi include trypsin 1, trypsin 2, and mesotrypsin.
  • trypsin-like serine proteases generally contain a catalytic triad of three amino acid residues (such as histidine, aspartate, and serine) that form a charge relay that serves to make the active site serine nucleophilic.
  • Eukaryotic trypsin-like serine proteases further include an “oxyanion hole” formed by the backbone amide hydrogen atoms of glycine and serine, which can assist in identification of the polypeptides as being trypsin-like serine proteases.
  • tsp1 trypsin-like serine protease has been identified in Trichoderma fungal cells: tsp1 (tre73897). As discussed in PCT/EP/2013/050186, tsp1 has been demonstrated to have a significant impact on expression of recombinant glycoproteins, such as immunoglobulins.
  • tsp1 proteases examples include, without limitation, Trichoderma reesei tsp1 (SEQ ID NO: 24) and homologs thereof. Examples of homologs of tsp1 proteases identified in other organisms are described in PCT/EP/2013/050186.
  • Subtilisin proteases are enzymes with substrate specificity similar to that of subtilisin. Subtilisin proteases use a serine residue for hydrolysis of the peptide bonds in polypeptides and proteins. Generally, subtilisin proteases are serine proteases that contain a catalytic triad of the three amino acids aspartate, histidine, and serine. The arrangement of these catalytic residues is shared with the prototypical subtilisin from Bacillus licheniformis . Subtilisin proteases from eukaryotic organisms such as Trichoderma fungi include furin, MBTPS1, and TPP2. Eukaryotic trypsin-like serine proteases further include an aspartic acid residue in the oxyanion hole.
  • subtilisin proteases have been identified in Trichoderma fungal cells: slp1 (tre51365); slp2 (tre123244); slp3 (tre123234); slp5 (tre64719), slp6 (tre121495), slp7 (tre123865), and slp8 (tre58698).
  • Subtilisin protease slp7 resembles also sedolisin protease tpp1.
  • slp proteases include, without limitation, Trichoderma reesei slp1 (SEQ ID NO: 25), slp2 (SEQ ID NO: 26); slp3 (SEQ ID NO: 27); slp5 (SEQ ID NO: 28), slp6 (SEQ ID NO: 29), slp7 (SEQ ID NO: 30), and slp8 (SEQ ID NO: 31), and homologs thereof. Examples of homologs of slp proteases identified in other organisms are described in PCT/EP/2013/050186.
  • Glutamic proteases are enzymes that hydrolyse the peptide bonds in polypeptides and proteins. Glutamic proteases are insensitive to pepstatin A, and so are sometimes referred to as pepstatin insensitive acid proteases. While glutamic proteases were previously grouped with the aspartic proteases and often jointly referred to as acid proteases, it has been recently found that glutamic proteases have very different active site residues than aspartic proteases.
  • gap1 Trichoderma fungal cells
  • gap2 Trichoderma reesei gap1
  • SEQ ID NO: 33 Trichoderma reesei gap2
  • homologs of gap proteases identified in other organisms are described in PCT/EP/2013/050186.
  • Sedolisin proteases are enzymes that use a serine residue for hydrolysis of the peptide bonds in polypeptides and proteins. Sedolisin proteases generally contain a unique catalytic triad of serine, glutamate, and aspartate. Sedolisin proteases also contain an aspartate residue in the oxyanion hole. Sedolisin proteases from eukaryotic organisms such as Trichoderma fungi include tripeptidyl peptidase.
  • tpp1 proteases examples include, without limitation, Trichoderma reesei tpp1 tre82623 (SEQ ID NO: 34) and homologs thereof. Examples of homologs of tpp1 proteases identified in other organisms are described in PCT/EP/2013/050186.
  • homolog refers to a protein which has protease activity and exhibit sequence similarity with a known (reference) protease sequence. Homologs may be identified by any method known in the art, preferably, by using the BLAST tool to compare a reference sequence to a single second sequence or fragment of a sequence or to a database of sequences. As described in the “Definitions” section, BLAST will compare sequences based upon percent identity and similarity.
  • a homologous protease has at least 30% identity with (optionally 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99% or 100% identity over a specified region, or, when not specified, over the entire sequence), when compared to one of the protease sequences listed above, including T. reesei pep1, pep2, pep3, pep4, pep5, pep7, pep8, pep 11, pep 12, tsp1, slp1, slp2, slp3, slp5, slp6, slp7, slp8, tpp1, gap1 and gap2.
  • Corresponding homologous proteases from N. crassa and M. thermophila are shown in SEQ ID NO: 136-169.
  • the filamentous fungal cells according to the invention have reduced activity of at least one endogenous protease, typically 2, 3, 4, 5 or more, in order to improve the stability and production of the protein with reduced O-mannosylation in said filamentous fungal cell, preferably in a PMT-deficient Trichoderma cell.
  • proteases found in filamentous fungal cells can be reduced by any method known to those of skill in the art.
  • reduced activity of proteases is achieved by reducing the expression of the protease, for example, by promoter modification or RNAi.
  • the reduced or eliminated expression of the proteases is the result of anti-sense polynucleotides or RNAi constructs that are specific for each of the genes encoding each of the proteases.
  • an RNAi construct is specific for a gene encoding an aspartic protease such as a pep-type protease, a trypsin-like serine proteases such as a tsp1, a glutamic protease such as a gap-type protease, a subtilisin protease such as a slp-type protease, or a sedolisin protease such as a tpp1 or a slp7 protease.
  • an RNAi construct is specific for the gene encoding a slp-type protease. In one embodiment, an RNAi construct is specific for the gene encoding slp2, slp3, slp5 or slp6. In one embodiment, an RNAi construct is specific for two or more proteases. In one embodiment, two or more proteases are any one of the pep-type proteases, any one of the trypsin-like serine proteases, any one of the slp-type proteases, any one of the gap-type proteases and/or any one of the sedolisin proteases.
  • RNAi construct comprises any one of the following nucleic acid sequences (see also PCT/EP/2013/050186).
  • RNAi Target sequence GCACACTTTCAAGATTGGC (SEQ ID NO: 15) GTACGGTGTTGCCAAGAAG (SEQ ID NO: 16) GTTGAGTACATCGAGCGCGACAGCATTGTGCACACCATGCTTCCCCTCGA GTCCAAGGACAGCATCATCGTTGAGGACTCGTGCAACGGCGAGACGGAGA AGCAGGCTCCCTGGGGTCTTGCCCGTATCTCTCACCGAGACGCTCAAC TTTGGCTCCTTCAACAAGTACCTCTACACCGCTGATGGTGGTGAGGGTGT TGATGCCTATGTCATTGACACCGGCACCAACATCGAGCACGTCGACTTTG AGGGTCGTGCCAAGTGGGGCAAGACCATCCCTGCCGGCGATGAGGACGAG GACGGCAACGGCCACGGCACTCACTGCTCTGGTACCGTTGCTGGTAAGAA GTACGGTGTTGCCAAGAAGGCCCACGTCTACGCCGTCAAGGTGCTCCGAT CCAACGTCTACGCCGTCAAGGTGCTCCGAT CC
  • reduced activity of proteases is achieved by modifying the gene encoding the protease.
  • modifications include, without limitation, a mutation, such as a deletion or disruption of the gene encoding said endogenous protease activity.
  • the invention relates to a filamentous fungal cell, such as a PMT-deficient Trichoderma cell, which has a first mutation that reduces or eliminates at least one endogenous protease activity compared to a parental filamentous fungal cell which does not have such protease deficient mutation, said filamentous fungal cell further comprising at least a second mutation in a PMT gene that reduces endogenous protein O-mannosyltransferase activity compared to a parental Trichoderma cell which does not have said second PMT-deficient mutation.
  • Deletion or disruption mutation includes without limitation knock-out mutation, a truncation mutation, a point mutation, a missense mutation, a substitution mutation, a frameshift mutation, an insertion mutation, a duplication mutation, an amplification mutation, a translocation mutation, or an inversion mutation, and that results in a reduction in the corresponding protease activity.
  • Methods of generating at least one mutation in a protease encoding gene of interest are well known in the art and include, without limitation, random mutagenesis and screening, site-directed mutagenesis, PCR mutagenesis, insertional mutagenesis, chemical mutagenesis, and irradiation.
  • a portion of the protease encoding gene is modified, such as the region encoding the catalytic domain, the coding region, or a control sequence required for expression of the coding region.
  • a control sequence of the gene may be a promoter sequence or a functional part thereof, i.e., a part that is sufficient for affecting expression of the gene.
  • a promoter sequence may be inactivated resulting in no expression or a weaker promoter may be substituted for the native promoter sequence to reduce expression of the coding sequence.
  • Other control sequences for possible modification include, without limitation, a leader sequence, a propeptide sequence, a signal sequence, a transcription terminator, and a transcriptional activator.
  • Protease encoding genes that are present in filamentous fungal cells may also be modified by utilizing gene deletion techniques to eliminate or reduce expression of the gene.
  • Gene deletion techniques enable the partial or complete removal of the gene thereby eliminating their expression.
  • deletion of the gene may be accomplished by homologous recombination using a plasmid that has been constructed to contiguously contain the 5′ and 3′ regions flanking the gene.
  • protease encoding genes that are present in filamentous fungal cells may also be modified by introducing, substituting, and/or removing one or more nucleotides in the gene, or a control sequence thereof required for the transcription or translation of the gene.
  • nucleotides may be inserted or removed for the introduction of a stop codon, the removal of the start codon, or a frame-shift of the open reading frame.
  • Such a modification may be accomplished by methods known in the art, including without limitation, site-directed mutagenesis and peR generated mutagenesis (see, for example, Botstein and Shortie, 1985, Science 229: 4719; Lo et al., 1985, Proceedings of the National Academy of Sciences USA 81: 2285; Higuchi et al., 1988, Nucleic Acids Research 16: 7351; Shimada, 1996, Meth. Mol. Bioi. 57: 157; Ho et al., 1989, Gene 77: 61; Horton et al., 1989, Gene 77: 61; and Sarkar and Sommer, 1990, BioTechniques 8: 404).
  • protease encoding genes that are present in filamentous fungal cells may be modified by gene disruption techniques by inserting into the gene a disruptive nucleic acid construct containing a nucleic acid fragment homologous to the gene that will create a duplication of the region of homology and incorporate construct nucleic acid between the duplicated regions.
  • a gene disruption can eliminate gene expression if the inserted construct separates the promoter of the gene from the coding region or interrupts the coding sequence such that a nonfunctional gene product results.
  • a disrupting construct may be simply a selectable marker gene accompanied by 5′ and 3′ regions homologous to the gene. The selectable marker enables identification of transformants containing the disrupted gene.
  • Protease encoding genes that are present in filamentous fungal cells may also be modified by the process of gene conversion (see, for example, Iglesias and Trautner, 1983, Molecular General Genetics 189:5 73-76).
  • gene conversion a nucleotide sequence corresponding to the gene is mutagenized in vitro to produce a defective nucleotide sequence, which is then transformed into a Trichoderma strain to produce a defective gene.
  • the defective nucleotide sequence replaces the endogenous gene. It may be desirable that the defective nucleotide sequence also contains a marker for selection of transformants containing the defective gene.
  • Protease encoding genes of the present disclosure that are present in filamentous fungal cells that express a recombinant polypeptide may also be modified by established anti-sense techniques using a nucleotide sequence complementary to the nucleotide sequence of the gene (see, for example, Parish and Stoker, 1997, FEMS Microbiology Letters 154: 151-157).
  • expression of the gene by filamentous fungal cells may be reduced or inactivated by introducing a nucleotide sequence complementary to the nucleotide sequence of the gene, which may be transcribed in the strain and is capable of hybridizing to the mRNA produced in the cells. Under conditions allowing the complementary anti-sense nucleotide sequence to hybridize to the mRNA, the amount of protein translated is thus reduced or eliminated.
  • Protease encoding genes that are present in filamentous fungal cells may also be modified by random or specific mutagenesis using methods well known in the art, including without limitation, chemical mutagenesis (see, for example, Hopwood, The Isolation of Mutants in Methods in Microbiology (J. R. Norris and D. W. Ribbons, eds.) pp. 363-433, Academic Press, New York, 25 1970). Modification of the gene may be performed by subjecting filamentous fungal cells to mutagenesis and screening for mutant cells in which expression of the gene has been reduced or inactivated.
  • the mutagenesis which may be specific or random, may be performed, for example, by use of a suitable physical or chemical mutagenizing agent, use of a suitable oligonucleotide, subjecting the DNA sequence to peR generated mutagenesis, or any combination thereof.
  • suitable physical or chemical mutagenizing agents include, without limitation, ultraviolet (UV) irradiation, hydroxylamine, N-methyl-N′-nitro-N-nitrosoguanidine (MNNG), N-methyl-N′-nitrosogaunidine (NTG) O-methyl hydroxylamine, nitrous acid, ethyl methane sulphonate (EMS), sodium bisulphite, formic acid, and nucleotide analogues.
  • the mutagenesis is typically performed by incubating the filamentous fungal cells, such as Trichoderma cells, to be mutagenized in the presence of the mutagenizing agent of choice under suitable conditions, and then selecting for mutants exhibiting reduced or no expression of the gene.
  • the at least one mutation or modification in a protease encoding gene of the present disclosure results in a modified protease that has no detectable protease activity. In other embodiments, the at least one modification in a protease encoding gene of the present disclosure results in a modified protease that has at least 25% less, at least 50% less, at least 75% less, at least 90%, at least 95%, or a higher percentage less protease activity compared to a corresponding non-modified protease.
  • the filamentous fungal cells or Trichoderma fungal cells of the present disclosure may have reduced or no detectable protease activity of at least three, or at least four proteases selected from the group consisting of pep1, pep2, pep3, pep4, pep5, pep8, pep11, pep12, tsp1, slp1, slp2, slp3, slp5, slp6, slp7, gap1 and gap2.
