Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF TYPE 1 DIABETES
Document Type and Number:
WIPO Patent Application WO/2017/122180
Kind Code:
A1
Abstract:
Provided herein are compositions and methods for the treatment of type 1 diabetes (T1D) in mammalian subjects. The compositions include lactic acid fermenting bacteria (LAB) expressing an IL-2 gene and a TID-specific self-antigen (e.g., proinsulin (PINS)) gene. Exemplary methods include: orally administering to a mammalian subject, a therapeutically effective amount of the composition. The composition can be administered to the subject mucosally, resulting in delivery of the LAB into the gastrointestinal tract, where the LAB is released. Bioactive polypeptides expressed by the LAB are thus administered via mucosal delivery. The LAB may be selected to deliver a low-dose of IL-2 to the subject. The methods may not require concomitant systemic anti-CD3 antibody treatment. The methods may be suited for subjects possessing residual beta-cell function, e.g., those with recent-onset T1D.

Inventors:
ROTTIERS PIETER (BE)
STEIDLER LOTHAR (BE)
Application Number:
PCT/IB2017/050204
Publication Date:
July 20, 2017
Filing Date:
January 13, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INTREXON ACTOBIOTICS N V (BE)
International Classes:
A61K35/744; A61K38/20; A61K39/00; A61P3/10; A61P37/06; C12N1/00; C12N1/20; C12N15/09; C12R1/01
Domestic Patent References:
WO2013036914A12013-03-14
WO2002090551A22002-11-14
WO1996032487A11996-10-17
WO1993017117A11993-09-02
WO2005086751A22005-09-22
WO2000018377A12000-04-06
WO2000022909A22000-04-27
Foreign References:
US20140004080A12014-01-02
US20140105863A12014-04-17
GB2278358B1995-07-26
US8759088B22014-06-24
US4518584A1985-05-21
US81065685A1985-12-17
EP0091539A11983-10-19
EP0088195A21983-09-14
US20120244112A12012-09-27
US7569215B22009-08-04
US5229109A1993-07-20
US20060269515A12006-11-30
EP1730184A22006-12-13
US5972685A1999-10-26
US20120039853A12012-02-16
US6387352B12002-05-14
US6348187B12002-02-19
US6171611B12001-01-09
US6165494A2000-12-26
US6117417A2000-09-12
US5993785A1999-11-30
US5695746A1997-12-09
US5470561A1995-11-28
US4919918A1990-04-24
US20040076590A12004-04-22
US20030152530A12003-08-14
US20020044910A12002-04-18
US5700782A1997-12-23
US5869118A1999-02-09
US5223285A1993-06-29
Other References:
TATIANA TAKIISHI ET AL: "Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice", JOURNAL OF CLINICAL INVESTIGATION, vol. 122, no. 5, 1 May 2012 (2012-05-01), US, pages 1717 - 1725, XP055308282, ISSN: 0021-9738, DOI: 10.1172/JCI60530
S. ROBERT ET AL: "Oral Delivery of Glutamic Acid Decarboxylase (GAD)-65 and IL10 by Lactococcus lactis Reverses Diabetes in Recent-Onset NOD Mice", DIABETES, vol. 63, no. 8, 27 March 2014 (2014-03-27), US, pages 2876 - 2887, XP055351010, ISSN: 0012-1797, DOI: 10.2337/db13-1236
R. MALLONE ET AL: "Of Bugs and Men: Antigen-Fortified Lactoccoccus lactis for Type 1 Diabetes Immunotherapy", DIABETES, vol. 63, no. 8, 24 July 2014 (2014-07-24), US, pages 2603 - 2605, XP055350900, ISSN: 0012-1797, DOI: 10.2337/db14-0587
AIXIN YU ET AL: "Selective IL-2 Responsiveness of Regulatory T Cells Through Multiple Intrinsic Mechanisms Supports the Use of Low-Dose IL-2 Therapy in Type 1 Diabetes", DIABETES, vol. 64, no. 6, 9 January 2015 (2015-01-09), US, pages 2172 - 2183, XP055350944, ISSN: 0012-1797, DOI: 10.2337/db14-1322
ROSENZWAJG MICHELLE ET AL: "Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients", JOURNAL OF AUTOIMMUNITY, vol. 58, 26 January 2015 (2015-01-26), pages 48 - 58, XP029204247, ISSN: 0896-8411, DOI: 10.1016/J.JAUT.2015.01.001
VAN BELLE, T.L. ET AL., PHYSIOL. REV., vol. 91, no. 1, 2011, pages 79 - 118
YU, A. ET AL., DIABETES, vol. 64, 2015, pages 2172 - 2183
HARTEMANN A ET AL., LANCET DIABETES ENDOCRINOL., vol. 1, 2013, pages 295 - 305
ROSENZWAJG M ET AL., JAUTOIMMUN., vol. 58, 2015, pages 48 - 58
STEIDLER, L. ET AL., NAT. BIOTECHNOL., vol. 21, no. 7, 2003, pages 785 - 789
ROBERT S.; STEIDLER L., MICROB. CELL FACT., vol. 13, no. 1, 2014, pages L 1
ROBERT, S. ET AL., DIABETES, vol. 63, 2014, pages 2876 - 2887
TAKIISHI, T ET AL., J. CLIN. INV., vol. 122, no. 5, 2012, pages 1717 - 1725
STEIDLER ET AL., NAT. BIOTECHNOL., vol. 21, no. 7, 2003, pages 785 - 789
JONES A.G.; HATTERSLEY A.T., DIABETIC MEDICINE, vol. 30, 2013, pages 803 - 817
LITTLE RR ET AL., CLIN. CHEM., vol. 54, 2008, pages 1023 - 1026
WIEDMEYER ET AL., CLIN. CHEM., vol. 53, 2007, pages 784 - 787
DEMEESTER ET AL., DIABETES CARE, vol. 38, no. 4, 2015, pages 644 - 651
STROBEL ET AL., IMMUNOLOGY, vol. 49, 1983, pages 451 - 456
MAYER; SHAO, NATURE REV. IMMUNOL., vol. 4, 2004, pages 407 - 419
GASSON, M.J., J. BACTERID., vol. 154, 1983, pages 1 - 9
SORENSEN ET AL., APPL. ENVIRON. MICROBIOL., vol. 66, 2000, pages 1253 - 1258
SAMBROOK ET AL.: "Molecular Cloning: a Laboratory Manual, 2nd ed.", 1989, COLD SPRING HARBOR LABORATORY PRESS
AUSUBEL ET AL.: "Short Protocols in Molecular Biology, 2nd ed.", 1992, JOHN WILEY & SONS
WATERFIELD NR ET AL., GENE, vol. 165, no. 1, 1995, pages 9 - 15
RAPOPORT, CURRENT OPINION IN BIOTECHNOLOGY, vol. 1, 1990, pages 21 - 27
HARWOOD; CUTTING: "Molecular Biological Methods for Bacillus", 1990, JOHN WILEY & CO., pages: 341 - 344
VAN ASSELDONK ET AL., MOL. GEN. GENET., vol. 240, 1993, pages 428 - 434
DAYHOFF: "The Atlas of Protein Sequence and Structure", vol. 5, 1978
ARGOS, EMBO J., vol. 8, 1989, pages 779 - 785
TANIGUCHI ET AL., NATURE, vol. 302, no. 5906, 1983, pages 305 - 10
DEVOS ET AL., NUCLEIC ACIDS RES., vol. 11, no. 13, 1983, pages 4307 - 23
STEIDLER ET AL., SCIENCE, vol. 289, no. 5483, 2000, pages 1352 - 1355
HARTEMANN ET AL., LANCET DIABETES ENDOCRINOL., vol. 1, no. 4, 2013, pages 295 - 305
ROBERT ET AL., BENEF. MICROBES, vol. 6, no. 4, 2015, pages 591 - 601
ARDEN ET AL., DIABETES, vol. 48, no. 3, 1999, pages 531 - 542
MARTIN ET AL., J. BIOL. CHEM., vol. 276, no. 27, 2001, pages 25197 - 207
DOGRA ET AL., DIABETOLOGIA, vol. 49, no. 5, 2006, pages 953 - 7
HANSEL ET AL.: "PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th ed.", 1990, WILLIAM AND WILKINS
CHIEN: "NOVEL DRUG DELIVERY SYSTEM, 2nd ed.", 1992, M. DEKKER
PRESCOTT ET AL.: "NOVEL DRUG DELIVERY", 1989, J. WILEY & SONS
CAZZANIGA ET AL., INT. J. PHARM., vol. 108, no. 1, 1994, pages 77 - 83
PRESCOTT ET AL.: "NOVEL DRUG DELIVERY", 1989, J.WILEY & SONS
BRUSCHI M. L.; DE FREITAS O., DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, vol. 31, 2005, pages 293 - 310
BATCHELOR ET AL., INT. J. PHARM., vol. 238, 2002, pages 123 - 32
GASSON MJ, J. BACTERIOL., vol. 154, no. 1, 1983, pages 1 - 9
STEIDLER L. ET AL., NAT. BIOTECHNOL., vol. 21, no. 7, 2003, pages 785 - 789
LAW J ET AL., J. BACTERIOL., vol. 177, no. 24, 1995, pages 7011 - 7018
SANDERS ET AL., J. BACTERIOL., vol. 177, no. 18, 1995, pages 5254 - 5260
TAKIISHI, T. ET AL., J. CLIN. INV., vol. 122, no. 5, 2012, pages 1717 - 1725
SCHOTTE ET AL., ENZYME MICROB. TECHNOL., vol. 27, no. 10, 2000, pages 761 - 765
DROUAULT S ET AL., APPL. ENVIRON. MICROBIOL., vol. 65, no. 11, 1999, pages 4881 - 6
GAGLIANI, N. ET AL., NAT. MED., vol. 19, no. 6, 2013, pages 739 - 746
ANTONIOLI, L. ET AL., TRENDS MOL. MED., vol. 19, no. 6, 2013, pages 355 - 367
TAKIISHI, T. ET AL., J CLIN.INVEST., vol. 122, no. 5, 2012, pages 1717 - 1725
SUAREZ-PINZON WL ET AL.: "Combination therapy with glucagon-like peptide-1 and gastrin restores normoglycemia in diabetic NOD mice", DIABETES, vol. 57, 2008, pages 3281 - 8
TANG Q; BLUESTONE JA, NAT. IMMUNOL., vol. 9, no. 3, 2008, pages 239 - 244
ZHENG Y; RUDENSKY AY., NAT. IMMUNOL., vol. 8, no. 5, 2007, pages 457 - 462
GRINBERG-BLEYER Y. ET AL., J. EXP. MED., vol. 207, no. 9, 2010, pages 1871 - 1878
Download PDF:
Claims:
What is claimed is:

Claim 1. A composition comprising:

(a) a LAB comprising an exogenous nucleic acid encoding an interleukin-2 (IL-2) polypeptide and an exogenous nucleic acid encoding a type-1 diabetes mellitus (T1D)- specific antigen polypeptide, or

(b) a first LAB comprising an exogenous nucleic acid encoding an interleukin-2 (IL- 2) polypeptide, and a second LAB comprising an exogenous nucleic acid encoding a type-1 diabetes mellitus (TlD)-specific antigen polypeptide.

Claim 2. The composition of claim 1 , wherein said LAB is selected from the group consisting of: a Lactococcus species, a Lactobacillus species, a Bifidobacterium species, a Streptococcus species, and an Enterococcus species.

Claim 3. The composition of claim 2, wherein said LAB is selected from the group consisting of: Lactococcus garvieae, Lactococcus lactis, Lactococcus lactis subsp. cremoris, Lactococcus lactis subsp. hordniae, Lactococcus lactis subsp. Lactis, Lactococcus piscium, Lactococcus plantarum, Lactococcus raffinolactis, Lactobacillus acetotolerans, Lactobacillus acidophilus, Lactobacillus agilis, Lactobacillus algidus, Lactobacillus alimentarius, Lactobacillus amylolyticus, Lactobacillus amylophilus, Lactobacillus amylovorus, Lactobacillus animalis, Lactobacillus aviariits, Lactobacillus aviarius subsp. araffinosus, Lactobacillus aviarius subsp. aviarius, Lactobacillus bavaricus, Lactobacillus bifermentans, Lactobacillus brevis, Lactobacillus buchneri, Lactobacillus bulgaricus, Lactobacillus carnis, Lactobacillus casei, Lactobacillus casei subsp. alactosus, Lactobacillus casei subsp. casei, Lactobacillus casei subsp. pseudoplantarum, Lactobacillus casei subsp. rhamnosus, Lactobacillus casei subsp. tolerans, Lactobacillus catenaformis, Lactobacillus cellobiosus, Lactobacillus collinoides, Lactobacillus confusus, Lactobacillus coryniformis, Lactobacillus coryniformis subsp. coryniformis, Lactobacillus coryniformis subsp. torquens, Lactobacillus crispatus, Lactobacillus curvatus, Lactobacillus curvatus subsp. curvatus, Lactobacillus curvatus subsp. melibiosus, Lactobacillus delbrueckii, Lactobacillus delbrueckii subsp. bulgaricus, Lactobacillus delbrueckii subsp. delbrueckii, Lactobacillus delbrueckii subsp. lactis, Lactobacillus divergens, Lactobacillus farciminis, Lactobacillus fermentum, Lactobacillus fornicalis, Lactobacillus fructivorans, Lactobacillus fructosus, Lactobacillus gallinarum, Lactobacillus gasseri, Lactobacillus graminis, Lactobacillus halotolerans, Lactobacillus hamsteri, Lactobacillus helveticus, Lactobacillus heterohiochii, Lactobacillus hilgardii, Lactobacillus homohiochii, Lactobacillus iners, Lactobacillus intestinalis, Lactobacillus jensenii, Lactobacillus johnsonii, Lactobacillus kandleri, Lactobacillus kefiri, Lactobacillus kefiranofaciens, Lactobacillus kefirgranum, Lactobacillus kunkeei, Lactobacillus lactis, Lactobacillus leichmannii, Lactobacillus lindneri,

Lactobacillus malefermentans, Lactobacillus mali, Lactobacillus maltaromicus,

Lactobacillus manihotivorans, Lactobacillus minor, Lactobacillus minutus, Lactobacillus mucosae, Lactobacillus murinus, Lactobacillus nagelii, Lactobacillus oris, Lactobacillus panis, Lactobacillus parabuchneri, Lactobacillus paracasei, Lactobacillus paracasei subsp. paracasei, Lactobacillus paracasei subsp. tolerans, Lactobacillus parakefiri, Lactobacillus paralimentarius, Lactobacillus paraplantarum, Lactobacillus pentosus, Lactobacillus perolens, Lactobacillus piscicola, Lactobacillus plantarum, Lactobacillus pontis,

Lactobacillus reuteri, Lactobacillus rhamnosus, Lactobacillus rimae, Lactobacillus rogosae, Lactobacillus ruminis, Lactobacillus sakei, Lactobacillus sakei subsp. camosus,

Lactobacillus sakei subsp. sakei, Lactobacillus salivarius, Lactobacillus salivarius subsp. salicinius, Lactobacillus salivarius subsp. salivarius, Lactobacillus sanfranciscensis, Lactobacillus sharpeae, Lactobacillus suebicus, Lactobacillus trichodes, Lactobacillus uli, Lactobacillus vaccinostercus, Lactobacillus vaginalis, Lactobacillus viridescens,

Lactobacillus vitulinus, Lactobacillus xylosus, Lactobacillus yamanashiensis, Lactobacillus yamanashiensis subsp. mali, Lactobacillus yamanashiensis subsp. Yamanashiensis, Lactobacillus zeae, Bifidobacterium adolescentis, Bifidobacterium angidatum,

Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium catenulatum,

Bifidobacterium longum, Bifidobacterium infantis, Enterococcus alcedinis, Enterococcus aquimarinus, Enterococcus asini, Enterococcus avium, Enterococcus caccae, Enterococcus camelliae, Enterococcus canintestini, Enterococcus canis, Enterococcus casseliflavus, Enterococcus cecorum, Enterococcus columbae, Enterococcus devriesei, Enterococcus diestrammenae, Enterococcus dispar, Enterococcus durans, Enterococcus eurekensis, Enterococcus faecalis, Enterococcus faecium, Enterococcus gallinarum, Enterococcus gilvus, Enterococcus haemoperoxidus, Enterococcus hermanniensis, Enterococcus hirae, Enterococcus italicus, Enterococcus lactis, Enterococcus lemanii, Enterococcus malodoratus, Enterococcus moraviensis, Enterococcus mundtii, Enterococcus olivae, Enterococcus pollens, Enterococcus phoeniculicola, Enterococcus plantarum, Enterococcus pseudoavium, Enterococcus quebecensis, Enterococcus raffinosus, Enterococcus ratti, Enterococcus rivorum, Enterococcus rotai, Enterococcus saccharolyticus, Enterococcus silesiacus, Enterococcus solitarius, Enterococcus sulfureus, Enterococcus termitis,

Enterococcus thailandicus, Enterococcus iireasiticus, Enterococcus ureilyticus, Enterococcus viikkiensis, Enterococcus villorum, Enterococcus xiangfangensis, Streptococcus agalactiae, Streptococcus anginosus, Streptococcus bovis, Streptococcus canis, Streptococcus constellatus, Streptococcus dysgalactiae, Streptococcus equinus, Streptococcus iniae, Streptococcus intermedius, Streptococcus milleri, Streptococcus mitis, Streptococcus mutans, Streptococcus oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus pyogenes, Streptococcus ratti, Streptococcus salivarius, Streptococcus tigurinus, Streptococcus thermophilus, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus suis, Streptococcus uberis, Streptococcus vestibularis, Streptococcus viridans, and Streptococcus zooepidemicus.

Claim 4. The composition of claim 2, wherein said LAB is a Lactococcus species.

Claim 5. The composition of claim 4, wherein said LAB is Lactococcus lactis.

Claim 6. The composition of any one of claims 1 to 5, wherein said composition further comprises a pharmaceutically acceptable carrier.

Claim 7. The composition of any one of claims 1 to 6, wherein said T1D- specific antigen is selected from the group consisting of: proinsulin (PINS), glutamic acid decarboxylase (GAD65), insulinoma-associated protein 2 (IA-2), islet-specific glucose-6- phosphatase catalytic subunit-related protein (IGRP), zinc transporter 8 (ZnT8),

chromogranin A, (prepro) islet amyloid polypeptide (ppIAPP), peripherin, and citrullinated glucose-regulated protein (GRP).

Claim 8. The composition of claim 7, wherein said TID-specific antigen is

PINS.

Claim 9. The composition of any one of claims 1 to 8, wherein a LAB comprises said exogenous nucleic acid encoding an IL-2 polypeptide and said exogenous nucleic acid encoding a T ID-specific antigen polypeptide. Claim 10. The composition of any one of claims 1 to 8, wherein a first LAB comprises said exogenous nucleic acid encoding an IL-2 polypeptide, and a second LAB comprises said exogenous nucleic acid encoding a TID-specific antigen polypeptide.

Claim 11. The composition of any one of claims 1 to 10, wherein said exogenous nucleic acid encoding an IL-2 polypeptide is integrated into the chromosome of said LAB.

Claim 12. The composition of any one of claims 1 to 11, wherein said exogenous nucleic acid encoding a T ID-specific antigen polypeptide is integrated into the chromosome of said LAB, or is present on a plasmid contained in said LAB.

Claim 13. The composition of any one of claims 1 to 11, wherein said exogenous nucleic acid encoding an IL-2 polypeptide and said exogenous nucleic acid encoding a TID- specific antigen polypeptide are both integrated into the chromosome of said LAB.

Claim 14. The composition of claim 9 or 13, wherein said exogenous nucleic acid encoding an IL-2 polypeptide and said exogenous nucleic acid encoding a TID-specific antigen polypeptide are part of a polycistronic expression unit, driven by the same promoter.

Claim 15. The composition of any one of claims 1 to 14, wherein said IL-2 is a membrane bound form of IL-2 or a soluble form of IL-2.

Claim 16. The composition of any one of claims 1 to 15, wherein said exogenous nucleic acid encoding an IL-2 polypeptide encodes an IL-2 variant polypeptide.

Claim 17. The composition of claim 16, wherein said IL-2 variant polypeptide has a diminished IL-2 activity or an enhanced IL-2 activity, when compared to a

corresponding wild-type IL-2 polypeptide.

Claim 18. The composition of claim 16, wherein said IL-2 variant polypeptide is selected from the group consisting of: aldesleukin, teceleukin, and bioleukin. Claim 19. The composition of claim 16, wherein said IL-2 variant polypeptide comprises:

(a) a first amino acid substitution relative to mature, wild-type IL-2, selected from the group consisting of L72G, L72A, L72S, L72T, L72Q, L72E, L72N, L72D, L72R, and L72K; or

(b) a second amino acid substitution relative to mature, wild-type IL-2, selected from the group consisting of F42A, F42G, F42S, F42T, F42Q, F42E, F42N, F42D, F42R, and F42K; or

(c) a third amino acid substitution relative to mature, wild-type IL-2, selected from the group consisting of Y45A, Y45G, Y45S, Y45T, Y45Q, Y45E, Y45N, Y45D, Y45R, and Y45K; or

(d) a combination thereof.

Claim 20. The composition of any one of claims 1 to 19, wherein said LAB is adapted for mucosal delivery of low-dose IL-2 when administered to a mammalian subject.

Claim 21. A composition comprising a Lactococcus lactis, wherein said

Lactococcus lactis comprises an exogenous nucleic acid encoding an IL-2 polypeptide and an exogenous nucleic acid encoding PINS, and wherein said Lactococcus lactis is adapted for mucosal delivery of low-dose IL-2 when administered to a mammalian subject.

Claim 22. Use of the composition of any one of claims 1 to 21 for the treatment of T1D in a mammalian subject in need thereof.

Claim 23. A method of treating type 1 diabetes mellitus (T1D) comprising: administering to a mammalian subject in need thereof a therapeutically effective amount of the composition of any one of claims 1 to 21.

Claim 24. The method of claim 23, wherein no anti-CD3 antibody is administered to said subject.

Claim 25. The method of claim 23, further comprising administering an anti-CD3 antibody to said subject. Claim 26. The method of claim 25, wherein said anti-CD3 antibody is administered in a low-dose simultaneously with said composition to said subject.

Claim 27. The method of claim 25 or 26, wherein said anti-CD3 antibody is administered intravenously to said subject.

Claim 28. The method of any one of claim 23 to 27, wherein said low-dose IL-2 delivery is the range of from about 0.01 M IU/day/subject to about 5.4 M IU/day/subject; from about 0.02 M IU/day/subject to about 3.0 M IU/day/subject; from about 0.1 M

IU/day/subject to about 3.0 M IU/day/subject; or from about 0.2 M IU/day/subject to 2.0 M IU/day/subject.

Claim 29. The method of any one of claims 23 to 28, wherein said subject has residual beta-cell function.

Claim 30. The method of any one of claims 23 to 29, wherein said subject has recent-onset T1D.

Claim 31. The method of any one of claims 23 to 30, wherein said subject has a blood or urine C-peptide concentration indicative of residual beta-cell function.

Claim 32. The method of any one of claims 23 to 31 , wherein said subject is a human patient having a fasting blood C-peptide concentration of less than about 1 nmol/L, but at least about 0.2 nmol/L; or has a stimulated blood C-peptide concentration of less than about 4 nmol/L, but at least about 0.5 nmol/L.

Claim 33. The method of any one of claims 23 to 32, wherein said subject has been diagnosed with T1D within the previous 12 months prior to administering said composition.

Claim 34. The method of any one of claims 23 to 33, wherein said composition is mucosally administered to said subject. Claim 35. The method of any one of claims 23 to 34, wherein said composition is administered to said subject in a liquid form.

Claim 36. The method of any one of claims 23 to 34, wherein said composition is administered to said subject in the form of a food product, a dietary supplement, or a suppository product.

Claim 37. The method of any one of claims 23 to 36, wherein said composition is administered in a unit dosage form comprising from about 1 x 104 to about 1 x 1012; about 1 x 106 to about 1 x 1012; or about 1 x 109 to about 1 x 1012 colony-forming units (cfu).

Claim 38. The method of claim 37, wherein said unit dosage form is selected from the group consisting of: a capsule, a tablet, a granule, a suppository, and a metered aerosol dose.

Claim 39. The method of any one of claims 23 to 34 and 36, wherein said composition is in a dry-powder form or a compressed version thereof.

Claim 40. A genetically modified microorganism comprising an exogenous nucleic acid encoding an IL-2 polypeptide; and an exogenous nucleic acid encoding a T ID- specific antigen polypeptide.

Claim 41. The genetically modified microorganism of claim 40, wherein said microorganism is a LAB.

Claim 42. The genetically modified microorganism of claim 40 or 41, wherein said exogenous nucleic acid encoding an interleukin-2 and/or TlD-specific antigen polypeptide is stably integrated into the chromosome of the microorganism.

Description:
COMPOSITIONS AND METHODS FOR THE TREATMENT

OF TYPE 1 DIABETES

CROSS REFERENCE TO RELATED APPLICATIONS

[001] This application claims benefit to U.S. Provisional Application No. 62/278,493, filed January 14, 2016, and U.S. Provisional Application No. 62/350,472, filed June 15, 2016, each of which is incorporated herein in its entirety.

REFERENCE TO SEQUENCE LISTING

[002] The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on January 10, 2017, is named 205350-0030-00-WO-549987_SL.txt and is 189,854 bytes in size.

