Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
TREATMENTS FOR SYSTEMIC SCLEROSIS
Document Type and Number:
WIPO Patent Application WO/2021/092079
Kind Code:
A1
Abstract:
In certain aspects, the present disclosure relates to methods of treating systemic sclerosis (SSc) using ΤβRΙΙ antagonists comprising a heterologous domain and a truncated, ligand -binding portion of the extracellular domain of ΤβRΙΙ polypeptide useful to selectively antagonize a ΤβRΙΙ ligand. The disclosure further provides methods for treating one or more complications of SSc, including interstitial lung disease (ILD), with ΤβRII antagonists of the present disclosure.

Inventors:
LI GANG (US)
DE OLIVEIRA PENA JANETHE (US)
KUMAR RAVINDRA (US)
REYNOLDS JOSEPH (US)
Application Number:
PCT/US2020/058967
Publication Date:
May 14, 2021
Filing Date:
November 04, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ACCELERON PHARMA INC (US)
International Classes:
A61K38/17; A61P37/00; C07K14/475; C07K19/00
Domestic Patent References:
WO2015027082A12015-02-26
WO2018204594A12018-11-08
WO1998048024A11998-10-29
WO1999065948A11999-12-23
Other References:
KATSUMOTO, T. R.: "Blocking TGFP via Inhibition of the alphavbeta6 Integrin: A Possible Therapy for Systemic Sclerosis Interstitial Lung Disease", INTERNATIONAL JOURNAL OF RHEUMATOLOGY, vol. 2011, pages 1 - 7, XP055350199, DOI: 10.1155/2011/208219
See also references of EP 4054617A4
Attorney, Agent or Firm:
VARMA, Anita et al. (US)
Download PDF:
Claims:
We Claim:

1. A method of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TpRII) antagonist to a subject in need thereof.

2. A method of treating systemic sclerosis, comprising administering a TbKII polypeptide to a subject in need thereof, 3. The method of claim 2, wherein the systemic sclerosis is SSc-ILD.

4. The method of any one of claims 1-3, wherein the systemic sclerosis is diffuse systemic sclerosis (dcSSc). 5. The method of any one of claims 1-4, wherein the subject has a forced vital capacity' (FVC) of greater than or equal to 50% of predicted.

6. The method of any one of claims 1 -4, wherein the subject has an FVC of between about 90% and about 80% of predicted.

7. The method of any one of claims 1-4, wherein the subject has an FVC of between about 80% and about 70% of predicted.

8. The method of any one of claims 1-4, wherein the subject has an FVC of between about 70% and about 60% of predicted,

9. The method of any one of claims 1-4, wherein the subject has an FVC of between about 60% and about 50% of predicted. 10. Hie method of any one of claims 1-4, wherein the subject has an FVC of between about 50% and about 40% of predicted.

11. The method of any one of claims 1 -4, wherein the subject has an FVC of between about 40% and about 30% of predicted.

12. The method of any one of claims 1-4, wherein the subject has an FVC of between about 30% and about 20% of predicted.

13. The method of any one of claims 6-12, wherein the FVC is measured by spirometry.

14. The method of any one of claims 1-13, wherein the administration reduces the annual rate of decline in FVC by between about 1% and about 10%. 15. The method of any one of claims 1-13, wherein the administration reduces the annual rate of decline in FVC by between about 10% and about 20%.

16. The method of any one of claims 1-13, wherein the administration reduces the annual rate of decline in FVC by between about 20% and about 30%.

17. The method of any one of claims 1-13, wherein the administration reduces the annual rate of decline in FVC by between about 30% and about 40%.

18. The method of any one of claims 1-13, wherein the admini stration reduces the annual rate of decline in FVC by between about 40% and about 50%.

19. The method of any one of claims 1-13, wherein the administration reduces the annual rate of decline in FVC by between about 50% and about 60%. 20. The method of any one of claims 1-13, wherein the administration reduces the annual rate of decline in FVC by between about 60% and about 70%.

21. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 1 mL and about 10 mL.

22. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 10 mL and about 20 ml,.

23. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 20 mL and about 30 mb.

24. The method of any one of claims 1 -20, wherein the administration reduces the annual rate of decline in FV C by between about 30 mL and about 40 mL.

25. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 40 mL and about 50 mL. 26. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 50 mL and about 60 mL.

27. The method of any one of claims 1-20, wherem the administration reduces the annual rate of decline in FVC by between about 60 mL and about 70 mL.

28. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 70 mL and about 80 ml,.

29. The method of any one of claims 1 -20, wherein the administration reduces the annual rate of decline in FVC by between about 80 mL and about 90 mL.

30. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by between about 90 mL and about 100 mL. 31. The method of any one of claims 1 -20, wherein the administration reduces the annual rate of decline in FVC by about 100 mL.

32. The method of any one of claims 1-20, wherein the administration reduces the annual rate of decline in FVC by more than 100 mL.

33. The method of any one of claims 1-20, wherein the administration slow's the annual rate of decline in FVC.

34. The method of any one of claims 1-33, wherein the subject is determined to have a slowing in the rate of decline in pulmonary function after the administering.

35. The method of any one of claims 14-34, wherein an annual rate of decline in forced vital capacity (FVC) of the subject is measured over a time period of at least one year after the administering and is compared to a baseline measurement.

36. The method of any one of claims 14-34, wherein an annual rate of decline in forced vital capacity (FVC) of the subject is measured over a time period of at least one year after the administering and is relative to a subject treated with standard of care (SOC).

37. The method of any one of claims 1-35, wherein the subject has at least between about 10% and about 20% fibrosis of the lungs. 38. The method of any one of claims 1-35, wherein the subject has at least between about 20% and about 30% fibrosis of the lungs.

39. The method of any one of claims 1 -35, wherein the subject has at least between about 30% and about 40% fibrosis of the lungs.

40. The method of any one of claims 1-35, wherein the subject has at least between about 40% and about 50% fibrosis of the lungs.

41. The method of any one of claims 1-35, wherein the subj ect has a pattern of nonspecific interstitial pneumonia (NSIP) in the lungs.

42. The method of any one of claims 1-35, wherein the subject has a pattern of usual interstitial pneumonia (UIP) in the Sungs. 43. The method of any one of claims 37-42, wherein the amount of fibrosis or pattern of interstitial pneumonia is determined by High Resolution Computed Topography (HRCT) scan of the lungs.