  • a filamentous fungal cell according to the invention is a PMT-deficient filamentous fungal cell which has a deletion or disruption in at least 3 or 4 endogenous proteases, resulting in no detectable activity for such deleted or disrupted endogenous proteases and further comprising another mutation in a PMT gene that reduces endogenous protein O-mannosyltransferase activity compared to a parental Trichoderma cell which does not have said mutation.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in pep1, tsp1, and slp1. In other embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in gap1, slp1, and pep1. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1 and gap1. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1, gap1 and pep4.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in slp2, pep1, gap1, pep4 and slp1. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, and slp3. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, and pep3.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, pep3 and pep2. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, pep3, pep2 and pep5.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, pep3, pep2, pep5 and tsp1. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, pep3, pep2, pep5, tsp1 and slp7.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, pep3, pep2, pep5, tsp1, slp7 and slp8. In certain embodiments, the PMT-deficient filamentous fungal cell or Trichoderma cell, has reduced or no detectable protease activity in slp2, pep1, gap1, pep4, slp1, slp3, pep3, pep2, pep5, tsp1, slp7, slp8 and gap2.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in at least three endogenous proteases selected from the group consisting of pep1, pep2, pep3, pep4, pep5, pep8, pep11, pep12, tsp1, slp2, slp3, slp7, gap1 and gap2.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in at least three to six endogenous proteases selected from the group consisting of pep1, pep2, pep3, pep4, pep5, tsp1, slp1, slp2, slp3, gap1 and gap2.
  • the PMT-deficient filamentous fungal cell or Trichoderma cell has reduced or no detectable protease activity in at least seven to ten endogenous proteases selected from the group consisting of pep1, pep2, pep3, pep4, pep5, pep7, pep8, tsp1, slp1, slp2, slp3, slp5, slp6, slp7, slp8, tpp1, gap1 and gap2.
  • the filamentous fungal cell of the invention does not comprise a deletion or disruption of an endogenous gene encoding a chaperone protein.
  • said filamentous fungal cell of the invention expresses functional endogenous chaperone protein Protein Disulphide Isomerase (PDI).
  • PDI Protein Disulphide Isomerase
  • O-mannosyltransferases are encoded by pmt genes in yeasts and filamentous fungi, which can be divided into three subfamilies, based on sequence homologies: PMT1, PMT2 and PMT4.
  • An identified O-mannosyltransferase may then be tested by deleting the gene encoding the identified O-mannosyltransferase from a filamentous fungal cell that expresses a recombinant O-mannosylated protein, such a heterologous or mammalian O-mannosylated protein, and determining whether the deletion results in a decrease in total O-mannosyltransferase activity of the cell, preferably not affecting the level of production of the expressed recombinant protein.
  • Methods for deleting genes and measuring levels of produced protein are well known in the art and include the methods described herein.
  • Trichoderma fungal cells pmt1, pmt2 and pmt3, belonging respectively based on sequence homologies to the PMT4, PMT1 and PMT2 subfamily.
  • O-mannosyltransferase examples include, without limitation, Trichoderma reesei pmt1 (SEQ ID NO: 2), Trichoderma reesei pmt2 (SEQ ID NO: 3), Trichoderma reesei pmt3 (SEQ ID NO: 4) and homologs thereof.
  • FIG. 5 shows phylogeny of pmt homologs in selected filamentous fungi and
  • FIG. 6 shows an alignment of pmt1 conserved domains among different species.
  • said PMT-deficient filamentous fungal cell e.g., a Trichoderma cell
  • said PMT-deficient filamentous fungal cell has at least one mutation in a PMT gene selected from the group consisting of:
  • said PMT-deficient filamentous fungal cell e.g., a Trichoderma cell
  • said PMT-deficient filamentous fungal cell has at least one mutation in a PMT gene which
  • Sequences of homologs of pmt1 in filamentous fungi can be found in the databases using sequence alignment search tools, such as BLAST algorithm. It includes without limitation, A. oryzae gi391865791, EIT75070.1 (SEQ ID NO:5), A. niger gi317036343, XP_001398147.2 (SEQ ID NO:6), A. nidulans gi67522004, XP_659063.1 (SEQ ID NO:7), T. virens gi358379774, EHK17453.1 (SEQ ID NO:8), T. atroviride gi358400594, EHK49920.1 (SEQ ID NO:9), F.
  • sequence alignment search tools such as BLAST algorithm. It includes without limitation, A. oryzae gi391865791, EIT75070.1 (SEQ ID NO:5), A. niger gi317036343, XP_001398147.2 (
  • oxysporum gi342879728, EGU80965.1 (SEQ ID NO:10), G. zeae gi46107450, XP_380784.1 (SEQ ID NO:11), M. thermophila gi367020262, XP_003659416.1 (SEQ ID NO:12), N. crassa gi164423013, XP_963926.2 (SEQ ID NO:13), and P. chrysogenum gi255953619, XP_002567562.1 (SEQ ID NO:14).
  • the PMT-deficient filamentous fungal cells according to the invention have reduced activity of at least one O-mannosyltransferase activity, in order to reduce or decrease O-mannosylation in said filamentous fungal cell, preferably Trichoderma cell.
  • the activity of said O-mannosyltransferases found in filamentous fungal cells can be reduced by any method known to those of skill in the art.
  • reduced activity of O-mannosyltransferases is achieved by reducing the expression of the O-mannosyltransferases, for example, by promoter modification or RNAi.
  • reduced activity of O-mannosyltransferases is achieved by modifying the gene encoding the O-mannosyltransferase.
  • modifications include, without limitation, a mutation, such as a deletion or disruption of the gene encoding said endogenous O-mannosyltransferase activity.
  • Deletion or disruption mutation can be performed as described in the above sections, in particular in relation to deletion or disruption of genes encoding proteases. These includes without limitation knock-out mutation, a truncation mutation, a point mutation, a missense mutation, a substitution mutation, a frameshift mutation, an insertion mutation, a duplication mutation, an amplification mutation, a translocation mutation, or an inversion mutation, and that results in a reduction in the corresponding O-mannosyltransferase activity.
  • the mutation or modification in an O-mannosyltransferase (PMT) encoding gene of the present disclosure results in a modified O-mannosyltransferase that has no detectable O-mannosyltransferase activity.
  • the at least one modification in a O-mannosyltransferase encoding gene of the present disclosure results in a modified O-mannosyltransferase that has at least 25% less, at least 50% less, at least 75% less, at least 90%, at least 95%, or a higher percentage less O-mannosyltransferase activity compared to a corresponding non-modified O-mannosyltransferase.
  • a mutation that reduces endogenous protein O-mannosyltransferase activity in a PMT-deficient filamentous fungal cell is a PMT-deficient cell which has a deletion or disruption of a PMT gene encoding said O-mannosyltransferase activity, resulting in no detectable expression for such deleted or disrupted PMT gene.
  • One specific embodiment of the present invention is a PMT-deficient Trichoderma reesei cell, comprising
  • the reduction (or decrease) of O-mannosyltransferase activity may be determined by comparing the O-mannosylation level of a heterologous protein in PMT-deficient filamentous fungal cell according to the invention, with the O-mannosylation level of a heterologous protein in the parental cell which does not have said PMT-deficient mutation.
  • the PMT-deficient filamentous fungal cell according to the invention expresses a heterologous protein which has reduced O-mannosylation due to said mutation in said PMT gene and the O-mannosylation level on the expressed heterologous protein is at least 20%, 40%, 50%, 60%, 70%, 80%, or 90% lower than the O-mannosylation level of the heterologous protein when expressed in the parental filamentous fungal cell which does not have said second PMT-deficient mutation.
  • O-mannosylation level may also be determined as mole % of O-mannosylated polypeptide per total polypeptide as produced by the host cell of the invention.
  • Analytical methods such as MALDI TOF MS analysis may be used to determine O-mannosylation level as described in detail in the Example 1 below, section entitled “Analyses of Dpmt1 strains M403, M404, M406 and M407.
  • a polypeptide as produced by the PMT-deficient filamentous fungal cell is purified to determine its O-mannoslyation level.
  • Non O-mannosylated, and O-mannosylated structure of the polypeptide are separated and quantified by MALDI-TOF MS analysis.
  • the quantification of O-mannosylation level may be performed by determining area values or intensity of the different peaks of MALDI-TOF MS spectrum.
  • An O-mannosylation level of 5% as determined by such method, using area values or intensity, reflects that about 95% (mol %) of the analysed polypeptides in the composition are not O-mannosylated
  • the PMT-deficient filamentous fungal cell expresses a heterologous protein which has reduced O-mannosylation due to said mutation in said PMT gene, and the O-mannosylation level on the expressed heterologous protein (for example, as defined above by determining area or intensity values of MALDI TOF MS spectrum peaks) is reduced to less than 25%, 20%, 17%, 15%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1%, or 0.5% (as mole % of mannose residues per polypeptide chain).
  • heterologous protein with reduced O-mannosylation is selected from the group consisting of:
  • a mutation that reduces endogenous O-mannosyltransferase activity is a deletion or a disruption of a PMT gene encoding said engogenous protein O-mannosyltransferase activity.
  • a mutation that reduces endogenous O-mannosyltransferase activity is a deletion or a disruption of a PMT1 gene.
  • filamentous fungal cells according to the present invention may be useful in particular for producing heterologous glycoproteins with reduced O-mannosylation and mammalian-like N-glycans, such as complex N-glycans.
  • the filamentous fungal cell is further genetically modified to produce a mammalian-like N-glycan, thereby enabling in vivo production of glycoprotein with no or reduced O-mannosylation and with mammalian-like N-glycan as major glycoforms.
  • this aspect includes methods of producing glycoproteins with mammalian-like N-glycans in a Trichoderma cell.
  • the glycoprotein comprises, as a major glycoform, the mammalian-like N-glycan having the formula [(Gal ⁇ 4) x GlcNAc ⁇ 2] z Man ⁇ 3([(Gal ⁇ 4) y GlcNAc ⁇ 2] w Man ⁇ 6)Man( ⁇ 4GlcNAc ⁇ GlcNAc, where ( ) defines a branch in the structure, where [ ] or ⁇ ⁇ define a part of the glycan structure either present or absent in a linear sequence, and where x, y, z and w are 0 or 1, independently. In an embodiment w and z are 1.
  • the glycoprotein comprises, as a major glycoform, mammalian-like N-glycan selected from the group consisting of:
  • the glycoprotein composition with mammalian-like N-glycans include glycoforms that essentially lack or are devoid of glycans Man ⁇ 3[Man ⁇ 6(Man ⁇ 3)Man ⁇ 6]Man ⁇ 4GlcNA ⁇ 4GlcNAc (Man5).
  • the filamentous fungal cell produces glycoproteins with, as major glycoform, the trimannosyl N-glycan structure Man ⁇ 3[Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc.
  • the filamentous fungal cell procudes glycoproteins with, as major glycoform, the G0 N-glycan structure GlcNAc ⁇ 2Man ⁇ 3[GlcNAc ⁇ 2Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc.
  • the PMT-deficient filamentous fungal cell of the invention produces glycoprotein composition with a mixture of different N-glycans.
  • Man3GlcNAc2 N-glycan (i.e. Man ⁇ 3[Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc) represents at least 10%, at least 20%, at least at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of total (mol %) neutral N-glycans of a heterologous protein with reduced O-mannosylation, as expressed in a filamentous fungal cells of the invention.
  • GlcNAc2Man3 N-glycan (for example G0 GlcNAc ⁇ 2Man ⁇ 3[GlcNAc ⁇ 2Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc) represents at least 10%, at least 20%, at least at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of total (mol %) neutral N-glycans of a heterologous protein with reduced O-mannosylation, as expressed in a filamentous fungal cells of the invention.
  • GalGlcNAc2Man3GlcNAc2 N-glycan represents at least 10%, at least 20%, at least at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of total (mol %) neutral N-glycans of a heterologous protein with reduced O-mannosylation, as expressed in a filamentous fungal cells of the invention.
  • GaI2GlcNAc2Man3GlcNAc2 N-glycan represents at least 10%, at least 20%, at least at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of total (mol %) neutral N-glycans of a heterologous protein with reduced O-mannosylation, as expressed in a filamentous fungal cells of the invention.
  • complex type N-glycan represents at least 10%, at least 20%, at least at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of total (mol %) neutral N-glycans of a heterologous protein with reduced O-mannosylation, as expressed in a filamentous fungal cells of the invention.
  • hybrid type N-glycan represents at least 10%, at least 20%, at least at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% or more of total (mol %) neutral N-glycans of a heterologous protein with reduced O-mannosylation, as expressed in a filamentous fungal cells of the invention.
  • N-glycan of the glycoprotein composition produced by the host cell of the invention comprises galactose. In certain embodiments, none of N-glycans comprise galactose.
  • the Neu5Gc and Gal ⁇ (non-reducing end terminal Gal ⁇ 3Gal ⁇ 4GlcNAc) structures are known xenoantigenic (animal derived) modifications of antibodies which are produced in animal cells such as CHO cells.
  • the structures may be antigenic and, thus, harmful even at low concentrations.
  • the filamentous fungi of the present invention lack biosynthetic pathways to produce the terminal Neu5Gc and Gal ⁇ structures.
  • less than 0.1%, 0.01%, 0.001% or 0% of the N-glycans and/or O-glycans of the glycoprotein composition comprises Neu5Gc and/or Gal ⁇ structure.
  • less than 0.1%, 0.01%, 0.001% or 0% of the N-glycans and/or O-glycans of the antibody composition comprises Neu5Gc and/or Gal ⁇ structure.
  • the filamentous fungal cells of the present invention lack genes to produce fucosylated heterologous proteins.
