Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HIGH-AFFINITY MYCOBACTERIUM TUBERCULOSIS CAPSULE-SPECIFIC HUMAN MONOCLONAL ANTIBODY
Document Type and Number:
WIPO Patent Application WO/2019/200255
Kind Code:
A1
Abstract:
Provided are high affinity Mycobacterium tuberculosis capsule-specific antibodies and fragments thereof, as well as methods of use and devices employing such antibodies and/or fragments.

Inventors:
ACHKAR JACQUELINE (US)
LAI JONATHAN (US)
ISHIDA ELISE (US)
HOFMANN DANIEL (DE)
CHEN TINGTING (US)
Application Number:
PCT/US2019/027218
Publication Date:
October 17, 2019
Filing Date:
April 12, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ALBERT EINSTEIN COLLEGE OF MEDICINE (US)
International Classes:
A61K39/04; A61K39/395; A61P31/06; C07K16/12
Foreign References:
US20020034763A12002-03-21
US20160083458A12016-03-24
US20160312314A12016-10-27
US20170002064A12017-01-05
Other References:
DATABASE Genbank [online] 11 September 2017 (2017-09-11), ULLY, BJ ET AL: "Hypothetical Protein", Database accession no. CMB92_01875
Attorney, Agent or Firm:
MACLEOD, Janet, M. et al. (US)
Download PDF:
Claims:
What is claimed is:

1. An anti -Mycobacterium tuberculosis arabinomannan (anti-Mtb AM) antibody, or Mycobacterium tuberculosis arabinomannan-binding fragment (Mtb AM-binding fragment) thereof, wherein said antibody or fragment thereof

(a) (i) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 3, but (ii) does not comprise complementarity determining region- 1 (CDRH1) of SEQ ID NO:l or does not comprise complementarity determining region- 1 (CDRH2) of SEQ ID NO:2; or

(b) (i) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 23, but (ii) does not comprise complementarity determining region-l (CDRH1) of SEQ ID NO:2l or does not comprise complementarity determining region-l (CDRH2) of SEQ ID NO:22; or

(c) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 31; or

(d) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 32.

2 The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of Claim 1, comprising VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

3. An anti -Mycobacterium tuberculosis arabinomannan (anti-Mtb AM) antibody, or Mycobacterium tuberculosis arabinomannan-binding fragment thereof, wherein said antibody or fragment thereof comprises:

(i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 1, 2 and 9; or SEQ ID NOS: 7, 8 and 3; or SEQ ID NOS: 21, 22 and 28; or SEQ ID NOS: 26, 27 and 23; or SEQ ID NOS: 21, 22 and 31; or SEQ ID NOS: 21, 22 and 32; SEQ ID NOS: 7, 8 and 32, and (ii) VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO 30.

4. The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of any of Claims 1- 3, comprising (i) VH CDR amino acid sequences of SEQ ID NOS: 7, 8 and 3; or SEQ ID NOS: 26, 27 and 23, and

(ii) VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

5. The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of any of Claims 1- 3, comprising (i) VH CDR amino acid sequences of SEQ ID NOS: 7, 8 and 32, and

(ii) VL CDR amino acid sequences of SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

6. The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of any of Claims 1-

3, comprising (i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 21, 22 and 31, and

(ii) VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

7. The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of any of Claims 1- 3, comprising (i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 21, 22 and 31, wherein in SEQ ID NO: 21, X = A, and

(ii) VL CDR amino acid sequences of SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25.

8. The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of any of Claims 1- 3, comprising (i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 21, 22 and 31, wherein in SEQ ID NO: 21, X = S, and (ii) VL CDR amino acid sequences of SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25.

9. The anti-Mtb AM antibody or Mtb AM-binding fragment thereof of any of Claims 1- 8, wherein the antibody is a monoclonal antibody.

10. The antibody, or Mtb AM-binding fragment thereof, of any of Claims 1 to 9, wherein said antibody comprises (i) a VH framework comprising the framework sequence of human germline VHl-2*02; and/or (ii) a VL framework comprising the framework sequence of human germline IGKV1-39.

11. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-10, comprising a VH that comprises the amino acid sequence of SEQ ID NO: 16 or 18 or 19 or 20.

12 The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-11, comprising a VL that comprises the amino acid sequence of SEQ ID NO: 15 or 17.

13. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-12, wherein the antibody, or Mtb AM-binding fragment thereof, binds Mtb AM with a binding affinity (Kd) of from about lxlO 6M to about lxlO 9M.

14. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-13, which is a monoclonal antibody.

15. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-4, which is a recombinant antibody.

16. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-15, which has a human framework region or a modified human constant region.

17. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-16, which has a human constant region or a modified human constant region.

18. The anti-Mtb AM antibody of any of Claims 1-17.

19. The anti-Mtb AM-binding fragment of any of Claims 1-18.

20. The anti-Mtb AM-binding fragment of Claim 19 which is an Fab, F(ab)2 orscFv.

21. An isolated nucleic acid molecule encoding the antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-20.

22 A vector comprising the nucleic acid molecule of Claim 21.

23. A host cell comprising the nucleic acid molecule of Claim 21, or the vector of Claim

22.

24. A method of producing an anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, comprising culturing the host cell of Claim 23, under conditions wherein the anti- Mtb AM antibody, or Mtb AM-binding fragment thereof, is produced by the host cell.

25. A pharmaceutical composition comprising an anti-Mtb AM antibody, or Mtb AM- binding fragment thereof, of any one of Claims 1-20, and a pharmaceutically acceptable excipient.

26. A method of reducing an activity of Mtb AM in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-20, or the pharmaceutical composition of Claim 25.

27. A method of treating a Mycobacterium tuberculosis infection in a subject, comprising administering to the subject an amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-20, or the pharmaceutical composition of Claim 25, effective to treat a Mycobacterium tuberculosis infection.

28. A method of reducing the likelihood of an Mycobacterium tuberculosis infection in a subject, comprising administering to the subject who does not have a Mycobacterium tuberculosis infection an amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1-20, or the pharmaceutical composition of Claim 25, effective to reduce the likelihood of an Mycobacterium tuberculosis infection.

29. A method of treating a disease, disorder, or condition mediated by, or related to increased activity of Mycobacterium tuberculosis in a subject, comprising administering to said subject a therapeutically effective amount of the anti-Mtb AM antibody, or Mtb AM- binding fragment thereof, of any one of Claims 1 -20 or the pharmaceutical composition of Claim 25.

30. An assay device for selectively detecting a virulent Mycobacterium tuberculosis in a biological sample comprising:

a first portion comprising a first plurality of anti- virulent Mycobacterium tuberculosis antibodies:

of any of Claims 1-20;

comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or

comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17,

wherein the antibodies are each attached to their own reporting entity; and

a second portion comprising a second plurality of anti -Mycobacterium tuberculosis antibodies.

31. The device of Claim 30, wherein the reporting entity comprises a gold nanoparticle. 32 The device of Claim 30, wherein the reporting entity comprises an enzyme.

33. The device of any of Claims 30 to 32, wherein the second plurality of virulent Mycobacterium tuberculosis antibodies is affixed to a solid support of the device.

34. The device of any of Claims 30 to 33, wherein the first plurality of virulent Mycobacterium tuberculosis antibodies is not affixed to a solid support of the device.

35. The device of any of Claims 30 to 34, wherein the solid support

comprises nitrocellulose.

36. The device of any of Claims 30 to 35, further comprising a fluid sample pad prior in sequential order to the first and second portions.

37. The device of any of Claims 30 to 36, further comprising a control portion subsequent in sequential order to the first and second portions.

38. The device of Claim 37, wherein the control portion comprises a third plurality of antibodies, immobilized on a solid support of the device, and which third plurality of antibodies are capable of binding the first plurality of virulent Mycobacterium tuberculosis antibodies each attached to their own reporting molecule.

39. The device of any of Claims 30 to 38, further comprising a fluid-absorbent wicking pad subsequent in sequential order to the first and second portions, and third portion if present.

40. The device of any of Claims 30 to 39, wherein the second plurality ofanti- Mycobacterium tuberculosis antibodies comprise antibodies of any of any of Claims 1-20; comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or

comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17,

41. The device of any of Claims 30 or 32-40, wherein the reporting entity is an enzyme which is horseradish peroxidase (HRP) or alkaline phosphatase (AP).

42 A lateral flow assay device for detecting a Mycobacterium tuberculosis in a biological sample comprising:

a first portion comprising a first plurality of anti -Mycobacterium tuberculosis antibodies: of any of Claims 1-20;

comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or

comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17,

wherein the antibodies are each attached to their own reporting entity; and a second portion comprising a second plurality of anti -Mycobacterium tuberculosis antibodies.

43. The device of Claim 42, wherein the reporting entity comprises a gold

nanoparticle, horseradish peroxidase (HRP) or alkaline phosphatase (AP).

44. The device of Claim 42, wherein the reporting entity comprises an enzyme.

45. The device of any of Claims 42 to 44, wherein the second plurality of

Mycobacterium tuberculosis antibodies is affixed to a solid support of the device.

46. The device of any of Claims 42 to 45, wherein the first plurality of

Mycobacterium tuberculosis antibodies is not affixed to a solid support of the device.

47. The device of any of Claims 42 to 46, wherein the solid support

comprises nitrocellulose.

48. The device of any of Claims 42 to 47, further comprising a fluid sample pad prior in sequential order to the first and second portions.

49. The device of any of Claims 42 to 48, further comprising a control portion subsequent in sequential order to the first and second portions.

50. The device of Claim 49, wherein the control portion comprises a third plurality of antibodies, immobilized on a solid support of the device, and which third plurality of antibodies are capable of binding the first plurality of virulent Mycobacterium tuberculosis antibodies each attached to their own reporting molecule.

51. The device of any of Claims 42 to 50, further comprising a fluid-absorbent wicking pad subsequent in sequential order to the first and second portions, and third portion if present.

52 The device of any of Claims 42 to 51 , wherein the second plurality of anti- Mycobacterium tuberculosis antibodies comprise antibodies

of any of Claims 1-20;

comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or

comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17.

53. A method of detecting a virulent Mycobacterium tuberculosis in a biological sample comprising contacting the device of any of Claims 30-41 with the sample and observing if virulent Mycobacterium tuberculosis- bound antibodies bind to the second plurality of virulent Mycobacterium tuberculosis- binding antibodies, wherein if such antibodies bind then virulent Mycobacterium tuberculosis has been detected in the biological sample and wherein if no virulent Mycobacterium tuberculosis- bound antibodies bind to the second plurality of virulent Mycobacterium tuberculosis- binding antibodies then virulent Mycobacterium tuberculosis has not been detected in the biological sample.

