Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MULTIPLE PROTEASES DEFICIENT FILAMENTOUS FUNGAL CELLS AND METHODS OF USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2015/004241
Kind Code:
A2
Abstract:
The present disclosure relates to compositions and methods useful for the production of heterologous proteins in filamentous fungal cells.

Inventors:
LANDOWSKI CHRISTOPHER (FI)
HUUSKONEN ANNE (FI)
WESTERHOLM-PARVINEN ANN (FI)
SALOHEIMO MARKKU (FI)
KANERVA ANNE (FI)
HILTUNEN JUKKA (FI)
Application Number:
PCT/EP2014/064820
Publication Date:
January 15, 2015
Filing Date:
July 10, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NOVARTIS AG (CH)
GLYKOS FINLAND OY (FI)
International Classes:
C12N9/14
Attorney, Agent or Firm:
CABINET PLASSERAUD (Lyon, FR)
Download PDF:
Claims:
CLAIMS

We claim: 1. A filamentous fungal cell comprising at least one endogenous protease having reduced or no protease activity, and a recombinant polynucleotide encoding a heterologous polypeptide, wherein the cell has reduced or no protease activity in one or more the following proteases: amp2, mpl, mp2, mp3, mp4, mp5, pep9, ampl, and sepl.

2. The filamentous fungal cell of Claim 1, wherein the fungal cell is a Trichoderma fungal cell, a Myceliophthora fungal cell, an Aspergillus fungal cell, a Neurospora fungal cell, a

Penicillium cell, a Fusarium cell, or a Chrysosporium fungal cell.

3. The filamentous fungal cell of any one of claims 1 to 2, wherein the total protease activity is reduced to 40% or less, preferably 6% or less, of the total protease activity of the corresponding parental filamentous fungal cell in which the proteases do not have the reduced activity.

4. The filamentous fungal cell of any preceding claim, wherein the cell further has reduced or no detectable protease activity in at least three or four proteases selected from the group consisting of pepl, pep2, pep3, pep4, pep5, pep8, pepl l, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2.

5. The filamentous fungal cell of any preceding claim, wherein the cell has reduced or no protease activity in at least eight proteases, each of the genes encoding the eight proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the eight proteases are either:

a. pepl, slpl, gapl, gap2, pep4, pep3, pep5, amp2 b. pepl, slpl, gapl, gap2, pep4, pep3, pep5, ampl, c. pepl, slpl, gapl, gap2, pep4, pep3, pep5, sepl d. pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep9, or e. pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2.

6. The filamentous fungal cell of any preceding claim, wherein the cell has reduced or no protease activity in at least nine proteases, each of the genes encoding the nine proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the nine proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl.

7. The filamentous fungal cell of any preceding claim, wherein the cell has reduced or no protease activity in at least ten proteases, each of the genes encoding the ten proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the ten proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8.

8. The filamentous fungal cell of any preceding claim, wherein the cell has reduced or no protease activity in at least eleven proteases, each of the genes encoding the eleven proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the eleven proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2.

9. The filamentous fungal cell of any preceding claim, wherein either,

(i) the cell has reduced or no protease activity in at least twelve proteases, each of the genes encoding the twelve proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the twelve proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7;

(ii) the cell has reduced or no protease activity in at least thirteen proteases, each of the genes encoding the thirteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the thirteen proteases are either

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, pep9; pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7; or, pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp3;

(iii) the cell has reduced or no protease activity in at least fourteen proteases, each of the genes encoding the fourteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fourteen proteases are pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2; or,

(iv) the cell has reduced or no protease activity in at least fifteen proteases, each of the genes encoding the fifteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fifteen proteases are either

pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mpl ; or, pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mp5. 10. The filamentous fungal cell according to Claim 9, wherein the cell has reduced or no protease activity in at least sixteen, at least seventeen, at least eighteen, at least nineteen, or at least twenty or more proteases, further comprising at least one or more additional mutations, each additional mutation reduces or eliminates the corresponding additional protease activity, and said at least additional protease being selected from the group consisting of

an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5;

Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

11. The filamentous fungal cell of any preceding claim, wherein the mammalian polypeptide is selected from the group consisting of an antibody and their antigen-binding fragments, a growth factor, an interferon, a cytokine, and an interleukin.

12. The filamentous fungal cell of any preceding claim, wherein the fungal cell further comprises ALG3 having reduced activity.

13. The filamentous fungal cell of claim 12, wherein the gene encoding ALG3 comprises a mutation that reduces or eliminates the corresponding activity.

14. The filamentous fungal cell of any preceding claim, further comprising

an N-acetylglucosaminyltransferase I catalytic domain, and, optionally, an N-acetylglucosaminyltransferase II catalytic domain.

15. A method of improving stability of a polypeptide, comprising

a) providing the filamentous fungal cell of any preceding claim, and b) culturing the cell such that the heterologous polypeptide is expressed, wherein the heterologous polypeptide exhibits increased stability compared to the heterologous polypeptide produced in a corresponding parental filamentous fungal cell in which the proteases do not have reduced activity.

16. A method of making a heterologous polypeptide, comprising

a) providing the filamentous fungal cell of any of claims 1-14, b) culturing the cell such that the heterologous polypeptide is expressed, and c) purifying the heterologous polypeptide.

Description:
MULTIPLE PROTEASES DEFICIENT FILAMENTOUS FUNGAL CELLS AND

METHODS OF USE THEREOF

FIELD OF THE INVENTION

The present disclosure relates to compositions and methods useful for the production of heterologous proteins in filamentous fungal cells.

BACKGROUND

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. Moreover, tools are available to manipulate the relatively simple genetic makeup of yeast and fungal cells as well as more complex eukaryotic cells such as mammalian or insect cells (De Pourcq et al., Appl Microbiol Biotechnol, 87(5): 1617-31). Despite these advantages, many therapeutic proteins are still being produced in mammalian cells, which produce therapeutic proteins with posttranslational modifications most resembling the native human proteins, whereas the posttranslational modifications naturally produced by yeast and fungi often differ from that found in mammalian cells.

To address this deficiency, new strains of yeast and fungi are being developed that produce posttranslational modifications that more closely resemble those found in native human proteins. Thus, there has been renewed interest in using yeast and fungal cells to express more complex proteins. However, due to the industry's focus on mammalian cell culture technology for such a long time, the fungal cell expression systems such as Trichoderma are not as well established as mammalian cell culture and therefore suffer from drawbacks when expressing mammalian proteins.

Thus, a need remains in the art for improved filamentous fungal cells, such as Trichoderma fungus cells, that can stably produce heterologous proteins, such as immunoglobulins, preferably at high levels of expression.

SUMMARY

Described herein are compositions including filamentous fungal cells, such as Trichoderma fungal cells having reduced or no detectable activity of at least three proteases, and having a recombinant polynucleotide encoding a heterologous polypeptide that is produced at increased levels. Further described herein are methods of improving heterologous polypeptide stability and methods of making heterologous polypeptides in which the proteases do have the reduced activity. Further described herein are compositions including filamentous fungal cells, such as Trichoderma fungal cells, having reduced or no detectable activity in one or more of the following proteases: pep9, ampl, amp2, and sepl. Thus one aspect includes filamentous fungal cells comprising at least one endogenous protease having reduced or no protease activity, and a recombinant polynucleotide encoding heterologous polypeptide, wherein the cell has reduced or no protease activity in one or more of the following proteases: pep9, ampl, amp2, and sep2. In one specific embodiment, the production level of the polypeptide is at least two-fold higher than the production level of the same polypeptide as produced in a corresponding filamentous fungal cell in which the proteases do not have the reduced activity.

In another aspect, that may be combined with the precedent embodiment, it includes filamentous fungal cells having reduced or no detectable activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a heterologous polypeptide produced at a level of at least 2-fold higher than the production level of the polypeptide in a corresponding parental filamentous fungal cell in which the proteases do not have the reduced activity. In certain embodiments, when the cell is an Aspergillus cell, the total protease activity is reduced to 50% or less of the total protease activity of the corresponding parental Aspergillus cell in which the protease do not have reduced activity.

In one embodiment that may be combined with the preceding embodiments, the total protease activity of the filamentous fungal cell is reduced to 49% or less, 40% or less, 31% or less, 6% or less, of the total protease activity of the corresponding parental filamentous fungal cell in which the proteases do not have the reduced activity.

In certain embodiments, the expression level of at least three proteases is reduced or eliminated. In certain embodiments, genes encoding the three proteases each comprise a mutation that reduces or eliminates the corresponding protease activity. In certain embodiments that may be combined with the preceding embodiments, the three protease encoding genes are pepl, tspl, and slpl. In other embodiments, the three protease encoding genes are gapl, slpl, and pepl.

In certain embodiments, the fungal cells have reduced or no detectable activity of four endogenous proteases; genes encoding the four proteases each comprise a mutation that reduces or eliminates the corresponding protease activity. In certain embodiments that may be combined with the preceding embodiments, the four protease encoding genes are pepl, tspl, slpl, and gapl.

In certain embodiments that may be combined with the preceding embodiments, the three or four protease encoding genes are selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2. In certain embodiments that may be combined with the preceding embodiments, the three or four protease encoding genes are selected from pepl, pep3, pep4, tspl, slpl, slp2, gapl, and gap2. In certain embodiments, the three or four protease encoding genes are selected from pepl, pep2, pep3, pep4, pep5, gapl, gap2, slpl, slp2, and tspl.

In other embodiments, the fungal cells have reduced or no detectable activity of five endogenous proteases; genes encoding the five proteases each comprise a mutation that reduces or eliminates the corresponding protease activity. In certain embodiments that may be combined with the preceding embodiments, the five protease encoding genes are pepl, tspl, slpl, gapl, and pep4. In other embodiments, the five protease encoding genes are pepl, tspl, slpl, gapl, and gap2. In certain embodiments, the fungal cells have reduced or no detectable activity of six endogenous proteases; genes encoding the six proteases each comprise a mutation that reduces or eliminates the corresponding protease activity. In certain embodiments, the cell has six protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the six protease encoding genes are pepl, tspl, slpl, gapl, gap2, and pep4.

In certain embodiments that may be combined with the preceding embodiments, the fungal cells have three to six proteases having reduced or no detectable activity in each of the three to six proteases selected from pepl, pep2, pep3, pep4, pep5, tspl, slpl, slp2, slp3, gapl, and gap2.

In certain embodiments that may be combined with the preceding embodiments, the cell has seven protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the seven protease encoding genes are pepl, tspl, slpl, gapl, gap2, pep4, and pep 3.

In certain embodiments that may be combined with the preceding embodiments, the cell has eight protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the eight protease encoding genes are pepl , tspl , slpl , gapl , gap2, pep4, pep3, and pep5.

In certain embodiments that may be combined with the preceding embodiments, the fungal cell has an additional protease having reduced activity, the gene encoding the additional protease comprises a mutation that reduces or eliminates the corresponding protease activity, and the additional protease is selected from pep7, pep8, pepll, pep!2, tppl, gap2, slp3, slp5, slp6, slp7, and slp8.

In certain embodiments that may be combined with the preceding embodiments, the cell has eight protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the eight protease encoding genes are either pepl, slpl, gapl, gap2, pep4, pep3, pep5, ampl, b) pepl, slpl, gapl, gap2, pep4, pep3, pep5, amp2, c) pepl, slpl, gapl, gap2, pep4, pep3, pep5, sepl, d) pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep9, or e) pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2.

Optionally, in one specific embodiment of the preceding emdodiment, the cell further comprises a mutation that reduces or eliminates the protease activity of tspl .

In certain embodiments that may be combined with the preceding embodiments, the cell has nine protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the nine protease encoding genes are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2 and sepl. Optionally, in one specific embodiment of such emdodiment, the cell further comprises a mutation that reduces or eliminates the protease activity of tspl .

In certain embodiments that may be combined with the preceding embodiments, the cell has ten protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the ten protease encoding genes are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2,sepl and slp8. Optionally, in one specific embodiment of such emdodiment, the cell further comprises a mutation that reduces or eliminates the protease activity of tspl.

In certain embodiments that may be combined with the preceding embodiments, the cell has eleven protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the eleven protease encoding genes are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2,sepl, slp8 and amp2. Optionally, in one specific embodiment of such emdodiment, the cell further comprises a mutation that reduces or eliminates the protease activity of tspl.

In certain embodiments that may be combined with the preceding embodiments, the cell has twelve protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the twelve protease encoding genes are pepl, slpl, gapl, gap2, pep4, pep3, pep 5, pep2,sepl, slp8, amp2 and slp7. In certain embodiments that may be combined with the preceding embodiments, the cell has reduced or no protease activity in at least thirteen proteases, each of the genes encoding the thirteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the thirteen proteases are either pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, pep9; pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7;

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp3.

In certain embodiments that may be combined with the preceding embodiments, the cell has reduced or no protease activity in at least fourteen proteases, each of the genes encoding the fourteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fourteen proteases are pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2;

In certain embodiments that may be combined with the preceding embodiments, the cell has reduced or no protease activity in at least fifteen proteases, each of the genes encoding the fifteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fifteen proteases are either pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mpl ; or, pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mp5.

In certain embodiments that may be combined with the precedent embodiments, the cell has reduced or no protease activity in at least sixteen, at least seventeen, at least eighteen, at least nineteen, or at least twenty or more proteases, and said cell comprises at least one mutation that reduces or eliminates the corresponding protease activity, selected from the group consisting of an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5; Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

In certain embodiments that may be combined with the preceding embodiments, the heterologous polypeptide is a mammalian polypeptide. In certain embodiments, the mammalian polypeptide is glycosylated.

In certain embodiments, the mammalian polypeptide is selected from an immunoglobulin, an antibody and their antigen-binding fragments, a growth factor, an interferon, a cytokine, and an interleukin. In certain embodiments, the mammalian polypeptide is an immunoglobulin or an antibody, or their Fc fragment. In certain embodiments, the mammalian polypeptide is selected from insulin-like growth factor 1 (IGFl), human growth hormone (hGH), and interferon alpha 2b (IFNa2b).

In certain embodiments that may be combined with the preceding embodiments, the heterologous polypeptide is a non-mammalian polypeptide. In certain embodiments, the non-mammalian polypeptide is an aminopeptidase, amylase, carbohydrase, carboxypeptidase, catalase, cellulase, chitinase, cutinase, deoxyribonuclease, esterase, alpha-galactosidase, beta-galactosidase, glucoamylase, alpha-glucosidase, beta-glucosidase, invertase, laccase, lipase, mutanase, oxidase, pectinolytic enzyme, peroxidase, phospholipase, phytase, polyphenoloxidase, proteolytic enzyme, ribonuclease, transglutaminase or xylanase. In certain embodiments that may be combined with the preceding embodiments, the fungal cell further contains reduced or no detectable activity of ALG3, a mannosyltransferase enzyme. In certain embodiments, the gene encoding ALG3 contains a mutation that reduces or eliminates the corresponding activity. In certain embodiments that may be combined with the preceding embodiments, the fungal cell further contains a polynucleotide encoding an a-l ,2-mannosidase.

In certain embodiments that may be combined with the preceding embodiments, the fungal cell has a mutation that reduces the expression of a protease desired to have reduced activity. In certain embodiments that may be combined with the preceding embodiments, the mutation is a deletion within the gene encoding the protease. In certain embodiments that may be combined with the preceding embodiments, the mutation is a deletion of the portion of the gene encoding the catalytic domain of the protease. In certain embodiments that may be combined with the preceding embodiments, the fungal cell has a point mutation in the portion of the gene encoding the catalytic domain of the protease.

In other embodiments, the reduction or elimination of protease activity of one or more proteases results from RNAi constructs specific for i) one protease or ii) two or more proteases selected from the group consisting of a pep-type, protease, a trypsin-like serine protease, a gap-type proteae, a sedolisin protease and a sip-type, protease. In certain embodiments, RNAi constructs are specific for slp2, slp3, slp5 and/or slp6.

In certain embodiments that may be combined with the preceding embodiments, the fungal cell further contains an N-acetylglucosaminyltransferase I catalytic domain and an N- acetylglucosaminyltransferase II catalytic domain. In certain embodiments, the N- acetylglucosaminyltransferase I catalytic domain and the N-acetylglucosaminyltransferase II catalytic domain are encoded by a polynucleotide. In certain embodiments, the N- acetylglucosaminyltransferase I catalytic domain is encoded by a first polynucleotide and the N- acetylglucosaminyltransferase II catalytic domain is encoded by a second polynucleotide. In certain embodiments that may be combined with the preceding embodiments, the fungal cell further contains a polynucleotide encoding a mannosidase II and/or a galactosyl transferase. In certain embodiments, the fungal cell contains enzymes selected from the group consisting of ocl,2 mannosidase, N-acetylglucosaminyltransferase I, N-acetylglucosaminyltransferase II, mannosidase II and/or galactosyltransferase, said enzymes further comprising a targeting peptide, for example a heterologous targeting peptide for proper localization of the corresponding enzyme. In certain embodiments, the targeting peptide is selected from SEQ ID NOs: 589-594. In certain embodiments that may be combined with the preceding embodiments, the fungal cell is a Trichoderma fungal cell, a Myceliophthora fungal cell, an Aspergillus fungal cell, a Neurospora fungal cell, a Fusarium or Penicilium fungal cell, or a Chrysosporium fungal cell. In certain embodiments that may be combined with the preceding embodiments, the fungal cell is Trichoderma reesei. In certain embodiments that may be combined with the preceding embodiments, the fungal cell is wild type for pep4 protease. In certain embodiments that may be combined with the preceding embodiments, the fungal cell is wild type for tspl protease.

Another aspect includes methods of improving heterologous polypeptide stability, by: a) providing the filamentous fungal cell of any of the preceding embodiments; and b) culturing the cell such that the heterologous polypeptide is expressed, where the heterologous polypeptide has increased stability compared to the heterologous polypeptide produced in a corresponding parental filamentous fungal cell in which the proteases do not have reduced activity, for example, as not containing the mutations of the genes encoding the proteases. Another aspect includes methods of making a heterologous polypeptide, by: a) providing the filamentous fungal cell of any of the preceding embodiments; b) culturing the host cell such that the heterologous polypeptide is expressed; and c) purifying the heterologous polypeptide.

In certain embodiments that may be combined with the preceding embodiments, the filamentous fungal cell further contains a carrier protein. In certain embodiments, the carrier protein is CBH1. In certain embodiments that may be combined with the preceding embodiments, the culturing is in a medium comprising a protease inhibitor. In certain embodiments, the culturing is in a medium having one or two protease inhibitors selected from SBTI and chymostatin. In certain embodiments, the protease activites are reduced or eliminated according to the invention with further co-expression or co-culture such as SBTI, or BBI, e.g. as described in WO2005047302, or sip inhibitor such as pepc inhibitor as described in WO2009071530.

In certain embodiments, the heterologous polypeptide produced according to the method is a glycosylated mammalian polypeptide, preferably an antibody or their Fc glycosylated fragments, and at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mole%) of the total N-glycans of the polypeptide consists of Man 3 GlcNAc 2 N-glycan or Man5GlcNAc2 N-glycan. In other embodiments, the heterologous polypeptide produced according to the method is a glycosylated mammalian polypeptide, preferably an antibody or their Fc glycosylated fragments, and at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mole%) of the total N-glycans of the polypeptide consists of complex N-glycan, such as GO, Gl or G2 glycoforms or their fucosylated forms, FG0, FG1 and FG2. In certain embodiments, the heterologous polypeptide produced according to the method is a glycosylated mammalian polypeptide, preferably an antibody or their Fc glycosylated fragments, and at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mole%) of the total N-glycans of the polypeptide consists of hybrid N-glycan. In certain embodiments, the heterologous polypeptide produced according to the method is a glycosylated mammalian polypeptide, preferably an antibody or their Fc glycosylated fragments, and at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mole%) of the total N-glycans of the polypeptide consists of Gl or G2 N-glycan. Another aspect includes the heterologous polypeptides, preferably an antibody or their Fc glycosylated fragments, obtainable by the methods as described above.

Another aspect includes Trichoderma fungal cells, or closely related species, including

Myceliophthora fungal cell, Aspergillus fungal cell, Neurospora fungal cell, Penicillium cell, Fusarium cell, or Chrysosporium fungal cell, said fungal cell having reduced or no detectable activity of at least three proteases selected from pepl, pep2, pep3, pep4, pep5, tspl, slpl, slp2, gapl, and gap2, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide produced at a level of at least 2-fold higher than the production level of the polypeptide in a corresponding parental fungal cell. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell, further contains reduced or no detectable activity of one or more of the following proteases: pep9, ampl, amp2 and sepl.

In certain embodiments, the expression level of the at least three proteases is reduced or eliminated in the Trichoderma or closely related species fungal cell. In certain embodiments, genes encoding the at least three proteases each comprise a mutation that reduces or eliminates the corresponding protease activity in the Trichoderma or closely related species cell. In certain embodiments, the Trichoderma or closely related species fungal cell includes three protease encoding genes with a mutation that reduces or eliminates protease activity, which are selected from gapl, slpl, and pepl. In certain embodiments that may be combined with the preceding embodiments, the mammalian polypeptide in the Trichoderma or closely related species fungal cell is an antibody, or their antigen-binding fragments, or an immunoglobulin, and the at least three proteases are selected from pepl, pep3, pep4, tspl, slpl, slp2, gapl, and gap2. In certain embodiments, the Trichoderma or closely related species fungal cell contains four protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the four protease encoding genes with such mutation are pepl, tspl, slpl, and gapl. In certain embodiments, the Trichoderma or closely related species fungal cell has five protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the five protease encoding genes with such mutation are pepl, tspl, slpl, gapl, and pep4. In certain embodiments that may be combined with the preceding embodiments, the mammalian polypeptide in the Trichoderma fungal cell is a growth factor, interferon, cytokine, or interleukin, and the three proteases with reduced activity are selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, gapl, gap2, slpl, slp2, slp7, and, optionally tspl. In certain embodiments, the Trichoderma or closely related species fungal cell has five protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the five protease encoding genes with such mutation are pepl, tspl, slpl, gapl, and gap2. In certain embodiments, the Trichoderma or closely related species fungal cell has six protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the six protease encoding genes with such mutation are pepl, tspl, slpl, gapl, gap2, and pep4. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has seven protease encoding genes, each of which comprise a mutation that reduces or eliminates the corresponding protease activity, and the seven protease encoding genes are pepl, tspl, slpl, gapl, gap2, pep4, and pep3. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has eight protease encoding genes, each of which comprise a mutation that reduces the corresponding protease activity, and the eight protease encoding genes with such mutation are pepl, tspl, slpl, gapl, gap2, pep4, pep3, and pep5. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has eight protease encoding genes, each of which comprise a mutation that reduces the corresponding protease activity, and the eight protease encoding genes with such mutation are selected from either

(i) pepl, slpl, gapl, gap2, pep4, pep3, pep5, ampl; (ii) pepl, slpl, gapl, gap2, pep4, pep3, pep5, amp2;

(iii) pepl, slpl, gapl, gap2, pep4, pep3, pep5, sepl;

(iv) pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep9; or,

(v) pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2

In such embodiment, the cell may further comprise an additional mutation that reduces or eliminates the protease activity of tspl .

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has nine protease encoding genes, each of which comprise a mutation that reduces the corresponding protease activity, and the nine protease encoding genes with such mutation are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl. In such embodiment, the cell may further comprise an additional mutation that reduces or eliminates the protease activity of tspl . In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has ten protease encoding genes, each of which comprise a mutation that reduces the corresponding protease activity, and the ten protease encoding genes with such mutation are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8. In such embodiment, the cell may further comprise an additional mutation that reduces or eliminates the protease activity of tspl .

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has eleven protease encoding genes, each of which comprise a mutation that reduces the corresponding protease activity, and the eleven protease encoding genes with such mutation are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2. In such embodiment, the cell may further comprise an additional mutation that reduces or eliminates the protease activity of tspl .

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has twelve protease encoding genes, each of which comprise a mutation that reduces the corresponding protease activity, and the twelve protease encoding genes with such mutation are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7.

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has reduced or no protease activity in at least thirteen proteases, each of the genes encoding the thirteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the thirteen proteases are either pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, pep9; pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7;

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp3. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has reduced or no protease activity in at least fourteen proteases, each of the genes encoding the fourteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fourteen proteases are pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2;

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell has reduced or no protease activity in at least fifteen proteases, each of the genes encoding the fifteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fifteen proteases are either P e pl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mpl ; or, pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mp5.

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell further contains reduced or no detectable activity of one or more additional proteases. In certain embodiments, the expression level of the one or more additional proteases in the Trichoderma or closely related species fungal cell is reduced or eliminated. In certain embodiments, genes encoding the one or more additional protease in the Trichoderma or closely related species fungal cell each have a mutation that reduces or eliminates the corresponding protease activity. In certain embodiments that may be combined with the preceding embodiments, the one or more additional protease encoding genes are selected from pep7, pep8, pepll, pepl2, tppl, gap2, slp3, slp5, slp6, slp7, and slp8. In certain embodiments that may be combined with the preceding embodiments, the one or more additional protease encoding genes are selected from the group consisting of an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5; Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell further contains reduced or no detectable activity of ALG3. In certain embodiments, the gene encoding ALG3 in the Trichoderma or closely related species fungal cell contains a mutation that reduces or eliminates the corresponding activity. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell further contains a polynucleotide encoding an a-l,2-mannosidase. In certain embodiments that may be combined with the preceding embodiments, the mutation reduces or eliminates the expression of the gene in the Trichoderma or closely related species fungal cell. In certain embodiments that may be combined with the preceding embodiments, the mutation is a deletion of the gene in the Trichoderma or closely related species fungal cell. In certain embodiments that may be combined with the preceding embodiments, the mutation is a deletion of the portion of the gene encoding the catalytic domain of the protease in the Trichoderma or closely related species fungal cell. In certain embodiments that may be combined with the preceding embodiments, the mutation is a point mutation in the portion of the gene encoding the catalytic domain of the protease in the Trichoderma or closely related species fungal cell. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma or closely related species fungal cell further contains a N- acetylglucosaminyltransferase I catalytic domain and an N-acetylglucosaminyltransferase II catalytic domain. In certain embodiments, the N-acetylglucosaminyltransferase I catalytic domain and the N-acetylglucosaminyltransferase II catalytic domain are encoded by a polynucleotide of the Trichoderma or closely related species fungal cell. In certain embodiments, the N- acetylglucosaminyltransferase I catalytic domain is encoded by a first polynucleotide and the N- acetylglucosaminyltransferase II catalytic domain is encoded by a second polynucleotide of the Trichoderma or closely related species fungal cell. In certain embodiments that may be combined with the preceding embodiments, the Trichoderma fungal cell further contains a polynucleotide encoding a mannosidase II. In certain embodiments, the proteases each have 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% sequence identity to an amino acid sequence selected from SEQ ID NOs: 1, 17, 37, 58, 66, 82, 98, 118, 129, 166, and 182. In certain embodiments, the total protease activity in the Trichoderma fungal cell is reduced to 49% or less, 40% or less, 31% or less, 6% or less of of the total protease activity of the corresponding Trichoderma parental cell in which the proteases do not have the reduced activity. In certain embodiments that may be combined with the preceding embodiments, the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide produced at a level of at least 2- fold higher than the production level of the polypeptide in a corresponding parental Trichoderma fungal cell. In certain embodiments that may be combined with the preceding embodiments, the mammalian polypeptide is produced in a full length version at a level higher than the production level of the full-length version of the polypeptide in a corresponding parental Trichoderma fungal cell.

Another aspect includes methods of improving heterologous polypeptide stability, by: a) providing the Trichoderma fungal cell of any of the preceding embodiments; and b) culturing the cell such that the heterologous polypeptide is expressed, where the heterologous polypeptide has increased stability compared to a host cell not containing the mutations of the genes encoding the proteases. Another aspect includes methods of making a heterologous polypeptide, e.g. an immunoglobulin or antibody or their glycosylated Fc fragments, by: a) providing the Trichoderma or closely related species fungal cell of any of the preceding embodiments; b) culturing the host cell such that the heterologous polypeptide is expressed; and c) purifying the heterologous polypeptide. In certain embodiments that may be combined with the preceding embodiments, the filamentous fungal cell further contains a carrier protein. In certain embodiments, the carrier protein is CBH1. DESCRIPTION OF THE FIGURES

Figure 1 depicts Southern blot analysis showing the generation of the 10-fold protease deletion strain M633 (48-70C; M659 is repurified M633). Figure A depicts the expected signal of sepl ORF: 7.5 kb from M124 (Δ0) and M574 (Δ9), no signal from the transformants. Figure B depicts the expected signal of sepl 5' flank: 7.5 kb from M124, 3.1 kb from transformants, 3.9 kb from pTTv255. Figure C depicts the expected signal of sepl 3' flank: 7.5 kb from M124, 3.9 kb from transformants, 3.9 kb from pTTv255.

Figure 2 depicts Southern blot analysis showing the generation of the 11 -fold protease deletion strain M750 (54-131-2). Figure A depicts the expected signal of slp8 ORF: 5.6 kb from M124 (Δ0) and M659 (Δ10), no signal from the transformants. The faint signal pattern seen in all samples is unspecific background. Figure B depicts the expected signal of slp8 5' flank: 5.6 kb from M659, 2.9 kb from transformants, 6.7 kb from pTTv330. Figure C depicts the expected signal of slp8 3' flank: 5.6 kb from M659, 4.2 kb from transformants, 6.7 kb from pTTv330. Strain M751 (54-159-1) shows multiple signals with both probes indicating integration of several deletion cassettes to the genome and putatively also genomic rearrangements. Figure 3 depicts Southern blot analysis showing the generation of the 12-fold protease deletion strain M893 (59-6A). Figure A depicts the expected signal of amp2 ORF: 5.5 kb from M124 (Δ0) and M750 (Δ11), no signal from the transformants. Figure B depicts the expected signal of amp2 5' flank: 5.5 kb from M124 and M750, 3.6 kb from transformants, 6.6 kb from pTTv327. Signal seen at approximately 7 kb for all samples is unspecific background. Figure C depicts the expected signal of amp2 3' flank: 5.5 kb from M124 and M750, 4.5 kb from transformants, 6.6 kb from pTTv327. Figure 4 depicts 24 well culture of 9 fold protease deletion strains. Cultures were grown in TrMM with diammonium citrate without ammonium sulfate, 100 mM PIPPS, 20 g/L spent grain extract, 40 g/L lactose at pH 4.5, shaking at 28°C. Immunoblot of interferon alpha 2b expression from 0.5 μΐ culture supernantat. Figure 5 depicts a fermentor cultivation of 9 protease deletion strains expressing interferon. Strains were grown in TrMM plus 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, 40 g/L sorbose, pH 4.5. Immunoblot detecting interferon alpha 2b from 0.1 μΐ of supernatant. Standard amounts of interferon 50, 100, and 200 ng were used to generate a standard curve.

Figure 6 depicts an immunoblot showing the interferon alpha 2b production level in the supernatant of the M674 cultivated in the Triabl25 and Triabl26 fermentations

Figure 7 depicts a 24 well culture of slp2 silencing strains and control M577 strain. Cultures were grown in TrMM with diammonium citrate without ammonium sulfate, 100 mM PIPPS, 20 g/L spent grain extract, 40 g/L lactose at pH 4.5, shaking at 28°C. Immunoblot of interferon alpha 2b expression from 0.2 μΐ culture supernantat. Figure 8 depicts cultivation of strains M960 and M961 in fermentor using TrMM in 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, and 40 g/L sorbose at pH 4.5 with the temperature shifting from 28° to 22° at 48h. Immunoblot detecting interferon alpha 2b from 0.05 μΐ of supernatant. Interferon standards were 400, 200, 100, 50, and 25 ng.

Figure 9 depicts a phylogenetic tree of ampl and amp2 of selected filamentous fungi. Figure 10 depicts a phylogenetic tree of sepl of selected filamentous fungi.

Figure 11 depicts a phylogenetic tree of pep9 of selected filamentous fungi.

Figure 12 FGF21 production in 24 well cultures treated with protease inhibitors. Supernatant samples from day 6 were assayed via immunoblotting with a FGF21 standard curve. The primary antibody and reference material was provided by Novartis. The primary antibody was diluted to 2 μg/ml in TBST before use. The goat anti-rabbit secondary (Biorad goat anti-rabbit AP #170- 6518) was diluted 1: 10,000 in TBST.

Figure 13 Immunoblot detecting FGF21 and CBHI expression from the M393 control strain and the new FGF21 transformants from day 4-6. The new transformants are from Ml 076

transformation. CBHI carrier was detected around 60 kD and the FGF21 free product was at 10 kl).

Figure 14 Immunoblot using anti-FGF21 antibody to detect expression of FGF21 from

fermentation samples from cultivation of strain M1200 and M1205 with and without inhibitors. Standard amounts of FGF21 are included on each blot for quantitation. Supernatants were diluted so that 0.1 μΐ was loaded per lane.

Figure 15 Immunoblot detecting FGF21 expression from the M1200 strain grown in 24 well culture with and without protease inhibitor treatment. Pepstatin, chymostatin, 1 , 10-phenathroline were used at a final concentration of 10 μΜ and SBTI was used at 0.1 mg/ml. 2 μΐ of the culture supernatant plus orange LSB was loaded per well. Whole anti-FGF21 antibody was generally used to detect all forms of FGF21 produced, but in the lower right blot an N-terminal antibody was used to detect the N-terminal containing forms. Also in the lower right blot anti-CBHI antibody was used for detection.

Figure 16 Immunoblot detecting FGF21 expression from the M1200 strain grown in 24 well culture with and without protease inhibitor treatment. Pepstatin, chymostatin, 1 , 10-phenathroline were used at a final concentration of 10 μΜ and SBTI was used at 0.1 mg/ml. 2 μΐ of the culture supernatant plus orange LSB was loaded per well. Whole anti-FGF21 antibody and a C-terminal antibody were used to detect specifically the C-terminus and all forms of the FGF21 protein. In the left blot anti-CBHI antibody was used for detection.

Figure 17. Immunoblots showing the MAB01 heavy chain expressed in 24 well cultures. 0.5 μΐ of each supernatant was loaded into a 4-15% gel. Detected with anti-IgG heavy chain AP conjugate (Sigma# A3188) diluted 1: 10,000 in TBST. Developed with Promega AP substrate. Figure 18 Immunoblots showing the MAB01 heavy chain. 0.05 μΐ of each supernatant was loaded into a 4-15% gel. The heavy chain was detected with an anti -human Fc antibody IRDye 700 DX conjugate (Rockland #609-130-003) diluted 1:30,000 in TBST. The fluorescence at 700 nm was detected using an Odyssey CLx near infrared imager (Li-Cor). Figure 19 show a phylogenetic tree of a subset of the proteases amenable to deletions.