  • less than 0.1%, 0.01%, 0.001%, or 0% of the N-glycan of the glycoprotein composition comprises core fucose structures.
  • less than 0.1%, 0.01%, 0.001%, or 0% of the N-glycan of the antibody composition comprises core fucose structures.
  • the terminal Gal ⁇ 4GlcNAc structure of N-glycan of mammalian cell produced glycans affects bioactivity of antibodies and Gal ⁇ 3GlcNAc may be xenoantigen structure from plant cell produced proteins.
  • less than 0.1%, 0.01%, 0.001%, or 0% of N-glycan of the glycoprotein composition comprises terminal galactose epitopes Gal ⁇ 3/4GlcNAc.
  • less than 0.1%, 0.01%, 0.001%, or 0% of the N-glycan of the antibody composition comprises terminal galactose epitopes Gal ⁇ 3/4GlcNAc.
  • Glycation is a common post-translational modification of proteins, resulting from the chemical reaction between reducing sugars such as glucose and the primary amino groups on protein. Glycation occurs typically in neutral or slightly alkaline pH in cell cultures conditions, for example, when producing antibodies in CHO cells and analysing them (see, for example, Zhang et al. (2008) Unveiling a glycation hot spot in a recombinant humanized monoclonal antibody. Anal Chem. 80(7):2379-2390). As filamentous fungi of the present invention are typically cultured in acidic pH, occurrence of glycation is reduced.
  • less than 1.0%, 0.5%, 0.1%, 0.01%, 0.001%, or 0% of the glycoprotein composition comprises glycation structures. In an embodiment that may be combined with the preceding embodiments, less than 1.0%, 0.5%, 0.1%, 0.01%, 0.001%, or 0% of the antibody composition comprises glycation structures.
  • the glycoprotein composition such as an antibody is devoid of one, two, three, four, five, or six of the structures selected from the group of Neu5Gc, terminal Gal ⁇ 3Gal ⁇ 4GlcNAc, terminal Gal ⁇ 4GlcNAc, terminal Gal ⁇ 3GlcNAc, core linked fucose and glycation structures.
  • such glycoprotein protein with mammalian-like N-glycan and reduced O-mannosylation, as produced in the filamentous fungal cell of the invention is a therapeutic protein.
  • Therapeutic proteins may include immunoglobulin, or a protein fusion comprising a Fc fragment or other therapeutic glycoproteins, such as antibodies, erythropoietins, interferons, growth hormones, albumins or serum albumin, enzymes, or blood-clotting factors and may be useful in the treatment of humans or animals.
  • the glycoproteins with mammalian-like N-glycan and reduced O-mannosylation as produced by the filamentous fungal cell according to the invention may be a therapeutic glycoprotein such as rituximab.
  • the filamentous fungal cell according to the invention as described above is further genetically modified to mimic the traditional pathway of mammalian cells, starting from Man5 N-glycans as acceptor substrate for GnTI, and followed sequentially by GnT1, mannosidase II and GnTII reaction steps (hereafter referred as the “traditional pathway” for producing G0 glycoforms).
  • a single recombinant enzyme comprising the catalytic domains of GnTI and GnTII, is used.
  • the filamentous fungal cell according to the invention as described above is further genetically modified to have alg3 reduced expression, allowing the production of core Man 3 GlcNAc 2 N-glycans, as acceptor substrate for GnTI and GnTII subsequent reactions and bypassing the need for mannosidase ⁇ 1,2 or mannosidase II enzymes (the reduced “alg3” pathway).
  • a single recombinant enzyme comprising the catalytic domains of GnTI and GnTII, is used.
  • a Man 5 expressing filamentous fungal cell such as T. reesei strain
  • T. reesei strain may be transformed with a GnTI or a GnTII/GnTI fusion enzyme using random integration or by targeted integration to a known site known not to affect Man5 glycosylation.
  • Strains that synthesise GlcNAcMan5 N-glycan for production of proteins having hybrid type glycan(s) are selected.
  • mannosidase II-type mannosidase capable of cleaving Man5 structures to generate GlcNAcMan3 for production of proteins having the corresponding GlcNAcMan3 glycoform or their derivative(s).
  • mannosidase II-type enzymes belong to glycoside hydrolase family 38 (cazy.org/GH38_all.html). Characterized enzymes include enzymes listed in seray.org/GH38_characterized.html.
  • Especially useful enzymes are Golgi-type enzymes that cleaving glycoproteins, such as those of subfamily ⁇ -mannosidase II (Man2A1;ManA2). Examples of such enzymes include human enzyme AAC50302, D.
  • the catalytic domain of the mannosidase is typically fused with an N-terminal targeting peptide (for example as disclosed in the above Section) or expressed with endogenous animal or plant Golgi targeting structures of animal or plant mannosidase II enzymes.
  • strains After transformation with the catalytic domain of a mannosidase II-type mannosidase, strains are selected that produce GlcNAcMan3 (if GnTI is expressed) or strains are selected that effectively produce GlcNAc2Man3 (if a fusion of GnTI and GnTII is expressed). For strains producing GlcNAcMan3, such strains are further transformed with a polynucleotide encoding a catalytic domain of GnTII and transformant strains that are capable of producing GlcNAc2Man3GlcNAc2 are selected.
  • the filamentous fungal cell is a PMT-deficient filamentous fungal cell as defined in previous sections, and further comprising one or more polynucleotides encoding a polypeptide selected from the group consisting of:
  • the filamentous fungal cell such as a Trichoderma cell
  • Dolichyl-P-Man:Man(5)GlcNAc(2)-PP-dolichyl mannosyltransferase (EC 2.4.1.130) transfers an alpha-D-mannosyl residue from dolichyl-phosphate D-mannose into a membrane lipid-linked oligosaccharide.
  • the dolichyl-P-Man:Man(5)GlcNAc(2)-PP-dolichyl mannosyltransferase enzyme is encoded by an alg3 gene.
  • the filamentous fungal cell for producing glycoproteins with mammalian-like N-glycans has a reduced level of expression of an alg3 gene compared to the level of expression in a parent strain.
  • the filamentous fungal cell comprises a mutation of alg3.
  • the ALG3 gene may be mutated by any means known in the art, such as point mutations or deletion of the entire alg3 gene.
  • the function of the alg3 protein is reduced or eliminated by the mutation of alg3.
  • the alg3 gene is disrupted or deleted from the filamentous fungal cell, such as Trichoderma cell.
  • the filamentous fungal cell is a T. reesei cell.
  • SEQ ID NOs: 36 and 37 provide, the nucleic acid and amino acid sequences of the alg3 gene in T. reesei , respectively.
  • the filamentous fungal cell is used for the production of a glycoprotein, wherein the glycan(s) comprise or consist of Man ⁇ 3[Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc, and/or a non-reducing end elongated variant thereof.
  • the filamentous fungal cell has a reduced level of activity of an alpha-1,6-mannosyltransferase compared to the level of activity in a parent strain.
  • Alpha-1,6-mannosyltransferase (EC 2.4.1.232) transfers an alpha-D-mannosyl residue from GDP-mannose into a protein-linked oligosaccharide, forming an elongation initiating alpha-(1->6)-D-mannosyl-D-mannose linkage in the Golgi apparatus.
  • the alpha-1,6-mannosyltransferase enzyme is encoded by an och1 gene.
  • the filamentous fungal cell has a reduced level of expression of an och1 gene compared to the level of expression in a parent filamentous fungal cell.
  • the och1 gene is deleted from the filamentous fungal cell.
  • the filamentous fungal cells used in the methods of producing glycoprotein with mammalian-like N-glycans may further contain a polynucleotide encoding an N-acetylglucosaminyltransferase I catalytic domain (GnTI) that catalyzes the transfer of N-acetylglucosamine to a terminal Man ⁇ 3 and a polynucleotide encoding an N-acetylglucosaminyltransferase II catalytic domain (GnTII), that catalyses N-acetylglucosamine to a terminal Man ⁇ 6 residue of an acceptor glycan to produce a complex N-glycan.
  • said polynucleotides encoding GnTI and GnTII are linked so as to produce a single protein fusion comprising both catalytic domains of GnTI and GnTII.
  • N-acetylglucosaminyltransferase I catalytic domain is any portion of an N-acetylglucosaminyltransferase I enzyme that is capable of catalyzing this reaction.
  • GnTI enzymes are listed in the CAZy database in the glycosyltransferase family 13 (cazy.org/GT13_all).
  • Enzymatically characterized species includes A. thaliana AAR78757.1 (U.S. Pat. No. 6,653,459), C. elegans AAD03023.1 (Chen S. et al J. Biol. Chem 1999; 274(1):288-97), D.
  • the N-acetylglucosaminyltransferase I catalytic domain is from the human N-acetylglucosaminyltransferase I enzyme (SEQ ID NO: 38) or variants thereof.
  • the N-acetylglucosaminyltransferase I catalytic domain contains a sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to amino acid residues 84-445 of SEQ ID NO: 38.
  • a shorter sequence can be used as a catalytic domain (e.g. amino acid residues 105-445 of the human enzyme or amino acid residues 107-447 of the rabbit enzyme; Sarkar et al. (1998) Glycoconjugate J 15:193-197). Additional sequences that can be used as the GnTI catalytic domain include amino acid residues from about amino acid 30 to 445 of the human enzyme or any C-terminal stem domain starting between amino acid residue 30 to 105 and continuing to about amino acid 445 of the human enzyme, or corresponding homologous sequence of another GnTI or a catalytically active variant or mutant thereof.
  • the catalytic domain may include N-terminal parts of the enzyme such as all or part of the stem domain, the transmembrane domain, or the cytoplasmic domain.
  • N-acetylglucosaminyltransferase II catalytic domain is any portion of an N-acetylglucosaminyltransferase II enzyme that is capable of catalyzing this reaction.
  • Amino acid sequences for N-acetylglucosaminyltransferase II enzymes from various organisms are listed in WO2012069593.
  • the N-acetylglucosaminyltransferase II catalytic domain is from the human N-acetylglucosaminyltransferase II enzyme (SEQ ID NO: 39) or variants thereof.
  • GnTII species are listed in the CAZy database in the glycosyltransferase family 16 (cazy.org/GT16_all). Enzymatically characterized species include GnTII of C. elegans, D. melanogaster, Homo sapiens (NP_002399.1), Rattus norvegicus, Sus scrofa (cazy.org/GT16_characterized).
  • the N-acetylglucosaminyltransferase II catalytic domain contains a sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to amino acid residues from about 30 to about 447 of SEQ ID NO: 39.
  • the catalytic domain may include N-terminal parts of the enzyme such as all or part of the stem domain, the transmembrane domain, or the cytoplasmic domain.
  • the fusion protein may further contain a spacer in between the N-acetylglucosaminyltransferase I catalytic domain and the N-acetylglucosaminyltransferase II catalytic domain.
  • the spacer is an EGIV spacer, a 2 ⁇ G4S spacer, a 3 ⁇ G4S spacer, or a CBHI spacer.
  • the spacer contains a sequence from a stem domain.
  • N-acetylglucosaminyltransferase I and/or N-acetylglucosaminyltransferase II catalytic domain is typically fused with a targeting peptide or a part of an ER or early Golgi protein, or expressed with an endogenous ER targeting structures of an animal or plant N-acetylglucosaminyltransferase enzyme.
  • the N-acetylglucosaminyltransferase I and/or N-acetylglucosaminyltransferase II catalytic domain contains any of the targeting peptides of the invention as described in the section entitled “Targeting sequences”.
  • the targeting peptide is linked to the N-terminal end of the catalytic domain.
  • the targeting peptide contains any of the stem domains of the invention as described in the section entitled “Targeting sequences”.
  • the targeting peptide is a Kre2/Mnt1 targeting peptide.
  • the targeting peptide further contains a transmembrane domain linked to the N-terminal end of the stem domain or a cytoplasmic domain linked to the N-terminal end of the stem domain. In embodiments where the targeting peptide further contains a transmembrane domain, the targeting peptide may further contain a cytoplasmic domain linked to the N-terminal end of the transmembrane domain.
  • the filamentous fungal cells may also contain a polynucleotide encoding a UDP-GlcNAc transporter.
  • the polynucleotide encoding the UDP-GlcNAc transporter may be endogenous (i.e., naturally present) in the host cell, or it may be heterologous to the filamentous fungal cell.
  • the filamentous fungal cell may further contain a polynucleotide encoding a ⁇ -1,2-mannosidase.
  • the polynucleotide encoding the ⁇ -1,2-mannosidase may be endogenous in the host cell, or it may be heterologous to the host cell. Heterologous polynucleotides are especially useful for a host cell expressing high-mannose glycans transferred from the Golgi to the ER without effective exo- ⁇ -2-mannosidase cleavage.
  • the ⁇ -1,2-mannosidase may be a mannosidase I type enzyme belonging to the glycoside hydrolase family 47 (cazy.org/GH47_all.html).
  • the ⁇ -1,2-mannosidase is an enzyme listed at Merriy.org/GH47_characterized.html.
  • the ⁇ -1,2-mannosidase may be an ER-type enzyme that cleaves glycoproteins such as enzymes in the subfamily of ER ⁇ -mannosidase I EC 3.2.1.113 enzymes.
  • examples of such enzymes include human ⁇ -2-mannosidase 1B (AAC26169), a combination of mammalian ER mannosidases, or a filamentous fungal enzyme such as ⁇ -1,2-mannosidase (MDS1) ( T. reesei AAF34579; Maras M et al J Biotech.
  • the catalytic domain of the mannosidase is typically fused with a targeting peptide, such as HDEL, KDEL, or part of an ER or early Golgi protein, or expressed with an endogenous ER targeting structures of an animal or plant mannosidase I enzyme.
  • a targeting peptide such as HDEL, KDEL, or part of an ER or early Golgi protein, or expressed with an endogenous ER targeting structures of an animal or plant mannosidase I enzyme.
  • the filamentous fungal cell may also further contain a polynucleotide encoding a galactosyltransferase.
  • Galactosyltransferases transfer ⁇ -linked galactosyl residues to terminal N-acetylglucosaminyl residue.
  • the galactosyltransferase is a ⁇ -1,4-galactosyltransferase.
  • ⁇ -1,4-galactosyltransferases belong to the CAZy glycosyltransferase family 7 (cazy.org/GT7_all.html) and include ⁇ -N-acetylglucosaminyl-glycopeptide ⁇ -1,4-galactosyltransferase (EC 2.4.1.38), which is also known as N-acetylactosamine synthase (EC 2.4.1.90).
  • Useful subfamilies include ⁇ 4-GalT1, ⁇ 4-GalT-II, -III, -IV, -V, and -VI, such as mammalian or human ⁇ 4-GalTI or ⁇ 4GalT-II, -III, -IV, -V, and -VI or any combinations thereof.
  • ⁇ 4-GalT1, ⁇ 4-GalTII, or ⁇ 4-GalTIII are especially useful for galactosylation of terminal GlcNAc ⁇ 2-structures on N-glycans such as GlcNAcMan3, GlcNAc2Man3, or GlcNAcMan5 (Guo S. et al. Glycobiology 2001, 11:813-20).