54. The method of Claim 53, further comprising obtaining the sample from asubject.

55. The method of Claim 53 or 54, wherein the sample is urine or blood.

56. The method of any of Claims 26-29 or 54-55, wherein the subject is human.

57 The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1 to 20, wherein the antibody comprises SEQ ID NO:l6 and SEQ IDNO:l7.

58. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1 to 20, wherein the antibody comprises SEQ ID NO: 18 and SEQ IDNO:l7.

59. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1 to 20, wherein the antibody comprises SEQ ID NO:l9 and SEQ IDNO:l5.

60. The antibody, or Mtb AM-binding fragment thereof, of any one of Claims 1 to 20, wherein the antibody comprises SEQ ID NO:20 and SEQ IDNO:l5.

Description:
HIGH-AFFINITY MYCOBACTERIUM TUBERCULOSIS CAPSULE-SPECIFIC

HUMAN MONOCLONAL ANTIBODY

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority under 35 U.S.C. §119(e) to U. S. Provisional

Application Serial No. 62/657,253 filed April 13, 2018 and U. S. Provisional Application Serial No. 62/739,428 filed October 1, 2018, the disclosures of which are incorporated herein in their entireties.

STATEMENT OF GOVERNMENT SUPPORT

[0002] This invention was made with government support under grant numbers All 25462 and AI127173 awarded by the National Institutes of Health. The government has certain rights in the invention.

FIELD OF THE INVENTION

[0003] This disclosure relates generally to antibodies against Mycobacterium tuberculosis capsular polysaccharides and methods of use thereof.

BACKGROUND OF THE INVENTION

[0004] The capsule of microorganisms is an important virulence factor. Antibodies (Abs) to capsular and surface polysaccharides are protective against infections with encapsulated extra- and intracellular pathogens. Some successful vaccines are based on inducing Abs to capsular polysaccharides. The mechanisms by which Abs protect against Mycobacterium tuberculosis (Mtb) have been insufficiently studied because of the general belief that Mtb, a predominantly intracellular organism, is outside the reach of extracellular located Abs. However, Abs contribute to the defense against many intracellular pathogens, including Mtb, through various functions, including interactions with Fc receptors (FcR) and the modulation of innate and other immune responses. Mycobacteria have a capsule, an important virulence factor. The majority of the mycobacterial capsule is composed of proteins and polysaccharides; lipids are a minor component of the capsule. The three major capsular polysaccharides are a-glucan, arabinomannan (AM) and mannan. The 13 kDa, immunogenic polysaccharide AM can be isolated from the capsule of Mtb. AM is structurally related to lipoarabinomannan (LAM), a glycolipid from the cell walls and membranes of mycobacteria, and both AM and LAM are very immunogenic. Some but not all murine mAbs to AM/LAM show protective in vivo efficacy, and immunization with AM/LAM- protein conjugates improves the outcome of Mtb infected mice. However, these studies are limited in capturing the tremendous complexity and heterogeneity of potentially Mtb protective Abs in humans, but are consistent with the data that not all‘anti- AM’ mAbs have the same binding specificity or protective ability. Several recent studies provide compelling data suggesting that Mtb specific antibodies have a role in controlling Mtb infection in humans and could be protective but very little is known about the functions of antigen-specific human mAbs in Mtb infection.

BRIEF SUMMARY OF THE INVENTION

[0005] An anti -Mycobacterium tuberculosis arabinomannan (anti-Mtb AM) antibody, or

Mycobacterium tuberculosis arabinomannan-binding fragment (Mtb AM-binding fragment) thereof, is provided, wherein said antibody or fragment thereof:

(a) (i) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 3, but (ii) does not comprise complementarity determining region-l (CDRH1) of SEQ ID NO:l or does not comprise complementarity determining region-l (CDRH2) of SEQ ID NO:2; or

(b) (i) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 23, but (ii) does not comprise complementarity determining region-l (CDRH1) of SEQ ID NO:2l or does not comprise complementarity determining region-l (CDRH2) of SEQ ID NO:22; or

(c) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 31; or

(d) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 32.

[0006] An anti -Mycobacterium tuberculosis arabinomannan (anti-Mtb AM) antibody, or

Mycobacterium tuberculosis arabinomannan-binding fragment thereof, is provided, wherein said antibody or fragment thereof comprises:

(i) VH complementarity determining region (CDR) amino acid sequences o SEQ ID NOS: 1, 2 and 9; or SEQ ID NOS: 7, 8 and 3; or SEQ ID NOS: 21, 22 and 28; or SEQ ID NOS: 26, 27 and 23; or SEQ ID NOS: 21, 22 and 31; or SEQ ID NOS: 21, 22 and 32; or SEQ ID NOS: 7, 8 and 32, and (ii) VL CDR amino acid sequences SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30; or SEQ ID NOS: 21, 22 and 32.

[0007] An isolated nucleic acid molecule encoding the antibody, or Mtb AM-binding fragment thereof, described herein is provided. In an embodiment, the nucleic acid is a DNA. In an embodiment, the nucleic acid is a cDNA. In an embodiment, the nucleic acid is an RNA.

[0008] A vector encoding the nucleic acid molecule described herein is provided.

A host cell comprising the nucleic acid molecule described herein, or the vector described herein, is provided.

[0009] A method of producing an anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, comprising culturing the host cell described herein, under conditions wherein the anti- Mtb AM antibody, or Mtb AM-binding fragment thereof, is produced by the host cell, is provided herein.

[0010] A pharmaceutical composition comprising an anti-Mtb AM antibody, or Mtb

AM-binding fragment thereof, described herein, and a pharmaceutically acceptable excipient, is provided.

[0011] A method of reducing an activity of Mtb AM in a subject in need thereof is provided, comprising administering to said subject a therapeutically effective amount of the anti- Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein.

[0012] A method of treating a Mycobacterium tuberculosis infection in a subject, comprising administering to the subject an amount of the anti-Mtb AM antibody, or Mtb AM- binding fragment thereof, as described herein, or the pharmaceutical composition described herein, effective to treat a Mycobacterium tuberculosis infection, is provided herein.

[0013] A method of reducing the likelihood of an Mycobacterium tuberculosis infection in a subject, comprising administering to the subject who does not have a Mycobacterium tuberculosis infection an amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein, or the pharmaceutical composition described herein, effective to reduce the likelihood of an Mycobacterium tuberculosis infection, is provided herein.

[0014] A method of treating a disease, disorder, or condition mediated by, or related to increased activity of Mycobacterium tuberculosis in a subject, comprising administering to said subject a therapeutically effective amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein, or the pharmaceutical composition described herein, is provided herein. [0015] An assay device is provided for selectively detecting a virulent Mycobacterium tuberculosis in a biological sample comprising: a first portion comprising a first plurality of anti- virulent Mycobacterium tuberculosis antibodies or Mtb AM-binding fragments thereof, as described herein, wherein the antibodies are each attached to their own reporting entity; and a second portion comprising a second plurality of anti -Mycobacterium tuberculosis antibodies.

[0016] Also provided is a lateral flow assay device for detecting a Mycobacterium tuberculosis in a biological sample comprising: a first portion comprising a first plurality of anti- Mycobacterium tuberculosis antibodies as described herein;

comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or

comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17,

wherein the antibodies are each attached to their own reporting entity; and

a second portion comprising a second plurality of anti -Mycobacterium tuberculosis.

BRIEF DESCRIPTION OF THE DRAWINGS

[0017] Figs. 1A, 1B, 1C, 1B, and 1E show that anti-AM IgG titers in sera significantly correlate with mycobacteria phagocytosis, growth rate, and reactivity to certain AM OS fragments (adapted from Chen, et al. 2016, J. Infect. Pis. 214(2) 300-10.). Fig. 1A shows anti- AM IgG responses after primary BCG vaccination increases Wilcoxon matched-pairs signed rank test. Fig. 1B shows the significant correlation between 4 weeks post-vaccination IgG responses to AM and enhanced BCG phagocytosis (in fold change compared to co-incubation with pre- vaccination sera) by THP-l cells co-incubated with corresponding 4 weeks post-vaccination sera using Spearman rank correlation test. Fig. 1C shows the significant BCG growth reduction in THP-l cells incubated with post- compared to pre-vaccination sera. Fig. 1D shows the correlation between 8 weeks post-vaccination IgG titers and mycobacterial growth reduction using the Spearman rank correlation test. Fig. 1E shows the significant correlations between increased IgG reactivity to certain AM epitopes at 4 weeks post vaccination and enhanced BCG phagocytosis by human macrophages co-incubated with corresponding sera.

[0018] Figs. 2A, 2B, 2C, and 2D show that serum from a PPD+ subject, coded V57, in the presence of AM-specific IgG has protective functions in vitro and in vivo. Figure 2A shows V57 serum recognition of AM oligosaccharides before depletion and after depletion of AM- specific IgG. Fig. 2B shows the decrease in Mtb phagocytosis after serum is depleted of AM- specific IgG. Fig. 2C shows the increase in intracellular Mtb growth after serum is depleted of AM-specific IgG. Fig. 2D shows that V57 serum decreases the bacterial burden in the lungs of mice infected with Mtb.

[0019] Figs. 3A, 3B, 3C, 3D, and 3E depict the characterization of monoclonal antibody

(mAb) AM009’s binding properties. Fig. 3A shows mAb AM009 binds whole Mtb bacteria with higher affinity than other anti-AM mAbs by whole bacteria ELISA. Fig. 3B shows that AM009 in an IgGl or IgG2 backbone significantly enhances opsonophagocytosis of Mtb; *p<0.05 one- way ANOVA; Mann- Whitney. Fig. 3C shows germline and mutation frequency information for AM009. Fig. 3D shows that AM009 binds with increased specificity to capsular arabinomannan (AM) isolated and purified from Mtb (strain H37Rv) compared to other surface polysaccharide components. Fig. 3E shows that TRITC-labeled anti-human IgG was used to detect AM009 binding to Mtb with an intact capsule (H37Rv grown without detergent).