Figure 20 graphically depicts normalized protease activity data from culture supernatants from each of the protease deletion supernatants and the parent strain Ml 24. Protease activity was measured at pH 5.5 in first 5 strains and at pH 4.5 in the last three deletion strains. Protease activity is against green fluorescent casein. The six protease deletion strain has only 6% of the wild type parent strain and the 7 protease deletion strain protease activity was about 40% less than the 6 protease deletion strain activity.

DETAILED DESCRIPTION

The present invention relates to improved methods of generating recombinant heterologous polypeptides in filamentous fungal cells that have reduced or no activity of at least three proteases. The present invention further relates to improved methods of generating recombinant heterologous polypeptides in filamentous fungal cells that have reduced or no activity in one or more of the following proteases: pep9, ampl, amp2 and sepl. The present invention is based in part upon the surprising discovery that reducing the activity of a specific combination of endogenous proteases in filamentous fungal cells increases the expression and stability of a variety of recombinantly expressed heterologous proteins, such as immunoglobulins and growth factors. While others have created Trichoderma fungal cells with one or more proteases inactivated, they have not provided guidance as to which proteases are most relevant to increasing the expression and stability of specific types of proteins, such as mammalian proteins. For example, WO2011/075677 discloses certain proteases that can be knocked out in Trichoderma and even discloses Trichoderma fungal cells that are deficient in multiple proteases.

However, WO2011/075677 does not provide any guidance regarding which of the proteases have an adverse impact on the expression and stability of mammalian proteins, such as immunoglobulins or growth factors, as no examples of expression of any mammalian proteins are described therein. Moreover, WO2011/075677 only discloses heterologous expression of a single fungal protein in each of three different fungal strains deficient in a single protease. Thus, one of skill in the art would likely read WO2011/075677 as teaching that inactivating each single protease would be sufficient for heterologous protein production. Yoon et al (2009, Appl. Microbiol Biotechnol 82: 691-701, 2010: Appl. Microbiol Biotechnol DOI 10.1007/s00253-010- 2937-0) reported the construction of quintuple and ten fold protease gene disruptants for heterologous protein production in Aspergillus oryzae. The 10 protease disruptant cells improve the production yield of chymosin by only 3.8 fold, despite the high number of disrupted protease genes. Van den Hombergh et al reported a triple protease gene disruptant of Aspergillus niger. While the data show a reduction in protease activity, there is no example of any mammalian protein production described herein. WO2002068623 further report Aspergillus niger ampl, sepl and pep9proteases, and WO2012048334 reports Myceliophtora thermophila amp2, sepl and pep9 proteases. Other reports describe the cloning and characterization of sepl protease in filamentous fungal strains (WO2011077359, WO2009144269, WO200762936 and WO2002045524). The cloning and characterization of pep9 has also been described in WO2012032472, and WO2006110677.

Applicants have surprisingly shown that multiple proteases are relevant to reduction of total protease activity, increasing production of heterologous proteins and stabilizing the heterologous proteins after expression, in filamentous fungal cells, such as Trichoderma fungal cells. In particular, the inventors have identified proteases that are actually expressed in Trichoderma fungal cells (as opposed to merely being coded for in the genome) by purifying these proteases and determining which have activities that are most relevant in degrading heterologous proteins, such as mammalian proteins. Additionally, the inventors confirmed that deleting the genes responsible for the particular protease activities achieved a substantial reduction in total protease activity, which correlates to an increase in protein stabilization in terms of both quantity and quality of proteins produced in filamentous fungal cells containing such deletions, and resulted in an increase in the production of full length heterologous proteins in the cells. It was also found that Trichoderma fungal cells engineered to reduce the activity of at least three protease genes resulted in an unexpected, synergistic increase in the production of full length mammalian proteins, such as antibodies, therapeutic protein or antibody variants such Fab or single domain antibodies. In other words, the amount of full length mammalian protein produced was greater than the sum of the amounts produced in Trichoderma fungal cells containing only one or two protease gene deletions. Thus, in contrast to WO2011/075677, the inventors have shown that production of intact heterologous proteins in filamentous fungal cells, such as Trichoderma fungal cells, can be achieved by reducing or eliminating the activity of at least three proteases in the cells.

Accordingly, certain aspects of the present disclosure provide filamentous fungal cells that produce increased levels of a heterologous protein by having reduced or no activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a heterologous polypeptide produced at a level of at least 2-fold higher than the production level of the polypeptide in a corresponding parental filamentous fungal cell in which the proteases do not have the reduced activity. In other words, the desired increase in the level of the heterologous protein production is determinable by comparing the production level of the heterologous protein in a filamentous fungal cell having the reduced activity of at least three proteases, to that of a filamentous fungal cell which does not have such reduced activity, but is otherwise identical to the cell exhibiting the increased level.

Other aspects of the present disclosure provide methods of improving heterologous polypeptide stability, by: a) providing a filamentous fungal cell of the present disclosure having reduced or no activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a heterologous polypeptide; and b) culturing the cell such that the heterologous polypeptide is expressed, where the heterologous polypeptide has increased stability compared to a host cell not containing the mutations of the genes encoding the proteases. Still other aspects of the present disclosure provide methods of making a heterologous polypeptide, by: a) providing a filamentous fungal cell of the present disclosure having reduced or no activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a heterologous polypeptide; b) culturing the host cell such that the heterologous polypeptide is expressed; and c) purifying the heterologous polypeptide.

Certain aspects of the present disclosure also provide Trichoderma fungal cells that produce increased levels of a mammalian polypeptide by having reduced or no activity of at least three proteases selected from pepl , pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, gapl, and gap2, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide produced at a level of at least 2-fold higher than the production level of the polypeptide in a corresponding parental Trichoderma fungal cell in which the proteases do not have the reduced activity. In other words, the desired increase in the level of the heterologous protein production is determinable by comparing the production level of the heterologous protein in a Trichoderma fungal cell having the reduced activity of at least three proteases, to that of a Trichoderma fungal cell which does not have such reduced activity, but is otherwise identical to the cell exhibiting the increased level.

Certain aspects of the present disclosure also provide Trichoderma fungal cells that produce increased levels of a mammalian polypeptide by having reduced or no activity of at least one or more proteases selected from pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl.

Other aspects of the present disclosure provide methods of improving mammalian polypeptide stability, by: a) providing a Trichoderma fungal cell of the present disclosure having reduced activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide; and b) culturing the cell such that the mammalian polypeptide is expressed, where the mammalian polypeptide has increased stability compared to a host cell not containing the mutations of the genes encoding the proteases.

Other aspects of the present disclosure provide methods of improving mammalian polypeptide stability, by: a) providing a Trichoderma fungal cell of the present disclosure having reduced activity of at least one or more proteases selected from pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide; and b) culturing the cell such that the mammalian polypeptide is expressed, where the mammalian polypeptide has increased stability compared to a host cell not containing the mutations of the genes encoding the proteases.

Further aspects of the present disclosure provide methods of making a mammalian polypeptide, by: a) providing a Trichoderma fungal cell of the present disclosure having reduced activity of at least three protease, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide; b) culturing the host cell such that the mammalian polypeptide is expressed; and c) purifying the mammalian polypeptide.

Definitions

As used herein, 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.

As used herein, 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 and F(ab) fragments and Fv molecules; non-covalent heterodimers [227, 228]; single- chain Fv molecules (scFv) (see, e.g., Huston et al. Proc. Natl. Acad. Sci. U.S.A. 85:5897-83, 1988); dimeric and trimeric antibody fragment constructs; minibodies (see, e.g., Pack et al. Biochem 31, 1579-84, 1992; and Cumber et al. J. Immunology 149B, 120-26, 1992); humanized antibody molecules (see e.g., Riechmann et al. Nature 332, 323-27, 1988; Verhoeyan et al. Science 239, 1534-36, 1988; and GB 2,276,169); and any functional fragments obtained from such molecules, as well as antibodies obtained through non-conventional processes such as phage display. Preferably, the antibodies are monoclonal antibodies. Methods of obtaining monoclonal antibodies are well known in the art. As used herein, a "peptide" and a "polypeptide" are amino acid sequences including a plurality of consecutive polymerized amino acid residues. For purpose of this invention, typically, peptides are those molecules including up to 50 amino acid residues, and polypeptides include more than 50 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. As used herein, "protein" may refer to a peptide or a polypeptide of any size. Proteases of the Invention

The invention described herein relates to filamentous fungal cells, such as Trichoderma fungal cells, that produce increased levels of a heterologous polypeptide, such as a mammalian polypeptide, by having reduced or no detectable activity of at least three proteases found in the cells. Such proteases found in filamentous fungal cells that express a heterologous polypeptide normally catalyze significant degradation of the expressed recombinant polypeptides. Thus, by reducing or eliminating the activity of proteases in filamentous fungal cells that express a heterologous polypeptide , the stability of the expressed polypeptide is increased, resulting in an increased level of production of the polypeptide, and in some circumstances, improved quality of the produced polypeptide (e.g., full-length instead of degraded). Proteases including, without limitation, aspartic proteases, trypsin-like serine proteases, subtilisin proteases, glutamic proteases, metalloproteases 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, for example, to a level of 49% or less, or 31% or less, of the total protease activity of the corresponding parental filamentous fungal cell; and an increase in the level of production of the expressed recombinant polypeptide, for example two-fold higher than the production level in the corresponding parental filamentous fungal cell. 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. The "corresponding parental filamentous fungal cell" refers to the corresponding cell in which the proteases do not have reduced or eliminated activity.

Aspartic Proteases 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 an 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.

Fifteen aspartic proteases have been identified in Trichoderma fungal cells: pepl (tre74156), pep2 (tre53961), pep3 (trel21133), pep4 (tre77579), pep5 (tre81004), pep6 (tre68662), pep7 (tre58669), pep8 (trel22076), pep9 (tre79807), peplO (tre78639), pepll (trel21306), pepl2 (trel l9876), pepl3 (tre76887), pepl4 (trel08686) and pepl6 (trel l0490). Pepl

Examples of suitable pepl proteases include, without limitation, Trichoderma reesei pepl (SEQ ID NO: 1), Hypocrea lixii gill 1558498 (SEQ ID NO: 2), Trichoderma asperellum gil47027997 (SEQ ID NO: 3), Trichoderma atroviride jgilTriat2l297887 (SEQ ID NO: 4), Trichoderma virens jgilTriviGv29_8_2l81777 (SEQ ID NO: 5), Aspergillus fumigatus jgilTrire2lafm:Afu5gl3300 (SEQ ID NO: 6), Aspergillus oryzae gil94730408 (SEQ ID NO: 7), Metarhizium anisopliae gil322712783 (SEQ ID NO: 8), Gibberella zeae gil46126795 (SEQ ID NO: 9), Fusarium venenatum gill8448713 (SEQ ID NO: 10), Fusarium oxysporum gil342879173 (SEQ ID NO: 11), Grosmannia clavigera gil320591399 (SEQ ID NO: 12), Verticillium alboatrum gil302422750 (SEQ ID NO: 13), Chaetomium globosum gill 16182964 (SEQ ID NO: 14), Neurospora crassa gil85110723 (SEQ ID NO: 15), Neurospora tetrasperma gil336463990 (SEQ ID NO: 16), Myceliophthora thermophila gi367030924 (SEQ ID NO: 491), Penicillium chrysogenum gi255953325 (SEQ ID NO: 492), Aspergillus niger gi350639535 (SEQ ID NO: 493), Aspergillus nidulans gi67541436 (SEQ ID NO: 494), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pepl protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 1-16, SEQ ID NOs:491-494. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 1-16, SEQ ID NOs:491-494.

In some embodiments, pepl is T. reesei pepl. The amino acid sequence encoded by T. reesei pepl is set forth in SEQ ID NO: 1. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 1. In further embodiments, the protease has 100% identity to SEQ ID NO: 1.

Pep2

Examples of suitable pep2 proteases include, without limitation, Trichoderma reesei pep2 (SEQ ID NO: 182), T atroviride jgilTriat2H42040 (SEQ ID NO: 183), T virens jgilTriviGv29_8_2l53481 (SEQ ID NO: 184), Cordyceps militaris CMOl gil346326575 (SEQ ID NO: 185), Neurospora crassa gi 85111370 (SEQ ID NO: 495), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep2 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 182-185, SEQ ID NO:495. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 182-185, SEQ ID NO:495.

In some embodiments, pep2 is T. reesei pep2. The amino acid sequence encoded by T. reesei pep2 is set forth in SEQ ID NO: 182. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 182. In further embodiments, the protease has 100% identity to SEQ ID NO: 182.

Pep3

Examples of suitable pep3 proteases include, without limitation, Trichoderma reesei pep3 (SEQ ID NO: 17), T atroviride jgilTriat2 (SEQ ID NO: 18), T virens, jgilTriviGv29_8_2 (SEQ ID NO: 19), Hypocrea lixii gill45583125 (SEQ ID NO: 20), Trichoderma asperellum gil51860175 (SEQ ID NO: 21), Aspergillus niger gil317025164 (SEQ ID NO: 22), Aspergillus fumigatus gill59122534 (SEQ ID NO: 23), Aspergillus niger gill34054572 (SEQ ID NO: 24), Cordyceps militaris, gil346318620 (SEQ ID NO: 25), Glomerella graminicola gil310800156 (SEQ ID NO: 26), Fusarium oxysporum gil342871221 (SEQ ID NO: 27), Grosmannia clavigera gil320591121 (SEQ ID NO: 28), Botryotinia fuckeliana gil 12002205 (SEQ ID NO: 29), Thielavia terrestris gil346997107 (SEQ ID NO: 30), Sclerotinia sclerotiorum gill56055954 (SEQ ID NO: 31), Chaetomium globosum gill 16197829 (SEQ ID NO: 32), Neurospora tetrasperma gil336472132 (SEQ ID NO: 33), Neurospora crassa gil85102020 (SEQ ID NO: 34), Neosartorya fischeri gil 119467426 (SEQ ID NO: 35), Penicillium marneffei gil212534792 (SEQ ID NO: 36), M. thermophila gi367025909 (SEQ ID NO: 496), P. chrysogenum gi255947264 (SEQ ID NO: 497), A. oryzae 391870123 (SEQ ID NO: 498), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep3 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 17-36, SEQ ID NOs:496-498. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 17-36, SEQ ID NOs: 496-498.

In some embodiments, pep3 is T. reesei pep3. The amino acid sequence encoded by T. reesei pep3 is set forth in SEQ ID NO: 17. In other embodiments, a protease of the present disclosure has an amino acid sequence having50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 17. In further embodiments, the protease has 100% identity to SEQ ID NO: 17.

Pep4

Examples of suitable pep4 proteases include, without limitation, Trichoderma reesei pep4 (SEQ ID NO: 37), T virens jgilTriviGv29_8_2 (SEQ ID NO: 38), T atroviride jgilTriat2 (SEQ ID NO: 39), Trichoderma aureoviride gill93735605 (SEQ ID NO: 40), Aspergillus niger gil 145232965 (SEQ ID NO: 41), Aspergillus fumigatus gil70999520 (SEQ ID NO: 42), Aspergillus clavatus gil 121705756 (SEQ ID NO: 43), Nectria haematococca gil302899226 (SEQ ID NO: 44), Glomerella graminicola gil310796316 (SEQ ID NO: 45), Cordyceps militaris gil346322842 (SEQ ID NO: 46), Gibberella zeae gil46138535 (SEQ ID NO: 47), Metarhizium anisopliae gil322708430 (SEQ ID NO: 48), Fusarium oxysporum gil342882947 (SEQ ID NO: 49), Metarhizium acridum gil322700747 (SEQ ID NO: 50), Verticillium dahliae, gil346973691 (SEQ ID NO: 51), Botryotinia fuckeliana gill54309857 (SEQ ID NO: 52), Chaetomium globosum gill 16203505 (SEQ ID NO: 53), Thielavia terrestris gil347001590 (SEQ ID NO: 54), Magnaporthe oryzae gil39973863 (SEQ ID NO: 55), Tuber melanosporum gil296417651 (SEQ ID NO: 56), Neurospora crassa gil85094599 (SEQ ID NO: 57), M. thermophila gi367031892 gi255947264 (SEQ ID NO: 499), P. chrysogenum gi255936729 gi255947264 (SEQ ID NO: 500), A. oryzae gil69770745 gi255947264 (SEQ ID NO: 501), A. nidulans gi67524891 gi255947264 (SEQ ID NO: 502), and homologs thereof. Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep4 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 37-57, SEQ ID NOs:499-502. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 37-57, SEQ ID NOs:499-502.

In some embodiments, pep4 is T. reesei pep4. The amino acid sequence encoded by T. reesei pep4 is set forth in SEQ ID NO: 37. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 37. In further embodiments, the protease has 100% identity to SEQ ID NO: 37.

Pep5 Examples of suitable pep5 genes include, without limitation, Trichoderma reesei pep5 (SEQ ID NO: 58), T virens jgilTriviGv29_8_2 (SEQ ID NO: 59), T atroviride jgilTriat2l277859 (SEQ ID NO: 60), Metarhizium acridum gil322695806 (SEQ ID NO: 61), Fusarium oxysporum gill56071418 (SEQ ID NO: 62), Cordyceps militaris gil346324830 (SEQ ID NO: 63), Gibberella zeae gil46124247 (SEQ ID NO: 64), Verticillium dahliae gil346978752 (SEQ ID NO: 65), M. thermophila gi367019798 (SEQ ID NO: 503), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep5 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 58-65, SEQ ID NO:503. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 58-65, SEQ ID NO:503.

In some embodiments, pep5 is T. reesei pep5. The amino acid sequence encoded by T. reesei pep5 is set forth in SEQ ID NO: 58. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 58. In further embodiments, the protease has 100% identity to SEQ ID NO: 58. Pep7

Examples of suitable pep7 genes include, without limitation, Trichoderma reesei pep7 (SEQ ID NO: 186), Trichoderma atroviride jgilTriat2 (SEQ ID NO: 187), Trichoderma virens jgilTriviGv29_8_2 (SEQ ID NO: 188), Glomerella graminicola gil310800487 (SEQ ID NO: 189), Metarhizium acridum gil322700577 (SEQ ID NO: 190), Thielavia terrestris gil347003264 (SEQ ID NO: 191), Podospora anserine gil 171680938 (SEQ ID NO: 192), Chaetomium thermophilum gil340905460 (SEQ ID NO: 193), Verticillium dahliae gil346975960 (SEQ ID NO: 194), Myceliophthora thermophila gil347009870, gi367026634 (SEQ ID NO: 195), Neurospora crassa gil85090078 (SEQ ID NO: 196), Magnaporthe oryzae gil39948622 (SEQ ID NO: 197), Chaetomium globosum gill 16191517 (SEQ ID NO: 198), Magnaporthe oryzae gil39970765 (SEQ ID NO: 199), A. nidulans gi67522232 (SEQ ID NO: 504), A. niger gi350630464 (SEQ ID NO: 505), A. oryzae gi317138074 (SEQ ID NO: 506), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep7 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 186-199, SEQ ID NOs:504-506. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 186-199, SEQ ID NOs:504-506.

In some embodiments, pep7 is T. reesei pep7. The amino acid sequence encoded by T. reesei pep7 is set forth in SEQ ID NO: 186. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 186. In further embodiments, the protease has 100% identity to SEQ ID NO: 186.

Pep8 Examples of suitable pep8 genes include, without limitation, Trichoderma reesei pep8 EGR48424 (SEQ ID NO: 507), Trichoderma virens EHK19238 (SEQ ID NO: 508), Trichoderma atroviride EHK40047 (SEQ ID NO: 509), Neurospora tetrasperma EG053367 (SEQ ID NO: 510), Myceliophthora thermophila XP_003658897 (SEQ ID NO: 511), Neurospora crassa XP_965343(SEQ ID NO: 512), Metarhizium anisopliae EFZ03501 (SEQ ID NO: 513), Thielavia terrestris XP_003656869 (SEQ ID NO: 514), Fusarium oxysporum EGU79769 (SEQ ID NO: 515), and Gibberella zeae XP_381566 (SEQ ID NO: 516), Magnaporthe oryzae XP_°°3714540.1 (SEQ ID NO:517), P. chrysogenum XP_002557331 (SEQ ID NO: 518), A. oryzae XP_001822899.1 (SEQ ID NO: 519), A. nidulans XP_664091.1 (SEQ ID NO: 520), A. niger EHA24387.1 (SEQ ID NO: 521), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep8 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 507-521. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 507-521.

In some embodiments, pep8 is T. reesei pep8. The amino acid sequence encoded by T. reesei pep8 is set forth in SEQ ID NO: 507. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 507. In further embodiments, the protease has 100% identity to SEQ ID NO: 507.

Pep9

Examples of suitable pep9 genes include, without limitation, Trichoderma reesei pep9 79807 (SEQ ID NO: 750), Trichoderma virens 158334 (SEQ ID NO: 751), Trichoderma atroviride 90832 (SEQ ID NO: 752), Fusarium graminicola XP_384573.1(SEQ ID NO: 753), Neurospora crassa XP_001727974.1 (SEQ ID NO: 754), Myceliophthora thermophila XP_003667167.1(SEQ ID NO: 528) , Aspergillus oryzae XP_001821372.2 (SEQ ID NO: 529), Aspergillus niger ABM05950.1 (SEQ ID NO: 755), Aspergillus fumigatus XP_752122.1 (SEQ ID NO: 756), Aspergillus nidulans XP_662026.1 (SEQ ID NO: 757), Penicillium Wisconsin XP_002565726.1 (SEQ ID NO: 758) and homologs thereof. Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep9 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 750-758. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 750-758.

In some embodiments, pep9 is T. reesei pep9. The amino acid sequence encoded by T. reesei pep9 is set forth in SEQ ID NO: 750. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 750. In further embodiments, the protease has 100% identity to SEQ ID NO: 750.

Pepll

Examples of suitable pepll genes include, without limitation, Trichoderma reesei pepll EGR49498 (SEQ ID NO: 522), Trichoderma virens EHK26120 (SEQ ID NO: 523), Trichoderma atroviride EHK41756 (SEQ ID NO: 524), Fusarium pseudograminearum EKJ74550 (SEQ ID NO: 525), Metarhizium acridum EFY91821 (SEQ ID NO: 526), and Gibberella zeae XP_384151(SEQ ID NO: 527), M. thermophila XP_003667387.1(SEQ ID NO: 528) , N. crassa XP_960328.1 (SEQ ID NO: 529), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pepll protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 522-529. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 522-529.

In some embodiments, pepll is T. reesei pepll. The amino acid sequence encoded by T. reesei pepll is set forth in SEQ ID NO: 522. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 522. In further embodiments, the protease has 100% identity to SEQ ID NO: 522. Pepl2

Examples of suitable pepl2 genes include, without limitation, Trichoderma reesei pepl2 EGR52517 (SEQ ID NO: 530), Trichoderma virens pepl2 EHK18859 (SEQ ID NO: 531), Trichoderma atroviride pepl2 EHK45753 (SEQ ID NO: 532), Fusarium pseudograminearum pepl2 EKJ73392 (SEQ ID NO: 533), Gibberella zeae pepl2 XP_388759 (SEQ ID NO: 534), and Metarhizium anisopliae pepl2 EFY95489 (SEQ ID NO: 535), N. crassa XP_964574.1 (SEQ ID NO: 536), M. thermophila XP_003659978.1 (SEQ ID NO: 537), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pepl2 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 530-537. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 530-537.

In some embodiments, pepl2 is T. reesei pepl2. The amino acid sequence encoded by T. reesei pepl2 is set forth in SEQ ID NO: 530. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 530. In further embodiments, the protease has 100% identity to SEQ ID NO: 530.

Pep6, peplO, pepl3, pepl4 and pepl6

Other aspartic proteases include, without limitation, T. reesei /?e/?<5_(Tre68662, SEQ ID NO: 880), /?e/?iO_(Tre78639, SEQ ID NO:881), /¾ > /?i3_(Tre76887, SEQ ID NO:882), pepl4_{Trel 08686, SEQ ID NO:883), or pepl6_{Trel 10490, SEQ ID NO:884), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a pep6, peplO, pepl3, pepl4, pepl6 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 880-884. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 880-884.

Trypsin-Like Serine Proteases

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. Such 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.

One trypsin-like serine protease has been identified in Trichoderma fungal cells: tspl (tre73897). As discussed below, tspl has been demonstrated to have a significant impact on expression of recombinant polypeptides, such as immunoglobulins.

As discussed in Example 3 of WO 2013/102674, serine proteases were purified from Trichoderma and shown to have multiple protease activities that degrade mammalian proteins. Of these activities, tspl was identified as a trypsin-like serine protease. The tspl protease gene was then deleted from Trichoderma fungal cells and it was demonstrated that deleting tspl achieved a significant reduction in total protease activity resulting in increased stabilization of mammalian proteins produced by the cells.

Examples of suitable tspl proteases include, without limitation, Trichoderma reesei tspl (SEQ ID NO: 66), Trichoderma atroviride jgilTriat2l298187 (SEQ ID NO: 67), jgilTriviGv29_8_2 (SEQ ID NO: 68), Hypocrea lixii gil 145583579 (SEQ ID NO: 69), Hypocrea lixii gil63025000 (SEQ ID NO: 70), Sclerotinia sclerotiorum gil 156052735 (SEQ ID NO: 71), Botryotinia fuckeliana gill54314937 (SEQ ID NO: 72), Phaeosphaeria nodorum gill69605891 (SEQ ID NO: 73), Leptosphaeria maculans gil312219044 (SEQ ID NO: 74), Verticillium dahliae gil37992773 (SEQ ID NO: 75), Cochliobolus carbonum gill072114 (SEQ ID NO: 76), Metarhizium acridum gil322695345 (SEQ ID NO: 77), Metarhizium anisopliae gil4768909 (SEQ ID NO: 78), gil464963 (SEQ ID NO: 79), Gibberella zeae gil46139299 (SEQ ID NO: 80), Metarhizium anisopliae (SEQ ID NO: 81), A. nidulans gi67523821 (SEQ ID NO: 538) and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically tspl protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 66-81, SEQ ID NO:538. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 66-81, SEQ ID NO:538.

In some embodiments, tspl is T. reesei tspl. The amino acid sequence encoded by T. reesei tspl is set forth in SEQ ID NO: 66. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 66. In further embodiments, the protease has 100% identity to SEQ ID NO: 66.

Subtilisin Proteases

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, MBTPS 1 , and TPP2. Eukaryotic trypsin-like serine proteases further include an aspartic acid residue in the oxyanion hole. Subtilisin protease slp7 resembles also sedolisin protease tppl. Seven subtilisin proteases have been identified in Trichoderma fungal cells: slpl (tre51365); slp2 (trel23244); slp3 (trel23234); slp5 (tre64719), slp6 (trel21495), slp7 (trel23865), and slp8 (tre58698).

Slpl Examples of suitable slpl proteases include, without limitation, Trichoderma reesei slpl (SEQ ID NO: 82), Trichoderma atroviride jgilTriat2 (SEQ ID NO: 83), Trichoderma atroviride jgilTriat2 (SEQ ID NO: 84), Trichoderma virens jgilTriviGv29_8_2 (SEQ ID NO: 85), Hypocrea lixii gil 145583581 (SEQ ID NO: 86), Metarhizium acridum gil322694632 (SEQ ID NO: 87), Fusarium oxysporum gil342877080 (SEQ ID NO: 88), Gibberella zeae gil46139915 (SEQ ID NO: 89), Epichloe festucae gil 170674476 (SEQ ID NO: 90), Nectria haematococca gil302893164 (SEQ ID NO: 91), Sordaria macrospore gil336266150 (SEQ ID NO: 92), Glomerella graminicola gil310797947 (SEQ ID NO: 93), Neurospora tetrasperma gil336469805 (SEQ ID NO: 94), Neurospora crassa gil85086707 (SEQ ID NO: 95), Magnaporthe oryzae gil 145608997 (SEQ ID NO: 96), Chaetomium globosum gil 116208730 (SEQ ID NO: 97), M. thermophila gi367029081 (SEQ ID NO: 539), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a slpl protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 82-97, SEQ ID NO:539. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 82-97, SEQ ID NO:539.

In some embodiments, slpl is T. reesei slpl. The amino acid sequence encoded by T. reesei slpl is set forth in SEQ ID NO: 82. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 82. In further embodiments, the protease has 100% identity to SEQ ID NO: 82.

Slp2 Examples of suitable slp2 proteases include, without limitation, Trichoderma reesei slp2 (SEQ ID NO: 98), T atroviride jgilTriat2 (SEQ ID NO: 99), T virens jgilTriviGv29_8_2 (SEQ ID NO: 100), Hypocrea lixii gill 15111226 (SEQ ID NO: 101), Aspergillus fumigatus gil70997972 (SEQ ID NO: 102), Nectria haematococca gil302915240 (SEQ ID NO: 103), Gibberella zeae gil46105128 (SEQ ID NO: 104), Isaria farinose gil68165000 (SEQ ID NO: 105), Glomerella graminicola gil310797854 (SEQ ID NO: 106), Epichloe festucae gill70674491 (SEQ ID NO: 107), Metarhizium acridum gil322697754 (SEQ ID NO: 108), Acremonium sp. F11177 gill47225254 (SEQ ID NO: 109), Ophiostoma piliferum gill5808807 (SEQ ID NO: 110), Neurospora tetrasperma gil336463649 (SEQ ID NO: 111), Chaetomium thermophilum gil340992600 (SEQ ID NO: 112), Metarhizium flavoviride gil254351265 (SEQ ID NO: 113), Podospora anserine gill71680111 (SEQ ID NO: 114), Magnaporthe oryzae gil39943180 (SEQ ID NO: 115), Sclerotinia sclerotiorum gill56058540 (SEQ ID NO: 116), Talaromyces stipitatus gil242790441 (SEQ ID NO: 117), M. thermophila gi367021472 (SEQ ID NO: 540), A. niger gil45237646 (SEQ ID NO: 541), A. oryzae gil69780712 (SEQ ID NO: 542), P. chrysogenum gi255955889 (SEQ ID NO: 543), A. nidulans gi259489544 (SEQ ID NO: 544), N. crassa gi85084841 (SEQ ID NO: 545), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a slp2 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 98-117, SEQ ID NOs:540-545. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 98-117, SEQ ID NOs:540-545.

In some embodiments, slp2 is T. reesei slp2. The amino acid sequence encoded by T. reesei slp2 is set forth in SEQ ID NO: 98. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 98. In further embodiments, the protease has 100% identity to SEQ ID NO: 98.

Slp3 Examples of suitable slp3 proteases include, without limitation, Trichoderma reesei slp2 (SEQ ID NO: 166), T. atroviride jgilTriat2 (SEQ ID NO: 167), T. virens jgilTriviGv29_8_2 (SEQ ID NO: 168), Hypocrea koningii gil 124295071 (SEQ ID NO: 169), Purpureocillium lilacinum gill30750164 (SEQ ID NO: 170), Metarhizium anisopliae gill6215677 (SEQ ID NO: 171), Hirsutella rhossiliensis gil90655148 (SEQ ID NO: 172), Tolypocladium inflation gil 18542429 (SEQ ID NO: 173), Metacordyceps chlamydosporia gil 19171215 (SEQ ID NO: 174), Cordyceps militaris gil346321368 (SEQ ID NO: 175), Fusarium sp. gil628051 (SEQ ID NO: 176), Neurospora tetrasperma gil336471881 (SEQ ID NO: 177), Chaetomium globosum gill 16197403 (SEQ ID NO: 178), Neurospora crassa gil85084841 (SEQ ID NO: 179), Fusarium oxysporum gil56201265 (SEQ ID NO: 180), Gibberella zeae gil46114268 (SEQ ID NO: 181), M. thermophila gi367026259 (SEQ ID NO: 546), A. nidulans gi67538776 (SEQ ID NO: 547), A. oryzae gil69771349 (SEQ ID NO: 222), A. niger gi470729 (SEQ ID NO: 223), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a slp3 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 166-181, SEQ ID NOs:546-547, SEQ ID NOs:222- 223. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 166-181, SEQ ID NOs:546-547, SEQ ID NOs:222-223. In some embodiments, slp3 is T. reesei slp3. The amino acid sequence encoded by T. reesei slp3 is set forth in SEQ ID NO: 166. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 166. In further embodiments, the protease has 100% identity to SEQ ID NO: 166. Slp5

Examples of suitable slp5 proteases include, without limitation, Trichoderma reesei slp5 (SEQ ID NO: 200), T. atroviride jgilTriat2 (SEQ ID NO: 201), T. virens jgilTriviGv29_8_2 (SEQ ID NO: 202), Hypocrea lixii gill 18161442 (SEQ ID NO: 203), Fusarium oxysporum gil342883549 (SEQ ID NO: 204), Gibberella zeae gil46135733 (SEQ ID NO: 205), Glomerella graminicola gil310796396 (SEQ ID NO: 206), Nectria haematococca gil302927954 (SEQ ID NO: 207), Cordyceps militaris gil346319783 (SEQ ID NO: 208), Neurospora crassa gil85094084 (SEQ ID NO: 209), Neurospora tetrasperma gil336467281 (SEQ ID NO: 210), Verticillium dahliae gil346971706 (SEQ ID NO: 211), Thielavia terrestris gil347001418 (SEQ ID NO: 212), Magnaporthe oryzae gill45605493 (SEQ ID NO: 213), M. thermophila gi367032200 (SEQ ID NO: 548), P. chrysogenum gi62816282 (SEQ ID NO: 549), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a slp5 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 200-213, SEQ ID NOs:548-549. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 200-213, SEQ ID NOs:548-549. In some embodiments, slp5 is T. reesei slp5. The amino acid sequence encoded by T. reesei slp5 is set forth in SEQ ID NO: 200. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 200. In further embodiments, the protease has 100% identity to SEQ ID NO: 200. Slp6

Examples of suitable slp6 proteases include, without limitation, Trichoderma reesei slp6 (SEQ ID NO: 214), T. atroviride jgilTriat2 (SEQ ID NO: 215), T. virens jgilTriviGv29_8_2 (SEQ ID NO: 216), Hypocrea virens gil29421423 (SEQ ID NO: 217), Hypocrea lixii gil 145583127 (SEQ ID NO: 218), Trichoderma hamatum gil30144643 (SEQ ID NO: 219), Aspergillus fumigatus gil2295 (SEQ ID NO: 220), Aspergillus terreus gill 15391147 (SEQ ID NO: 221), Aspergillus oryzae gil 169771349 (SEQ ID NO: 222), Aspergillus niger gil470729 (SEQ ID NO: 223), Glomerella graminicola gil310794714 (SEQ ID NO: 224), Gibberella zeae gil46114946 (SEQ ID NO: 225), Fusarium oxysporum gil342873942 (SEQ ID NO: 226), Nectria haematococca gil302884541 (SEQ ID NO: 227), Neosartorya fischeri gill 19500190 (SEQ ID NO: 228), Verticillium alboatrum gil302413161 (SEQ ID NO: 229), Glomerella graminicola gil310790144 (SEQ ID NO: 230), N. crassa gi85090020 (SEQ ID NO: 550), and homologs thereof. Accordingly, in certain embodiments, a protease of the present disclosure, typically a slp6 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 214-230, SEQ ID NO:550. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 214-230, SEQ ID NO:550.