44. The method of claim 43, wherein the HRCT scan reveals lung parenchymal changes in the subject including any round glass opacity and fibrotic peripheral reticulations and honey combing without abnormalities. 45. The method of any one of claims 1 -43 , wherein the subj ect has at least 10% fibrosis of the lungs as determined by HRCT.

46. The method of claim 43, wherein the HRCT scan is further paired with functional respiratory7 imaging (FRI).

47. The method of any one of claims 1 -46, wherein the subject has an increased level of one or more antibodies selected from the group consisting of antinuclear (ANA), anticentromere (ACA), anti-topoisomerase I (anti-ScI-70), and anti-RNA polymerase III antibodies.

48. The method of any one of claims 1-46, wherein the subject has antinuclear antibodies.

49. The method of any one of claims 1 -46, wherein the subject has anti- topoisomerase I antibodies.

50. The method of any one of claims 1-49, wherein the subject has a diffusing capacity for carbon monoxide (DLCO) of greater than or equal to 40% of predicted. 51. The method of any one of claims 1 -49, wherein the subject has a DLCO of between about 40% and about 89% of predicted.

52. The method of any one of claims 1-49, wherein the subject has a DLCO of between about 90% and about 80% of predicted.

53. The method of any one of claims 1-49, wherein the subject has a DLCO of between about 80% and about 70% of predicted.

54. The method of any one of claims 1-49, wherein the subject has a DLCO of between about 70% and about 60% of predicted.

55. The method of any one of claims 1 -49, wherein the subject has a DLCO of between about 60% and about 50% of predicted.

56. The method of any one of claims 1-49, wherein the subject has a DLCO of between about 50% and about 40% of predicted. 57. The method of any one of claims 1-49, wherein the subject has a DLCO of between about 40% and about 30% of predicted,

58. The method of any one of claims 1-49, wherein the subject has a DLCO of between about 30% and about 20% of predicted.

59. The method of any one of claims 50-58, wherein the DLCO is measured by a rapidly responding gas analyzer (RGA).

60. The method of any one of claims 1-59, wherein the administration improves a modified rodnan skin score (mRSS) of the subject.

61 . The method of any one of claims 1-59, wherein the administration improves the mRSS by between about 1% and about 3%. 62. The method of any one of claims 1 -59, wherein the administration improves the mRSS by between about 1% and about 5%.

63. The method of any one of claims 1-59, wherein the administration improves the mRSS by between about 1% and about 8%.

64. The method of any one of claims 1-59, wherein the administration impro ves the mRSS by between about 1% and about 10%.

65. The method of any one of claims 1-59, wherein the administration improves the mRSS by between about 10% and about 15%.

66. The method of any one of claims 1 -59, wherein the administration improves the mRSS by between about 15% and about 20%.

67. The method of any one of claims 1-59, wherein the administration improves the mRSS by between about 20% and about 25%. 68. The method of any one of claims 1-67, wherein the mRSS is improved after the administering, compared to a baseline measurement.

69. The method of any one of claims 1-67, wherein the mRSS is decreased after the administering, compared to a baseline measurement.

70. The method of any one of claims 1-69, wherein the subject is evaluated for skin thickness using a modified rodnan skin score (mRSS).

71. The method of any one of claims 1 -70, wherein the mRSS is an average of measurements of multiple areas on the subject.

72. The method of any one of claims 1-71 , wherein the subject has a total mRSS of greater than or equal to 15 at baseline. 73. The method of any one of claims 1 -72, wherein the administration improves a

St. George’s Respiratory Questionnaire (SGRQ) score of the subject.

74. The method of any one of claims 1-73, wherein the administration improves the SGRQ by between about 1 % and about 3%.

75. The method of any one of claims 1-73, wherein the administration improves the SGRQ by between about 1% and about 5%.

76. The method of any one of claims 1-73, wherein the administration improves the SGRQ by between about 1% and about 8%.

77. The method of any one of claims 1 -73, wherein the administration improves the SGRQ by between about 1% and about 10%.

78. The method of any one of claims 1-73, wherein the administration improves the SGRQ by between about 10% and about 15%. 79. The method of any one of claims 1-73, wherein the administration improves the SGRQ by between about 15% and about 20%.

80. The method of any one of claims 1-73, wherein the administration improves the SGRQ by between about 20% and about 25%.

81. Hie method of any one of claims 1-80, wherein the SGRQ score is improved after the administering, compared to a baseline measurement.

82. The method of any one of claims 1 -80, wherein the SGRQ is decreased after the administering, compared to a baseline measurement.

83. The method of any one of claims 1-82, wherein the subject is evaluated for respiratory function using a St. George’s Respiratory Questionnaire (SGRQ). 84. The method of any one of claims 1 -83, wherein the administration improves a

Composite Response Index in Systemic Sclerosis (CRISS) score of the subject.

85. The method of any one of claims 1-84, wherein the CRISS score is improved after the administering, compared to a baseline measurement.

86. The method of any one of claims 1-85, wherein the CRISS score is increased after the administering, compared to a baseline measurement.

87. The method of any one of claims 1-86, wherein the administration improves a King’s Brief Interstitial Lung Disease (KB1LD) score of the subject.

88. The method of any one of claims 1 -87, wherein the KBILD score is improved after the administering, compared to a baseline measurement.

89. The method of any one of claims 1-88, wherein the KBILD score is increased after the administering, compared to a baseline measurement. 90. The method of any one of claims 1-89, wherein the administration improves a

Health Assessment Questionnaire-Diability Index (HAQ-DI) score of the subject.

91. Tire method of any one of claims 1-90, wherein the HAQ-DI score is improved after the administering, compared to a baseline measurement.

92. The method of any one of claims 1-91, wherein the administration improves a physical/physician global assessment score of the subject.

93. The method of any one of claims 1 -92, wherein the physical/physician global assessment score is improved after the administering, compared to a baseline measurement.

94. The method of any one of claims 1-93, wherein the administration improves a patient global assessment score of the subject. 95. The method of any one of claims 1 -94, wherein the patient global assessment score is improved after die administering, compared to a baseline measurement.