BACKGROUND

[003] Approximately 10-15 million people suffer from type 1 diabetes mellitus (T1D), the most common metabolic disorder in infancy and adolescence, affecting 1 12,000 children younger than 16 years of age in Europe alone. T1D results from a progressive immune- mediated destruction of the pancreatic insulin-producing islet beta-cells ("beta-cells") in genetically-susceptible individuals, leading to chronic hyperglycemia which instigates micro- and macrovascular complications. See, e.g. , van Belle, T.L. et al. , Physiol. Rev. 2011 , 91(1): 79-1 18. While therapeutic options are available for some autoimmune diseases, no therapies are currently approved for T1D. Patients with T1D require lifelong treatment with insulin. Moreover, long-term management requires a multidisciplinary approach that includes physicians, nurses, dieticians, and other specialists.

[004] Blocking autoreactive effector T cells using generalized immunosuppression, either in the context of short-term therapy or chronic regimens, has been the sole therapeutic strategy in autoimmune diseases. It is believed that activating or expanding regulatory T (Treg) cells can restore a balance between effector T cells and Treg cells, and may achieve the same objective without the toxicity associated with immunosuppression.

[005] Interleukin-2 ("IL-2") has key functions of the immune system, primarily via its direct effects on T cells. In the thymus, where T cells mature, it prevents autoimmune diseases by promoting the differentiation of certain immature T cells into regulatory T cells, which suppress other T cells that are otherwise primed to attack healthy cells in the body. At higher concentrations, IL-2 also promotes the differentiation of T cells into effector T cells and into memory T cells when the initial T cell is also stimulated by an antigen, thus helping the body fight off infections.

[006] Native IL-2 was initially identified as a lymphocyte growth factor, and thought to primarily promote effector T cell responses in vivo, and recombinant IL-2 was developed for the treatment of conditions calling for the boosting of effector T cells, i.e. cancer and infectious diseases. However, it was shown that IL-2 is dispensable for the differentiation, survival and function of effector T cells, as IL-2 knockout mice develop T-cell-mediated autoimmune disease. IL-2 is now known to be a cytokine critical for Treg cell development, expansion, survival and peripheral activity. A deficiency in IL-2 production or lack of IL-2 responsiveness leads to a loss of Treg cell function and an increase in autoimmunity. Treg cells constitutively express the trimeric high affinity receptor for IL-2 (IL-2RaPy) at higher levels than CD4 + and CD8 + effector T cells, NK cells, and eosinophils. Induction of STAT5a signaling occurs at lower doses of IL-2 in Treg cells than in effector T cells. Hence, low dose IL-2 appears to stimulate preferential activation and promote the survival of Treg cells in vivo. See, e.g., Yu, A., et al., Diabetes 2015, 64: 2172-2183.

[007] In clinical studies, administration of IL-2 induced immunological changes, but did not change variables of glucose metabolism. See, e.g., Hartemann A. et ah, Lancet Diabetes Endocrinol. 2013; 1 :295-305; and Rosenzwajg M. et al., J Autoimmun. 2015; 58:48-58.

[008] Pharmaceutical IL-2 preparations are administered by injection. Although oral delivery is attractive, e.g., as a result of the ease of administration, gastrointestinal degradation and low levels of absorption generally render this route ineffective for the delivery of polypeptides. Alternative routes such as nasal, rectal, pulmonary, and ocular routes are being investigated for polypeptide-based therapeutics.

[009] Genetically modified bacteria have been used to deliver therapeutic molecules to the mucosal tissues. See, e.g., Steidler, L., et al, Nat. Biotechnol. 2003, 21(7): 785-789; and Robert S. and Steidler L., Microb. Cell Fact. 2014, 13 Suppl. 1 : SI 1.

[0010] Intestinal introduction of antigens implicated in beta-cell autoimmunity via genetically-altered Lactococcus lactis, has been shown to arrest T1D in NOD mice via induction of Foxp3+ Treg cells. Oral administration of genetically-altered Lactococcus lactis targets human pro-insulin (PINS) along with human IL-10 to the gastrointestinal (GI) mucosa, and in combination with systemic low-dose anti-CD3 antibody, resets the immune system towards long-term tolerance in nearly 60% of new-onset diabetic NOD mice. See, e.g., Robert, S. et al , Diabetes 2014, 63: 2876-2887; and Takiishi, T. et al. , J. Clin. Inv. 2012, 122(5): 1717-1725. However, clinical translation of such antigen-specific combination therapy involving additional immuno-modulators, such as anti-CD3 antibodies, is being hampered, e.g., because Fc-modified anti-human CD3 antibodies have not been approved by regulatory agencies for the treatment of TID.

[0011] There is a need in the art for efficacious, targeted, and controlled methods for the treatment of TID, without off-target activities and systemic toxicities. Such strategies should facilitate administration, increase safety, and ideally, improve efficacy and reduce therapeutically effective doses. The present disclosure addresses these needs.

INCORPORATION BY REFERENCE

[0012] All publications, patents, and patent applications herein are incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference. In the event of a conflict between a term herein and a term in an incorporated reference, the term herein controls.

BRIEF SUMMARY

[0013] Accordingly, provided are compositions and methods involving live lactic acid fermenting bacteria (LAB), e.g. , genetically modified Lactococcus lactis (LL) strains, as delivery vehicles for the mucosal delivery of low-dose IL-2, e.g., in combination with T1D- specific self-antigens, such as proinsulin (PINS). The LAB are genetically modified to express the bioactive polypeptides, which induce biological responses, which in turn, block further autoimmune destruction of pancreatic beta-cells. Such strategy can reverse established TID, e.g., in subjects with sufficient residual beta-cell function, and thus represent a "true" treatment for auto-immune diabetes. The compositions may be administered orally, e.g., in the form of an enterically coated pharmaceutical formulation which transports the bacteria to the gastrointestinal tract, e.g., to the lower part of the gastrointestinal tract {e.g., distal parts of the colon), where they will secrete a suitable low- dose of IL-2, optionally in combination with a TlD-specific antigen {e.g., PINS). [0014] The provided composition comprises a lactic acid fermenting bacterium (LAB) comprising an exogenous nucleic acid encoding an interleukin-2 (IL-2) polypeptide and an exogenous nucleic acid encoding a type-1 diabetes mellitus (TlD)-specific antigen polypeptide. Alternatively, the provided composition comprises a first LAB comprising an exogenous nucleic acid encoding an interleukin-2 (IL-2) polypeptide, and a second LAB comprising an exogenous nucleic acid encoding a type-1 diabetes mellitus (TlD)-specific antigen polypeptide. The composition may further comprise a pharmaceutically acceptable carrier. Said LAB may be adapted for mucosal delivery of low-dose IL-2 when administered to a mammalian subject.

[0015] In one aspect, said LAB may be selected from the group consisting of: a

Lactococcus species, a Lactobacillus species, a Bifidobacterium species, a Streptococcus species, and an Enterococcus species. Said LAB may be a Lactococcus species. For example, said LAB may be Lactococcus lactis. Alternatively, said LAB may be selected from the group consisting of: Lactococcus garvieae, Lactococcus lactis, Lactococcus lactis subsp. cremoris, Lactococcus lactis subsp. hordniae, Lactococcus lactis subsp. Lactis, Lactococcus piscium, Lactococcus plantarum, Lactococcus raffinolactis, Lactobacillus acetotolerans, Lactobacillus acidophilus, Lactobacillus agilis, Lactobacillus algidus, Lactobacillus alimentarius,

Lactobacillus amylolyticus, Lactobacillus amylophilus, Lactobacillus amylovorus,

Lactobacillus animalis, Lactobacillus aviarius, Lactobacillus aviarius subsp. araffinosus, Lactobacillus aviarius subsp. aviarius, Lactobacillus bavaricus, Lactobacillus bifermentans, Lactobacillus brevis, Lactobacillus buchneri, Lactobacillus bulgaricus, Lactobacillus carnis, Lactobacillus casei, Lactobacillus casei subsp. alactosus, Lactobacillus casei subsp. casei, Lactobacillus casei subsp. pseudoplantarum, Lactobacillus casei subsp. rhamnosus,

Lactobacillus casei subsp. tolerans, Lactobacillus catenaformis, Lactobacillus cellobiosus, Lactobacillus collinoides, Lactobacillus confusus, Lactobacillus coryniformis, Lactobacillus coryniformis subsp. coryniformis, Lactobacillus coryniformis subsp. torquens, Lactobacillus crispatus, Lactobacillus curvatus, Lactobacillus curvatus subsp. curvatus, Lactobacillus curvatus subsp. melibiosus, Lactobacillus delbrueckii, Lactobacillus delbrueckii subsp.

bulgaricus, Lactobacillus delbrueckii subsp. delbrueckii, Lactobacillus delbrueckii subsp. lactis, Lactobacillus divergens, Lactobacillus farciminis, Lactobacillus fermentum,

Lactobacillus fornicalis, Lactobacillus fructivorans, Lactobacillus fructosus, Lactobacillus gallinarum, Lactobacillus gasseri, Lactobacillus graminis, Lactobacillus halotolerans, Lactobacillus hamsteri, Lactobacillus helveticus, Lactobacillus heterohiochii, Lactobacillus hilgardii, Lactobacillus homohiochii, Lactobacillus iners, Lactobacillus intestinalis,

Lactobacillus jensenii, Lactobacillus johnsonii, Lactobacillus kandleri, Lactobacillus kefiri, Lactobacillus kefiranofaciens, Lactobacillus keflrgranum, Lactobacillus kunkeei, Lactobacillus lactis, Lactobacillus leichmannii, Lactobacillus lindneri, Lactobacillus malefermentans, Lactobacillus mali, Lactobacillus maltaromicus, Lactobacillus manihotivorans, Lactobacillus minor, Lactobacillus minutus, Lactobacillus mucosae, Lactobacillus murinus, Lactobacillus nagelii, Lactobacillus oris, Lactobacillus panis, Lactobacillus parabuchneri, Lactobacillus paracasei, Lactobacillus paracasei subsp. paracasei, Lactobacillus paracasei subsp. tolerans, Lactobacillus parakefiri, Lactobacillus paralimentarius, Lactobacillus paraplantarum, Lactobacillus pentosus, Lactobacillus perolens, Lactobacillus piscicola, Lactobacillus plantarum, Lactobacillus pontis, Lactobacillus reuteri, Lactobacillus rhamnosus,

Lactobacillus rimae, Lactobacillus rogosae, Lactobacillus ruminis, Lactobacillus sakei, Lactobacillus sakei subsp. camosus, Lactobacillus sakei subsp. sakei, Lactobacillus salivarius, Lactobacillus salivarius subsp. salicinius, Lactobacillus salivarius subsp. salivarius,

Lactobacillus sanfranciscensis, Lactobacillus sharpeae, Lactobacillus suebicus, Lactobacillus trichodes, Lactobacillus uli, Lactobacillus vaccinostercus, Lactobacillus vaginalis,

Lactobacillus viridescens, Lactobacillus vitulinus, Lactobacillus xylosus, Lactobacillus yamanashiensis, Lactobacillus yamanashiensis subsp. mali, Lactobacillus yamanashiensis subsp. Yamanashiensis, Lactobacillus zeae, Bifidobacterium adolescentis, Bifidobacterium angulatum, Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium catenulatum, Bifidobacterium longum, Bifidobacterium infantis, Enterococcus alcedinis, Enterococcus aquimarinus, Enterococcus asini, Enterococcus avium, Enterococcus caccae, Enterococcus camelliae, Enterococcus canintestini, Enterococcus canis, Enterococcus casseliflavus, Enterococcus cecorum, Enterococcus columbae, Enterococcus devriesei, Enterococcus diestrammenae, Enterococcus dispar, Enterococcus durans, Enterococcus eurekensis, Enterococcus faecalis, Enterococcus faecium, Enterococcus gallinarum, Enterococcus gilvus, Enterococcus haemoperoxidus, Enterococcus hermanniensis, Enterococcus hirae,

Enterococcus italicus, Enterococcus lactis, Enterococcus lemanii, Enterococcus malodoratus, Enterococcus moraviensis, Enterococcus mundtii, Enterococcus olivae, Enterococcus pallens, Enterococcus phoeniculicola, Enterococcus plantarum, Enterococcus pseudoavium,

Enterococcus quebecensis, Enterococcus raffinosus, Enterococcus ratti, Enterococcus rivorum, Enterococcus rotai, Enterococcus saccharolyticus, Enterococcus silesiacus, Enterococcus solitarius, Enterococcus sulfureus, Enterococcus termitis, Enterococcus thailandicus, Enterococcus ureasiticus, Enterococcus ureilyticus, Enterococcus viikkiensis, Enterococcus villorum, Enterococcus xiangfangensis, Streptococcus agalactiae, Streptococcus anginosus, Streptococcus bovis, Streptococcus canis, Streptococcus constellatus,

Streptococcus dysgalactiae, Streptococcus equinus, Streptococcus iniae, Streptococcus intermedius, Streptococcus milleri, Streptococcus mitis, Streptococcus mutans, Streptococcus oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus pyogenes, Streptococcus ratti, Streptococcus salivarius, Streptococcus tigurinus, Streptococcus thermophilus, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus suis, Streptococcus uberis, Streptococcus vestibularis, Streptococcus viridans, and Streptococcus zooepidemicus.

[0016] In another aspect, said TlD-specific antigen may be selected from the group consisting of: proinsulin (PINS), glutamic acid decarboxylase (GAD65), insulinoma-associated protein 2 (IA-2), islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), zinc transporter 8 (ZnT8), chromogranin A, (prepro) islet amyloid polypeptide (ppIAPP), peripherin, and citrullinated glucose-regulated protein (GRP). For example, said TlD-specific antigen may be PINS.

[0017] In another aspect, a LAB may comprise said exogenous nucleic acid encoding an IL-2 polypeptide and said exogenous nucleic acid encoding a TlD-specific antigen polypeptide. Alternatively, a first LAB may comprise said exogenous nucleic acid encoding an IL-2 polypeptide, and a second LAB may comprise said exogenous nucleic acid encoding a TlD-specific antigen polypeptide. Said exogenous nucleic acid encoding an IL-2 polypeptide may be integrated into the chromosome of said LAB. Said exogenous nucleic acid encoding a TlD-specific antigen polypeptide may be integrated into the chromosome of said LAB, or may be present on a plasmid contained in said LAB. Said exogenous nucleic acid encoding an IL-2 polypeptide and said exogenous nucleic acid encoding a TlD-specific antigen polypeptide may be both integrated into the chromosome of said LAB. Said exogenous nucleic acid encoding an IL-2 polypeptide and said exogenous nucleic acid encoding a TlD-specific antigen polypeptide may be part of a polycistronic expression unit, driven by the same promoter.

[0018] In yet another aspect, said IL-2 may be a membrane bound form of IL-2 or a soluble form of IL-2. Said exogenous nucleic acid encoding an IL-2 polypeptide may encode an IL-2 variant polypeptide. Said IL-2 variant polypeptide may have a diminished IL-2 activity or an enhanced IL-2 activity, when compared to a corresponding wild-type IL-2 polypeptide. Said IL-2 variant polypeptide may be selected from the group consisting of: aldesleukin, teceleukin, and bioleukin. For example, said IL-2 variant polypeptide comprises: (a) a first amino acid substitution relative to mature, wild-type IL-2, selected from the group consisting of L72G, L72A, L72S, L72T, L72Q, L72E, L72N, L72D, L72R, and L72K; or

(b) a second amino acid substitution relative to mature, wild-type IL-2, selected from the group consisting of F42A, F42G, F42S, F42T, F42Q, F42E, F42N, F42D, F42R, and F42K; or

(c) a third amino acid substitution relative to mature, wild-type IL-2, selected from the group consisting of Y45A, Y45G, Y45S, Y45T, Y45Q, Y45E, Y45N, Y45D, Y45R, and Y45K; or

(d) a combination thereof.

[0019] The provided composition may comprise a Lactococcus lactis, wherein said Lactococcus lactis comprises an exogenous nucleic acid encoding an IL-2 polypeptide and an exogenous nucleic acid encoding PINS, and wherein said Lactococcus lactis is adapted for mucosal delivery of low-dose IL-2 when administered to a mammalian subject. Said low-dose IL-2 delivery may be the range of from about 0.01 M IU/day/subject to about 5.4 M

IU/day/subject; from about 0.02 M IU/day/subject to about 3.0 M IU/day/subject; from about 0.1 M IU/day/subject to about 3.0 M IU/day/subject; or from about 0.2 M IU/day/subject to 2.0 M IU/day/subject.

[0020] Also provided is the use of the composition for the treatment of T1D in a mammalian subject in need thereof. The provided method of treating type 1 diabetes mellitus (T1D) comprising administering to a mammalian subject in need thereof a therapeutically effective amount of the composition.

[0021] In one aspect, no anti-CD3 antibody is administered to said subject in the method of treating T1D. Alternatively, the method of treating T1D further comprises administering an anti-CD3 antibody to said subject. Said anti-CD3 antibody may be administered in a low-dose simultaneously with said composition to said subject. Said anti-CD3 antibody may be administered intravenously to said subject.

[0022] In another aspect, said subject may have residual beta-cell function. Said subject may have recent-onset T1D. Said subject may have a blood or urine C-peptide concentration indicative of residual beta-cell function. Said subject may be a human patient having a fasting blood C-peptide concentration of less than about 1 nmol/L, but at least about 0.2 nmol/L; or has a stimulated blood C-peptide concentration of less than about 4 nmol/L, but at least about 0.5 nmol/L. Said subject may have been diagnosed with TID within the previous 12 months prior to administering said composition.

[0023] In a further aspect, said composition may be mucosally administered to said subject. Said composition may be administered to said subject in a liquid form. Said composition may be administered to said subject in the form of a food product, a dietary supplement, or a suppository product. Said composition may be administered in a unit dosage form comprising from about 1 x 10 4 to about 1 x 10 12 ; about 1 x 10 6 to about 1 x 10' 2 ; or about 1 10 9 to about 1 x 10 12 colony- forming units (cfu). Said unit dosage form may be selected from the group consisting of: a capsule, a tablet, a granule, a suppository, and a metered aerosol dose. Said composition may be in a dry-powder form or a compressed version thereof.

[0024] Further provided is a genetically modified microorganism comprising an exogenous nucleic acid encoding an IL-2 polypeptide; and an exogenous nucleic acid encoding a T1D- specific antigen polypeptide. For example, said microorganism may be a LAB. Said exogenous nucleic acid encoding an interleukin-2 and/or TID-specific antigen polypeptide may be stably integrated into the chromosome of the microorganism.

BRIEF DESCRIPTION OF THE DRAWINGS

[0025] FIG. 1 depicts a nucleotide sequence (SEQ ID NO: 46) encoding a fusion of usp45 secretion leader (SSusp45) and the hIL-2 gene, encoding human interleukin-2 (hIL-2; UniProt: P60568, aa 21-153), downstream of the highly expressed phosphopyruvate hydratase gene {eno; Gene ID: 4797432; location: NC_009004.1 (606184..607485)) comprising an intergenic region preceding the highly expressed L. lactis MG1363 50S ribosomal protein L30 gene {rpmD; Gene ID: 4797873; location: NC_009004.1 (2316732..231691 1 , complement)) between eno and SSusp45.

[0026] FIG. 2 depicts a schematic overview of genetic loci of LL-IL-2 (AthyA, trePTS (AtrePP), otsB, AptcC, and eno»hIL-2) with intergenic regions, and PCR amplification product sizes (bp).

[0027] FIG. 3 depicts an exemplary carrier plasmid with a backbone that exists of a pORI19 fragment to which a PhllA»P-glucuronidase (uidA; Gene ID: 946149) expression module was added; a cargo region comprising pins downstream of gapB coupled by the rpmD intergenic region, flanked by cross over (XO) areas, positioned 5' and 3' of eno»hil- 2; as well as an erythromycin selection marker: erythromycin resistant 23S RNA methylase gene (ermC). [0028] FIG. 4 depicts a 1.2 % agarose gel analysis of PCR fragments generated from LL- IL-2.

[0029] FIG. 5 depicts a Western blot showing the presence of hIL-2 in LL-IL-2 culture supernatants.

[0030] FIG. 6 depicts a nucleotide sequence (SEQ ID NO: 57) encoding a fusion of usp45 secretion leader (SSusp45) with the pins gene, encoding human proinsulin (PINS; UniProt: P01308, aa 25-110), downstream of the highly expressed glyceraldehyde 3-phosphate dehydrogenase gene (gapB; Gene ID: 4797877; location: NC_009004.1 (2492509..2493519, complement)) comprising an intergenic region preceding the highly expressed L. lactis MG1363 50S ribosomal protein L30 gene (rpmD; Gene ID: 4797873; location: NC_009004.1

(2316732..231691 1 , complement), see, e.g., Steidler et al, Nat. Biotechnol. 2003; 21(7):785- 789) between gapB and pins.

[0031] FIG. 7 depicts a schematic overview of relevant genetic loci of LL-PINS/IL-2: AthyA, trePTS (AtrePP), otsB, ptcC-, gapB»pins and eno»hil-2 with indication of the relevant oligonucleotide binding sites, EcoRI restriction site, (/truncated/) genetic characters, intergenic regions (IR), PCR amplification product sizes (bp).

[0032] FIG. 8 depicts an exemplary carrier plasmid with a backbone that exists of a pORI19 fragment to which a PhllA»P-glucuronidase (uidA; Gene ID: 946149) expression module was added; a cargo region containing pins downstream of gapB coupled by intergenic region rpmD, and flanked by cross over (XO) areas, positioned 5' and 3' of gapB»pins; as well as an erythromycin selection marker: erythromycin resistant 23 S RNA methylase gene (ermC).

[0033] FIG. 9 depicts a 1.2% agarose gel analysis of PCR fragments generated from LL- PINS/IL-2.

[0034] FIGs. 10A and 10B depict Western blots showing (1) the presence of PINS (black arrowhead) in LL-PINS/IL-2 culture supernatants (FIG. 10A), and (2) the presence of hIL-2 (open arrowhead) in LL-PINS/IL-2 culture supernatants (FIG. 10B).

[0035] FIG. 11 depicts a stable reversal of hyperglycemia in new-onset diabetic NOD mice in an exemplary antigen-specific therapy according to the present disclosure. New-onset diabetic NOD mice were treated as described herein, e.g., in Example 3, and blood glucose concentrations were followed up for 14 weeks post-treatment initiation. Shown is the percentage of mice that remained diabetic upon treatment with mucosally delivered LL-PINS or mucosally delivered LL-PINS+LL-IL-2.

[0036] FIG. 12 depicts the effectiveness of an exemplary antigen-specific therapy according to the present disclosure in recent-onset diabetic mice with an initial blood glucose concentration of more than or less than 350 mg/dL. Shown is the percentage of mice that remained diabetic upon treatment with mucosally delivered LL-PINS+LL-IL-2 as described herein, e.g., in Example 3.

[0037] FIG. 13 depicts a comparison of the biological activities between LL-IL-2 and recombinant human IL-2 (rhIL-2).

[0038] FIGs. 14A and 14B depict the concentrations of live bacteria (FIG. 14A:

CFU/tissue; FIG. 14B: CFU/g, respectively) in different tissues of the GI tract at different time-points after administration of a single dose (10 10 CFU) of LL-IL-2 to non-obese diabetic mice. All bars represent an average of 3 mice (n=3). SIP = proximal small intestine; SID = distal small intestine; CAE = caecum; COP = proximal colon; COD = distal colon.

[0039] FIG. 15 depicts the structure of plasmid pAGX0053. The plasmid backbone exists of a pTINX fragment to which a PthyA»SSusp45::hpins expression module was added. PthyA, promoter of the thymidylate synthase gene. EmR: erythromycin selection marker; repD, repE: Replication genes.

[0040] FIG. 16 depicts a western blot showing the presence of full-length, plasmid derived PINS in LL-PINS culture supernatants.

[0041] FIG. 17 depicts diabetes remission rates in new-onset diabetic NOD mice treated with various recombinant bacteria. Results demonstrate that mucosally delivered LL-IL-2 (e.g., providing low-dose IL-2), optionally in combination with an exemplary TID-specific antigen (i.e., PINS) according to the present disclosure induces diabetes remission, and stably reverses hyperglycemia in new-onset diabetic NOD mice. Mice were treated for 6 weeks as described herein, and blood glucose concentrations were measured, including 14 weeks post- treatment initiation. Shown is the percentage of mice that remained diabetic after treatment (Mantel-Cox log-rank test; ** p<0.01).

[0042] FIG. 18 depicts the diabetes remission rates according to starting blood glucose concentrations. Results indicate that starting blood glucose concentrations can predict therapeutic success in mice. Recent onset diabetic mice were stratified based on an initial (prior to treatment) blood glucose level of less than or greater than 350 mg/dL. Results demonstrate that mucosally delivered LL-IL-2, optionally in combination with an exemplary antigen- specific therapy (e.g., PINS) according to the present disclosure is particularly effective in recent-onset diabetic mice with an initial blood glucose concentration of less than 350 mg/dL. Shown is the percentage of mice that remained diabetic upon treatment with LL- IL-2 or LL-PINS + LL-IL-2 as described herein. It is noted that 6 mice treated with LL-IL-2 alone and having less than 350 mg/dL glucose, indicated as "LL-IL2 < 350 (n=9)" were sacrificed after 6 weeks of treatment and 3 mice were observed for the full 14 week period. A data timepoint for LL-IL-2 > 350 (n=6) (solid triangle) is hidden behind a data point for Untreated > 350 (n=16) (solid square). All LL-IL-2 treated mice having > 350 mg/dL of glucose were still diabetic after 6 weeks of treatment and after the 14 week follow-up period (Mantel-Cox log-rank test; **** pO.0001).