[0020] Figs. 4A, 4B, 4C, and 4D show that a point mutation in the CDRH3 increases the affinity of mAb AM009. Fig.4A is a schematic of alanine scanning of AM009 CDRH3 (GILLNGIGAFDY, SEQ ID NO:3) showing that point mutation GlOOb to AlOOb is important for binding of AM009 to AM. Fig. 4B shows that the mean fluorescence intensity (MFI) on the synthetic AM oligosaccharide glycan array is increased in AM009 GlOObA (m8) compared to AM009 (wt). Fig. 4C shows that the mAb recognition of oligosaccharide glycan fragments is distinct when AM009 GlOObA (m8) is compared to murine mAb CS-35. This indicates that AM009 recognizes a different AM glycan epitope and has different binding properties than murine anti-LAM mAb CS-35. CS-35 is a murine mAb generated from M. leprae LAM. CS-35 cross-reacts with LAM and AM from many strains and is the standard positive control used in the field. Fig. 4D depicts raw images of the glycan array showing mAb binding reactivity to AM-OS fragments printed in triplicate.

[0021] Figs. 5A and 5B show that AM009 has novel binding kinetics and polysaccharide binding patterns specific for a glycan epitope in virulent H37Rv and CDC1551 AM. Fig. 5 A depicts Bio layer Interferometry (BLI) data showing the unusually high binding affinity of AM009 to the mycobacterial capsular polysaccharide arabinomannan (AM). Kinetic characterization of mAb-AM (H37Rv) interaction with solid lines representing experimental data and dashed lines representing the statistical fitting of curves is shown. The mutation in AM009 GlOObA resulted in an overall higher affinity binding to AM. Data also show distinct binding kinetics of AM009 versus murine mAb CS-35. Fig. 5B shows MAb binding by ELISA to AM isolated from five mycobacterial strains. It shows that AM009 has greater specificity for AM isolated from virulent strains CDC1551 and H37Rv compared to AM isolated avirulent strains H37Ra and BCG. One- way ANVOA was used to test a group comparison of the EC50s from two independent experiments. AM009 GlOObA and murine mAb, CS-35 show significant difference in binding AM from different mycobacterial strains

DETAILED DESCRIPTION OF THE INVENTION

[0022] An anti -Mycobacterium tuberculosis arabinomannan (anti-Mtb AM) antibody, or

Mycobacterium tuberculosis arabinomannan-binding fragment (Mtb AM-binding fragment) thereof, is provided, wherein said antibody or fragment thereof

(a) (i) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 3, but (ii) does not comprise complementarity determining region-l (CDRH1) of SEQ ID NO:l or does not comprise complementarity determining region-l (CDRH2) of SEQ ID NO:2; or

(b) (i) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 23, but (ii) does not comprise complementarity determining region-l (CDRH1) of SEQ ID NO:2l or does not comprise complementarity determining region-l (CDRH2) of SEQ ID NO:22; or

(c) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 31; or

(d) comprises VH complementarity determining region-3 (CDRH3) amino acid sequence of SEQ ID NO: 32.

[0023] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

[0024] An anti -Mycobacterium tuberculosis arabinomannan (anti-Mtb AM) antibody, or

Mycobacterium tuberculosis arabinomannan-binding fragment thereof, is provided, wherein said antibody or fragment thereof comprises:

(i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 1, 2 and 9; or SEQ ID NOS: 7, 8 and 3; or SEQ ID NOS: 21, 22 and 28; or SEQ ID NOS: 26, 27 and 23; or SEQ ID NOS: 21, 22 and 31; or SEQ ID NOS: 21, 22 and 32; SEQ ID NOS: 7, 8 and 32 and (ii) VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NOS: 29, the sequence GIS and SEQ ID NO: 30.

[0025] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises (i) VH CDR amino acid sequences of SEQ ID NOS: 7, 8 and 3; or SEQ ID NOS: 26, 27 and 23, and

(ii) VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

[0026] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises (i) VH CDR amino acid sequences of SEQ ID NOS: 7, 8 and 32, and

(ii) VL CDR amino acid sequences of SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

[0027] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises (i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 21, 22 and 31, and

(ii) VL CDR amino acid sequences of SEQ ID NOS: 4, 5 and 6; or SEQ ID NOS: 10, 11 and 12; or SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25; or SEQ ID NO: 29, the sequence DAS and SEQ ID NO: 30.

[0028] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises (i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 21, 22 and 31, wherein in SEQ ID NO: 21, X = A, and

(ii) VL CDR amino acid sequences SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25.

[0029] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises (i) VH complementarity determining region (CDR) amino acid sequences of SEQ ID NOS: 21, 22 and 31, wherein in SEQ ID NO: 21, X = S, and

(ii) VL CDR amino acid sequences of SEQ ID NO: 24, the sequence GIS and SEQ ID NO: 25.

[0030] In embodiments, the antibody is a monoclonal antibody, or the fragment thereof is a fragment of a monoclonal antibody.

[0031] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises (i) a VH framework comprising the framework sequence of human germline VH1- 2*02; and/or (ii) a VL framework comprising the framework sequence of human germline IGKV1-39. [0032] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises a VH that comprises the amino acid sequence of SEQ ID NO: 16 or 18 or 19 or 20.

[0033] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, comprises a VL that comprises the amino acid sequence of SEQ ID NO: 15 or 17.

[0034] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, binds Mtb AM with a binding affinity (KD) of from about lxlO 6 M to about lxlO 9 M.

[0035] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, is a monoclonal antibody.

[0036] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, is a recombinant antibody.

[0037] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, has a human framework region.

[0038] In embodiments, the anti-Mtb AM antibody or Mtb AM-binding fragment thereof, has a human constant region or modified constant region.

[0039] Modified IgG Fc regions are well known in the art. For example, see any of the mutations listed in Table 1 of Wang et al. Protein Cell (2018), 9(l):63-73. In embodiments, the modified Fc region, relative to the unmodified Fc region, has enhanced complement-based effector function, increased or decreased FcyR-based effector function, reduced effector function, enhanced coengagement of antigen and FcyRs, and/or increased serum half life.

[0040] Examples of Fc modifications to modulate antibody effector function for IgGl (see

Wang et al. Protein Cell (2018), 9(l):63-73) included within the scope of the invention are:

Increased FcyRIIIa binding: F243L/R292P/Y300L/V305I/P396L

Increased FcyRIIIa binding: S239D/I332E

Increased FcyRIIIa binding: decreased FcyRIIb binding S239D/I332E/A330L

Increased FcyRIIIa binding S298A/E333A/K334A In one heavy chain: L234Y/L235Q/G236W/ S239M/H268D/D270E/S298A and in the opposing heavy chain: D270E/ K326D/A330M/K334E

Increased FcyRIIa binding, increased FcyRIIIa binding G236A/S239D/I332E Enhance CDC Increased Clq binding K326W/E333S

Increased Clq binding S267E/H268F/S324T

Increased Clq binding IgGl/IgG3 cross subclass

Hexamerization E345R/E430G/S440Y

Reduce effector function - Aglycosylated N297A or N297Q or N297G

Reduced FcyR and Clq binding L235E

Reduced FcyR and Clq binding IgGl: L234A/L235A; IgG4:F234A/L235A

Reduced FcyR and Clq binding IgG2/IgG4 cross isotype

Reduced FcyR and Clq binding IgG2: H268Q/V309L/A330S/P331 S

Reduced FcyR and Clq binding IgG2: V234A/G237A/P238S/H268A/ V309L/ A330S/P331S

Increased FcRn binding at pH 6.0: M252Y/S254T7T256E

Increased FcRn binding at pH 6.0: M428L/N434S (Zalevsky et al., 2010)

Increased FcyRIIb binding: S267E/L328F (Chu et al., 2008)

Increased FcyRIIa binding, decreased FcyRIIIa binding: N325S/L328F.

[0041] In embodiments, an anti-Mtb AM antibody is provided.

[0042] In embodiments, an anti-Mtb AM-binding fragment of the antibody is provided.

[0043] In embodiments, the anti-Mtb AM-binding fragment is an Fab, F(ab)2 or scFv.

[0044] An isolated nucleic acid molecule encoding the antibody, or Mtb AM-binding fragment thereof, described herein is provided. In an embodiment, the nucleic acid is a DNA. In an embodiment, the nucleic acid is a cDNA. In an embodiment, the nucleic acid is an RNA.

[0045] In an embodiment, the disclosure provides a vector encoding the nucleic acid molecule described herein. In an embodiment, a host cell comprising the nucleic acid molecule described herein, or the vector described herein, is provided.

[0046] In an embodiment, the disclosure provides a method of producing an anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, comprising culturing the host cell described herein under conditions wherein the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, is produced by the host cell. [0047] In an embodiment, a composition is provided comprising the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof as described herein, and a carrier. In an embodiment, the composition is a pharmaceutical composition, and the carrier is a pharmaceutical carrier. In an embodiment, a pharmaceutical composition comprising an anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, described herein, and a pharmaceutically acceptable excipient, is provided.

[0048] In an embodiment, the disclosure provides a method of reducing an activity of Mtb

AM in a subject in need thereof, comprising administering to said subject a therapeutically effective amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein.

[0049] In an embodiment, the disclosure provides a method of treating a Mycobacterium tuberculosis infection in a subject, comprising administering to the subject an amount of the anti- Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein, or the pharmaceutical composition described herein, effective to treat a Mycobacterium tuberculosis infection.

[0050] In an embodiment, the disclosure provides a method of reducing the likelihood of an Mycobacterium tuberculosis infection in a subject, comprising administering to the subject who does not have a Mycobacterium tuberculosis infection an amount of the anti-Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein, or the pharmaceutical composition described herein, effective to reduce the likelihood of an Mycobacterium tuberculosis infection.

[0051] In an embodiment, the disclosure provides a method of treating a disease, disorder, or condition mediated by, or related to increased activity of Mycobacterium tuberculosis in a subject, comprising administering to said subject a therapeutically effective amount of the anti- Mtb AM antibody, or Mtb AM-binding fragment thereof, as described herein, or the pharmaceutical composition described herein.

[0052] In an embodiment, an assay device is provided for selectively detecting a virulent

Mycobacterium tuberculosis in a biological sample comprising:

a first portion comprising a first plurality of anti-virulent Mycobacterium tuberculosis antibodies or Mtb AM-binding fragments thereof, as described herein, wherein the antibodies are each attached to their own reporting entity;

a second portion comprising a second plurality of anti -Mycobacterium tuberculosis antibodies. [0053] In embodiments, the reporting entity comprises a nanoparticle. In an embodiment the nanoparticle is a gold nanoparticle.

[0054] In embodiments, the reporting entity comprises an enzyme.

[0055] In embodiments, the second plurality of virulent Mycobacterium tuberculosis antibodies is affixed to a solid support of the device.

[0056] In embodiments, the first plurality of virulent Mycobacterium tuberculosis antibodies is not affixed to a solid support of the device.

[0057] In embodiments, the solid support comprises nitrocellulose.

[0058] In embodiments, the device further comprises a fluid sample pad prior in sequential order to the first and second portions.