  • the three-dimensional structure of the catalytic region is known (e.g. (2006) J. Mol. Biol. 357: 1619-1633), and the structure has been represented in the PDB database with code 2FYD.
  • the CAZy database includes examples of certain enzymes. Characterized enzymes are also listed in the CAZy database at malariay.org/GT7_characterized.html. Examples of useful ⁇ 4GalT enzymes include ⁇ 4GalT1, e.g. bovine Bos taurus enzyme AAA30534.1 (Shaper N. L. et al Proc. Natl. Acad. Sci. U.S.A. 83 (6), 1573-1577 (1986)), human enzyme (Guo S. et al.
  • ⁇ 4GalTII enzymes such as human ⁇ 4GalTII BAA75819.1, Chinese hamster Cricetulus griseus AAM77195, Mus musculus enzyme BAA34385, and Japanese Medaka fish Oryzias latipes BAH36754
  • ⁇ 4GalTIII enzymes such as human ⁇ 4GalTIII BAA75820.1, Chinese hamster Cricetulus griseus AAM77196 and Mus musculus enzyme AAF22221.
  • the galactosyltransferase may be expressed in the plasma membrane of the host cell.
  • a heterologous targeting peptide such as a Kre2 peptide described in Schwientek J. Biol. Chem 1996 3398, may be used.
  • Promoters that may be used for expression of the galactosyltransferase include constitutive promoters such as gpd, promoters of endogenous glycosylation enzymes and glycosyltransferases such as mannosyltransferases that synthesize N-glycans in the Golgi or ER, and inducible promoters of high-yield endogenous proteins such as the cbh1 promoter.
  • the filamentous fungal cell contains a polynucleotide encoding a galactosyltransferase
  • the filamentous fungal cell also contains a polynucleotide encoding a UDP-Gal 4 epimerase and/or UDP-Gal transporter.
  • lactose may be used as the carbon source instead of glucose when culturing the host cell.
  • the culture medium may be between pH 4.5 and 7.0 or between 5.0 and 6.5.
  • the filamentous fungal cell contains a polynucleotide encoding a galactosyltransferase and a polynucleotide encoding a UDP-Gal 4 epimerase and/or UDP-Gal transporter
  • a divalent cation such as Mn2+, Ca2+ or Mg2+ may be added to the cell culture medium.
  • the filamentous fungal cell of the invention for example, selected among Neurospora, Trichoderma, Myceliophthora or Chrysosporium cell, and more preferably Trichoderma reesei cell, may comprise the following features:
  • recombinant enzymes such as ⁇ 1,2 mannosidases, GnTI, or other glycosyltransferases introduced into the filamentous fungal cells, include a targeting peptide linked to the catalytic domains.
  • the term “linked” as used herein means that two polymers of amino acid residues in the case of a polypeptide or two polymers of nucleotides in the case of a polynucleotide are either coupled directly adjacent to each other or are within the same polypeptide or polynucleotide but are separated by intervening amino acid residues or nucleotides.
  • a “targeting peptide”, as used herein, refers to any number of consecutive amino acid residues of the recombinant protein that are capable of localizing the recombinant protein to the endoplasmic reticulum (ER) or Golgi apparatus (Golgi) within the host cell.
  • the targeting peptide may be N-terminal or C-terminal to the catalytic domains. In certain embodiments, the targeting peptide is N-terminal to the catalytic domains.
  • the targeting peptide provides binding to an ER or Golgi component, such as to a mannosidase II enzyme. In other embodiments, the targeting peptide provides direct binding to the ER or Golgi membrane.
  • Components of the targeting peptide may come from any enzyme that normally resides in the ER or Golgi apparatus.
  • Such enzymes include mannosidases, mannosyltransferases, glycosyltransferases, Type 2 Golgi proteins, and MNN2, MNN4, MNN6, MNN9, MNN10, MNS1, KRE2, VAN1, and OCH1 enzymes.
  • Such enzymes may come from a yeast or fungal species such as those of Acremonium, Aspergillus, Aureobasidium, Cryptococcus, Chrysosporium, Chrysosporium lucknowense, Filobasidium, Fusarium, Gibberella, Humicola, Magnaporthe, Mucor, Myceliophthora, Myrothecium, Neocallimastix, Neurospora, Paecilomyces, Penicillium, Piromyces, Schizophyllum, Talaromyces, Thermoascus, Thielavia, Tolypocladium , and Trichoderma . Sequences for such enzymes can be found in the GenBank sequence database.
  • the targeting peptide comes from the same enzyme and organism as one of the catalytic domains of the recombinant protein.
  • the targeting peptide of the recombinant protein is from the human GnTII enzyme.
  • the targeting peptide may come from a different enzyme and/or organism as the catalytic domains of the recombinant protein.
  • Examples of various targeting peptides for use in targeting proteins to the ER or Golgi that may be used for targeting the recombinant enzymes include: Kre2/Mnt1 N-terminal peptide fused to galactosyltransferase (Schwientek, J B C 1996, 3398), HDEL for localization of mannosidase to ER of yeast cells to produce Man5 (Chiba, J B C 1998, 26298-304; Callewaert, FEBS Lett 2001, 173-178), OCH1 targeting peptide fused to GnTI catalytic domain (Yoshida et al, Glycobiology 1999, 53-8), yeast N-terminal peptide of Mns1 fused to ⁇ 2-mannosidase (Martinet et al, Biotech Lett 1998, 1171), N-terminal portion of Kre2 linked to catalytic domain of GnTI or ⁇ 4GalT (Vervecken, Appl.
  • the targeting peptide is an N-terminal portion of the Mnt1/Kre2 targeting peptide having the amino acid sequence of SEQ ID NO: 40 (for example encoded by the polynucleotide of SEQ ID NO:41).
  • the targeting peptide is selected from human GNT2, KRE2, KRE2-like, Och1, Anp1, Van1 as shown in the Table 1 below:
  • sequences that may be used for targeting peptides include the targeting sequences as described in WO2012/069593.
  • Uncharacterized sequences may be tested for use as targeting peptides by expressing enzymes of the glycosylation pathway in a host cell, where one of the enzymes contains the uncharacterized sequence as the sole targeting peptide, and measuring the glycans produced in view of the cytoplasmic localization of glycan biosynthesis (e.g. as in Schwientek J B C 1996 3398), or by expressing a fluorescent reporter protein fused with the targeting peptide, and analysing the localization of the protein in the Golgi by immunofluorescence or by fractionating the cytoplasmic membranes of the Golgi and measuring the location of the protein.
  • filamentous fungal cells as described above are useful in methods for producing a protein having reduced O-mannosylation.
  • the invention relates to a method for producing a protein having reduced O-mannosylation, comprising:
  • the produced protein has reduced O-mannosylation due to said mutation in said PMT gene as described in the previous sections.
  • the PMT-deficient Trichoderma cell may optionally have reduced endogenous protease activity as described in the previous sections.
  • the filamentous fungal cells and methods of the invention are useful for the production of protein with serine or threonine which may be O-mannosylated.
  • it is particularly useful for the production of protein which are O-mannosylated when produced in a parental PMT-functional filamentous fungal host cell, for example, in at least one Trichoderma cell which is wild type for PMT1 gene, such as SEQ ID NO:1.
  • certain growth media include, for example, common commercially-prepared media such as Luria-Bertani (LB) broth, Sabouraud Dextrose (SD) broth or Yeast medium (YM) broth.
  • LB Luria-Bertani
  • SD Sabouraud Dextrose
  • YM Yeast medium
  • Other defined or synthetic growth media may also be used and the appropriate medium for growth of the particular host cell will be known by someone skilled in the art of microbiology or fermentation science.
  • Culture medium typically has the Trichoderma reesei minimal medium (Penttila et al., 1987, Gene 61, 155-164) as a basis, supplemented with substances inducing the production promoter such as lactose, cellulose, spent grain or sophorose.
  • the pH of cell culture is between 3.5 and 7.5, between 4.0 and 7.0, between 4.5 and 6.5, between 5 and 5.5, or at 5.5.
  • the filamentous fungal cell or Trichoderma fungal cell is cultured at a pH range selected from 4.7 to 6.5; pH 4.8 to 6.0; pH 4.9 to 5.9; and pH 5.0 to 5.8.
  • the protein which may be O-mannosylated is a heterologous protein, preferably a mammalian protein.
  • the heterologous protein is a non-mammalian protein.
  • the protein which may be O-mannosylated is a glycoprotein with N-glycan posttranslational modifications.
  • a mammalian protein which may be O-mannosylated is selected from an immunoglobulin, immunoglobulin or antibody heavy or light chain, a monoclonal antibody, a Fab fragment, an F(ab′)2 antibody fragment, a single chain antibody, a monomeric or multimeric single domain antibody, a camelid antibody, or their antigen-binding fragments.
  • a fragment of a protein consists of at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100 consecutive amino acids of a reference protein.
  • an “immunoglobulin” refers to a multimeric protein containing a heavy chain and a light chain covalently coupled together and capable of specifically combining with antigen.
  • Immunoglobulin molecules are a large family of molecules that include several types of molecules such as IgM, IgD, IgG, IgA, and IgE.
  • an “antibody” refers to intact immunoglobulin molecules, as well as fragments thereof which are capable of binding an antigen. These include hybrid (chimeric) antibody molecules (see, e.g., Winter et al. Nature 349:293-99225, 1991; and U.S. Pat. No. 4,816,567 226); F(ab′)2 molecules; non-covalent heterodimers; dimeric and trimeric antibody fragment constructs; humanized antibody molecules (see e.g., Riechmann et al. Nature 332, 323-27, 1988; Verhoeyan et al.
  • the antibodies are classical antibodies with Fc region. Methods of manufacturing antibodies are well known in the art.
  • the yield of the mammalian glycoprotein is at least 0.5, at least 1, at least 2, at least 3, at least 4, or at least 5 grams per liter.
  • the mammalian glycoprotein is an antibody, optionally, IgG1, IgG2, IgG3, or IgG4.
  • the yield of the antibody is at least 0.5, at least 1, at least 2, at least 3, at least 4, or at least 5 grams per liter.
  • the mammalian glycoprotein is an antibody, and the antibody contains at least 70%, at least 80%, at least 90%, at least 95%, or at least 98% of a natural antibody C-terminus and N-terminus without additional amino acid residues.
  • the mammalian glycoprotein is an antibody, and the antibody contains at least 70%, at least 80%, at least 90%, at least 95%, or at least 98% of a natural antibody C-terminus and N-terminus that do not lack any C-terminal or N-terminal amino acid residues.
  • the culture containing the mammalian glycoprotein contains polypeptide fragments that make up a mass percentage that is less than 50%, less than 40%, less than 30%, less than 20%, or less than 10% of the mass of the produced polypeptides.
  • the mammalian glycoprotein is an antibody
  • the polypeptide fragments are heavy chain fragments and/or light chain fragments.
  • the culture containing the antibody contains free heavy chains and/or free light chains that make up a mass percentage that is less than 50%, less than 40%, less than 30%, less than 20%, or less than 10% of the mass of the produced antibody.
  • the culture contains at least 70%, 80%, 90%, 95% or 100% of the proteins that is not O-mannosylated (mol %, as determined for example by MALDI TOF MS analysis, and measuring area or intensity of peaks as described in the Example 1 below).
  • the culture further comprises at least 5%, 10%, 15%, 20%, 25%, 30% of secreted complex neutral N-glycans (mol %) compared to total secreted neutral N-glycans (as measured for example as described in WO2012069593).
  • the heterologous protein with reduced O-mannosylation comprises the trimannosyl N-glycan structure Man ⁇ 3[Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc.
  • the Man ⁇ 3[Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc structure represents at least 20%, 30%; 40%, 50%; 60%, 70%, 80% (mol %) or more, of the total N-glycans of the heterologous protein with reduced O-mannosylation.
  • the heterologous protein with reduced O-mannosylation comprises the G0 N-glycan structure GlcNAc ⁇ 2Man ⁇ 3[GlcNAc ⁇ 2Man ⁇ 6]Man ⁇ 4GlcNAc ⁇ 4GlcNAc.
  • the non-fucosylated G0 glycoform structure represents at least 20%, 30%; 40%, 50%; 60%, 70%, 80% (mol %) or more, of the total N-glycans of the heterologous protein with reduced O-mannosylation.
  • galactosylated N-glycans represents less (mol %) than 0.5%, 0.1%, 0.05%, 0.01% of total N-glycans of the culture, and/or of the heterologous protein with reduced O-mannosylation, for example an antibody.
  • the culture or the heterologous protein, for example an antibody comprises no galactosylated N-glycans.
  • the heterologous (purified) protein is an antibody, a light chain antibody, a heavy chain antibody or a Fab, that comprises Man3, GlcNAcMan3, Man5, GlcNAcMan5, G0, core G0, G1, or G2 N-glycan structure as major glycoform and less than 35%, 20%, 17%, 15%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1%, or less than 0.5% of O-mannosylation level (as mole % as determined for example by MALDI TOF MS analysis, and measuring area or intensity of peaks as described in Example 1).
  • the invention therefore relates to a method for producing an antibody having reduced O-mannosylation, comprising:
  • At least 70%, 80%, 90%, 95%, 97%, 98%, 99% or 100% of the produced antibody is not O-mannosylated (mol %, as determined for example by MALDI TOF MS analysis, and measuring area or intensity of peaks as described in Example 1.
  • the method includes the further step of providing one or more, two or more, three or more, four or more, or five or more protease inhibitors.
  • the protease inhibitors are peptides that are co-expressed with the mammalian polypeptide.
  • the inhibitors inhibit at least two, at least three, or at least four proteases from a protease family selected from aspartic proteases, trypsin-like serine proteases, subtilisin proteases, and glutamic proteases.
  • the filamentous fungal cell or Trichoderma fungal cell also contains a carrier protein.
  • a “carrier protein” is portion of a protein that is endogenous to and highly secreted by a filamentous fungal cell or Trichoderma fungal cell.
  • Suitable carrier proteins include, without limitation, those of T. reesei mannanase I (Man5A, or MANI), T. reesei cellobiohydrolase II (CeI6A, or CBHII) (see, e.g., Paloheimo et al Appl. Environ. Microbiol. 2003 December; 69(12): 7073-7082) or T.
  • the carrier protein is CBH1.
  • the carrier protein is a truncated T. reesei CBH1 protein that includes the CBH1 core region and part of the CBH1 linker region.
  • a carrier such as a cellobiohydrolase or its fragment is fused to an antibody light chain and/or an antibody heavy chain.