In some embodiments, slp6 is T. reesei slp6. The amino acid sequence encoded by T. reesei slp6 is set forth in SEQ ID NO: 214. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 214. In further embodiments, the protease has 100% identity to SEQ ID NO: 214.

Slp7

Examples of suitable slp7 proteases include, without limitation, Trichoderma reesei slp7 (SEQ ID NO: 231), T. atroviride jgilTriat2 (SEQ ID NO: 232), T. virens jgilTriviGv29_8_2 (SEQ ID NO: 233), Metarhizium anisopliae gil322710320 (SEQ ID NO: 234), Nectria haematococca gil302915000 (SEQ ID NO: 235), Myceliophthora thermophila gil347009020, gi367024935 (SEQ ID NO: 236), Gibberella zeae gil46137655 (SEQ ID NO: 237), Thielavia terrestris gil346996549 (SEQ ID NO: 238), Magnaporthe oryzae gil 145610733 (SEQ ID NO: 239), A. nidulans gi67541991 (SEQ ID NO: 551), P. chrysogenum gi255933786 (SEQ ID NO: 552), A. niger gi317036543 (SEQ ID NO: 553), A. oryzae gil69782882 (SEQ ID NO: 554), N. crassa gi85109979 (SEQ ID NO: 555), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a slp7 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 231-239, SEQ ID NOs:551-555. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 231-239, SEQ ID NOs:551-555. In some embodiments, slp7 is T. reesei slp7. The amino acid sequence encoded by T. reesei slp7 is set forth in SEQ ID NO: 231. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 231. In further embodiments, the protease has 100% identity to SEQ ID NO: 231. Slp8

Examples of suitable slp8 proteases include, without limitation, Trichoderma reesei slp8 (SEQ ID NO: 240), T. atroviride jgilTriat2H98568 (SEQ ID NO: 241), T. virens jgilTriviGv29_8_2l33902 (SEQ ID NO: 242), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 240-242. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 240-242.

In some embodiments, slp8 is T. reesei slp8. The amino acid sequence encoded by T. reesei slp8 is set forth in SEQ ID NO: 240. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 240. In further embodiments, the protease has 100% identity to SEQ ID NO: 240.

Sip like proteases Other sip-like proteases include, without limitation, slp57433 (Tre57433 SEQ ID NO: 885), slp35726 _(Tre35726 SEQ ID NO:886), slp60791 _(Tre60791 SEQ ID NO:887) or slpl09276_(Trel09276 SEQ ID NO:888), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a slp57433, slp35726, slp60791 or slpl09276 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 885-888. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 885-888. Glutamic Proteases

Glutamic proteases are enzymes that hydrolyze 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.

Two glutamic proteases have been identified in Trichoderma fungal cells: gapl (tre69555) and gap2 (trel 06661).

Gapl

Examples of suitable gapl proteases include, without limitation, Trichoderma reesei gapl (SEQ ID NO: 118), T. atroviride jgilTriat2l40863 (SEQ ID NO: 119), T. virens jgilTriviGv29_8_2l 192684 (SEQ ID NO: 120), Aspergillus flavus gil238499183 (SEQ ID NO: 121), Aspergillus niger gill45251555 (SEQ ID NO: 122), Aspergillus terreus gill 15491521 (SEQ ID NO: 123), gil37154543 (SEQ ID NO: 124), gil48425531 (SEQ ID NO: 125), gil351873 (SEQ ID NO: 126), Thielavia terrestris gil346997245 (SEQ ID NO: 127), Penicillium chrysogenum gil255940586 (SEQ ID NO: 128), M. thermophila gi367026504 (SEQ ID NO: 574), A. oryzae gi317150886 (SEQ ID NO: 575), N. crassa gi85097968 (SEQ ID NO: 576), A. niger gil31056 (SEQ ID NO: 577), P. chrysogenum gi255930123 (SEQ ID NO: 578), A. niger gil45236956 (SEQ ID NO: 579), A. oryzae gil69772955 (SEQ ID NO: 580), A. niger gil45249222 (SEQ ID NO: 581), A. nidulans gi67525839 (SEQ ID NO: 582), A. oryzae gil69785367 (SEQ ID NO: 583), P. chrysogenum gi255955319 (SEQ ID NO: 584), M. thermophila gi367019352 (SEQ ID NO: 585), A oryzae gi391863974 (SEQ ID NO: 586), M. thermophila gi367024513 (SEQ ID NO: 587), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a gapl protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 118-128, SEQ ID NOs:574-587. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 118-128, SEQ ID NOs:574-587.

In some embodiments, gapl is T. reesei gapl. The amino acid sequence encoded by T. reesei gapl is set forth in SEQ ID NO: 118. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 118. In further embodiments, the protease has 100% identity to SEQ ID NO: 118.

Gap2

Examples of suitable gap2 proteases include, without limitation, Trichoderma reesei gap2 (SEQ ID NO: 129), T. atroviride jgilTriat2l298116 (SEQ ID NO: 130), T. virens jgilTriviGv29_8_2l30331 (SEQ ID NO: 131), jgilTriviGv29_8_2l225131 (SEQ ID NO: 132), Aspergillus flavus gil238499183 (SEQ ID NO: 133), Aspergillus niger gill45251555 (SEQ ID NO: 134), Aspergillus nidulans gil67901056 (SEQ ID NO: 135), Aspergillus clavatus gill21711990 (SEQ ID NO: 136), Aspergillus fumigatus gil70986250 (SEQ ID NO: 137), Penicillium marneffei gil212534108 (SEQ ID NO: 138), Talaromyces stipitatus gil242789335 (SEQ ID NO: 139), Grosmannia clavigera gil320591529 (SEQ ID NO: 140), Neosartorya fischeri gill 19474281 (SEQ ID NO: 141), Penicillium marneffei gil212527274 (SEQ ID NO: 142), Penicillium chrysogenum gil255940586 (SEQ ID NO: 143), gill31056 (SEQ ID NO: 144), M. thermophila gi367030275 (SEQ ID NO: 588), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a gap2 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 129-144, SEQ ID NO:588. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 129-144, SEQ ID NO:588.

In some embodiments, gap2 is T. reesei gap2. The amino acid sequence encoded by T. reesei gap2 is set forth in SEQ ID NO: 129. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 129. In further embodiments, the protease has 100% identity to SEQ ID NO: 129.

Other gap-like proteases include, without limitation, gap3 (Tre70927 SEQ ID NO: 889), or gap4_(Tre57575 SEQ ID NO: 890), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a gap3 or gap4 protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NO:889 or SEQ ID NO:890 respectively. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 889-889.

Sedolisin Proteases

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.

Examples of suitable tppl proteases include, without limitation, Trichoderma reesei tppl (SEQ ID NO: 145), T. atroviride jgilTriat2l 188756 (SEQ ID NO: 146), T. virens jgilTriviGv29_8_2l217176 (SEQ ID NO: 147), Aspergillus fumigatus gil70993168 (SEQ ID NO: 148), Aspergillus oryzae gill69776800 (SEQ ID NO: 149), Aspergillus niger gill45236399 (SEQ ID NO: 150), Aspergillus clavatus gill21708799 (SEQ ID NO: 151), Aspergillus niger gill45239871 (SEQ ID NO: 152), Aspergillus clavatus gill21714541 (SEQ ID NO: 153), Aspergillus terreus gill 15387645 (SEQ ID NO: 154), Aspergillus fumigatus gil70982015 (SEQ ID NO: 155), Sclerotinia sclerotiorum gill56045898 (SEQ ID NO: 156), Botryotinia juckeliana gill54321758 (SEQ ID NO: 157), Neosartorya fischeri gilll9499774 (SEQ ID NO: 158), Talaromyces stipitatus gil242798348 (SEQ ID NO: 159), Penicillium marneffei gil212541546 (SEQ ID NO: 160), Gibberella zeae gil46114460 (SEQ ID NO: 161), Fusarium oxysporum gil342890694 (SEQ ID NO: 162), Grosmannia clavigera gil320592937 (SEQ ID NO: 163), Verticillium alboatrum gil302406186 (SEQ ID NO: 164), Verticillium dahliae gil346971444 (SEQ ID NO: 165), A. fumigatus CAE51075.1 (SEQ ID NO: 556), A. oryzae XP_001820835.1 (SEQ ID NO: 557), P. chrysogenum XP_002564029.1 (SEQ ID NO: 558), A. nidulans XP_664805.1 (SEQ ID NO: 559), P. chrysogenum XP_002565814.1 (SEQ ID NO: 560), M. thermophila XP_003663689.1 (SEQ ID NO: 561), N. crassa XP_958412.1 (SEQ ID NO: 562), A. niger XP_001394118.1 (SEQ ID NO: 563), A. fumigatus CAE17674.1 (SEQ ID NO: 564), A. niger XP_001400873.1 (SEQ ID NO: 565), A. fumigatus CAE46473.1 (SEQ ID NO: 566), A. oryzae XP_002373530.1 (SEQ ID NO: 567), A. nidulans XP_660624.1 (SEQ ID NO: 568), P. chrysogenum XP_002562943.1 (SEQ ID NO: 569), A. fumigatus CAE17675.1 (SEQ ID NO: 570), A. fumigatus EAL86850.2 (SEQ ID NO: 571), N. crassa XP_961957.1 (SEQ ID NO: 572), A. oryzae BAB97387.1 (SEQ ID NO: 573), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a tppl protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%,

80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 145-165, SEQ ID NOs:556-573. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 145-165, SEQ ID NOs:556-573.

In some embodiments, tppl is T. reesei tppl. The amino acid sequence encoded by T. reesei tppl is set forth in SEQ ID NO: 145. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 145. In further embodiments, the protease has 100% identity to SEQ ID NO: 145.

Other sedolisin-like proteases include, without limitation, sed2 (Tre70962, SEQ ID NO: 891), sed3 _(Tre81517 SEQ ID NO:892), or sed5_(Trel 11838 SEQ ID NO:893), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a sed2, sed3 or sed5 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 891-893. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 891-893.

Aminopeptidase Proteases

Aminopeptidases catalyze the cleavage of amino acids from the amino terminus of protein or peptide substrates. They are widely distributed throughout the animal and plant kingdoms and are found in many subcellular organelles, in cytoplasm, and as membrane components. Many, but not all, of these peptidases are zinc metalloenzymes. Amp2 is a bifunctional enzyme. It is a leukotriene A4 hydrolase with aminopeptidase activity (EC 3.3.2.6).

Two aminopeptidases have been identified in Trichoderma fungal cells: ampl (tre81070) and amp2 (trel08592).

Ampl Examples of suitable ampl proteases include, without limitation, Trichoderma reesei ampl 81070 (SEQ ID NO: 759), T. virens 74747 (SEQ ID NO: 760), T. atroviride 147450 (SEQ ID NO: 761), F. graminicola XP_386703.1 (SEQ ID NO: 762), A. nidulans CBF75094.1 (SEQ ID NO: 763), A.niger EHA21022.1 (SEQ ID NO: 764), A. oryzae XP_001727175.1 (SEQ ID NO: 765), A.fumigatus XP_749158.1 (SEQ ID NO: 766), M. thermophila XP_003667354.1 (SEQ ID NO: 767), F .graminicola XP_385112.1 (SEQ ID NO: 768), P. Chrysogenum XP_002567159.1 (SEQ ID NO: 769), A. fumigatus XP_748386.2 (SEQ ID NO: 770), A. oryzae XP_001819545.1 (SEQ ID NO: 771), A. nidulans XP_681714.1 (SEQ ID NO: 772), N. crassa XP_957507.1 (SEQ ID NO: 773), M. thermo XP_003665703.1 (SEQ ID NO: 774), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a ampl protease, has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 759-774. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 759-774.

In some embodiments, ampl is T. reesei ampl. The amino acid sequence encoded by T. reesei ampl is set forth in SEQ ID NO: 759. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity (e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 759. In further embodiments, the protease has 100% identity to SEQ ID NO: 759.

Amp2

Examples of suitable amp2 proteases include, without limitation, Trichoderma reesei amp2 108592 (SEQ ID NO: 775), T. virens 73611(SEQ ID NO: 776), T. atroviride 284076 (SEQ ID NO: 777), F. graminicola XP_390364.1 (SEQ ID NO: 778), N. crassa XP_960660.1 (SEQ ID NO: 779), M. thermophila XP_003662184.1 (SEQ ID NO: 780), A. oryzae XP_001826499.2 (SEQ ID NO: 781), A. niger XP_001390581.1 (SEQ ID NO: 782), A. nidulans XP_663416.1 (SEQ ID NO: 783), A. fumigatus XP_755088.1 (SEQ ID NO: 784), P. chrysogenum XP_002558974.1 (SEQ ID NO: 785) and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a amp2 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 775-785. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 775-785.

In some embodiments, amp2 is T. reesei amp2. The amino acid sequence encoded by T. reesei amp2 is set forth in SEQ ID NO: 775. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 775. In further embodiments, the protease has 100% identity to SEQ ID NO: 775.

Sep Proteases

Sep proteases are serine proteases belonging to the S28 subtype. They have a catalytic triad of serine, aspartate, and histidine: serine acts as a nucleophile, aspartate as an electrophile, and histidine as a base. These serine proteases include several eukaryotic enzymes such as lysosomal Pro-X carboxypeptidase, dipeptidyl-peptidase II, and thymus-specific serine peptidase.

Examples of suitable sepl proteases include, without limitation, Trichoderma reesei sepl 124051 (SEQ ID NO: 786), T. virens 39211 (SEQ ID NO: 787), T. atroviride 296922 (SEQ ID NO: 788), A. niger CAK45422.1 (SEQ ID NO: 789), A. fumigatus EDP53789.1 (SEQ ID NO: 790), N. crassa XP . 958301.1 (SEQ ID NO: 791), M. thermophila XP_003664601.1 (SEQ ID NO: 792), M. graminicola XP_384993.1 (SEQ ID NO: 793), M. thermophila XP_003658945.1 (SEQ ID NO: 794), F. graminicola XP_382380.1 (SEQ ID NO: 795), A. niger XP_001395660.1 (SEQ ID NO: 796), M. thermophila XP_003659734.1 (SEQ ID NO: 797), N. crassa XP_964374.1 (SEQ ID NO: 798), A. fumigatus XP_756068.1 (SEQ ID NO: 799), A. oryzae EIT77098.1 (SEQ ID NO: 800), P. chrysogenum XP_002560028.1 (SEQ ID NO: 801), A. oryzae EIT71569.1 (SEQ ID NO: 802), A. nidulans CBF79006.1 (SEQ ID NO: 803), A. niger XP_001400740.2 (SEQ ID NO: 804), A. oryzae BAE57999.1 (SEQ ID NO: 805), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a sepl protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 786-805. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 786-805.

In some embodiments, sepl is T. reesei sepl. The amino acid sequence encoded by T. reesei sepl is set forth in SEQ ID NO: 786. In other embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more) to SEQ ID NO: 786. In further embodiments, the protease has 100% identity to SEQ ID NO: 786.

Zinc metalloprotease

Zinc metalloproteases are protease enzymes that require zinc for catalytic activity. Five metalloproteases have been identified in Trichoderma fungal cells: mpl (trel22703), mp2 (trel22576), mp3 (tre4308), mp4 (tre53343), mp5 (tre73809). mpl, mp2, mp3, mp4 and mp5

Examples of suitable mpl, mp2, mp3, mp4 and mp5 proteases include, without limitation, Trichoderma reesei mpl (SEQ ID NO: 875), Trichoderma reesei mp2 (SEQ ID NO: 876), Trichoderma reesei mp3 (SEQ ID NO:877), Trichoderma reesei mp4 (SEQ ID NO:878), Trichoderma reesei mp5 (SEQ ID NO:879), and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure, typically a mpl, mp2, mp3, mp4 or mp5 protease, has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 875-879. In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 875-879.

Other proteases

Examples of other suitable proteases include, without limitation,

Trichoderma reesei Group A protease selected from the group of protease65735 (SEQ ID NO: 894), protease77577 (SEQ ID NO: 895), protease81087 (SEQ ID NO:896), protease56920 (SEQ ID NO:900), proteasel22083 (SEQ ID NO:911), protease79485 (SEQ ID NO:910), protease 120998 (SEQ ID NO:901), or protease61127 (SEQ ID NO:912);

Trichoderma reesei Group B protease selected from the group of protease21659 (SEQ ID NO:905), protease58387 (SEQ ID NO:921), protease75159 (SEQ ID NO:918), protease56853 (SEQ ID NO:914), or protease64193 (SEQ ID NO:908); Trichoderma reesei Group C protease selected from the group of protease82452 (SEQ ID NO:906), protease80762 (SEQ ID NO:913), protease21668 (SEQ ID NO:919), protease81115 (SEQ ID NO:907), protease82141 (SEQ ID NO:902), protease23475 (SEQ ID NO:909);

Trichoderma reesei Group D protease selected from the group of proteasel21890 (903), protease22718 (SEQ ID NO:904), protease47127 (SEQ ID NO:899), protease61912 (SEQ ID NO:920), protease80843 (SEQ ID NO:897), protease66608 (SEQ ID NO:923), protease72612 (SEQ ID NO:898), protease40199 (SEQ ID NO:917); or

Trichoderma reesei Group E protease selected from the group of protease22210 (SEQ ID NO:915), proteasel 11694 (SEQ ID NO:916), protease82577 (SEQ ID NO: 922),

and homologs thereof.

Accordingly, in certain embodiments, a protease of the present disclosure has an amino acid sequence having 50% or more identity {e.g. 60%, 65%, 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, 99.5% or more to an amino acid sequence selected from SEQ ID NOs: 894-923). In some embodiments, the protease has 100% identity to an amino acid sequence selected from SEQ ID NOs: 894-923.

Homologous Proteases

Other embodiments of the present disclosure relate to reducing the activity of proteases that are homologous to the proteases of the present disclosure. "Homology" as used herein refers to sequence similarity between a reference sequence and at least a fragment of a second 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 below, BLAST will compare sequences based upon percent identity and similarity.

The terms "identical" or percent "identity," in the context of two or more 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. Optionally, 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.

For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, 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. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters. When comparing two sequences for identity, it is not necessary that the sequences be contiguous, but any gap would carry with it a penalty that would reduce the overall percent identity. For blastn, the default parameters are Gap opening penalty=5 and Gap extension penalty=2. For blastp, the default parameters are Gap opening penal ty=l 1 and Gap extension penal ty= 1.

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, WI), 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)].

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. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always < 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) or 10, M=5, N=-4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix [see Henikoff and Henikoff, (1992) Proc Natl Acad Sci USA 89(22): 10915-10919] alignments (B) of 50, expectation (E) of 10, M=5, N=-4, and a comparison of both strands.

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. For example, 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.

Other than percentage of sequence identity noted above, another indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross-reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below. Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.

As disclosed herein, proteases of the present disclosure may also include proteases that are conservatively modified variants of proteases encoded by the protease genes disclosed above. "Conservatively modified variants" as used herein 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)). Phylogenetic trees of aspartic, subtilisin, glutamic and sedolisin proteases of selected filamentous fungi are described in WO2013/102674.

Methods of Reducing the Activity of Proteases of the Invention

Further aspects of the present disclosure relate to reducing the activity of proteases found in filamentous fungal cells that express a heterologous polypeptide, such a mammalian polypeptide. The activity of proteases found in filamentous fungal cells can be reduced by any method known to those of skill in the art.

In some embodiments reduced activity of proteases is achieved by reducing the expression of the protease, for example, by promoter modification or RNAi.

In other embodiments, reduced activity of proteases is achieved by modifying the gene encoding the protease. Examples of such modifications include, without limitation, a 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.

In certain embodiments, 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. Such 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. For example, 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 of the present disclosure that are present in filamentous fungal cells that express a recombinant polypeptide 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. In such methods, 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.

The protease encoding genes of the present disclosure that are present in filamentous fungal cells that express a recombinant polypeptide 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. For example, 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. Biol 57: 157; Ho et al., 1989, Gene 77: 61; Horton et al., 1989, Gene 77: 61 ; and Sarkar and Sommer, 1990, BioTechniques 8: 404). Additionally, protease encoding genes of the present disclosure that are present in filamentous fungal cells that express a recombinant polypeptide 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 DNA between the duplicated regions. Such 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 of the present disclosure that are present in filamentous fungal cells that express a recombinant polypeptide may also be modified by the process of gene conversion (see, for example, Iglesias and Trautner, 1983, Molecular General Genetics 189:5 73-76). For example, in the 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. By homologous recombination, 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). In particular, 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.

In addition, 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 RNA interference (RNAi) techniques (see, for example, WO 2005/056772 and WO 2008/080017).

Protease encoding genes of the present disclosure that are present in filamentous fungal cells that express a recombinant polypeptide 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. Examples of physical and chemical mutagenizing agents include, without limitation, ultraviolet (UV) irradiation, hydroxyl amine, 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. When such agents are used, the mutagenesis is typically performed by incubating the 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.

In certain embodiments, 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%, at least 100%, at least 200%, at least 300%, at least 400%, at least 500%, at least 600%, at least 700%, at least 800%, at least 900%, at least 1,000%, or a higher percentage less protease activity compared to a corresponding non-modified protease.

In certain embodiments, for example, in a Trichoderma cell, the at least one mutation or modification in a protease encoding gene of the present disclosure results in a reduction of total protease activity to 49% or less, typically with a mutation in at least 2 distinct protease genes, or 31% or less, typically with a mutation in at least 3 distinct protease genes, or 13% or less, typically with a mutation in at least 4 distinct protease genes, or 10% or less, typically with a mutation in at least 5 distinct protease genes, or 6.3% or less, typically with a mutation in at least 6 distinct protease genes, or 5.5% or less, typically with a mutation in at least 7 distinct protease genes, of the total protease activity of the corresponding parental Trichoderma cell.

Heterologous Polypeptides of the Invention

The invention herein further relates to increasing the production of heterologous polypeptides in filamentous fungal cells that express such heterologous polypeptides by reducing the activity of proteases found in the cells. As used herein a "heterologous polypeptide" refers to a polypeptide that is not naturally found in (i.e., endogenous) a filamentous fungal cell of the present disclosure, or that is expressed at an elevated level in a filamentous fungal cell as compared to the endogenous version of the polypeptide. In certain embodiments, the heterologous polypeptide is a mammalian polypeptide. In other embodiments, the heterologous polypeptide is a non-mammalian polypeptide. Mammalian Polypeptides

Mammalian polypeptides of the present disclosure may be any mammalian polypeptide having a biological activity of interest. As used herein, a "mammalian polypeptide" is a polypeptide that is natively expressed in a mammal, a polypeptide that is derived from a polypeptide that is natively expressed in a mammal, or a fragment thereof. A mammalian polypeptide also includes peptides and oligopeptides that retain biological activity. Mammalian polypeptides of the present disclosure may also include two or more polypeptides that are combined to form the encoded product. Mammalian polypeptides of the present disclosure may further include fusion polypeptides, which contain a combination of partial or complete amino acid sequences obtained from at least two different polypeptides. Mammalian polypeptides may also include naturally occurring allelic and engineered variations of any of the disclosed mammalian polypeptides and hybrid mammalian polypeptides.

The mammalian polypeptide may be a naturally glycosylated polypeptide or a naturally non- glycosylated polypeptide.

Examples of suitable mammalian polypeptides include, without limitation, immunoglobulins, antibodies, antigens, antimicrobial peptides, enzymes, growth factors, hormones, interferons, cytokines, interleukms, immunodilators, neurotransmitters, receptors, reporter proteins, structural proteins, and transcription factors.

Specific examples of suitable mammalian polypeptides include, without limitation, immunoglobulins, immunoglobulin heavy chains, immunoglobulin light chains, monoclonal antibodies, hybrid antibodies, F(ab')2 antibody fragments, F(ab) antibody fragments, Fv molecules, single-chain Fv antibodies, dimeric antibody fragments, trimeric antibody fragments, functional antibody fragments, immunoadhesins, insulin-like growth factor 1, growth hormone, insulin, interferon alpha 2b, fibroblast growth factor 21, human serum albumin, camelid antibodies and/or antibody fragments, single domain antibodies, multimeric single domain antibodies, and erythropoietin. Other examples of suitable mammalian proteins include, without limitation, an oxidoreductase, a transferase, a hydrolase, a lyase, an isomerase, a ligase, an aminopeptidase, an amylase, a carbohydrase, a carboxypeptidase, a catalase, a glycosyltransferase, a deoxyribonuclease, an esterase, a galactosidase, a betagalactosidase, a glucosidase, a glucuronidase, a glucuronoyl esterase, a haloperoxidase, an invertase, a lipase, an oxidase, a phospholipase, a proteolytic enzyme, a ribonuclease, a urokinase, an albumin, a collagen, a tropoelastin, and an elastin.

Non-mammalian Polypeptides Non-mammalian polypeptides of the present disclosure may be any non-mammalian polypeptide having a biological activity of interest. As used herein, a "non-mammalian polypeptide" is a polypeptide that is natively expressed in a non-mammalian organism, such as a fungal cell, a polypeptide that is derived from a polypeptide that is natively expressed in a non-mammal organism, or a fragment thereof. A non-mammalian polypeptide also includes peptides and oligopeptides that retain biological activity. Non-mammalian polypeptides of the present disclosure may also include two or more polypeptides that are combined to form the encoded product. Non-mammalian polypeptides of the present disclosure may further include fusion polypeptides, which contain a combination of partial or complete amino acid sequences obtained from at least two different polypeptides. Non-mammalian polypeptides may also include naturally occurring allelic and engineered variations of any of the disclosed non-mammalian polypeptides and hybrid non-mammalian polypeptides.

Examples of suitable non-mammalian polypeptides include, without limitation, aminopeptidases, amylases, carbohydrases, carboxypeptidases, catalases, cellulases, chitinases, cutinases, deoxyribonucleases, esterases, alpha-galactosidases, beta-galactosidases, glucoamylases, alpha- glucosidases, beta-glucosidases, invertases, laccases, lipases, mutanases, oxidases, pectinolytic enzymes, peroxidases, phospholipases, phytases, polyphenoloxidases, proteolytic enzymes, ribonucleases, transglutaminases and xylanases.

Heterologous Polypeptide Production A heterologous polypeptide of interest is produced by filamentous fungal cells of the present disclosure containing at least three proteases having reduced activity by cultivating the cells in a nutrient medium for production of the heterologous polypeptide using methods known in the art. For example, the cells may be cultivated by shake flask cultivation, small-scale or large-scale fermentation (including continuous, batch, fed-batch, or solid state fermentations) in laboratory or industrial fermentors performed in a suitable medium and under conditions allowing the polypeptide to be expressed and/or isolated. The cultivation takes place in a suitable nutrient medium comprising carbon and nitrogen sources and inorganic salts, using procedures known in the art. Suitable media are available from commercial suppliers or may be prepared according to published compositions (e.g., in catalogues of the American Type Culture Collection). The secreted polypeptide can be recovered directly from the medium. If the polypeptide is not secreted, it may be obtained from cell lysates.

A heterologous polypeptide of interest produced by a filamentous fungal cell of the present disclosure containing at least three proteases having reduced activity may be detected using methods known in the art that are specific for the heterologous polypeptide. These detection methods may include, without limitation, use of specific antibodies, high performance liquid chromatography, capillary chromatography, formation of an enzyme product, disappearance of an enzyme substrate, and SDS-PAGE. For example, an enzyme assay may be used to determine the activity of an enzyme. Procedures for determining enzyme activity are known in the art for many enzymes (see, for example, O. Schomburg and M. Salzmann (eds.), Enzyme Handbook, Springer- Verlag, New York, 1990).

The resulting heterologous polypeptide may be isolated by methods known in the art. For example, a heterologous polypeptide of interest may be isolated from the cultivation medium by conventional procedures including, without limitation, centrifugation, filtration, extraction, spray- drying, evaporation, and precipitation. The isolated heterologous polypeptide may then be further purified by a variety of procedures known in the art including, without limitation, chromatography (e.g., ion exchange, affinity, hydrophobic, chromatofocusing, and size exclusion), electrophoretic procedures (e.g., preparative isoelectric focusing (IEF), differential solubility (e.g., ammonium sulfate precipitation), or extraction (see, for example, Protein Purification, J.-C. Janson and Lars Ryden, editors, VCH Publishers, New York, 1989).

Preparation of Polynucleotides Encoding Heterologous Polypeptides

Sequences of the heterologous polynucleotides of the present disclosure are prepared by any suitable method known in the art, including, without limitation, direct chemical synthesis or cloning. For direct chemical synthesis, formation of a polymer of nucleic acids typically involves sequential addition of 3 '-blocked and 5 '-blocked nucleotide monomers to the terminal 5'- hydroxyl group of a growing nucleotide chain, wherein each addition is effected by nucleophilic attack of the terminal 5'-hydroxyl group of the growing chain on the 3 '-position of the added monomer, which is typically a phosphorus derivative, such as a phosphotriester, phosphoramidite, or the like. Such methodology is known to those of ordinary skill in the art and is described in the pertinent texts and literature [e.g., in Matteucci et al., (1980) Tetrahedron Lett 21:719-722; U.S. Pat. Nos. 4,500,707; 5,436,327; and 5,700,637]. In addition, the desired sequences may be isolated from natural sources by splitting DNA using appropriate restriction enzymes, separating the fragments using gel electrophoresis, and thereafter, recovering the desired nucleic acid sequence from the gel via techniques known to those of ordinary skill in the art, such as utilization of polymerase chain reactions (PCR; e.g., U.S. Pat. No. 4,683,195).

Each heterologous polynucleotide of the present disclosure can be incorporated into an expression vector. "Expression vector" or "vector" refers to a compound and/or composition that transduces, transforms, or infects a host cell, thereby causing the cell to express nucleic acids and/or proteins other than those native to the cell, or in a manner not native to the cell. An "expression vector" contains a sequence of nucleic acids (ordinarily RNA or DNA) to be expressed by the host cell. Optionally, the expression vector also includes materials to aid in achieving entry of the nucleic acid into the host cell, such as a virus, liposome, protein coating, or the like. The expression vectors contemplated for use in the present disclosure include those into which a nucleic acid sequence can be inserted, along with any preferred or required operational elements. Further, the expression vector must be one that can be transferred into a host cell and replicated therein. Preferred expression vectors are plasmids, particularly those with restriction sites that have been well documented and that contain the operational elements preferred or required for transcription of the nucleic acid sequence. Such plasmids, as well as other expression vectors, are well known in the art.

Incorporation of the individual polynucleotides may be accomplished through known methods that include, for example, the use of restriction enzymes (such as BamHI, EcoRI, Hhal, Xhol,

Xmal, and so forth) to cleave specific sites in the expression vector, e.g., plasmid. The restriction enzyme produces single stranded ends that may be annealed to a polynucleotide having, or synthesized to have, a terminus with a sequence complementary to the ends of the cleaved expression vector. Annealing is performed using an appropriate enzyme, e.g., DNA ligase. As will be appreciated by those of ordinary skill in the art, both the expression vector and the desired polynucleotide are often cleaved with the same restriction enzyme, thereby assuring that the ends of the expression vector and the ends of the polynucleotide are complementary to each other. In addition, DNA linkers maybe used to facilitate linking of nucleic acids sequences into an expression vector.

A series of individual polynucleotides can also be combined by utilizing methods that are known t in the art {e.g., U.S. Pat. No. 4,683,195).

For example, each of the desired polynucleotides can be initially generated in a separate PCR. Thereafter, specific primers are designed such that the ends of the PCR products contain complementary sequences. When the PCR products are mixed, denatured, and reannealed, the strands having the matching sequences at their 3' ends overlap and can act as primers for each other. Extension of this overlap by DNA polymerase produces a molecule in which the original sequences are "spliced" together. In this way, a series of individual polynucleotides may be "spliced" together and subsequently transduced into a host cell simultaneously. Thus, expression of each of the plurality of polynucleotides is affected.

Individual polynucleotides, or "spliced" polynucleotides, are then incorporated into an expression vector. The present disclosure is not limited with respect to the process by which the polynucleotide is incorporated into the expression vector. Those of ordinary skill in the art are familiar with the necessary steps for incorporating a polynucleotide into an expression vector. A typical expression vector contains the desired polynucleotide preceded by one or more regulatory regions, along with a ribosome binding site, e.g., a nucleotide sequence that is 3-9 nucleotides in length and located 3-11 nucleotides upstream of the initiation codon in E. coli. See Shine and Dalgarno (1975) Nature 254(5495):34-38 and Steitz (1979) Biological Regulation and Development (ed. Goldberger, R. R), 1:349-399 (Plenum, New York).

The term "operably linked" as used herein refers to a configuration in which a control sequence is placed at an appropriate position relative to the coding sequence of the DNA sequence or polynucleotide such that the control sequence directs the expression of a polypeptide. Regulatory regions include, for example, those regions that contain a promoter and an operator. A promoter is operably linked to the desired polynucleotide, thereby initiating transcription of the polynucleotide via an RNA polymerase enzyme. An operator is a sequence of nucleic acids adjacent to the promoter, which contains a protein-binding domain where a repressor protein can bind. In the absence of a repressor protein, transcription initiates through the promoter. When present, the repressor protein specific to the protein-binding domain of the operator binds to the operator, thereby inhibiting transcription. In this way, control of transcription is accomplished, based upon the particular regulatory regions used and the presence or absence of the corresponding repressor protein. Examples include lactose promoters (Lad repressor protein changes conformation when contacted with lactose, thereby preventing the Lad repressor protein from binding to the operator) and tryptophan promoters (when complexed with tryptophan, TrpR repressor protein has a conformation that binds the operator; in the absence of tryptophan, the TrpR repressor protein has a conformation that does not bind to the operator). Another example is the tac promoter (see de Boer et al, (1983) Proc Natl Acad Sci USA 80(l):21-25). As will be appreciated by those of ordinary skill in the art, these and other expression vectors may be used in the present disclosure, and the present disclosure is not limited in this respect.

Although any suitable expression vector may be used to incorporate the desired sequences, readily available expression vectors include, without limitation: plasmids, such as pSClOl, pBR322, pBBRlMCS-3, pUR, pEX, pMRlOO, pCR4, pBAD24, pUC19, pRS426; and bacteriophages, such as Ml 3 phage and λ phage. Of course, such expression vectors may only be suitable for particular host cells. One of ordinary skill in the art, however, can readily determine through routine experimentation whether any particular expression vector is suited for any given host cell. For example, the expression vector can be introduced into the host cell, which is then monitored for viability and expression of the sequences contained in the vector. In addition, reference may be made to the relevant texts and literature, which describe expression vectors and their suitability to any particular host cell.