[0043] FIG. 19 depicts insulitis scoring of islet beta-cells in diabetic NOD mice. Results demonstrate that mucosally delivered LL-IL-2 (e.g., providing low-dose IL-2), optionally in combination with an exemplary antigen-specific therapy according to the present disclosure (e.g., LL-PINS+LL-IL-2) did not only prevent worsening of insulitis (normally seen during progression to long-standing diabetes when untreated), but reduces insulitis of islet beta-cells when compared to recent-onset and untreated longstanding diabetic mice (e.g., reduces insulitis to a degree comparable to insulitis found in longstanding "normoglycemic" NOD mice). The degree of heavy insulitis improved upon treatment when compared to untreated longstanding diabetic mice. The percentage of insulitis-free islet beta-cells dramatically increased when compared to recent-onset and untreated longstanding mice. A significant percentage of islets with mild-insulitis improved to "peri-insulitis" or "no insulitis." Unexpectedly, this significant reduction in insulitis was observed in all treated recent-onset mice (with and without remission - classified as "cured" and "uncured").

[0044] FIG. 20 depicts the random C-peptide concentrations in the plasma of diabetic NOD mice. Results demonstrate that low-dose IL-2 with or without proinsulin preserves beta- cell function in diabetic NOD mice. After 6 weeks of LL-IL-2 treatment, optionally in combination with an exemplary antigen-specific therapy according to the present disclosure (e.g., LL-PINS+LL-IL-2) plasma C-peptide concentrations in recent-onset diabetic mice showing remission (classified as "cured") increased when compared to recent-onset and untreated long-standing diabetic mice (Mann-Whitney T-test; *p<0.05). C-peptide concentrations in the plasma of treated, but "uncured" mice were not detected (ND) indicating that such mice may have little or no remaining active beta cells. The C-peptide levels measured for 3 out of the 4 analyzed untreated longstanding diabetic mice were also not detected.

[0045] FIG. 21 depicts the expansion of foxp3+CTLA4+ regulatory T cells in different immune cell subsets, locally (i.e., mesenteric draining lymph nodes; MLN), systemically (i.e., spleen and blood), and at the target organ (i.e., pancreatic LN), of diabetic NOD mice measured by flow cytometry after 6 weeks of treatment. Results demonstrate that mucosally delivered LL-IL-2, optionally in combination with an exemplary antigen-specific therapy according to the present disclosure (e.g., LL-PINS+LL-IL-2) increases the number of foxp3+CTLA4+ regulatory T cells in the pancreatic LN when compared to recent-onset mice. Unexpectedly, increases in the pancreatic LN were observed in mice exhibiting remission ("cured") and those not exhibiting remission ("uncured") mice (Mann- Whitney T-test; * p<0.05, ** p<0.01 , *** pO.001).

[0046] FIGs. 22A and 22B depict the blood glucose concentrations (mg/dL) in recent onset-diabetic mice treated with LL-IL-2 alone (FIG. 22A) and treated with LL-IL-2+LL-PINS (FIG. 22B).

[0047] FIG. 23 depicts diabetes remission rate according to starting blood glucose concentrations under 350 mg/dl at study entry. Mice were allocated to 5 experimental treatment groups: untreated controls, LL-PINS, LL-IL2, a mixture of LL-PINS +LL-IL2 and a mixture of LL-PINS+LL-IL2 combined with a systemic immunomodulatory anti-CD3, as described in Example 9. Shown is the percentage of mice that remained diabetic at various time points after treatment.

DETAILED DESCRIPTION

Abbreviations and Acronyms used in herein may include:

BD Becton Dickinson

BSA bovine serum albumin

CAE caecum

CDS coding sequence

CFU colony forming units

COD or DCO distal colon

COP or PCO proximal colon

DCO distal colon

EDTA ethylenediaminetetraacedic acid ELISA enzyme-linked immunosorbent assay

FACS fluorescence-activated cell sorting

FCS fetal calf serum

FOS fructo-oligosaccharides

GAD65 glutamic acid decarboxylase

GRP citrullinated glucose-regulated protein

GUS glucuronidase

HRP horseradish peroxidase

IA-2 insulinoma-associated protein 2

IAA insulin auto-antibodies

IC insulin content determination

IGRP islet-specific glucose-6-phosphatase catalytic subunit-related protein

IL-2 interleukin-2

INS insulin

IU international unit

LAB lactic acid fermenting bacterium/bacteria

LL Lactococcus lactis

LLOQ limit of quantification

MCT oil medium chain triglycerides

MLN mesenteric lymph nodes

MMTT mixed meal tolerance test

MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide

MWM molecular weight marker

NCBI National Center for Biotechnology Information

NIBSC National Institute for Biological Standards and Control

NK cells natural killer cells

NOD mice non-obese diabetic mice

PBS phosphate-buffered saline

PCR polymerase chain reaction

PFA paraformaldehyde

PINS proinsulin

PLN pancreatic draining lymph nodes

ppIAPP (prepro) islet amyloid polypeptide PTPRN protein tyrosine phosphatase, receptor type N

PTS phosphotransferase system

RIA Radioimmunoassay (RIA)

SID/DSI distal small intestine

SIP/PSI proximal small intestine

SPL spleen

TID type-1 diabetes mellitus

TSLP Thymic stromal lymphopoietin (TSLP)

WHO World Health Organization

XO cross over

ZnT8 zinc transporter 8

[0049] Currently provided are compositions and methods for the treatment of TID, for the induction of Tregs and/or for restoring tolerance to TlD-specific antigens (i.e., self- antigens) in a subject.

[0050] Provided herein are compositions comprising (1) a LAB comprising an interleukin-2 (IL-2) gene and a TlD-specific antigen gene, or (2) a first LAB comprising an Interleukin-2 (IL-2) gene, and a second LAB comprising a TlD-specific antigen. In some examples, the LAB expresses the IL-2 gene and/or the TlD-specific antigen gene to produce IL-2 and TlD-specific antigen (e.g., PINS). In some embodiments, the compositions are pharmaceutical compositions comprising the LAB and a pharmaceutically acceptable carrier. Exemplary carriers are described herein. In some examples, the pharmaceutical composition is adapted for mucosal delivery of the composition to the subject.

[0051] Methods are provided for the treatment of TID in a mammalian subject in need thereof. The methods include administering (e.g., via a mucosal route) to the subject a composition according to the present disclosure. Exemplary methods include: administering to the subject a therapeutically effective amount of the LAB capable of expressing IL-2 and a TlD-specific antigen (e.g., PINS).

[0052] Unexpectedly, it is discovered that subjects with significant residual beta-cell function respond particularly well to the therapeutic methods described herein. Thus, in some embodiments, the mammalian subject in the herein described methods, has recently been diagnosed with TID and/or has recent-onset TID. In some examples, the subject may have been diagnosed with TID within the previous 12 months, the previous 24 months, or the previous 36 months prior to administering the composition comprising the LAB described herein.

[0053] In some examples in the herein described methods, the IL-2 and antigen polypeptides are delivered to the mucosa. This approach may result in delivering low-dose IL-2 concentrations that are even lower than those required for low-dose systemic administration. Off-target toxicities associated with systemic delivery of IL-2 may thus be avoided.

Definitions

[0054] As used herein, the singular forms "a," "an" and "the" include plural references unless the context clearly dictates otherwise. For example, the term "a cell" includes a plurality of cells, including mixtures thereof. Similarly, use of "a compound" for treatment or preparation of medicaments as described herein contemplates using one or more compounds for such treatment or preparation unless the context clearly dictates otherwise.

[0055] As used herein, the term "comprising" is intended to mean that the compositions and methods include the recited elements, but not excluding others. "Consisting essentially of when used to define compositions and methods, shall mean excluding other elements of any essential significance to the combination. Thus, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method and pharmaceutically acceptable carriers, such as phosphate buffered saline, preservatives, and the like. "Consisting of shall mean excluding any other component in more than trace elements of other ingredients and substantial method steps for administering the compositions described herein. Embodiments defined by each of these transition terms are within the scope of present disclosure.

[0056] As used herein, the term "expressing" a gene or polypeptide or "producing" a polypeptide (e.g., an IL-2 polypeptide or TlD-specific antigen polypeptide) is meant to include "capable of expressing" and "capable of producing," respectively. For example, a microorganism, which contains an exogenous nucleic acid can under sufficient conditions (e.g., sufficient hydration and/or in the presence of nutrients) produce a polypeptide encoded by the exogenous nucleic acid). However, the microorganism may not always actively produce the encoded polypeptide. The LAB (e.g., Lactococcus lactis) may be dried (e.g., freeze-dried), and in that state can be considered dormant (i.e., is not actively producing polypeptide). However, once the LAB is subjected to sufficient conditions, e.g., is administered to a subject and is released (e.g., into the gastro-intestinal tract of a subject) it may begin producing polypeptide. Thus, a LAB "expressing" a gene or polypeptide or "producing" a polypeptide of the current disclosure includes the LAB in its "dormant" state.

[0057] The term "about" in relation to a reference numerical value, and its grammatical equivalents as used herein, can include the reference numerical value itself and a range of values plus or minus 10% from that reference numerical value. For example, the term "about 10" includes 10 and any amounts from and including 9 to 11. In some cases, the term "about" in relation to a reference numerical value can also include a range of values plus or minus 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1 % from that reference numerical value.

[0058] An "IL-2 gene" refers to an interleukin 2 gene encoding an "IL-2 polypeptide." The term "IL-2 gene" includes "IL-2 variant genes" encoding "IL-2 variant polypeptides."

[0059] The term "IL-2" or "IL-2 polypeptide" refers to a functional, e.g., full-length, interleukin 2 polypeptide (e.g., human IL-2 polypeptide), including membrane-bound forms and soluble forms, as well as "IL-2 variant polypeptides."

[0060] An "IL-2 variant" or "IL-2 variant polypeptide" refers to a modified (e.g., truncated or mutated), but functional IL-2 polypeptide, e.g., a truncated or mutated version of human IL-2. The term "IL-2 variant polypeptide" includes IL-2 polypeptides with enhanced activity or diminished activity when compared to a corresponding wild-type IL-2 polypeptide. An "IL-2 variant polypeptide" retains at least some IL-2 activity.

TID-Speciflc Antigen

[0061] The terms "TID-specific self-antigen," "TID-specific antigen," "disease-specific antigen," "self-antigen," "auto-antigen," or "antigen" are used interchangeably herein. The terms are used herein in accordance with the art recognized meaning of self-antigen or auto- antigen, and generally refer to a polypeptide/protein originating from within a subjects own body (produced by the subject's own body), wherein the antigen is recognized by the subject's own immune system, and typically produces antibodies against such antigen.

Autoimmune diseases are generally associated with certain disease-specific self-antigens. In T1D a subject's immune system may produce antibodies against at least one antigen associated with the beta-cell destruction process. Such self-antigens include proinsulin (PINS), glutamic acid decarboxylase (GAD65), insulinoma-associated protein 2 (IA-2), islet- specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) and zinc transporter (ZnT) 8. Clinical T1D may further be associated with additional candidate target molecules expressed by beta-cells such as chromogranin A, (prepro) islet amyloid polypeptide

(ppIAPP), peripherin and citrullinated glucose-regulated protein (GRP).

[0062] The term "TlD-specific antigen gene" refers to a gene encoding the above "TlD- specific antigen." The term "TlD-specific antigen gene" includes "TlD-specific antigen variant genes" encoding "TlD-specific antigen variant polypeptides."

[0063] The term "TlD-specific antigen polypeptide" refers to a functional, e.g. , full- length, polypeptide, as well as "TlD-specific antigen variant polypeptides," which may have enhanced activity or diminished activity when compared to a corresponding wild-type polypeptide.

[0064] The term "TlD-specific antigen variant" or "TlD-specific antigen variant polypeptide" refers to a modified (e.g., truncated or mutated), but functional polypeptide, e.g., a truncated or mutated version of human PINS. The term "variant polypeptide" includes polypeptides with enhanced activity or diminished activity when compared to a

corresponding wild-type polypeptide. A "variant polypeptide" retains at least some biological activity (functional polypeptide). Exemplary variants of GAD65 and IA-2 include trimmed versions thereof (e.g., GAD65 3 7 0 -575, and IA-2635-97 , respectively; relative to NCBI accession numbers NP_000809.1 (SEQ ID NO: 7) and NP_002837.1 (SEQ ID NO: 9, respectively) retaining antigenic properties, and are thus useful in the compositions and methods of the current disclosure, e.g., in stimulating Tregs and inducing tolerance in a subject. Generally, trimmed or truncated versions of a TlD-specific antigen are efficiently expressed and secreted by the LAB (e.g., Lactococcus lactis).

[0065] The term "operably linked" refers to a juxtaposition wherein the components described are in a relationship permitting them to function in their intended manner. A control sequence "operably linked" to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under condition compatible with the control sequences.

Subject

[0066] A "subject" is an organism, which may benefit from being administered a composition of the present disclosure, e.g., according to methods of the present disclosure. The subject may be a mammal ("mammalian subject"). Exemplary mammalian subjects include humans, farm animals (such as cows, pigs, horses, sheep, goats), pets (such as a dogs, cats, and rabbits), and other animals, such as mice, rats, and primates. In some examples, the mammalian subject is a human patient.

Low-Dose IL-2

[0067] The term "low-dose IL-2" refers to a dose or a concentration of IL-2 polypeptide which can promote competence and stability of regulatory T (Treg) cell populations and/or promotes the development of naive CD4+ T cells into Treg cells in the respective subject, but is below a threshold dose/concentration, which stimulates the differentiation of naive T cells into effector T cells and/or memory T cells in a subject. It has been shown that Treg cells have a 10-20 fold lower activation threshold for IL-2 than effector T cells, e.g., when measured in terms of STAT5 (pSTAT5). Downstream of pSTAT5, the activation of numerous genes important for cell function require IL-2 doses that are 100-times lower for Treg cells than for effector T-cells (see, e.g., Yu, A., et al, Diabetes 2015, 64: 2172-2183). However, in connection with known treatment regimens in humans, a minimum dose of IL-2 necessary to stimulate Treg cells has not been established.

[0068] In some embodiments, e.g., in the context of human treatment, low-dose IL-2 typically refers to a dose of IL-2 polypeptide or IL-2 variant polypeptide that can be in the range of from about 0.01 M IU/day/subject to about 5.4 M IU/day/subject. The low dose can be in the range of from about 0.01 M IU/day/subject to about 3.0 M IU/day/subject. The low dose can be in the range of from about 0.02 M IU/day/subject to about 3 M IU/day/subject, from about 0.03 M IU/day/subject to about 3 M IU/day/subject, from about 0.04 M

IU/day/subject to about 3 M IU/day/subject, from about 0.05 M IU/day/subject to about 3 M IU/day/subject, from about 0.06 M IU/day/subject to about 3 M IU/day/subject, from about 0.07 M IU/day/subject to about 3 M IU/day/subject, from about 0.08 M IU/day/subject to about 3 M IU/day/subject, from about 0.09 M IU/day/subject to about 3 M IU/day/subject, from about 0.1 M IU/day/subject to about 3 M IU/day/subject, from about 0.2 M

IU/day/subject to about 3 M IU/day/subject, from about 0.3 M IU/day/subject to about 3 M IU/day/subject, from about 0.4 M IU/day/subject to about 3 M IU/day/subject, from about 0.5 M IU/day/subject to about 3 M IU/day/subject, from about 0.6 M IU/day/subject to about 3 M IU/day/subject, from about 0.7 M IU/day/subject to about 3 M IU/day/subject, from about 0.8 M IU/day/subject to about 3 M IU/day/subject, from about 0.9 M IU/day/subject to about 3 M IU/day/subject, or from about 1.0 M IU/day/subject to about 3 M IU/day/subject. The low dose can be in the range of from about 0.02 M IU/day/subject to about 2.5 M

IU/day/subject. The low dose also can be in the range of from about 0.05 M IU/day/subject to about 2.0 M IU/day/subject. The low dose can be in the range of from about 0.1 M IU/day/subject to about 1.5 M IU/day/subject. In still other embodiments, the low dose can be in the range of from about 0.3 MlU/day/subject to about 1.0 M IU/day/subject. The low dose can be in the range of from 0.5 M IU/day/subject to about 1.0 M IU/day/subject.

[0069] The term "international unit" (IU) is used herein in accordance with its art- recognized meaning and represents an amount of a substance (e.g., polypeptide). The mass or volume that constitutes one international unit varies based on which substance is being measured. The World Health Organization (WHO) provides unit characterizations for bioactive polypeptides. For example, 1 IU of human IL-2 is equivalent to about 73 pg of bioactive polypeptide (WHO International Standard; NIBSC 86/500).

Low-Dose Anti-CD3

[0070] The term "low-dose anti-CD3" refers to a cumulative dose or a concentration of anti-CD3 antibody which is below a standard dose of anti-CD3 antibody, or a regulatory approved dose of anti-CD3 antibody in humans to treat disease such as T1D or cancer. For example, in humans, a low-dose anti-CD3 treatment can comprise a dose of less than 50 mg (cumulative) of anti-CD3 antibody in a human. For example, a low-dose anti-CD3 can comprise about 1 mg to about 50 mg; about 5 mg to about 40 mg; about 10 mg to about 30 mg; about 15 mg to about 25 mg; about 20 mg to about 30 mg; about 15 mg to about 20 mg; or about 30 mg to about 35 mg of cumulative anti-CD antibody treatment. A lose-dose anti- CD3 can comprise less than about 50 mg; about 45 mg; about 40 mg; about 35 mg; about 30 mg; about 25 mg; about 20 mg; about 15 mg; about 10 mg; or about 5 mg of cumulative anti- CD antibody treatment.

[0071] For example, in some cases, the cumulative dosage of anti-CD3 antibody dosed in in humans can be about 34 mg or about 17 mg, given over a specific time periods, e.g., in a 14 day period. This means that about 2.43 mg or 1.21 mg of anti-CD3 antibody is given over the course of the 14 day period.

[0072] A low-dose anti-CD3 treatment can also comprise a dose of about 80%; about 70%; about 60%; about 50%; about 40%; about 30%; about 20%; about 10%; about 5%; about 2%; about 1 %; from about 80% to about 70%; from about 70% to 60%; from about 60% to 50%; from about 50% to 25%; from about 40% to 15%; or from about 30% to 5% of a regulatory approved dose of anti-CD3 antibody to treat T1D or cancer. [0073] In other mammals such as mice, the low-dose anti-CD3 can refer to a dosage less than about 5 μg; 2.5 μg; or 1 μg. For example, about 5 μg can be used for the treatment in a mouse. In some instances, about 2.5 μg can be used for the treatment in a mouse. In other cases, 1 μg can be used for the treatment in a mouse. Total dosage for mice can be 12.5 μg or 6 μg of anti-CD3.

[0074] Anti-CD3 can be given at least once a day to up to 5 times a day. For example, once a day, 2 times a day, or 3 times a day, as long as the long as the cumulative dosage is met. For example, if the dose to be given is 2.43 mg/day, and dosing occurs twice a day, then 1.21 mg per dose can be given.

[0075] To achieve a low dose anti-CD3 regime, dosages can be given at least once a day continuously for at least 3 days; 4 days; 5 days; 6 days; 7 days; 8 days; 9 days; 10 days; 1 1 days; 12 days; 13 days; 14 days; 15 days; 16 days; 17 days, 18 days; 19 days; 20 days; 30 days; or 40 days as long as the long as the cumulative dosage is met. For example, if a 34 mg anti-CD3 dosage regime is given (a low-dose anti-CD3) for 14 days, approximately 2.43 mg/day can be given to the subject. In some cases, the low-dose anti-CD can be given at least once a day continuously for at least 1 month, 2 months, 3 months, 6 months, 1 year, or more.

[0076] Low-dose anti-CD3 can be given intravenously simultaneously with the administration of the composition described herein. Optionally, low-dose anti-CD3 can be given 1 day; 2 days; 3 days; 4 days; 5 days; 6 days; 7 days; 2 weeks; 3 weeks; or 1 month after the first administration of the composition described herein. Additionally, in some instances, low-dose anti-CD3 can be given 1 day; 2 days; 3 days; 4 days; 5 days; 6 days; 7 days; 2 weeks; 3 weeks; or 1 month before the first administration of the composition described herein.

[0077] In some cases, the standard dose of anti-CD3 antibody, or a regulatory approved dose of anti-CD3 antibody in humans to treat disease such as TID and cancer, can be given to the patients before or after the administration of the compositions described herein.

[0078] In certain embodiments, anti-CD3 antibody can be teplizumab. Patient Sub-Populations

[0079] The subject being treated using the methods described herein can have significant (e.g., measurable) residual beta-cell function. Under such circumstances, the subject may maintain disease remission, even after treatment is interrupted or stopped altogether. Newly diagnosed patients often have a certain minimal number of pancreatic islet beta-cells (beta- cells) remaining at the time of diagnosis, so that such patients are able to produce a certain minimal amount of endogenous insulin. Such patient population can benefit particularly well when treated with the compositions and methods of the current disclosure (e.g., low-dose IL- 2 and PINS therapy). The treatments described herein can prevent further destruction of beta-cells and may thus induce disease remission. It was found that initial beta-cell mass may affect the efficacy of treatment. For example, in 57% of recent-onset NOD mice treated with the compositions of the current disclosure, and having a blood glucose concentration of about 350 mg/dL or less at treatment initiation, diabetes could be reversed. Reversal of disease was accomplished in only 22% of mice having an initial glucose concentration of more than 350 mg/dL. Further, in recent-onset mice, reversal of disease remained stable after treatment was stopped, indicating that the methods of the current disclosure (involving mucosal delivery of the bioactive polypeptides) can effectively correct hyperglycemia and restore long-term tolerance to beta-cells. However, once a subject's beta-cells are destroyed, such subject may no longer benefit from the described treatment in the same manner.

Treating

[0080] The terms "treatment", "treating", and the like, as used herein means ameliorating or alleviating characteristic symptoms or manifestations of a disease or condition, e.g., T1D. For example, treatment of T1D can result in the restoration or induction of antigen-specific immune tolerance in the subject. In other examples, treatment means arresting auto-immune diabetes, or reversing autoimmune diabetes. For example, treatment may result in the maintenance of remaining beta-cell mass. In other examples, treatment of T1D involves increasing the frequency or activation of Treg cells. In other examples, treatment may expand antigen-specific Treg cells (e.g., in the thymus), and/or induces migration of Treg cells into peripheral blood. In yet other examples, treatment involves improving at least one of a subject's (a human patients) clinical marker. For example, treatment may raise blood and/or urine C-peptide levels. In other examples, treatment may lower the subject's (e.g., a human patient's) blood glucose levels (e.g., in response to food ingestion or fasting glucose levels); reduce the amount of injected insulin required to maintain appropriate blood glucose levels in the subject, reduce diabetes-related auto-antibody levels in a subject, and/or increase/preserve C-peptide levels (e.g., following an oral glucose tolerance test). Treatment can mean continuous/chronic treatment, or treatment, in which the subject is free of clinical symptoms of the disease or condition for a significant amount of time (e.g., at least 6 months, at least 1 year, at least 2 years, at least 3 years, at least 4 years, or at least 5 years), after the treatment is stopped.

[0081] As used herein, these terms also encompass, preventing or delaying the onset of a disease or condition or of symptoms associated with a disease or condition, including reducing the severity of a disease or condition or symptoms associated therewith prior to affliction with said disease or condition. Such prevention or reduction prior to affliction refers to administration of the compound or composition described herein to a patient that is not at the time of administration afflicted with the disease or condition. "Preventing" also encompasses preventing the recurrence or relapse-prevention of a disease or condition or of symptoms associated therewith, for instance after a period of improvement.

Therapeutically Effective Amount

[0082] As used herein, the term "therapeutically effective amount" refers to an amount of a non-pathogenic microorganism or a composition of the present disclosure that will elicit a desired therapeutic effect or response when administered according to the desired treatment regimen. The compounds or compositions are typically provided in a unit dosage form, for example a tablet or capsule, which contains an amount of the active component equivalent with the therapeutically effective amount when administered once, or multiple times per day.

[0083] A person of ordinary skill in the art will appreciate that a therapeutically effective amount of a recombinant microorganism, which is required to achieve a desired therapeutic effect (e.g., for the effective treatment of T1D), will vary, e.g., depending on the nature of the IL-2 polypeptide expressed by the microorganism, the nature of the antigen polypeptide expressed by the LAB, the route of administration, and the age, weight, and other characteristics of the recipient.

Recent-Onset T1D

[0084] In some embodiments, the subject has recent-onset T1D. The term "recent-onset T1D," new-onset T1D," or "recent-onset disease" refers to a subject's (e.g., a human patient's) condition, which has recently been diagnosed with T1D (e.g., within about 3 months, within about six months, within about 9 months, within about 12 months, within about 15 months, within about 18 months, within about 24 months, within about 30 months, within about 36 months, within about 42 months, within about 48 months, within about 54 months, or within about 60 months). [0085] In humans, the decline of beta-cell function, which occurs prior to and after the diagnosis of T1D, can be measured using diagnostic marker compounds. For example, C- peptide is produced in equal amounts to insulin (during enzymatic cleavage of pro-insulin) and can therefore be used as a measure of endogenous insulin secretion (including in patients being treated with insulin). C-peptide has been used in the clinical management of patients with diabetes, and assay systems for measuring C-peptide are known to those of skill in the art. See, e.g., Jones A.G. and Hattersley A.T., Diabetic Medicine 2013, 30: 803-817; Little KR et al., Clin. Chem. 2008, 54: 1023-1026; Wiedmeyer et al, Clin. Chem. 2007, 53: 784- 787.

[0086] C-peptide values can be measured in nmol/L (wherein 1 nmol/L is 1000 pmol/L, and is equivalent to about 3 ng/mL). C-peptide can be measured in the blood or the urine of a subject. Blood C-peptide levels can be determined in non-fasting subjects (random C- peptide), in fasting subjects (fasting C-peptide), or in subjects stimulated with a dietary stimulator, such as a mixed liquid meal, or glucagon (stimulated C-peptide). C-peptide in the urine can be measured as the total amount of C-peptide secreted by the subject over a period of 24 hours. Often, C-peptide contained in the urine is measured as a ratio between C-peptide and creatinine.