  • a carrier-antibody fusion polypeptide comprises a Kex2 cleavage site.
  • Kex2, or other carrier cleaving enzyme is endogenous to a filamentous fungal cell.
  • carrier cleaving protease is heterologous to the filamentous fungal cell, for example, another Kex2 protein derived from yeast or a TEV protease. In certain embodiments, carrier cleaving enzyme is overexpressed. In certain embodiments, the carrier consists of about 469 to 478 amino acids of N-terminal part of the T. reesei CBH1 protein GenBank accession No. EGR44817.1.
  • the filamentous fungal cell of the invention overexpress KEX2 protease.
  • the heterologous protein is expressed as fusion construct comprising an endogenous fungal polypeptide, a protease site such as a Kex2 cleavage site, and the heterologous protein such as an antibody heavy and/or light chain.
  • Useful 2-7 amino acids combinations preceding Kex2 cleavage site have been described, for example, in Mikosch et al. (1996) J. Biotechnol. 52:97-106; Goller et al. (1998) Appl Environ Microbiol. 64:3202-3208; Spencer et al. (1998) Eur. J. Biochem.
  • the invention further relates to the protein composition, for example the antibody composition, obtainable or obtained by the method as disclosed above.
  • such antibody composition obtainable or obtained by the methods of the invention, comprises at least 70%, 80%, 90%, 95%, or 100% of the antibodies that are not O-mannosylated (mol %, as determined for example by MALDI TOF MS analysis, and measuring area or intensity of peaks as described in Example 1).
  • such antibody composition further comprises as 50%, 60%, 70% or 80% (mole % neutral N-glycan), of the following glycoform:
  • the N-glycan glycoform according to iii-v comprises less than 15%, 10%, 7%, 5%, 3%, 1% or 0.5% or is devoid of Man5 glycan as defined in i) above.
  • the invention also relates to a method of reducing O-mannosylation level of a recombinant glycoprotein composition produced in a Trichoderma cell, said method consisting of using a Trichoderma cell having a mutation in a PMT gene wherein said PMT gene is either:
  • said Trichoderma cell is Trichoderma reesei.
  • said recombinant glycoprotein comprises at least a light chain antibody or its fragments comprising at least one serine or threonine residue and with at least one N-glycan.
  • Example 2 As more specifically exemplified in Example 2, after deletion of pmt1, almost 95% of purified mAb and 70% of Fab molecules no longer contained any O-mannose residues. In contrast, as exemplified in Examples 3 to 4, O-mannosylation level analysis performed on pmt2 and pmt3 deletion strains did not exhibit any appreciable reduction in O-mannosylation. Together with the titer and growth analysis set forth in Example 2, these results demonstrate that filamentous fungal cells, such as Trichoderma cells, can be genetically modified to reduce or suppress O-mannosylation activity, without adversely affecting viability and yield of produced glycoproteins. As such, pmt1 is identified a valuable target to reduce O-mannosylation of secreted proteins and to improve product quality of biopharmaceuticals produced by Trichoderma reesei.
  • pmt1 is a valuable target to reduce O-mannosylation of secreted proteins and to improve product quality of biopharmaceuticals produced by Trichoderma reesei.
  • plasmids Three different deletion plasmids (pTTv36, pTTv124, pTTv185) were constructed for deletion of the protein O-mannosyltransferase gene pmt1 (TrelD75421). All the plasmids contain the same 5′ and 3′ flanking regions for correct integration to the pmt1 locus.
  • pTTv36 contains a gene encoding acetamidase of Aspergillus nidulans (amdS)
  • pTTv124 contains a loopout version (blaster cassette) of the amdS marker
  • pTTv185 a loopout version (blaster cassette) of a gene encoding orotidine-5′-monophosphate (OMP) decarboxylase of T. reesei (pyr4).
  • the third deletion construct, pTTv185, for the protein O-mannosyltransferase gene pmt1 was designed to enable removal of the selection marker from the Trichoderma reesei genome after successful integration and thereby recycling of the selection marker for subsequent transformations.
  • the recycling of the marker i.e. removal of pyr4 gene from the deletion construct, resembles so called blaster cassettes developed for yeasts (Hartl, L. and Seiboth, B., 2005, Curr Genet 48:204-211; and Alani, E. et al., 1987, Genetics 116:541-545).
  • the TrelD number refers to the identification number of a particular protease gene from the Joint Genome Institute Trichoderma reesei v2.0 genome database. Primers for construction of deletion plasmids were designed either manually or using Primer3 software (Primer3 website, Rozen and Skaletsky (2000) Bioinformatics Methods and Protocols: Methods in Molecular Biology . Humana Press, Totowa, N.J., pp 365-386).
  • the principle of the blaster cassette using pyr4 as the marker gene is as follows: pyr4, encoding orotidine-5′-monophosphate (OMP) decarboxylase of T. reesei (Smith, J. L., et al., 1991, Current Genetics 19:27-33) is needed for uridine synthesis. Strains deficient for OMP decarboxylase activity are unable to grow on minimal medium without uridine supplementation (i.e. are uridine auxotrophs).
  • OMP orotidine-5′-monophosphate
  • 5-fluoroorotic acid in generation of mutant strains lacking OMP decarboxylase activity (pyr4 ⁇ strains) is based on the conversion of 5-FOA to a toxic intermediate 5-fluoro-UMP by OMP decarboxylase. Therefore, cells which have a mutated pyr4 gene are resistant to 5-FOA, but in addition are also auxotrophic for uridine.
  • the 5-FOA resistance can in principle result also from a mutation in another gene (pyr2, orotate phosphoribosyltransferase), and therefore the spontaneous mutants obtained with this selection need to be verified for the pyr4 ⁇ genotype by complementing the mutant with the pyr4 gene.
  • the pyr4 gene can be used as an auxotrophic selection marker in T. reesei .
  • pyr4 is followed by a 310 by direct repeat of pyr4 5′ untranslated region (5′UTR) and surrounded by 5′ and 3′ flanking regions of the gene to be deleted. Integration of the deletion cassette is selected via the pyr4 function. Removal of the pyr4 marker is then forced in the presence of 5-FOA by recombination between the two homologous regions (direct repeat of 5′UTR) resulting in looping out of the selection marker and enabling the utilisation of the same blaster cassette (pyr4 loopout) in successive rounds of gene deletions. After looping out, only the 310 bp sequence of 5′UTR remains in the locus.
  • the pyr4 selection marker and the 5′ direct repeat (DR) fragment (310 bp of pyr4 5′UTR) were produced by PCR using plasmid containing a genomic copy of T. reesei pyr4 as a template. Both fragments contained 40 bp overlapping sequences needed to clone the plasmid with the loopout cassette using homologous recombination in yeast (see below). To enable possible additional cloning steps, an AscI digestion site was placed between the pyr4 marker and the 5′ direct repeat and NotI sites to surround the complete blaster cassette.
  • flanking region fragments were produced by PCR using a T. reesei wild type strain QM6a (ATCC13631) as the template.
  • T. reesei wild type strain QM6a ATCC13631
  • overlapping sequences for the vector and the selection marker were placed to the appropriate PCR-primers.
  • NotI restriction sites were introduced between the flanking regions and the selection marker.
  • PmeI restriction sites were placed between the vector and the flanking regions for removal of vector sequence prior to transformation into T. reesei .
  • Vector backbone pRS426 was digested with restriction enzymes (EcoRI and XhoI).
  • First deletion plasmid for pmt1 (plasmid pTTv36, Table 2) used amdS, a gene encoding acetamidase of Aspergillus nidulans , as the marker.
  • the marker cassette was digested from an existing plasmid pHHO1 with NotI. All fragments used in cloning were separated with agarose gel electrophoresis and correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods.
  • the vector backbone and the appropriate marker and flanking region fragments were transformed into Saccharomyces cerevisiae (strain H3488/FY834).
  • the yeast transformation protocol was based on the method for homologous yeast recombination described in the Neurospora knockouts workshop material of Colot and Collopy, (Dartmouth Neurospora genome protocols website), and the Gietz laboratory protocol (University of Manitoba, Gietz laboratory website).
  • the plasmid DNA from the yeast transformants was rescued by transformation into Escherichia coli . A few clones were cultivated, plasmid DNA was isolated and digested to screen for correct recombination using standard laboratory methods. A few clones with correct insert sizes were sequenced and stored.
  • the amdS marker was removed from the deletion plasmid pTTv36 with NotI digestion and replaced by another variant of the blaster cassette, amdS loopout cassette containing the amdS selection marker gene, followed by AscI restriction site and a 300 bp direct repeat of amdS 5′UTR.
  • the amdS blaster cassette functions in a similar manner to the pyr4 blaster cassette. The clones containing the amdS blaster cassette are able to grow on acetamide as sole nitrogen source.
  • a functional amdS gene will convert 5-FAA to a toxic fluoroacetate and therefore, in the presence of 5-FAA, removal of amdS gene is beneficial to the fungus. Removal of amdS blaster cassette is enhanced via the 300 bp DRs in the cassette like in the pyr4 blaster cassette, which enables the amdS gene to loop out via single crossover between the two DRs. Resulting clones are resistant to 5-FAA and unable to grow on acetamide as the sole nitrogen source.
  • the fragments needed for the amdS blaster cassette were produced by PCR using a plasmid p3SR2 (Hynes M. J. et al, 1983, Mol. Cell. Biol. 3:1430-1439) containing a genomic copy of the amdS gene as the template.
  • a plasmid p3SR2 containing a genomic copy of the amdS gene as the template.
  • yeast homologous recombination system used in cloning see above
  • NotI restriction sites were kept between the flanking regions and the blaster cassette. Additional restriction sites FseI and AsiSI were introduced to the 5′ end of amdS and an AscI site between amdS and amdS 5′DR.
  • the plasmid pTTv124 was constructed using the yeast recombination system described above.
  • the plasmid DNA from the yeast transformants was rescued by transformation into Escherichia coli .
  • a few clones were cultivated, plasmid DNA was isolated and digested to screen for correct recombination using standard laboratory methods.
  • a few clones with correct insert sizes were sequenced and stored.
  • the amdS marker was removed from the deletion plasmid pTTv36 with NotI digestion and replaced by the pyr4 blaster cassette described above.
  • the pyr4 blaster cassette was obtained from another plasmid with NotI digestion, ligated to NotI cut pTTv36 and transformed into E. coli using standard laboratory methods. A few transformants were cultivated, plasmid DNA isolated and digested to screen for correct ligation and orientation of the pyr4 blaster cassette using standard laboratory methods. One clone with correct insert size and orientation was sequenced and stored.
  • T. reesei strain M304 comprises MAB01 light chain fused to T. reesei truncated CBH1 carrier with NVISKR Kex2 cleavage sequence, MAB01 heavy chain fused to T.
  • Purified clones were sporulated on plates containing 39 g/l potato dextrose agarose. These clones were tested for uridine auxotrophy by plating spores onto minimal medium plates (20 g/l glucose, 1 ml/l Triton X-100, pH 4.8) with and without 5 mM uridine supplementation. No growth was observed on plates without uridine indicating the selected clones were putative pyr4 ⁇ . Clones were stored for future use and one of them was designated with strain number M317.
  • Pmt1 was deleted from M317 (pyr4 ⁇ of the strain M304) using the pmt1 deletion cassette from plasmid pTTv185 described above.
  • plasmid pTTv185 ⁇ pmt1-pyr4 was digested with PmeI+XbaI and the correct fragment was purified from an agarose gel using QIAquick Gel Extraction Kit (Qiagen). Approximately 5 ⁇ g of the pmt1 deletion cassette was used to transform strain M317.
  • Preparation of protoplasts and transformation for pyr4 selection were carried out essentially according to methods in Penttilä et al. (1987, Gene 61:155-164) and Gruber et al (1990 , Curr. Genet. 18:71-76).
  • Southern analyses were essentially performed according to the protocol for homologous hybridizations in Sambrook et al. (1989, Molecular Cloning: A laboratory manual. 2 nd Ed., Cold Spring Harbor Laboratory Press) using radioactive labeling ( 32 P-dCTP) and DecaLabel Plus kit (Fermentas). Southern digestion schemes were designed using Geneious Pro software (Geneious website). Fragments for probes were produced by PCR using the primers listed in Table 4 using a T. reesei wild type strain QM6a (ATCC13631) as the template. PCR products were separated with agarose gel electrophoresis and correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods.
  • O-mannosylation status analysis was performed to shake flask cultivations of T. reesei M304, eight pmt1 disruptants (pTTv185: 26-8A, 26-8B, 26-16A, 26-16B, 26-19A, 26-19B, 26-21A, 26-21B). All were cultivated in TrMM—40 g/l lactose—20 g/l SGE—100 mM PIPPS—9 g/l casamino acids, pH 5.5 at +28° C. and samples were taken on time point days 3, 5, 7 and 10.
  • MAB01 antibody from each sample from day 7 was purified from supernatants using Protein G HP MultiTrap 96-well plate (GE Healthcare) according to manufacturer's instructions. The antibody was eluted with 0.1 M citrate buffer, pH 2.6 and neutralized with 2 M Tris, pH 9. The concentration was determined via UV absorbance in spectrophotometer against MAB01 standard curve. For O-mannosylation analysis, 10 ⁇ g of protein was incubated in 6 M Guanidinium HCl for 30 minutes at +60° C. after which 5 ⁇ l of fresh 0.1 M DTT was added and incubated again as above. The samples were purified using Poros R1 96-well plate and the resulting light chains were analysed using MALDI-TOF MS. All were made as duplicates.
  • FIG. 2 Spectra of light chain of flask cultured parental T. reesei strain M317 (pyr4 ⁇ of M304) (A) and ⁇ pmt1 disruptant clone 26-8A (B), day 7).
  • Fermentation was carried out with ⁇ pmt1 strain M403 (clone 26-8A; pTTv185 in M317). Fermentation culture medium contained 30 g/l glucose, 60 g/l lactose, 60 g/l whole spent grain at pH 5.5. Lactose feed was started after glucose exhaustion. Growth temperature was shifted from +28° C. to +22° C. after glucose exhaustion. Samples were collected by vacuum filtration. Supernatant samples were stored to ⁇ 20° C.
  • FIG. 3 is shown the Western analyses of supernatant samples.