Suitable expression vectors for the purposes of the invention, including the expression of the desired heterologous polypeptide, enzyme, and one or more catalytic domains described herein, include expression vectors containing the polynucleotide encoding the desired heterologous polypeptide, enzyme, or catalytic domain(s) operably linked to a constitutive or an inducible promoter. Examples of particularly suitable promoters for operable linkage to such polynucleotides include promoters from the following genes: gpdA, cbhl, Aspergillus oryzae TAKA amylase, Rhizomucor miehei aspartic proteinase, Aspergillus niger neutral alpha-amylase, Aspergillus niger acid stable alpha-amylase, Aspergillus niger glucoamylase (glaA), Aspergillus awamori glaA, Rhizomucor miehei lipase, Aspergillus oryzae alkaline protease, Aspergillus oryzae triose phosphate isomerase, Aspergillus nidulans acetamidase, Aspergillus oryzae acetamidase, Fusarium oxysporum trypsin-like protease, fungal endo a-L-arabinase (abnA), fungal α-L-arabinofuranosidase A (abfA), fungal α-L-arabinofuranosidase B (abfB), fungal xylanase (xlnA), fungal phytase, fungal ATP-synthetase, fungal subunit 9 (oliC), fungal triose phosphate isomerase (tpi), fungal alcohol dehydrogenase (adhA), fungal a-amylase (amy), fungal amyloglucosidase (glaA), fungal acetamidase (amdS), fungal glyceraldehyde-3-phosphate dehydrogenase (gpd), yeast alcohol dehydrogenase, yeast lactase, yeast 3-phosphoglycerate kinase, yeast triosephosphate isomerase, bacterial a-amylase, bacterial Spo2, and SSO. Examples of such suitable expression vectors and promoters are also described in WO2012/069593, the entire contents of which is hereby incorporated by reference herein.

Pharmaceutical Compositions Containing Heterologous Polypeptides Produced by Filamentous Fungal Cells of the Invention

In another aspect, the present invention provides a composition, e.g., a pharmaceutical composition, containing one or more heterologous polypeptides of interest, such as mammalian polypeptides, produced by the filamentous fungal cells of the present disclosure having reduced activity of at least three proteases and further containing a recombinant polynucleotide encoding the heterologous polypeptide, formulated together with a pharmaceutically acceptable carrier. Pharmaceutical compositions of the invention also can be administered in combination therapy, i.e., combined with other agents. For example, the combination therapy can include a mammalian polypeptide of interest combined with at least one other therapeutic agent.

As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound, i.e., the mammalian polypeptide of interest, may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.

The pharmaceutical compositions of the invention may include one or more pharmaceutically acceptable salts. A "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S.M., et al. (1977) J. Pharm. Sci. 66: 1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as Ν,Ν'-dibenzylethylenediamine, N- methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.

A pharmaceutical composition of the invention also may also include a pharmaceutically acceptable antioxidant. Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

Examples of suitable aqueous and nonaqueous carriers that may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of presence of microorganisms may be ensured both by sterilization procedures, and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. The use of such media and agents for pharmaceutically active substances is known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the pharmaceutical compositions of the invention is contemplated. Supplementary active compounds can also be incorporated into the compositions.

Therapeutic compositions typically must be sterile and stable under the conditions of manufacture and storage. The composition can be formulated as a solution, microemulsion, liposome, or other ordered structure suitable to high drug concentration. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin. Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by sterilization microfiltration. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the certain methods of preparation are vacuum drying and freeze-drying (lyophilization) that yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.

The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the subject being treated, and the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the composition which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.01 percent to about ninety-nine percent of active ingredient, preferably from about 0.1 percent to about 70 percent, most preferably from about 1 percent to about 30 percent of active ingredient in combination with a pharmaceutically acceptable carrier.

Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such an active compound for the treatment of sensitivity in individuals. For administration of a mammalian polypeptide of interest, in particular where the mammalian polypeptide is an antibody, the dosage ranges from about 0.0001 to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight. For example, dosages can be 0.3 mg/kg body weight, 1 mg/kg body weight, 3 mg/kg body weight, 5 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10 mg/kg. An exemplary treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to 6 months. Certain dosage regimens for an antibody may include 1 mg/kg body weight or 3 mg/kg body weight via intravenous administration, with the antibody being given using one of the following dosing schedules: (i) every four weeks for six dosages, then every three months; (ii) every three weeks; (iii) 3 mg/kg body weight once followed by 1 mg/kg body weight every three weeks.

Alternatively a mammalian polypeptide of interest can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the administered substance in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.

Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present disclosure may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.

A "therapeutically effective dosage" of an immunoglobulin of the present disclosure preferably results in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction. For example, for the treatment of tumors, a "therapeutically effective dosage" preferably inhibits cell growth or tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. The ability of a compound to inhibit tumor growth can be evaluated in an animal model system predictive of efficacy in human tumors. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit, such inhibition in vitro by assays known to the skilled practitioner. A therapeutically effective amount of a therapeutic compound can decrease tumor size, or otherwise ameliorate symptoms in a subject. One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.

A composition of the present disclosure can be administered via one or more routes of administration using one or more of a variety of methods known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results. Certain routes of administration for binding moieties of the invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase "parenteral administration" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion. Alternatively, a mammalian polypeptide according to the present disclosure can be administered via a nonparenteral route, such as a topical, epidermal or mucosal route of administration, for example, intranasally, orally, vaginally, rectally, sublingually or topically.

The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release formulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. (see, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978).

Therapeutic compositions can be administered with medical devices known in the art. For example, in a certain embodiment, a therapeutic composition of the invention can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Patent Nos. 5,399,163; 5,383,851 ; 5,312,335; 5,064,413; 4,941,880; 4,790,824; or 4,596,556. Examples of well-known implants and modules useful in the present invention include: U.S. Patent No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Patent No. 4,486,194, which discloses a therapeutic device for administering medicants through the skin; U.S. Patent No. 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Patent No. 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Patent No. 4,439,196, which discloses an osmotic drug delivery system having multi-chamber compartments; and U.S. Patent No. 4,475,196, which discloses an osmotic drug delivery system.

In certain embodiments, the use of mammalian polypeptides according to the present disclosure is for the treatment of any disease that may be treated with therapeutic antibodies. Filamentous Fungal Cells of the Invention

The invention herein also relates to increasing the levels of production of heterologous polypeptides, such as mammalian polypeptides, in filamentous fungal cells by reducing or eliminating the activity of at least three proteases found in cells that express heterologous polypeptides, and that catalyze the degradation of the heterologous polypeptides. Reducing or eliminating the activity of proteases found in the filamentous fungal cells that express heterologous polypeptides increases the stability of the expressed recombinant polypeptides, which results in an increased level of production of the heterologous polypeptides. The activity of the proteases found in the filamentous fungal cells may be reduced, for example, by modifying the genes encoding the proteases.

"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.

Any filamentous fungal cell may be used in the present disclosure so long as it remains viable after being transformed with a sequence of nucleic acids and/or being modified or mutated to decrease protease activity. Preferably, 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.

Examples of suitable filamentous fungal cells include, without limitation, cells from an Acremonium, Aspergillus, Fusarium, Humicola, Mucor, Myceliophthora, Neurospora, Penicillium, Scytalidium, Thielavia, Tolypocladium, or Trichoderma strain. In certain embodiments, 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, P enicillium, Piromyces, Schizophyllum, Talaromyces, Thermoascus, Thielavia, or Tolypocladium 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.

In certain embodiments, the filamentous fungal cell is not an Aspergillus cell.

In certain embodiments, the filamentous fungal cell is selected from the group consisting of Trichoderma (T. reesei), Neurospora (N. crassa), Penicillium (P. chrysogenum), Aspergillus (A. nidulans, A. niger and A. oryzae), Myceliophthora (M. thermophila) and Chrysosporium (C. lucknowense).

In certain embodiments, 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. Examples of 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).

General methods to disrupt genes of and cultivate filamentous fungal cells are disclosed, for example, for Penicillium, in Kopke et al. (2010) Application of the Saccharomyces cerevisiae FLP/FRT recombination system in filamentous fungi for marker recycling and construction of knockout strains devoid of heterologous genes. Appl Environ Microbiol. 76(14):4664-74. doi: 10.1128/AEM.00670-10, for Aspergillus, in Maruyama and Kitamoto (2011), Targeted Gene Disruption in Koji Mold Aspergillus oryzae, in James A. Williams (ed.), Strain Engineering: Methods and Protocols, Methods in Molecular Biology, vol. 765, DOI 10.1007/978-1-61779- 197-0_27; for Neurospora, in Collopy et al. (2010) High-throughput construction of gene deletion cassettes for generation of Neurospora crassa knockout strains. Methods Mol Biol. 2010;638:33- 40. doi: 10.1007/978-1-60761-611-5_3; and for Myceliophthora or Chrysosporium PCT/NL2010/000045 and PCT/EP98/06496.

Filamentous Fungal Cell Components

Certain aspects of the present disclosure relate to filamentous fungal cells having reduced or no detectable activity of at least three proteases and having a recombinant polynucleotide encoding a heterologous polypeptide that is produced at increased levels, for example at least two-fold increased levels. Other aspects of the present disclosure relate to Trichoderma or closely related species fungal cells that has reduced or no detectable protease activity of at least two, three or four proteases selected from pep9, ampl, amp2 and sepl. Other aspects of the present disclosure relate to Trichoderma or closely related species fungal cells that has reduced or no detectable protease activity in one or more of the following proteases selected from pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl. Other aspects of the present disclosure relate to Trichoderma or closely related species fungal cells that has reduced or no detectable protease activity of at least two, three or four proteases selected from proteases selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pep!2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide produced at a level of at least 2- fold higher than the production level of the polypeptide in a corresponding parental Trichoderma fungal cell. In certain embodiments, the filamentous fungal cells or Trichoderma fungal cells have reduced or no activity of at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, at least eleven, at least twelve, or more proteases. Reduced expression of proteases

The reduced activity of the at least three proteases in filamentous fungal cells or Trichoderma fungal cells of the present disclosure can be the result of reduced or eliminated expression of the proteases. In some embodiments, the reduced or eliminated expression of the at least three proteases is the result of a modification to the catalytic domain, the coding region, or a control sequence required for expression of the coding region of the genes encoding each of the proteases. In other embodiments, the reduced or eliminated expression of the proteases is the result of introducing, substituting, and/or removing one or more nucleotides in the genes, or a control sequence thereof required for the transcription or translation of the genes encoding each of the proteases. In some embodiments, the reduced or eliminated expression of the proteases is the result of introducing, recombining, or replacing the endogenous promoter, or part thereof, of the protease with a heterologous promoter. The "heterologous promoter" herein means an operatively linked promoter DNA sequence which is non-native for the protease. In some embodiments, the heterologous promoter reduces protease activity compared to the activity in the corresponding parental filamentous fungal cell in which the protease is expressed by its endogenous promoter. If deleting the protease affects growth, sporulation or function of the fungus, the heterologous promoter be selected in such a way that the protease is expressed, for example, during essential phase of the cell, for example sporulation, but the protease expression is reduced or eliminated during production of heterologous protein compared to the corresponding parental strain in which the protease is expressed by its endotenous promoter.

In some embodiments, the heterologous promoter is a constitutive promoter or an inducible promoter. In some embodiments, the heterologous promoter is selected from the group of flavin containing monooxygenase gene (Tre76230), the RNA polymerase gene (Tre49048), and the slp8 protease gene (Tre58698).

In some embodiments, the reduced or eliminated protease with a heterologous promoter is slp2, e.g., slp2 in Trichoderma fungal cell or closely related species. In some specific embodiments, the endogenous promoter of slp2, e.g. in Trichoderma or closely related species, is replaced with a heterologous promoter selected from the group of flavin containing monooxygenase gene (Tre76230), the RNA polymerase gene (Tre49048), and the slp8 protease gene (Tre58698). More details are given in Example 41. In some embodiments, a filamentous fungal cell of the invention comprises at least one endogenous protease having a heterologous promoter and having reduced or no protease activity, and a recombinant polynucleotide encoding a heterologous polypeptide, wherein the cell has reduced or no protease activity in one or more of following proteases: pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl, and, optionnaly one or more additional proteases selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2.

In further embodiments, the reduced or eliminated expression of the proteases is the result of inserting into the genes encoding each of the proteases disruptive nucleic acid constructs each containing a nucleic acid fragment homologous to each of the genes that will create a duplication of the region of homology and incorporate construct DNA between the duplicated regions. In other embodiments, the reduced or eliminated expression of the proteases is the result of gene conversion of the genes encoding each of the proteases. In still other embodiments, the reduced or eliminated expression of the proteases is the result of by anti-sense polynucleotides or RNAi constructs that are specific for the each of the genes encoding each of the proteases. In one embodiment, 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 tspl, a glutamic protease such as a gap-type protease, a subtilisin protease such as a sip-type, protease, or a sedolisin protease such as a tppl or a slp7 protease. In one embodiment, an RNAi construct is specific for the gene encoding a sip-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 proteasess, any one of the sip-type proteases, any one of the gap-type proteases, any one of the metalloproteases and/or any one of the sedolisin proteases. In one embodiment, two or more proteases are slp2, slp3, slp5 and/or slp6. In one embodiment, RNAi construct comprises any one of nucleic acid sequences of Table 22.2.

In some embodiments, the genes encoding the proteases each contain a mutation that reduces or eliminates the corresponding protease activity. In other embodiments, the mutation reduces or eliminates the expression of each of the proteases. In further embodiments, the mutation is a 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, an inversion mutation that reduces or eliminates the corresponding protease activity.

In some embodiments, the mutation is a deletion of the protease encoding gene. In other embodiments, the mutation is a deletion of the portion of the protease encoding gene encoding the catalytic domain of the protease. In still other embodiments, the mutation is point mutation in the portion of the protease encoding gene encoding the catalytic domain of the protease.

Another method to disrupt protease genes of filamentous fungal cells include CRISPR-CAS system, or clustered regularly interspaced short palindromic repeats. CRISPR-Cas system is a novel technique of gene editing (silencing, enhancing or changing specific genes). By inserting a plasmid containing cas9 genes and specifically designed CRISPRs, the organism's genome can be cut at any desired location. Cas9 gene originates from the type II bacterial CRISPR system of Streptococcus pyogenes. Gene product, CAS9 nuclease, complexes with a specific genome targeting CRISPR guideRNA and has high site specificity of the DNA cutting activity. It has been shown recently that CAS9 can function as an RNA-guided endonuclease in various heterologous organisms (Mali et al. 2013: Rna guided human genome engineering via Cas9. Science 339:823-826; Cong et al 2013: Multiplex genome engineering using CRISPR-Cas systems. Science 339:819-823; Jiang et al 2013: RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat Biotechnol 31 :233-239; Jinek et al. 2013: RNA programmed genome editing in human cells. eLife 2:e00471 ; Hwang et al. 2013: Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat Biotech 31:227-279. DiCarlo et al 2013: Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems. NAR 41:4336-4343).

GuideRNA synthesis have been usually carried out from promoters transcribed by RNA polymerase III, most commonly used being SNR52 snoRNA promoter in yeasts and U3/U6 snoRNA promoters in plants and animals. Promoters transcribed by RNA polymerase II have been considered to be unsuitable for guideRNA synthesis because of the posttranscriptional modifications, 5'capping, 573' UTR's and poly A tailing. However, it has been recently demonstrated that RNA polymerase II type promoters can be used if the guideRNA sequence is flanked with self-processing ribozyme sequences. Primary transcript then undergoes self- catalyzed cleavage and generates desired gRNA sequence (Gao and Zhao 2014: Self processing of ribozyme-flanked RNAs into guide RNA's in vitro and in vivo for CRISPR-mediated genome editing. Journal of Integrative Plant Biology e-publication ahead of print; March 2014). Example 21 exemplifies methods to disrupt various protease encoding genes that affect and/or hinder efficient production of heterologous proteins in T. reesei. GuideRNA sequences as shown in Table 21.1 and their use for disrupting corresponding protease genes in Trichoderma cells are also part of the invention.

Combinations of protease genes The filamentous fungal cells or Trichoderma fungal cells of the present disclosure may contain at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or more aspartic proteases, trypsin-like serine proteases, subtilisin proteases, glutamic proteases, and metallopro teases. In certain embodiments, the proteases are encoded by pep-type protease genes, gap-type protease genes, sip-type proteases genes, amp-type protease,sep-type protease and mp-type proteases. In some embodiments, the pep-type protease genes are selected from pepl, pep2, pep3, pep4, pep5, pep6, pep8, pep 9, peplO, pepll, and pepl2, pepl3, pepl4, pepl6. In other embodiments, the gap-type protease genes are selected from gapl, and gap2. In other embodiments, the gap-type protease genes are selected from gapl, gap2, gap3 or gap4. In further embodiments, the sip-type, proteases genes are selected from slpl, slp2, slp3, and slp7; or are selected from slpl, slp2, slp3, slp5, slp6, slp7, and slp8, or are selected from slpl, slp2, slp3, slp5, slp6, slp7, and slp8 and slp57433, slp35726, slp60791, or slp!09276. In certain preferred embodiments, the sip-type proteases gene is slpl. In certain embodiments, the amp-type proteases are selected from ampl and amp2. In further embodiments, the sep-type proteases are selected from sepl, sed2, sed3, and sed5. In further emboduments, the mp-type metalloproteases are selected from mpl, mp2, mp3, mp4 and mp5. In other embodiments, the proteases are encoded by genes selected from pep9, ampl, amp2, sepl, pepl, pep2, pep3, pep4, pep5, pep7, pep8, pepll, pep!2, tspl, slpl, slp2, slp3, slp5, slp6, slp7, slp8, gapl, gap2, and tppl. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of at least two, three or at least four protease of a first group of protease selected from pep9, ampl, amp2 and sepl, optionally in combination with a second group of protease encoding genes selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels in one or more of the following protease selected from pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl, optionally in combination with a second group of protease encoding genes selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2. In certain embodiments, the filamentous fungal cell, for example a Trichoderma cell, has reduced or no expression levels of at least three protease encoding genes selected from pepl, tspl, and slpl. In other embodiments, the filamentous fungal cell, or Trichoderma cell, has reduced or no expression levels of at least three protease encoding genes selected from gapl, slpl and pepl. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, and gapl. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, and pep4. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, and slpl. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, and slp3. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, slp3, and pep3. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, slp3, pep3, and pep2. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl , pep4, slpl, slp3, pep3, pep2, and pep5. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, slp3, pep3, pep2, pep5, and tspl. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, slp3, pep3, pep2, pep5, tspl, and slp7. In some embodiments, the cell, for example a Trichoderma cell, has reduced or no protease activity in at least twelve proteases, each of the genes encoding the twelve proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the twelve proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7. In other embodiments, the cell, for example a Trichoderma cell has either

(i) reduced or no protease activity in at least thirteen proteases, each of the genes encoding the thirteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the thirteen proteases are either pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, pep9; pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7; or pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp3;

(ii) the cell has reduced or no protease activity in at least fourteen proteases, each of the genes encoding the fourteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fourteen proteases are pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2;

(iii) the cell has reduced or no protease activity in at least fifteen proteases, each of the genes encoding the fifteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fifteen proteases are either

pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mpl ; or, pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mp5.

In other embodiments, the filamentous fungal cell of the invention, for example a Trichoderma cell, has reduced or no protease activity in at least sixteen, at least seventeen, at least eighteen, at least nineteen, or at least twenty or more proteases, with 13, 14, or 15 mutations in the above recited proteases, and further comprising one or more additional mutations, each additional mutation reduces or eliminates a corresponding additional protease activity, said additional protease being selected from the group consisting of

an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5;

Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, slp3, pep3, pep2, pep5, tspl, slp7, and slp8. In some embodiments, the filamentous fungal cell, for example, a Trichoderma cell has reduced or no expression levels of protease encoding genes slp2, pepl, gapl, pep4, slpl, slp3, pep3, pep2, pep5, tspl, slp7, slp8, and gap2.

In certain embodiments, the filamentous fungal cell has at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least ten, or more proteases with reduced protease activity, wherein the corresponding proteases with wild type activity each have an amino acid 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 the amino acid sequences of SEQ ID NOs: 1-16; 17-36; 37-57; 58-65; 66-81 ; 82-97; 98-117; 118-128; 129-144; 166-181; 182-185; 491-588, SEQ ID NOs 750-805 or SEQ ID NOs 875-923. In embodiments where the filamentous fungal cell is a Trichoderma fungal cell with reduced protease activity in one or more proteases, wherein the corresponding proteases with wild type activity each have an amino acid 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 the amino acid sequences of SEQ ID NOs: 1, 17, 37, 58, 66, 82, 98, 118, 129, 166, 182, 507, 522, 530, 750, 759, 775, SEQ ID NO:786, or SEQ ID NOs 875-923.

Heterologous polypeptides The filamentous fungal cells or Trichoderma fungal cells of the present disclosure contain a recombinant polynucleotide encoding a heterologous polypeptide. In certain embodiments, the heterologous polypeptide is a mammalian polypeptide. In other embodiments, heterologous polypeptide is a non-mammalian polypeptide.

In embodiments where the filamentous fungal cell contains a recombinant polynucleotide encoding a mammalian polypeptide, the mammalian polypeptide can be a non-glycosylated mammalian polypeptide, a glycosylated mammalian polypeptide, or combinations thereof, including, without limitation, an immunoglobulin, an antibody, a growth factor, and an interferon. In some embodiments, the mammalian polypeptide is an immunoglobulin or antibody. In embodiments where the filamentous fungal cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody, the filamentous fungal cell, for example, a Trichoderma fungal cell may have reduced or no expression of at least two, three or at least four protease encoding genes selected from pep9, ampl, amp2 and sepl, optionally in combination with at least three or four proteases selected from pepl, pep3, pep4, pep8, pepll, pepl2, tspl, slpl, slp2, slp7, gapl, and gap2. In some embodiments where the filamentous fungal cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody, for example, a Trichoderma cell, said cell has reduced or no expression levels in one or more of the following protease selected from pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl, optionally in combination with one or more additional proteases encoding genes selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2. In certain preferred embodiments, the cell, for example a Trichoderma fungal cell, contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced or no expression of the protease encoding genes pepl, tspl, slpl, and gapl. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes pepl, tspl, slpl, gapl, and pep4. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, and slp3. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, and tspl. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, tspl, and pepl. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, tspl, pepl, and gapl. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, tspl, pepl, gapl, and pep4. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, tspl, pepl, gapl, pep4, and pep3. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, tspl, pepl, gapl, pep4, pep3, and pep2. In other embodiments, the cell contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced expression of the protease encoding genes slpl, slp2, slp3, tspl, pepl, gapl, pep4, pep3, pep2, and pep5. In some embodiments, the cell, for example a Trichoderma cell, contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced or no protease activity in at least twelve proteases, each of the genes encoding the twelve proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the twelve proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7. In other embodiments, the cell, for example a Trichoderma cell, contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has either

(i) reduced or no protease activity in at least thirteen proteases, each of the genes encoding the thirteen proteases comprises a mutation that reduces or eliminates the

corresponding protease activity, and the thirteen proteases are either

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, pep9; pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7;

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp3;

(ii) reduced or no protease activity in at least fourteen proteases, each of the genes encoding the fourteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fourteen proteases are pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2; or,

(iii) reduced or no protease activity in at least fifteen proteases, each of the genes encoding the fifteen proteases comprises a mutation that reduces or eliminates the

corresponding protease activity, and the fifteen proteases are either

pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mpl ; or, pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mp5. In other embodiments, the filamentous fungal cell, for example a Trichoderma cell, contains a recombinant polynucleotide encoding an immunoglobulin or antibody and has reduced or no protease activity in at least sixteen, at least seventeen, at least eighteen, at least nineteen, or at least twenty or more proteases, with 13, 14 or 15 mutations in the above recited proteases, and further comprising one or more additional mutation, each additional mutation reduces or eliminates the corresponding additional protease activity, and said at least additional protease being selected from the group consisting of

an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5; Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

In other embodiments, the filamentous fungal cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, or interleukin. In embodiments where the filamentous fungal cell, for example a Trichoderma fungal cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin, the filamentous fungal cell may have reduced or no expression of at least two, three or four proteases selected from pep9, ampl, amp2 and sepl, optionally in combination with at least three or four proteases selected from pepl, pep3, pep4, pep8, pepll, pepl2, tspl, slpl, slp2, slp7, gapl, and gap2 or at least three or at least four protease encoding genes selected from pepl, pep2, pep3, pep4, pep5, pep8, gapl, gap2, slpl, slp2, slp7, and tspl.

In some embodiments where the filamentous fungal cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin, for example, a Trichoderma cell, said cell has reduced or no expression levels in one or more of the following protease selected from pep9, ampl, amp2, mpl, mp2, mp3, mp4, mp5 and sepl, optionally in combination with a second group of protease encoding genes selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pepl2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukm and has reduced expression of the protease encoding genes pepl, tspl, slpl, gapl, and gap2.

In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukm and has reduced expression of the protease encoding genes slpl, slp2, pepl, gapl, pep4, slp7, pep2, pep3, pep5, tspl, and gap2. In other embodiments, the cell, for example a Trichoderma fungal cell, contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, tspl, slpl, gapl, gap2, and pep4. In a further embodiment, the cell contains a recombinant polynucleotide encoding a growth factor, and has reduced expression of a pep-type protease genes are selected from pepl, pep2, pep3, pep4, and pep5.

In certain preferred embodiments, the growth factor is IGF-1 or the interferon is interferon-oc 2b. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, and pep4. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, and slp7. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, slp7, and slp2. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, slp7, slp2, and pep2.

In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, slp7, slp2, pep2, and pep3. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, slp7, slp2, pep2, pep3, and pep5. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, slp7, slp2, pep2, pep3, pep5, and slpl. In certain embodiments, the cell contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced expression of the protease encoding genes pepl, gapl, pep4, slp7, slp2, pep2, pep3, pep5, slpl, and tspl.

In some embodiments, the cell, for example a Trichoderma cell, contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced or no protease activity in at least twelve proteases, each of the genes encoding the twelve proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the twelve proteases are pepl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7. In other embodiments, the cell, for example a Trichoderma cell, contains a recombinant polynucleotide encoding a growth factor, interferon, cytokine, human serum albumin, or interleukin and has either

(i) reduced or no protease activity in at least thirteen proteases, each of the genes encoding the thirteen proteases comprises a mutation that reduces or eliminates the

corresponding protease activity, and the thirteen proteases are either

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, pep9; pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp7;

pepl, tspl, slpl, gapl, gap2, pep4, pep3, pep5, pep2, sepl, slp8, amp2, slp3;

(ii) reduced or no protease activity in at least fourteen proteases, each of the genes encoding the fourteen proteases comprises a mutation that reduces or eliminates the corresponding protease activity, and the fourteen proteases are pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2; or,

(iii) reduced or no protease activity in at least fifteen proteases, each of the genes encoding the fifteen proteases comprises a mutation that reduces or eliminates the

corresponding protease activity, and the fifteen proteases are either pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mpl ; or, pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2 pep9 slp2 mp5.

In other embodiments, the filamentous fungal cell, for example a Trichoderma cell, contains a recombinant polynucleotide encoding growth factor, interferon, cytokine, human serum albumin, or interleukin and has reduced or no protease activity in at least sixteen, at least seventeen, at least eighteen, at least nineteen, or at least twenty or more proteases, with mutations in the above recited 13, 14 or 15 proteases, and further comprising one or more additional mutation, each additional mutation reduces or eliminates the corresponding additional protease activity, and said at least additional protease being selected from the group consisting of

an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5;

Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

In certain embodiments, the mammalian polypeptide is produced at a level that is at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 75-fold, at least 80-fold, at least 90-fold, at least 100-fold, or a greater fold higher than the production level of the polypeptide in a corresponding parental filamentous fungal cell without the reduced protease activity. In other embodiments, the mammalian polypeptide is produced in a full length version at a level higher than the production level of the full-length version of the polypeptide in a corresponding parental filamentous fungal cell.

In embodiments where the filamentous fungal cell contains a recombinant polynucleotide encoding a non-mammalian polypeptide, the non-mammalian polypeptide may be an aminopeptidase, amylase, carbohydrase, carboxypeptidase, catalase, cellulase, chitinase, cutinase, deoxyribonuclease, esterase, alpha-galactosidase, beta-galactosidase, glucoamylase, alpha- glucosidase, beta-glucosidase, invertase, laccase, lipase, mutanase, oxidase, pectinolytic enzyme, peroxidase, phospholipase, phytase, polyphenoloxidase, proteolytic enzyme, ribonuclease, transglutaminase or xylanase. In embodiments where the filamentous fungal cell contains a recombinant polynucleotide encoding a non-mammalian polypeptide, the filamentous fungal cell may have reduced or no detectable expression of at least two, three or four proteases selected from pep9, ampl, amp2 mpl, mp2, mp3, mp4, mp5 and sepl, optionally in combination with three, at least four, at least five, or at least six protease encoding genes selected from pepl, pep2, pep3, pep4, pep5, pep8, pepll, pep!2, tspl, slpl, slp2, slp3, slp7, gapl, and gap2. In certain embodiments, the non-mammalian polypeptide is produced at a level that is at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15-fold, at least 20-fold, at least 25-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 75-fold, at least 80-fold, at least 90-fold, at least 100-fold, or a greater fold higher than the production level of the polypeptide in a corresponding parental filamentous fungal cell. In other embodiments, the non-mammalian polypeptide is produced in a full length version at a level higher than the production level of the full-length version of the polypeptide in a corresponding parental filamentous fungal cell.

Reduced activity of additional proteases In some embodiments, the filamentous fungal cells or Trichoderma fungal cells of the present disclosure also have reduced activity of one or more additional proteases. In certain

embodiments, the expression level of the one or more additional proteases is reduced. In certain preferred embodiments, genes encoding the one or more additional proteases each comprise a mutation that reduces the corresponding protease activity. The one or more additional protease encoding genes may be pep7, tppl, gap2, slp3, slp5, slp6, slp7, slp8, or a gene encoding protease selected from the group consisting of

an aspartic protease pep6, peplO, pepl3, pepl4, or pepl6;

> sip like protease slp57433, slp35726, slp60791, or slpl09276;

gap like protease gap3or gap4;

sedolisin like protease sed2, sed3, or sed5;

Group A protease selected from the group of protease65735, protease77577, protease81087, protease56920, proteasel22083, protease79485, proteasel20998, or pro tease61127;

Group B protease selected from the group of protease21659, protease58387, protease75159, protease56853, or protease64193;

Group C protease selected from the group of protease82452, protease80762, protease21668, protease81115, protease82141, protease23475;

Group D protease selected from the group of proteasel21890, protease22718, protease47127, protease61912, protease80843, protease66608, protease72612, protease40199; or

Group E protease selected from the group of protease22210, proteasel 11694, protease82577.

In certain embodiments, when the filamentous fungal cells is an Aspergillus cell, the total protease activity is reduced to 50% or less of the total protease activity in the corresponding parental Aspergillus cell in which the proteases do not have reduced activity.

In certain embodiments, total protease activity is reduced in the cell of the present disclosure, for example a Trichoderma cell, to 49% or less, 31% or less, 13% or less, 10% or less, 6.3% or less, or 5.5% or less, of the total protease activity in the corresponding parental filamentous fungal cell in which the proteases do not have reduced activity.

Additional recombinant modifications

In certain embodiments, the filamentous fungal cells or Trichoderma fungal cells of the present disclosure also have reduced activity of a dolichyl-P-Man:Man(5)GlcNAc(2)-PP-dolichyl mannosyltransferase. 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. Typically, the dolichyl-P-Man:Man(5)GlcNAc(2)-PP- dolichyl mannosyltransferase enzyme is encoded by an alg3 gene. Thus, in certain embodiments, the filamentous fungal cell has reduced activity of ALG3, which is the activity encoded by the alg3 gene. In some embodiments, the alg3 gene contains a mutation that reduces the corresponding ALG3 activity. In certain embodiments, the alg3 gene is deleted from the filamentous fungal cell.

In other embodiments, the filamentous fungal cells or Trichoderma fungal cells of the present disclosure further contain a polynucleotide encoding an a-l,2-mannosidase. The polynucleotide encoding the a-l,2-mannosidase may be endogenous in the host cell, or it may be heterologous to the host cell. These polynucleotides are especially useful for a filamentous fungal cell expressing high-mannose glycans transferred from the Golgi to the ER without effective exo-a-2- mannosidase cleavage. The a-l,2-mannosidase may be a mannosidase I type enzyme belonging to the glycoside hydrolase family 47 (cazy.org/GH47_all.html). In certain embodiments the a- 1 ,2-mannosidase is an enzyme listed at cazy.org/GH47_characterized.html. In particular, the a- 1 ,2-mannosidase may be an ER-type enzyme that cleaves glycoproteins such as enzymes in the subfamily of ER a-mannosidase I EC 3.2.1.113 enzymes. Examples of such enzymes include human α-2-mannosidase IB (AAC26169), a combination of mammalian ER mannosidases, or a filamentous fungal enzyme such as a-l,2-mannosidase (MDS1) (T. reesei AAF34579; Maras M et al J Biotech. 77, 2000, 255). For ER/Golgi expression 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, see, for example, Callewaert et al. 2001 Use of HDEL-tagged Trichoderma reesei mannosyl oligosaccharide 1 ,2-a-D-mannosidase for N-glycan engineering in Pichia pastoris. FEBS Lett 503: 173-178.

In further embodiments, the filamentous fungal cells or Trichoderma fungal cells of the present disclosure also contain an N-acetylglucosaminyltransferase I catalytic domain and an N- acetylglucosaminyltransferase II catalytic domain. Such catalytic domains are useful for expressing complex N-glycans in non-mammalian cells. N-acetylglucosaminyltransferase I (GlcNAc-TI; GnTI; EC 2.4.1.101) catalyzes the reaction UDP-N-acetyl-D-glucosamine + 3- (alpha-D-mannosyl)-beta-D-mannosyl-R <=> UDP + 3-(2-(N-acetyl-beta-D-glucosaminyl)- alpha-D-mannosyl)-beta-D-mannosyl-R, where R represents the remainder of the N-linked oligosaccharide in the glycan acceptor. An N-acetylglucosaminyltransferase I catalytic domain is any portion of an N-acetylglucosaminyltransferase I enzyme that is capable of catalyzing this reaction. N-acetylglucosaminyltransferase II (GlcNAc-TII; GnTII; EC 2.4.1.143) catalyzes the reaction UDP-N-acetyl-D-glucosamine + 6-(alpha-D-mannosyl)-beta-D-mannosyl-R <=> UDP + 6-(2-(N-acetyl-beta-D-glucosaminyl)-alpha-D-mannosyl)-beta-D -mannosyl-R, where R represents the remainder of the N-linked oligosaccharide in the glycan acceptor. An N- acetylglucosaminyltransferase II catalytic domain is any portion of an N- acetylglucosaminyltransferase II enzyme that is capable of catalyzing this reaction. Examples of suitable N-acetylglucosaminyltransferase I catalytic domains and an N- acetylglucosaminyltransferase II catalytic domains can be found in International Patent Publication WO2012/069593. The N-acetylglucosaminyl transferase I catalytic domain and N- acetylglucosaminyltransferase II catalytic domain can be encoded by a single polynucleotide. In certain embodiments, the single polynucleotide encodes a fusion protein containing the N- acetylglucosaminyltransferase I catalytic domain and the N-acetylglucosaminyltransferase II catalytic domain. Alternatively, the N-acetylglucosaminyltransferase I catalytic domain can be encoded by a first polynucleotide and the N-acetylglucosaminyltransferase II catalytic domain can be encoded by a second polynucleotide.