96. The method of any one of claims 1-95, wherein the administration increases the subject’s length of life.

97. The method of any one of claims 1-95, wherein the administration reduces the subject’s risk of death due to SSe.

98. The method of any one of claims 1-95, wherein the administration reduces the subject’s risk of hospitalization due to SSc.

99. The method of any one of claims 1 -98, wherein the administration lengthens the time to clinical worsening of the subject.

100. The method of any one of claims 1-99, wherein the administration prevents a decline in FVC of the subject of greater than 10% of predicted relative to a baseline measurement.

10 i , The method of any one of claims 1 - 100, wherein the administration preven ts at least a greater than 20% increase in mRSS of the subject.

102. The method of any one of claims 1-101, wherein the administration prevents at least a greater than 5 point increase in total mRSS of the subject.

103. The method of any one of claims 1-102, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of aSMA, ADAM12, Anti-HSP70 IgG, BAFF, BLyS, C3M, C4M, C6M, Cal5.3, CC16, CCL2, CCL18, Collal, Co33al, CTGF, CXCL4, E-selectin, ET-1, fibronectin, 1CAM, 1L-6, 1L-8, KL-6, MCP-1,

MMP7, MMP12, MueSB, Osteopontin, PAI-1, periostim, pro-C3, pro-C4, SP-A, SP-D, Tnfa, VC AM, VEGF, WFDC2 (HE4) and Y K 1.-40.

104. The method of any one of claims 1-102, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of Ca15.3, CC16, KL-6,

SP-A, and SP-D.

105. The method of any one of claims 1-102, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of Collal , Col3al, CTGF, fibronectin, P AI- 1 , and Tnfa.

106. The method of any one of claims 1-102, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of pro-C3, pro-C4, pro- C6, C3M, C4M, C6M, Collal, and aSMA.

107. The method of any one of claims 1-102, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of WFDC2 (HE4), CCL18, osteopontin, periostin, KL-6, YKL-40, and SP-D. 108. The method of any one of claims 1-102, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of pro-C3, pro-C4, pro- C6, C3M, C4M, C6M, Coiial, aSMA, WFDC2 (HE4), CCL18, osteopontin, periostin, KL-6, YKL-40, and SP-D. 109. The method of any one of claims 1-108, wherein the subject has a C-reactive protein (CRP) level of greater than or equal to 3 mg/L.

110. The method of any one of claims 1-109, wherein the subject has one or more clinical complications of the skin.

111. The method of any one of the preceding claims, wherein one or more clinical complications of the skin is selected from the group consi sting of calcinosi s cutis, capi llar}7 changes at the nail beds, depigmentation, digital tip ulcers and/or pitting at fingertips, dryness, edema, hyperpigmentation, lipoatrophy, loss of appendicular hair, pruritus, telangiectasia, and/or traumatic skin ulcerations over finger joints.

112. The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc. 113, The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc according to the European League Against Rheumatism (EULAR) criteria.

114. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of fibrosis of the skin and/or other internal organs, production of specific autoantibodies (e.g., antinuclear antibody (ANA), anticentromere, anti-topoisomerase I, anti-RNA polymerase III), and evidence of vasculopathy.

115. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of skin thickening of fingers extending proximal to metacarpophalangeal joints. 116. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of skm thickening of the fingers, fingertip lesions, telangiectasia, Raynaud’s phenomenon, abnormal nailfold capillaroscopy, and/or presence of pulmonary' arterial hypertension (PAH) and interstitial lung disease (ILD.) 117. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of heartburn, erectile dysfunction in men, and dyspnea.

118. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of fatigue, breathlessness, dry- cough, and bibasilar fine respiratory crackles.

119. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of arrythmia, kidney damage (scleroderma renal crisis), increased risk of cancers, myocardial disease (e.g., myocardial fibrosis, myocardial ischemia), muscle atrophy (e.g., sarcopenia), muscle weakness, myopathy, pericardial disease, and thromboembolic risk.

120. The method of any one of the preceding claims, wherein one or more clinical complications of SSc is selected from the group consisting of musculoskeletal complications, gastrointestinal complications, cardiac complications, renal complications, neuromuscular complications (e.g., muscle atrophy, muscle weakness, myopathy), genitourinary complications (e.g,, erectile dysfunction in men), increased risk of cancer (e.g., lung cancer), and increased risk of thromboembolic complications,

121. The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc comprising one or more complications of the lungs.

122. The method of any one of the preceding claims, wherein one or more complications of the lungs is selected from the group consisting of alveolitis, interstitial pulmonary' fibrosis or interstitial lung disease (ILD), recurrent aspiration, and pulmonary' vasculopathy.

123. The method of any one of the preceding claims, wherein one or more complications of the lungs is selected from the group consisting of ground glass opacities on HRCT, a neutrophilic or eosinophilic BAL, and declining FVC and/or DLCO scores, 124, The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc comprising one or more musculoskeletal complications.

125. The method of any one of the preceding claims, wherein one or more musculoskeletal complications of SSc is selected from the group consisting of arthritis, tendinitis, tendon friction rubs, and/or joint contractures.

126. The method of any one of the preceding claims, wherein one or more musculoskeletal complications of SSc is selected from the group consisting of swelling of the hands, arthralgia, myalgia, and fatigue.

127. The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc comprising one or more gastrointestinal complications.

128. The method of any one of the preceding claims, wherein one or more gastrointestinal complications of SSc is selected from the group consisting of dysphagia, choking, heartburn, hoarseness, cough after swallowing, early satiety, bloating, alternating constipation and/or diarrhea, episodic pseudo-obstruction and/or bacterial small bowel overgrowth with malabsorption, fecal incontinence, chronic gastroesophageal reflux, and recurrent episodes of microaspiration,

129. The method of any one of the preceding claims, wherein the subject has one or more of chronic gastroesophageal reflux and/or recurrent episodes of microaspiration.

130. The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc comprising one or more cardiac complications,

131, The method of any one of the preceding claims, wherein one or more cardiac complications of SSc is selected from the group consisting of complications in myocardium, pericardium, and/or conduction system

132. The method of any one of the preceding claims, wherein the subject has one or more clinical complications of SSc comprising one or more renal complications,

133, The method of any one of the preceding claims, wherein one or more renal complications of SSc is selected from the group consisting of kidney damage, vascular fibrosis of the kidney, interstitial collagen accumulation of the kidney, glomerulonephritis, impaired renal reserve, microalbuminuria, and scleroderma renal crisis (SRC).