  • MAB01 heavy and light chains were detected from supernatant after day three.
  • pmt1 that could also reduce O-mannosylation of the linker and thus aid KEX2 cleavage
  • substantial amount of light chain remains attached to the carrier in the early days of the fermentation.
  • the cleavage is more complete but the yield may be affected by the degradation of the heavy chain.
  • Results on antibody titres (Table 7 below) indicate fairly steady expression between days 7 to 10.
  • the pmt1 deletion strain produced approximately equal antibody levels as the parental strain. Higher titres were obtained when the same strain was fermented using a different fermenter.
  • M403 (clone 26-8A) was cultivated in fermenter in TrMM, 30 g/l glucose, 60 g/l lactose, 60 g/l spent grain, pH 5.5 with lactose feed. Samples were harvested on days 2, 3 and 5-11. 0-mannosylation level analysis was performed as to flask cultures. The O-mannosylation status was greatly decreased also in fermenter culture ( FIG. 4 , Table 5).
  • the O-mannosylation level was calculated from average of area and intensity (Table 5). Area (Table 6) seems to give more commonly higher rate of non-O-glycosylated LC than intensity (Table 7). In all time points the O-mannosylation level was below 5%.
  • pmt1 Deletion of pmt1 diminished dramatically MAB01 0-mannosylation; the amount of O-mannosylated LC was ⁇ 61% in parental strain, 3% in the best ⁇ pmt1 clone in shake flask culture and practically 0% in fermenter culture in time point day 9.
  • pmt1 disruption cassette (pmt1 amdS) was released from its backbone vector pTTv124 described above by restriction digestion and purified through gel extraction. Using protoplast transformation the deletion cassette was introduced to T. reesei strains M304 (3-fold protease deletion strain expressing MAB01) and M307 (4-fold protease deletion strain ⁇ pep1 ⁇ tsp1 ⁇ slp1 ⁇ gap1, also described in PCT/EP2013/050126 that has been transformed to express a Fab). Transformants were plated to acetamidase selective medium (minimal medium containing acetamide as the sole carbon source).
  • Transformants were screened by PCR for homologous integration of the acetamidase marker to the pmt1 locus using a forward primer outside the 5′ flanking region fragment of the construct and the reverse primer inside the AmdS selection marker (5′ integration) as well as a forward primer inside the AmdS selection marker and a reverse primer outside the 3′ flanking region fragment (3′ integration).
  • Three independent transformants of each transformation (MAB01 and Fab expressing strains), which gave PCR results displaying correct integration of the construct to the pmt1 locus were selected for single spore purification to obtain uninuclear clones.
  • strains were grown in batch fermentations for 7 days, in media containing 2% yeast extract, 4% cellulose, 4% cellobiose, 2% sorbose, 5 g/L KH2PO4, and 5 g/L (NH4)2SO4.
  • Culture pH was controlled at pH 5.5 (adjusted with NH4OH). The starting temperature was 30° C., which was shifted to 22° C. after 48 hours.
  • mAb fermentations (strains M304, M403, M406 and M407) were carried out in 4 parallel 2 L glas reactor vessels (DASGIP) with a culture volume of 1 L and the Fab fermentation (TR090#5) was done in a 15 L steel tank reactor (Infors) with a culture volume of 6 L.
  • Fab strains (TR090#5, TR090#3, TR090#17) were additionally cultured in shake flasks for 4 days at 28° C.
  • Main media components were 1% yeast extract, 2% cellobiose, 1% sorbose, 15 g/L KH2PO4 and 5 g/L (NH4)2SO4 and the pH was uncontrolled (pH drops from 5.5 to ⁇ 3 during a time course of cultivation). Culture supernatant samples were taken during the course of the runs and stored at ⁇ 20° C. Samples were collected daily from the whole course of these cultivations, and production levels were analyzed by affinity liquid chromatography. Samples with maximum production levels were subject to purification and further O-mannosylation analysis.
  • O-mannosylation was analyzed on mAb and Fab molecules expressed from both, the pmt1 deletion and parental strains.
  • the mAb and Fab was purified from culture supernatants using Lambda Select Sure and CaptureSelect Fab Lambda (BAC) affinity chromatography resin, respectively, applying conditions as described by the manufactures protocols. Both purified molecules including, the purified mAb and Fab were subjected to RP-LC-QTOF-MS either as intact and/or reduced/alkylated samples.
  • Reversed-phase chromatography separation was carried out on a 2.1 ⁇ 150 mm Zorbax C3 column packed with 5 ⁇ m particles, 300 ⁇ pore size the eluents were: eluent A 0.1% TFA in water and eluent B 0.1% TFA in 70% IPA, 20% ACN, 10% water.
  • the column was heated at 75° C. and the flo rate was 200 ⁇ L/min.
  • the gradient used for the sample separation is shown in Table 9.
  • the HPLC was directly coupled with a Q-TOF Ultima mass spectrometer (Waters, Manchester, UK).
  • the ESI-TOF mass spectrometer was set to run in positive ion mode.
  • the data evaluation of intact and reduced/alkylated analyses was performed using MassLynx analysis software (Waters, Manchester, UK).
  • the deconvolution of the averaged mass spectra from the main UV signals was carried out using the MaxEnt algorithm, a part of the MassLynx analysis software (Waters, Manchester, UK).
  • the deconvolution parameters were the following: “max numbers of iterations” are 8; resolution is 0.1 Da/channel; Uniform Gaussian—width at half height is 1 Da for intact and 0.5 for the reduced chains and minimum intensity ratios are left 30% and right 30%.
  • the estimated level of O-mannosylation was determined using the peak signal height after deconvolution.
  • the observed O-mannosylation levels (%) of mAbs and Fabs from independent pmt1 deletion strains are compared to the ones of the respective parental wild-type strains in Tables 10 and 11.
  • the O-mannosylation level was found to be 70% on intact Fab derived from the parental strain and reduced to ⁇ 34% in all three pmt1 deletion strains.
  • the transfer of mannoses was more efficiently diminished on the Fab light chains (10% of residual O-mannosylation on light chains obtained from pmt1 deletion strains vs. 59% for the parental strain), as compared to the heavy chains, for which it decreased from 43% to ⁇ 26%.
  • the O-mannosylation level was found to be 50% on the light chain of mAb derived from parental strains and reduced to 5.7-5.8% in all three pmt1 deletion strains.
  • the O-mannosylation level was found to be 4.8% on the heavy chain of mAb derived from parental strains and was completely reduced (below the limit of detection by LC-MS) in all three pmt1 deletion strains.
  • pmt1 is a valuable target to reduce O-mannosylation of secreted proteins and to improve product quality of biopharmaceuticals produced by Trichoderma reesei.
  • plasmids Three different deletion plasmids (pTTv34, pTTv122, pTTv186) were constructed for deletion of the protein O-mannosyltransferase gene pmt2 (TrelD22005). All the plasmids contain the same 5′ and 3′ flanking regions for correct integration to the pmt2 locus.
  • pTTv34 contains a gene encoding acetamidase of Aspergillus nidulans (amdS)
  • pTTv122 contains a loopout version (blaster cassette) of the amdS marker
  • pTTv186 a loopout version (blaster cassette) of a gene encoding orotidine-5′-monophosphate (OMP) decarboxylase of T. reesei (pyr4).
  • the amdS marker was removed from the deletion plasmid pTTv34 with NotI digestion and replaced by amdS blaster cassette for which the fragments were produced by PCR (see Example 1 above for details).
  • the plasmid pTTv122 was constructed using the yeast recombination system described in Example 1. The plasmid DNA from the yeast transformants was rescued by transformation into Escherichia coli . A few clones were cultivated, plasmid DNA was isolated and digested to screen for correct recombination using standard laboratory methods. A few clones with correct insert sizes were sequenced and stored.
  • the third deletion plasmid for pmt2, pTTv186 (Table 12) was cloned like the third plasmid for pmt1; the amdS blaster cassette was removed from the deletion plasmid pTTv122 with NotI digestion and replaced by the pyr4 blaster cassette described in Example 1.
  • the pyr4 blaster cassette was obtained from another plasmid with NotI digestion, ligated to NotI cut pTTv122 and transformed into E. coli using standard laboratory methods. A few transformants were cultivated, plasmid DNA isolated and digested to screen for correct ligation and orientation of the pyr4 blaster cassette using standard laboratory methods. One clone with correct insert size and orientation was sequenced and stored.
  • These deletion plasmids for pmt2 (pTTv34, pTTv122 and pTTv186, Table 12) result in 3186 bp deletion in the pmt2 locus and cover the complete coding sequence of PMT2.
  • Southern analyses were essentially performed as described in Example 1. Fragments for probes were produced by PCR using the primers listed in Table 14 using a T. reesei strain M124 as the template for the ORF probe and plasmid pTTv122 for the 5′ and 3′ flank probes. PCR products were separated with agarose gel electrophoresis and correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods.
  • Southern analyses were essentially performed as described in Example 1. Fragments for probes were produced by PCR using the primers listed in Table 16 using a T. reesei wild type strain QM6a (ATCC13631) as the template for pmt2 ORF probe and plasmid pTTv186 for 5′ and 3′ flank probes. PCR products were separated with agarose gel electrophoresis and correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods.
  • O-mannosylation level analysis was performed to pmt2 deletion strains as to flask cultures of pmt1 deletion strains. No difference was observed in O-mannosylation compared to parental strain M304.
  • plasmids Three different deletion plasmids (pTTv35, pTTv123, pTTv187) were constructed for deletion of the protein O-mannosyltransferase gene pmt3 (TrelD22527). All the plasmids contain the same 5′ and 3′ flanking regions for correct integration to the pmt3 locus.
  • pTTv35 contains a gene encoding acetamidase of Aspergillus nidulans (amdS)
  • pTTv123 contains a loopout version (blaster cassette) of the amdS marker
  • pTTv187 a loopout version (blaster cassette) of a gene encoding orotidine-5′-monophosphate (OMP) decarboxylase of T. reesei (pyr4).
  • the amdS marker was removed from the deletion plasmid pTTv35 with NotI digestion and replaced by amdS blaster cassette for which the fragments were produced by PCR (see Example 1 above for details).
  • the plasmid pTTv123 was constructed using the yeast recombination system described in Example 1.
  • the plasmid DNA from the yeast transformants was rescued by transformation into Escherichia coli .
  • a few clones were cultivated, plasmid DNA was isolated and digested to screen for correct recombination using standard laboratory methods. A few clones with correct insert sizes were sequenced and stored.
  • the third deletion plasmid for pmt3, pTTv187 (Table 17) was cloned like the third plasmid for pmt1; the amdS blaster cassette was removed from the deletion plasmid pTTv123 with NotI digestion and replaced by the pyr4 blaster cassette described in Example 1.
  • the pyr4 blaster cassette was obtained from another plasmid with NotI digestion, ligated to NotI cut pTTv123 and transformed into E. coli using standard laboratory methods. A few transformants were cultivated, plasmid DNA isolated and digested to screen for correct ligation and orientation of the pyr4 blaster cassette using standard laboratory methods. One clone with correct insert size and orientation was sequenced and stored.
  • These deletion plasmids for pmt3 (pTTv35, pTTv123 and pTTv187, Table 17) result in 2495 bp deletion in the pmt3 locus and cover the complete coding sequence of PMT3.
  • Southern analyses were essentially performed as described in Example 1. Fragments for probes were produced by PCR using the primers listed in Table 19 using a T. reesei strain M124 as the template for the ORF probe and plasmid pTTv123 for the 5′ and 3′ flank probes. PCR products were separated with agarose gel electrophoresis and correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods.
  • Southern analyses were essentially performed as described in Example 1. Fragments for probes were produced by PCR using the primers listed in Table 21 using a T. reesei wild type strain QM6a (ATCC13631) as the template for the ORF probe and plasmid pTTv187 for the 5′ and 3′ flank probes. PCR products were separated with agarose gel electrophoresis and correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods.
  • T. reesei strain M304 and eight pmt3 deletion strains (33-34S/M522, 33-34T, 33-34U, 33-34 ⁇ , 33-188A-a/M523, 33-188B-a, 33-188C-a and 33-188D-a) was carried out in Trichoderma minimal medium with 40 g/l lactose, 20 g/l spent grain extract, 100 mM PIPPS, 9 g/l casamino acids, pH 5.5 at +28° C., 800 rpm with humidity control. Samples were collected on days 3, 5 and 6 by centrifugation. Supernatant samples were stored to ⁇ 20° C. Mycelia for cell dry weight determinations were rinsed once with DDIW and dried at +100° C. for 20-24 h. Mycelia for genomic DNA extraction were rinsed twice with DDIW and stored to ⁇ 20° C.
  • O-mannosylation level analysis was performed to pmt3 deletion strains as to flask cultures of pmt1 deletion strains. No difference was observed in O-mannosylation compared to parental strain M304.
  • T. reesei pmt homologs were identified from other organisms.
  • BLAST searches were conducted using the National Center for Biotechnology Information (NCBI) non-redundant amino acid database using the Trichoderma reesei PMT amino acid sequences as queries. Sequence hits from the BLAST searches were aligned using the ClustalW2 alignment tool provided by EBI. Phylogenetic trees were generated using average distance with BLOSUM62 after aligning the sequences in the Clustal Omega alignment tool.
  • NCBI National Center for Biotechnology Information
  • FIGS. 5 and 6 A phylogenetic tree and a partial sequence alignment of the results of the PMT BLAST searches are depicted in FIGS. 5 and 6 , respectively.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
US14/902,492 2013-07-04 2014-07-03 O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof Abandoned US20160376570A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP13175141 2013-07-04
EP13175141.4 2013-07-04
PCT/EP2014/064248 WO2015001049A1 (fr) 2013-07-04 2014-07-03 Cellules fongiques filamenteuses déficientes en o-mannosyltransférase et leurs procédés d'utilisation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/064248 A-371-Of-International WO2015001049A1 (fr) 2013-07-04 2014-07-03 Cellules fongiques filamenteuses déficientes en o-mannosyltransférase et leurs procédés d'utilisation