In embodiments where, the filamentous fungal cell or Trichoderma fungal cell contains an N- acetylglucosaminyltransferase I catalytic domain and an N-acetylglucosaminyltransferase II catalytic domain, the cell can also contain a polynucleotide encoding a mannosidase II. Mannosidase II enzymes are capable of cleaving Man5 structures of GlcNAcMan5 to generate GlcNAcMan3, and if combined with action of a catalytic domain of GnTII, to generate GO; and further, with action of a catalytic domain of a galactosyltransferase, to generate Gl and G2. In certain embodiments mannosidase II-type enzymes belong to glycoside hydrolase family 38 (cazy.org/GH38_all.html). Examples of such enzymes include human enzyme AAC50302, D. melanogaster enzyme (Van den Elsen J.M. et al (2001) EMBO J. 20: 3008-3017), those with the 3D structure according to PDB-reference 1HTY, and others referenced with the catalytic domain in PDB. For ER/Golgi expression, the catalytic domain of the mannosidase is typically fused with an N-terminal targeting peptide, for example using targeting peptides listed in the International Patent Publication No. WO2012/069593 or of SEQ ID NOs 589-594. After transformation with the catalytic domain of a mannosidase II-type mannosidase, a strain effectively producing GlcNAc2Man3, GlcNAclMan3 or GO is selected.

In certain embodiments that may be combined with the preceding embodiments, the filamentous fungal cell further contains a polynucleotide encoding a UDP-GlcNAc transporter.

In certain embodiments that may be combined with the preceding embodiments, the filamentous fungal cell further contains a polynucleotide encoding a -l,4-galactosyltransferase. Generally, β- 1,4-galactosyltransferases belong to the CAZy glycosyltransferase family 7 (cazy.org/GT7_all.html). Examples of useful 4GalT enzymes include 4GalTl, 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. Glycobiology 2001, 11:813-20), and Mus musculus enzyme AAA37297 (Shaper, N.L. et al. 1998 J. Biol. Chem. 263 (21), 10420-10428). In certain embodiments of the invention where 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. In certain embodiments of the invention where the filamentous fungal cell contains a polynucleotide encoding a galactosyltransferase, 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. In certain embodiments of the invention where the filamentous fungal cell contains a polynucleotide encoding a galactosyltransferase and, optionally, 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.

In certain embodiments that may be combined with the preceding embodiments, the level of activity of alpha- 1,6-mannosyltransferase in the host cell is reduced compared to the level of activity in a wild-type host cell. In certain embodiments, the filamentous fungal has a reduced level of expression of an ochl gene compared to the level of expression in a wild-type filamentous fungal cell.

Another aspect includes methods of producing a Man3GlcNAc2 N-glycan [i.e. Mana3(Manoc6)ManP4GlcNAcP4GlcNAc] in a filamentous fungal cell including the steps of providing a filamentous fungal cell with a recombinant polynucleotide encoding a heterologous polypeptide and a reduced level of activity of an alg3 mannosyltransferase compared to the level of activity in a wild-type filamentous fungal cell and culturing the filamentous fungal cell to produce a Man3GlcNAc2 glycan, where the Man3GlcNAc2 glycan constitute at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the neutral N-glycans secreted by the filamentous fungal cell. In certain embodiment, Man3GlcNAc2 N-glycan represents at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the total N-glycans of the heterologous polypeptide. Another aspect includes methods of producing a complex N-glycan (i.e an N-glycan comprising a terminal GlcNAc2Man3 structure), for example GlcNAc2Man3GlcNAc2 {i.e. GO, i.e. GlcNAc 2Man 3(GlcNAcP2Man 6)ManP4GlcNAcP4GlcNAc} glycan in a filamentous fungal cell including the steps of providing a filamentous fungal cell with a recombinant polynucleotide encoding a heterologous polypeptide, a reduced level of activity of an alg3 mannosyltransferase compared to the level of activity in a wild-type filamentous fungal cell and comprising further a polynucleotide encoding an N-acetylglucosaminyltransferase I catalytic domain and a polynucleotide encoding an N-acetylglucosaminyltransferase II catalytic domain and culturing the filamentous fungal cell to produce the complex N-glycan, for example GlcNAc2Man3GlcNAc2 glycan, where the GlcNAc2Man3GlcNAc2 glycan constitutes at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the neutral N-glycans secreted by the filamentous fungal cell. In certain embodiments, the complex N-glycan, for example GlcNAc2Man3GlcNAc2 glycan, represents at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %)of the total N-glycans of the polypeptide. In certain embodiments, said complex N- gl yeans are GlcNAcMan3 and/or GlcNAc2Man3.

Another aspect includes methods of producing a Gl or G2 N-glycan or mixture thereof, for example GalGlcNAc2Man3GlcNAc2 {i.e. Gl, i.e.

Gai 4GlcNAc 2Mana3(GlcNAc 2Mana6)Man 4GlcNAc 4GlcNAc } or GlcNAc 2Mana3(Gai 4GlcNAc 2Mana6)Man 4GlcNAc 4GlcNAc } and/or Gal2GlcNAc2Man3GlcNAc2 {i.e. G2, i.e. Gai 4GlcNAc 2Mana3(Gai 4 GlcNAc 2Mana6)Man 4GlcNAc 4GlcNAc} glycan in a filamentous fungal cell including the steps of providing a filamentous fungal cell with a recombinant polynucleotide encoding a heterologous polypeptide and a reduced level of activity of an alg3 mannosyltransferase compared to the level of activity in a wild-type filamentous fungal cell and comprising further a polynucleotide encoding an N-acetylglucosaminyltransferase I catalytic domain, a polynucleotide encoding an N-acetylglucosaminyltransferase II catalytic domain, and a polynucleotide encoding a GalT catalytic domain and culturing the filamentous fungal cell to produce the Gl or G2 N- glycan or mixture thereof, where Gl glycan constitutes at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the neutral N-glycans secreted by the filamentous fungal cell, or where the G2 glycan constitutes at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the neutral N-glycans secreted by the filamentous fungal cell. In certain embodiment, Gl glycan constitutes at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the total N-glycans of the polypeptide. In certain embodiment, G2 glycan constitutes at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the total N- glycans of the polypeptide.

In certain embodiments, the method of producing a complex N-glycan will generate a mixture of different glycans. The complex N-glycan or Man3GlcNAc2 may constitute at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%), or at least 90% or more of such a glycan mixture. In certain embodiments, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%), or at least 90% or more of the N-glycans of the polypeptide consists of such a glycan mixture. In certain embodiments, the method of producing a complex and Gl and/or G2 N- glycan will generate a mixture of different glycans. The complex N-glycan, Man3GlcNAc2, Gl and/or G2 may constitute at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%), or at least 90% or more of such a glycan mixture. In certain embodiments, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%), or at least 90% or more of of the N- glycans of the polypeptide consists of such a glycan mixture.

In certain embodiments, methods of producing a hybrid N-glycan are desirable. As used herein, the term "hybrid" means a glycan containing both unsubstituted terminal mannose residues (as are present in high-mannose glycans) and substituted mannose residues with an N- acetylglucosaminelinkage,forexampleGlcNAc 2Mana3[Mana3(Mana6)Mana6]Man 4GlcNAc 4GlcNAc. In such embodiments, a Man5 {i.e Mana3[Mana3(Mana6) Mana6]Man 4GlcNAc 4GlcNAc} expressing filamentous fungal cell such as T. reesei strain is transformed with a recombinant polynucleotide encoding a heterologous polypeptide and a polynucleotide encoding an N-acetylglucosaminyltransferase I catalytic domain and the filamentous fungal cell is cultured to produce the hybrid N-glycan where the hybrid N-glycan constitutes at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least

60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the neutral N-glycans secreted by the filamentous fungal cell. In certain embodiment, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100% (mol %) of the N-glycans of the polypeptide consists of a hybrid N-glycan. The Man3GlcNAc2, complex, hybrid, Gl, and G2 N-glycan may be attached to a molecule selected from an amino acid, a peptide, and a polypeptide. In certain embodiments, the Man3GlcNAc2, complex, hybrid, Gl, and G2 N-glycan is attached to a heterologous polypeptide. In certain embodiments, the heterologous polypeptide is a glycosylated protein. In certain embodiment, the glycosylated polypeptide is a mammalian polypeptide. In certain embodiments, mammalian polypeptide is an antibody or its antigen-binding fragment.

In certain embodiments, glycosyltransferases, for example, GnTI, GnTII, or GalT or glycosylhydrolases, for example, a-l,2-mannosidase or mannosidase II, 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 filamentous fungal 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. In certain 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, MNSl, KRE2, VANl, and OCHl enzymes. Suitable targeting peptides are described in the International Patent Publication No. WO2012/069593. In one embodiment, the targeting peptide of GnTI or GnTII is human GnTII enzyme. In other embodiments, targeting peptide is derived from Trichoderma Kre2, Kre2-\ike,, Ochl, Anpl, and Vanl. In one embodiment, the targeting peptide is selected from the group of SEQ ID NOs: 589-594. Uses of the Filamentous Fungal Cells of the Invention

The invention herein further relates to methods of using any of the filamentous fungal cells of the present disclosure, such as Trichoderma fungal cells, that have reduced or no protease activity of at least three proteases and that contain a recombinant polynucleotide encoding a heterologous polypeptide, such as a mammalian polypeptide, that is produced at increased levels, for improving heterologous polypeptide stability and for making a heterologous polypeptide. Methods of measuring protein stability and for making a heterologous polypeptide are well known, and include, without limitation, all the methods and techniques described in the present disclosure. Accordingly, certain embodiments of the present disclosure relate to methods of improving heterologous polypeptide stability, by: a) providing a filamentous fungal cell of the present disclosure having reduced or no activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a heterologous polypeptide; and b) culturing the cell such that the heterologous polypeptide is expressed, where the heterologous polypeptide has increased stability compared to a host cell not containing the mutations of the genes encoding the proteases. Other embodiments of the present disclosure relate to methods of improving mammalian polypeptide stability, by: a) providing a Trichoderma fungal cell of the present disclosure having reduced or no activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide; and b) culturing the cell such that the mammalian polypeptide is expressed, where the mammalian polypeptide has increased stability compared to a host cell not containing the mutations of the genes encoding the proteases. The filamentous fungal cell or Trichoderma fungal cell may be any cell described in the section entitled "Filamentous Fungal Cells of the Invention". Methods of measuring polypeptide stability and for culturing filamentous fungal and Trichoderma fungal cells are well known in the art, and include, without limitation, all the methods and techniques described in the present disclosure.

In certain embodiments, the stability of the heterologous polypeptide or mammalian polypeptide is increased by at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 6-fold, at least 7-fold, at least 8-fold, at least 9-fold, at least 10-fold, at least 15 -fold, at least 20-fold, at least 25- fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 75- fold, at least 80-fold, at least 90-fold, at least 100-fold, or a greater fold higher compared to a heterologous polypeptide or mammalian polypeptide expressed in a corresponding parental filamentous fungal or Trichoderma fungal cell.

Other embodiments of the present disclosure relate to methods of making a heterologous polypeptide, by: a) providing a filamentous fungal cell of the present disclosure having reduced or no activity of at least three proteases, where the cell further contains a recombinant polynucleotide encoding a heterologous polypeptide; b) culturing the host cell such that the heterologous polypeptide is expressed; and c) purifying the heterologous polypeptide. Further embodiments of the present disclosure relate to methods of making a mammalian polypeptide, by: a) providing a Trichoderma fungal cell of the present disclosure having reduced or no activity of at least three protease, where the cell further contains a recombinant polynucleotide encoding a mammalian polypeptide; b) culturing the host cell such that the mammalian polypeptide is expressed; and c) purifying the mammalian polypeptide. The filamentous fungal cell or Trichoderma fungal cell may be any cell described in the section entitled "Filamentous Fungal Cells of the Invention". Methods of culturing filamentous fungal and Trichoderma fungal cells and purifying polypeptides are well known in the art, and include, without limitation, all the methods and techniques described in the present disclosure. In certain embodiments, the filamentous fungal cell or Trichoderma fungal cell is cultured at a pH range selected from pH 3.5 to 7; pH 3.5 to 6.5; pH 4 to 6; pH 4.3 to 5.7; pH 4.4 to 5.6; and pH 4.5 to 5.5. In certain embodiments, to produce an antibody 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.

In some embodiments, the heterologous polypeptide is a mammalian polypeptide, embodiments, the heterologous polypeptide is a non-mammalian polypeptide. In certain embodiments, the mammalian polypeptide is selected from an immunoglobulin, immunoglobulin heavy chain, an immunoglobulin light chain, a monoclonal antibody, a hybrid antibody, an F(ab')2 antibody fragment, an F(ab) antibody fragment, an Fv molecule, a single- chain Fv antibody, a dimeric antibody fragment, a trimeric antibody fragment, a functional antibody fragment, a single domain antibody, multimeric single domain antibodies, an immunoadhesin, insulin- like growth factor 1, a growth hormone, insulin, and erythropoietin. In other embodiments, the mammalian protein is an immunoglobulin or insulin-like growth factor 1. In yet other embodiments, the mammalian protein is an antibody. In further embodiments, the yield of the mammalian polypeptide is at least 0.5, at least 1, at least 2, at least 3, at least 4, or at least 5 grams per liter. In certain embodiments, the mammalian polypeptide is an antibody, optionally, IgGl, IgG2, IgG3, or IgG4. In further embodiments, 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. In still other embodiments, the mammalian polypeptide is a growth factor or a cytokine. In further embodiments, the yield of the growth factor or cytokine is at least 0.1, at least 0.2, at least 0.3, at least 0.4, at least 0.5, at least 1, at least 1.5, at least 2, at least 3, at least 4, or at least 5 grams per liter. In further embodiments, the mammalian polypeptide 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. In other embodiments, the mammalian polypeptide 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

In certain embodiments where the mammalian polypeptide is purified from cell culture, the culture containing the mammalian polypeptide 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. In certain preferred embodiments, the mammalian polypeptide is an antibody, and the polypeptide fragments are heavy chain fragments and/or light chain fragments. In other embodiments, where the mammalian polypeptide is an antibody and the antibody purified from cell culture, 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. Methods of determining the mass percentage of polypeptide fragments are well known in the art and include, measuring signal intensity from an SDS-gel.

In further embodiments, the non-mammalian polypeptide is selected from an aminopeptidase, amylase, carbohydrase, carboxypeptidase, catalase, cellulase. chitinase, cutinase, deoxyribonuclease, esterase, alpha-galactosidase, beta-galactosidase, glucoamylase, alpha- glucosidase, beta-glucosidase, invertase, laccase, lipase, mutanase, oxidase, pectinolytic enzyme, peroxidase, phospholipase, phytase, polyphenoloxidase, proteolytic enzyme, ribonuclease, transglutaminase, and xylanase. In certain embodiments of any of the disclosed methods, 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. In certain embodiments, the protease inhibitors are peptides that are co-expressed with the mammalian polypeptide. In other embodiments, 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.

In certain embodiments of any of the disclosed methods, the filamentous fungal cell or Trichoderma fungal cell also contains a carrier protein. As used herein, 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 (Cel6A, or CBHII) (see, e.g., Paloheimo et al Appl. Environ. Microbiol. 2003 December; 69(12): 7073-7082) or T. reesei cellobiohydrolase I (CBHI). In some embodiments, the carrier protein is CBH1. In other embodiments, the carrier protein is a truncated T. reesei CBHI protein that includes the CBHI core region and part of the CBHI linker region. In some embodiments, a carrier such as a cellobiohydrolase or its fragment is fused to an antibody light chain and/or an antibody heavy chain. In some embodiments, a carrier such as a cellobiohydrolase or its fragment is fused to insulin-like growth factor 1, growth hormone, insulin, interferon alpha 2b, fibroblast growth factor 21, or human serum albumin. In some embodiments, a carrier-antibody fusion polypeptide comprises a Kex2 cleavage site. In certain embodiments, Kex2, or other carrier cleaving enzyme, is endogenous to a filamentous fungal cell. In certain embodiments, 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. In one embodiment, the polynucleotide encoding the heterologous glycoprotein (e.g. the antibody) further comprises a polynucleotide encoding CBH1 catalytic domain and linker as a carrier protein, and/or cbhl promoter. In certain embodiments, the filamentous fungal cell of the invention overexpress KEX2 protease. In an embodiment the heterologous glycoprotein (e.g. the antibody) 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. 258: 107-112; Jalving et al. (2000) Appl. Environ. Microbiol. 66:363-368; Ward et al. (2004) Appl. Environ. Microbiol. 70:2567-2576; Ahn et al. (2004) Appl. Microbiol. Biotechnol. 64:833-839; Paloheimo et al. (2007) Appl Environ Microbiol. 73:3215-3224; Paloheimo et al. (2003) Appl Environ Microbiol. 69:7073- 7082; and Margolles-Clark et al. (1996) Eur J Biochem. 237:553-560. It is to be understood that, while the invention has been described in conjunction with the certain specific embodiments thereof, the foregoing description is intended to illustrate and not limit the scope of the invention. Other aspects, advantages, and modifications within the scope of the invention will be apparent to those skilled in the art to which the invention pertains.

The invention having been described, the following examples are offered to illustrate the subject invention by way of illustration, not by way of limitation. EXAMPLES

The identification of certain proteases in Trichoderma reesei and corresponding T. reesei cells with deficient protease activity have been described in Examples 1-23 of WO 2013/102674 which contents is incorporated herein by reference. EXAMPLE 1 - Generation of deletion plasmids

The deletion plasmid below were constructed essentially as described for pepl deletion plasmid pTTv41 in Example 1 of WO2013/102674, except that the marker used for selection was pyr4- hgh or pyr4 from pTTv213 or pTTvl94. Deletion plasmid for ampl

958 bp of 5' flanking region and 994 bp of 3' flanking region were selected as the basis of the ampl deletion plasmid pTTv240. A 367 bp stretch from the end of ampl 5' flank was used as the direct repeat fragment. These fragments were amplified by PCR using the primers listed in Table 1-1. Template used in the PCR of the flanking regions was from the T. reesei wild type strain QM6a. The products were separated with agarose gel electrophoresis and the correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods. The pyr4-hph selection marker used in pTTv240 was obtained from pTTv213 (Apep2-pyr-hph see Example 14 of WO2013/102674) with Noil digestion. To enable removal of the pyr4-hgh marker cassette, Noil restriction sites were introduced on both sides of the cassette. Ascl site was introduced between the ampl 5 'direct repeat and 3' flank. Vector backbone was EcoRI/XhoI digested pRS426 and the plasmid pTTv240 was constructed with the 5' flank, 3' flank, the 5' direct repeat, pyr4-hph marker, and vector backbone using the yeast homologous recombination method as described in Example 1 of WO2013/102674). The deletion plasmid for ampl (pTTv240, Table 1-1) results in a deletion in the ampl locus and covers the complete coding sequence of AMP1 (tre81070).

Table 1-1 : Primers for generating ampl deletion plasmid. Deletion plasmid pTTv240 (Aampl-pyr4-hph), vector backbone pRS426

Primer Sequence

T832_ampl_5flkfw_vector GTAACGCCAGGGTTTTCCCAGTCACGACGGTTTAAACC

ATGGAAGATGCGAGCTACA (SEQ ID NO: 806)

T833_amp l_5flkrev_pyr4Pr GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGC om TAGGCGGCCGCGGAGAGGAGATGGGTGTTGA (SEQ ID

NO: 807)

T836_amp l_3flkfw_5DR_e CCCCCCTTTCTCTCTCTCTTTCAACACCCATCTCCTCTC nd CGGCGCGCCGCGAGGTGCGTTTCTGTAGC (SEQ ID NO:

808)

T837_amp l_3flkrev_vector GCGGATAACAATTTCACACAGGAAACAGCGTTTAAAC

CGGCAAATACTACGACGACA (SEQ ID NO: 809)

T834_ampl_5 DR fwd GTACACTTGTTTAGAGGTAATCCTTCTTTCTAGAAGGA

GAGCGGCCGCGTCGAGTGCATCAATGACGA (SEQ ID NO: 810)

T835_ampl_5 DR rev CAAACAGCATGCTCGTAAATGCTACAGAAACGCACCT

CGCGGCGCGCCGGAGAGGAGATGGGTGTTGA (SEQ ID NO: 811)

Deletion plasmid for amp2

918 bp of 5' flanking region and 978 bp of 3' flanking region were selected as the basis of the amp2 deletion plasmid pTTv271. These fragments were amplified by PCR using the primers listed in Table 1-2. Template used in the PCR of the flanking regions was from the T. reesei wild type strain QM6a. The products were separated with agarose gel electrophoresis and the correct fragments were isolated from the gel with a gel extraction kit (Qiagen) using standard laboratory methods. The pyr4-hph cassette was obtained from pTTvl94 (Apep4-pyr-hph) with Noil digestion. To enable removal of the marker cassette, Noil restriction sites were introduced on both sides of the cassette. Vector backbone was EcoRI/XhoI digested pRS426 and the plasmid pTTv271 was constructed with the 5' flank, 3' flank, pyr4-hph marker, and vector backbone using the yeast homologous recombination method.

Another deletion plasmid for amp2 (pTTv327) was generated as follows. A 311 bp stretch from the end of amp2 5' flank was used as the direct repeat fragment. The fragments were amplified by PCR using the primers listed in Table 1-3. The products were separated with agarose gel electrophoresis and the correct fragments were isolated from the gel. The pyr4-hph cassette was obtained from pTTv210 (Asepl -pyr4-hph) with Noil digestion. To enable removal of the marker cassette, Noil restriction sites were introduced on both sides of the cassette. Ascl site was introduced between the amp2 5 'direct repeat and 3' flank. Vector backbone was EcoRI/XhoI digested pRS426 and the plasmid was constructed as described in Example 1 of WO2013/102674.

These deletion plasmids for amp2 (pTTv271 and pTTv327) result in 2143 bp deletion in the amp2 locus and cover the complete coding sequence of AMP2 (trel 08592).

Table 1-2: Primers for generating amp2 deletion plasmid.

Deletion plasmid pTTv271 (Aamp2-pyr4-hph), vector backbone pRS426

Primer Sequence

Tl 079_amp2_5flkfw_vecto GTAACGCCAGGGTTTTCCCAGTCACGACGGTTTAAACCC r ATTCTCGTCGTTGTTTCC (SEQ ID NO: 812)

Tl 080_amp2_5flkre GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGCTAGGC v_pyr4Prom GGCCGCTGGAGGAGTAGCTGCACTGA (SEQ ID NO: 813)

T1081_amp2_3flkf CAACCAGCCGCAGCCTCAGCCTCTCTCAGCCTCATCAGCCGCG w_pyr41oop GCCGCACAGCCAGTGGAAACCAAAC (SEQ ID NO: 814)

Tl 082_amp2_3flkre GCGGATAACAATTTCACACAGGAAACAGCGTTTAAACTAGAGC v_vector TTGGAGGGAACAGG (SEQ ID NO: 815)

Table 1-3: Primers for generating amp2 deletion plasmid.

Deletion plasmid pTTv327 (Aamp2-pyr4-hph), vector backbone pRS426

Primer Sequence

Tl 079_amp2_5flkfw_vec GTAACGCCAGGGTTTTCCCAGTCACGACGGTTTAAAC tor CCATTCTCGTCGTTGTTTCC (SEQ ID NO: 812)

Tl 080_amp2_5flk GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGAT

rev_pyr4Prom GCTAGGCGGCCGCTGGAGGAGTAGCTGCACTGA (SEQ ID NO:

813)

Tl 194_amp2_5dr GTACACTTGTTTAGAGGTAATCCTTCTTTCTAGAAGGA

J_ GAGCGGCCGCGAGTCGGTCCTACTGCTTGA (SEQ ID NO: 816)

Tl 195_amp2_5dr TTTTACTTGTTTTGATAAGGGTTTGGTTTCCACTGGCTG

_j TGGCGCGCCTGGAGGAGTAGCTGCACTGA (SEQ ID NO: 817)

T1196_amp2_3f_f ACAGCCAGTGGAAACCAAAC (SEQ ID NO: 818)

2

Tl 082_amp2_3flk GCGGATAACAATTTCACACAGGAAACAGCGTTTAAAC vector TAGAGCTTGGAGGGAACAGG (SEQ ID NO: 815)

Deletion plasmid for sepl

First deletion plasmid for sepl, pTTv210, contained 1099 bp 5' flanking region and 806 bp 3' flanking region, a 300 bp strech from the end of sepl 5' flank as the direct repeat fragment, pyr4- hph double selection marker and an expression construct for T. reesei native kex2 (trel23561 ; promoter cDNAl - kex2 - terminator cbh2). All other fragments except pyr4 were amplified by PCR using the primers listed in Table 1-4. Template used in the PCR of the flanking regions, direct repeat, kex2 and terminator was from the T. reesei wild type strain QM6a. Templates for cDNAl promoter and hph marker were plasmids. The pyr4 marker was obtained from pTTvl81 with Noil digestion (pTTvl81 is described in Example 4 of WO2013/102674). To enable removal of the pyr4-hph marker cassette, Noil restriction sites were introduced on both sides of the cassette. Ascl site was introduced between the sepl 5 'direct repeat and 3' flank. The products were separated with agarose gel electrophoresis and the correct fragments were isolated from the gel with a gel extraction kit (Qiagen). Vector backbone was EcoRI/XhoI digested pRS426. Plasmid pTTv210 was constructed with all eight fragments above and vector backbone using the yeast homologous recombination method as described above.

The second deletion plasmid for sepl, pTTv247, was constructed by removing the KEX2 overexpression cassette from pTTv210 with Ascl digestion. The fragments were separated with agarose gel electrophoresis and the vector part isolated from the gel with a gel extraction kit (Qiagen). pTTv247 was cloned by self-ligation using T4 DNA ligase and standard laboratory methods.

Third deletion plasmid pTTv255 for sepl was constructed as follows. 1099 bp of 5' flanking region and 806 bp of 3' flanking region were selected as the basis and a 300 bp stretch from the end of sepl 5' flank was used as the direct repeat fragment. These fragments were amplified by PCR using the primers listed in Table 1-4. The pyr4 cassette was obtained from pTTvl81 (Apep4-pyr4) with Noil digestion. To enable removal of the marker cassette, Noil restriction sites were introduced on both sides of the cassette. Ascl site was introduced between the sepl 5 'direct repeat and 3' flank. Vector backbone was EcoRI/XhoI digested pRS426 and the plasmid was constructed using the yeast homologous recombination as above.

The deletion plasmids pTTv210, pTTv247 and pTTv255 result in 2519 bp deletion in the sepl locus and cover the complete coding sequence of SEP1 (trel24051).

Table 1-4: Primers for generating sepl deletion plasmids.

Deletion plasmid pTTv210 (Asepl-pyr4-hph, kex2), vector backbone pRS426

Primer Primer sequence

T489_ _serendo_ _5f_for GGTAACGCCAGGGTTTTCCCAGTCACGACGGTTTAAAC

ATGGGCTGAAACCGGCGCAA (SEQ ID NO: 819)

T490_ _serendo_ _5f_rev GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGC

TAGGCGGCCGCGACAGCGCCTCGCCAAGTGT (SEQ ID NO: 820)

T493_ _serendo_ _DR_for GTACACTTGTTTAGAGGTAATCCTTCTTTCTAGAAGGA

GAGCGGCCGCAGCAGCCTGCCCAGAGAATC (SEQ ID NO: 821)

T494 _serendo_ _DR_rev GTCATTAAGTCCATCATTCCACGTCCTTCAGACCGAATT

CGGCGCGCCGACAGCGCCTCGCCAAGTGT (SEQ ID NO: 822)

T498_ _serendo_ _3f_for ATGATGCCTTTGCAGAAATGGCTTGCTCGCTGACTGATA

CGGCGCGCCTGGCGCTTCCGTTCCCTTCC (SEQ ID NO: 823)

T499 _serendo_ _3f_rev AGCGGATAACAATTTCACACAGGAAACAGCGTTTAAAC

TGTTGAGACGGGCGAGTGCT (SEQ ID NO: 824)

T49 l_hph_recpyr4_for3 TGATTGTACCCCAGCTGCGATTGATGTGTATCTTTGCAT

GATTTAAATTCTCCTTAGCTCTGTACAGT (SEQ ID NO: 607)

T492_hph_rev2 GCGGCCGCTCTCCTTCTAGAAAGAAGGA (SEQ ID NO:

608)

T495_ _cDNAl_ _for GAATTCGGTCTGAAGGACGT (SEQ ID NO: 609)

T138_ _cDNAl_ _Rev GTTGAGAGAAGTTGTTGGATTG (SEQ ID NO: 392)

T139_ _123561For_cDNAl

CATGAAGATTTCCTCGATCCTTG (SEQ ID NO: 393)

T516_123561Rev TCAGCGCCGTAACCTCTGC (SEQ ID NO: 394) T496_tcbh2_for TGATGGTGATGAGGCGGAAAAGCAGAGGTTACGGCG

CTGAGGCTTTCGTGACCGGGCTTC (SEQ ID NO: 610)

T497_tcbh2_rev GTATCAGTCAGCGAGCAAGC (SEQ ID NO: 611)

Deletion plasmid pTTv247 (Asepl-pyr4-hph), vector backbone pTTv210

Primer Primer sequence

No new primers; pTTv210 digested with Ascl (to remove kex2 cassette)

and -12 kb fragment self-li^ >ated

Deletion plasmid pTTv255 (Asepl-pyr4), vector backbone pRS426

Primer Primer sequence

T489_serendo_5f_for GGTAACGCCAGGGTTTTCCCAGTCACGACGGTTTAAAC

ATGGGCTGAAACCGGCGCAA (SEQ ID NO: 819)

T490_serendo_5f_rev GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGC

TAGGCGGCCGCGACAGCGCCTCGCCAAGTGT (SEQ ID

NO: 820)

Tl 000_serendo_5dr_for2 TGATTGTACCCCAGCTGCGATTGATGTGTATCTTTGCA

TGGCGGCCGCAGCAGCCTGCCCAGAGAATC (SEQ ID

NO: 825)

T 1001 _serendo_5 dr_rev2 GACAATCAGAGGCCTCAATTGGAAGGGAACGGAAGC

GCCAGGCGCGCCGACAGCGCCTCGCCAAGTGT (SEQ ID

NO: 826)

T502_serendo_3f_probef TGGCGCTTCCGTTCCCTTCC (SEQ ID NO: 827)

T499_serendo_3f_rev AGCGGATAACAATTTCACACAGGAAACAGCGTT

TAAACTGTTGAGACGGGCGAGTGCT (SEQ ID NO: 824)

Deletion plasmids for pep9

918 bp of 5' flanking region and 978 bp of 3' flanking region were selected as the basis of the pep9 deletion plasmid pTTv267. These fragments were amplified by PCR using the primers listed in Table 1-5. Template used in the PCR of the flanking regions was from the T. reesei wild type strain QM6a. The products were separated with agarose gel electrophoresis and the correct fragments were isolated from the gel with a gel extraction kit (Qiagen). The pyr4-hph cassette was obtained from pTTvl94 (Apep4-pyr-hph) with Noil digestion. To enable removal of the marker cassette, Noil restriction sites were introduced on both sides of the cassette. Vector backbone was EcoRVXhoI digested pRS426 and the plasmid was constructed with the 5' flank, 3' flank, pyr4-hph marker, and vector backbone using the yeast homologous recombination method. This deletion plasmid for pep9 (pTTv267, Table 1-5) results in a 1448 bp deletion in the pep9 locus and covers majority of the coding sequence PEP9 (tre79807).

Table 1-5: Primers for generating pep9 deletion plasmid.

Deletion plasmid pTTv267 (Apep9-pyr4-hph), vector backbone pRS426

Primer Sequence

Tl 027_pep9_5flkfw_vector GTAACGCCAGGGTTTTCCCAGTCACGACGGTTTAAACA

ACCACGACGAAAATCAAGG (SEQ ID NO: 828)

Tl 028_pep9_5flkrev_pyr4Pr GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGC om TAGGCGGCCGCAATGGACCCAGATGTCAAGG (SEQ ID

NO: 829)

Tl 029_pep9_3flkfw_pyr41oo CAACCAGCCGCAGCCTCAGCCTCTCTCAGCCTCATCAG p CCGCGGCCGCGATCTAGGATTCGCCAAACG (SEQ ID

NO: 830)

Tl 030_pep9_3flkrev_vector GCGGATAACAATTTCACACAGGAAACAGCGTTTAAAC

ACGACATGAACAAACGGACA (SEQ ID NO: 831)

The second deletion plasmid for pep9, pTTv421, was constructed using the plasmid pTTv267 above as the backbone. The pyr4-hph double marker with pyr4 5DR was removed from pTTv267 with Noil digestion. New marker, pyr4-hph, was obtained from a plasmid derived from pTTv264 with egl2 deletion with Noil digestion. To enable removal of the marker cassette, Noil restriction sites were introduced on both sides of the cassette. Fsel site was introduced between the pep9 3'direct repeat and 5' flank. A 321 bp stretch from the beginning of pep9 3' flank was used as the direct repeat fragment. Part of the cbh2 terminator was used as a bridge in cloning. These two fragments were amplified by PCR using the primers listed in Table 1-6.

The third deletion plasmid for pep9, pTTv426, was constructed using the plasmid pTTv421 above as the backbone. The pyr4-hph double marker was removed from pTTv421 with Noil digestion. The pyr4 marker gene was obtained from pTTvl81 (Apep4-pyr4) with Noil digestion. Cloning of the plasmid pTTv426 was done with quick ligation using T4 DNA ligase at room temperature. Part of the ligation mixture was transformed into E. coli with electroporation. A few clones were cultivated, plasmid DNA was isolated and digested to screen for correct ligation using standard laboratory methods. Correct ligation and orientation of the marker was further verified by sequencing. These deletion plasmids for pep9 (pTTv421 and pTTv426, Table 1-6) result in a 1448 bp deletion in the pep9 locus and cover majority of the coding sequence of PEP9 (tre79807).

Table 1-6: Primers for generating pep9 deletion plasmids pTTv421 and pTTv426.