[0059] In embodiments, the device further comprises a control portion subsequent in sequential order to the first and second portions.

[0060] In embodiments, the control portion comprises a third plurality of antibodies, immobilized on a solid support of the device, and which third plurality of antibodies are capable of binding the first plurality of virulent Mycobacterium tuberculosis antibodies each attached to their own reporting molecule.

[0061] In embodiments, the device further comprises a fluid-absorbent wicking pad subsequent in sequential order to the first and second portions, and third portion if present.

[0062] In embodiments, the second plurality of anti -Mycobacterium tuberculosis

antibodies comprise antibodies or Mtb AM-binding fragments thereof, as described herein.

[0063] In embodiments, the reporting entity is an enzyme which is horseradish peroxidase

(HRP) or alkaline phosphatase (AP).

[0064] Also provided is a lateral flow assay device for detecting a Mycobacterium tuberculosis in a biological sample comprising:

a first portion comprising a first plurality of anti -Mycobacterium tuberculosis antibodies as disclosed herein comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17, wherein the antibodies are each attached to their own reporting entity; and a second portion comprising a second plurality of anti- Mycobacterium tuberculosis antibodies.

[0065] In an embodiment, the reporting entity comprises an enzyme. [0066] In an embodiment, the reporting entity comprises a gold nanoparticle, horseradish peroxidase (HRP), or alkaline phosphatase (AP). In an embodiment, the reporting entity comprises a gold nanoparticle.

[0067] In an embodiment, the second plurality of Mycobacterium tuberculosis antibodies is affixed to a solid support of the device.

[0068] In an embodiment, the first plurality of Mycobacterium tuberculosis antibodies is not affixed to a solid support of the device.

[0069] In an embodiment, the solid support comprises nitrocellulose.

[0070] In an embodiment, the device further comprises a fluid sample pad prior in sequential order to the first and second portions.

[0071] In an embodiment, the device further comprises a control portion subsequent in sequential order to the first and second portions.

[0072] In an embodiment, the control portion comprises a third plurality of antibodies, immobilized on a solid support of the device, and which third plurality of antibodies are capable of binding the first plurality of virulent Mycobacterium tuberculosis antibodies each attached to their own reporting molecule.

[0073] In an embodiment, the device further comprises a fluid-absorbent wicking pad subsequent in sequential order to the first and second portions, and third portion if present.

[0074] In an embodiment, the second plurality of anti -Mycobacterium tuberculosis antibodies comprise antibodies as described herein comprising a heavy chain variable region of SEQ ID NOS: 13 or 14 and a light chain variable region of SEQ ID NO: 15; or comprising a heavy chain variable region of SEQ ID NO: 33 and a light chain variable region of SEQ ID NO: 17

[0075] In an embodiment, a method is provided for detecting a virulent Mycobacterium tuberculosis in a biological sample comprising contacting the device described herein with the sample and observing if virulent Mycobacterium tuberculosis- bound antibodies bind to the second plurality of virulent Mycobacterium tuberculosis- binding antibodies, wherein if such antibodies bind then virulent Mycobacterium tuberculosis has been detected in the biological sample and wherein if no virulent Mycobacterium tuberculosis- bound antibodies bind to the second plurality of virulent Mycobacterium tuberculosis- binding antibodies then virulent

Mycobacterium tuberculosis has not been detected in the biological sample. [0076] In embodiments, the method further comprises obtaining the sample from a subject.

[0077] In embodiments, the sample is urine or blood.

[0078] In embodiments, the subject is human.

[0079] As used herein, the term "antibody" refers to an intact antibody, i.e., with complete

Fc and Fv regions.“Fragment” refers to any portion of an antibody, or portions of an antibody linked together, such as, in non-limiting examples, a Fab, F(ab)2, a single-chain Fv (scFv), which is less than the whole antibody but which is an antigen-binding portion and which competes with the intact antibody of which it is a fragment for specific binding. In this case, the antigen is the Mtb capsule AM.

[0080] As such a fragment can be prepared, for example, by cleaving an intact antibody or by recombinant means. See generally, Fundamental Immunology, Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989), hereby incorporated by reference in its entirety). Antigen-binding fragments may be produced by recombinant DNA techniques or by enzymatic or chemical cleavage of intact antibodies or by molecular biology techniques. In some embodiments, a fragment is an Fab, Fab', F(ab')2, Fd , Fv, complementarity determining region (CDR) fragment, single-chain antibody (scFv), (a variable domain light chain (VL) and a variable domain heavy chain (VH) linked via a peptide linker. In an embodiment, the scFv comprises a variable domain framework sequence having a sequence identical to a human variable domain FR1, FR2, FR3 or FR4. In an embodiment, the scFv comprises a linker peptide from 5 to 30 amino acid residues long. In an embodiment, the scFv comprises a linker peptide comprising one or more of glycine, serine and threonine residues.

[0081] In an embodiment the linker of the scFv is 10-25 amino acids in length. In an embodiment the peptide linker comprises glycine, serine and/or threonine residues. For example, see Bird et al., Science, 242: 423-426 (1988) and Huston et al., Proc. Natl. Acad. Sci. USA, 85:5879-5883 (1988), each of which are hereby incorporated by reference in their entirety, or a polypeptide that contains at least a portion of an antibody that is sufficient to confer Mtb capsular AM-specific antigen binding on the polypeptide, including a diabody. From N-terminus to C- terminus, both the mature light and heavy chain variable domains comprise the regions FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is in accordance with the definitions of Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987), or Chothia et al., Nature 342:878-883 (1989) , each of which are hereby incorporated by reference in their entirety). As used herein, the term "polypeptide" encompasses native or artificial proteins, protein fragments and polypeptide analogs of a protein sequence. A polypeptide may be monomeric or polymeric. As used herein, an Fd fragment means an antibody fragment that consists of the VH and CH1 domains; an Fv fragment consists of the VI and VH domains of a single arm of an antibody; and a dAb fragment (Ward et al., Nature 341:544-546 (1989) hereby incorporated by reference in its entirety) consists of a VH domain. In some embodiments, fragments are at least 5, 6, 8 or 10 amino acids long. In other embodiments, the fragments are at least 14, at least 20, at least 50, or at least 70, 80, 90, 100, 150 or 200 amino acids long.

[0082] The term "monoclonal antibody" as used herein refers to an antibody member of a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible mutations, e.g., naturally occurring mutations, that may be present in minor amounts. Thus, the modifier "monoclonal" indicates the character of the antibody as not being a mixture of discrete antibodies. In certain embodiments, such a monoclonal antibody typically includes an antibody comprising a polypeptide sequence that binds a target an Mtb capsular AM, wherein the target-binding polypeptide sequence was obtained by a process that includes the selection of a single target binding polypeptide sequence from a plurality of polypeptide sequences. For example, the selection process can be the selection of a unique clone from a plurality of clones, such as a pool of hybridoma clones, phage clones, or recombinant DNA clones. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody of a monoclonal antibody preparation is directed against a single determinant on an antigen. In addition to their specificity, monoclonal antibody preparations are advantageous in that they are typically uncontaminated by other immunoglobulins. Thus an identified monoclonal antibody can be produced by non-hybridoma techniques, e.g., by appropriate recombinant means once the sequence thereof is identified.

[0083] In an embodiment of the inventions described herein, the antibody is isolated. As used herein, the term "isolated antibody" refers to an antibody that by virtue of its origin or source of derivation has one, two, three or four of the following: (1) is not associated with naturally associated components that accompany it in its native state, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, and (4) does not occur in nature.

[0084] As used herein, a "human antibody" unless otherwise indicated is one whose sequences correspond to (i.e., are identical in sequence to) an antibody that could be produced by a human and/or has been made using any of the techniques for making human antibodies as disclosed herein, but not one which has been made in a human. This definition of a human antibody specifically excludes a humanized antibody. A“human antibody” as used herein can be produced using various techniques known in the art, including phage-display libraries (Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581 (1991), hereby incorporated by reference in their entireties, by methods described in Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p. 77 (1985) hereby incorporated by reference in its entirety; Boemer et al., J. Immunol., l47(l):86-95 (1991) hereby incorporated by reference in its entirety, van Dijk and van de Winkel, Curr. Opin. Pharmacol., 5: 368-74 (2001) hereby incorporated by reference in its entirety, and by administering the antigen (e.g., Mtb capsular AM or an entity comprising such) to a transgenic animal that has been modified to produce such antibodies in response to antigenic challenge, but whose endogenous loci have been disabled, e.g., immunized xenomice (see, e.g., U.S. Pat. Nos. 5,939,598; 6,075,181; 6,114,598; 6,150,584 and 6,162,963 to Kucherlapati et al. regarding XENOMOUSETM technology, each of which patents are hereby incorporated by reference in their entireties), e.g., Veloclmmune® (Regeneron, Tarrytown, NY), e.g., UltiMab® platform (Medarex, now Bristol Myers Squibb, Princeton, NJ). See also, for example, Li et al., Proc. Natl. Acad. Sci. USA, 103:3557-3562 (2006) regarding human antibodies generated via a human B-cell hybridoma technology. See also KM Mouse® system, described in PCT Publication WO 02/43478 by Ishida et al., in which the mouse carries a human heavy chain transchromosome and a human light chain transgene, and the TC mouse system, described in Tomizuka et al. (2000) Proc. Natl. Acad. Sci. USA 97:722-727, in which the mouse carries both a human heavy chain transchromosome and a human light chain transchromosome, both of which are hereby incorporated by reference in their entireties. In each of these systems, the transgenes and/or transchromosomes carried by the mice comprise human immunoglobulin variable and constant region sequences.

[0085] The term "human antibody", as used herein, is intended to include antibodies having variable regions in which both the framework and CDR regions are sequences of human origin or identical thereto other than antibodies naturally occurring in a human or made in a human. Furthermore, if the antibody (e.g., an intact antibody rather than, for example, an Fab fragment) contains a constant region, the constant region also is derived from such human sequences, e.g., human germline sequences, or mutated versions of human germline sequences. The human antibodies of the invention may include amino acid residues not encoded by human sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term "human antibody", as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. In one non- limiting embodiment, where the human antibodies are human monoclonal antibodies, such antibodies can be produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and a light chain transgene fused to an immortalized cell.

[0086] In an embodiment, the Mtb capsular AM antibody described herein is a recombinant human antibody. The term "recombinant human antibody", as used herein, includes all human antibodies that are prepared, expressed, created or isolated by recombinant means, such as antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom, antibodies isolated from a host cell transformed to express the human antibody, e.g., from a transfectoma, antibodies isolated from a recombinant, combinatorial human antibody library, and antibodies prepared, expressed, created, or isolated by any other means that involve splicing of all or a portion of a human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable regions in which the framework and CDR regions are derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo.