134. The method of any one of the preceding claims, wherein the subject has one a complication of SSc comprising SRC,

135, The method of any one of the preceding claims, wherein SRC comprises one or more complications selected from the group consisting of abrupt onset of marked or malignant hypertension, acute onset of oliguric renal failure, urinalysis that reveals only mild proteinuria with few cells or casts, microangiopathic hemolysis anemia and thrombocytopenia. 136, The method of any one of claims 1-135, wherein a dose of the THRU antagonist comprises between about 0.75 mg/to about 6.0 mg/kg of the antagonists.

137. The method of any one of claims 1-135, wherein the TbKII antagonist is administered in a dose of about 0.75 mg/kg once every two weeks to a subject in need thereof.

138, The method of any one of claims 1-135, wherein the TbKII antagonist is administered in a dose of about 2,25 mg/kg once even' four weeks to a subject in need thereof.

139. The method of any one of claims 1-135, wherein the TflRII antagonist is administered in a dose of about 1.5 mg/kg once every two weeks to a subject in need thereof.

140. The method of any one of claims 1-135, wherein the TbMI antagonist is administered in a dose of about 4.5 mg/kg once every four weeks to a subject in need thereof.

141. The method of any one of claims 1-135, wherein the TbKίί antagonist is administered in a dose of about 3.0 mg/kg once every two weeks to a subject in need thereof. 142, The method of any one of claims 1-135, wherein the TbKII antagonist is administered in a dose of about 6,0 mg/kg once every- four weeks to a subject in need thereof.

143. The method of any one of claims 1-142, wherein the TbKII antagonist is administered subcutaneously,

144. The method of any one of claims 1-143, wherein the TbKII antagonist is admini stered at a dose that achi eves a serum concentration of antagonist of between about 10 and about 20 ug/mL in the subject. 145. The method of any one of claims 1-144, wiierein the TbKP antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 30 and about 50 ug/mL in the subject.

146. The method of any one of claims 1-144, wherein the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, ambrisentan, AVID200,

AV1D300, azathioprine, BCD-089, helimumab, BG0QQ11, BMS-986Q20, bortezomib, bosentan, brentuximab, carlumab, CC-90001, clazakizumab, CQR-OGI, cyclophosphamide (CYC), cyclosporine A, dectrekumab, EHR-10Ϊ, elzonris/SL-401, etanercept, FCX-013, fresoiimumab, GLPG169Q, GASK2126458, GSK2330811, GSK3008348, IBIO-CFB03, ifetroban, IFNy, imatinib, immune globulin, IW001, lanifibranor, lebrikizumab, levilimab, losartan, macitentan, MEDI-5117, methotrexate, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decanoate, nintedanib (Ofev), olokizumab, pamrevlumab, pirfenidone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, rituximab, SAR156597, sildenafil, siituximab, simtuzumab, sirolimus, sirukumab, tacrolimus, tadalafil, tanzisertib. TD139, tetrathiomolybdate, tocilizumab, tralokimimab, treprostinil, vobarilizumab, warfarin, ziieuton, and ziltivekimab.

147, The method of any one of claims 1-144, wherein the method further comprises administration of one or more of abatacept, abituzumab, ajulemic acid, ambrisentan,

AVTD200, AV1D300, azatluoprme, BCD-089, belimumab, BG00011, BMS-986020, bortezomib, bosentan, brentuximab, carlumab, CC-90001, clazakizumab, COR-OOl, cyclophosphamide (CYC), cyclosporine A, dectrekumab, EHP-101, elzonris/SL-401, etanercept, FCX-013, fresolimumab, GLPG1690, GASK2126458, GSK2330811, GSK3008348, IBIO-CFB03, ifetroban, IFNy, imatinib, immune globulin, IW001, lanifibranor, lebrikizumab, levilmiab, losartan, macitentan, MEDT-5117, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decanoate, olokizumab, pamrevlumab, pirfemdone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, ntuximab, SARI 56597, sildenafil, siltuximab, simtuzumab, sirolimus, sirukumab, tacrolimus, tadalafrl, tanzisertib, TD 139, tetrathiomolybdate, tocilizumab, tralokinumab, treprostinil, vobarilizumab, warfarin, ziieuton, and ziltivekimab.

148, The method of any one of claims 1-144, wherein the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, AVTD200, AVID300, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IB1Ό- CFB03, ifetroban, imatinib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SAR156597, tacrolimus, tadaiafii, and tocilizumab.

149, The method of any one of claims 1-144, wherein the method further comprises administration of one or more of abatacept, abituzumab, ajulemic acid, AV1D200, AVID300. azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CY C), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, TBIO- CFBQ3, ifetroban, imatinib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, ntuximab, SARI 56597, tacrolimus, tadaiafii, and tocilizumab.

150. The method of any one of claims 1-144, wherein the subject has been treated with one or more of azathiopnne, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and nintedanib. 151. The method of any one of claims 1 - 144, wherein the method further comprises administration of one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and nintedanib.

152. The method of any one of claims 1-144, wherein the method further comprises administration of one or more of cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), nintedanib, and rituximab.

153. The method of any one of claims 1-144, wherein tire subject has been treated with one or more of IL-6 and/or IL-6R antibodies,

154. The method of any one of claims 1-144, wherein the method further comprises administration of one or more of IL-6 and/or IL-6R antibodies.

155. The method of any one of claims 1-144, wherein the subject has been treated with one or more of IL-6 antibodies comprising clazakizumab, COR-001, MEDI-5117, olokizumab, siltuximab, simkumab, and ziltivekimab.

156. The method of any one of claims 1-144, wherein the method further comprises administration of one or more of IL-6 antibodies comprising clazakizumab, COR-001, MEDI-5117, olokizumab, siltuximab, simkumab, and ziltivekimab.

157. The method of any one of claims 1-144, wherein the subject has been treated with one or more of IL-6R antibodies comprising BCD-089 (levilimab), tocilizumab, and vobarilizumab.

158. The method of any one of claims 1-144, wherein the method further comprises administration of one or more of 11.-6 K antibodies comprising BCD-089 (levilimab), tocilizumab, and vobarilizumab.