Deletion plasmid pTTv421 (Apep9-pyr4-hph), vector backbone pTTv267

Primer Primer sequence

Tl 520_pep9_3dr_fo TAATCTCTAATCACCTAATACCTTGACATCTGGGTCCATT _r GGCCGGCCGATCTAGGATTCGCCAAACG (SEQ ID NO: 832)

Tl 521_pep9_3dr_re GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGC

_v TAGGCGGCCGCAGTATCGCAGGAGCGAGCAT (SEQ ID NO: 833)

T1404_cbh2term_fo CCGTCTAGCGCTGTTGATTG (SEQ ID NO: 834)

Tl 522_tcbh2_recpe GCTGGATGAACGTTTGGCGAATCCTAGATCGCGGCCGCG p9_rev TGCTGCGGAATCATTATCA (SEQ ID NO: 835)

Deletion plasmid pTTv426 (Apep9-pyr4), vector backbone pTTv421

Primer Primer sequence

No new primers; pTTv421 digested with Noil and ligated with pyr4 from pTTv!81.

Deletion plasmid for slp7

The deletion plasmid pTTv329 for slp7 (trel23865) was constructed essentially as described for pTTv269 except the additional primers used in Table 1-7, the pyr4-hph cassette was obtained from pTTv210 (Asepl -pyr4-hph), and Ascl site was introduced between the slp7 5'direct repeat and 3' flank.

Table 1-7: Additional primers for generating slp7 deletion plasmid pTTv329.

Deletion plasmid pTTv329 (Aslp7-pyr4-hph), vector backbone pRS426

Primer Primer sequence

T1200_slp7_5dr_f GTACACTTGTTTAGAGGTAATCCTTCTTTCTAGAAGGAGAGCG

G CCGCGGGGAAACAATGACATACGC (SEQ ID NO: 836)

T1201_slp7_5dr_r CTCTTGGAGAGTCTTCTCCAAAACCCAAGCTTATCACCCAGGC

GCGCCTTTGCAGCAAGATGTCGTTC (SEQ ID NO: 837)

T1202_slp7_3f_f2 TGGGTGATAAGCTTGGGTTT (SEQ ID NO: 838)

Deletion plasmid for slp3

The deletion plasmid pTTvl l6 for slp3 (trel 23234) is described in Example 3 of WO2013/102674. This plasmid results in 1597 bp deletion in the slp3 locus and covers the complete coding sequence of SLP3.

Second deletion plasmid for slp3, pTTv420, was cloned using pTTvl l6 as backbone. Marker from pTTvl l6 was removed by Noil digestion. New marker, pyr4-hph, was obtained from a plasmid derived from pTTv264with egl2 deletion with Noil digestion. To enable removal of the marker cassette, Noil restriction sites were introduced on both sides of the cassette. Fsel site was introduced between the slp3 3 'direct repeat and 5' flank. A 300 bp stretch from the beginning of slp3 3' flank was used as the direct repeat fragment. Part of the cbh2 terminator was used as a bridge in cloning. These two fragments were amplified by PCR using the primers listed in Table 1-8. The products were separated with agarose gel electrophoresis and the correct fragments were isolated from the gel with a gel extraction kit (Qiagen). The plasmid was constructed using the yeast homologous recombination method as described above.

The third deletion plasmid for slp3, pTTv425, was constructed using the plasmid pTTv420 above as the backbone. The pyr4-hph double marker was removed from pTTv420 with Noil digestion. The pyr4 marker gene was obtained from pTTvl81 (Apep4-pyr4) with Noil digestion. Cloning of the plasmid pTTv425 was done with quick ligation using T4 DNA ligase at room temperature. Part of the ligation mixture was transformed into E. coli with electroporation. A few clones were cultivated, plasmid DNA was isolated and digested to screen for correct ligation using standard laboratory methods. Correct ligation and orientation of the marker was further verified by sequencing. These three deletion plasmids for slp3 (pTTvl l6, pTTv420 and pTTv425; Table 1-8) result in 1597 bp deletion in the slp3 locus and cover the complete coding sequence of SLP3. Table 1-8: Primers for generating slp3 deletion plasmids pTTv420 and pTTv425.

Deletion plasmid pTTv420 (Aslp3-pyr4-hph), vector backbone pTTyll6

Primer Primer sequence

T1523_slp3_3dr_for TCTTCTTGACACAATGTTCCTGTTCTCCTCATCCTTGGATG

GCCGGCCACCTAATGGTTTCTTCGTTT (SEQ ID NO: 839)

T1524_slp3_3dr_rev GCGCTGGCAACGAGAGCAGAGCAGCAGTAGTCGATGC

TAGGCGGCCGCCTGGAGAGGATATGGTTCTG (SEQ ID

NO: 840)

T1404_cbh2term_for CCGTCTAGCGCTGTTGATTG (SEQ ID NO: 834)

T 1525_tcbh2_recslp3_rev CACCAAAGAAAAACGAAGAAACCATTAGGTGCGGCCGC

GTGCTGCGGAATCATTATCA (SEQ ID NO: 841)

Deletion plasmid pTTv425 (Aslp3-pyr4), vector backbone pTTv420

Primer Primer sequence

No new primers; pTTv420 digested with Noil and ligated with pyr4 from pTTvl81.

Deletion plasmids for pep2 with slp2 RNAi

The selection markers in vectors pTTv217 and pTTv263 (both vectors silencing slp2) were switched to a pyr4-hgh double marker. The vectors were digested with Noil restriction enzyme and the vector was purified from agarose gel. The pyr4-hgh cassette was obtained from pTTvl94 (Apep4-pyr-hgh) with Noil digestion. The new double marker and the vectors were ligated together and transformed to DH5a chemically competent E.coli. The resulting vectors pTTv376 and pTTv405 were amplified and purified. EXAMPLE 2 - Generation of 10-fold protease deletion strain with sepl deletion (M659)

To generate a 10-fold protease deletion strain, removal of the pyr4 marker was applied to the 9- fold deletion strain M574 (described in Example 14 of WO2013/102674) essentially as described in Example 3 of WO2013/102674 for removal of the pyr4 blaster cassette from the strain M195 (Apepl). Consecutive 5-FOA selection steps were carried out to ensure that the clones selected were originating from single cells.

Final clones were verified by PCR using the primers listed in Table 25-1. Signal corresponding to successful removal of the blaster cassette was obtained for majority of the clones. Removal of the blaster cassette was further verified by plating the clones onto minimal medium plates with or without 5 mM uridine. Resulting strain used in generation of 10-fold protease deletion strain was designated with strain number M597.

To remove vector sequence, plasmid pTTv255 (Asepl-pyr4) was digested with Mssl and the correct fragment purified from an agarose gel using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform 9-fold protease deletion strain M597 (Apepl Atspl Aslpl Agapl Agap2Apep4Apep3Apep5Apep2, pyr4-). Preparation of protoplasts and transformation were carried out essentially as described for the strains Ml 81 and Ml 95 using pyr4 selection in WO2013/102674.

Transformants were picked as first streaks. Growing streaks were screened by PCR (using the primers listed in Table 2-1) for correct integration. Clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 2-1. Deletion of sepl was verified by Southern analyses from selected clones (Figure 1 A, B, C) using methods described above. Clone 48-70C was designated with strain number M633. An additional single cell purification step was applied to strain M633 to obtain 10-fold protease deletion strain M659 (clone 48-70C-2).

Table 2-1 : Primers for screening removal of pyr4 blaster cassette from 9-fold protease deletion strain and for screening pTTv2551 As epl-pyr4 integration and strain purity.

For screening removal ofpyrf blaster cassette from M574 and strain purity

Primer Sequence

T1162_pep2_5f_f2 CTGTAAAGGCAGCATCGG (SEQ ID NO: 638)

T1163_pep2_3f_r2 TCAGAACGGCTTCAATCATT (SEQ ID NO: 639)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T601_pep2 fwd GACGTGGTACGACAACATCG (SEQ ID NO: 269)

T623_pep2 rev TATCAAGGTACCGGGGACAG (SEQ ID NO: 270)

For screening integration of pTTv255 (Asepl-pyr4)

Primer Sequence

T519_serendo_5int AACCACCTTGTTCTGTCCGT (SEQ ID NO: 842)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428) T061 _pyr4_orf_screen_2F TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

T521 _serendo_3int GGAACTGTCAAGATCTGGGA (SEQ ID NO: 843)

For screening deletion of sepl ORF

Primer Sequence

T 1059_serendo_orf_probef CTTCTGCAAGGAGGACGATT (SEQ ID NO: 844)

2

T 1060_serendo_orf_prober GCGATGCCGTACGTGTAC (SEQ ID NO: 845)

2

EXAMPLE 3 - Generation of 11 -fold protease deletion strain with slp8 deletion (M750)

The deletion plasmid pTTv330 for subtilisin-like protease slp8 (tre58698) is described in Example 12 of WO2013/102674.

To generate a 11 -fold protease deletion strain, removal of the pyr4 marker was applied to the 10- fold deletion strain M633 essentially as described above. Consecutive 5-FOA selection steps were carried out to ensure that the clones selected were originating from single cells.

Final clones were verified by PCR using the primers listed in Table 3-1. Signal corresponding to successful removal of the blaster cassette was obtained for majority of the clones. Removal of the blaster cassette was further verified by plating the clones onto minimal medium plates with or without 5 mM uridine, and resulting strain used in generation of 11 -fold protease deletion strain was designated with strain number M637 (clone 1-G).

To remove vector sequence, plasmid pTTv330 (Aslp8-pyr4-hph) was digested with Mssl+Sbfl and the correct fragment purified from an agarose gel using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform 10-fold protease deletion strain M637 (ApeplAtsplAslplAgaplAgap2Apep4Apep3Apep5Apep2Asepl, pyr4-). Preparation of protoplasts and transformation were carried out essentially as described above.

Transformants were picked as first streaks. Growing streaks were screened by PCR (using the primers listed in Table 3-1) for correct integration and clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 3-1. Deletion of slp8 was verified by Southern analyses from selected clones (Figure 2 A, B, C) using methods described above. Clone 54-131-2 was designated with strain number M750.

Table 3-1 : Primers for screening removal of pyr4 blaster cassette from 10-fold protease deletion strain and for screening pTTv330/ Aslp8-pyr4-hph integration and strain purity.

For screening removal ofpyrf blaster cassette from M633 and strain purity

Primer Sequence

T1173_sepl_5f_f2 TAGGTCTCGGCTGACAAGG (SEQ ID NO: 846)

T1174_sepl_3f_r2 GCTTCTTCCACTGAATGCTC (SEQ ID NO: 847)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T 1059_serendo_orf_probef2 CTTCTGCAAGGAGGACGATT (SEQ ID NO: 844)

T 1060_serendo_orf_prober2 GCGATGCCGTACGTGTAC (SEQ ID NO: 845)

For screening integration of pTTv330 (Aslp8-pyr4-hph)

Primer Sequence

T1298_slp8_5int CATCACCAAGAAGGTCCCTC (SEQ ID NO: 848)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T061_pyr4_orf_screen_2F TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

T1299_slp8_3int ACTCAATGGAACCACATCGA (SEQ ID NO: 849)

For screening deletion of slp8 ORF

Primer Sequence

T1300_slp8_orf_for CTGTGGTTGAGTGCAGATG (SEQ ID NO: 850)

T1301_slp8_orf_rev TCCCACACATCAACACAAGT (SEQ ID NO: 851)

EXAMPLE 4 - Generation of 12-fold protease deletion strain with amp2 deletion (M893)

To generate a 12-fold protease deletion strain, removal of the pyr4-hph double marker was applied to the 11 -fold deletion strain M750 essentially as described above. Consecutive 5-FOA selection steps were carried out to ensure that the clones selected were originating from single cells.

Final clones were verified by PCR using the primers listed in Table 4-1. Signal corresponding to successful removal of the blaster cassette was obtained for majority of the clones. Removal of the blaster cassette was further verified by plating the clones onto minimal medium plates with or without 5 mM uridine. In addition, clones were tested also on hygromycin plates with or without 5 niM uridine. No growth was observed on the plates without uridine supplementation or if plates contained hygromycin. Resulting marker-free strain used in generation of 12-fold protease deletion strain was designated with strain number M780 (clone 1AH).

To remove vector sequence, plasmid pTTv327 (Aamp2-pyr4-hph) was digested with Mssl+Sbfl and the correct fragment purified from an agarose gel using a QIAquick Gel Extraction Kit (Qiagen). More than 6 μg of the deletion cassette was used to transform 11 -fold protease deletion strain M780 (ApeplAtsplAslplAgaplAgap2Apep4Apep3Apep5Apep2AseplAslp8, pyr4-, hph-). Preparation of protoplasts and transformation were carried out essentially as above.

Transformants were picked as first streaks. Growing streaks were screened by PCR (using the primers listed in Table 4-1) for correct integration. Clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 4-1. Deletion of amp2 was verified by Southern analyses from selected clones (Figure 3 A, B, C). Clone 59-6A was designated with strain number M893.

Table 4-1: Primers for screening removal of pyr4-hph blaster cassette from 11 -fold protease deletion strain and for screening pTTv32T/Aamp2-pyr4-hph integration and strain purity.

For screening removal of pyr4-hph blaster cassette from M750 and strain purity

Primer Sequence

T047_trpC_term_end_F CCTATGAGTCGTTTACCCAGA (SEQ ID NO: 426)

T1299_slp8_3int ACTCAATGGAACCACATCGA (SEQ ID NO: 849)

T061_pyr4_orf_scree TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

n_2F

T624_gpdA_seqRl CTCCATATTCTCCGATGATGC (SEQ ID NO: 852)

T1420_slp8_5f_f2 TGAATTTGTTGGATCCCTGC (SEQ ID NO: 853)

T1421_slp8_3f_r2 TACCCAGGTCAAAAGAGCAG (SEQ ID NO: 854)

T1292_Hygrorf_For GCCTGAACTCACCGCGACG (SEQ ID NO: 855)

T1293_Hygrorf_Rev CCTTTGCCCTCGGACGAGTG (SEQ ID NO: 856)

For screening integration of pTTv327 (Aamp2-pyr4-hph)

Primer Sequence

T1083_amp2_screen CCACTGAAGGGGAGTTTTCA (SEQ ID NO: 857)

_5flk_fwd

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T047_trpC_term_end CCTATGAGTCGTTTACCCAGA (SEQ ID NO: 426) T1085_amp2_screen TCGCGGTATCGTATGAGATG (SEQ ID NO: 858)

_3flk_rev

For screening deletion of amp2 ORF

Primer Sequence

T1086_amp2_orf_fw GCCAGCTTCAACATCGACTT (SEQ ID NO: 859)

d

T1087_amp2_orf_re CAGCACGAGCACGTTGTACT (SEQ ID NO: 860)

EXAMPLE 5 - The 13-fold protease deletion strain having deletions ApeplAtsplAslplAgaplAgap2Apep4Apep3Apep5Apep2AseplAslp8Aamp2 Aslp7 To generate a 13 -fold protease deletion strain, removal of the pyr4-hph double marker was applied to the 12-fold deletion strain M893 (59-6A, pTTv327 in M780) as described above. Consecutive 5-FOA selection steps were carried out to ensure that the clones selected were originating from single cells.

Final clones were verified by PCR using the primers listed in Table 5-1. Signal corresponding to successful removal of the blaster cassette was obtained for majority of the clones. Removal of the blaster cassette was further verified by plating the clones onto minimal medium plates with or without 5 mM uridine. In addition, clones were tested also on hygromycin plates with or without 5 mM uridine. No growth was observed on the plates without uridine supplementation or if plates contained hygromycin. Resulting marker-free strain used in generation of 13-fold protease deletion strain was designated with strain number M901 (clone 1A2).

To remove vector sequence, plasmid pTTv329 (Aslp7-pyr4-hph) was digested with Mssl+Sbfl and the correct fragment purified from an agarose gel. Approximately 10 μg of the deletion cassette was used to transform 12-fold protease deletion strain M901 (ApeplAtsplAslplAgaplAgap2Apep4Apep3Apep5Apep2AseplAslp8Aamp 2, pyr4-, hph-). Preparation of protoplasts was carried out as above. Transformation was carried out using modified transformation method for protoplasts. In this method DNA was introduced to the protoplasts as described above but media and method for plating was changed. Transformation plates and first top agar were potato dextrose agar medium containing 1 M sorbitol for osmotic stabilisation. Protoplasts were added to the first top agar, mixed, poured onto the PD agar plates and let to regenerate for a few (2-4) hours at room temperature. After the short regeneration, a second top agar containing 10 g/1 sorbose, 10 g/1 cellobiose, 10 g/1 yeast extract, 1 M sorbitol, 5 g/1 (NH4)2S04 (+ agar & salts) and 150 μg ml Hygromycin B was poured on top of the first top agar.

Transformants were picked as first streaks. Growing streaks were screened by PCR (using the primers listed in Table 5-1) for correct integration. Clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 5-1. Table 5-1 : Primers for screening removal of pyr4-hph blaster cassette from 12-fold protease deletion strain and for screening pTTv329/Aslp7-pyr4-hph integration and strain purity.

For screening removal of pyr4-hph blaster cassette from M893 and strain purity

Primer Sequence

T1556_amp2_5f_f2 AAGTGTGCTGATGTGATGGA (SEQ ID NO: 861)

T1557_amp2_3f_r2 GCATGCGAAGTACCTTACGA (SEQ ID NO: 862)

T061_pyr4_orf_scree TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

n_2F

T624_gpdA_seqRl CTCCATATTCTCCGATGATGC (SEQ ID NO: 852)

For screening integration of pTTv329 (Aslp7-pyr4-hph)

Primer Sequence

T 1092_slp7_screen_5 TTGGTTTGAACAGCTGCAAG (SEQ ID NO: 677)

flk_fwd

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T047_trpC_term_end CCTATGAGTCGTTTACCCAGA (SEQ ID NO: 426)

_F

T1093_slp7_screen_3 ATGGTCAGCCAGAACCTGAC (SEQ ID NO: 680)

flk_rev

For screening deletion o(slp7 ORF

Primer Sequence

T1094_slp7_orf_fwd TCTTGAGCCGTTTCTCGAAT (SEQ ID NO: 681)

T1095_slp7_orf_rev CCGCTCTTAGATCGATGGTC (SEQ ID NO: 682) EXAMPLE 6 - Generation of 13-fold protease deletion strain with slp3

To generate a 13 -fold protease deletion strain, removal of the pyr4-hph double marker was applied to the 12-fold deletion strain M893 as described above. Consecutive 5-FOA selection steps were carried out to ensure that the clones selected were originating from single cells.

Final clones were verified by PCR using the primers listed in Table 6-1. Signal corresponding to successful removal of the blaster cassette was obtained for majority of the clones. Removal of the blaster cassette was further verified by plating the clones onto minimal medium plates with or without 5 mM uridine. In addition, clones were tested also on hygromycin plates with or without 5 mM uridine. Resulting marker-free strain used in generation of 13-fold protease deletion strain was designated with strain number M901 (clone 1A2).

To remove vector sequence, plasmid pTTv425 (Aslp3-pyr4) was digested with Mssl and the correct fragment purified from an agarose gel. Approximately 5 μg of the deletion cassette was used to transform 12-fold protease deletion strain M901 (ApeplAtsplAslplAgaplAgap2Apep4Apep3Apep5Apep2AseplAslp8Aamp 2, pyr4-, hph-). Preparation of protoplasts and transformation were carried out as above. Transformants were picked as first streaks. Growing streaks were screened by PCR (using the primers listed in Table 6-1) for correct integration and clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 6-1. Table 6-1: Primers for screening removal of pyr4-hph blaster cassette from 12-fold protease deletion strain and for screening pTTv425/Aslp3-pyr4 integration and strain purity.

For screening removal of pyr4-hph blaster cassette from M893 and strain purity

Primer Sequence

T1556_amp2_5f_f2 AAGTGTGCTGATGTGATGGA (SEQ ID NO: 861)

T1557_amp2_3f_r2 GCATGCGAAGTACCTTACGA (SEQ ID NO: 862)

T061_pyr4_orf_scree TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

n_2F

T624_gpdA_seqRl CTCCATATTCTCCGATGATGC (SEQ ID NO: 852)

For screening integration of pTTv425 (Aslp3-pyr4)

Primer Sequence T056_slp3_5screen_F GTGAATGGGTGGCAACATGA (SEQ ID NO: 333)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T061_pyr4_orf_scree TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

n_2F

T057_slp3_3screen_ CATCAAGTTGACCACCATTGT (SEQ ID NO: 336)

R

For screening deletion of slp3 ORF

Primer Sequence

T1581_slp3_ORF_pr AGTTAATGATGCCCGTCTTG (SEQ ID NO: 663)

obe2_f

T1582_slp3_ORF_pr GAGCGTCTCCTGTTAGCTTG (SEQ ID NO: 664)

obe2_r

EXAMPLE 7 - Generation of 13-fold protease deletion strain with pep9

To generate a 13 -fold protease deletion strain, removal of the pyr4-hph double marker was applied to the 12-fold deletion strain M893 essentially as described above. Consecutive 5-FOA selection steps were carried out to ensure that the clones selected were originating from single cells.

Final clones were verified by PCR using the primers listed in Table 7-1. Signal corresponding to successful removal of the blaster cassette was obtained for majority of the clones. Removal of the blaster cassette was further verified by plating the clones onto minimal medium plates with or without 5 mM uridine. In addition, clones were tested also on hygromycin plates with or without 5 mM uridine. Resulting marker-free strain used in generation of 13-fold protease deletion strain was designated with strain number M901 (clone 1A2).

To remove vector sequence, plasmid pTTv426 was digested with Mssl and the correct fragment purified from an agarose gel. Approximately 5 μg of the deletion cassette was used to transform 12-fold protease deletion strain M901

(ApeplAtsplAslplAgaplAgap2Apep4Apep3Apep5Apep2AseplAslp8A amp2, pyr4-, hph-). Preparation of protoplasts and transformation were carried out as above. Transformants were picked as first streaks. Growing streaks were screened by PCR (using the primers listed in Table 7-1) for correct integration and clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 7-1.

Table 7-1: Primers for screening removal of pyr4-hph blaster cassette from 12-fold protease deletion strain and for screening pTTv426/ Apep9-pyr4 integration and strain purity.

For screening removal of pyr4-hph blaster cassette from M893 and strain purity

Primer Sequence

T1556_amp2_5f_f2 AAGTGTGCTGATGTGATGGA (SEQ ID NO: 861)

T1557_amp2_3f_r2 GCATGCGAAGTACCTTACGA (SEQ ID NO: 862)

T061_pyr4_orf_screen_2F TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

T624_gpdA_seqRl CTCCATATTCTCCGATGATGC (SEQ ID NO: 852)

For screening integration of pTTv426 (Apep9-pyr4)

Primer Sequence

T1031_pep9_screen_5flk_fwd GGGTTGGAGATGTTGGAAGA (SEQ ID NO: 865)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T061_pyr4_orf_screen_2F TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

T 1032_pep9_screen_3flk_rev TTGACGAGACGGGGAGTTAC (SEQ ID NO: 866)

For screening deletion of pep9 ORF

Primer Sequence

T1033_pep9_orf_fwd CAGCCCTGACACCACTCTCT (SEQ ID NO: 867)

T 1034_pep9_orf_rev TCCAGTCCTTGGGAGAAATG (SEQ ID NO: 868)

EXAMPLE 8 - Generation of 9-fold protease deletion strains from the interferon producing strain M577

Generation of 9-fold deletion strain with ampl deletion To remove the deletion cassette, plasmid pTTv240 (Aampl -pyr4-hph) was digested with Pmel and the correct fragment was purified using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform the 8-fold protease deletion strain M577 (Apepl Atspl Aslpl Agapl Agap2Apep4Apep3Apep5; see Example 18 of WO2013/102674), which produces interferon alpha 2b. Preparation of protoplasts and transformation were carried out essentially as described above using hygromycin selection. Transformants were picked and streaked on selection plates. Growing streaks were screened by PCR (using the primers listed in Table 8-1) for correct integration. Transformants giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 8-1. The clone 108A was designated with strain number M669. Table 8-1 : Primers for screening pTTv240/ Aampl -pyr4-hgh integration and strain purity.

For screening integration of pTTv240 (Aamp I -pyr4-hph )

Primer Sequence

T840_ampl_screen_5 flk fwd TGGCATTGATCTAGAACCTCCT (SEQ ID NO: 869) T1084_screen_5flk_pyr_rev TCTTGAGCACGACAATCGAC (SEQ ID NO: 662)

T1015_screen_3flk_hygro_fwd GCATGGTTGCCTAGTGAATG (SEQ ID NO: 663)

T843_ampl_scrn_rev 3 flk GACGACTTGGTGGAGCTCAT (SEQ ID NO: 870)

For screening deletion of ampl ORF

T940_ampl_orf2_fw GACTACCCCCAGAACGTCAA (SEQ ID NO: 871)

T941_ampl_orf2_rev AAGAGGCGGATCTTTTGGTT (SEQ ID NO: 872)

Generation of 9-fold deletion strain with slp7 deletion

The deletion plasmid pTTv269 for the serine protease slp7 (trel23865) is described in Example 11 of WO2013/102674. To remove the deletion cassette, plasmid pTTv269 (Aslp7-pyr4-hph) was digested with Pmel and the correct fragment was purified using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform the 8-fold protease deletion strain M577 (Apepl Atspl Aslpl Agapl Agap2Apep4Apep3Apep5 see above). Preparation of protoplasts and transformation were carried out essentially as described above using hygromycin selection. Transformants were picked and streaked on selection plates. Growing streaks were screened by PCR (using the primers listed in Table 8-2) for correct integration. Transformants giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 8-2. The clone 5.64 was designated with strain number M673.

Table 8-2: Primers for screening pTTv269/Aslp7-pyr4-hgh integration and strain purity. For screening integration of pTTv269 (Aslp7-pyr4-hgh)

Primer Sequence

T1092_slp7_screen_5flk_fwd TTGGTTTGAACAGCTGCAAG (SEQ ID NO: 677)

T1084_screen_5flk_pyr_rev TCTTGAGCACGACAATCGAC (SEQ ID NO: 662) T1015_screen_3flk_hygro_fwd GCATGGTTGCCTAGTGAATG (SEQ ID NO: 663)

T1093_slp7_screen_3flk_rev ATGGTCAGCCAGAACCTGAC (SEQ ID NO: 680)

For screening deletion of slp7 ORF

T1094_slp7_orf_fwd TCTTGAGCCGTTTCTCGAAT (SEQ ID NO: 681)

T1095_slp7_orf_rev CCGCTCTTAGATCGATGGTC (SEQ ID NO: 682)

Generation of 9-fold deletion strain with amp2 deletion

To remove the deletion cassette, plasmid pTTv271 (Aamp2-pyr4-hgh) was digested with Pmel and the correct fragment was purified using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform the 8-fold protease deletion strain M577.

Transformants were picked and streaked on selection plates. Growing streaks were screened by PCR (using the primers listed in Table 8-3) for correct integration. Transformants giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 8-3. Clone 24A was designated with strain number M674.

Table 8-3: Primers for screening ρΎΎγΠ I /Aamp2-pyr4-hgh integration and strain purity.

For screening integration of pTTv271 ( amp2-pyr4-hph)

Primer Sequence

Tl 083_amp2_screen_5flk_fwd CCACTGAAGGGGAGTTTTCA (SEQ ID NO: 857)

Tl 084_screen_5flk_pyr_rev TCTTGAGCACGACAATCGAC (SEQ ID NO: 662)

T 1015_screen_3 flk_hy gr o_fwd GCATGGTTGCCTAGTGAATG (SEQ ID NO: 663)

Tl 085_amp2_screen_3flk_rev TCGCGGTATCGTATGAGATG (SEQ ID NO: 858)

For screening deletion of amp2 ORF

T1086_amp2_orf_fwd GCCAGCTTCAACATCGACTT (SEQ ID NO: 859)

T1087_amp2_orf_rev CAGCACGAGCACGTTGTACT (SEQ ID NO: 860) Generation of 9-fold deletion strain with sepl deletion

To remove the deletion cassette, plasmid pTTv247 (Asepl-pyr4-hgh) was digested with Pmel and the correct fragment was purified using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform the 8-fold protease deletion strain M577. Transformants were picked and streaked on selection plates. Growing streaks were screened by PCR (using the primers listed in Table 8-4) for correct integration. Clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 8-4. Clone 47.1 was designated with strain number M668.

Table 8-4: Primers for screening pTTv24T/ As epl-pyr4-hgh integration and strain purity.

For screening integration of pTTv247 (Asepl-pyr4-hgh)

Primer Sequence

T519_serendo_5int AACCACCTTGTTCTGTCCGT (SEQ ID NO: 842)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T521_serendo_3int GGAACTGTCAAGATCTGGGA (SEQ ID NO: 843)

T1015_screen_3flk_hygro_f GCATGGTTGCCTAGTGAATG (SEQ ID NO: 663)

wd

For screening deletion of sepl ORF

T504_serendo_orf_probef GCCTCCGCCCTCCTCTTCCA (SEQ ID NO: 873)

T505_serendo_orf _prober GCTTTGTCGAGCGGAGCGGT (SEQ ID NO: 874)

Generation of 9-fold deletion strain with pep9 deletion

To remove the deletion cassette, plasmid pTTv267 (Apep9-pyr4-hgh) was digested with Pmel and the correct fragment was purified using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform the 8-fold protease deletion strain M577.

Transformants were picked and streaked on selection plates. Growing streaks were screened by PCR (using the primers listed in Table 8-5) for correct integration. Clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 8-5. Transformant 77 was designated with strain number M671.

Table 8-5 : Primers for screening pTTv267 /Apep9-pyr4-hgh integration and strain purity.

For screening integration of pTTv267 (Apep9-pyr4-hgh)

Primer Sequence

T 1031 _pep9_screen_5flk_fwd GGGTTGGAGATGTTGGAAGA (SEQ ID NO: 865)

Tl 084_screen_5flk_pyr_rev TCTT GAGCACGACAATCGAC (SEQ ID NO: 662)

T 1015_screen_3 flk_hy gr o_fwd GCATGGTTGCCTAGTGAATG (SEQ ID NO: 663)

T 1032_pep9_screen_3flk_rev TTGACGAGACGGGGAGTTAC (SEQ ID NO: 866)

For screening deletion of pep9 ORF

T1033_pep9_orf_fwd CAGCCCTGACACCACTCTCT (SEQ ID NO: 867)

T1034_pep9_orf_rev TCCAGTCCTTGGGAGAAATG (SEQ ID NO: 868) EXAMPLE 9 - 9-fold protease deletion strains producing interferon in 24 well culture

The 9-fold protease deletion strains were grown in 24 well culture in TrMM with diammonium citrate without ammonium sulfate, 100 mM PIPPS, 20 g/L spent grain extract, 40 g/L lactose at pH 4.5, shaking at 28°C. Immunoblotting was done to detect interferon alpha 2b. The supernatant was diluted with water, so that 0.5 μΐ of each supernatant could be loaded into the 4-20% Criterion gel. Mixed with LSB+BME and heated at 95°C for 5 minutes. The proteins were transferred to nitrocellulose with the Turbo semi-dry blotter for 7 minutes. The nitrocellulose membrane was blocked with 5% milk in TBST for 1 hour. The interferon protein was detected with a mouse anti-interferon alpha 2b antibody (Abeam #ab9386) diluted 0.5 μg ml in TBST. The primary antibody was incubated with the membrane for 1 hour shaking at room temperature. The primary antibody was removed and the membrane washed with TBST. The secondary antibody was goat anti-mouse AP conjugate (BioRad cat#170-6520) diluted 1 :10,000 in TBST. The secondary antibody was incubated for 1 hour shaking at room temperature, the antibody was removed, and membrane washed for 1 hour shaking at room temperature. The blot was developed using AP substrate (Promega). Observing the day 7 culture samples there was a dramatic effect on the stability of interferon. The slp7 and amp2 deletion strains continued to produce interferon, while interferon was not stable in the M577 control and the ampl and sepl deletion strains. Results are shown in Figure 4.

EXAMPLE 10 - 9-fold protease deletion strains producing interferon in fermentor culture

The 9-fold protease deletion strains expressing interferon were cultivated in fermentors as batch cultivations. They were grown in TrMM plus in 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, and 40 g/L sorbose at pH 4.5 with the temperature shifting from 28° to 22° at 48h. These cultivations were done and assigned the culture codes FTR108_R1 (M668), FTR108_R2 (M669), FTR108_R6 (M673), and FTR108_R7 (M674).

The supernatant was diluted in water and sample buffer so that 0.1 μΐ could be loaded per well. The immunoblotting procedure to detect interferon alpha 2b was carried out as described above with the 24 well cultures.

The results of the fermentor cultivations can be seen in Figure 5. Standard amounts representing 50, 100, and 200 ng of interferon were used to generate a standard curve. The parental M577 control cultivation produced 0.84 g/L of interferon on day 3. The M674 strain with the amp2 protease deletion was the most improved strain overall providing 2.4 g/L on day 3. Deletion of amp2 protease provided a 2.9 fold improvement in interferon production. The slp7 protease deletion strain M673 achieved 2.1 g/L on day 4. This was a 2.5 fold improvement over the parent strain M577. The sepl protease deletion in M668 secreted 1.38 g/L of interferon. The pep9 deletion in strain M671 also improved the interferon expression level to 1.03 g/L. The M669 strain with ampl deletion produced 0.36 g/L under these culture conditions (data not shown).

The strain M674 (Aamp2) was also cultivated with and without SBTI inhibitor addition. The medium was TrMM plus 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, and 40 g/L sorbose at pH 4.5 with the temperature shifting from 28° to 22° at 48h. The Triab 125 cultivation was done without SBTI inhibitor and Triab 126 was done with SBTI inhibitor feeding (0.4 mg/ml target concentration). The SBTI inhibitor improved the interferon expression level, see Figure 6. The base level was 1.4 g/L on day 4, but with inhibitor treatment the interferon expression could be increased to 4.5 g/L on day 5. The addition of inhibitor shifted the peak expression day until day 5, which indicated higher stability of the interferon in the supernatant. The supernatant was diluted in water so that 0.025 μΐ could be loaded in 10 μΐ volume into a 4- 20% SDS-PAGE gel. Immunodetection done with Abeam (#ab9386) anti-IFN-a 2b antibody diluted to 1 μg/ml in TBST. The secondary antibody from Bio-rad (#170-6520) goat anti-mouse IgG AP conjugated secondary antibody diluted 1:5000 in TBST. The protein standards were loaded in the gel corresponding 200 ng, 100 ng, and 50 ng of full length IFN-a 2b. EXAMPLE 11 - Generation of 9-fold protease deletion strain M960 with pep2 deletion and slp2 RNAi from the interferon producing strain M788 (pyr4- of M577)

The interferon production strain M577 had the pyr4 marker in the pep5 locus, where the last protease deletion was made. To remove the pyr4 loopout marker, the M577 strain was plated on 5FOA plates. The surviving colonies were screened by PCR to check the presence and integration of the pyr4 marker. The clones that did not have the double marker by PCR were tested on minimum medium agar plates with and without uridine. Those clones that could not grow on plates without uridine were selected as marker loopout clones. One pyr4- negative clone was selected to become strain M788. Primers used in the screening are shown below in table 11-1. Table 11-1:

For screening integration of pyr4-hgh marker

Primer Sequence

T627_pep5_5int_new GTCGAAGATGTCCTCGAGAT (SEQ ID NO: 432)

T488_pyr4_5utr_rev GGAGTTGCTTTAATGTCGGG (SEQ ID NO: 428)

T061_pyr4_orf_screen_2F TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

T628_pep5_3int_new TAGTCCATGCCGAACTGC (SEQ ID NO: 435)

For screening deletion of marker

T858_pep5_5f_f3 GGAATCGTCACCAAGGAG (SEQ ID NO: 612)

T755_pep5_3 f_rev3 CTTCTGGTGACATTCCGAC (SEQ ID NO: 613) To remove the deletion cassette, the plasmids were digested with Pmel and the correct fragment was purified using a QIAquick Gel Extraction Kit (Qiagen). Approximately 5 μg of the deletion cassette was used to transform the 8-fold protease deletion strain M788 (Apepl Atspl Aslpl Agapl Agap2Apep4Apep3Apep5, pyr4-), which produces interferon alpha 2b. Preparation of protoplasts and transformation were carried out essentially as described above. The silencing cassettes were designed to integrate into the pep2 locus (tre53961).