[0087] “Humanized” forms of non-human (e.g., murine) antibodies are chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin. In one embodiment, a humanized antibody is a human immunoglobulin (recipient antibody) in which residues from a hypervariable region (HVR) of the recipient are replaced by residues from a HVR of a non-human species (donor antibody) such as mouse, rat, rabbit, or nonhuman primate having the desired specificity, affinity, and/or capacity. In some instances, FR residues of the human immunoglobulin variable domain are replaced by corresponding non-human residues. These modifications may be made to further refine antibody performance. Furthermore, in a specific embodiment, humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. In an embodiment, the humanized antibodies do not comprise residues that are not found in the recipient antibody or in the donor antibody. In general, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non- human immunoglobulin, and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody optionally will also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. See, e.g., Jones et al., Nature 321:522-525 (1986); Riechmann et al., Nature 332:323-329 (1988); Presta, Curr. Op. Struct. Biol. 2:593-596 (1992); Vaswani and Hamilton, Ann. Allergy, Asthma & Immunol. 1:105-115 (1998); Harris, Biochem. Soc. Transactions 23:1035-1038 (1995); Hurle and Gross, Curr. Op. Biotech. 5:428-433 (1994); and U.S. Pat. Nos. 6,982,321 and 7,087,409, the contents of each of which references and patents are hereby incorporated by reference in their entirety. In one embodiment where the humanized antibodies do comprise residues that are not found in the recipient antibody or in the donor antibody, the Fc regions of the antibodies are modified as described in WO 99/58572, the content of which is hereby incorporated by reference in their entireties.

[0088] Techniques to humanize a monoclonal antibody are described in U.S. Pat. Nos.

4,816,567; 5,807,715; 5,866,692; 6,331,415; 5,530,101; 5,693,761; 5,693,762; 5,585,089; and 6, 180,370, the content of each of which is hereby incorporated by reference in its entirety.

[0089] A number of "humanized" antibody molecules comprising an antigen-binding site derived from a non-human immunoglobulin have been described, including antibodies having rodent or modified rodent V regions and their associated complementarity determining regions (CDRs) fused to human constant domains. See, for example, Winter et al. Nature 349: 293-299 (1991), Lobuglio et al. Proc. Nat. Acad. Sci. USA 86: 4220-4224 (1989), Shaw et al. J. Immunol. 138: 4534-4538 (1987), and Brown et al. Cancer Res. 47: 3577-3583 (1987), the content of each of which is hereby incorporated by reference in its entirety. Other references describe rodent hypervariable regions or CDRs grafted into a human supporting framework region (FR) prior to fusion with an appropriate human antibody constant domain. See, for example, Riechmann et al. Nature 332: 323-327 (1988), Verhoeyen et al. Science 239: 1534-1536 (1988), and Jones et al. Nature 321: 522-525 (1986), the content of each of which is hereby incorporated by reference in its entirety. Another reference describes rodent CDRs supported by recombinantly veneered rodent framework regions - European Patent Publication No. 0519596 (incorporated by reference in its entirety). These "humanized" molecules are designed to minimize unwanted immunological response toward rodent anti-human antibody molecules which limits the duration and effectiveness of therapeutic applications of those moieties in human recipients. The antibody constant region can be engineered such that it is immunologically inert (e.g., does not trigger complement lysis). See, e.g. PCT Publication No. W099/58572; UK Patent Application No. 9809951.8. Other methods of humanizing antibodies that may also be utilized are disclosed by Daugherty et al., Nucl. Acids Res. 19: 2471-2476 (1991) and in U.S. Pat. Nos. 6,180,377; 6,054,297; 5,997,867; 5,866,692; 6,210,671; and 6,350,861; and in PCT Publication No. WO 01/27160 (each incorporated by reference in its entirety).

[0090] Other forms of humanized antibodies have one or more CDRs (CDR Ll, CDR L2,

CDR L3, CDR Hl, CDR H2, or CDR H3) which are altered with respect to the original antibody, which are also termed one or more CDRs "derived from" one or more CDRs from the original antibody.

[0091] In embodiments, the antibodies or fragments herein can be produced recombinantly, for example antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes.

[0092] In an embodiment, the Mtb capsular AM antibody described herein is capable of specifically binding or specifically binds an Mtb capsular AM. As used herein, the terms "is capable of specifically binding" or "specifically binds" refers to the property of an antibody or fragment of binding to the specified antigen with a dissociation constant that is <1 mM, preferably <1 nM and most preferably <10 pM. In an embodiment, the Kd of the antibody (or fragment) for Mtb capsular AM is better than 1.0 nM. In an embodiment, the Kd of the antibody (or fragment) for Mtb capsular AM is better than 1.5 nM. In an embodiment, the Kd of the antibody (or fragment) for Mtb capsular AM is 1.8 nM or more. In an embodiment, the Kd of the antibody (or fragment) for avirulent Mtb capsular AM is a lower affinity than its Kd for virulent Mtb capsular AM. An epitope that "specifically binds" to an antibody or a polypeptide is a term well understood in the art. A molecular entity is said to exhibit "specific binding" or "preferential binding" if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular cell or substance than it does with alternative cells or substances. An antibody“specifically binds" or "preferentially binds" to a target if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an antibody that specifically or preferentially binds to a Mtb capsular AM conformational epitope is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other Mtb capsular AM epitopes or non-Mtb capsular AM epitopes. It is also understood by reading this definition that, for example, an antibody (or moiety or epitope) that specifically or preferentially binds to a first target may or may not specifically or preferentially bind to a second target. As such, "specific binding" or "preferential binding" does not necessarily require, although it can include, exclusive binding.

[0093] The term "compete", as used herein with regard to an antibody, means that a first antibody, or an antigen-binding portion thereof, binds to an epitope in a manner sufficiently similar to the binding of a second antibody, or an antigen-binding portion thereof, such that the result of binding of the first antibody with its cognate epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody. The alternative, where the binding of the second antibody to its epitope is also detectably decreased in the presence of the first antibody, can, but need not be the case. That is, a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope. However, where each antibody detectably inhibits the binding of the other antibody with its cognate epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to "cross-compete" with each other for binding of their respective epitope(s). Both competing and cross-competing antibodies are encompassed by the present invention. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate, based upon the teachings provided herein, that such competing and/or cross-competing antibodies are encompassed and can be useful for the methods disclosed herein.

[0094] Depending on the amino acid sequences of the constant domains of their heavy chains, antibodies (immunoglobulins) can be assigned to different classes. The antibody or fragment can be, e.g., any of an IgG, IgD, IgE, IgA or IgM antibody or fragment thereof, respectively. In an embodiment the antibody is an immunoglobulin G. In an embodiment the antibody fragment is a fragment of an immunoglobulin G. In an embodiment the antibody is an IgGl, IgG2, IgG2a, IgG2b, IgG3 or IgG4. In an embodiment the antibody comprises sequences from a human IgGl, human IgG2, human IgG2a, human IgG2b, human IgG3 or human IgG4. A combination of any of these antibodies subtypes can also be used. One consideration in selecting the type of antibody to be used is the desired serum half-life of the antibody. For example, an IgG generally has a serum half-life of 23 days, IgA 6 days, IgM 5 days, IgD 3 days, and IgE 2 days. (Abbas AK, Lichtman AH, Pober JS. Cellular and Molecular Immunology, 4th edition, W.B. Saunders Co., Philadelphia, 2000, hereby incorporated by reference in its entirety).

[0095] The "variable region" or "variable domain" of an antibody refers to the amino- terminal domains of the heavy or light chain of the antibody. The variable domain of the heavy chain may be referred to as "VH." The variable domain of the light chain may be referred to as "VL." These domains are generally the most variable parts of an antibody and contain the antigen-binding sites. The term "variable" refers to the fact that certain portions of the variable domains differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. However, the variability is not evenly distributed throughout the variable domains of antibodies. It is concentrated in three segments called hypervariable regions (HVRs) both in the light chain and the heavy chain variable domains. The more highly conserved portions of variable domains are called the framework regions (FR). The variable domains of native heavy and light chains each comprise four FR regions, largely adopting a beta-sheet configuration, connected by three HVRs, which form loops connecting, and in some cases forming part of, the beta-sheet structure. The HVRs in each chain are held together in close proximity by the FR regions and, with the HVRs from the other chain, contribute to the formation of the antigen-binding site of antibodies (see Kabat et al., Sequences of Proteins of Immunological Interest, Fifth Edition, National Institute of Health, Bethesda, Md. (1991)). The constant domains are not involved directly in the binding of an antibody to an antigen, but exhibit various effector functions, such as participation of the antibody in antibody-dependent cellular toxicity.

[0096] The "light chains" of antibodies (immunoglobulins) from any vertebrate species can be assigned to one of two clearly distinct types, called kappa (K) and lambda (l), based on the amino acid sequences of their constant domains.

[0097] "Framework" or "FR" residues are those variable domain residues other than the

HVR residues as herein defined.

[0098] The term "hypervariable region" or "HVR" when used herein refers to the regions of an antibody variable domain which are hypervariable in sequence and/or form structurally defined loops. Generally, antibodies comprise six HVRs; three in the VH (Hl, H2, H3) and three in the VL (Ll, L2, L3). In native antibodies, H3 and L3 display the most diversity of the six HVRs, and H3 in particular is believed to play a unique role in conferring fine specificity to antibodies. See, e.g., Xu et al., Immunity 13:37-45 (2000); Johnson and Wu, in Methods in Molecular Biology 248:1-25 (Lo, ed., Human Press, Totowa, N.J., 2003). Indeed, naturally occurring camelid antibodies consisting of a heavy chain only are functional and stable in the absence of light chain. See, e.g., Hamers-Casterman et al., Nature 363:446-448 (1993); Sheriff et al., Nature Struct. Biol. 3:733-736 (1996). A number of HVR delineations are in use and are encompassed herein. The Kabat Complementarity Determining Regions (CDRs) are based on sequence variability and are the most commonly used (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. (1991) hereby incorporated by reference in its entirety). Chothia refers instead to the location of the structural loops (Chothia and Lesk, J. Mol. Biol. 196:901-917 (1987)). The AbM HVRs represent a compromise between the Kabat HVRs and Chothia structural loops, and are used by Oxford Molecular's AbM antibody modeling software. The "contact" HVRs are based on an analysis of the available complex crystal structures. HVRs may comprise "extended HVRs" as follows: 24-36 or 24-34 (Ll), 46-56 or 50-56 (L2) and 89-97 or 89-96 (L3) in the VL and 26-35 (Hl), 50-65 or 49-65 (H2) and 93-102, 94-102, or 95-102 (H3) in the VH. The variable domain residues are numbered according to Kabat et al., supra, for each of these definitions.