159. A method of treating systemic sclerosis associated with interstitial lung disease (SSc-lLD), steps comprising: measuring at least one initial point of lung function in a subject; administering a Transforming Growth Factor-b Receptor II (TbKP) fusion protein to the subject; re-measuring the at least one point of lung function m the subject; and based on the measuring, determining a change in the rate of decline in lung function of the subject. 160, The method of claim 159, wherein the rate of decline in lung function is measured by Forced Vital Capacity (FVC) of the subject.

161. The method of any of claims 159-160, wherein the rate of decline in lung function is measured as an annual rate of decline in Forced Vital Capacity (FVC) of the subject

162, The method of any one of claims 159-161, wherein the initial point of lung function comprises an FVC of greater than or equal to 50% of predicted. 163. The method of any of claims 159-161, wherein the initial point of lung function comprises an FVC of between about 90% and about 80% predicted.

164. The method of any of claims 159-161, wherein the initial point of lung function comprises an FVC of between about 80% and about 70% predicted.

165. The method of any of claims 159-161, wherein the initial point of lung function comprises an FVC of between about 70% and about 60% predicted.

166. The method of any of claims 159-161, wherein the initial point of lung function comprises an FVC of between about 60% and about 50% predicted.

167. The method of any of claims 159-161, wherein the initial point of king function comprises an FVC of between about 50% and about 40% predicted.

168. The method of any of claims 159-161, wherein the initial point of lung function comprises an FVC of between about 40% and about 30% predicted.

169. The method of any of claims 159-161, wherein the initial point of lung function comprises an FVC of between about 30% and about 20% predicted.

170. The method of any of claims 159-169, wherein the annual rate of decline in FVC is between about 1% and about 10%. 171. The method of any- of claims 159-169, wherein the annual rate of decline in FVC is between about 10% and about 20%.

172. The method of any of claims 159-169, wherein the annual rate of decline in FVC is between about 20% and about 30%.

173. The method of any' of claims 159-169, wherein the annual rate of decline in FVC is between about 30% and about 40%.

174. The method of any of claims 159-169, wherein the annual rate of decline in FVC is between about 40% and about 50%.

175. The method of any of claims 159-169, wherein the annual rate of decline in FVC is between about 50% and about 60%. 176, The method of any of claims 159-169, wherein the annual rate of decline in FV C is between about 60% and about 70%.

177. The method of any one of claims 159-176, wherein the annual rate of decline in FVC is reduced by about 100 mL,

178. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 1 mL and about 10 mL.

179. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 10 inL and about 20 mL.

180. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 20 mL and about 30 mL.

181. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 30 mL and about 40 mL. 182. The method of any of claims 159-176, wherein the annual rate of decline in

FVC is reduced by between about 40 mL and about 50 mL.

183. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 50 mL and about 60 mL.

184. Tire method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 60 mL and about 70 mL.

185. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 70 mL and about 80 mL.

186. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by between about 80 mL and about 90 mL. 187, The method of any of claims 159-176, wherein the annual rate of decline in

FVC is reduced by between about 90 mL and about 100 mL.

188. The method of any of claims 159-176, wherein the annual rate of decline in FVC is reduced by more than 100 mL.

189. The method of any one of claims 161-188, wherein an annual rate of decline in forced vital capacity (FVC) of the subject is measured over a time peri od of at least one year after the administering and is relative to a subject treated with standard of care (SOC),

190. The method of any one of claims 159-189, wherein a dose of the TbKII antagonist comprises between about 0.75 mg/to about 6.0 mg/kg of the antagonists.

191. The method of any one of claims 159-189, wherein the TbKII antagonist is administered in a dose of about 0.75 mg/kg once every two weeks to a subject in need thereof.

192. The method of any one of claims 159-189, wherein the TbKII antagonist is administered in a dose of about 2.25 mg/kg once every four weeks to a subject in need thereof.

193. The method of any one of claims 159-189, wherein the TfiRII antagonist is administered in a dose of about 1.5 mg/kg once every two weeks to a subject in need thereof. 194. The method of any one of claims 159-189, wherein the TbKII antagonist is administered in a dose of about 4.5 mg/kg once every four weeks to a subject in need thereof.

195. The method of any one of claims 159-189, wherein the TbKII antagonist is administered in a dose of about 3.0 mg/kg once every two weeks to a subject in need thereof.

196. The method of any one of claims 159-189, wherein the TbKP antagonist is administered in a dose of about 6.0 mg/kg once every four weeks to a subject in need thereof.

197. The method of any one of claims 159-196, wherein the TbKII antagonist is administered subcutaneously.

198. The method of any one of claims 159-196, wherein the Tj!RII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 10 and about 20 ug/mL in the subject.

199. The method of any one of claims 159-196, wherein the 'TbKH antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 30 and about 50 ug/mL in the subject.

200. The method of any one of claims 159-199, wherein the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, ambnsenian, AV1D200, AVID300, azathioprine, BCD-089, belimumab, BG00011, BMS-986020, bortezomib, bosentan, brentuximab, carlumab, CC-90001, clazakizumab, COR-001, cyclophosphamide (CYC), cyclosporine A, dectrekumab, EHP-101, elzonris/SL-401, etanercept, FCX-013, fresolimumab, GLPG1690, GASK2126458, GSK2330811, GSK3008348, 1BIO-CFB03, ifetroban, IFNy, imatinib, immune globulin, IW001, lanifibranor, lebrikizumab, levilimab, losartan, macitentan, MEDI-5117, methotrexate, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decanoate, nintedanib (Qfev), olokizumab, pamrevlumab, pirfemdone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, rituxiniab, SAR156597, sildenafil, siltuximab, simtuzumab, sirolimus, sirukumab, tacrolimus, tadalafil, tanzisertib, TD139, tetrathiomolybdate, tocilizumab, tralokinumab, treprostinil, vobarilizumab, warfarin, zileuton, and ziitivekimab. 201. Tlie method of any one of claims 159-199, wherein the method further comprises administration of one or more of abatacept, abituzumab, ajulemic acid, am b risen inn. AVID200, AVID300, azathioprine, BCD-089, belimumab, BG00011, BMS- 986020, bortezomib, bosentan, brentuximab, carlumab, CC-90001, clazakizumab, COR-001, cyclophosphamide (CYC), cyclosporine A, dectrekumab, EHP-101, elzonris/SL-401, etanercept, FCX-013, fresolimumab, GLPG1690, GASK2126458, GSK2330811, GSK3008348, IBIO-CFB03, ifetroban, IFNy, imatinib, immune globulin, IW001, lanifibranor, lebrikizumab, levilimab, losartan, macitentan, MEDI-5117, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decanoate, olokizumab, pamrevlumab, pirfenidone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, rituximab, SAR156597, sildenafil, siltuximab, simtuzumab, sirolimus, sirukumab, tacrolimus, tadalafil, tanzisertib, TD139, tetrathiomolybdate, tocilizumab, tralokinumab, treprostinil, vobarilizumab, warfarin, zileuton, and ziitivekimab.