[0087] In some embodiments, a subject (e.g., human) prior to administering the composition of the present disclosure (e.g., a subject with recent-onset T1D) has a fasting blood C-peptide concentration of less than about 1 nmol/L, but at least about 0.5 nmol/L, at least about 0.4 nmol/L, at least about 0.3 nmol/L, or at least about 0.2 nmol/L. In other embodiments, a subject (e.g., human) has a stimulated blood C-peptide concentration of less than about 4 nmol/L, but at least about 1 nmol/L, at least about 0.9 nmol/L, at least about 0.8 nmol/L, at least about 0.7 nmol/L, at least about 0.6 nmol/L, or at least about 0.5 nmol/L. In yet other embodiments, a subject (e.g., human) with recent-onset T1D has a post-meal urine C-peptide:creatinine ratio (nmol/mmol) of less than about 4, but at least about 1 , at least about 0.9, at least about 0.8, at least about 0.7, at least about 0.6, at least about 0.5, at least about 0.4, or at least about 0.3.

[0088] In other embodiments, a recent onset T1D subject (e.g., human patient) can be identified by measuring insulin auto-antibodies (IAA) in the serum or blood of the subject. In some examples, the subjects tests positive for IAA. Serum IAA concentration may also be used to measure disease progression or treatment progress. Methods for measuring insulin auto-antibodies have been described. See, e.g., Demeester et al, Diabetes Care 2015, 38(4): 644-651.

Mucosa

[0089] The term "mucosa" or "mucous membrane" is used herein in accordance with its art recognized meaning. The "mucosa" can be any mucosa found in the body, such as oral mucosa, rectal mucosa, gastric mucosa, intestinal mucosa, urethral mucosa, vaginal mucosa, ocular mucosa, buccal mucosa, bronchial or pulmonary mucosa, and nasal or olfactory mucosa.

[0090] The term "mucosal delivery" as used herein is used in accordance with its art recognized meaning, i.e., delivery to the mucosa, e.g., via contacting a composition of the present disclosure with a mucosa. Oral mucosal delivery includes buccal, sublingual and gingival routes of delivery. Accordingly, in some embodiments, "mucosal delivery" includes gastric delivery, intestinal delivery, rectal delivery, buccal delivery, pulmonary delivery, ocular delivery, nasal delivery, vaginal delivery and oral delivery.

[0091] The term "mucosal tolerance" refers to the inhibition of specific immune responsiveness to an antigen in a mammalian subject (e.g., a human patient), after the subject has been exposed to the antigen via the mucosal route. Typically, said mucosal tolerance is systemic tolerance. Low dose oral tolerance is oral tolerance induced by low doses of antigens, and is characterized by active immune suppression, mediated by cyclophosphamide sensitive regulatory T-cells that can transfer tolerance to naive hosts. High dose oral tolerance is oral tolerance induced by high doses of antigens, is insensitive to

cyclophosphamide treatment, and proceeds to induction of T cell hyporesponsiveness via anergy and/or deletion of antigen specific T-cells. The difference in sensitivity to cyclophosphamide can be used to make a distinction between low dose and high dose tolerance. Strobel et al., Immunology 1983, 49:451-456. An exemplary oral tolerance is low dose oral tolerance as described in Mayer and Shao, Nature Rev. Immunol. 2004, 4:407-419.

Immuno-Modulating Compound

[0092] In some embodiments, the present disclosure provides methods for the treatment of T1D, in which the subject is not concomitantly treated with an additional immuno- modulating compound (i.e., in addition to IL-2). Thus the subject is treated with the T1D- specific antigen and the IL-2 alone. [0093] In some embodiments, the present disclosure provides methods for the treatment of T1D, in which the subject is concomitantly treated with an additional immuno-modulating compound. Thus the subject is treated with the TID-specific antigen, the IL-2, and the additional immune-modulating compound.

[0094] The terms "immuno-modulating compound" or immuno-modulator" are used herein in accordance with their art-recognized meaning. The immuno-modulating compound can be any immuno-modulating compound known to a person skilled in the art. A skilled person in the art may opt to include or not include an immune-modulating compound in the treatment described herein. The decision to include an immune-modulating compound in a treatment regimen can be determined by the performance of the treatment described herein, a subject's genetic traits, and/or physiological conditions, among other factors.

[0095] In some embodiments, the immuno-modulating compound is a tolerance inducing compound. Tolerance induction can be obtained, e.g., by inducing regulatory T-cells, or in an indirect way, e.g., by activation of immature dendritic cells to make tolerant dendritic cells and/or inhibiting Th2 immune response inducing expression of "co-stimulation" factors on mature dendritic cells. Immuno-modulating and immuno-suppressing compounds are known to the person skilled in the art and include, but are not limited to, bacterial metabolites such as spergualin, fungal and streptomycal metabolites such as tacrolimus or cyclosporin, immuno-suppressing cytokines such as IL-4, IL-10, IFNa, TGFp (as selective adjuvant for regulatory T-cells) Flt3L, TSLP and Rank-L (as selective tolerogenic DC inducers), antibodies and/or antagonist such as anti-CD40L, anti-CD25, anti-CD20, anti-IgE, anti-CD3, and proteins, peptides or fusion proteins such as the CTL-41 g or CTLA-4 agonist fusion protein. In some embodiments, the immuno-modulating compound is an immuno- suppressing compound. The immuno-suppressing compound can be an immuno-suppressing cytokine or antibody. In other embodiments, the immuno-suppressing cytokine is a tolerance-enhancing cytokine or antibody. It will be appreciated by the person skilled in the art that the term "immuno-modulating compound" also includes functional homologues thereof. A functional homologue is a molecule having essentially the same or similar function for the intended purposes, but can differ structurally. In some examples, the immuno-modulating compound is anti-CD3, or a functional homologue thereof. LAB

[0096] The present disclosure relates to the use of genetically modified lactic acid fermenting bacteria (LAB). The LAB strain can be a Lactococcus species, a Lactobacillus species, a Bifidobacterium species, a Streptococcus species, or an Enterococcus species.

[0097] As used herein, Lactococcus or Lactobacillus is not limited to a particular species or subspecies, but meant to include any of the Lactococcus or Lactobacillus species or subspecies. Exemplary Lactococcus species include Lactococcus garvieae, Lactococcus lactis, Lactococcus piscium, Lactococcus plantarum, and Lactococcus raffmolactis. In some examples, the Lactococcus lactis is Lactococcus lactis subsp. cremoris, Lactococcus lactis subsp. hordniae, or Lactococcus lactis subsp. lactis.

[0098] Exemplary Lactobacillus species include Lactobacillus acetotolerans,

Lactobacillus acidophilus, Lactobacillus agilis, Lactobacillus algidus, Lactobacillus alimentarius, Lactobacillus amylolyticus, Lactobacillus amylophilus, Lactobacillus amylovorus, Lactobacillus animalis, Lactobacillus aviarius, Lactobacillus aviarius subsp. araffinosus, Lactobacillus aviarius subsp. aviarius, Lactobacillus bavariciis, Lactobacillus bifermentans, Lactobacillus brevis, Lactobacillus buchneri, Lactobacillus bulgaricus, Lactobacillus carnis, Lactobacillus casei, Lactobacillus casei subsp. alactosus, Lactobacillus casei subsp. casei, Lactobacillus casei subsp. pseudoplantarum, Lactobacillus casei subsp. rhamnosus, Lactobacillus casei subsp. tolerans, Lactobacillus catenaformis, Lactobacillus cellobiosus, Lactobacillus collinoides, Lactobacillus confusus, Lactobacillus coryniformis, Lactobacillus coryniformis subsp. coryniformis, Lactobacillus coryniformis subsp. torquens, Lactobacillus crispatus, Lactobacillus curvatus, Lactobacillus curvatus subsp. curvatus, Lactobacillus curvatus subsp. melibiosus, Lactobacillus delbrueckii, Lactobacillus delbrueckii subsp. bulgaricus, Lactobacillus delbrueckii subsp. delbrueckii, Lactobacillus delbrueckii subsp. lactis, Lactobacillus divergens, Lactobacillus farciminis, Lactobacillus fermentum, Lactobacillus fornicalis, Lactobacillus fructivorans, Lactobacillus fructosus, Lactobacillus gallinarum, Lactobacillus gasseri, Lactobacillus graminis, Lactobacillus halotolerans, Lactobacillus hamsteri, Lactobacillus helveticus, Lactobacillus heterohiochii, Lactobacillus hilgardii, Lactobacillus homohiochii, Lactobacillus iners, Lactobacillus intestinalis, Lactobacillus jensenii, Lactobacillus johnsonii, Lactobacillus kandleri, Lactobacillus kefiri, Lactobacillus kefiranofaciens, Lactobacillus kefirgranum, Lactobacillus kunkeei, Lactobacillus lactis, Lactobacillus leichmannii, Lactobacillus lindneri,

Lactobacillus malefermentans, Lactobacillus mali, Lactobacillus maltaromicus, Lactobacillus manihotivorans, Lactobacillus minor, Lactobacillus minutus, Lactobacillus mucosae, Lactobacillus murinus, Lactobacillus nagelii, Lactobacillus oris, Lactobacillus panis, Lactobacillus parabuchneri, Lactobacillus paracasei, Lactobacillus paracasei subsp. paracasei, Lactobacillus paracasei subsp. tolerans, Lactobacillus parakefiri, Lactobacillus paralimentarius, Lactobacillus paraplantarum, Lactobacillus pentosus, Lactobacillus perolens, Lactobacillus piscicola, Lactobacillus plantarum, Lactobacillus pontis,

Lactobacillus reuteri, Lactobacillus rhamnosus, Lactobacillus rimae, Lactobacillus rogosae, Lactobacillus ruminis, Lactobacillus sakei, Lactobacillus sakei subsp. camosus,

Lactobacillus sakei subsp. sakei, Lactobacillus salivarius, Lactobacillus salivarius subsp. salicinius, Lactobacillus salivarius subsp. salivarius, Lactobacillus sanfranciscensis, Lactobacillus sharpeae, Lactobacillus suebicus, Lactobacillus trichodes, Lactobacillus uli, Lactobacillus vaccinostercus, Lactobacillus vaginalis, Lactobacillus viridescens,

Lactobacillus vitulinus, Lactobacillus xylosus, Lactobacillus yamanashiensis, Lactobacillus yamanashiensis subsp. mali, Lactobacillus yamanashiensis subsp. Yamanashiensis,

Lactobacillus zeae, Bifidobacterium adolescentis, Bifidobacterium angulatum,

Bifidobacterium bifidum, Bifidobacterium breve, Bifidobacterium catenulatum,

Bifidobacterium longum, and Bifidobacterium infantis. In some examples, the LAB is Lactococcus lactis (LL).

[0099] In further examples, the bacterium is selected from the group consisting of Enterococcus alcedinis, Enterococcus aquimarinus, Enterococcus asini, Enterococcus avium, Enterococcus caccae, Enterococcus camelliae, Enterococcus canintestini, Enterococcus canis, Enterococcus casseliflavus, Enterococcus cecorum, Enterococcus columbae,

Enterococcus devriesei, Enterococcus diestrammenae, Enterococcus dispar, Enterococcus durans, Enterococcus eurekensis, Enterococcus faecalis, Enterococcus faecium,

Enterococcus gallinarum, Enterococcus gilvus, Enterococcus haemoperoxidus, Enterococcus hermanniensis, Enterococcus hirae, Enterococcus italicus, Enterococcus lactis, Enterococcus lemanii, Enterococcus malodoratus, Enterococcus moraviensis, Enterococcus mundtii, Enterococcus olivae, Enterococcus pallens, Enterococcus phoeniculicola, Enterococcus plantarum, Enterococcus pseudoavium, Enterococcus quebecensis, Enterococcus raffinosus, Enterococcus ratti, Enterococcus rivorum, Enterococcus rotai, Enterococcus

saccharolyticus, Enterococcus silesiacus, Enterococcus solitarius, Enterococcus sulfureus, Enterococcus termitis, Enterococcus thailandicus, Enterococcus ureasiticus, Enterococcus ureilyticus, Enterococcus viikkiensis, Enterococcus villorum, and Enterococcus

xiangfangensis,

[00100] In further examples, the bacterium is selected from the group consisting of Streptococcus agalactiae, Streptococcus anginosus, Streptococcus bovis, Streptococcus canis, Streptococcus constellatus, Streptococcus dysgalactiae, Streptococcus equinus, Streptococcus iniae, Streptococcus intermedius, Streptococcus milleri, Streptococcus mitis, Streptococcus mutatis, Streptococcus oralis, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus pseudopneumoniae, Streptococcus pyogenes, Streptococcus ratti, Streptococcus salivarius, Streptococcus tigurinus,

Streptococcus thermophilus, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus suis, Streptococcus uberis, Streptococcus vestibularis, Streptococcus viridans, and

Streptococcus zooepidemicus.

[00101] The exemplary LAB strain may be Lactococcus lactis or any of its subspecies, including Lactococcus lactis subsp. cremoris, Lactococcus lactis subsp. hordniae,

Lactococcus lactis and Lactococcus lactis subsp. lactis. In another aspect, the LAB strain may be a biologically contained system, such as the plasmid free Lactococcus lactis strain MG1363, that lost the ability of normal growth and acid production in milk as described in Gasson, MJ. (1983) J. Bacterid. 154: 1-9; or the threonine- and pyrimidine-auxotroph derivative L. lactis strains as described in Sorensen et al. (2000) Appl. Environ. Microbiol. 66: 1253-1258; and Glenting et al. (2002) 68:5051 -5056.

[00102] The recombinant bacterial host-vector system can be a biologically contained system. Biological containment is known to the person skilled in the art and can be realized by the introduction of an auxotrophic mutation, for example, a suicidal auxotrophic mutation such as the ThyA mutation, or its equivalents. Alternatively, the biological containment can be realized at the level of the plasmid carrying the gene encoding the IL-2 polypeptide or IL- 2 variant, such as, for example, by using an unstable episomal construct, which is lost after a few generations. Several levels of containment, such as plasmid instability and auxotrophy, can be combined to ensure a high level of containment, if desired.

Constructs

[00103] As described herein, the LAB delivers the IL-2 polypeptide and the TID-specific antigen at the intended site, i.e., the mucosa. For example, the LAB expresses the IL-2 polypeptide, after which the IL-2 polypeptide is exposed on the cell surface (if a membrane- bound form of IL-2 is used) or secreted (if a secreted form of IL-2 is used). Hence, in a particular embodiment the LAB, such as L. lactis, comprises an expression vector capable of expressing the IL-2 polypeptide and the T ID-specific antigen, intracellularly. For example, the polypeptides is exposed on the cell surface under conditions present at the intended mucosa, e.g., in the gastrointestinal tract. The LAB can comprise expression vectors capable of expressing the IL-2 polypeptide intracellularly, such that the IL-2 polypeptide is exposed on the cell surface to a degree sufficient to provide a low-dose of IL-2 that is effective in treating T1D in the recipient. When using LAB strains expressing higher amounts of IL-2 polypeptide and T ID-specific antigen, less frequent and lower LAB doses may be required for the treatment of T1D. Thus, one of skill in the art may adjust the amount of LAB strains provided to deliver the desired amount of IL-2 polypeptide and TlD-specific antigen.

[00104] Usually, the expression system will comprise a genetic construct comprising at least one nucleotide sequence encoding an IL-2 polypeptide and/or a TD1 -specific antigen polypeptide, typically operably linked to a promoter capable of directing expression of the sequence(s) in the hosting microorganism. Suitably the IL-2 polypeptide and the TlD- specific antigen to be expressed can be encoded by a nucleic acid sequence that is adapted to the preferred codon usage of the host. The construct may further contain (all) other suitable element(s), including enhancers, transcription initiation sequences, signal sequences, reporter genes, transcription termination sequences, etc., operable in the selected host, as is known to the person skilled in the art.

[00105] The construct is typically in a form suitable for transformation of the host and/or in a form that can be stably maintained in the host, such as a vector, plasmid or mini- chromosome. Suitable vectors comprising nucleic acid for introduction into LAB strains, e.g., L. lactis, can be chosen or constructed, containing appropriate regulatory sequences, including promoter sequences, terminator fragments, enhancer sequences, marker genes and other sequences as appropriate. Vectors may be plasmids, viral e.g. , phage, or phagemid, as appropriate. Further details can be found in, for example, Molecular Cloning: a Laboratory Manual 2nd edition, Sambrook et al., 1989, Cold Spring Harbor Laboratory Press.

[00106] Many known techniques and protocols for manipulation of nucleic acid, for example in preparation of nucleic acid constructs, mutagenesis, sequencing, introduction of DNA into cells and gene expression, and analysis of proteins, are described in detail in Short Protocols in Molecular Biology, Second Edition, Ausubel et al, eds., John Wiley & Sons, 1992. In one embodiment, the coding sequence for the IL-2 polypeptide can be contained in an operon, i.e., a nucleic acid construct for multi-cistronic expression. In an operon, transcription from the promoter results in a mRNA which comprises more than one coding sequence, each with its own suitably positioned ribosome binding site upstream. Thus, more than one polypeptide can be translated from a single mRNA. Use of an operon enables expression of the IL-2 polypeptide and TlD-specific antigen polypeptide to be coordinated. Polycistronic expression systems in bacterial host cells are described, e.g., in U.S. Patent Application No. 2014/0105863.

[00107] To obtain stably transfected LAB strains, i.e., the gene coding for the IL-2 polypeptide and/or the TlD-specific antigen gene can be integrated into the host LAB's genome. Techniques for establishing stably transfected LAB strains are known in the art. For instance, the IL-2 polypeptide and/or the TlD-specific antigen gene may be cloned into the host's genome via homologous recombination. Typically, an essential gene of the host is disrupted by the homologous recombination event, such as deletion of the gene, one or more amino acid substitutions leading to an inactive form of the protein encoded by the essential gene, or to a frameshift mutation resulting in a truncated form of the protein encoded by the essential gene. In an embodiment, the essential gene is the thyA gene. An exemplary technique is described in WO 02/090551. The transforming plasmid is not particularly limited, as long as it cannot complement the disrupted essential gene, e.g., thyA gene. The plasmid may be a self-replicating, typically carrying one or more genes of interest and one or more resistance markers, or the plasmid is an integrative plasmid. In the latter case, the integrative plasmid itself may be used to disrupt the essential gene, by causing integration at the locus of the essential gene, e.g. , thyA site, because of which the function of the essential gene, e.g., the thyA gene, is disrupted. Typically, the essential gene, such as the thyA gene, is replaced by double homologous recombination by a cassette comprising the gene or genes of interest, flanked by targeting sequences that target the insertion to the essential gene, such as the thyA target site. It will be appreciated that that these targeting sequences are sufficiently long and sufficiently homologous to enable integration of the gene of interest into the target site.

[00108] The genetic construct encoding the IL-2 polypeptide and or the TlD-specific antigen may thus be present in the host cell extra-chromosomally, typically autonomously replicating using an own origin of replication, or may be integrated into the LAB genomic DNA, e.g., Lactococcus chromosome. In the latter case, a single or multiple copies of the nucleic acid may be integrated; the integration may occur at a random site of the chromosome or, as described above, at a predetermined site thereof, for example, in the thyA locus of Lactococcus, e.g., Lactococcus lactis.

[00109] Hence, the genetic construct encoding the IL-2 polypeptide and/or the T1D- specific antigen may further comprise sequences configured to effect insertion of the genetic construct into the genome, e.g. , a chromosome, of a host LAB cell.

[00110] In an example, insertion of the genetic construct into particular sites within a genome, e.g., chromosome, of a host LAB cell may be facilitated by homologous recombination. For instance, the genetic constructs described herein may comprise one or more regions of homology to the said site of integration within the genome e.g. , a chromosome, of the host LAB cell. The sequence at the said genome, e.g., chromosome, site may be natural, i.e. , as occurring in nature, or may be an exogenous sequence introduced by previous genetic engineering.

[00111] For instance, the region(s) of homology may be at least 50 bp, 100 bp, 200 bp, 300 bp, 400 bp, 500 bp, 600 bp 700 bp, 800 bp, 900 bp, 1000 bp, or more.

[00112] In one example, two regions of homology may be included, one flanking each side of the relevant expression units present in the genetic constructs described herein. Such configuration may advantageously insert the relevant sequences, . e. , at least the ones encoding and effecting the expression of the antigen of interest, in host cells. Ways of performing homologous recombination, especially in bacterial hosts, and selecting for recombinants, are generally known in the art.

[00113] Transformation methods of LAB strains are known to the person skilled in the art, for example, protoplast transformation and electroporation.

[00114] A high degree of expression can be achieved by using homologous expression and/or secretion signals on the expression vectors present in the LAB, e.g., L. lactis.

Expression signals will be apparent to the person skilled in the art. The expression vector can be optimized for expression depending on the LAB, e.g. , L. lactis, it is incorporated in. For instance, specific expression vectors that gave sufficient levels of expression in Lactococcus, Lactobacillus lactis, casei and plantarum are known. Moreover, systems are known which have been developed for the expression of heterologous antigens in the non-pathogenic, non- colonizing, non- invasive food-grade bacterium Lactococcus lactis {e.g., UK patent

GB2278358B). An exemplary construct comprises the multi-copy expression vector described in PCT/NL95/00135 (WO 96/32487), in which the nucleotide sequence encoding the IL-2 polypeptide T ID-specific antigen has been described. Such a construct may be suitable for expression of a desired antigen in a lactic acid bacterium, in particular in a Lactobacillus, at a high level of expression, and also can be used advantageously to direct the expressed product to the surface of the bacterial cell. The constructs (e.g. , of

PCT/NL95/00135) may be characterized in that the nucleic acid sequence encoding the IL-2 polypeptide and/or TID-specific antigen is preceded by a 5' non-translated nucleic acid sequence comprising at least the minimal sequence required for ribosome recognition and RNA stabilization. This can be followed by a translation initiation codon which may be (immediately) followed by a fragment of at least 5 codons of the 5' terminal part of the translated nucleic acid sequence of a gene of a lactic acid bacterium or a structural or functional equivalent of the fragment. The fragment may also be controlled by the promoter. One aspect of the present disclosure provides a method which permits the high level regulated expression of heterologous genes in the host and the coupling of expression to secretion. In another embodiment, the T7 bacteriophage RNA polymerase and its cognate promoter are used to develop a powerful expression system according to WO 93/171 17. In one embodiment, the expression plasmid may be derived from pTl NX.

[00115] A promoter employed herein is typically expressed constitutively in the bacterium. The use of a constitutive promoter avoids the need to supply an inducer or other regulatory signal for expression to take place. Typically, the promoter directs expression at a level at which the bacterial host cell remains viable, i.e., retains some metabolic activity, even if growth is not maintained. Advantageously then, such expression may be at a low level. For example, where the expression product accumulates intracellularly, the level of expression may lead to accumulation of the expression product at less than about 10% of cellular protein, optionally about or less than about 5%, for example about 1-3%. The promoter may be homologous to the bacterium employed, i.e., one found in that bacterium in nature. For example, a Lactococcal promoter may be used in a Lactococcus. An exemplary promoter for use in Lactococcus lactis (or other Lactococci) is "PI" derived from the chromosome of Lactococcus lactis (Waterfield NR et al., Gene 1995, 165(1):9-15). Another example of a promoter is the usp45 promoter. Other useful promoters are described in U.S. Patent 8,759,088 and U.S. Patent Application No. 2014/0105863.

[00116] The nucleic acid construct or constructs may comprise a secretory signal sequence. Thus, in some embodiments the nucleic acid encoding IL-2 and/or the TID- specific antigen may provide for secretion of the polypeptides, e.g. , by appropriately coupling a nucleic acid sequence encoding a signal sequence to the nucleic acid sequence encoding the polypeptide). Ability of a bacterium harboring the nucleic acid to secrete the antigen may be tested in vitro in culture conditions which maintain viability of the organism. Exemplary secretory signal sequences include those with activity in LAB strains. Such sequences may include the a-amylase secretion leader of Bacillus amyloliquetaciens or the secretion leader of the Staphylokinase enzyme secreted by some strains of Staphylococcus, which is known to function in both Gram-positive and Gram-negative hosts (Rapoport, Current Opinion in Biotechnology 1990, 1 : 21-27), or leader sequences from numerous other Bacillus enzymes or S-layer proteins {see pp 341-344 of Harwood and Cutting, "Molecular Biological Methods for Bacillus " John Wiley & Co. 1990). In one embodiment, said secretion signal is derived from usp45 (Van Asseldonk et al. (1993) Mol. Gen. Genet. 240:428-434). In some embodiments, the IL-2 polypeptide or IL-2 variant may be constitutively secreted.

IL-2 Polypeptides

[00117] Examples of IL-2 polypeptides include wild-type human IL-2 in either membrane bound or secreted forms, and any IL-2 variant polypeptide, e.g., polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 98% sequence identity with wild-type IL-2, or the corresponding mature IL-2 polypeptide. An exemplary amino acid sequence of wild-type human IL-2 is represented by SEQ ID NO: 1 , while an exemplary IL-2 encoding nucleic acid sequence is represented by SEQ ID NO: 2. Mature wild-type human IL-2 is represented by SEQ ID NO: 3.

[00118] The signal peptide for IL-2 (SEQ ID NO: 4) is underlined and represents amino acids 1-20 of SEQ ID NO: 1. The signal peptide of IL-2 may be substituted with a bacterial secretory signal sequence {e.g., SSusp45) as described herein. An exemplary nucleotide sequence according to this embodiment is represented by SEQ ID NO: 5.