[0099] The term "Fc region" herein is used to define a C-terminal region of an immunoglobulin heavy chain, including native sequence Fc regions and variant Fc regions. Although the boundaries of the Fc region of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc region is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The C-terminal lysine of the Fc region may be removed, for example, during production or purification of the antibody, or by recombinantly engineering the nucleic acid encoding a heavy chain of the antibody. Accordingly, an intact antibody as used herein may be an antibody with or without the otherwise C-terminal lysine.

[00100] Compositions or pharmaceutical compositions comprising the antibodies, ScFvs or fragments of antibodies disclosed herein preferably comprise stabilizers to prevent loss of activity or structural integrity of the protein due to the effects of denaturation, oxidation, or aggregation over a period of time during storage and transportation prior to use. The compositions or pharmaceutical compositions can comprise one or more of any combination of salts, surfactants, pH and tonicity agents such as sugars that contribute to overcoming aggregation problems. Where a composition or pharmaceutical composition of the present invention is used as an injection, it is desirable to have a pH value in an approximately neutral pH range. It is also advantageous to minimize surfactant levels to avoid bubbles in the formulation which are detrimental for injection into subjects. In an embodiment, the composition or pharmaceutical composition is in liquid form and stably supports high concentrations of bioactive antibody in solution and is suitable for inhalatuional or parenteral administration. In an embodiment, the composition or pharmaceutical composition is suitable for intravenous, intramuscular, intraperitoneal, intradermal and/or subcutaneous injection. In an embodiment, the composition or pharmaceutical composition is in liquid form and has minimized risk of bubble formation and anaphylactoid side effects. In an embodiment, the composition or pharmaceutical composition is isotonic. In an embodiment, the composition or pharmaceutical composition has a pH or 6.8 to 7.4.

[00101] In an embodiment the ScFvs or fragments of antibodies disclosed herein are lyophilized and/or freeze dried and are reconstituted for use.

[00102] Examples of pharmaceutically acceptable carriers include, but are not limited to, phosphate buffered saline solution, sterile water (including water for injection USP), emulsions such as oil/water emulsion, and various types of wetting agents. Preferred diluents for aerosol or parenteral administration are phosphate buffered saline or normal (0.9%) saline, for example 0.9% sodium chloride solution, USP. Compositions comprising such carriers are formulated by well known conventional methods (see, for example, Remington's Pharmaceutical Sciences, 18th edition, A. Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990; and Remington, The Science and Practice of Pharmacy 20th Ed. Mack Publishing, 2000, the content of each of which is hereby incorporated in its entirety). In non-limiting examples, the carrier can comprise one or more of dibasic sodium phosphate, potassium chloride, monobasic potassium phosphate, polysorbate 80 (e.g. 2-[2-[3,5-bis(2-hydroxyethoxy)oxolan-2-yl]-2-(2-hydroxyethox y)ethoxy]ethyl (E)-octadec- 9-enoate), disodium edetate dehydrate, sucrose, monobasic sodium phosphate monohydrate, and dibasic sodium phosphate dihydrate.

[00103] The antibodies, or fragments of antibodies, or compositions, or pharmaceutical compositions described herein can also be lyophilized or provided in any suitable forms including, but not limited to, injectable solutions or inhalable solutions, gel forms, and tablet forms.

[00104] The term "Kd", as used herein, is intended to refer to the dissociation constant of an antibody-antigen interaction. One way of determining the Kd or binding affinity of antibodies to Mtb capsular AM is by measuring binding affinity Using a Dip and Read assay using an immobilized antigen and monoclonal antibodies (Octet Red96 ForteBio, Fremont, CA). (The affinity constant is the inverted dissociation constant). Biotinylated Mtb capsular AM can be diluted into PBS + 0.1% BSA, 0.02% Tween20 and 0.05% sodium azide (Kinetics Buffer, ForteBio) and dipped in to wells containing serial diluted mAbs starting from 37.75 nM. The concentrations of the Fab proteins are determined by ELISA and/or SDS-PAGE electrophoresis using a IgGl standard monoclonal antibody of known concentration as a standard. Kinetic association rates (kon) and dissociation rates (koff) are obtained simultaneously by fitting the data to a 1:1 Langmuir binding model (Karlsson, R. Roos, H. Fagerstam, L. Petersson, B. (1994). Methods Enzymology 6. 99-110, the content of which is hereby incorporated in its entirety) using the BIA evaluation program. Equilibrium dissociation constant (Kd) values are calculated as koff/kon. This protocol is suitable for use in determining binding affinity of an antibody or fragment to any Mtb capsular AM. Other protocols known in the art may also be used. For example, ELISA of Mtb capsular AM with mAb can be used to determine the kD values.

[00105] The term "Fc domain" herein is used to define a C-terminal region of an immunoglobulin heavy chain, including native sequence Fc regions and variant Fc regions. Although the boundaries of the Fc domain of an immunoglobulin heavy chain might vary, the human IgG heavy chain Fc domain is usually defined to stretch from an amino acid residue at position Cys226, or from Pro230, to the carboxyl-terminus thereof. The C-terminal lysine of the Fc domain may be removed, for example, by recombinantly engineering the nucleic acid encoding it.

[00106] In embodiments, the antibody comprises an Fc domain. In an embodiment, the Fc domain has the same sequence or 99% or greater sequence similarity with a human IgGl Fc domain. In an embodiment, the Fc domain has the same sequence or 99% or greater sequence similarity with a human IgG2 Fc domain. In an embodiment, the Fc domain has the same sequence or 99% or greater sequence similarity with a human IgG3 Fc domain. In an embodiment, the Fc domain has the same sequence or 99% or greater sequence similarity with a human IgG4 Fc domain. In an embodiment, the Fc domain is not mutated. In an embodiment, the Fc domain is mutated at the CH2-CH3 domain interface to increase the affinity of IgG for FcRn at acidic but not neutral pH. In an embodiment, the Fc domain has the same sequence as a human IgGl Fc domain.

[00107] The invention encompasses modifications to the variable regions disclosed herein. For example, the invention includes antibodies comprising functionally equivalent variable regions and CDRs which do not significantly affect their properties as well as variants which have enhanced or decreased activity and/or affinity. For example, the amino acid sequence may be mutated to obtain an antibody with the desired binding affinity to Mtb AM. Modification of polypeptides is routine practice in the art and need not be described in detail herein. Examples of modified polypeptides include polypeptides with conservative substitutions of amino acid residues, one or more deletions or additions of amino acids which do not significantly deleteriously change the functional activity, or which mature (enhance) the affinity of the polypeptide for its ligand, or use of chemical analogs.

[00108] Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to an epitope tag. Other insertional variants of the antibody molecule include the fusion to the N- or C- terminus of the antibody of an enzyme or a polypeptide which increases the half-life of the antibody in the blood circulation.

[00109] Substitution variants have at least one amino acid residue in the antibody molecule removed and a different residue inserted in its place. The sites of greatest interest for substitutional mutagenesis include the hypervariable regions, but framework alterations are also contemplated. Conservative substitutions are shown in Table 1 under the heading of "conservative substitutions." If such substitutions result in a change in biological activity, then more substantial changes, denominated "exemplary substitutions" in Table 1, or as further described below in reference to amino acid classes, may be introduced and the products screened.

[00110]

Table 1 : Amino Acid Substitutions

[00111] Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a b-sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Naturally occurring residues are divided into groups based on common side-chain properties:

(1) Non-polar: Norleucine, Met, Ala, Val, Leu, Ile;

(2) Polar without charge: Cys, Ser, Thr, Asn, Gln;

(3) Acidic (negatively charged): Asp, Glu;

(4) Basic (positively charged): Lys, Arg;

(5) Residues that influence chain orientation: Gly, Pro; and

(6) Aromatic: Trp, Tyr, Phe, His.

[00112] Non-conservative substitutions are made by exchanging a member of one of these classes for another class.

[00113] One type of substitution, for example, that may be made is to change one or more cysteines in the antibody, which may be chemically reactive, to another residue, such as, without limitation, alanine or serine. For example, there can be a substitution of a non-canonical cysteine. The substitution can be made in a CDR or framework region of a variable domain or in the constant region of an antibody. In some embodiments, the cysteine is canonical. Any cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant cross-linking. Conversely, cysteine bond(s) may be added to the antibody to improve its stability, particularly where the antibody is an antibody fragment such as an Fv fragment.

[00114] The antibodies may also be modified, e.g., in the variable domains of the heavy and/or light chains, e.g., to alter a binding property of the antibody. Changes in the variable region can alter binding affinity and/or specificity. In some embodiments, no more than one to five conservative amino acid substitutions are made within a CDR domain. In other embodiments, no more than one to three conservative amino acid substitutions are made within a CDR domain. For example, a mutation may be made in one or more of the CDR regions to increase or decrease the KD of the antibody for Mtb AM, to increase or decrease koff, or to alter the binding specificity of the antibody. Techniques in site-directed mutagenesis are well-known in the art..

[00115] A modification or mutation may also be made in a framework region or constant region to increase the half-life of an anti-Mtb AM antibody. See, e.g., PCT Publication No. WO 00/09560. A mutation in a framework region or constant region can also be made to alter the immunogenicity of the antibody, to provide a site for covalent or non-covalent binding to another molecule, or to alter such properties as complement fixation, FcR binding and antibody- dependent cell-mediated cytotoxicity. In embodiments, a single antibody may have mutations in any one or more of the CDRs or framework regions of the variable domain or in the constant region.

[00116] In an embodiment, an antibody described herein is recombinantly produced. In an embodiment, the fusion protein is produced in a eukaryotic expression system.

[00117] In an embodiment, the fusion protein produced in the eukaryotic expression system comprises glycosylation at a residue on the Fc portion corresponding to Asn297.

[00118] In an embodiment, the disclosure provides a composition comprising an antibody, or antigen-binding fragment thereof, as described herein. In an embodiment, the composition is a pharmaceutical composition. In an embodiment the composition or pharmaceutical composition comprising the antibody, or antigen-binding fragment thereof, described herein is substantially pure with regard to the antibody, or antigen-binding fragment thereof. A composition or pharmaceutical composition comprising the antibody, or antigen-binding fragment thereof, described herein is "substantially pure" with regard to the antibody or fragment when at least 60% to 75% of a sample of the composition or pharmaceutical composition exhibits a single species of the antibody, or antigen-binding fragment thereof. A substantially pure composition or pharmaceutical composition comprising the antibody, or antigen-binding fragment thereof, described herein can comprise, in the portion thereof which is the antibody, or antigen-binding fragment, 60%, 70%, 80% or 90% of the antibody, or antigen-binding fragment, of the single species, more usually about 95%, and preferably over 99%. Purity or homogeneity may be tested by a number of means well known in the art, such as polyacrylamide gel electrophoresis or HPLC.