202. The method of any one of claims 159-199, wherein the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, AYID200, AVID3Q0, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IBIG- CFBQ3, ifetroban, imatinib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SAR156597, tacrolimus, tadaiafii, and tocilizumab.

203, The method of any one of claims 159-199, wherein the method further comprises administration of one or more of abatacept, abituzumab, ajulemic acid, AVID200, AVTD300. azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-I01, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IBIO-CFB03, ifetroban, imatinib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SARI 56597, tacrolimus , tadaiafii , and toci lizumab .

204, The method of any one of claims 159-199, wherein the subject has been treated with one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and nintedanib.

205, The method of any one of claims 159-199, wherein the method further comprises administration of one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and nintedanib, 206. The method of any one of claims 159-199, wherein the method further comprises admini stration of one or more of cyclophosphamide (CY C), methotrexate, mycophenolate mofetil (MMF), nintedanib, and rituximab,

207. The method of any one of claims 159-199, wherein the subject has been treated with one or more of IL-6 and/or IL-6R antibodies,

208. The method of any one of claims 159-199, wherein the method further comprises admini stration of one or more of IL-6 and/or IL-6R antibodies. 209. The method of any one of claims 159-199, wherein the subject has been treated with one or more of IL-6 antibodies comprising clazakizumab, COR-001, MED1- 5117, olokizumab, siltuximab, sirukumab, and ziltivekimab.

210. The method of any one of claims 159-199, wherein the method further comprises administration of one or more of 1L-6 antibodies comprising clazakizumab, COR- 001, MEDI-5117, olokizumab, siituximab, sirukumab, and ziltivekimab. 211. The method of any one of claims 159-199, wherein the subject has been treated with one or more of 1L-6R antibodies comprising BCD-089 (levilimab), tocilizumab, and vobarilizumab.

212. The method of any one of claims 159-199, wherein the method further comprises administration of one or more of IL-6R antibodies comprising BCD-089

(levilimab), tocilizumab, and vobarilizumab,

213. The method of any of the preceding claims, wherein the TpRil antagonist comprises a TbKίί extracellular domain, wherein the TbKP extracellular domain comprises an amino acid sequence at least 80% identical to: i) a sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1 or; ii) a sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2.

214. The method of claim 213, wherein the TbKII extracellular domain comprises an amino acid sequence at least 90% identical to a sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1. 215, The method of claim 213, wherein the TbKII extracellular domain comprises an amino acid sequence at least 95% identical to a sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1.

216. The method of claim 213, wherein the TbKP extracellular domain comprises an amino acid sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1.

217. The method of claim 213, wherein the TbKII extracellular domain comprises an amino acid sequence at least 90% identical to a sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2. 218. The method of claim 213, wherein the TflRII extracellular domain comprises an amino acid sequence at least 95% identical to a sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2.

219. The method of claim 213, wherein tire TbKII extracellular domain comprises an amino acid sequence beginning at any of positions 23 to 60 of SF1Q ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2.

220. The method of claim 213, wherein the TbKII extracellular domain comprises an amino acid sequence at least 90% identical to SEQ ID NO: 18.

221. The method of claim 213, wherein the TbKII extracellular domain comprises an amino acid sequence at least 95% identical to SP!Q ID NO: 18.

222. The method of claim 213, wherein the TbKII extracellular domain comprises the amino acid sequence of SEQ ID NO: 18.

223. The method of any of the preceding claims, wherein the TbKII antagonist is a fusion protein further comprising a heterologous domain. 224, The method of claim 223, wherein the heterologous domain comprises an immunoglobulin Fc domain.

225. The method of claim 223, wherein the immunoglobulin Fc domain is a human immunoglobulin Fc domain.

226. The method of claim 223, wherein the heterologous domain comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 20.

227. The method of claim 223, wherein the heterologous domain comprises an amino acid sequence that is at least 95% identical to SEQ ID NO: 20.

228. The method of claim 223, wherein the heterologous domain comprises the amino acid sequence of SEQ ID NO: 20.

229. The method of claim 223, wherein the heterologous domain comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 72. 2.30, The method of claim 223, wherein the heterologous domain comprises an amino acid sequence that is at least 95% identical to SEQ ID NO: 72.

231. The method of claim 223, wherein the heterologous domain comprises the amino acid sequence of SEQ ID NO: 72.

232. The method of any of the preceding claims, wherein the TbKK antagonist or fusion protein further comprises a linker

233. The method of any one of claims 159-232, wherein the linker comprises (GGGGS)r., wTierein n = > 4.

234. Tire method of any one of claims 159-233, wherein the linker comprises the amino acid sequence of SEQ ID NO: 6. 235, The method of any one of claims 159-134, wherein the TbKII antagonist or fusion protein does not include amino acids 185-592 of SEQ ID NO: 2.

236. The method of any one of claims 159-235, wherein the TbKP antagonist or fusion protein does not include amino acids 1-22 of SEQ ID NO: 2.

237. The method of claim 223, wherein the fusion protein consists of or consists essentially of: a) a TpRII polypeptide portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids; b) a linker portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3,

2 or 1 additional amino acids; e) a heterologous portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 20 or SEQ ID NO: 72 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids; and d) optionally a leader sequence (e.g., SEQ ID NO: 23).

238. The method of claim 223 wherein the fusion protein consists of or consists essentially of: a) a TbMII polypeptide portion comprising the amino acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids; b) a linker portion comprising the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or 1 additional amino acids; c) a heterologous portion comprising the amino acid sequence of SEQ ID NO: 20 or SEQ ID NO: 72 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids; and d) optionally a leader sequence (e.g., SEQ ID NO: 23).