[00119] The term "IL-2 variant" includes IL-2 polypeptides characterized by amino acid insertions, deletions, substitutions, and/or modifications at one or more sites of the native IL- 2 polypeptide chain. In accordance with this disclosure any such insertions, deletions, substitutions, and modifications result in an IL-2 variant polypeptide that retains at least some IL-2RP binding activity. Exemplary variants include polypeptides with substitutions of 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10 or more amino acids. IL-2 variants can have conservative modifications and substitutions at other positions of IL-2 {i.e., those that have a minimal effect on the secondary or tertiary structure of the variant polypeptide). Such conservative substitutions include those described by Dayhoff in 'The Atlas of Protein Sequence and Structure 5' (1978), and by Argos in EMBO J., 8:779-785 (1989). For example, amino acids belonging to one of the following groups represent conservative changes: Group I: ala, pro, gly, gin, asn, ser, thr; Group II: cys, ser, tyr, thr; Group II: val, ile, leu, met, ala, phe; Group IV: lys, arg, his; Group V: phe, tyr, trp, his; and Group VI: asp, glu.

[00120] In some examples, the IL-2 is a variant as described in U.S. Pat. No. 4,518,584, in which the cysteine normally occurring at position 125 of the wild-type or native molecular has been replaced by a neutral amino acid such as serine or alanine. Alternatively or conjunctively, the IL-2 variant may be one as described in U.S. application No. 06/810,656 filed Dec. 17, 1985, in which the methionine normally occurring at position 104 of the wild- type or native molecule has been replaced by a neutral amino acid such as alanine. In some examples, the IL-2 variant may have one or more of the first five N-terminal amino acids of the native IL 2 deleted. IL-2 muteins were also generated with decreased binding affinity to CD122 (to achieve lower IL-2 toxicity), such as BAY 50-4798 (containing an N88R mutation of IL 2).

[00121] Other forms of IL-2 that may be used include IL-2 variant sequences such as those found in aldesleukin, or proleukin (Prometheus Laboratories), teceleukin (Roche), bioleukin (Glaxo), as well as variants as described in Taniguchi et al, Nature 1983, 302(5906):305-10 and Devos et al, Nucleic Acids Res. 1983, 11(13): 4307-23; European Patent Application Nos. 91 ,539 and 88,195; U.S. Pat. No. 4,518,584. U.S. Patent Publication No. 2012/0244112; U.S. Patent No. 7,569,215; U.S. Patent No. 5,229,109; U.S. Patent Publication No.

2006/0269515; EP Patent Publication No. EP 1730184A2; and PCT Publication WO

2005/086751.

[00122] In some embodiments, the IL-2 variant has diminished capacity to bind to the high-affinity IL-2 receptor, but preserves affinity of the variant IL-2 to bind intermediate- affinity IL-2 receptor compared to wild-type IL-2 polypeptide. In some embodiments, the mature IL-2 polypeptide is characterized by one, two, or three amino acid substitutions, e.g., wherein the substituted amino acid residues are selected from L72, F42, and Y45. In some embodiments, the IL-2 variant is characterized by a substitution of L72, e.g., comprises a first amino acid substitution selected from the group consisting of L72G, L72A, L72S, L72T, L72Q, L72E, L72N, L72D, L72R, and L72K. The IL-2 variant can be characterized by a substitution of F42, e.g., comprises a second amino acid substitution selected from the group consisting of F42A, F42G, F42S, F42T, F42Q, F42E, F42N, F42D, F42R, and F42K. In further embodiments, the IL-2 variant is characterized by a substitution of Y45, e.g., comprises a third amino acid substitution selected from the group consisting of Y45A, Y45G, Y45S, Y45T, Y45Q, Y45E, Y45N, Y45D, Y45R, and Y45K. The IL-2 variant of the present disclosure may contain any combination of the above recited first, second, and third amino acid substitutions.

[00123] The IL-2 variants as described herein may be about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99% identical to a corresponding wild type IL-2, provided that the IL-2 variant polypeptide retains some IL-2 activity (functional polypeptide).

[00124] The percentage identity of polypeptide sequences can be calculated using commercially available algorithms which compare a reference sequence (e.g., SEQ ID NO: 1 of the present disclosure) with a query sequence.

[00125] A person of ordinary skill in the art will appreciate that the optimal amount of IL-2 to be delivered to the subject using the methods of the present disclosure varies, e.g. , with the LAB expressing the IL-2 polypeptide, and the genetic construct, e.g. , the strength of the promoter used in the genetic construct. Typically, the LAB may be administered in an amount equivalent to a particular amount of expressed IL-2 polypeptide, or in an amount, which generates a desired PK profile for the respective IL-2 polypeptide in the respective subject. Exemplary daily IL-2 polypeptide doses are from about 10 fg to about 100 μg of active polypeptide per day. Other exemplary dose ranges are from about 1 pg to about 100 μg per day; or from about 1 ng to about 100 μg per day.

[00126] The above doses may be realized by administering to the subject effective amounts of the microorganism per day, wherein the microorganism is adapted to express a sufficient amount of IL-2 to realize the desired dose, such as those above. The LAB secreting the IL-2 polypeptide may be delivered in a dose of from about 10 4 colony forming units (cfu) to about 10 12 cfu per day, in particular from about 10 6 cfu to about 10 12 cfu per day, more in particular from about 10 9 cfu to about 10 12 cfu per day. The amount of secreted IL-2 polypeptide can be determined based on cfu, for example in accordance with the methods described in Steidler ei a/., Science 2000; 289(5483): 1352-1355, or by using ELISA. For example, a LAB may secrete at least about 1 ng to about 1 μg of active polypeptide per 10 9 cfu. Based thereon, the skilled person can calculate the range of IL-2 polypeptide secreted at other cfu doses. [00127] Each of the above doses/dose ranges may be administered in connection with any dosing regimen as described herein. The daily dose may be administered in 1 , 2, 3, 4, 5, or 6 portions throughout the day. Further the daily doses may be administered for any number of days, with any number of rest periods between administration periods. For example, the subject may be administered microorganism at a dose equivalent to about 0.1 to about 3 MlU/day or every other day, for a period of at least about 1 week, at least about 2 weeks, at least about 3 weeks, at least about 4 weeks, at least about 5 weeks, or at least about 6 weeks. In some examples, the subject is administered the LAB at a dose equivalent to about 0.1 to about 5 MlU/day, or about 0.3 to about 3 MIU, e.g., for about 5 days, about 7 days, or about 14 days. Exemplary doses are described, e.g., in Hartemann et al., Lancet Diabetes

Endocrinol. 2013, 1 (4): 295-305.

TID-Speciflc Antigen Polypeptides

[00128] The LAB of the present disclosure contains at least one disease-specific {i.e., TlD-specific) self-antigen gene, and can express such gene under conditions sufficient for expression. Exemplary T ID-specific self-antigens include islet antigens associated with the beta-cell destruction process. Examples include but are not limited to: proinsulin (PINS), glutamic acid decarboxylase (GAD65), insulinoma-associated protein 2 (IA-2), islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP) and zinc transporter 8 (ZnT8) 8. Other examples include molecules expressed by beta beta-cells, such as chromogranin A, (prepro) islet amyloid polypeptide (ppIAPP), peripherin and citrullinated glucose-regulated protein (GRP).

[00129] Examples of PINS polypeptides include wild-type human PINS and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type human PINS. An exemplary amino acid sequence of wild-type human PINS is represented by SEQ ID NO: 6, while an exemplary PINS encoding nucleic acid sequence is represented by SEQ ID NO: 7 {see CDS contained in accession number NM_000207.2).

[00130] Additional exemplary PINS nucleotide sequences are represented by the coding sequences of NCBI accession numbers AY899304 (complete CDS, alternatively spliced; SEQ ID NO: 8); NM_000207 (transcript variant 1 ; SEQ ID NO: 9); NM_001185097 (transcript variant 2; SEQ ID NO: 10); NM_001 185098 (transcript variant 3; SEQ ID NO: 11); NMJ301291897 (transcript variant 4; SEQ ID NO: 12), and partial functional sequences thereof. Exemplary PINS amino acid sequences include those encoded by any one of the above PINS nucleic acid sequences.

[00131] Any nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 6, or any nucleotide sequence encoding at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, or at least about 100 consecutive amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 6 may be used.

[00132] Additional PINS polypeptides are described, e.g., in UniProtKB - P01308 and links therein. In some examples, the PINS polypeptide is represented by amino acid residues 25-1 10 (numbering according to SEQ ID NO: 6).

[00133] Exemplary GAD (e.g., GAD65) polypeptides include wild-type human GAD65, and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type GAD65. An exemplary amino acid sequence of wild-type human GAD65 is represented by SEQ ID NO: 13, while an exemplary GAD65 encoding nucleic acid sequence is represented by SEQ ID NO: 14 (see, e.g., CDS contained in accession number M81882.1).

[00134] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 13, or any nucleotide sequence encoding at least about 100, at least about 200, at least about 300, at least about 400, or at least about 500 consecutive amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 13 may be used.

[00135] Other exemplary glutamate decarboxylase (e.g., GAD65) sequences are described, e.g., in UniProtKB - Q05329 and links therein. In some example, the GAD polypeptide is a trimmed variant containing less than about 500, less than about 400, or less than about 300 of the wild-type amino acids. Exemplary polypeptide fragments (trimmed GAD65 variants) are described, e.g., in Robert et al., Benef. Microbes 2015, 6(4): 591-601. In some examples, the trimmed GAD variants are efficiently expressed and secreted by a LAB (i.e., Lactococcus lactis). An exemplary trimmed GAD variant is GAD65 3 7 0 -575 (amino acid numbering relative to NCBI accession number P_000809.1 , i.e., SEQ ID NO: 13).

[00136] Other exemplary GAD nucleotide sequences are represented by NCBI accession numbers M81882 (GAD65; SEQ ID NO: 15); M81883 (GAD67; SEQ ID NO: 16);

NM_000818 (GAD2 variant 1 ; SEQ ID NO: 17); and NM_001 134366 (GAD2 variant 2; SEQ ID NO: 18); and open reading frames (CDS) contained therein. Exemplary amino acid sequences include sequences encoded by the above nucleotide sequences of accession numbers M81882, M81883, NM_001 134366, and NM_000818.

[00137] Examples of IA-2 polypeptides include wild-type human IA-2 and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%), at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type IA-2. An exemplary amino acid sequence of wild-type human IA-2 is represented by SEQ ID NO: 19, while an exemplary IA-2 encoding nucleic acid sequence is represented by SEQ ID NO: 20 (see, e.g., open reading frame of accession number NM_002846.3).

[00138] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 19, or any nucleotide sequence encoding at least about 100, at least about 200, at least about 300, at least about 400, at least about 500, at least about 600, or at least about 800 consecutive amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 19 may be used.

[00139] Exemplary IA-2 nucleotide sequences are represented by NCBI accession numbers NM_002846 (human IA-2 or protein tyrosine phosphatase, receptor type N

(PTPRN), transcript variant 1 ; SEQ ID NO: 21); NM_001199763 (Human IA-2 or protein tyrosine phosphatase, receptor type, N (PTPRN), transcript variant 2; SEQ ID NO: 22); NM_001 199764 (Human IA-2 or protein tyrosine phosphatase, receptor type, N (PTPRN), transcript variant 3; SEQ ID NO: 23). Exemplary IA-2 amino acid sequences include those encoded by the above nucleotide sequences.

[00140] Other exemplary IA-2 sequences are described, e.g., in UniProtKB - Q16849 and links therein. In some example, the IA-2 polypeptide can be a trimmed variant containing less than about 700, less than about 600, less than about 500, or less than about 400 of the wild- type amino acids. Exemplary polypeptide fragments (trimmed IA-2 variants) are described, e.g., in Robert et al, Benef. Microbes 2015, 6(4): 591-601. In some examples, the trimmed IA-2 variants can be efficiently expressed and secreted by a LAB {i.e., Lactococcus lactis). In one example, the trimmed IA-2 variant is IA-2635.979 (amino acid numbering relative to NCBI accession number NP 002837.1 ; i.e., SEQ ID NO: 19).

[00141] Examples of IGRP polypeptides include wild-type human IGRP, and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type IGRP. An exemplary amino acid sequence of wild- type human IGRP is represented by SEQ ID NO: 24, while an exemplary IGRP encoding nucleic acid sequence is represented by SEQ ID NO: 25 (see open reading frame of NCBI accession number BC 113376.1).

[00142] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 24, or any nucleotide sequence encoding at least 50, at least 100, at least 200, or at least 300 amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 24 may be used.

[00143] Further exemplary nucleotide sequences are represented by NCBI accession numbers NM_021176 (G6PC2 transcript variant 1 ; SEQ ID NO: 26); NM_001081686 (human glucose-6-phosphatase, catalytic, 2 (G6PC2) transcript variant 2; SEQ ID NO: 27); and NM_001270397 (G6PC, transcript variant 2; SEQ ID NO: 28). Exemplary IGRP amino acid sequences include those encoded by the above nucleotide sequences.

[00144] Other exemplary sequences are described, e.g., in UniProtKB - Q9NQR9 and links therein, as well as in Arden et al, Diabetes 1999; 48(3):531-542; Martin et al, J. Biol. Chem. 2001 ; 276(27):25197-207; and Dogra et al, Diabetologia 2006; 49(5):953-7. In some examples, the IGRP polypeptide is a trimmed variant containing less than about 300, less than about 200, less than about 100, or less than about 50 of the wild-type amino acids. In some examples, the trimmed IGRP variants are selected to be efficiently expressed and secreted by a LAB (i.e., Lactococcus lactis). [00145] Examples of ZnT8 polypeptides include wild-type human ZnT8, and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type ZnT8. An exemplary amino acid sequence of wild- type human ZnT8 is represented by SEQ ID NO: 29, while an exemplary ZnT8 encoding nucleic acid sequence is represented by SEQ ID NO: 30 (see, e.g., open reading frame contained in NCBI accession number NM_173851.2).

[00146] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 29, or any nucleotide sequence encoding at least 50, at least 100, at least 200, at least 250, or at least 300 amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 29 may be used.

[00147] Further exemplary ZnT8 nucleotide sequences are represented by NCBI accession numbers AY212919.1 (human zinc transporter 8, complete cds; SEQ ID NO: 31);

NM_173851.2 (human zinc transporter 8, transcript variant 1 ; SEQ ID NO: 32);

NM_001 172814.1 (human zinc transporter 8, transcript variant 2; SEQ ID NO: 33);

NM_001 172811.1 (human zinc transporter 8, transcript variant 3; SEQ ID NO: 34);

NM_001172813.1 (human zinc transporter 8, transcript variant 4; SEQ ID NO: 35);

NM_001172815.2 (human zinc transporter 8, transcript variant 5; SEQ ID NO: 36), and partial sequences thereof. Exemplary ZnT8 amino acid sequences include those encoded by the above nucleotide sequences.

[00148] Other exemplary sequences are described, e.g., in UniProtKB - Q8IWU4 and links therein. In some examples, the ZnT8 polypeptide is a trimmed variant containing less than about 300, less than about 200, or less than about 100 of the wild-type amino acids. In some examples, the trimmed ZnT8 variants are selected to be efficiently expressed and secreted by a LAB (i.e., Lactococcus lactis).

[00149] Examples of ppIAPP polypeptides include wild-type human ppIAPP, and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type ppIAPP. An exemplary amino acid sequence of wild-type human ppIAPP is represented by SEQ ID NO: 37, while an exemplary ppIAPP encoding nucleic acid sequence is represented by SEQ ID NO: 38 (see, e.g., open reading frame of NCBI accession number NM_000415.2).

[00150] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 37, or any nucleotide sequence encoding at least 50, at least 100, at least 200, at least 250, or at least 300 amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 37 may be used.

[00151] Other exemplary ppIAPP polypeptide sequences are disclosed, e.g., in UniProtKB - PI 0997 and links therein. In some examples, the ppIAPP polypeptide can be a trimmed variant containing less than about 80, less than about 60, less than about 40, or less than about 20 of the wild-type amino acids. In some examples, the trimmed ppIAPP variants are selected to be efficiently expressed and secreted by a LAB strain (i.e., Lactococcus lactis).

[00152] Examples of peripherin polypeptides include wild-type human peripherin, and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type. An exemplary amino acid sequence of wild-type human peripherin is represented by SEQ ID NO: 39, while an exemplary peripherin encoding nucleic acid sequence is represented by SEQ ID NO: 40 (see, e.g., open reading frame of NCBI accession number NM_006262.3).

[00153] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 39, or any nucleotide sequence encoding at least about 100, at least about 200, at least about 300, or at least about 400 consecutive amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 39 may be used.

[00154] Other exemplary peripherin sequences are disclosed, e.g., in UniProtKB - P41219 and links therein. In some examples, the peripherin polypeptide is a trimmed variant containing less than about 400, less than about 300, less than about 200, or less than about 100 of the wild-type amino acids. In some examples, the trimmed peripherin variants are selected to be efficiently expressed and secreted by a LAB (i.e., Lactococcus lactis). [00155] Further exemplary nucleotide sequences are represented by NCBI accession numbers NM_006262.3 (human peripherin; PRPH; SEQ ID NO: 41); XM_005269025.1 (predicted human peripherin, transcript variant XI ; SEQ ID NO: 42); XR 944623.1

(predicted human peripherin, transcript variant X2; SEQ ID NO: 43), and partial sequences thereof. Exemplary peripherin amino acid sequences include those encoded by the above nucleotide sequences.

[00156] Examples of GRP polypeptides include wild-type human GRP78/BiP, and polypeptides having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with such wild-type GRP. An exemplary amino acid sequence of wild-type human GRP is represented by SEQ ID NO: 44, while an exemplary GRP encoding nucleic acid sequence is represented by SEQ ID NO: 45 (see, e.g., open reading frame in NCBI accession number X87949.1).

[00157] Any nucleotide sequence encoding the above amino acid sequence represented by SEQ ID NO: 44, or any nucleotide sequence encoding at least about 100, at least about 200, at least about 300, at least about 400, or at least about 500 consecutive amino acids thereof, or any nucleotide sequence encoding a polypeptide having at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity with SEQ ID NO: 44 may be used.

[00158] Other exemplary GRP sequences are disclosed, e.g., in UniProtKB - PI 1021 and links therein. In some examples, the GRP polypeptide is a trimmed variant containing less than about 500, less than about 400, less than about 300, or less than about 200 of the wild- type amino acids. In some examples, the trimmed GRP variants are selected to be efficiently expressed and secreted by a LAB strain (i.e., Lactococcus lactis).

[00159] A person of ordinary skill in the art will appreciate that the optimal amount of self- antigen to be delivered to the subject using the methods of the present disclosure varies, e.g., with the type of antigen, the microorganism expressing the antigen, and the genetic construct, e.g., the strength of the promoter used in the genetic construct. Typically, the microorganism will be administered in an amount equivalent to a particular amount of expressed antigen, or in an amount, which generates a desired PK profile for the respective antigen polypeptide in the respective subject. Exemplary daily antigen doses can be from about 10 fg to about 100 μg of active polypeptide per day. Other exemplary dose ranges can be from about 1 pg to about 100 μg per day; or from about 1 ng to about 100 μg per day.

[00160] The above antigen doses may be realized by administering to the subject effective amounts of the LAB per day, wherein the LAB is adapted to express a sufficient amount of bioactive polypeptide to realize the desired dose, such as those above. The LAB secreting the antigen polypeptide may be delivered in a dose of from about 10 4 colony forming units (cfu) to about 10 12 cfu per day, e.g. , from about 10 6 cfu to about 10 12 cfu per day, or from about 10 9 cfu to about 10 12 cfu per day.

[00161] The amount of secreted antigen polypeptide can be determined based on cfu, for example in accordance with the methods described in Steidler et al, Science 2000;

289(5483): 1352-1355, or by using ELISA. For example, a LAB may secrete at least about 1 ng to about 1 μg of active polypeptide per 10 9 cfu. Based thereon, the skilled person can calculate the range of antigen polypeptide secreted at other cfu doses.

[00162] Each of the above doses/dose ranges may be administered in connection with any dosing regimen as described herein. The daily dose of active polypeptide may be administered in 1 , 2, 3, 4, 5, or 6 portions throughout the day. Further the daily doses may be administered for any number of days, with any number of rest periods between administration periods. For example, a dose of from about 0.1 to about 3.0 M IU/day/subject may be administered every other day for a total of 6 weeks.

Formulations and Regimens

[00163] In the methods described herein, the IL-2 and the T1D may be expressed by the same or different LAB. When the two polypeptides are expressed by different

microorganisms, those may be administered to the subject in the same (e.g., combined) formulation or may be administered in separate (e.g., different) formulations. Separate formulations may be administered at the same time or at different time points. For example, the of IL-2 and TlD-specific antigen producing microorganisms in their respective formulations can be administered to the subject simultaneously or may be administered sequentially, e.g., with a rest period between administrations.

[00164] The IL-2 and TlD-specific antigen producing LAB strains can be administered simultaneously. In some examples, the IL-2-producing microorganism, and the TlD-specific antigen-producing microorganism can be comprised in the same pharmaceutical formulation, or in more than one pharmaceutical formulation taken at the same time. In exemplary embodiments, the two bioactive polypeptides are delivered to the subject using a single LAB strain producing both the IL-2 and the TlD-specific antigen.

[00165] In some embodiments, the composition described herein will be administered, once, twice, three, four, five, or six times daily, e.g. , using an oral formulation. In some embodiments, the LAB strains are administered every day, every other day, once per week, twice per week, three times per week, or four times per week. In other embodiments, treatment occurs once every two weeks. In other embodiments, treatment occurs once every three weeks. In other embodiments, treatment occurs once per month.

[00166] The duration of a treatment cycle for the method may be, for example, 7 days to the subject's lifetime, as needed to treat or reverse T1D, or prevent relapse. A treatment cycle can last for about 30 days to about 2 years. In other embodiments, the subject can have a treatment cycle that lasts from 30 days to 1.5 years. In other embodiments, the subject can have a treatment cycle that lasts from 30 days to 1 year. In other embodiments, the subject can have a treatment cycle that lasts from 30 days to 1 1 months. In other embodiments, the subject can have a treatment cycle that lasts from 30 days to 10 months. In other embodiments, the subject can have a treatment cycle that lasts from 30 days to 9 months. The subject can have a treatment cycle that lasts from 30 days to 8 months. The subject can have a treatment cycle that lasts from 30 days to 7 months. The subject can have a treatment cycle that lasts from 30 days to 6 months. The subject can have a treatment cycle that lasts from 30 days to 5 months. The subject can have a treatment cycle that lasts from 30 days to 4 months. The subject can have a treatment cycle that lasts from 30 days to 3 months. The subject can have a treatment cycle that lasts from 30 days to 2 months.

[00167] Daily maintenance doses can be given for a period clinically desirable in the subject, for example from 1 day up to several years (e.g. for the subject's entire remaining life); for example from about (2, 3 or 5 days, 1 or 2 weeks, or 1 month) upwards and/or for example up to about (5 years, 1 year, 6 months, 1 month, 1 week, or 3 or 5 days).

Administration of the daily maintenance dose for about 3 to about 5 days or for about 1 week to about 1 year is typical. Nevertheless, unit doses optionally may be administered from twice daily to once every two weeks until a therapeutic effect is observed.

[00168] The LAB strains producing the IL-2 polypeptide and the antigen polypeptide may be delivered in mono- or combination therapy for the treatment of T1D. In some embodiments, the compositions of the present disclosure include additional therapeutically active agents. In some embodiments, the treatment of the subject does not involve other active components, e.g. , does not involve additional immune-modulating substances, such as antibodies (e.g., anti-CD3). Thus, in some examples, the pharmaceutical compositions of the present disclosure consist essentially of the LAB as described herein (expressing the therapeutic IL-2 and antigen polypeptides), and a pharmaceutically acceptable carrier.

Pharmaceutical Compositions and Carriers

[00169] The LAB strains described herein (e.g., L. lactis) may be administered in pure form, combined with other active ingredients, and/or combined with pharmaceutically acceptable (i.e., nontoxic) excipients or carriers. The term "pharmaceutically acceptable" is used herein in accordance with its art-recognized meaning and refers to carriers that are compatible with the other ingredients of a pharmaceutical composition, and are not deleterious to the recipient thereof.

[00170] The compositions described herein can be prepared in any known or otherwise effective dosage or product form suitable for use in providing systemic delivery of the LAB strains (e.g., L. lactis) to the mucosa, which would include pharmaceutical compositions and dosage forms as well as nutritional product forms.

[00171] In some embodiments, the formulation is an oral formulation or pharmaceutical composition. In some examples according to this embodiment, the formulation or pharmaceutical composition comprises the LAB strains in a dry-powder form (e.g., freeze- dried form) or in compacted form thereof, optionally in combination with other dry carriers. Oral formulations will generally include an inert diluent carrier or an edible carrier.

[00172] In some examples, the oral formulation comprises a coating or utilizes an encapsulation strategy, which facilitates the delivery of the formulation into the intestinal tract, and/or allows the microorganism be released and hydrated in the intestinal tract (e.g., the ileum, small intestine, or the colon). Once the LAB is released from the formulation and sufficiently hydrated, it begins expressing the bioactive polypeptide, which is subsequently released into the surroundings, or expressed on the surface of the microorganism. Such coating and encapsulation strategies (i.e., delayed-release strategies) are known to those of skill in the art. See, e.g., U.S. 5,972,685; WO 2000/18377; and WO 2000/22909.