[00119] In embodiments, the antibody, or antigen-binding fragment thereof, binds to a linear epitope. In embodiments, the antibody, or antigen-binding fragment thereof, binds to a linear oligosaccharide epitope.

[00120] In an embodiment, “determining” as used herein means experimentally determining.

[00121] “And/or” as used herein, for example, with option A and/or option B, encompasses the separate embodiments of (i) option A, (ii) option B, and (iii) option A plus option B.

[00122] All combinations of the various elements described herein are within the scope of the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

[00123] All references cited herein are incorporated herein in their entireties.

[00124] The following non-limiting example serves to further illustrate embodiments of the invention.

EXAMPLE

[00125] Individuals at different stages of Mtb infection and exposure were recruited and their sera screened for AM reactivity by ELISA. The individuals were living in the US (mostly immigrants from TB endemic regions) and categorized by the results of their Tuberculin skin- tests (TST) and Interferon Gamma Release Assays (IGRA), or TB diagnostic test results. PPD+ individuals were TST-positive (TST+), had a history of BCG vaccination and were IGRA- negative (IGRA-); latent (controlled) Mtb infection (LTBI) were TST+ and IGRA+; and subjects diagnosed with active TB had symptomatic microbio logically confirmed disease. PPD- volunteers were TST-negative and their sera were used as a baseline control.

[00126] It was observed that AM serum titers correlated with protective functions against Mtb such as enhanced Mtb opsonophagocytosis and intracellular growth restriction in vitro (Figs. 1A-E and Figs. 2A-D). Human serum IgG titers to AM and the lipidated cell wall antigen lipoarabinomanan (LAM) were significantly correlated with protective effects such as enhanced Mtb opsonophagocytosis and intracellular growth restriction by human macrophages in vitro. These protective effects were associated with antibody reactivity to certain AM oligosaccharides using a glycan microarray (Figs. 1A-E and Fig. 2A-D). This indicates that the Mtb capsule exposes different polysaccharide epitopes and some could be more pertinent than others. Passive transfer of polyclonal AM IgG from a few subjects showed significant reduction in the bacterial burden in the lungs of mice infected with a low dose of Mtb resembling human infection (25 CFU; Fig. 2D).

[00127] Subjects with high AM IgG titer in their sera were identified. High titer subjects (ELISA OD405 > 1.0) were depleted of AM-specific IgG by incubating the serum with magnetic beads conjugated to biotinylated AM. Recombinant human mAbs were generated from isolated single memory B cells. Single memory B cell sorting was performed for AM (isolated from the virulent Mtb strain H37Rv) with PBMCs from a high anti-AM IgG titer Tuberculin skin test (TST)+ healthy individual with polyclonal anti- AM Ab functions against Mtb. Immunoglobulin genes were cloned, sequenced and re-expressed in 293T HEK cells.

[00128] The antibody given lab designation AM009 was generated from the memory B cells obtained from a PPD+ individual with high AM serum titers, and whose serum demonstrated protective functions against Mtb. This subject’s sera restricted Mtb growth intracellularly in vitro by a THP-l (monocyte cell line) based growth inhibition assay and reduced the bacterial burden in the lungs of mice infected with Mtb in vivo (Figs. 2A-D). The binding of AM009 to AM was characterized and the mAb’s functions in vitro investigated (Figs. 3A-E). It was determined that some of the regions of the heavy or light chain are essential for OS-binding and activity. Mutants with substitutions in the complementarity determining regions (CDRs) and framework regions (FRs) were designed and expressed (Figs. 4A-C).

[00129] AM009 was not remarkably somatically mutated, and shared sequence similarity in both heavy (91%) and light chains (89%) at the nucleotide level. The AM009 antibody sequence also contained features that were consistent with polysaccharide specific antibodies such as multiple repeating hydrophobic residues and a moderately long CDRH3 (14 amino acids long). The heavy chain is derived from the VH 1-2*02 germline gene (QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEWMGWINPNSG GTNYAQKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYCAR (SEQ ID NO:43)). The light chain was IGKV1-39 derived (DIQMTQSPSSLSASVGDRVTITCRASQSISSYLN WYQQKPGKAPKLLIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYST E IK (SEQ ID NO:44). Notably, there is an overrepresentation of polysaccharide specific antibodies from the VH3 family reported.

[00130] BioLayer Interferometry (BLI) was used to measure the affinity of the mAb to AM. The binding specificity of mAbs to different Mtb strains (whole bacteria as well as capsular AM and cell wall LAM) was determined by immunofluorescence and ELISA. [00131] To further characterize the polysaccharide epitopes, mAbs were tested for their binding reactivity to 30 synthesized AM oligosaccharide fragments. Thirty oligosaccharide fragments representing the components of AM were designed and synthesized by Dr. Todd Lowary’s group (University of Alberta, Canada). In binding characterization, AM009 binds with very high affinity to AM by BLI (KD = 5.12 x 10 8 M) and binds to the whole bacteria with an intact capsule. This is an exceptionally high affinity for a polysaccharide mAb . In addition, AM009 binds a novel polysaccharide epitope which could be specific for capsular AM (Figs. 5A and B). AM009 has increased specificity for the virulent Mtb H37Rv AM (but less for the avirulent H37Ra AM) isolated from the capsule with some degree of crossreactivity with H37Rv LAM (Figs. 3A-E).

[00132] Murine mAb CS-35 to LAM was used as a positive control for high affinity binding to LAM and AM isolated from various mycobacteria strains. The epitope for CS-35 is very well defined and the mAb binds a conserved terminal arabinofuranose epitope on LAM and AM from several strains of mycobacteria. Preliminary data suggest that both AM009 and CS-35 bind the fixed Mtb with the capsule, but recognize different epitopes. In cross-competition ELISA, CS-35 and AM009 do not compete - when CS-35 was used as a competitor mAb, AM009 binding to AM remained unchanged. Moreover, AM009 had very distinct reactivity to certain AM OS. AM009 has high affinity for compounds 18-19 and 21-22 . Murine mAb to LAM, CS-35 was the positive control. Most other murine mAbs that bind AM and LAM compete with CS-35, but AM009 exhibits preferential binding to epitopes that are different than CS-35, suggesting that the epitope specificities of antibodies in humans and mice recognized different Mtb capsular AM epitopes.

[00133] Human mAbs targeting polysaccharides from the Mtb capsule can restrict Mtb growth using in vitro cell-based assays and in vivo using humanized FcyR mice. In preliminary studies, AM009 and one of the mutants enhanced Mtb opsonophagocytosis in human macrophages in vitro. These data indicate that AM009 has functions consistent with the functions observed in the serum (Figs. 2A-D). Collectively, the data indicate that AM0009 is unique and has exceptionally high binding specificity for virulent Mtb.

[00134] AM009-1 Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGYTFS TYWIHWMRQA PGQGPEWMGW IIPKSGGTNY AQKFQGRVAM TRDTSLNTVY MELSRLTSDD TAVYYCARGI LLNGIGAFDY WGQGTLVTVS S (SEQ ID NO: 13)

[00135] AM009-1 Heavy Chain Variable Region (nucleotide sequence)

CAGGTGCAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGGTACACCTTCTCCACCTACTGGATCCACTGGATGCG G

CAGGCCCCTGGACAAGGGCCTGAGTGGATGGGGTGGATCATCCCTAAGAGTGGCGG

CACAAACTATGCACAGAAGTTTCAGGGCAGGGTCGCCATGACCAGGGACACGTCCCT

CAATACAGTCTACATGGAGTTGAGCAGGCTGACATCGGACGACACGGCCGTTTATTA

TTGTGCGAGAGGTATTCTGTTGAACGGAATTGGGGCCTTTGACTACTGGGGCCAGGG

AACCCTGGTCACCGTCTCCTCA

(SEQ ID NO:34)

[00136] AM009-2 Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGYTFA TYWIHWMRQA PGQGPEWMGW IIPKSGGTNY AQKFQGRVAM TRDTSLNTVY MELSRLTSDD TAVYYCARGI LLNGIGAFDY WGQGTLVTVS S

(SEQ ID NO: 14)

[00137] AM009-2 Heavy Chain Variable Region (nucleotide sequence)

CAGGTGCAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGGTACACCTTCGCCACCTACTGGATCCACTGGATGCG G

CAGGCCCCTGGACAAGGGCCTGAGTGGATGGGGTGGATCATCCCTAAGAGTGGCGG

CACAAACTATGCACAGAAGTTTCAGGGCAGGGTCGCCATGACCAGGGACACGTCCCT

CAATACAGTCTACATGGAGTTGAGCAGGCTGACATCGGACGACACGGCCGTTTATTA

TTGTGCGAGAGGTATTCTGTTGAACGGAATTGGGGCCTTTGACTACTGGGGCCAGGG

AACCCTGGTCACCGTCTCCTCA

(SEQ ID NO:35)

[00138] AM009 Kappa Chain Variable Region (amino acid sequence)

DIVMTQSPSS LSASVGDRVT ITCRTSQTVS SNLNWYQQRP GKAPKLLISG ISDLHSGVPS RFSGSGSGTD FTLTISSLQP EDSATYYCQQ SYSLPRTFGQ GTKVEIK SEQ ID NO: 15) [00139] AM009 Kappa Chain Variable Region (nucleotide sequence)

GATATTGTGATGACCCAGTCTCCATCCTCCCTGTCCGCATCTGTTGGAGACAGAGTCA

CCATCACTTGCCGGACGAGTCAGACCGTTTCCAGTAATTTAAATTGGTATCAGCAGA

GACCAGGGAAAGCCCCTAAACTCCTGATCTCTGGTATATCCGATCTGCATAGTGGGG

TCCCATCCAGGTTCAGTGGCAGTGGGTCTGGGACAGATTTCACTCTCACCATCAGCA

GTCTGCAGCCTGAAGATTCTGCAACTTACTACTGTCAACAGAGTTACAGTCTCCCTC G

GACGTTCGGCCAAGGGACCAAGGTGGAAATCAAA

(SEQ ID NO:36)