239. The method of claim 223, wherein the fusion protein comprises: a) an extracellular domain of a TbKII portion; wherein the extracellular domain comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the sequence of SEQ ID NO: 18; b) a heterologous portion, wherein the heterologous portion comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the sequence of SEQ ID NO: 20 or SEQ ID NO: 72; and c) a linker portion connecting the extracellular domain and the heterologous portion; wherein the linker comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or

99% identical to the ammo acid sequence of SEQ ID NO: 6.

240. The method of claim 223, wherein the fusion protein comprises: a) an extracellular domain of a TpRII portion; wherein the extracellular domain comprises the amino acid sequence of SEQ ID NO: 18; b) a heterologous portion, wherein the heterologous portion comprises the amino acid sequence of SEQ ID NO: 20 or SEQ ID NO: 72; and c) a linker portion connecting the extracellular domain and the heterologous portion; wherein the linker comprises the amino acid sequence of SEQ ID NO: 6.

241. The method of any of the preceding claims, wherein the TbKII antagonist comprises an amino acid sequence at least 90% identical to SE1Q ID NO: 48.

242. The method of any of the preceding claims, wherein the TbKII antagonist comprises an amino acid sequence at least 95% identical to SEQ ID NO: 48. 243. The method of any of the preceding claims, wherein the TbKII antagonist comprises the amino acid sequence of SEQ ID NO: 48.

244. The method of any of the preceding claims, wherein the TbR.P antagonist consists of the amino acid sequence of SEQ ID NO: 48.

245. The method of any of the preceding claim s, wherein the TbKίί antagonist comprises an amino acid sequence at least 90% identical to SEQ ID NO: 67.

246. The method of any of the preceding claims, wherein the TbKII antagonist comprises an amino acid sequence at least 95% identical to SEQ ID NO: 67.

247. The method of any of tire preceding claims, wherein tire TbKII antagonist comprises the amino acid sequence of SEQ ID NO: 67. 248. The method of any of the preceding claims, wherein the TbKII antagonist consists of the ammo acid sequence of SEQ ID NO: 67.

249, The method of any one of claims 159-248, wherein the antagonist includes one or more modified amino acid residues selected from: a glycosylated amino acid, a PEGylated amino acid, a famesylated amino acid, an acetylated amino acid, a biotinylated amino acid, an amino acid conjugated to a lipid moiety, and an amino acid conjugated to an organic derivatizing agent.

250, The method of claim 249, wherein the antagonist is glycosylated.

Description:
TREATMENTS FOR SYSTEMIC SCLEROSIS

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of and priority to U.S. Provisional Application No. 62/930,941, filed November 5, 2019, Tire foregoing application is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

Members of the transforming growth factor-beta (TORb) superfamily are pleiotropic cytokines involved in essential cellular functions such as proliferation, differentiation, apoptosis, motility, extracellular matrix production, tissue remodeling, angiogenesis, immune response, cell adhesion, and also play a key role in pathophysiology of disease states as different as chronic inflammatory' conditions and cancer. Members of the TORb superfamily have been classified into major family groupings, which include TGF[¾. bone morphogenetic proteins (BMP), osteogenic proteins (OP), growth and differentiation factors (GDF), inhibins/activins, muilerian inhibitory' substances (MIS) and glial derived neurotrophic factors (GDNF).

TGFβ superfamily members transduce their signals across the plasma membrane by inducing the formation of heteromeric complexes of specific type I and type P serine/threonine kinase receptors, which in turn activate a particular subset of SMAD proteins (some inhibitory and some excitatory). Hie SMAD molecule compounds relay the signals into the nucleus where they direct transcriptional responses in concert with other proteins.

Dysfunctional TGFp superfamily signaling has been linked to several clinical disorders including cancer, fibrosis, bone diseases, diabetic nephropathy, as well as chronic vascular diseases such as atherosclerosis.

Systemic sclerosis (SSc) is a rare fibrotic disease with an incidence of between about 20 to 24 people per 1 million people in the US every year. A common manifestation of SSc is interstitial lung disease (ILD), which affects approximately 80% of SSc patients and is one of the leading causes of death of those with SSc. Currently, nintedamb is the only approved therapy available in the US for SSc-ILD, but it failed to meet key secondary endpoints such as an improvement in modified Rodnan Skin Score (mRSS) and St. George’s Respiratory' Questionnaire (SGRQ) score. Therefore, a need exists to develop new treatments for SSc-ILD SUMMARY OF THE INVENTION

In part, the disclosure relates to TGFfi type II receptor (TbKίί) antagonists (inhibitors) that can be used to treat systemic sclerosis (SSc), particularly clinical complications of SSc including, for example, interstitial lung disease (ILD). Accordingly, the disclosure provides methods for treating SSc comprising administering to a patient in need thereof one or more TbKIΊ antagonist. In some embodiments, the disclosure provides methods for treating one or more complications of SSc comprising administering to a patient in need thereof one or more TbIIII antagonist. In some embodiments, the disclosure provides methods for treating SSc- ILD compri sing admini stering to a patient in need thereof one or more TpRII antagonist. Optionally, such methods further comprise administering to the patient one or more additional active agents and/or supportive therapies for treating SSc or a complication of SSc (e.g, SSc-ILD).