[00173] A pharmaceutical composition is provided that can comprise the LAB stains in a lyophilized or freeze dried form, optionally in conjunction with other components, such as dextranes, sodium glutamate, and polyols. Exemplary freeze dried compositions are described, e.g., in U.S. Pub. No. 2012/0039853. Exemplary formulations comprise freeze- dried bacteria (e.g., a therapeutically effective amount of the bacteria) and a pharmaceutically acceptable carrier. Freeze-dried bacteria may be prepared in the form of capsules, tablets, granulates and powders, each of which may be administered orally. Alternatively, freeze- dried bacteria may be prepared as aqueous suspensions in suitable media, or lyophilized bacteria may be suspended in a suitable medium, such as a drink, just prior to use.

[00174] For oral administration, the formulation may be a gastro-resistant oral dosage form. For example, the oral dosage form (e.g., capsules, tablets, pellets, micro-pellets, granulates, and the like) may be coated with a thin layer of excipient (usually polymers, cellulosic derivatives and/or lipophilic materials) that resists dissolution or disruption in the stomach, but not in the intestine, thereby allowing transit through the stomach in favor of disintegration, dissolution and absorption in the intestine (e.g., the small intestine, or the colon).

[00175] In some examples, oral formulations may include compounds providing controlled release, sustained release, or prolonged release of the microorganism, and thereby provide controlled release of the desired protein encoded therein. These dosage forms (e.g. , tablets or capsules) typically contain conventional and well known excipients, such as lipophilic, polymeric, cellulosic, insoluble, and swellable excipients. Controlled release formulations may also be used for any other delivery sites including intestinal, colon, bioadhesion or sublingual delivery (i.e., dental mucosal delivery), and bronchial delivery. When the compositions described herein are to be administered rectally or vaginally, pharmaceutical formulations may include suppositories and creams. In this instance, the host cells are suspended in a mixture of common excipients also including lipids. Each of the

aforementioned formulations are well known in the art and are described, for example, in the following references: Hansel et al. (1990, PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th edition, William and Wilkins); Chien 1992, NOVEL DRUG DELIVERY SYSTEM, 2nd edition, M. Dekker); Prescott et al. (1989, NOVEL DRUG DELIVERY, J. Wiley & Sons); Cazzaniga et al , (1994, Int. J. Pharm. 108(1): 77-83).

[00176] The oral formulations and compositions described herein can further include compounds that can enhance mucosal delivery and/or mucosal uptake of the bioactive polypeptides expressed by the LAB. The formulations / compositions described herein can also include compounds, which enhance the viability of the microorganism within the formulation, and/or once released. [00177] The LAB as described herein can be suspended in a pharmaceutical formulation for administration to the human or animal having the disease to be treated. Such pharmaceutical formulations include but are not limited to live LAB and a medium suitable for administration. The LAB may be lyophiiized in the presence of common excipients such as lactose, other sugars, alkaline and/or alkali earth stearate, carbonate and/or sulphate (e.g. , magnesium stearate, sodium carbonate and sodium sulphate), kaolin, silica, flavorants and aromas. Bacteria so-lyophilized may be prepared in the form of capsules, tablets, granulates and powders (e.g., a mouth rinse powder), each of which may be administered by the oral route. Alternatively, the LAB strains may be prepared as aqueous suspensions in suitable media, or lyophiiized bacteria may be suspended in a suitable medium just prior to use, such medium including the excipients referred to herein and other excipients such as glucose, glycine, and sodium saccharinate.

[00178] In some examples, the LAB is locally delivered to the gastrointestinal tract of the subject using any suitable method. For example, microsphere delivery systems could be employed to enhance delivery to the gut. Microsphere delivery systems include

microparticles having a coating that provides localized release into the gastrointestinal tract of the subject (e.g., controlled release formulations such as enteric-coated formulations and colonic formulations).

[00179] For oral administration, gastroresistant oral dosage forms may be formulated, which dosage forms may also include compounds providing controlled release of the LAB strains and thereby provide controlled release of the desired protein encoded therein at different points in digestion (e.g., IL-2). For example, the oral dosage form (including capsules, tablets, pellets, granulates, powders) may be coated with a thin layer of excipient (e.g., polymers, cellulosic derivatives and/or lipophilic materials) that resists dissolution or disruption in the stomach, but not in the intestine, thereby allowing transit through the stomach in favor of disintegration, dissolution and absorption in the intestine.

[00180] The oral dosage form may be designed to allow slow release of the LAB strains and of the produced exogenous proteins, for instance as controlled release, sustained release, prolonged release, sustained action tablets or capsules. These dosage forms usually contain conventional and well-known excipients, such as lipophilic, polymeric, cellulosic, insoluble, and swellable excipients. Such formulations are described, for example, in the following references: Hansel et al, PHARMACEUTICAL DOSAGE FORMS AND DRUG DELIVERY SYSTEMS, 5th edition, William and Wilkins, 1990; Chien 1992, NOVEL DRUG DELIVERY SYSTEM, 2nd edition, M. Dekker; Prescott et al, NOVEL DRUG DELIVERY, J.Wiley & Sons, 1989; and Cazzaniga et al. nt. J. Pharm. 108(l):77-83 (1994).

[00181] The pharmaceutical dosage form {e.g. capsule) is typically coated with pH- dependent Eudragit® polymers to obtain gastric juice resistance and for the intended delivery at the terminal ileum and colon, where the polymers dissolve at pH 6.5. By using other Eudragit® polymers or a different ratio between the polymers, the delayed release profile could be adjusted, to release the bacteria for example in the duodenum or jejunum.

[00182] Pharmaceutical compositions commonly contain at least one pharmaceutically acceptable carrier. Non-limiting examples of suitable excipients, diluents, and carriers include preservatives, inorganic salts, acids, bases, buffers, nutrients, vitamins, fillers and extenders such as starch, sugars, mannitol, and silicic derivatives; binding agents such as carboxymethyl cellulose and other cellulose derivatives, alginates, gelatin, and polyvinyl pyrolidone; moisturizing agents such as glycerol/ disintegrating agents such as calcium carbonate and sodium bicarbonate; agents for retarding dissolution such as paraffin;

resorption accelerators such as quaternary ammonium compounds; surface active agents such as acetyl alcohol, glycerol monostearate; adsorptive carriers such as kaolin and bentonite; carriers such as propylene glycol and ethyl alcohol, and lubricants such as talc, calcium and magnesium stearate, and solid polyethyl glycols.

[00183] Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. Tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a dispersing agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. When the dosage unit form is a capsule, it can contain, in addition to material of the above type, a liquid carrier such as a fatty oil. In addition, dosage unit forms can contain various other materials that modify the physical form of the dosage unit, for example, coatings of sugar, shellac, or enteric agents. Further, a syrup may contain, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes, colorings, and flavorings. It will be appreciated that the form and character of the pharmaceutically acceptable carrier is dictated by the amount of active ingredient with which it is to be combined, the route of administration and other well-known variables. The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.

[00184] Alternative preparations for administration include sterile aqueous or nonaqueous solutions, suspensions, and emulsions. Examples of non-aqueous solvents are

dimethylsulfoxide, alcohols, propylene glycol, polyethylene glycol, vegetable oils such as olive oil and injectable organic esters such as ethyl oleate. Aqueous carriers include mixtures of alcohols and water, buffered media, and saline. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers, such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present such as, for example, antimicrobials, anti-oxidants, chelating agents, inert gases, and the like. Various liquid formulations are possible for these delivery methods, including saline, alcohol, DMSO, and water based solutions.

[00185] Oral aqueous formulations include excipients, such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and/or the like. These compositions take the form of solutions such as mouthwashes and mouth rinses, further comprising an aqueous carrier such as for example water, alcoholic/aqueous solutions, saline solutions, parenteral vehicles such as sodium chloride, Ringer's dextrose, and the like.

[00186] Aqueous mouthwash formulations are well4cnown to those skilled in the art. Formulations pertaining to mouthwashes and oral rinses are discussed in detail, for example, in U.S. Patent 6,387,352, U.S. Patent 6,348,187, U.S. Patent 6,171 ,611, U.S. Patent

6,165,494, U.S. Patent 6,117,417, U.S. Patent 5,993,785, U.S. Patent 5,695,746, U.S. Patent 5,470,561 , U.S. Patent 4,919,918, U.S. Patent Appl. No. 2004/0076590, U.S. Patent Appl. No. 2003/0152530, and U.S. Patent Appl. No. 2002/0044910.

[00187] Other additives may be present in the formulations of the present disclosure, such as flavoring, sweetening or coloring agents, or preservatives. Mint, such as from peppermint or spearmint, cinnamon, eucalyptus, citrus, cassia, anise and menthol are examples of suitable flavoring agents. Flavoring agents are optionally present in the oral compositions in an amount in the range of from 0 to 3%; optionally up to 2%, such as up to 0.5%, optionally around 0.2%, in the case of liquid compositions.

[00188] Sweeteners include artificial or natural sweetening agents, such as sodium saccharin, sucrose, glucose, saccharin, dextrose, levulose, lactose, mannitol, sorbitol, fructose, maltose, xylitol, thaumatin, aspartame, D-tryptophan, dihydrochalcones, acesulfame, and any combinations thereof, which may be present in an amount in the range of from 0 to 2%, optionally up to 1 % w/w, such as 0.05 to 0.3% w/w of the oral composition.

[00189] Coloring agents are suitable natural or synthetic colors, such as titanium dioxide or CI 42090, or mixtures thereof. Coloring agents may be present in the compositions in an amount in the range of from 0 to 3%; optionally up to 0.1 %, such as up to 0.05%, optionally around 0.005- 0.0005%, in the case of liquid compositions. Of the usual preservatives, sodium benzoate is typically used in concentrations insufficient substantially to alter the pH of the composition, otherwise the amount of buffering agent may need to be adjusted to arrive at the desired pH.

[00190] Other optional ingredients include humectants, surfactants (non- ionic, cationic or amphoteric), thickeners, gums and binding agents. A humectant adds body to the formulation and retains moisture in a dentifrice composition. In addition, a humectant helps to prevent microbial deterioration during storage of the formulation. It also assists in maintaining phase stability and provides a way to formulate a transparent or translucent dentifrice.

[00191] Suitable humectants include glycerin, xylitol, glycerol and glycols such as propylene glycol, which may be present in an amount of up to 50% w/w each, but total humectant may be no more than about 60-80% w/w of the composition. For example, liquid compositions may comprise up to about 30% glycerin plus up to about 5%, optionally about 2% w/w xylitol. Surfactants may be not anionic and may include polysorbate 20 or cocoamidobetaine or the like in an amount up to about 6%, optionally about 1.5 to 3%, w/w of the composition.

[00192] When the oral compositions as described herein are in a liquid form, said compositions typically may include a film- forming agent up to about 3% w/w of the oral composition, such as in the range of from 0 to 0.1%, optionally about 0.001 to 0.01%, such as about 0.005% w/w of the oral composition. Suitable film-formers include (in addition to sodium hyaluronate) those sold under the tradename, Gantrez™.

[00193] Liquid nutritional formulations for oral or enteral administration may comprise one or more nutrients such as fats, carbohydrates, proteins, vitamins, and minerals. Many different sources and types of carbohydrates, lipids, proteins, minerals and vitamins are known and can be used in the nutritional liquid embodiments described herein, provided that such nutrients are compatible with the added ingredients in the selected formulation, are safe and effective for their intended use, and do not otherwise unduly impair product performance.

[00194] These nutritional liquids are typically formulated with sufficient viscosity, flow, or other physical or chemical characteristics to provide a more effective and soothing coating of the mucosa while drinking or administering the nutritional liquid. These nutritional embodiments also may represent a balanced nutritional source suitable for meeting the sole, primary, or supplemental nutrition needs of the individual. Non-limiting examples of suitable nutritional liquids are described, e.g., in U. S. Patent 5,700,782; U. S. Patent 5,869,118; and U. S. Patent 5,223,285.

[00195] Nutritional proteins suitable for use herein can be hydrolyzed, partially hydrolyzed or non-hydrolyzed, and can be derived from any known or otherwise suitable source such as milk (e.g., casein, whey), animal (e.g., meat, fish), cereal (e.g., rice, corn), vegetable (e.g., soy), or combinations thereof.

[00196] Fats or lipids suitable for use in the nutritional liquids include, but are not limited to, coconut oil, soy oil, corn oil, olive oil, safflower oil, high oleic safflower oil, MCT oil (medium chain triglycerides), sunflower oil, high oleic sunflower oil, structured triglycerides, palm and palm kernel oils, palm olein, canola oil, marine oils, cottonseed oils, and combinations thereof. Carbohydrates suitable for use in the nutritional liquids may be simple or complex, lactose- containing or lactose-free, or combinations thereof. Non-limiting examples of suitable carbohydrates include hydrolyzed corn starch, maltodextrin, glucose polymers, sucrose, corn syrup, corn syrup solids, rice-derived carbohydrate, glucose, fructose, lactose, high fructose corn syrup and indigestible oligosaccharides such as fructo- oligosaccharides (FOS), and combinations thereof.

[00197] The nutritional liquids as described herein may further comprise any of a variety of vitamins, non-limiting examples of which include vitamin A, vitamin D, vitamin E, vitamin K, thiamine, riboflavin, pyridoxine, vitamin B 12, niacin, folic acid, pantothenic acid, biotin, vitamin C, choline, inositol, salts and derivatives thereof, and combinations thereof.

[00198] The nutritional liquids as described herein may further comprise any of a variety of minerals known or otherwise suitable for us in patients at risk of or suffering from T1D, non-limiting examples of which include calcium, phosphorus, magnesium iron, selenium, manganese, copper, iodine, sodium, potassium, chloride, and combinations thereof. [00199] The LAB strains described herein can also be formulated as elixirs or solutions for convenient oral or rectal administration or as solutions appropriate for parenteral administration, for instance by intramuscular, subcutaneous or intravenous routes.

Additionally, the nucleoside derivatives are also well suited for formulation as a sustained or prolonged release dosage forms, including dosage forms that release active ingredient only or optionally in a particular part of the intestinal tract, optionally over an extended or prolonged period of time to further enhance effectiveness. The coatings, envelopes, and protective matrices in such dosage forms may be made, for example, from polymeric substances or waxes well known in the pharmaceutical arts.

[00200] The compositions as described herein may include pharmaceutical dosage forms such as lozenges, troches or pastilles. These are typically discoid-shaped solids containing the active ingredient in a suitably flavored base. The base may be a hard sugar candy, glycerinated gelatin, or the combination of sugar with sufficient mucilage to give it form. Troches are placed in the mouth where they slowly dissolve, liberating the active ingredient for direct contact with the mucosa.

[00201] The troche embodiments can be prepared, for example, by adding water slowly to a mixture of the powdered active, powdered sugar, and a gum until a pliable mass is formed. A 7% acacia powder can be used to provide sufficient adhesiveness to the mass. The mass is rolled out and the troche pieces cut from the flattened mass, or the mass can be rolled into a cylinder and divided. Each cut or divided piece is shaped and allowed to dry, to thus form the troche dosage form.

[00202] If the active ingredient is heat labile, it may be made into a lozenge preparation by compression. For example, the granulation step in the preparation is performed in a manner similar to that used for any compressed tablet. The lozenge is made using heavy compression equipment to give a tablet that is harder than usual as it is desirable for the dosage form to dissolve or disintegrate slowly in the mouth. Ingredients are typically selected to promote slow-dissolving characteristics.

[00203] In an exemplary formulation, the LAB strains may be incorporated in a bioadhesive carrier containing pregelatinized starch and cross-linked poly (acrylic acid) to form a bioadhesive tablet and a bioadhesive gel suitable for buccal application (i.e., having prolonged bioadhesion and sustained drug delivery). [00204] A powder mixture of the LAB strains, bioadhesive polymers (pregelatinized starch and cross-linked poly (acrylic acid) coprocessed via spray drying), sodium stearyl fumarate (lubricant) and silicon dioxide (glidant) may be processed into tablets (weight: 100 mg; diameter: 7 mm). The methods for the production of these tablets are well known to the person skilled in the art and has been described before for the successful development of bioadhesive tablets containing various drugs (miconazol, testosterone, fluoride,

ciprofloxacin) (Bruschi M. L. and de Freitas O., Drug Development and Industrial

Pharmacy, 2005 31 : 293-310).

[00205] To optimize the formulation, the drug load in the tablets and the ratio between starch and poly (acrylic acid) can be varied. Based on previous research, the maximum drug load in the co-processed bioadhesive carrier is about 60% (w/w) and the starch/poly (acrylic acid) ratio can be varied between 75/25 and 95/5 (w/w). During the optimization study the bioadhesive properties of the tablets and the drug release from the tablets are the main evaluation parameters, with the standard tablet properties (hardness, friability) as secondary evaluation criteria.

[00206] The LAB strains may be incorporated into an aqueous dispersion of pregelatinized starch and cross-linked poly (acrylic acid). This polymer dispersion is prepared via a standard procedure using a high shear mixer.

[00207] Similar to the tablet, the drug load of the gel and the starch/poly (acrylic acid) ratio may need to be optimized in order to obtain a gel having optimal adherence to the esophageal mucosa. For a gel, the concentration of the polymers in the dispersion is an additional variable as it determines the viscosity of the gel, hence its muco-adhesive properties.

[00208] The model to screen the bioadhesive properties of polymer dispersions to the mucosa of esophagus has been described in detail by Batchelor et al. {Int. J. Pharm., 238: 123- 32, 2002).

[00209] Other routes and forms of administration include food preparations containing the live LAB strains. In some examples, the bioactive polypeptide-expressing LAB strains may be included into a dairy product.

[00210] The pharmaceutical compositions described herein may be prepared by any known or otherwise effective method for formulating or manufacturing the selected dosage form. For example, the LAB strains can be formulated along with common, e.g., pharmaceutically acceptable carriers, such as excipients and diluents, formed into oral tablets, capsules, sprays, lozenges, treated substrates (e.g., oral or topical swabs, pads, or disposable, non-digestible substrate treated with the compositions described herein); oral liquids (e.g., suspensions, solutions, emulsions), powders, suppositories, or any other suitable dosage form. In some embodiments, the present disclosure provides a method for the manufacture of a pharmaceutical composition. Exemplary methods include: contacting the LAB strains (e.g., L. lactis) containing the IL-2 gene and the TlD-specific antigen gene (or which is capable of expressing the IL-2 and the TlD-specific antigen) with a pharmaceutically acceptable carrier, thereby forming the pharmaceutical composition. In some examples, the method may further include: growing the LAB strains in a medium. The method may further include freeze- drying a liquid containing the microorganism, wherein the liquid optionally includes the pharmaceutically acceptable carrier.

Unit Dosage Forms

[00211] The current disclosure further provides unit dosage forms comprising a certain amount of the LAB strain optionally in combination with a food-grade or pharmaceutically acceptable carrier, wherein the LAB strain comprises: an interleukin-2 (IL-2) gene; and a type-1 diabetes mellitus (TlD)-specific antigen gene. Exemplary unit dosage forms contain from about 1 * 10 3 to about 1 x 10 14 colony-forming units (cfu) of the LAB (e.g., L. lactis). Other exemplary unit dosage forms contain from about 1 x 10 4 to about 1 x 10 13 colony- forming units (cfu) of the LAB (e.g., L. lactis), or from about 1 x 10 4 to about 1 x 10 12 colony-forming units (cfu) of the LAB (e.g., L. lactis). In other embodiments, the unit dosage form comprises from about 1 10 s to about 1 10 12 colony-forming units (cfu), or from about 1 x 10 6 to about 1 x 10 12 colony-forming units (cfu) of the LAB (e.g., L. lactis). In other embodiments, the unit dosage form comprises from about 1 x 10 8 to about 1 x 10 12 colony-forming units (cfu), or from about 1 x 10 9 to about 1 x 10 12 colony- forming units (cfu) of the LAB (e.g., L. lactis). In yet other embodiments, the unit dosage form comprises from about 1 10 9 to about 1 x 10 1 1 colony-forming units (cfu), or from about 1 x 10 9 to about 1 x 10 10 colony- forming units (cfu) of the LAB (e.g., L. lactis). In yet other embodiments, the unit dosage form comprises from about 1 x 10 7 to about 1 x 10 1 1 colony- forming units (cfu), or from about 1 x 10 s to about 1 x 10 10 colony-forming units (cfu) of the LAB (e.g., L. lactis). [00212] In yet other embodiments, the unit dosage form comprises from about 1 x 10 9 to about 1 x 10 10 colony-forming units (cfu), or from about 1 x 10 9 to about 100 x 10 9 colony- forming units (cfu) of the LAB (e.g., L. lactis).

[00213] The unit dosage form can have any physical form or shape. In some

embodiments, the unit dosage form may be adapted for oral administration. In some examples according to these embodiments, the unit dosage form may be in the form of a capsule, a tablet, or a granule. Exemplary capsules include capsules filled with micro- granules. In some embodiments, the LAB (e.g., L. lactis) contained in the dosage form is in a dry-powder form. For example, the LAB is in a freeze-dried powder form, which is optionally compacted and coated.

EXAMPLES

[00214] The following examples are provided in order to demonstrate and further illustrate certain representative embodiments and aspect of the present disclosure and are not to be construed as limiting the scope of the specification or claims.

Example 1. Construction of Lactococcus lactis Secreting hIL-2 (LL-IL-2)

[00215] A Lactococcus lactis strain, which can secrete human IL-2 (LL-IL-2) was constructed relative to Lactococcus lactis MG1363 (parent strain). See, e.g., Gasson MJ, J. Bacteriol. 1983, 154(l):l -9. In LL-IL-2, the following modifications were introduced into the genome of the bacteria:

[00216] (a) The thymidylate synthase gene (thyA; Gene ID: 4798358; location:

NC_009004.1 (930251..931090)) was removed to ascertain environmental containment.

[00217] (b) Trehalose-6-phosphate phosphorylase gene (trePP; Gene ID: 4797140;

location: NC_009004.1 (449195..451504)) was removed to allow accumulation of exogenous trehalose.

[00218] (c) Trehalose-6-phosphate phosphatase (otsB; Gene ID: 1036914; Locus tag c2311) was positioned downstream of unidentified secreted 45-kDa protein gene (usp45; Gene ID: 4797218; location: NC_009004.1 (2462440..2463825, complement)) to facilitate conversion of trehalose-6-phosphate to trehalose.

[00219] (d) The constitutive promoter of the HU-like DNA-binding protein gene (PhllA; Gene ID: 4797353; location: NC_009004.1 (490275..490550)) was added to precede the putative phosphotransferase genes in the trehalose operon (trePTS; ptsl and ptsll; LLMG_RS02300 and LLMG_RS02305; Gene ID: 4797778; location: NC_009004.1 (446937..447422) and Gene ID: 4797093; location: NC_009004.1 (447563..449128), respectively) to potentiate trehalose uptake.

[00220] (e) The gene encoding cellobiose-specific PTS system IIC component (Gene ID: 4796893; location: NC_009004.1 (430271..431608)), ptcC, was deleted to increase trehalose retention.

[00221] (f) A gene encoding a fusion of usp45 secretion leader (Ssusp45) with the hIL-2 gene, encoding human interleukin-2 (hIL-2; UniProt: P60568, aa 21-153) was positioned downstream of the phosphopyruvate hydratase gene (eno; Gene ID: 4797432; location: NC_ 009004.1 (606184..607485)), to allow expression and secretion of hIL-2. The hIl-2 expression unit was transcriptionally and translationally coupled to eno by use of the intergenic region preceding the highly expressed L. lactis MG1363 50S ribosomal protein L30 gene (rpmD; Gene ID: 4797873; location: NC_009004.1 (2316732..2316911, complement)). An exemplary nucleotide sequence encoding the above fusion of Ssusp45 and hIL-2 downstream of enoA, linked by rpmD is depicted in FIG. 1 (SEQ ID NO: 46).

[00222] FIG. 2 provides a schematic overview of the above described genetic loci.

[00223] The experiments also involve a control strain (LL-Control) having genetic traits comparable to LL-IL-2, except that the control strain does not contain the constructs for the expression of IL-2. The genetic traits for LL-IL-2 and the LL-Control strains are summarized in Table 1 below.

Table 1. Overview of Genetic Characteristics of Various LL Strains

[00224] Referring to Table 1 , trePTS expression (at the trehalose operon) can be as in the wild type (wt) or driven by the hllA promoter (PhllA»PTS); trePP can be wt or deleted (Δ); ptcC can be wt or Δ; otsB can be absent (-) or located downstream of and expressed from usp45 (usp45»otsB); thyA can be wt or Δ; eno locus can be wt (-) or contain hIL-2. All gapB loci are wt, in contrast to LL-PINS/IL-2, which carries gapB»pins as described herein below.

[00225] The genetic modifications were carried out using double homologous

recombination at the 5' and 3' end of these genetic traits. A similar method has been described for the construction of L. lactis Thy 12 (see, e.g., Steidler L., et al, Nat. Biotechnol. 2003, 21(7):785-789), with the difference that the helper plasmid pVE6007 was not used. The procedure involved erythromycin selection as an intermediate step, and the erythromycin selection marker was subsequently removed. As a result, LL-IL-2 has substantially no residual erythromycin resistance.

Carrier Plasmids

[00226] The modification method makes use of carrier plasmids derived from the conditionally non-replicative pORI19. See, e.g., Law J., et al., J. Bacteriol. 1995;

177(24):7011-7018. This replication protein A gene deficient (repA)- plasmid, as all of its repA- derivatives, cannot replicate in repA- . lactis. The repA+ L. lactis strain LL108 (see Sanders et al., J. Bacteriol. 1995, 177(18):5254-5260) was used as a construction host.

Carrier plasmids were designed so that up to 1 kb cross over (XO) areas, identical to the ones flanking the wild type sequence on the bacterial chromosome, are positioned 5' and 3' of the plasmid borne modification. Exemplary plasmids are used to insert hIL-2 downstream of eno in such way that both are coupled by the rpmD intergenic region. All plasmid construction was performed by use of standard molecular biological methods.