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/124,679 Continuation US10724013B2 (en) 2013-07-04 2018-09-07 O-mannosyltransferase deficient filamentous fungal cells and methods of use thereof

Publications (1)

Publication Number Publication Date
US20160376570A1 true US20160376570A1 (en) 2016-12-29

Family

ID=48740957

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/902,492 Abandoned US20160376570A1 (en) 2013-07-04 2014-07-03 O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof
US16/124,679 Active US10724013B2 (en) 2013-07-04 2018-09-07 O-mannosyltransferase deficient filamentous fungal cells and methods of use thereof
US16/901,175 Abandoned US20200362319A1 (en) 2013-07-04 2020-06-15 O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof
US17/699,302 Pending US20220213451A1 (en) 2013-07-04 2022-03-21 O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US16/124,679 Active US10724013B2 (en) 2013-07-04 2018-09-07 O-mannosyltransferase deficient filamentous fungal cells and methods of use thereof
US16/901,175 Abandoned US20200362319A1 (en) 2013-07-04 2020-06-15 O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof
US17/699,302 Pending US20220213451A1 (en) 2013-07-04 2022-03-21 O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof

Country Status (15)

Country Link
US (4) US20160376570A1 (fr)
EP (1) EP3016970B1 (fr)
KR (1) KR102111078B1 (fr)
CN (1) CN105492457A (fr)
BR (1) BR112015032695A2 (fr)
CA (1) CA2916594C (fr)
DK (1) DK3016970T3 (fr)
ES (1) ES2739280T3 (fr)
HR (1) HRP20191069T1 (fr)
MX (1) MX364532B (fr)
PL (1) PL3016970T3 (fr)
RU (1) RU2016103373A (fr)
SG (1) SG11201510814YA (fr)
SI (1) SI3016970T1 (fr)
WO (1) WO2015001049A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10513724B2 (en) 2014-07-21 2019-12-24 Glykos Finland Oy Production of glycoproteins with mammalian-like N-glycans in filamentous fungi

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3113661A1 (fr) * 2018-09-20 2020-03-26 Perfect Day, Inc. Procedes et compositions de production de cellules fongiques filamenteuses homocaryotiques
BR112021010338A2 (pt) * 2018-11-28 2021-11-16 Novozymes As Células hospedeiras fúngicas filamentosas modificadas
CN110885810B (zh) * 2019-12-16 2021-03-23 华中农业大学 一种利用真核发酵制备得到的抑菌蛋白酶CmYC1及其应用
CN115976033B (zh) * 2022-12-14 2024-05-17 西南大学 瓜实蝇tssk1和tssk3基因及其应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009105357A1 (fr) * 2008-02-20 2009-08-27 Glycofi, Inc. Vecteurs et souches de levure pour la production de protéines
WO2013062886A1 (fr) * 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Contrôle de l'o-glycosylation dans des eucaryotes inférieurs