Transformants were picked and streaked on selection plates. Growing streaks were screened by PCR (using the primers listed in Table 11-2) for correct integration. Clones giving the expected signals were purified to single cell clones and rescreened by PCR using the primers listed in Table 11-2. The transformation of pTTv376 produced a strain designated as M960 and pTTv405 produced the M961 strain.

Table 11-2: Primers for screening pTTv376 and pTTv405/ Apep2-pyr4-hgh integration and strain purity.

For screening integration of pTTv376 and pTTv405 (Apep2-pyr4-hgh)

Primer Sequence

T596_pep2 fwd 5 'flank screen CCTCTGCGTTGAGCAACATA (SEQ ID NO: 614)

T624_gpdA_seqRl CTCCATATTCTCCGATGATGC (SEQ ID NO: 852)

T600_pep2 rev 3 'flank screen CGAAAGCGTGGAGTCTTCTC (SEQ ID NO: 615)

T047_trpC_term_end_F CCTATGAGTCGTTTACCCAGA (SEQ ID NO: 426)

For screening deletion of pep2 ORF

T601_pep2 fwd GACGTGGTACGACAACATCG (SEQ ID NO: 269)

T623_pep2 rev TATCAAGGTACCGGGGACAG (SEQ ID NO: 270)

Culturing in 24 well plates Two transformants from the pTTv376 and pTTv405 transformations were grown in 24 well cultures to compare their interferon production against the control strain M577. The strains were grown in TrMM with diammonium citrate without ammonium sulfate, 100 mM PIPPS, 20 g/L spent grain extract, 40 g/L lactose at pH 4.5, shaking at 28°C. Duplicate wells were used for each transformant. Samples from the 24 well cultures taken on day 5 were used for immunoblotting. The supernatant was diluted with water, so that 0.2 μΐ of each supernatant could be loaded into the 4-20% Criterion gel. Mixed with LSB+BME and heated at 95 °C for 5 minutes. The proteins were transferred to nitrocellulose with the BioRad Turbo semi-dry blotter for 7 minutes. The nitrocellulose membrane was blocked with 5% milk in TBST for 1 hour. The interferon protein was detected with a mouse anti-interferon alpha 2b antibody (Abeam #ab9386) diluted 0.5 μg/ml in TBST. The primary antibody was incubated with the membrane for 1 hour shaking at room temperature. The primary antibody was removed and the membrane washed with TBST. The secondary antibody goat anti-mouse IRDye 680RD conjugate (Li-cor #926-68070) diluted 1 :30,000 in TBST. The secondary antibody was incubated for 1 hour shaking at room temperature. The secondary antibody was removed and membrane washed for 1 hour in TBST before scanning the membrane. The membrane was scanned at 700 nm using the Odyssey CLx near infrared imaging system (Li-cor, Inc.).

The 24 well cultivation results can be seen in Figure 7. Both strains with slp2 silencing produced high amounts of interferon. The best pTTv405 transformant 405b produced 427 mg/L of interferon which was assigned strain number M960. The best pTTv376 transformant 376a produced up to 534 mg/L of interferon and was called strain M961. The control strain (M577) reached up to 200 mg/L. The M961 strain produced 2.7 times more interferon in this cultivation and 2.1 times more with M960 compared to control strain, respectively.

Fermentation

The two strains were cultivated in 1L ferment or s as batch cultures in TrMM in 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, and 40 g/L sorbose at pH 4.5 with the temperature shifting from 28° to 22° at 48h. Triabl52 and Triabl53 cultivations were done with strains M960 and M961, respectively. The supernatant samples were diluted in water and loading dye so that 0.05 μΐ of supernatant was loaded per well. Immunoblotting for detection was done as described above for interferon. Standard amounts of interferon representing 400, 200. 100, 50 and 25 ng were used to construct a standard curve to determine expressed concentration. In cultivation Triabl52 the M960 strain produced 1.4 g/L on day 3 (Figure 8) and the M577 strain produced 1.2 g/L on day 3 (data not shown). The strain M961 achieved 3.2 g/L on day 4 which was 2.7 times more interferon than the parental strain M577.

EXAMPLE 12 - Introduction of heterologous proteins into the protease deletion strains

To generate the MAB01 antibody producing T. reesei strain in any of the protease deleted strain(s) of the invention a strain is transformed with MAB01 light and heavy chain constructs (pTTv98, pTTv99, pTTv67, pTTvlOl, pTTvl02, and/or pTTv223, see also Examples 1-3 of WO2013/102674) using hygromycin and acetamide in selection as described above. To produce rituximab antibody, constructs having harmonised heavy chain fused with CBHI carrier and light chain fused with CBHI carrier are transformed to protease deletion strains of T. reesei as described above.

To express various antibody fragments (Fabs, multimeric single domain antibodies (sd-Ab's) and scFVs) in different protease deletion backgrounds of the invention methods of Example 23 are applicable. The architecture of the genetic expression cassettes applied for this purpose is usually based on the regulatory elements (promoter and terminator) of the cellobiohydrolase I {cbhl) gene. The catalytic domain of the CBHI protein is modified to remove intron sequences and used as fusion partner to enhance antibody fragment expression and secretion. A recognition motif for the Kex2 protease is inserted in between the fusion partners to promoter co-secretory release of the antibody fragments from the CBHI carrier protein and the expression cassettes are flanked by homologous regions to allow targeted integration to the T. reesei cbhl locus.

T. reesei protease deletion strains are transformed with the purified expression cassettes as decribed and selected for appropriate selection marker. Transformants are screened by PCR for homologous integration of the expression cassette to the cbhl locus using a forward primer outside the 5' flanking region fragment of the construct and the reverse primer inside the modified CBHI catalytic domain (5' integration) as well as a forward primer inside the selection marker gene(s), respectively, and a reverse primer outside the 3' flanking region fragment (3' integration). Proper integration of the disruption cassette is reconfirmed by PCR using the same primer combinations as described above and the absence of the parental CBHI locus is verified by using a primer combination targeted to the cbhl open reading frame. Correct integration of the disruption cassette is additionally verified for all clones applying Southern hybridization.

Transformed T. reesei strains are cultivated in fermentation and shake flask conditions as described earlier. Samples are collected in the course of the cultivations, and production levels are analysed, for example, by affinity liquid chromatography as described above. The quality of the purified samples is checked by SDS-PAGE.

EXAMPLE 13 - Glycoengineering of the protease deletion strains

Generation of GO producing strains The generation of alg3 deletion plasmids pTTgl56 (human full length GnTl and human full length GnT2) and pTTgl73 (Kre2 N-terminal fusion with catalytic domain of human GnTl and full length human GnT2) are described in Example 19 of WO2013/102674.

A T. reesei MAB01 expression strain is transformed with the Pmel fragments of pTTgl56 and pTTgl73. Variable amount of transformants (100-170 depending on the construct) are picked onto selective plates and on the basis of PCR screening clones with positive results concerning 5'- and 3 '-integration are selected for single spore platings and re-screening for integration and alg3 deletion using primers as described in Table 19.3 of Example 19 of WO2013/102674. PCR- screened strains are subjected to shake flask and fermentation cultivations, samples are obtained in appropriate days and the samples are subjected to antibody concentration and glycan analyses. Generation of GlcNacMan5 producing strains

The generation of plasmids for fusion proteins of targeting peptide and catalytic domain of human GnTl plasmids pTTv274 (N-terminal portion of human GnT2), pTTv275 (N-terminal portion of T. reesei Kre2), and pTTv278 (N-terminal portion of T. reesei Ochl) are described in Example 21 of WO2013/102674. Fragments for transformations are released from the above plasmids with Pmel. All fragments are transformed individually to a MAB01 (or an antibody such as rituximab) expressing strain and protoplast transformations are carried out essentially as described above.

Well growing clones on selective streaks are screened for the 5 ' and 3 ' integration into the egl2 locus. Double integration-positive clones are additionally screened for the loss of the egl2 ORF. The clones giving the desired results are purified through single spore platings, and the single spore-derived clones are verified by PCR to be pure integration strains. Selected strains are subjected to shake flask and fermentation cultivations, samples are obtained in appropriate days and the samples are subjected to antibody concentration and glycan analyses. For N-glycan analysis MAB01 is purified from culture supernatants using Protein G HP MultiTrap 96-well filter plate (GE Healthcare) according to manufacturer's instructions and the antibody concentrations are determined via UV absorbance against MAB01 standard curve. N- glycans are released from EtOH precipitated and SDS denatured antibody using PNGase F (ProZyme Inc.) in 20 mM sodium phosphate buffer, pH 7.3, in overnight reaction at +37°C. The released N-gl yeans are purified with Hypersep C-18 and Hypersep Hypercarb (Thermo Scientific) and analysed with MALDI-TOF MS.

EXAMPLE 14 - Protease Homologs

T. reesei sepl, ampl, amp2, and pep9 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 protease amino acid sequences as queries. Trichoderma virens and Trichoderma atroviride BLAST searches were conducted using the DOE Joint Genome Institute's web site (Trichoderma virens Gv29-8 v2.0 and Trichoderma atroviride v2.0, respectively). Sequence hits from the BLAST searches were aligned using the ClustalW2 or Clustal Omega alignment tool provided by EBI. Phylogenetic trees were also generated using the sequence alignments.

Figure 9 depicts a phylogenetic tree of ampl and amp2 of selected filamentous fungi. Figure 10 depicts a phylogenetic tree of sepl of selected filamentous fungi. Figure 11 depicts a phylogenetic tree of pep9 of selected filamentous fungi.

EXAMPLE 15 - Protease activity measurement of protease deficient T. reesei strains The protein concentrations were determined from supernatant samples from day 2-7 of lx-7x protease deficient strains according to EnzChek protease assay kit (Molecular probes #E6638, green fluorescent casein substrate). Briefly, the supernatants were diluted in sodium citrate buffer to equal total protein concentration and equal amounts of the diluted supernatants were added into a black 96 well plate, using 3 replicate wells per sample. Casein FL diluted stock made in sodium citrate buffer was added to each supernatant containing well and the plates were incubated covered in plastic bag at 37°C. The fluorescence from the wells was measured after 2, 3, and 4 hours. The readings were done on the Varioskan fluorescent plate reader using 485 nm excitation and 530 nm emission. Some protease activity measurements were performed using succinylated casein (QuantiCleave protease assay kit, Pierce #23263) according to the manufacturer's protocol.

The pepl single deletion reduced the protease activity by 1.7-fold, the pepl/tspl double deletion reduced the protease activity by 2-fold, the pepl/tspl/slpl triple deletion reduced the protease activity by 3.2-fold, the pepl/tspl/slpl/gapl quadruple deletion reduced the protease activity by 7.8-fold compared to the wild type M124 strain, the pepl/tspl/slpl/gapl/gap2 5-fold deletion reduced the protease activity by 10-fold, the pepl/tspl/slpl/gapl/gap2/pep4 6-fold deletion reduced the protease activity by 15.9-fold, and the pepl/tspl/slpl/gapl/gap2/pep4/pep3 7-fold deletion reduced the protease activity by 18.2-fold.

The Figure 20 graphically depicts normalized protease activity data from culture supernatants from each of the protease deletion supernatants (from 1-fold to 7-fold deletion mutant) and the parent strain M124. Protease activity was measured at pH 5.5 in first 5 strains and at pH 4.5 in the last three deletion strains. Protease activity is against green fluorescent casein. The six-fold protease deletion strain has only 6% of the wild type parent strain and the 7-fold protease deletion strain protease activity was about 40% less than the 6-fold protease deletion strain activity. EXAMPLE 16- IGFl production in the 13-fold protease deletion strain

An IGFl expression cassette was created from a plasmid, which contained cbhl promoter, terminator and 3 'flank and amdS marker, to express IGFl as a fusion protein with CBHI carrier and without any carrier cleavage sites or purification tags. The construct was designed to be integrated into the cbhl locus under control of the native cbhl promoter and terminator. The expression construct was cut out of the vector with Pmel and purified via standard methods. The Trichoderma transformation was made into the 13 -fold protease deletion strain Ml 076 using AmdS selection marker (M1076 was generated from M901 by deleting pep9 (tre79807)). Transformants were screened by PCR for loss of cbhl orf and proper integration of the expression cassette into the locus.

The CBHI carrier sequence:

(SEQ ID NO :924)

The IGFl protein sequence expressed: (SEQ ID NO :925) The Ml 140 was grown in the fermentator cultivation T189 without the use of protease inhibitors in TrMM with 20 g/L yeast extract, 40 g/L cellulose, 100 g/L cellobiose, 40 g/L sorbose at pH 4.5 in a 1 liter fermentor. The temperature was shifted from 28°C to 22°C after 48 hours.

The expression level was checked via immunoblotting. The samples were diluted in orange LSB so that 0.0125 μΐ of supernatant could be loaded per well in a 4-20% Criterion gel. Immunoblotting was done to dual detect the IGFl and CBHI expression. Detection was done with rabbit anti-IGFl diluted to 0.25 μg ml in TBST (Abeam rabbit polyclonal antibody, ab9572) and anti-CBHI mab261 diluted 1: 10,000 in TBST. The secondary antibody was goat anti-rabbit IgG 1:30,000 dilution in TBST (Li-Cor #926-68071, IRDye 680RD goat anti-rabbit IgG). The anti-mouse CBHI antibody was detected with Li-Cor #926-32210 IRDye 800CW goat anti- mouse IgG diluted 1:30,000 in TBST. Washed for 1 hour in TBST and rinsed with TBS. The filters were scanned with LI-COR Odyssey CLx Infrared Imaging System at 700 nm and 800 nm. The expression of IGFl was measured from 44 hours to 162 hours compared to an IGFl standard curve. The expressed fusion protein was around 70 kD. At 71 hours the expression level was 3.5 g/L. The highest expression level measured was 7.9 g/L at 92 hours. At 116 hours the expression level reduced to 6.6 g/L. The IGFl expression reduced significantly after 116 hours in this batch cultivation. The C0 2 peak was around 92 hours, which correlated well with the expression peak. After 116 hours the IGFl protein was degraded off the carrier and only CBHI protein could be seen in the samples. CBHI expression could be observed from 71 hours until 162 hours. No protease inhibitors were used in this cultivation. The 13 protease deletions improved the stability of the IGFl . Protease inhibitors may be needed to reach higher production levels in strains having less protease deletions. With Ml 140 it would be possible to achieve higher production levels if more cellulose was used in the cultivation.

The M231 production strain was made to produce a CBHI-TEV site- IGFl fusion protein in a strain with no protease deletions, M124. The TEV protease cleavage site was included in between the CBHI carrier and IGFl . IGFl was very stable and remained predominately in the fusion form after secretion into the supernatant. The expression construct contained the CBHI carrier-TEV cleavage site- IGFl protein sequence and was designed to be integrated into the CBHI locus under control of the native promoter and terminator. After transformation transformants were screened by PCR for loss of cbhl orf and proper integration of the expression cassette into the cbhl locus. The final transformant was named M231. CBHI carrier sequence used to create M231 was the same as for IGFl above except after C- terminal amino acids "...TTTGSS" of the CBHI amino acids "PGP" were included before TEV cleavage site (ENLYFQ).

A second strain that produced an IGFl variant called BVS857 was made in a similar way as M231 but the expression construct included a strepll tag and spacer before the TEV cleavage site. The expression cassette was integrated into the cbhl locus under control of the native cbhl promoter and terminator. The vector was digested with Pmel, the expression cassette was gel purified, and was transformed into the Ml 94 strain and selected for using AmdS selection. Transformants were screened by PCR for loss of cbhl orf and proper integration of the expression cassette into the cbhl locus. The resulting strain M236 strain produced a CBHI- streptll tag-3x(GGGS)-TEV site-BVS857 fusion protein. The strepll tag was added to allow for purification, which could be conveniently removed afterward with TEV protease treatment.

The IGF1 strain M231 has no protease deletions, while the BVS857 strain M236 has the pepl and tspl protease deletions. Both these strains were expressed as stable CBHI-IGFl fusions into the supernatant. The M231 and M236 were cultivated in 1 L fermentors in the presence of chymostatin (20 μΜ) and pepstatin (10 μΜ) inhibitors. Both strains were cultivated in TrMM with 20 g/L yeast extract, 40 g/L cellulose, 100 g/L cellobiose, 40 g/L sorbose at pH 4.5 with chymostatin and pepstatin inhibitors added on day 3, 4, and 5. The temperature was shifted from 28°C to 22°C after 48 hours.

The production levels were assessed by immunoblotting. The M231 culture supernatant was diluted with orange LSB so that 0.025 μΐ could be loaded into a 10 μΐ volume into a 4-20% criterion gel with IGF1 standards. The M236 culture supernatant was diluted so that 0.05 μΐ of supernatant was loaded to each well along with BVS857 standards onto a 4-20% criterion gel. Both blots were detected with rabbit anti-IGFl diluted to 0.25 μg ml in TBST (Abeam rabbit polyclonal antibody, ab9572) and anti-CBHI mab261 diluted 1: 10,000 in TBST. The secondary antibody was goat anti-rabbit IgG 1 :30000 dilution in TBST (Li-Cor #926-68071, IRDye 680RD goat anti-rabbit IgG). The anti-mouse CBHI antibody was detected with Li-Cor #926-32210 IRDye 800CW goat anti-mouse IgG diluted 1:30,000 in TBST. Washed for 1 hour in TBST and rinsed with TBS. The filters were scanned with Li-Cor Odyssey CLx Infrared Imaging System at 700 nm and 800 nm.

The strain M231 demonstrated that up to 19 g/L of IGF1 could be produced as a fusion to CBHI carrier at 118 hours. The fusion protein ran around 75 kD on the immunoblot. The CBHI-IGFl fusion protein expression level was 2 g/L at 72 hours, 10 g/L at 95 hours, 16 g/L at 101 hours, and 19 g/L at 118 hours. The CBHI expression level accumulated throughout the batch cultivation from 49 hours to 118 hours. With strain M236 up to 7 g/L of the BVS857 variant could be produced as a CBHI carrier fusion at 101 hours. The fusion protein ran around 75 kD on the immunoblot. The CBHI-BVS857 fusion protein expression level was 2.7 g/L at 72 hours, 6.2 g/L at 95 hours, 7.0 g/L at 101 hours, 5.1 g/L at 118 hours, and 2.5 g/L at 140 hours. As a fusion with CBHI carrier both these IGF1 proteins are stably expressed into the supernatant.

The M236 IGF1-BVS857 productions strain was further cultivated in fermentors with and without chymostatin and pepstatin inhibitors to see the effect of proteases. This strain was cultivated previously with inhibitors and using 40 g/L cellulose. The M236 strain was cultivated in TrMM with 20 g/L yeast extract, 80 g/L cellulose, 100 g/L cellobiose, 40 g/L sorbose at pH 4.5 with and without chymostatin and pepstatin inhibitors added on day 3, 4, and 5. The temperature was shifted from 28°C to 22°C after 48 hours.

No CBHI-BVS857 fusion protein or free protein could be detected in the supernatant samples taken from 71 to 115 hours in strain M236 (no added protease inhibitors). Only the CBHI carrier protein could be detected accumulating in the supernatant during that time. The same strain grown in the presence of chymostatin and pepstain inhibitors produced up to 9 g/L of CBHI- BVS857 fusion at 104 hours. The BVS857-CBHI fusion protein was detected at 75 kD. The expression level determined was 3.0 g/L at 71 hours, 6.3 g/L at 92 hours, 4.9 g/L at 98 hours, 9.0 g/L at 104 hours, and 7.8 g/L at 115 hours. The CBHI carrier levels increased throughout the culture and was maximal at 115 hours. Thus, the improvement in BVS857-CBHI production levels was dramatic and due to protease inhibition.

With the use of 40 g/L of cellulose production levels of 7.0 g/L was reached at 101 hour for BVS857-CBHI and with 80 g/L of cellulose production levels were 9.0 g/L at 104 hours. This demonstrates that higher production levels are achieved by adding more cellulose to the culture medium. CBHI-streptll tag-3x(GGGS)-TEV site-BVS857 fusion protein from M236 was purified via strep-tag affinity column (IB A GmbH) according the manufacture's protocol. The culture supernatant (600 μΐ) was applied to the column and washed with 10 volumes of wash buffer. The column was eluted with elution buffer containing 2.5 mM desthiobiotin. The fractions were run on a 4-20% criterion gel and stained with gel code blue Coomassie strain. The eluted fractions #2 and #3 contained concentrated protein that was slightly less than 75 kD, where the BVS857 fusion protein should run. The eluted fraction #2 was used for testing TEV protein cleavage efficiency. The AcTEV protease (Invitrogen, catalog# 12575-015) was used for overnight incubation at 8°C with different amounts of enzyme. Standard amounts of IGFl were used on the immunoblot to quantify the amounts IGFl present in each reaction. The amount of TEV was varied to evaluate its effectiveness (0, 1 μΐ, 5 μΐ, 10 μΐ, or 15 μΐ). TEV (5 μΐ) plus BVS857 control (4 μg) showed that there did not seem to be any significant TEV protease activity against BVS857. The baseline amount of IGFl in the carrier fusion sample was 687 ng. Using 1 μΐ (10 units) TEV enzyme converted 46% of the fusion after 16 hours at 8°C. The 5 μΐ (50 units) amount converted about 95% of the fusion. This peaked at 97% when 10 μΐ (100 units) of TEV was used. The TEV enzyme gave a background staining in the immunoblots at 25 kDa. The released BVS857 product at the size of the native IGFl could be easily observed around 10 kDa, but was below the standard curve range. PMSF was used in the reaction buffer, but it did not seem to neutralize the protease activity responsible for degrading the BVS857 after it was released from the carrier. The strain producing this material had only 2 protease deletions so protease inhibitors were used to try to control the protease activity. This experiment showed that the internal strepll tag can be used to affinity purify the CBHI fusion protein from supernatant and the TEV cleavage site is efficiently cut in order to release the model protein such as IGFl .

EXAMPLE 17 - FGF21 expression strains in the 13-fold protease deletion strains

The first FGF21 production strain was made in M369 (Apepl Atspl Aslpl Agapl Agap2, see Example 4 of WO2013/102674) using vector with an FGF21 expression cassette targeted to the cbhl locus, which contained a hygromycin marker. The vector was digested with Pmel and processed for transformation into the M369 strain, and transformants were selected for with hygromycin. The expression construct produced a CBHI carrier- NVISKR kex2 site-FGF21 fusion protein, so that the secreted protein would be the free FGF21 protein. Correct integration into the locus and absence of the cbhl orf was checked by PCR.

The CBHI carrier sequence was the same as for IGF1 above, followed by NVISKR Kex2 cleavage site and FGF21 sequence: (SEQ ID NO :926)

The M393 strain was grown with and without protease inhibitors pepstatin A, chymostatin, or soybean trypsin inhibitor. Independent wells were chosen for control wells where no inhibitors were added. This strain was grown in 3 ml of TrMM with diammonium citrate without ammonium sulfate, 100 mM PIPPS, 20 g/L spent grain extract, 40 g/L lactose adjusted to pH 4.5. The 24 well plates were shaken at 800 rpm at 85% humidity. The plate was covered with an air permeable membrane and the cultures were grown for 6 days.

Inhibitors were added first on day 3 and then added daily until day 6. 200 μΐ samples were taken from the culture wells beginning on day 3. The mycelium was spun down for 5 minutes at 13k and the supernatant collected. From the culture supernatant 5 μΐ plus LSB was loaded in a 4-20% SDS PAGE gel and immunoblotting made on nitrocellulose with rabbit anti-FGF21 (2 μg/ml) and goat anti-rabbit IgG AP conjugated secondary antibody diluted 1:10,000. FGF21 standard were included on the blot for quantification.

FGF21 was very sensitive to protease degradation. On day 6 without treatment only about 2 mg/L of total protein could be produced of which very little was full length material (Figure 12). With 10 μΜ pepstatin A added to the cultures the production could be improved up to 160 mg/L on day 6. In the presence of inhibitor a major degradation product around 18 kD was detected.

Adding SBTI or chymostatin, in addition to pepstatin A, did not produce an additional benefit.

SBTI addition resulted in a much more acidic culture sample and thus produced a negative effect on the production level. Combination of pepstatin A and SBTI provided the most benefit as it allowed for 204 mg/L of total FGF21 to be produced. The expression levels were in the 1-12 mg/L range in the presence of single inhibitors and untreated cultures reached about 1 mg/L. It was important to inhibit both the aspartic proteases and the serine proteases to effectively produce FGF21.

The M393 strain was cultured in the fermentor in TrMM plus 2% yeast extract, 4% cellulose, 8% cellobiose, 4% sorbose, pH 4.5 with temperature shifted from 28° to 22° at 48h. When FGF21 expression was analysed via immunoblotting the highest expression level was seen on day 3 where there was a 10 kD degradation product produced at 130 mg/L. No full length FGF21 was observed. The FGF21 was very protease sensitive when produced in the 5-fold protease deletion strain. More protease activity reduction would be required via inhibitor treatment or protease deletions. When inhibitors were used in 24 well cultures lots of protein was observed, suggesting FGF21 seemed to be well secreted but was just sensitive to degradation.

An FGF21 expression vector with AmdS marker was transformed into the strain M1076 and Ml 085 (slp7 deletion with /?jr4-hygromycin double marker).

The pTTv470 vector was created by taking the AmdS marker as a Notl fragment from pTTv249 and adding it to the previously made pTTvl74 FGF21 expression vector. The selection marker was simply exchanged to create the new vector. The expression cassettes in pTTvl74 and pTTv470 were identical. The pTTv470 vector was digested with Pmel, the expression cassette was gel purified, and the FGF21 expression cassette with AmdS marker was transformed into the two of the 13-fold protease deletion strains Ml 076 (pep9 deletion) and Ml 085 (M901 with slp7 deletion). Following the AmdS selection, the resulting transformants were screened by PCR for correct 5' and 3' integration and presence or absence of cbhl open reading frame. The primer pairs used are shown below in table 17.1.

Table 17.1. Primers for screening integration into the cbhl orf.

5' integration, T095 + T096, -2.8 kb (2776 bp), 58°C

T095_Annl 12_F_cbhI_Ben GCTGTTCCTACAGCTCTTTC (SEQ ID

NO :927)

T096_Annl 13_R_cbhI_Exon_Ben AGCCGCACGGCAGC (SEQ ID NO :928)

3' integration, T008 + T022, -1.9 kb, 60°C

T008_pHHOl-CBHIloc_cbhl3'flankOutRev GGTTGACTTACTCCAGATCG (SEQ ID NO :929)

T022_Amds_start_rc_seg CTGAAGCAACAGGTGCCAAG (SEQ ID

NO :930)

ORF, T1720+T1721, gives a PCR band of 770 bp if the cbhl gene is not deleted. Pure transformants do not give signal. 68° C

T 1720_cbh 1 intronfor CCTGACGCTATCTTCTTGTTGG(SEQ ID

NO :931)

T 1721 _cbh 1 intronrev CGCGCATGTTTGTCCATCAAAC(SEQ ID

NO :932)

Positive transformants were found for both transformed strains. The M1076 transformation produced 13 good transformants and the M1085 produced 2 good transformants. These transformants were cultivated in 24 well cultures and compared to the earlier FGF21 production strain M393. The transformants and the control were grown in 10 g/L yeast extract, 20 g/L cellobiose, 10 g/L sorbose with PIPPS, pH 4.5. The cultures were started by adding 1 x 10 spores into 50 ml of culture medium and adding 3 ml of culture medium plus spores to each well (6 x 10 5 spores /well). 100 μΐ of each culture supernatant was collected on days 4, 5, and 6. The mycelium was spun down and orange LSB was added to the supernatant and the samples were heated for 5 minutes. 5 μΐ and 2 μΐ of the supernatant + LSB was loaded per well into 4-20% Criterion TGX gel. The gels were run and the proteins were transferred to nitrocellulose membrane.

The expressed FGF21 was detected by immunoblotting. The rabbit anti-FGF21 antibody was diluted to 2 μg/ml in TBST. The stock concentration provided was 3600 μg ml (from Novartis). The goat anti-rabbit secondary IRDye 680 was diluted 1:30,000 in TBST. The primary and secondary antibody incubations were done for 1 hour at room temperature with shaking. To detect the carrier CBHI levels the anti-CBHI antibody mab261 was diluted 1: 10,000 in TBST. The goat anti-mouse IRDye 800 secondary antibody was diluted 1:30,000 in TBST. Washed the membranes with TBST for 1 hour and rinsed with TBS. The blots were scanned at 700 and 800 nm. The day 4, day 5, and day 6 samples were analysed for FGF21 and CBHI expression. The difference between the M393 control strain and the new transformants with 13 protease deletions was dramatic. The secreted FGF21 was primarily observed as a 10 kD product. This product was only faintly visible in the M393 supernatants from day 4, but was massively expressed in supernatant from all of the 13 new transformants (Figure 13). On later days there was no FGF21 detected from the M393 strain. There was free CBHI carrier expressed in all the M393 control samples as well as in the new FGF21 transformants. This indicated that all the strains secreted both the CBHI carrier as well as the FGF21 protein, but there was too much protease activity for the FGF21 in the M393 strain. The eight additional proteases deleted in the new transformants dramatically improved the stability of the FGF21. Four of the transformants #24, 36, 48, and 73 seemed to produce higher amounts of CBHI and FGF21 compared to the nine other transformatants. The difference was particularly obvious on days 5 and 6. After Southern analysis this was determined to be due to multiple integration of the FGF21 expression cassette. The lower expression levels correspond to single copy integration. The M393 strain has 5 protease deletions, while the new strains produced have 13 proteases deleted. The transformants from the transformation of the M1085 strain were done in parallel. The results were similar as those seen in Figure 16. The M1085 based strain also produced FGF21 product mainly at 10 kD, but there was a weaker 17 kD product visible on day 4.

Southern blot analysis was done on the several transformants. Transformant numbers #20, 24, 36, 48, 55, 73 from the M1076 strain and #4 and 39 from the M1085 were analysed via Southern blot using radioactive detection. The genomic DNA was digested with Pstl for integration checking. Two PCR fragments were used as the probes for the cbhl promoter and for the cbhl 3' flank. The primers used to create the fragments are listed below. cbhl promoter probe, 799 bp fragment, 35 ng/μΐ (tube labelled 5 ' probe)

T 173_pcbh 1 _seq_r 1 CAAAGGCCGAAGGCCCGAGG (SEQ ID NO :933)

T1679 CAACCTTTGGCGTTTCCCTG (SEQ ID NO :934) cbhl 3' flank probe, 796 bp fragment, 32.2 ng/μΐ (tube labelled 3' probe) T178_cbhl 3flank_seq_f2 GGCCGCAGGCCCATAACCAG (SEQ ID NO :935)

T1680 TGAGTGGGGGATGACAGACA (SEQ ID NO :936)

The PstI digest and detection with both probes should give two bands at 3.9 kb and 4 kb if correctly integrated into the cbhl locus. Effectively there will only be one band at 4 kb. The native cbhl locus gives a signal around 6.7 kb. The transformant numbers 20, 55, 4, and 39 showed the expected integration band at 4 kb. These look to be clean single integrations into the cbhl locus. As seen from the 24 well culture studies, these transformants displayed a lower expression level than many of the other transformants. The highest expression levels were seen from transformants #24, 36, 48, and 73, which coincidentally showed extra integration signals in the Southern blotting. There was a nonspecific band in all the transformants at 2.3 kb.

The multiple copy containing transformants #24 and #48 were named Ml 200 and Ml 201. The single copy strains from transformant #20 and #55 were named M1202 and M1203. From the M1085 based strain transformants #4 and #39 were named M1204 and M1205, which were single copy strains. The FGF21 production strains were cultivated in the 1L fermentors with and without protease inhibitors. Cultivations were done with M1200, M1201, M1204, and M1205 strains. These strains were cultivated in TrMM with 20 g/L yeast extract, 40 g/L cellulose, 100 g/L cellobiose, 40 g/L sorbose at pH 4.5. Pepstatin, chymostatin, and SBTI inhibitors were added to the M1200 and Ml 205 cultures on day 3, 4, and 5. The supernatant samples were diluted in orange sample buffer so that 0.1 μΐ of supernatant could be loaded per well into a 4-20% Criterion PAGE gel.

The expressed FGF21 was detected by immunoblotting as above.

The cultivation of Ml 200 primarily produced a 10 kD product, but there were higher molecular weight products up to 17 kD (Figure 14). The full length FGF21 ran at 20 kD. On day 5 there was 2.0 g/L of the 10 kD product. Use of inhibitors increased the stability of the secreted FGF21. The major product was now 17 kD and produced up to 2.0 g/L on day 5. With inhibitor treatment the full length version of FGF21 could be produced at 0.2 g/L. This appeared to be full length. Cultivation of Ml 205 produced a 10 kD band at 2.4 g/L on day 5. Inhibitor treatment increased the stability of the FGF21 product to generate a predominate 17 kD form at 3.5 g/L and a full length product at 0.2 g/L on day 4 (Figure 14). Samples from the cultivation of M1201 had a technical problem with the fermentor system (analysis not shown). The M1204 strain was analysed and gave similar expression levels, 2.5 g/L, of the 10 kD product on day 5. The 10 kD product was lowest on day 2 and was highest on day 5.

The inhibitor treatment demonstrated that it would be possible to fully stabilize the FGF21 protein. Further protease deletions to produce FGF21 (without added protease inhibitors) include but are not limited to slp2, pep8, and pepll. Inhibitor studies on FGF21 production strain

To investigate which classes of proteases are contributing most to the degradation of the FGF21 protein individual protease inhibitors we tested in 24 well cultures.