[00140] AM009 HEAVY CHAIN - KABAT NUMBERING

[00141] CDRH1

[00142] TYWIH

[00143] (SEQ ID NO: 1)

[00144] CDRH2

[00145] WIIPKSGGTNYAQKFQG

[00146] (SEQ ID NO:2)

[00147] CDRH3

[00148] GILLNGIGAFDY

[00149] (SEQ ID NO:3)

[00150] AM009 KAPPA CHAIN - KABAT NUMBERING

[00151] CDRL1

[00152] RTSQTVSSNLN

[00153] (SEQ ID NO:4)

[00154] CDRL2

[00155] GISDLHS

[00156] (SEQ ID NO:5)

[00157] CDRL3

[00158] QQSYSLPRT [00159] (SEQ ID NO:6)

[00160] AM009-1 HEAVY CHAIN - IMGT NUMBERING

[00161] CDRH1

[00162] GYTFSTYW

[00163] (SEQ ID NO:2l, where sequence is GYTFXTYW

[00164] and X = S)

[00165] CDRH2

[00166] IIPKSGGT

[00167] (SEQ ID NO:22)

[00168] CDRH3

[00169] ARGILLNGIGAFDY

[00170] (SEQ ID NO:23)

[00171] AM009-2 HEAVY CHAIN - IMGT NUMBERING

[00172] CDRH1

[00173] GYTFATYW

[00174] (SEQ ID NO:2l, where sequence is GYTFXTYW

[00175] and X = A)

[00176] CDRH2

[00177] IIPKSGGT

[00178] (SEQ ID NO:22)

[00179] CDRH3

[00180] ARGILLNGIGAFDY

[00181] (SEQ ID NO:23)

[00182] AM009 KAPPA CHAIN - IMGT NUMBERING

[00183] CDRL1 [00184] QTVSSN

[00185] (SEQ ID NO:24)

[00186] CDRL2

[00187] GIS

[00188] CDRL3

[00189] QQSYSLPRT

[00190] (SEQ ID NO:25)

[00191] Additional Mutants

[00192] mutated CDRH3

[00193] SRGILLNGIGAFDY

[00194] (SEQ ID NO:32)

[00195] mutated CDRH3

[00196] ARGILLNGIAAFDY

[00197] (SEQ ID NO:3l)

[00198] AMO 16 - Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGFTFT DYYIHWVRQA PGQGLEWIGW INPHSGDTNS AQKFQGRVTM TRDTSISTAY MELSRLRSYD TAVYYCSRDH

YYDTSAYNPS DFWGQGTLVT VSS

(SEQ ID NO:33)

[00199] AMO 16 - Heavy Chain Variable Region (nucleotide sequence)

CAGGTACAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGATTCACCTTCACCGACTACTATATACACTGGGTGCG A

CAGGCCCCTGGACAAGGGCTTGAGTGGATTGGATGGATCAACCCTCACAGTGGTGAC

ACAAACTCTGCACAGAAGTTTCAGGGCAGGGTCACCATGACCAGGGACACGTCCATC

AGCACAGCCTACATGGAGCTGAGCAGGCTGAGATCTTACGACACGGCCGTCTATTAC

TGTTCGAGAGATCACTACTATGATACTAGTGCTTATAACCCCAGTGACTTCTGGGGC C

AGGGAACCCTGGTCACCGTCTCCTCA (SEQ ID NO:37)

[00200] AMO 16 Kappa Chain (amino acid sequence)

EIVLTQSPAT LSLSPGERAT LSCRTSQSVS SNLAWYQQKA GQTPRLIIYD ASNRATGTPA RFSGSGSGTD FTLTISSLEP EDFAVYYCQQ RTHWPPFTFG GGTKVEIK

(SEQ ID NO: 17)

[00201] AMO 16 Kappa Chain (nucleotide sequence)

GAAATTGTGTTGACGCAGTCTCCAGCCACCCTGTCTTTGTCTCCAGGGGAAAGAGCC

ACCCTCTCCTGTAGGACCAGTCAGAGTGTTAGCAGCAACTTAGCCTGGTACCAGCAG

AAAGCTGGCCAGACTCCCAGGCTCATCATCTATGATGCATCCAACAGGGCCACTGGC

ACCCCAGCCAGGTTCAGTGGCAGTGGGTCTGGGACAGACTTCACTCTCACCATCAGC

AGCCTAGAGCCTGAAGATTTTGCGGTTTATTACTGTCAGCAGCGTACCCACTGGCCT C

CGTTCACTTTCGGCGGAGGGACCAAGGTGGAAATCAAA

(SEQ ID NO:38)

[00202] AM016 HEAVY CHAIN - RABAT NUMBERING

[00203] CDRH1

[00204] DYYIH

[00205] (SEQ ID NO:7)

[00206] CDRH2

[00207] WINPHSGDTNSAQKFQG

[00208] (SEQ ID NO: 8)

[00209] CDRH3

[00210] DHYYDTSAYNPSDF

[00211] (SEQ ID NO:9)

[00212] AM016 KAPPA CHAIN - RABAT NUMBERING

[00213] CDRL1

[00214] RTSQSVSSNLA

[00215] (SEQ ID NO: 10) [00216] CDRL2

[00217] DASNRAT

[00218] (SEQ ID NO:l l)

[00219] CDRL3

[00220] QQRTHWPPFT

[00221] (SEQ ID NO: 12)

[00222] AM016 HEAVY CHAIN - IMGT NUMBERING

[00223] CDRH1

[00224] GFTFTDYY

[00225] (SEQ ID NO:26)

[00226] CDRH2

[00227] INPHSGDT

[00228] (SEQ ID NO:27)

[00229] CDRH3

[00230] SRDHYYDTSAYNPSDF

[00231] (SEQ ID NO:28)

[00232] AM016 KAPPA CHAIN - IMGT NUMBERING

[00233] CDRL1

[00234] QSVSSN

[00235] (SEQ ID NO:29)

[00236] CDRL2

[00237] DAS

[00238] CDRL3

[00239] QQRTHWPPFT

[00240] (SEQ ID NO:30) [00241] AMO 16 mutant Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGFTFT DYYIHWVRQA PGQGLEWIGW INPHSGDTNS AQKFQGRVTM TRDTSISTAY MELSRLRSYD TAVYYCARGI

LLNGIGAFDY WGQGTLVTVS S

(SEQ ID NO: 16)

[00242] AMO 16 mutant Heavy Chain Variable Region (nucleotide sequence)

CAGGTACAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGATTCACCTTCACCGACTACTATATACACTGGGTGCG A

CAGGCCCCTGGACAAGGGCTTGAGTGGATTGGATGGATCAACCCTCACAGTGGTGAC

ACAAACTCTGCACAGAAGTTTCAGGGCAGGGTCACCATGACCAGGGACACGTCCATC

AGCACAGCCTACATGGAGCTGAGCAGGCTGAGATCTTACGACACGGCCGTCTATTAC

TGTGCGAGAGGTATTCTGTTGAACGGAATTGGGGCCTTTGACTACTGGGGCCAGGGA

ACCCTGGTCACCGTCTCCTCAGC

(SEQ ID NO:39)

[00243] AMO 16 mutant A93S Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGFTFT DYYIHWVRQA PGQGLEWIGW INPHSGDTNS AQKFQGRVTM TRDTSISTAY MELSRLRSYD TAVYYCSRGI LLNGIGAFDY WGQGTLVTVS S

(SEQ ID NO: 18)

[00244] AMO 16 mutant A93S Heavy Chain Variable Region (nucleotide sequence)

CAGGTACAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGATTCACCTTCACCGACTACTATATACACTGGGTGCG A

CAGGCCCCTGGACAAGGGCTTGAGTGGATTGGATGGATCAACCCTCACAGTGGTGAC

ACAAACTCTGCACAGAAGTTTCAGGGCAGGGTCACCATGACCAGGGACACGTCCATC

AGCACAGCCTACATGGAGCTGAGCAGGCTGAGATCTTACGACACGGCCGTCTATTAC

TGTTCGAGAGGTATTCTGTTGAACGGAATTGGGGCCTTTGACTACTGGGGCCAGGGA

ACCCTGGTCACCGTCTCCTCAGC

(SEQ ID NO:40) [00245] AM009_l_Gl00bA Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGYTFS TYWIHWMRQA PGQGPEWMGW IIPKSGGTNY AQKFQGRVAM TRDTSLNTVY MELSRLTSDD TAVYYCARGI LLNGIAAFDY WGQGTLVTVS S

(SEQ ID NO: 19)

[00246] AM009_l_Gl00bA Heavy Chain Variable Region (nucleotide sequence)

CAGGTGCAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGGTACACCTTCTCCACCTACTGGATCCACTGGATGCG G

CAGGCCCCTGGACAAGGGCCTGAGTGGATGGGGTGGATCATCCCTAAGAGTGGCGG

CACAAACTATGCACAGAAGTTTCAGGGCAGGGTCGCCATGACCAGGGACACGTCCCT

CAATACAGTCTACATGGAGTTGAGCAGGCTGACATCGGACGACACGGCCGTTTATTA

TTGTGCGAGAGGTATTCTGTTGAACGGAATTGCGGCCTTTGACTACTGGGGCCAGGG

AACCCTGGTCACCGTCTCCTCA

(SEQ ID NO:4l)

[00247] AM009_2_Gl00bA Heavy Chain Variable Region (amino acid sequence)

QVQLVESGAE VKKPGASVKV SCKASGYTFA TYWIHWMRQA PGQGPEWMGW IIPKSGGTNY AQKFQGRVAM TRDTSLNTVY MELSRLTSDD TAVYYCARGI LLNGIAAFDY WGQGTLVTVS S

(SEQ ID NO:20)

[00248] AM009_2_Gl00bA Heavy Chain Variable Region (nucleotide sequence)

CAGGTGCAGCTGGTGGAGTCTGGGGCTGAGGTGAAGAAGCCTGGGGCCTCAGTGAA

GGTCTCCTGCAAGGCTTCTGGGTACACCTTCGCCACCTACTGGATCCACTGGATGCG G

CAGGCCCCTGGACAAGGGCCTGAGTGGATGGGGTGGATCATCCCTAAGAGTGGCGG

CACAAACTATGCACAGAAGTTTCAGGGCAGGGTCGCCATGACCAGGGACACGTCCCT

CAATACAGTCTACATGGAGTTGAGCAGGCTGACATCGGACGACACGGCCGTTTATTA

TTGTGCGAGAGGTATTCTGTTGAACGGAATTGCGGCCTTTGACTACTGGGGCCAGGG

AACCCTGGTCACCGTCTCCTCA

(SEQ ID NO:42)