Family Cites Families (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4743546A (en) 1985-02-13 1988-05-10 Biotechnica International, Inc. Controlled gene excision
GB2276169A (en) 1990-07-05 1994-09-21 Celltech Ltd Antibodies specific for carcinoembryonic antigen
DE4226971C2 (de) 1992-08-14 1997-01-16 Widmar Prof Dr Tanner Modifizierte Pilzzellen und Verfahren zur Herstellung rekombinanter Produkte
AU4424597A (en) 1996-09-13 1998-04-02 Novo Nordisk Biotech, Inc. Cells having dna insertion mutations which produce altered amounts of a polypeptide
DE19754622A1 (de) 1997-12-09 1999-06-10 Antje Von Dr Schaewen Pflanzliche GntI-Sequenzen und Verwendung derselben zur Gewinnung von Pflanzen mit verminderter oder fehlender N-Acetylglucosaminyltransferase I (GnTI)-Aktivität
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
GB0021421D0 (en) 2000-08-31 2000-10-18 Oxford Glycosciences Uk Ltd Compounds
EP1379667A2 (fr) 2000-12-05 2004-01-14 The Penn State Research Foundation Procedes et compositions permettant d'obtenir des proteines heterologues dans la levure de maniere tres efficace
WO2005003296A2 (fr) 2003-01-22 2005-01-13 Human Genome Sciences, Inc. Proteines hybrides d'albumine
CN100383251C (zh) 2002-12-20 2008-04-23 荷兰联合利华有限公司 抗冻蛋白的制备
ZA200605449B (en) 2003-12-03 2007-11-28 Delta Biotechnology Ltd Interleukin-11 fusion proteins
CN102816241B (zh) 2004-02-09 2015-07-22 人类基因科学公司 清蛋白融合蛋白
WO2006036817A2 (fr) 2004-09-24 2006-04-06 Microbia, Inc. Variants fongiques et utilisations correspondantes
DK1941025T3 (da) * 2005-10-17 2011-09-26 Novozymes As Brug af svampemutanter til ekspression af antistoffer
EP1954815B1 (fr) 2005-11-15 2015-02-25 GlycoFi, Inc. Production de glycoproteines a o-glycosylation reduite
AU2007250735A1 (en) 2006-05-16 2007-11-22 Daiichi Sankyo Company, Limited Method for high-level secretory production of protein
US8680252B2 (en) 2006-12-10 2014-03-25 Dyadic International (Usa), Inc. Expression and high-throughput screening of complex expressed DNA libraries in filamentous fungi
GB0711424D0 (en) 2007-06-13 2007-07-25 Novozymes Delta Ltd Recombinant transferrin mutants
EP2594583A1 (fr) 2007-08-08 2013-05-22 Novozymes Biopharma DK A/S Variants de transferrine et conjugues
US20090253173A1 (en) * 2008-04-08 2009-10-08 Danisco Us Inc., Genencor Division Filamentous fungi with inactivated protease genes for altered protein production
MX2010012626A (es) 2008-05-20 2011-02-22 Merck Sharp & Dohme Produccion eficiente de proteinas heterologas utilizando inhibidores de manosil transferasa.
AU2009282228A1 (en) * 2008-08-12 2010-02-18 Glycofi, Inc. Improved vectors and yeast strains for protein production: Ca2+ ATPase overexpression
WO2010034708A1 (fr) 2008-09-23 2010-04-01 Unilever Plc Cassette d’expression de polynucléotide
EP2396347B1 (fr) 2009-02-11 2017-04-12 Albumedix A/S Variants et conjugés de l'albumine
DK2427486T3 (en) 2009-05-07 2015-05-26 Novozymes Biopharma Dk As A process for purifying albumin
US20120059155A1 (en) 2009-05-07 2012-03-08 Novozymes Biopharma Dk A/S Method of Controlling O-Linked Glycosylation of Antibodies
US8440456B2 (en) 2009-05-22 2013-05-14 Vib, Vzw Nucleic acids of Pichia pastoris and use thereof for recombinant production of proteins
BR112012008862A2 (pt) 2009-10-16 2019-09-24 Merck Sharp & Dohme métodos para produzir uma glicoproteína recombinante e uma eritropoietina humana madura, e, composição
EP2513290B1 (fr) 2009-12-18 2015-11-11 Novozymes Inc. Procédés de production de polypeptides dans des mutants de trichoderma présentant une carence en protéase
JP5860413B2 (ja) 2010-02-10 2016-02-16 バイオコン リミテッド タンパク質のグリコシル化を低下させる方法、その産生方法およびタンパク質
CA2788992A1 (fr) 2010-02-24 2011-09-01 Merck Sharp & Dohme Corp. Procede pour augmenter l'occupation des sites de n-glycosylation sur des glycoproteines therapeutiques produites dans pichia pastoris
MX339809B (es) 2010-05-27 2016-06-09 Merck Sharp & Dohme Corp * Metodo para preparar anticuerpos con propiedades mejoradas.
US20120135461A1 (en) 2010-07-30 2012-05-31 William James Cook Production of glycoproteins with reduced o-glycosylation comprising the use of an alpha-1,2-mannosidase
DK2643456T3 (en) 2010-11-24 2016-08-22 Glykos Finland Oy FUSION enzymes with N-ACETYLGLUKOSAMINYLTRANSFERASE ACTIVITY
JP5885191B2 (ja) 2011-01-04 2016-03-15 国立研究開発法人産業技術総合研究所 糖鎖改変酵母及びそれを用いた糖タンパク質の製造方法
CN103764837A (zh) 2011-02-25 2014-04-30 默沙东公司 用于生产具有修饰的o-糖基化的蛋白的酵母菌株
CA2834589A1 (fr) 2011-05-25 2012-11-29 Merck Sharp & Dohme Corp. Procede de preparation de polypeptides contenant fc a proprietes ameliorees
AR087433A1 (es) 2011-08-08 2014-03-26 Merck Sharp & Dohme Analogos de insulina n-glicosilados
US20140286946A1 (en) 2011-10-31 2014-09-25 Merck Sharp & Dohme Corp. Method for preparing antibodies having improved properties
WO2013066765A1 (fr) 2011-11-01 2013-05-10 Merck Sharp & Dohme Corp. Mutation de tup1 dans de la levure glycomodifiée
WO2013074598A1 (fr) 2011-11-18 2013-05-23 Merck Sharp & Dohme Corp. Polypeptides contenant fc ayant des propriétés anti-inflammatoires améliorées et une liaison améliorée à fcrn
EP2852610B1 (fr) * 2012-05-23 2018-07-11 Glykos Finland Oy Production de glycoprotéines fucosylées
US20150275222A1 (en) 2012-10-22 2015-10-01 Bo Jiang Crz1 mutant fungal cells
CA2888645A1 (fr) 2012-12-17 2014-06-26 Merck Sharp & Dohme Corp. Cellules mutantes pmt2-, och1-, pmt5-
EP2956158A4 (fr) 2013-02-13 2016-10-05 Merck Sharp & Dohme Procédés pour augmenter l'occupation des n-glycanes et réduire la production des n-glycanes hybrides dans des souches de pichia pastoris n'exprimant pas alg3
US9926570B2 (en) 2013-03-06 2018-03-27 Glaxosmithkline Llc Host cells and methods of use
US20160207978A1 (en) 2013-05-06 2016-07-21 Cell Machines Inc. Methods and compositions related to large scale production of proteins
CN105308067B (zh) 2013-06-07 2020-07-24 诺和诺德股份有限公司 制备成熟胰岛素多肽的方法
WO2015013116A1 (fr) 2013-07-25 2015-01-29 Merck Sharp & Dohme Corp. Procédé de réduction de l'étendue d'o-mannosylation de glycoprotéines
WO2015054039A1 (fr) 2013-10-08 2015-04-16 Merck Sharp & Dohme Corp. Polypeptides contenant fc présentant une liaison accrue à fcgammariib
WO2015073307A2 (fr) 2013-11-13 2015-05-21 Merck Sharp & Dohme Corp. Polypeptides contenant fc et présentant une liaison améliorée à dc-sign humaine

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009105357A1 (fr) * 2008-02-20 2009-08-27 Glycofi, Inc. Vecteurs et souches de levure pour la production de protéines
US20110312032A1 (en) * 2008-02-20 2011-12-22 Byung-Kwon Choi Vectors and yeast strains for protein production
WO2013062886A1 (fr) * 2011-10-27 2013-05-02 Merck Sharp & Dohme Corp. Contrôle de l'o-glycosylation dans des eucaryotes inférieurs

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10513724B2 (en) 2014-07-21 2019-12-24 Glykos Finland Oy Production of glycoproteins with mammalian-like N-glycans in filamentous fungi

Also Published As

Publication number Publication date
MX364532B (es) 2019-04-29
EP3016970A1 (fr) 2016-05-11
US20200362319A1 (en) 2020-11-19
CA2916594A1 (fr) 2015-01-08
MX2016000128A (es) 2016-03-01
RU2016103373A3 (fr) 2018-03-26
HRP20191069T1 (hr) 2019-11-01
US20200040314A1 (en) 2020-02-06
KR102111078B1 (ko) 2020-05-18
BR112015032695A2 (pt) 2017-10-24
DK3016970T3 (da) 2019-06-24
RU2016103373A (ru) 2017-08-10
CA2916594C (fr) 2020-03-10
US10724013B2 (en) 2020-07-28
WO2015001049A1 (fr) 2015-01-08
SG11201510814YA (en) 2016-01-28
US20220213451A1 (en) 2022-07-07
KR20160030127A (ko) 2016-03-16
PL3016970T3 (pl) 2019-09-30
EP3016970B1 (fr) 2019-05-22
CN105492457A (zh) 2016-04-13
ES2739280T3 (es) 2020-01-30
SI3016970T1 (sl) 2019-08-30

Similar Documents

Publication Publication Date Title
US20220213451A1 (en) O-Mannosyltransferase Deficient Filamentous Fungal Cells and Methods of Use Thereof
EP3172333B1 (fr) Production de glycoprotéines avec des n-glycanes de type mammifères dans des champignons filamenteux
EP3019602B1 (fr) Cellules fongiques filamenteuses déficientes en plusieurs protéases et procédés d'utilisation de celles-ci
US9695454B2 (en) Production of fucosylated glycoproteins
US9399764B2 (en) Fusion enzymes
US20110207214A1 (en) Novel tools for the production of glycosylated proteins in host cells
KR20140114860A (ko) 프로테아제 결핍 사상형 진균 세포들 및 그의 이용방법
Kainz et al. N-glycan modification in Aspergillus species
US20190276867A1 (en) Production of Glycoproteins Having Increased N-Glycosylation Site Occupancy

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NATUNEN, JARI;HILTUNEN, JUKKA;REEL/FRAME:040941/0421

Effective date: 20140407

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TEKNOLOGIAN TUTKIMUSKESKUS VTT;REEL/FRAME:040941/0605

Effective date: 20140407

Owner name: TEKNOLOGIAN TUTKIMUSKESKUS VTT, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HUUSKONEN, ANNE;SALOHEIMO, MARKKU;SIGNING DATES FROM 20140320 TO 20140324;REEL/FRAME:040941/0510

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GLYKOS FINLAND OY;REEL/FRAME:040941/0771

Effective date: 20140414

Owner name: NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD., BERMUD

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GLYKOS FINLAND OY;REEL/FRAME:040941/0771

Effective date: 20140414

Owner name: NOVARTIS PHARMA AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OSTERMEIER, CHRISTIAN;SOMMER, BENJAMIN PATRICK;WAHL, RAMON;REEL/FRAME:040941/0812

Effective date: 20140429

Owner name: NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD., BERMUD

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS PHARMA AG;REEL/FRAME:040941/0848

Effective date: 20140512

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD.;REEL/FRAME:040941/0898

Effective date: 20140616

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NOVARTIS AG;GLYKOS FINLAND OY;SIGNING DATES FROM 20160627 TO 20160630;REEL/FRAME:040941/0988

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOVARTIS PHARMA AG;REEL/FRAME:040941/0848

Effective date: 20140512

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNORS:NATUNEN, JARI;HILTUNEN, JUKKA;REEL/FRAME:052657/0623

Effective date: 20140407

Owner name: TEKNOLOGIAN TUTKIMUSKESKUS VTT, FINLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNORS:HUUSKONEN, ANNE;SALOHEIMO, MARKKU;SIGNING DATES FROM 20140320 TO 20140324;REEL/FRAME:052657/0691

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:GLYKOS FINLAND OY;REEL/FRAME:052657/0768

Effective date: 20140414

Owner name: NOVARTIS PHARMA AG, SWITZERLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNORS:OSTERMEIER, CHRISTIAN;SOMMER, BENJAMIN PATRICK;WAHL, RAMON;REEL/FRAME:052657/0809

Effective date: 20140429

Owner name: NOVARTIS AG, SWITZERLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD.;REEL/FRAME:052657/0916

Effective date: 20140616

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:TEKNOLOGIAN TUTKIMUSKESKUS VTT;REEL/FRAME:052657/0719

Effective date: 20140407

Owner name: NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD., BERMUDA

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:NOVARTIS PHARMA AG;REEL/FRAME:052657/0865

Effective date: 20140512

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:NOVARTIS PHARMA AG;REEL/FRAME:052657/0865

Effective date: 20140512

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNORS:NOVARTIS AG;GLYKOS FINLAND OY;SIGNING DATES FROM 20160627 TO 20160630;REEL/FRAME:052658/0001

Owner name: NOVARTIS INTERNATIONAL PHARMACEUTICAL LTD., BERMUDA

Free format text: NUNC PRO TUNC ASSIGNMENT;ASSIGNOR:GLYKOS FINLAND OY;REEL/FRAME:052657/0768

Effective date: 20140414

AS Assignment

Owner name: GLYKOS FINLAND OY, FINLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:NOVARTIS AG;GLYKOS FINLAND OY;SIGNING DATES FROM 20160627 TO 20160630;REEL/FRAME:056061/0610