Chromosomal Modifications

[00227] Derivatives of plasmid pORIl 9 carry an erythromycin selection marker (ermC, 23S RNA methylase gene; Gene ID: 1263245) and cannot replicate in MG1363 or any of its derivatives. Upon introduction of such plasmid into MG1363, erythromycin selection was applied to the culture. Resistant colonies were selected on solid agar plates containing erythromycin. Because of the replication incompetence of the carrier plasmids, erythromycin- resistant bacteria can only arise following a first homologous recombination either at the 5' or 3' target site. Homologous recombination can be verified further by PCR.

[00228] Release of erythromycin selection enabled the excision of the carrier plasmid from the bacterial chromosome by a second homologous recombination, at either the 5' or 3' target site. For some erythromycin sensitive progeny, the second homologous recombination can occur at the target site alternative to the one of the first homologous recombination. This event replaces the wild type with the mutant on the bacterial chromosome and can be identified by PCR. Adequate subculture will rapidly dilute out all remnants of the carrier plasmid.

[00229] The presence of the β-glucuronidase gene (uidA, Gene ID: 946149) in the carrier plasmids, where it propagates along with ermC enables the identification of erythromycin sensitive and erythromycin resistant colonies. For example, bacterial suspensions were plated on 5-bromo-4-chloro-3-indolyl-beta-D-glucuronic acid (X-Gluc) containing solid agar plates. Glucuronidase (GUS) expressing (and therefore erythromycin resistant) clones will appear blue by conversion of X-gluc to its insoluble, blue reaction product

dichlorodibromoindigo, while erythromycin sensitive clones have also lost the uidA gene and therefore remain white. The identification of blue and white clones at relevant stages in the above described process greatly facilitated this approach.

PCR Analysis

[00230] Colonies showing the appropriate homologous recombination either at the 3' or 5' target site were analyzed by PCR. DNA fragments were purified using the Qiagen MinElute PCR Purification Kit. DNA sequences generated were identical to those predicted. FIG. 4 shows a 1.2% agarose gel of PCR fragments generated using the oligonucleotides listed in Table 2, Herculase II Fusion DNA polymerase (Agilent Technologies; #600677), and appropriate temperature cycles 50/120/30. Results demonstrated the presence of the desired genetic traits in LL-IL-2.

Table 2. Oligonucleotides Used for the Construction of LL-IL-2

[00231] In FIG. 4, molecular weight markers (MWM; Invitrogen 10488-85 Trackit 1 kb plus DNA Ladder) indicate base pairs: 100, 200, 300, 400, 500, 650, 850, 1000, 1650, 2000, 3000, 4000, 5000, and higher. Expected sizes of DNA fragments are also indicated in base pairs.

[00232] The bacterial genome of LL-IL-2 was further sequenced. The experimentally determined DNA sequences of all genetic traits in LL-IL-2 that differ from those of the parent strain MG1363 were found to be identical as expected.

Expression of hIL-2

[00233] Expression of hIL-2 by LL-IL-2 was measured using ELISA and western blot. In the ELISA experiment (utilizing R&D systems huIL-2 # DY202), 47.1 ng/mL of hIL-2 was measured in the culture supernatant, while a control strain did not produce hIL-2.

[00234] FIG. 5 is a Western blot showing the presence of hIL-2 in the culture supernatant of LL-IL-2. The Western blot was generated using goat anti-human IL-2 (1/1000 R&D systems AF-202-NA) as the first antibody, incubation with rabbit anti-goat - AP (1/1000 Southern Biotech #6160-04) as the detection antibody, and subsequent NBT/BCIP staining (Roche NBT BCIP tablets, #11 697 471 001). Equivalents of 1 ml bacterial cultures of LL- IL-2 and control strains were loaded onto the protein gel. Invitrogen SeeBlue® Plus2 Pre- Stained standard was used as molecular weight marker (MWM). The data indicates that LL- IL-2 secretes full length hIL-2 (i.e., as encoded by SEQ ID NO: 46).

[00235] Bacteria were cultured in GM 17 media, which is M 17 broth (Oxoid; #CM0817) supplemented with 0.5% glucose or GM17T medium (GM17 supplemented with 200 μΜ thymidine).

Example 2. Lactococcus lactis Secreting PINS and hIL-2 (LL-PINS/IL-2)

[00236] The construction and selection of strain LL-PINS/IL-2, a derivative of

Lactococcus lactis (L. lactis) MG1363, is described. LL-PINS/IL-2 includes the following genetic traits: (a) the thymidylate synthase gene (thyA; Gene ID: 4798358; location:

NC_009004.1 (930251..931090)) was removed to warrant environmental containment; (b) the trehalose-6-phosphate phosphorylase gene (trePP; Gene ID: 4797140; location:

NC 009004.1 (449195..451504)) was removed to allow accumulation of exogenously added trehalose; (c) the trehalose-6-phosphate phosphatase gene (otsB; Gene ID: 1036914; Locus tag c231 1) was positioned downstream of unidentified secreted 45-kDa protein gene (usp45; Gene ID: 4797218; location: NC_009004.1 (2462440..2463825, complement)) to facilitate conversion of trehalose-6-phosphate to trehalose; (d) the otsB expression unit was transcriptionally and translationally coupled to gapB using the intergenic region preceding the highly expressed L. lactis MG1363 50S ribosomal protein L30 gene (rpnxD; Gene ID: 4797873; location: NC_009004.1 (2316732..2316911 , complement)); (e) the constitutive promoter of the HU-like DNA-binding protein gene (PhllA; Gene ID: 4797353; location: NC_009004.1 (490275..490550)) precedes the putative phosphotransferase genes in the trehalose operon (trePTS; ptsl and ptsll; LLMG_RS02300 and LLMG_RS02305; Gene ID: 4797778; location: NC_009004.1 (446937..447422) and Gene ID: 4797093; location:

NC_009004.1 (447563..44 128) respectively) to potentiate trehalose uptake; (f) the gene encoding cellobiose-specific PTS system IIC component (Gene ID: 4796893; location:

NC_009004.1 (430271..431608)), ptcC, was disrupted (tga at codon position 30 of 446; tga30) to ascertain trehalose retention after accumulation; (g) a gene encoding a fusion of usp45 secretion leader (SSusp45) with the pins gene, encoding human proinsulin (PINS; UniProt: P01308, aa 25-1 10) is positioned downstream of the glyceraldehyde 3-phosphate dehydrogenase gene (gapB; Gene ID: 4797877; location: NC_009004.1 (2492509..2493519, complement)), to allow expression and secretion of proinsulin; (h) the pins expression unit was transcriptionally and translationally coupled to gapB by use of the intergenic region preceding the highly expressed L. lactis MG1363 50S ribosomal protein L30 gene (rpmD; Gene ID: 4797873; location: NC_009004.1 (2316732..2316911 , complement) ); and (i) a gene encoding a fusion of usp45 secretion leader (SSusp45) with the hIL-2 gene, encoding human interleukin-2 (hIL-2; UniProt: P60568, aa 21-153) was positioned downstream of the phosphopyruvate hydratase gene (eno; Gene ID: 4797432; location: NC_009004.1

(606184..607485)), to allow expression and secretion of hIL-2. The hil-2 expression unit was transcriptionally and translationally coupled to eno by use of the intergenic region preceding the highly expressed L. lactis MG1363 50S ribosomal protein L30 gene (rpmD; Gene ID: 4797873; location: NC_009004.1 (2316732..231691 1 , complement)).

[00237] All genetic traits of LL-PINS/IL-2 reside on the bacterial chromosome. The genetic background of this strain warrants: constitutive secretion of PINS and hIL-2; strict dependence on exogenously added thymidine for growth and survival; and the capacity to accumulate and retain trehalose to resist, e.g., bile acid lysis. [00238] An exemplary nucleotide sequence encoding the above fusion of SSusp45 and PINS downstream of the gapB gene, linked by the intergenic region rpmD is depicted in FIG. 6 (SEQ ID NO: 57). FIG. 7 provides a schematic overview of the above described genetic loci. Genetic traits were introduced into the bacterial genome as outlined in Example 1. Bacterial strains were grown and analyzed as described in Example 1 above.

Oligonucleotides used in the construction and analysis of LL-PINS/IL-2 are summarized in Table 3 below.

Table 3. Oligonucleotides Used for the Construction of LL-PINS/IL-2

[00239] FIG. 9 depicts a 1.2% agarose gel analysis of PCR fragments from LL-PINS/IL-2 indicating the presence of the desired genetic traits: trePTS, ptcC-, eno»hil-2, otsB, gapB»pins. In FIG. 9, molecular weight markers (MWM; Invitrogen 10488-85 Trackit 1 kb plus DNA Ladder) indicate base pairs: 100, 200, 300, 400, 500, 650, 850, 1000, 1650, 2000, 3000, 4000, 5000, and higher. Expected sizes of DNA fragments are also indicated in base pairs.

[00240] The bacterial genome of LL-PINS/IL-2 was further sequenced. The

experimentally determined DNA sequences of all genetic traits in LL-PINS/IL-2 that differ from those of the parent strain MG1363 were found to be identical to the predicted. [00241] Homologous recombination methods involved carrier plasmids derived from the conditionally non-replicative pORI19, described above. Carrier plasmids were designed in such way that up to 1 kb cross over (XO) areas, identical to the ones flanking the wild type sequence on the bacterial chromosome, are positioned 5' and 3' of the plasmid borne modification. An example of a carrier plasmid is pAGXl 145, a diagram of which is shown in FIG. 8. The plasmid is used to insert pins downstream of gapB in such way that both are coupled by the rpmD intergenic region. A similar plasmid, pAGX1372 {see annex:

pAGX1372.gbk) is used to insert hil-2 downstream of eno. All plasmid construction was performed by use of standard molecular biological methods.

PINS and hIL-2 Expression

[00242] Expression of PINS and hIL-2 by LL-PINS/IL-2 was measured using ELISA and western blot. Culture supematants from LL-PINS/IL-2 contained 0.6 ng/mL PINS and 28.2 ng/mL of hIL-2, while a control strain (LL-Control) did not produce either polypeptide. A MG 1363 bacterial strain expressing PINS from a plasmid vector (LL-PINS) was used as a positive control. PINS content in the supematants was determined using Mercodia cat. No. 10-1 1 18-01 , and hIL-2 content was determined by use of R&D system's huIL-2 # DY202.

[00243] FIG. 10 is a Western blot showing the presence of PINS and hIL-2 in the culture supernatant of LL-PINS/IL-2. Equivalents of 1 ml bacterial cultures were loaded onto the protein gel. The Western blot was generated using goat polyclonal anti-insulin B (Santa Cruz N-20: sc-7838) and goat anti-human IL-2 (1/1000 R&D systems AF-202-NA) as first antibodies for PINS and hIL-2 respectively, incubation with rabbit anti-goat - AP (1/1000 Southern Biotech #6160-04) detection antibody, and subsequent NBT/BCIP staining (Roche NBT/BCIP tablets, #11 697 471 001. Invitrogen SeeBlue® Plus2 Pre-Stained standard was used as molecular weight marker (MWM). The data indicates that LL-PINS/IL-2 secretes full length PINS and hIL-2.

[00244] Bacteria were cultured in GM 17 media, which is M 17 broth (Oxoid; #CM0817) supplemented with 0.5% glucose or GM17T medium (GM17 supplemented with 200 μΜ thymidine). Example 3. Pharmacodynamic studies to examine the effect of two bacterial strains on diabetes progression, Lactococcus lactis (LL) secreting proinsulin (LL-PINS) and hIL-2 (LL-IL-2)

[00245] Bacteria were cultured as described in Takiishi, T. et ah, J. Clin. Inv. 2012, 122(5): 1717-1725.

[00246] For example, single colonies of the respective L. lactis were inoculated in GM17T (M17, Oxoid, Hampshire, UK, supplemented with 0.5% glucose, 200 μΜ thymidine) and grown overnight to saturation. A 1/25 dilution of this culture was pre-grown for 3 hours in GM17T. Bacteria were harvested by centrifugation and further incubated for 3 hours in buffered culture medium (1 x BM9 salts, 0.5% casitone (Difco, BD Biosciences), 0.5% glucose, 25 mM NaHC0 3 , 25 mM Na 2 C0 3 , 2 mM MgS0 4 , 0.1 μΜ CaCl 2 ((BM9 Medium) (Schotte, et ah (2000) Enzyme Microb. Technoh 27(10):761 -765) supplemented with 200 μΜ thymidine) (BM9T). Bacteria were removed by centrifugation and supernatant samples were taken for analysis by Western blot and ELISA. For the western blot, proteins were prepared from crude BM9T L. lactis supernatants by deoxycholate/TCA acetone precipitation and were dissolved in SDS-PAGE sample buffer. Bacterial cell pellets were disrupted to obtain intracellular fractions. Culture supernatants (equivalent of 1 ml culture) and intracellular (equivalent of 50 μΐ culture) protein fractions were separated by SDS-12% PAGE, immunoblotted and revealed by goat anti-hIL-2 and detected using a rabbit anti-goat antibody and NBT/BCIP.

[00247] Stock solutions of all strains are stored in -20°C in 50% glycerol in GM17.

Bacteria are cultured in GM17 medium, i.e. Ml 7 (Difco Laboratories, Detroit, MI) supplemented with 0.5% glucose.

[00248] New-onset diabetic NOD mice, having positive glycosuria and two consecutive blood glucose measurements exceeding 200 mg/dl were used in the experimental set-up. Mice were allocated to three experimental treatment groups: (1) untreated controls, (2) LL- PINS-treated, and (3) mice treated with a combination of LL-PINS and LL-IL-2, for a period of 6 weeks.

[00249] This experiment involved two different LL strains. One strain constitutively expresses PINS and the other strain constitutively expresses IL-2. Mice were treated at a dose of 2 x 10 9 CFU by oral gavage 5 times weekly for six (6) weeks.

[00250] Mice were followed for either 42 days (therapy stop) or 100 days (8 weeks after therapy stop). Besides the initial follow up for disease remission until 100 days, additional mice (untreated and LL-PINS + LL-IL-2 treated) were euthanized at 42 days after treatment initiation and peripheral blood and different organs were used for further analyses. Serum samples for measuring insulin autoantibodies (IAA), inflammatory cytokines, and glucose- stimulated C-peptide were collected prior to treatment and after stopping therapy (day 42). In all experimental groups (both disease remitters and non-remitters) the peripheral immune system (phenotype and function) was assessed. Pancreas samples were taken for histology (insulitis) and insulin content determination (IC) at therapy stop (day 42).

T1D and Insulitis Assessment

[00251] NOD mice were screened for the onset of diabetes by evaluating glucose concentrations in the urine (Clinistix; Bayer Diagnostics) and venous blood (Accu-Chek® Aviva, Roche Diagnostics). Random-fed blood glucose measurements were collected between 8 and 11 am. Mice were classified as diabetic when having positive glycosuria and two consecutive blood glucose measurements exceeding 200 mg/dl. Diabetes remission was defined as an absence of glycosuria and glycemia values < 250 mg/dl on two consecutive days.

[00252] Pancreatic samples were fixed in formaldehyde solution and processed for paraffin embedding. 7^m-thick sections were stained with hematoxylin/eosin and the degree of insulitis was evaluated microscopically. Damages to the islets were graded as follows: 0: no infiltration; 1 : peri-insulitis; 2: islets with lymphocyte infiltration in less than 50% of the area; 3: islets with lymphocyte infiltration in more than 50% of the area; 4: islets completely destroyed.

Auto-antibody Detection

[00253] Serum IAA were measured at disease onset and at therapy discontinuation (day 42) by RIA assay. It was tested whether LL-PINS + LL-IL-2 vaccination can correct

hyperglycemia (disease remission) and maintain normoglycemia in new-onset diabetic NOD mice. Blood glucose concentrations were followed for 14 weeks post-treatment initiation.

Results - Disease Remission

[00254] FIG. 1 1 shows the percentage of mice that remained diabetic after treatment. After NOD mice developed hyperglycemia (2 consecutive days of blood glucose concentrations >200 mg/dl), they generally progressed to severe hyperglycemia with minimal spontaneous remissions and most died within 3-6 weeks (n=27). [00255] Mono-therapy with LL-PINS inoculation (2 x 10 9 CFU/day, 5 days per week for 6 weeks; n=8) corrected hyperglycemia in 15% of mice. Remarkably, 43% of newly diabetic mice (n=45) treated with a combination of LL-PINS and LL-IL-2 rapidly re-established normoglycemia. LL-PINS + LL-IL-2 therapy induced stable and permanent diabetes remission as cured mice maintained normoglycemia during an additional follow-up period of 8 weeks after stopping therapy.

[00256] Clinical efficacy of LL-PINS and LL-IL-2 therapy is clearly affected by the blood glucose concentrations at treatment initiation. Recent-onset diabetic NOD mice were stratified based on initial blood glucose concentrations under or above 350 mg/dL. The LL-PINS plus LL-IL-2 therapy not only cured 57% of mice with starting glycemia below 350 mg/dL, but also 22% of mice with a starting glycemia above 350 mg/dL (FIG. 12). These data demonstrate for the first time that mucosal delivery of PINS with IL-2 by recombinant L. lactis bacteria effectively corrects hyperglycemia and restores immune tolerance to β-cells in NOD mice with overt recent-onset T1D.

[00257] In all Kaplan-Meier survival curves, statistical significance between groups was determined by Mantel-Cox log-rank test (*: p<0.05).

Example 4. hIL-2 secreted by L. lactis (LL-IL-2) has biological activity comparable to recombinant hIL-2

[00258] This experiment involved the Lactococcus lactis strain expressing human IL-2 (LL- IL-2) as described herein {see, e.g., Example 1), and a control strain.

[00259] Bioactivity of LL-IL-2 was measured based on IL-2 dependent

survival/proliferation of a mouse T lymphocyte cell line HT2 clone A5E. HT2 cells were washed three times with medium without IL-2 and seeded at a density of 4 x 10 3 cells/96-well. A serial dilution series of recombinant hIL-2 {e.g. R&D systems # 202-IL-010) or supernatant from LL-IL-2 and a control strain was added to the plated cells and incubated for 24 hrs at 37°C, 5% C0 2 and high humidity. Cell viability was measured using CellTiter96®AQueous One Solution (Promega #G3582). 20 μΐ MTT solution was added per well and after an incubation period of 4 hrs at 37°C, 5% C0 2 and high humidity, the plates were read at 490 nm using 700 nm as reference wavelength. Recombinant hIL-2 (R&D systems) and hIL-2 derived from LL-IL-2 show comparable dose-dependent responses, while the supernatant of the L. lactis control strain was inactive. The results are shown in FIG. 13. Example 5. LL-IL-2 delivers low doses of hIL-2 to the GI tract of non-obese diabetic mice after oral administration

[00260] This experiment involved the Lactococcus lactis strain expressing human IL-2 (LL- IL-2) as described herein, e.g. , in Example 1.

Live Bacteria

[00261] The concentrations of live bacteria (CFU/tissue and CFU/g) in different tissues of the GI tract were measured at different time-points after a single dose administration of 10 10 CFU of LL-IL-2 by oral gavage. The results are depicted in FIGs. 14A and 14B, respectively, in which each bar represents an average of 3 mice (n=3). Referring to FIG. 14A (CFU/tissue), significant amounts of LL-IL-2 bacteria were found in the caecum (CAE), the proximal colon (COP), and the distal colon (COD) after 2, 4, 6, and 8 hours. The bacterial concentrations in the small intestine were found to be below 10 6 CFU/tissue. Referring to FIG. 14B (CFU/g), concentrations of LL-IL-2 bacteria were found in the proximal small intestine (SIP), the distal small intestine (SID), the caecum (CAE), the proximal colon (COP), and the distal colon (COD) after 2, 4, 6, and 8 hours, respectively. No bacteria were detected in the blood.

hIL-2 Protein

[00262] The concentrations of hIL-2 protein (pg/tissue and pg/g) in different tissues of the GI tract were measured after administration of a single dose of LL-IL-2 bacteria (10 10 CFU). hIL-2 protein concentrations were found in the caecum (CAE), the proximal colon (COP), and the distal colon (COD) after 2 and 4 hours. The hIL-2 protein concentrations in the small intestine were found to be below the limit of quantification (LLOQ=10 pg/mL). No hIL-2 protein was detected in the blood stream of the tested mice. The measured hIL-2 protein concentrations are summarized in Table 4 and Table 5 below.

[00263] At sacrifice, the complete tissue (SIP, SID, CAE, COP or COD) was weighed and homogenized. A sample of the homogenate was used for plating (to determine CFU) and for ELISA (to determine hIL-2). Total tissue in this context means that the concentration of bacteria or protein determined in the homogenate sample is recalculated to the weight of the total tissue. Table 4. Concentration of hIL-2 protein (pg/tissue) in different tissues of the GI tract after single dose administration of 10 10 CFU of LL-IL-2

CAE = caecum; COP = proximal colon; COD = distal colon; LLOQ = 10 pg/mL

Table 5. Concentration of hIL-2 protein (pg/g) in different tissues of the GI tract after administration of a single dose (10 10 CFU) of LL-IL-2

CAE = caecum; COP = proximal colon; COD = distal colon; LLOQ = 10 pg/mL

[00264] Viable L. lactis were found throughout the GI tract, with most bacteria located in the proximal and distal part of the large intestine and in the caecum. The bacterial concentration was a 1000-fold higher here than in the distal and proximal part of small intestine. This may be explained by the large amount of mucus and low motility in these parts of the intestine. About 50% of the administered L. lactis could be recovered from the distal parts of the colon 2 hours after administration. This finding is surprising because it had previously been reported that only about 10-30% of orally administered L. lactis survived the duodenal transit (Drouault S, et al, Appl. Environ. Microbiol. 1999; 65(11): 4881-6). It was speculated that inoculating the bacteria with BM9 inoculation buffer may protect the bacteria (at least partially) against GI conditions.

[00265] It was estimated that after dosing about 10 10 CFU LL-IL-2, about 90 pg IL-2 was delivered to the tissue which corresponds to about 1.2 IU of IL-2 (based on 1 IU = 73 pg). Example 6. Pharmacodynamic and mechanistic studies to examine the effect of a clinical grade Lactococcus lactis (LL) strain secreting both proinsulin (PINS) and hIL-2

[00266] This experiment involves a Lactococcus lactis strain expressing both PINS and IL-2 (LL-PINS/IL-2) as described herein {see, e.g., Example 2). Bacteria can be cultured as previously described. See Takiishi, T. et al, J. Clin. Inv. 2012, 122(5): 1717-1725. NOD Mice can be screened, treated, and analyzed as described in Example 3 above. Mice can be treated with the bacteria, e.g., at a dose of 2 x 10 9 CFU by oral gavage 5 times weekly for six (6) weeks.

Phenotypic analysis of the local and peripheral immune system

[00267] Peripheral organs {i.e., blood, mesenteric and pancreatic lymph nodes and pancreas) can be isolated at therapy discontinuation {e.g., on day 42) and can be

phenotypically examined, e.g., by flow cytometric analysis for canonical and non-canonical Treg makers {i.e., CD3, CD4, CD25, Foxp3, CD39, CD49b, LAG-3, and CD73). For example, co-expression of CD49b and LAG-3 enables the characterization of highly suppressive IL-10 producing Trl cells {see, e.g., Gagliani, N. et al, Nat. Med. 2013, 19(6): 739-746), while Tregs expressing both the ecto-enzymes CD39 and CD73 produce high concentrations of adenosine which is thought to be one of the Treg mechanisms of suppression. See, e.g., Antonioli, L., et al., Trends Mol. Med. 2013, 19(6): 355-367.

[00268] For intracellular cytokine staining, immune cells can be re-stimulated, e.g., with 1 μg/ml phorbol myristic acid (PMA, Sigma-Aldrich) and 0.5 μg ml ionomycin (Sigma- Aldrich) for 4 hours in the presence of 1 μΐ/ml GolgiPlug™ (BD). After cell surface staining, intracellular staining can be performed, e.g., using the Cytofix/Cytoperm™ kit (BD) {i.e. CD3, CD4, CD8, IL-2, IL-4, IL-17, and IFN-γ).

[00269] For pSTAT-5 detection, cell suspensions can be rapidly fixed after sacrifice or after in vitro culture, e.g., in 10 volumes of a solution of PBS 1.5% formaldehyde for 10 minutes at room temperature. Cells can be washed, e.g., in a solution of PBS containing 0.2% of BSA, and permeabilized, e.g., with 100% methanol for 10 minutes on ice. The cells may be washed further, e.g., with PBS 0.2% BSA, and can be incubated with a phospho- specific antibody in combination with an antibodies of interest {e.g., anti CD3, CD4, CD8, CD25, CD69, CD44, CD122), e.g., for 30 minutes in the dark at room temperature. In some cases, anti-Ki67 antibody can be added together with an anti-Foxp3 antibody. The pSTAT5 negative threshold can be defined on unstimulated cells or on cells stained with all fluorescent antibodies minus pSTAT5.

[00270] Multi-parameter analyses can be performed, e.g., using FACS Gallios (Beckman Coulter), FACS Canto II (Becton Dickinson (BD)), or FACS Fortessa (BD) and analyzed with Flow Jo® software (Tree Star). Dead cells (live dead yellow 405 staining) and doublets can be excluded from all analyses.

In vitro polyclonal suppression assay and IFN-y detection

[00271] Suppressive function of peripheral Tregs isolated from spleen and lymph nodes (ideally isolated from hCD2.Foxp3 NOD mice) can be assessed, e.g., in an in vitro polyclonal suppression assay were conducted as described. See, e.g., Takiishi, T., et al, J Clin. Invest. 2012. 122(5): 1717-1725. IFN-γ are measured in cell- free supernatants.