Chymostatin and SBTI are known to inhibit SLP2 and SLP7 subtilisin proteases that are still expressed in the M1200 strain. Pepstatin inhibits aspartic proteases such as PEP8, PEP11, PEP12 which are known to be secreted into the supernatant. 1,10-phenathroline targets mainly zinc metalloproteases which are secreted by Trichoderma. The Ml 200 strain was cultivated in 24 well format in TrMM with 10 g/L yeast extract, 20 g/L cellobiose, 10 g/L sorbose, PIPPS buffered at pH 4.5. Chymostatin, phenathroline, and pepstatin were used at a final concentration of 10 μΜ and SBTI was used at 0.1 mg/ml. Each treatment was done in duplicate wells and the untreated control was cultured in 4 wells.

η

The cultures were started by adding 1 x 10 spores into 50 ml of culture medium and adding 3 ml of culture medium plus spores to each well (6 x 10 5 spores /well). 100 μΐ of each culture supernatant was collected on days 4, 5, and 6. The mycelium was spun down to remove only the supernatant. Orange LSB was added and the samples were heated for 5 minutes. 2 μΐ of the supernatant + LSB was loaded per well into 4-20% Criterion TGX gel. The gel was run and proteins were transferred to nitrocellulose membranes. The expressed FGF21 was detected by immunoblotting as above. A commercially purchased N- terminal antibody rabbit polyclonal from Abeam (#ab66564) and C-terminal rabbit polyclonal antibody from Abeam (#abl37715) were used in some cases.

Without inhibitor treatment the Ml 200 strain produced mainly a 10 kD product, but there were some larger forms visible in the day 4 samples (Figure 15). Chymostatin treatment improved the stability of the protein allowing a 17 kD major product. There were small amounts of the full length form visible and a dimer form around 37 kD after chymostatin treatment. Pepstatin, SBTI, and 1 ,10-phenathroline treatment only improved the expression level of the 10 kD form. However, when chymostatin and pepstatin were combined there was a synergetic effect on the stabilization. The combined treatment promoted almost complete stabilization of the full length form and reduced the lower molecular weight products. This was most apparent in the 6 day culture samples, where mainly the full length FGF21 could be visualized. The appearance of the dimer was strongest with this treatment. Proper formation of the dimer indicates that the molecule has a C terminal cysteine residue needed for dimerization.

The culture samples were probed also with an N- and C-terminal antibody. The dual treatment with chymostatin and pepstatin generated a full length product that reacted to the N-terminal antibody as well as the FGF21 standard (Figure 15). When the C-terminal antibody was used to detect FGF21 produced in the 5 day samples two products were observed when treated with chymostatin or chymostatin/pepstatin (Figure 16). These appear to be the full length protein and the lowest 10 kD form. Chymostatin treatment was enough to stabilize the C-terminus of the protein. To maintain the N-terminus both chymostatin and pepstatin were necessary.

The data from the day 4 immunoblots was quantified to make comparisons of which inhibitors were most effective. The 1,10-phenathroline treatment best improved the total amount of FGF21 produced, thus indicating that zinc metalloproteases were likely involved (Table 17.2). There are many candidates such as mpl, mp2, mp3, mp4, and mp5. The full length form seems to be degraded by a subtilisin and an aspartic protease. The SLP2 protease is most likely to be the primary problem for the stability and can be addressed by, for example, deleting the gene, silencing the gene, or switching the promoter of the gene. Deleting or silencing aspartic proteases PEP8, PEP11 , and PEP12 found in the supernatant may further increase the stability of FGF21. In some embodiment, full-length production of FGF212 may only need deletion of two proteses, slp2 and pep8, in the strain Ml 200.

Table 17.2. Fluorescent units of FGF21 expression measured from the day 4 immunoblot shown in Figure 15.

EXAMPLE 18 - Reducing slp2 expression by replacing its promoter slp2 affects in some level growth and sporulation and therefore slp2 promoter was replaced with a promoter which is expressed in lower levels (of all identified proteases slp2 mRNA was expressed the highest levels in the conditions described in the last Example).

The promoters from the sporulation induced flavin containing monooxygenase gene tre76230, the RNA polymerase gene tre49048, and the slp8 protease gene tre58698 were chosen for replacement into the slp2 promoter locus. All genes appeared to be moderately expressed at levels less than those seen with slp2. A promoter region from the 3 genes was amplified by PCR and inserted into a vector with slp2 flank sequences that would direct the promoter into the correct position as the new slp2 promoter. The promoter cassette contains the hygromycin marker upstream of the new promoter. The vectors were digested with Pmel, the replacement promoter constructs were gel purified, and transformed in the M507 MABOl production strain under hygromycin selection conditions. Two transformants from each Transformants were isolated and named M773/M774, M775/M776, and M777/M778, respectively.

Promoter sequence for tre76230 promoter replacement: (SEQ ID NO :937) Promoter sequence for tre49048 promoter replacement: (SEQ ID NO :938)

Promoter sequence for tre58698 promoter replacement: (SEQ ID NO :939)

These transformants and the M507 control strain were cultivated in 24 well plates using TrMM with 1% yeast extract, 2% cellobiose, 1% sorbose, pH 5.5. Samples were taken on days 5, 6, and 7. Immunoblotting was done with AP conjugate antibodies to detect the heavy and light chain. The culture supernatants were diluted so that 0.5 μΐ was loaded per lane. Detection was visualized with AP substrate.

The heavy chain was clearly more stable in the M773/M774 and M777/M778 transformants (Figure 17). The most striking point is on day 7. The M507 heavy chain is almost fully degraded, while the M773/M774 heavy chain survived very well. The other remarkable feature was that there was no upper degradation product at 37 kD. There are only some lower degradation products around 28 kD. The M773/M774 strains grew well, but the M777/M778 strains were not growing that well and had trouble sporulating. Thus, the preferred strain would be M773 or M774. Total protease activity measurements with casein correlated and indicated that M773/M774 and M777/M778 have low protease activity.

When the light chains were detected via immunblotting there was less light chain in M773 and

M774 compared to control. There was particularly less light chain with Mill and M778 strains.

The M775 and M776 light chain amounts were similar to control. There seemed to be good agreement between the light chain amounts seen on the blots and the total immunoglobulin detected. The total IgG was measured from these cultures and showed that all of the promoter exchange strains had lower antibody expression, except for M775 and M776, which were very similar to control (Table 18.1). The M773/M774 strains had levels less than half M507. Thus, there might be something affecting the growth of some of the promoter replacement strains, in this small culture format.

Table 18.1. Total antibody concentrations from 24 well culture day 7.

The M774 and M775 strains were fermented. The two strains were cultivated in TrMM plus 20 g/1 yeast extract, 40 g/1 cellulose, 80 g/1 cellobiose, and 40 g/1 sorbose at pH 5.5 with the temperature shifting from 28° to 22° at 48 hours for 10 days. The control strain M667 was cultivated under the same conditions and under similar conditions where 120 g/1 cellulose was used. Samples were taken for immunoblotting and total immunoglobulin determination. The supernatants were diluted so that 0.05 μΐ was loaded per well in a 4-15% gel. Immunoblotting to detect the light chain was done with an AP conjugated antibody (A3818) diluted 1: 10,000 in TBST. The heavy chain was detected with an anti-human Fc antibody IRDye 700 DX conjugate (Rockland #609-130-003) diluted 1:30,000 in TBST. The fluorescence at 700 nm was detected using an Odyssey CLx near infrared imager (Li-Cor).

The MAB01 heavy chain produced from M774 looked remarkably good (Figure 18). The results were similar to those seen in the 24 well cultures. There was only one major degradation product around 25 kD. Typically, the heavy chain looks like that produced by M775. Normally there are degradation bands at 38 kD, 28 kD, and 10 kD. The disappearance of the 10 and 38 kD products may be explained by reduction of SLP2 activity because there are subtilisin cleavage site closer to the N terminus of the MAB01 heavy chain and without high slp2 expression the heavy chain is no longer cleaved at that position. The 28 kD product comes when proteases cleave the heavy chain in the hinge region. The cleavage sites in the hinge are more typical of sedolisin or trypsin like proteases. They don't appear to be caused by SLP2. The 10 kD product results when the hinge and the near N terminal sites are both cleaved.

No obvious differences were seen between the M774 and M775 light chain amounts or carrier bound percentages. While M775 is not optimal control strain, it resembles M507 very much, as seen in the 24 well culture data described above. This data also suggests that SLP2 does not appear to be cleaving the carrier-light chain fusion. Otherwise the amount of carrier bound material would increase dramatically when the slp2 expression level was reduced.

M774 strain slightly outperformed M775 when the total antibody amounts are compared (Table 18.2). The total antibody amounts are also listed on the heavy chain immunoblots. The biggest difference can be seen on day 7 and 8, where M774 produced 5.3 g/L on both days. The M775 produced 4.4 and 4.6 g/L on those days. The most important difference was observed once the amounts of full length antibody are measured and compared. The M774 produced a far more intact heavy chain. On day 7, the M774 strain produced 71 % full length, while the M775 was 37% full length. The M774 strain could maintain 65% full length antibody up to 10 days, whereas the M775 strain produced only 14% full length material. In terms of quality, the control strain M667 (with a slp2 gene silencing construct) was similar to M774 (Table 18.2). However, the M667 production strain produced slightly higher amounts of total antibody, at levels reaching 6.1 g/L on day 10 under the same culture conditions. With increased cellulose (120 g/L) the M667 strain could produce as high as 7.1 g/L of total antibody on day 10 (Table 18.2). From these data, another approach to reduce SLP2 activity was generated. Replacing SLP2 promoter led to less expression of the SLP2 protease and resulted in remarkably low heavy chain degradation. Some effects on growth and sporulation were still observed, but they were far milder than those seen with the complete deletion of the SLP2 gene. By giving slp2 a sporulation induced promoter it may have addressed this sporulation related problem. Any growth defect seen in M774 can be compensated with modifications in fermentation processes.

Table 18.2. Total antibody and full length antibody quantitation from fermentor cultivation supernatants of strains M774, M775, and M667 cultivations.

13 6435 - -

14 6175 - -

The M507 parental strain was compared to the M646 slp2 deletion strain and the M774 slp2 promoter switch strain in a separate fermentor cultivation series. They were grown in TrMM plus 20 g/1 yeast extract and 120 g/1 cellulose with 50% glucose/12.5% sorbose feed at pH 5.5 with the temperature shifting from 28° to 22° at 48 hours. Immunoblotting with anti-heavy and anti-light chain antibodies was done from the day 5 and day 6 samples to visualize the quality and relative amount of heavy and light chain produced.

The M507 started growing a faster than the M646 and M774 strains, but they caught up to M507 later in the culture. There was no significant difference in the amount of free light chain produced by these three strains, as seen on the immunoblot. There was a slightly higher amount of light chain bound to the CBHI carrier in the M646 strain. A small amount of carrier bound heavy chain was detected from the M646 samples and even less was seen in the M774 strain. This could not be detected in the M507 strain. This observations suggests that SLP2 may be involved in processing the CBHI carrier-antibody fusion proteins to some extent in the supernatant or potentially intracellularly.

The M774 strain showed a similar heavy chain degradation pattern to the slp2 deletion strain M646. Both strains were lacking the 10 and 38 kD degradation products that were seen in the M507 parental strain. In the immunoblot the M646 and M774 heavy chains showed only the full sized product at 50 kD and one major degradation product at 25 kD. The amount of full length heavy chain in the M646 and M774 strains was higher compared to M507. The control M507 heavy chain showed two additional degradation products at 10 and 38 kD. This shows that indeed SLP2 protease was responsible for producing these products. The amount of total MAB01 antibody produced on the day 6 time point was similar between the M774 strain and M507 strain, reaching 2.8 g/L under these culture conditions (Table 18.3). Even though the M774 and M507 strains produced the same amount of total immunoglobulin, the quality of the M774 material was much better considering the lack of heavy chain degradation products. Table 18.3. Fermentation data from cultivation of M507, M646, M774. The total antibody titers are shown in the table as g/L.

EXAMPLE 19 - 14-fold protease deletion strain - slp2 (trel23244) promoter switch to tre76230 promoter To generate a 14-fold protease deletion strain with reduced SLP2 activity, the 13-fold protease deletion strain M1077 (pyr4- of M1076) was transformed with Mssl fragment of a plasmid targeted to the slp2 (trel 23244) locus to replace slp2 promoter with sporulation induced gene tre76230 promoter.

Transformation was carried out using standard protoplast transformation method for pyr4 selection. Transformation was scaled to 2x (i.e. using 500 μΐ protoplasts). Total amount of DNA used was -14.6 μg. Colonies growing on transformation plates were picked on selective plates and screened for correct integration of the deletion cassette using primers shown in Table 19.1. Selected clones giving integration signals were purified via single spore purification and rescreened. Clones from three transformants seemed to be pure after one purification round with strong integration signals. Two pure sibling clones from each transformant were plated onto PD + 1 M sorbitol for spore suspensions.

Table 19.1. Primers used in screening correct integration of the deletion cassette to the genome and change of slp2 promoter (trel 23244) with tre76230 promoter.

Southern analyses of slp2 promoter replacement strains

Three biological clones with two replicates of the slp2 promoter replacement transformation along with their parental strain Ml 076 were cultivated for DNA extraction in 24-well plate. After three days mycelia were harvested by vacuum filtration, frozen and lyophilised. Genomic DNA was extracted from mycelia using Easy-DNA kit (Invitrogen) and samples were analysed by PCR using the primers in Table 19.1. None of the clones gave strong signal for slp2 promoter, but a very low amount of product corresponding to expected size was seen for a few clones.

Genomic DNA from M124, M1076 and six Aslp2 promoter replacement strains were digested with Hindlll for all analyses (Aslp2 promoter, 5' and 3 'flank). Strain Ml 24 was omitted from flank analyses. A control plasmid was digested with Mssl for both flank analyses. Primers used to generate probes for hybridisations are shown in Table 19.2.

Table 19.2. Primers used to produce probes for Southern analyses of slp2 (trel23244) promoter change with tre76230 clones.

Primer Sequence Size Target

T1393-slp2-5utr-F CACAACGTACTCGAAGTACC(SEQ 541 bp slp2 promoter ID NO :945)

T 1586_slp2_promch_r AGGTCACTCAGCCTTGTACC(SEQ

ID NO :946)

T 1764_slp2_5 fprobe_f or TCAGATGGAGTCCCTTGAAC 647 bp slp2 5' flank

(SEQ ID NO :947)

T1765_slp2_5fprobe_rev CTGAATCTTGCTGGTCCG(SEQ ID

NO :948)

T 1766_slp2_3 fprobe_f or CAGCACATTCCAGATTGGC (SEQ 853 bp slp2 3' flank

ID NO :949)

T 1767_slp2_3 fprobe_rev TGCTCAATGTGGGAGAGAGC(SEQ

ID NO :950)

The Southern analyses confirmed that all clones were pure slp2 promoter change clones. In addition, majority of the clones give only the expected signal with 5' and 3' flank probes verifying single integration of the deletion cassette to the genome. One clone (78-28B) may have an extra copy of the cassette integrated to the genome. Clone 78-6A has been stored for collection and designated with the code Ml 162. Clones with slp2 promoter change appear to have somewhat delayed and lower ability to sporulate.

Fermentor cultivation of Ml 162

The Ml 162 strain was cultivated and compared to the strain M1076. The preculture was grown for 3 days, instead of the normal 2 days. The Ml 162 was fermented in TrMM with 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, 40 g/L sorbose at pH 4.5. The Ml 162 strain grew a bit slower than its predecessor M1076. The C0 2 peak for M1076 came at 75 hours while the Ml 162 was at 94 hours.

Protease activity measurements The protease activity from the fermentor cultivation samples was analysed. The total protein concentrations from the cultivation samples were measured so that the samples could be adjusted to 1 mg/ml total protein. 100 μΐ of all the diluted supernatants was added into 96 well plate. Three replicate wells were used per sample. 100 μΐ of casein FL diluted stock (10 μg ml) made in sodium citrate buffer pH 4.5 was added to each well of supernatant. The casein stock solution from the vial was 1000 μg ml and initially resuspended in 200 μL· of PBS. For each sample a background control was used with 100 μΐ of diluted supernatant and 100 μΐ of sodium citrate buffer pH 4.5. The plates covered in plastic bags were incubated at 37°C. The fluorescence from the plates was measured after 2, 3, and 4 hours. The readings were done on a fluorescent plate reader using 485 nm excitation and 530 nm emission.

The protease activity of the supernatant coming from the Ml 162 strain was extremely low, compared to the M1076 strain. The SLP2 protease activity was seriously affected by switching the slp2 promoter. The resulting protease activity upon casein was 3.6 times lower on day 3 and 2.5 times lower on day 4 (Table 19.3).

Table 19.3. Protease activity measurements upon casein substrate for M1076 and Ml 162 fermentation supernatants. Samples were diluted so that 1 mg/ml of total protein was used per sample. Casein FL substrate was added to measure the protease activity.

EXAMPLE 20 - Generation of 13-fold protease deletion strain expressing IFN-a 2b The 13-fold deletion strain expressing interferon alpha 2b was created in two steps. First, an interferon producing strain from M893 was generated as described above, which contained 12 protease deletions A(pepl tspl slpl gapl gap2 pep4 pep3 pep5 pep2 sepl slp8 amp2). The M893 protoplasts were transformed with pTTv401 (derived from a plasmid having complete cbhl promoter and cbhl terminator sequences but lacking CBH1 encoding gene sequence inserted with IFN-a 2B, GGGGG-NVISKR) Mssl fragment. The pTTv401 cassette carried the acetamide selection marker and was targeted to cbhl locus. M893 transformants were streaked on selection plates and PCR screening for correct integration into cbhl locus. PCR screenings were done using Phire Plant Direct kit (Thermo Scientific, F- 130). Screening primers are listed on Table 43.1. A strain was named M1012.

Table 20.1. Screening primers for pTTv401 into M893 transformation.

Ml 012 was confirmed by Southern analysis to carry one IFN-a 2b expression cassette at cbhl locus. Ml 012 spores were plated on 5-FOA plates to loopout pyr4 marker. Colonies were picked and streaked on 5-FOA plates. Single cell plating was done for ten clones and subclones were screened by PCR to confirm pyr4 loopout. Screening primers are listed in Table 20.2. Clones which gave correct signals for pyr4 loopout were streaked on PD plates and +/- uridine plate test was done for selected clones. M1012 pyr4 negative clone 1-1 was named as M1065 and pyr4 negative clone 2-1 was named as M1066.

Table 20.2. Primers for screening pyr4 loopout from M1012

Primer pair 1:

T1556_amp2_5f_f2 AAGTGTGCTGATGTGATGGA (SEQ ID NO :956)

T1557_amp2_3f_r2 GCATGCGAAGTACCTTACGA (SEQ ID NO :957) Primer pair 2:

T061_pyr4_orf_screen_2F TTAGGCGACCTCTTTTTCCA (SEQ ID NO: 382)

T624_gpdA_seqRl CTCCATATTCTCCGATGATGC (SEQ ID NO: 852)

To generate 13-fold protease deletion strain with IFN-a 2b expression, Ml 065 protoplasts were transformed with Mssl fragment of slp2 (trel23244) deletion vector pTTv457 (generated from pTTvl l5 by introducing/changing pyr4-hygromycin selection marker). Protoplasting and transformation was carried out as above. pTTv457 Ml 065 transformants were streaked on selection plates and PCR screened for correct integration into slp2 locus. PCR Screening primers are listed on Table 20.3. Three transformants which did not give slp2 ORF signal were streaked on PD+1M sorbitol plates.

Table 20.3. Primers for screening slp2 deletion cassette integration and slp2 ORF deletion. 5' integration screening

T054_slp2_5screen_F GATGCACCGCTGCGGCC (SEQ ID NO: 327)

T026_Pyr4_orf_5rev2 CCATGAGCTTGAACAGGTAA (SEQ ID NO: 328)

3' integration screening

T047_trpC_term_end_F CCTATGAGTCGTTTACCCAGA (SEQ ID NO: 426)

T055_slp2_3screen_R GGCGTTGCTCCCCATGCG (SEQ ID NO: 330)

slp2 orf

T1231_slp2 seqF2 AACGGATCCGGCACCATGTC (SEQ ID NO :958)

Tl 12_slp2_ORF_R TTACTCGGAGAGCTCAGAGA (SEQ ID NO: 332)

The best clone was designated Ml 106 and cultivated with the control strain M961 in a 1 L fermentor in TrMM with 20 g/L yeast extract, 40 g/L cellulose, 100 g/L cellobiose, and 40 g/L sorbose at pH 4.5. The previous standard amount of cellobiose was 80 g/L, so the M961 strain was also cultivated in the control medium (TrMM with 20 g/L yeast extract, 40 g/L cellulose, 80 g/L cellobiose, and 40 g/L sorbose at pH 4.5).

The culture samples were analysed via immunoblotting to quantitate the expression of interferon. The samples were diluted so that 0.05 μΐ of supernatant could be loaded in 10 μΐ in a 4-20% criterion page gel. Standard amounts of interferon corresponding to 400, 200, 100, 50, and 25 ng were also loaded to the same gel. The interferon antibody (Abeam #ab9386; diluted to 1 μg/ml in TBST) was incubated with the blot membrane for 1 hour and washed with TBST. The secondary antibody IRDye 680 (Li-Cor #926-68070; diluted 1 :30,000 in TBST) was incubated for 1 hour, washed with TBST, and scanned at 700 nm. In all the immunoblots interferon was detected as one band about 17 kD. There was a small amount of carrier bound interferon detected about 75 kD, but the majority was in the free form. The M961 strain achieved an interferon production level of 3.2 g/L at 95 hours (Table 20.2). In the presence of higher concentration of cellobiose the interferon production level reached 7.9 g/L at 95 hours and the expression was stable from 89 hours through 99 hours where levels were 7.4 g/L or greater. To check what the maximum production level would be if more proteases were deleted, the M961 strain was grown in the same medium with chymostatin (20 μΜ) and pepstatin (10 μΜ) added on days 3-5. The protease inhibitor treatment raised the interferon production level to 10.7 g/L at 140 hours. During Ml 106 cultivation the expression of interferon was seen from 90 hours up to 121 hours. The highest amount was measured to be 4.3 g/L (Table 20.4). The strain Ml 106 grew somewhat slower: the Ml 106 reached 4.3 g/L at 121 hours, whereas the M961 reached 7.4 g/L at 90 hours.

Table 20.4. Expression level of interferon alpha 2b detected in the fermentor cultivations in g/L.

EXAMPLE 21 - Using CRISPR-CAS system to generate gene deficient strains of T. reesei

Cas9 nuclease sequence with C-terminally tagged nuclear localization signal (nls) is codon optimized for expression in Trichoderma reesei. Sequence is cloned under the control of constitutive gpdA promoter and trpC terminator sequences, using basic cloning vector and standard procedures. Final Cas9 nuclease expression vector is constructed from following components: pep4 protease (or any other suitable protease) locus 5' flanking sequence + pgpdA- Cas9-nls-ttrpC cassette + pyr4-hyg R double selection cassette and pyr4 loop-out sequence + pep4 protease locus 3 ' flanking sequence. Vector is constructed to pRS426 backbone by utilizing yeast recombination methodology; overlaps between the vector components are generated with PCR primers. Cas9 nuclease expression vector is transformed with peg-mediated protoplast transformation method to wild-type T. reesei Ml 24 strain or any other T. reesei strain generated above or in WO/2013/174927 or WO/2013/102674, generating simultaneously pep4 protease deletion. Generated strain Cas9_M124 is then used as a background strain for transfection of transient gRNA cassettes generated by PCR, as described in DiCarlo et al. 2013 (Genome engineering in Saccharomyces cerevisiae using CRISPR-Cas systems; NAR 41:4336-4343). Alternatively, RNA polymerase III SNR52 promoter- and SUP4 3' flanking region from Saccharomyces are replaced with Trichoderma homologues. Guide RNA needed for precise genomic targeting of CAS9 nuclease is located between the promoter and 3' flanking region. Guide RNA is composed of 20 nt's long sequence complementary to desired genomic target, followed by 3 nt's complementary with NGG PAM (protospacer-adjacent motif) -sequence and constant 3' portion required for CAS9 activity. Exemplary guide RNAs are shown in Table 21.1 for various proteases and glycoenzymes harmful for heterologous protein production. The genomic targets are selected among hydrolytic enzymes or enzymes from glycan biosynthesis pathway of Trichoderma reesei. Transient guide RNA cassettes (single and multiple) are introduced to Cas9_M124 protoplasts by electroporation or by other basic gene transfer method. Protease deficient clones are selected on the basis of reduced protease activity, caused by CAS9- generated point mutations to desired genomic target sequences. Clones with point mutations targeted to glycan biosynthesis pathway can be selected by glycan profiling. After single spore purification, selected clones are characterized by PCR amplification of genomic target locus and sequencing of the PCR product, to verify the point mutation inactivating the gene.

Alternative way to produce guide RNA is to express the sequence or multiple sequences from promoter transcribed by RNA polymerase II and flank the guide RNA's with self -processing ribozyme sequences, as described in Gao and Zhao 2014 (Self-processing of ribozyme flanked RNA's into guide RNA's in vitro and in vivo for CRISPR mediated genome editing; Journal of Integrative plant biology, 56:343-349). Figure 19 show a phylogenetic tree of a subset of the proteases amenable to deletions.

Table 21.1. Guide RNA sequences targeted to T. reesei proteases and ALG3.

amp2 108592 CCTCGACTCGCGCTTCGTCA (SEQ ID NO :985) sepl 124051 GCAGCCAGCACTCCCACCTA (SEQ ID NO :986) slp7 123865 TCTCCGACCCCTCAAGCCCA (SEQ ID NO :987) tppl/sed3 82623 GCAGTTCTGCCGTCGAGTCT (SEQ ID NO :988) sed2 70962 GAGATACCAGCAACGCGCGA (SEQ ID NO :989) sed5 111838 GATCCTTCATCAGAAACATT (SEQ ID NO :990) sed3 81517 GCAGCCATATATCGACAGCC (SEQ ID NO : 991) mpl 122703 CAGACGACGACGCTCAAGAA (SEQ ID NO :992) mp2 122576 GACGCTGCCTCATCTAGTCG (SEQ ID NO :993) mp3 4308 GCTGCGCGATCTCGACTTCA (SEQ ID NO :994) mp4 53343 CTCACATTCTCTATTCACGA (SEQ ID NO :995) mp5 73809 TGTGCTCCTGACCGACAAGC (SEQ ID NO :996)

ALG3 104121 ACTGCCGTGGACATTGCCAA (SEQ ID NO :997) a protease 80843 CAGTCACCAGCAAGACAAAG (SEQ ID NO :998) a protease 72612 CGACCTCCACGATGTCATCA (SEQ ID NO :999) a protease 47127 AAGACGAAGCTCCGCCAATC (SEQ ID NO : 1000) a protease 77577 AAGAGCACGACCGTTTCGTC (SEQ ID NO : 1001) pep 13 76887 TCTGACGCTGCTCCTCGCGA (SEQ ID NO :1002) a protease 56920 ATCACCGACACGCGAGACCT (SEQ ID NO : 1003) a protease 120998 GTCGCTGGCCTCGTCCCTCA (SEQ ID NO : 1004) a protease 65735 GAGCTCGTCCGACCCCCGCC (SEQ ID NO : 1005) a protease 82141 CCCTGTCCCTGACCTTACAA (SEQ ID NO : 1006) a protease 121890 GAAATCACAACACTGCCAAA (SEQ ID NO :1007) a protease 22718 GAACTCAACCTCCAAGACGC (SEQ ID NO : 1008) a protease 21659 GAGTATGTTGCCATGTTCCT (SEQ ID NO : 1009) a protease 82452 CACGGAGACTGCTGCCGCTC (SEQ ID NO : 1010) a protease 81115 CTACTTCACCTACGACATCC (SEQ ID NO : 1011) a protease 64193 CATCCTCACCATCCTCACCA (SEQ ID NO : 1012) a protease 23475 GCTCTCACGAAATCCTCGAC (SEQ ID NO : 1013) a protease 79485 GCTCTCTGAGCCTGCAAGAC (SEQ ID NO : 1014) a protease 122083 CCGCGTCTCCTGCACGTAGT (SEQ ID NO :1015) a protease 61127 TGCGCGACCCCGTCATCGTC (SEQ ID NO : 1016) a protease 80762 ACCTCTCTGGTCCACGACCT (SEQ ID NO :1017) a protease 56853 GACTCCTCCCTCCACACCGT (SEQ ID NO : 1018) a protease 22210 TCTGTCGAGGAGAGCAACAT (SEQ ID NO : 1019) a protease 111694 ACGCGCAACAACCGCCGCAC (SEQ ID NO : 1020) a protease 40199 GCCCGACCGGTTCAACGTCC (SEQ ID NO : 1021) a protease 75159 TTCGACAAGCTCACTTACAA (SEQ ID NO : 1022) a protease 21668 CTTCGACTCCCACTCCAAGA (SEQ ID NO :1023) a protease 61912 CGCCGCTGCCCTCTTCGAAA (SEQ ID NO :1024) a protease 58387 CGTCACAGAGCACTTCTTCC (SEQ ID NO :1025) a protease 82577 CAGCACGACTCCATCTACGC (SEQ ID NO : 1026) a protease 81087 AACCACATCGCCGAGAACAA (SEQ ID NO : 1027) pep 10 78639 CCTTGTCTATGCGAATGACC (SEQ ID NO :1028) pep 16 110490 AGCAGCAGCAGCACGAGCAG (SEQ ID NO :1029) pep 14 108686 TCCACGTTTGAGCTGCGTGT (SEQ ID NO : 1030) pep6 68662 ATCCCCATCCACCAGAAGCG (SEQ ID NO : 1031) a protease 66608 CGTCTTCGACCGAATACAAG (SEQ ID NO : 1032)

EXAMPLE 22 - Transcriptome analysis with Trichoderma reesei strains M629 and M507

Trichoderma reesei strains M629 (MAB01, pcDNAl-(Kre2)huGntl, pgpdA-(nat)huGnt2, Apepl tspl slpl gapl gap2 pep4 pep3) (as described in Example 19 of WO2013/102674) and M507 (MAB01, Apepl tspl slpl gapl gap2 pep4 pep3) (as described in Example 6 of WO2013/102674) were cultivated in fermentor with strandard Yeast extract- and Spent grain extract culture medias. Total RNA was purified with standard methods, from samples collected on days 1, 3 and 4 (Yeast exract media) and on days 1, 3 and 5 (Spent grain extract media). mRNA was purified from total RNA samples with Machery-Nagel nucleotrap mRNA kit according to Kits instructions. Conversion to cDNA and preparing for sequencing was made with IWuminaTruSeq Stranded mRNA Sample Prep Kit. 250-450 base pare products were collected for sequencing, with Illumina hiScanSQ sequencer (100+8(index)+100 cycles, paired end run).

For statistical analysis the sequence reads were manipulated as following. The 9134 gene reads originating from the sequencing were cleaned (i.e. genes with all values with zero or average all over conditions below 0.1 were removed) and this resulted 8525 gene reads. Of the 8525 genes potential protease genes were identified based on sequence similarity to other identified Trichoderma proteases or the ones of other filamentous fungi (Aspergillus, Neurospora). The following proteases either show constant or regulated expression levels in different time points and/or culture conditions (based on FPKM values; fragments per kilobase of ex on per million fragments mapped) and should therefore be deleted: a metalloprotease mpl (TR122703), a protease (TR80843), a peptidase (TR72612), a protease (TR47127), a peptidase (TR77577), pepl3 (TR76887), a protease (TR56920), a carboxypeptidase (TR120998), a protease (TR65735), a peptidase (TR82141), a metalloprotease (TR121890), a peptidase (TR22718), a peptidase (TR21659), mp5 (TR73809), a protease (TR82452), a peptidase (TR81115), a peptidase (TR64193), a protease (TR23475), a peptidase (TR79485), mp3 (TR4308), a protease (TR122083), a carboxy peptidase (TR61127), a peptidase (TR80762), a peptidase (TR56853), a peptidase (TR22210), a protease (TRl 11694), mp4 (TR53343), mp2 (TR122576), a protease (TR40199), a protease (TR75159), amp2 (TR108592), a protease (TR21668), ampl (TR81070), a protease (TR61912), a protease (TR58387), a protease (TR82577), a protease (TR81087), peplO (TR78639), pepl6 (TRl 10490), pep7 (TR58669), pepl4 (TR108686), pep6 (TR68662) and a protease (TR66608).

EXAMPLE 23 - Generation of mpl or mp5 deletion strains

The deletion plasmid pTTv468 for the metalloprotease mpl (tre 122703) was constructed using yeast homologous recombination. The plasmid has mpl 3' direct repeat for looping out the pyr4 marker. A Notl digested plasmid (from a plasmid with hygr-pyr4 double marker derived from plasmid pTTvl94) was used as backbone, mpl 3' direct repeat and pyr4 marker were constructed by PCR using oligos listed in Table 23.1. pTTv468 vector was assembled using yeast homologous recombination cloning as described above.

Table 23.1. Primers used for cloning deletion vector pTTv468 for mpl (trel 22703).

To generate a 15 -fold protease deletion strain, the 14-fold protease deletion strain Ml 199 (pyr4- of Ml 162) was transformed with MssI fragment of pTTv468. Transformation was carried out using standard protoplast transformation method for pyr4 selection. Colonies growing on transformation plates were being picked on selective plates and screened for correct integration of the deletion cassette using primers shown in Table 23.2.

Table 23.2. Primers used for screening.

The deletion plasmid pTTv469 for the metalloprotease mp5 (tre73809) was constructed using yeast homologous recombination. The plasmid has mp5 3' direct repeat for looping out the pyr4 marker. mp5 3' direct repeat and pyr4 marker were constructed by PCR using primers listed on Table 23.3.

Table 23.3. Primers used for cloning deletion vector pTTv469 for mp5 (tre73809).

To generate another 15-fold protease deletion strain, the 14-fold protease deletion strain Ml 199 (pyr4- of Ml 162) was transformed with MssI fragment of pTTv469. Transformation was carried out using standard protoplast transformation method for pyr4 selection. Colonies growing on transformation plates were being picked on selective plates and screened for correct integration of the deletion cassette using primers shown in Table 23.4.

Table 23.4. Primers used for cloning deletion vector pTTv469.

EXAMPLE 47 - generation of 13-fold protease deletion strain with slp3 deletion

To generate a 13-fold protease deletion strain with Aslp3, the 12-fold protease deletion strain M901 (pyr4- of M893) was transformed with MssI fragment of pTTv425. Transformation and screening of the clones were performed as described above. One original transformant was obtained and after three purification rounds clones were rechecked by PCR (see Table 6-1). A subclone was named as Ml 075.