Results

[00272] LL-PINS/IL-2 treatment is expected to stimulate and recruit Tregs, and have biological activities comparable to LL-IL-2+LL-PINS treatments as described herein, e.g., in Examples 3 and 8.

Example 7. Construction of Lactococcus lactis Secreting Proinsulin (LL-PINS)

[00273] The DNA sequence encoding human proinsulin (hpins) was retrieved from

GenBank (Accession number NM_00207.2). The hpins DNA sequence was extended at the 3' end with a TAA stop codon and Spel restriction site. The DNA fragment was synthesized by PCR assembly of 40-mer oligonucleotides (oAGX0362 to oAGX0377) and Accu Prime DNA polymerase was used for amplification. The amplified fragment was fused to the usp45 secretion signal (SSusp45), downstream of the thyA lactococcal promoter (PthyA), which was extended at the 5' end with an EcoRI restriction site. The amplified product, which has a 5' EcoRI end and a 3' Spel end, was inserted between the EcoRI and Spel restriction sites of plasmid pTINX (GenBank accession number HM585371.1) and ligated. The ligation was introduced in L. lactis MG1363 by electroporation and colonies were screened by PCR analysis. The resulting plasmid was designated pAGX0053 (FIG. 15).

[00274] From MG1363[pAGX0053], a PCR fragment that contains the

PthyA»SSusp45::hpins expression module was generated using oAGX0169 and oAGX0170. The fragment was purified and it was confirmed that the DNA sequence of MG 1363 [pAGX0053] is identical to the predicted sequence. Plasmid construction was performed by use of standard molecular biological methods.

[00275] PINS expression was tested on culture supernatant (SN) from

[MG1363]pAGX0053 by Elisa and western blot. MG1363 [pAGX0053] secretes 2.47 ng/m PINS, as determined by use of Pro-Insulin Elisa (Mercodia #10-1 118-01). Crude SN samples were prepared for western blot Equivalents of 1 ml bacterial culture of [MG1363]pAGX0053 and reference strains MG1363[pTlNX] and sAGX0037 were loaded on the protein gel. Samples were incubated with goat polyclonal anti-insulin B (Santa Cruz N-20: sc-7838, 1/500). Detection was done by incubation with rabbit anti-goat AP (Southern Biotech #6160- 04, 1/1000) and subsequent NBT/BCIP staining (Roche NBT/BCIP tablets #1 1 697 471 001 ; used as indicated by the manufacturer). Invitrogen SeeBlue© Plus-2 pre-stained standard was used as molecular weight marker (MWM). Data is presented in FIG. 16 showing secretion of full-length PINS by LL-PINS.

[00276] Lactococcus lactis strains containing exogenous nucleic acids encoding T1D- specific antigens other than PINS, such as GAD65, IA-2, IGRP, ZnT8, ppIAPP, peripherin, chromogranin A, and GRP can be made in accordance with the above procedure using appropriate nucleic acids instead of hpins.

Example 8. Pharmacokinetic Profiling of Orally Administered LL-IL-2 Alone in Comparison to LL-PINS + LL-IL-2

Bacterial Culture

[00277] Bacteria were cultured as previously described in Takiishi, T. et al, J. Clin. Inv. 2012, 122(5): 1717-1725. For example, LL-pTlNX and LL-PINS were cultured in GM17TE medium (Ml 7 broth supplemented with 0.5% glucose, 200 μΜ thymidine, and 5 μg/mL erythromycin). LL-IL-2 was cultured in GM17T medium (Ml 7 broth supplemented with 0.5% glucose, and 200 μΜ thymidine). Stock suspensions of the LL strains were stored at -80°C in glycerol. Stock suspensions were diluted 1/1000 in growth media (GM17TE or GM17T, respectively) and incubated for 16 hours at 30°C, reaching a saturation density of 2 χ 10 9 CFU/mL. Bacteria were collected by centrifugation for 10 minutes at 4°C and 2900 rpm and concentrated 10-fold in BM9T medium (5 x M9 salts, 10% casitone, 10% glucose, 0.5 M NaHC0 3 , 0.5 M Na 2 C0 3 , 1 M MgCl 2 , 100M CaCl 2) and 100 mM thymidine) for intragastric inoculations. Treatment consisted of 100 μΐ, of this suspension for LL-pTlNX, LL-PINS and LL-IL-2. LL-PINS+LL-IL-2 were prepared by mixing equal parts of LL-PINS and LL-IL-2 suspensions. Treatment then consisted of 200 \xL of this suspension.

Administration Schedule and Dosing

[00278] New-onset diabetic NOD mice (2 consecutive blood glucose measurements over 200 mg/dL and positive glucosuria) received 2 χ 10 9 colony forming units (CFU) of live genetically modified Lactococcus lactis bacteria 5 times a week (weekdays) for 6 weeks. Mice treated with LL-PINS + LL-IL-2 received 2 x 10 9 CFU of LL-PINS and 2 x 10 9 CFU of LL-IL- 2. Control 1 mice received no treatment, and control 2 mice received bacteria carrying an empty vector (LL-pTlNX). After a 6 week treatment period, some mice were reserved for further analysis after 14 weeks post treatment initiation.

[00279] Normoglycemic NOD mice (22 weeks of age) received one dose of 2 x 10 9 CFU of LL-IL-2. Two, four, six or eight hours after dose administration mice were euthanized and whole blood and serum were collected. Proximal small intestine (PSI), distal small intestine (DSI), caecum (CAE), proximal colon (PCO), and distal colon (DCO) were collected in homogenization buffer (10 x M9 salts, 0.5 M NaHC0 3 , 0.5 M Na 2 C0 3 , 10% bovine serum albumin, distilled water) at a concentration of 100 mg/mL and mechanically dissociated. Homogenates of gut tissues and whole blood were plated in serial dilutions on GM17 plates to quantify bacterial recovery. Concentration of IL-2 in serum and homogenates were measured by ELISA.

Organ Harvesting

[00280] Six weeks after treatment initiation, mice were euthanized with C0 2 . Blood was collected by cardiac puncture with a heparinized needle. The blood was aliquoted (200 uL) for processing to single cells for flow cytometry and for plasma separation (centrifuged for 10 min at 2000 g at room temperature). Pancreata were harvested for histological analyses and stored in 5% paraformaldehyde (PFA). The following lymphoid organs were removed for analysis by flow cytometry; spleen (SPL), mesenteric lymph nodes (MLN) and pancreatic draining lymph nodes (PLN).

Histology of Pancreas and Insulitis Grading

[00281] PFA-fixed paraffin embedded pancreata were cut into sections of 6 um and collected 100 μπι apart, then stained with hematoxylin and eosin. Paraffin was removed using xylene followed by ethyl alcohol dehydration with 100%-90%-70%-50% ethanol solutions. Sections were rehydrated with tap water and distilled water. Sections were stained for 3 minutes in hematoxylin and rinsed with tap water. The sections were briefly rinsed in acid ethanol 3 times followed by an extensive wash with tap water. Samples were placed into saturated aqueous L12CO3, and rinsed in tap water. Then samples were put in eosin Y-solution (0.5% aqueous) and rinsed in tap water. Slides were dehydrated with 50% ethanol, 70% ethanol, 90% ethanol, twice with 100% ethanol and twice with xyleen, for 10 seconds during each step. The cover glass was mounted and islets were observed under a light microscope at 20x or 40x and graded objectively. Islet infiltration was scored as follows: 0 - no infiltration; 1 - peri-insulitis; 2 - islets with infiltration in less than 50% of the area; 3 - islets with infiltration in more than 50% of the area; 4 - completely destroyed islets / heavy insulitis.

C-Peptide ELISA

[00282] A commercially available ELISA kit for rat/mouse C-peptide (EZRMCP2-21 K, EMD Millipore, St. Charles, MO) was used to determine C-peptide levels in plasma. Briefly, the 96-well plate was washed 3 times with 1 x HRP wash buffer (50 mM Tris buffered saline containing Tween-20 diluted 1 : 10 with distilled water). Matrix solution (serum matrix containing 0.008% sodium azide) was added to blank, standards and quality control wells. Assay buffer (0.05 M phosphosaline, 0.025 M ethylenediaminetetraacedic acid (EDTA), 0.08% sodium azide, 1% bovine serum albumin (BSA), pH 7.4) was then added to all wells. Standards and quality controls, containing known levels of rat C-peptide, were added to the respective wells and undiluted mouse plasma was added. Antibody solution mixture (1 : 1 mixture of pre-titered capture and biotinylated detection antibody to C-peptide) was added and the plate was incubated at room temperature for 2 hours on an orbital microtiter plate shaker at moderate speed. Wells were washed 3 times with 1 x HRP wash buffer, and enzyme solution (pre-titered streptavidin-horseradish peroxidase) was added to each well to conjugate horseradish peroxidase to the immobilized biotinylated antibodies. The plate was incubated for 30 minutes at room temperature on a micro-titer plate shaker set to moderate speed. Wells were washed extensively with 1 HRP wash buffer. Substrate solution, containing 3,3'5,5'- tetra-methylbenzidine, was added to each well and the plate was shaken 15 minutes. The enzyme activity was stopped with stop solution (0.3 M HC1) and absorbance was measured at 450 nm within 5 minutes on a Victor spectrophotometer (Perkin Elmer). Lymphocyte Isolation from Lymphoid Organs

[00283] Organs were harvested and placed in ice-cold wash medium (RPMI 1640 medium (Life Technologies/Invitrogen) supplemented with 4.5% antibiotics (G418 sulfate) and 2% fetal calf serum (FCS). Organs were mashed through a 70 μπι strainer with wash medium. Cells were pelleted by centrifugation. For the spleen, NH4CI was added for 3 minutes at 37°C followed by washing with PBS. Cells were resuspended in 150 FACS buffer (1 x PBS, 0.1% BSA, 2 mM EDTA). The following fluorochrome-conjugated antibodies were used for staining: CD3-PerCP-Cy5.5 (145-2C11), CD4-APC-H7 (GK1.5, BD), CD8a-eFluor450 (53- 6.7), CD25-PE-Cy7 (PC61.5), CTLA4-PE (UClO-489), Foxp3-APC (FJK-165). All antibodies came from eBioscience unless mentioned otherwise. Anti-mouse CD16/CD32 (93, eBioscience) was used to block the Fc-γ II and III receptor to reduce non-specific binding of the fluorochrome-conjugated antibodies. Zombie Yellow Fixable Viability Dye (BioLegend) was used according to the manufacturer's specifications to stain dead cells.

Treg Cell Staining

[00284] Cells were washed with 1 x PBS prior to staining. Approximately 1 x 10 9 cells were pelleted in a 96 well plate, and resuspended in 50 μΐ of Zombie Yellow Fixable Viability dye diluted 1/500 in 1 x PBS. Cells were incubated at room temperature in the dark for 20 minutes. Cells were subsequently washed with 200 \xL FACS buffer, and incubated with antibodies against extracellular epitopes diluted in FACS buffer in the dark for 30 minutes at 4°C. Cells were washed with 200 μΙ_ FACS buffer. Cells were fixed and permeabilized in Fixation Permeabilization solution (Foxp3/Transcription Factor Staining Buffer Set, eBioscience) for 30 minutes at room temperature. Antibodies against intracellular epitopes were diluted in 1 x permeabilization buffer and incubated with cells in the dark for 30 minutes at 4°C. The cells were washed and resuspended in FACS buffer and filtered for acquisition. The following antibodies were used in the following dilutions: CD3 (PerCP-Cy5.5; 1/100); CD25 (PE-Cy7; 1/625); CD4 (APC-H7; 1/160); CD8a (eFluor450; 1/300); CTLA4 (PE; 1/200); Foxp3 (APC 1/200).

Flow Cytometry

[00285] Flow cytometry data was acquired on a BD Canto II with FACSDiva and were analyzed with Flow Jo® software (TreeStar). UltraComp eBeads™ (eBioscience) were used for compensation settings. Statistical Analysis

[00286] Statistical analyses were performed using GraphPad Prism 6 software. Results

[00287] Results generally indicate that (a) low dose IL2 (mucosally administered via LL-IL- 2) with or without proinsulin as autoantigen (e.g., administered as LL-PINS) can safely revert new-onset diabetes in mice, (b) induction and activation of Tregs

(CD4+CD25+Foxp3+CTLA4+) are possible mechanisms of action of LL-IL-2 and LL-IL- 2+LL-PINS therapy, and (c) initial blood glucose concentrations are a predictive factor for therapeutic success in mice.

Mucosal Delivery of LL-IL-2 Induces Long-Lasting Diabetes Remission in NOD Mice

[00288] Diabetes onset was diagnosed when mice had blood glucose measurements over 200 mg/dL on two consecutive days, in combination with positive glucosuria. Treatment success ("remission") was defined as having two consecutive blood glucose measurements below 200 mg/dL and complete absence of positive glucosuria.

[00289] Untreated diabetic NOD mice remained hyperglycemic and were euthanized when they had lost more than 20% of their initial body weight. Treatment with LL-pTlNX did not restore normoglycemia in diabetic mice. Mucosal delivery of LL-PINS caused diabetes reversal in about 12% of new-onset diabetic mice. Unexpectedly, after 6 weeks of treatment, LL-IL-2 alone caused diabetes reversal {i.e., resulted in re-established normoglycemia) in about 27% of new-onset diabetic mice. A combination therapy consisting of mucosal delivery of LL-IL-2 in combination with LL-PINS (LL-IL-2 + LL-PINS) restored normoglycemia after diabetes in about 30% of mice. LL-IL-2 and LL-IL-2+LL-PINS induced long-lasting diabetes remission for at least an additional 8 weeks of follow-up after treatment termination.

Compared to LL-IL-2 therapy alone, LL-IL-2+ LL-PINS therapy may reverse diabetes faster. Results are illustrated in FIG. 17.

Blood Glucose Concentrations at the Beginning of Therapy Impact Therapeutic Success

[00290] Therapy with LL-IL-2 and LL-IL-2+LL-PINS for 6 weeks reverted diabetes in about 45% and about 50%, respectively, of mice starting with blood glucose measurements below 350 mg/dL. For comparison, only about 8% of mice with a starting blood glucose measurement above 350 mg/dL were in remission. These results indicate that residual beta- cell mass at the initiation of treatment may predict therapeutic success. Results are illustrated in FIG. 18. FIGs. 22A and 22B illustrates blood glucose concentrations (mg/dL) in recent onset diabetic mice treated with LL-IL-2 alone (FIG. 22A) and LL-IL-2+LL-PINS (FIG. 22B) over the treatment and follow-up periods.

Mucosal Delivery of LL-IL-2 Preserves Functional Beta-Cell Mass

[00291] Plasma C-peptide can reflect pancreatic insulin content {see, e.g., Suarez-Pinzon WL et al, Combination therapy with glucagon-like peptide- 1 and gastrin restores

normoglycemia in diabetic NOD mice. Diabetes 2008; 57:3281-8.). C-peptide levels in mice treated and cured with LL-IL-2 (113.23 ± 28.16 pM, n = 3) and LL-IL-2+LL-PINS (167.63 ± 64.38 pM, n = 9) was 19% and 28%, respectively, of the C-peptide values measured in longstanding normoglycemic NOD mice (602.93 ± 293.43 pM, n = 6) and about twice as high as the values found in untreated long-standing diabetic NOD mice (n = 4; 58.23 ± 100.86 pM). C-peptide levels can be a measure for the amount of endogenous insulin produced by remaining islets. When NOD mice turn diabetic C-peptide levels drop, and in untreated longstanding diabetic mice, C-peptide levels were expected to be very low or non-detectable. This was in fact observed in 3 out of the 4 analyzed mice. Only one mouse had detectable levels of C-peptide. This observation suggests that functional beta-cells are still present at diabetes diagnosis and are preserved by the therapy. Mice treated with LL-IL-2+LL-PINS and exhibiting diabetic remission had statistically significantly higher C-peptide levels when compared to new-onset diabetic mice (p<0.05). Non-cured mice had undetectable C-peptide values, similar to what was found in most long-standing diabetic mice. Results indicate that recent onset animals have some beta cell function left. Cured mice had C-peptide levels comparable to the level found in recent onset diabetic mice. Non cured animals lost beta cell function represented by non-detectable C-peptide levels. Results are illustrated in FIG. 19.

Mucosal Delivery of LL-IL-2 Halts Insulitis Progression

[00292] Histological analysis of the pancreas after diabetes onset revealed pancreatic islets heavily infiltrated by leukocytes and only a few islets with remaining beta-cells. LL-IL-2 and LL-IL-2+LL-PINS halted {i.e., prevented worsening) of insulitis. Worsening of insulitis is typically observed during progression from new-onset to longstanding diabetes. Unexpectedly, both therapies improved the degree of insulitis when compared to insulitis found in longstanding normoglycemic NOD mice. While the percentage of islets with heavy insulitis was not significantly affected, both therapies dramatically increased the insulitis- free area. Even more unexpectedly, this improvement was also observed in animals not reaching normoglycemia ("non-cured" animals). Results are illustrated in FIG. 20.

[00293] The hematoxylin and eosin staining did not allow for determining which immune cells infiltrated the pancreas, i.e. effector T cells (Teff) versus Treg cells.

Mucosal delivery of LL-IL-2 Induces Expansion of CD4+Foxp3+CTLA4+ Treg-cells

[00294] Effects of LL-IL-2 on different immune cell subsets, both locally (i.e., MLN), systemically (i.e., spleen and blood) and at the target organ (i.e., PLN) were measured using flow cytometric analysis. Because low-dose IL-2 given systemically can induce expression of Treg cell-associated proteins including Foxp3, CD25, and CTLA4, the frequencies of CD4+, CD8+, and CD4+Foxp3+ cells or their activation status (CD44, CD62L) were determined.

[00295] CD4+ and CD8+ T-cell populations within live CD3+ T-cells were assessed. A small decrease of CD4+ T-cells in MLN was detected with LL-IL-2 and LL-IL-2+LL-PINS therapy in non-cured mice compared to cured animals (p = 0.044 and p = 0.068 respectively). In the examined peripheral organs (i.e. blood and spleen) and target organ (i.e. PLN), there were no statistically significant changes in the number of these leukocytes. Differences in CD4+ T-cell frequencies were limited to sites exposed to LL-IL-2 and LLIL-2+LL-PINS locally. Inversely, the CD8+ T-cell population was increased in the MLN of LL-IL-2+LL- PINS therapy non-cured mice compared to cured mice (p = 0.012). This trend was also present in LL-IL-2 therapy. Furthermore, LL-IL-2 and LL-IL-2+LL-PINS treated non-cured mice had significantly higher CD8+ splenic T-cells (p = 0.025 and p = 0.01 1 respectively) compared to new-onset diabetic NOD mice. This systemic change may have been a consequence of disease progression rather than of L. lactis-based therapies.

[00296] The presence of Treg cells was also assessed using flow cytometry. At 6 weeks after therapy initiation, PLN of LL-IL-2 and LL-IL-2+LL-PINS cured mice showed a trend for higher frequencies of Foxp3+CTLA-4+ Treg cells, compared to new-onset diabetic mice (both about 17% compared to about 10% in recent-onset mice). This trend was also observed in LL- IL-2 and LL-IL-2+LL-PINS treated non-cured mice (about 14% and about 17%, respectively).

[00297] In the spleen, non-cured mice exhibited a decrease of this Treg cell compartment when compared to cured mice, and compared to untreated recent-onset mice. In blood, both therapies induced a trend towards an increase in circulating Foxp3+CTLA-4+ Treg cells when compared to new-onset diabetic mice. Again, this increase was independent of therapy outcome and in non-cured LL-IL-2+LL-PINS treated mice this increase was significant (p = 0.009). In the MLN, LL-IL-2+LL-PINS non-cured mice also have significantly higher frequencies of Foxp3+CTLA4+ Treg cells compared to new-onset diabetic mice (p= 0.002). The above results are illustrated in FIG. 21.

[00298] Treg cell compartments were further defined measuring CD25 expression. A considerable number of Foxp3+CTLA4+ Treg cells did not express CD25 (CD25 ), but therapy with LL-IL-2 and LL-IL-2+LL-PINS still induced changes in this population. CD25- Foxp3+CTLA-4+ Treg cells were relatively abundant in the blood and were increased in LL- IL-2+LL-PINS non-cured mice (p = 0.004), and in LL-IL-2 cured mice. In the spleen, a trend towards a decrease in this subset was observed in LL-IL-2 and LL-IL-2+LL-PINS treated non- cured mice compared to cured mice (p = 0.06 and p = 0.06 respectively). In mesenteric lymph nodes, there was little effect of LL-IL-2 or LL-IL-2+LL-PINS on CD25Toxp3+CTLA-4+ Tregs. In the target lymph nodes (i.e. PLN), this Treg cell subset was increased in LL-IL-2 and LL-IL-2+LL-PINS cured mice compared to new-onset diabetic mice.

[00299] The following trends were observed for CD25+ cells: In MLN,

CD25+Foxp3+CTLA-4+ Treg cells were increased in non-cured mice treated with LL-IL-2 and LL-IL-2+LL-PINS when compared to new-onset diabetic mice (12% and 14% vs 7%, respectively). This was statistically significant in the latter group (p = 0.001). In PLN, there was a visible trend towards an increase of this subset in LL-IL-2 and LL-IL-2+LL-PINS treated groups.

[00300] The distribution pattern of Foxp3+CTLA-4+ Treg cells seen in blood and spleen was similar upon further classification based on CD25 expression. In the local and target lymph nodes (i.e. the MLN and PLN), Foxp3+CTLA-4+ Treg cell increases were seen following both LL-IL-2 and LL-PINS+IL-2 therapies, which could be attributed largely to the CD25+Treg cell compartment.

[00301] The spleen size in new onset diabetic NOD mice treated with LL-PINS+IL-2 were assessed for the total period of 6 weeks (n = 3). No splenomegaly was observed. Treatment with LL-IL-2 as well as LL-IL-2+LL-PINS was safe and well tolerated.

Conclusion

[00302] LL-IL-2 and LL-IL-2+LL-PINS therapies are safe and well-tolerated, and induced diabetes remission in new-onset diabetic NOD mice. Both therapies have beneficial metabolic and immune effects. Unexpectedly, some effects are also present in non-cured mice, suggesting a potential benefit for subjects, which did not reach normoglycemia during the course of the experiments, or for which the treatment comes too late because the remaining beta cell mass was below a certain threshold when treatment began (in NOD mice the process of beta cell loss is faster when compared to the loss observed in humans). Thus, an immune effect may have been observed in all treated animals, but this effect may have translated into the defined therapeutic effect only in those animals that had sufficient beta cell mass left at the beginning of treatment.

[00303] LL-IL-2 and LL-IL-2+LL-PINS therapies halted insulitis progression, possibly through a mechanism involving expansion of Treg cells. Differences in the percentage of Foxp3+CTLA4+ Treg cells between treated and untreated new-onset diabetic mice were found in the PLN after LL-IL-2 and LL-IL-2+LL-PINS therapy. The increase in the number of Treg cells induced by the therapies may be a result of an improvement of Treg cell survival {see, e.g., Tang Q, Bluestone JA. Nat. Immunol. 2008; 9(3): 239-244). Alternatively, the increased number of Treg cells could be the result of the conversion of effector T cells (Teff) into induced Treg cells (Zheng Y, Rudensky AY. Nat. Immunol. 2007; 8(5): 457-462), or the increased recruitment of Treg cells to the PLN (Grinberg-Bleyer Y. et al, J. Exp. Med. 2010; 207(9): 1871-1878).

Example 9. Effect of anti-CD3 antibody administration upon diabetes remission rate

[00304] An anti-CD3 antibody dose finding study was performed in mice. Mice with a starting blood glucose level of greater than 200 mg/dl were dosed with 2.5 μg/day; 5 μg/day; 10 μg/day; 25 μg/day; or 50 μg/day of an anti-CD3 antibody (teplizumab) through IV injections. Dosages of 25 μg/day or 50 μg/day exhibited some toxicity. Treatments were given over a 5 day period, which means that a cumulative anti-CD3 antibody dosage was 12.5ig; 25 μg; 50 μg; 125 μg; or 250 μg. The treatment group receiving 10 μg/day resulted in a maximal remission induction of approximately 50%. However, in order to reduce toxicity over a longer period of treatment, the dosage of 2.5 μg/day (12.5 μg cumulative) was selected, which is considered a sub-therapeutic dose (low-dose). Treatment with 2.5 μg/day of anti-CD3 antibody was less effective than at the higher 10 μg/day, and resulted in a remission induction of approximately 20%.

[00305] Similar to Example 8, LL-PINS, LL-IL2, LL-PINS+LL-IL2, were given by intragastric inoculation (2x10 9 CFU/d), 5 times per week for 6 weeks to newly diabetic mice. Additionally, hamster anti-mouse CD3 monoclonal antibody (mAb) (145-2C11 , BioXCell, New Hampshire, USA) was administered intravenously (2.5 μg/d) for 5 consecutive days to mice received LL-PINS+LL-IL2 inoculation. All tested newly diabetic mice had starting blood glucose concentrations below 350 mg/dl. Weight and glycemia were measured 3 times per week. Diabetes remission was defined as absence of glycosuria and glycemia values < 200 mg/dl on two consecutive days.

[00306] As shown in FIG. 23, therapies with LL-PINS (n=5), LL-IL2 (n=l 3), and a mixture of LL-PINS+LL-IL2 (n=31) inoculation corrected hyperglycemia in 20%, 30.8%, and 45.2% of mice, respectively. Newly diabetic mice (n=3) treated with a combination of LL-PINS + LL-IL2 and anti-CD3 re-established normoglycemia in 66.7% of the mice. These data suggest that an additional immune-modulating substance, for example, an anti-CD3 antibody, improves the outcome of LL-IL-2+LL-PINS therapy in subjects having a lower starting blood glucose concentration.