In part, the disclosure provides TbKII polypeptides and the use of such polypeptides as selective antagonists for TORbί or TORb3. As described herein, fusion proteins comprising part or all of the TpRH extracellular domain (ECD), with or without additional mutations, bind to and/or inhibit ' TORbI or TORb3 with varying affinities. In particular, TpRIf fusion proteins comprising a heterologous portion (e.g., an Fc immunoglobulin domain) and a linker of at least 10 amino acids in length (e.g., a linker having the amino acid sequence of SEQ ID NO: 6) are associated with superior TORbI and TORb3 binding properties as compared to TbKII polypeptides having a shorter linker. Thus, the disclosure provides TbKII polypeptides, and fusion proteins thereof, for use in antagonizing the TOR If signaling pathway as well as uses of such TbKII antagonists in treating TGFp-associated disorders including, for example SSc as well as complications of SSc (e.g., SSc-ILD). While TbKII antagonists, and fusion proteins comprising the same, may affect SSc, including complications of SSc (e.g., SSc-ILD), through a mechanism other that TbIIII antagonism, the disclosure nonetheless provides that desirable therapeutic agents may be selected on the basis of TbKII antagonism. Therefore, while not wishing to be bound to a particular mechanism of action, it is expected that other TbKII antagonists may he useful in the treatment of SSc, particularly complications of SSc such as SSc-ILD. For example, agents that inhibit the activity and/or expression(e.g., transcription, translation, secretion from a cell, or combinations thereof) of one or more of: i) the ' TbMII receptor, ii) one or more TbKIΐ-Mhά^ ligand (e.g., TORbI , TORb2, and/or TORb3); iii) one or more TbKII- associated type I receptor (e.g., ALK5): iv) one or more T^RII-associated co-receptor (e.g., betaglycan); and/or v) one or more TpRJI downstream signaling component (e.g., Smad proteins), as well as combinations thereof, may be useful in the treatment of SSc, particularly complications of SSc such as SSc-ILD. Such agents are collectively referred to herein as “TbKII antagonists” or “TpRII inhibitors”. In some embodiments, the disclosure provides for methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor P ( ' TbKII) antagonist to a subject in need thereof. In some embodiments, the disclosure provides for methods of treating systemic sclerosis, comprising administering a TbIIII polypeptide to a subject in need thereof. In some embodiments, the systemic sclerosis is diffuse systemic sclerosis (deSSe),

In some embodiments of the present disclosure, forced vital capacity (FV C) of the subject is measured, among other characteristics. In some embodiments of the present disclosure, forced vital capacity (FVC) of the subject is measured in liters, milliliters, and/or percentage of predicted. In some embodiments, a “normal” range for FVC is typically considered to be between 80% and 100% of predicted. In some embodiments, “of predicted” refers to reporting of the subject’s results as a percentage of known predicted values for a subject of similar characteristics (e.g. height, sex, age, race, weight). In some embodiments, the subject has an FVC of greater than or equal to 50% of predicted. In some embodiments, the subject has an FVC of between about 100% and about 90% of predicted. In some embodiments, the subject has an FVC of between about 90% and about 80% of predicted. In some embodiments, the subject has an FVC of between about 80% and about 70% of predicted. In some embodiments, the subject has an FVC of between about 70% and about 60% of predicted. In some embodiments, the subject has an FVC of between about 60% and about 50% of predicted. In some embodiments, the subject has an FVC of between about 50% and about 40% of predicted. In some embodiments, the subject has an FV C of between about 40% and about 30% of predicted. In some embodiments, the subject has an FVC of between about 30% and about 20% of predicted. In some embodiments, FVC is measured by spirometry.

In some embodiments of the present disclosure, a general measurement of disease progression of the subject may be presented as an annual rate of decline in FVC. In some embodiments, an annual rate of decline m FVC of the subject is reduced by between about 1% and about 10%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 5% and about 10%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10% and about 15%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 15% and about 20%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20% and about 25%. in some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 25% and about 30%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30% and about 35%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 35% and about 40%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40% and about 45%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 45% and about 50%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50% and about 55%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 55% and about 60%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60% and about 65%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 65% and about 70%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10% and about 20%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20% and about 30%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30% and about 40%. In some embodiments, an annual rate of decline in FV C of the subject is reduced by between about 40% and about 50%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50% and about 60%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60% and about 70%.

In some embodiments, an annual rate of decline is measured in “mL”. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 1 mL and about 10 rnL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 5 mL and about 10 mL. In some embodiments, an amiual rate of decline in FVC of the subject is reduced by between about 10 mL and about 15 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 15 mL and about 20 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20 mL and about 25 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 25 ml, and about 30 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30 mL and about 35 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 35 rnL and about 40 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40 mL and about 45 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 45 mL and about 50 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50 mL and about 55 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 55 ml, and about 60 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60 mL and about 65 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 65 mL and about 70 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 70 mL and about 75 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 75 mL and about 80 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 85 mL and about 90 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 90 mL and about 95 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 95 mL and about 100 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by about 100 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10 rnL and about 20 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20 mL and about 30 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30 mL and about 40 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40 rnL and about 50 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50 mL and about 60 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60 mL and about 70 rnL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 70 mL and about 80 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 80 mL and about 90 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 90 mL and about 100 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by more than about 100 mL.

In some embodiments, the administration slows the annual rate of decline in FVC. In some embodiments, the subject is determined to have a slowing m the rate of decline in pulmonary function after the administering. In some embodiments, an annual rate of decline in forced vital capacity (FVC) of the subject is measured over a time period of at least one year after the administering and is compared to a baseline measurement. In some embodiments, an annual rate of decline in forced vital capacity (FVC) of the subject is measured over a time period of at least one year after the administering and is compared to a subject treated with standard of care (SOC).

In some embodiments of the present disclosure, High Resolution-Computer Topography (HRCT) can be utilized for diagnosing and evaluating an amount of fibrosis of the lungs. In some embodiments, the subject has at least between about 1% and about 10% fibrosis of tire lungs. In some embodiments, the subject has at least between about 1% and about 5% fibrosis of the lungs. In some embodiments, the subject has at least between about 5% and about 10% fibrosis of the lungs. In some embodiments, the subject has between about 10% and about 15% fibrosis of the lungs. In some embodiments, the subject has at least between about 10% and about 20% fibrosis of the lungs. In some embodiments, the subject has at least between about 2.0% and about 30% fibrosis of the lungs. In some embodiments, the subject has at least between about 30% and about 40% fibrosis of the lungs. In some embodiments, the subject has at least between about 40% and about 50% fibrosis of the lungs. In some embodiments, the subject has a patern of nonspecific interstitial pneumonia (NSIP) in the lungs. In some embodiments, the subject has a pattern of usual interstitial pneumonia (UIP) in the lungs. In some embodiments, the amount of fibrosis or pattern of interstitial pneumonia is determined by High Resolution Computed Topography (HRCT) scan of the lungs. In some embodiments, an HRCT scan reveals lung parenchymal changes in the subject including any round glass opacity and fibrotic peripheral reticulations and honey combing without abnormalities. In some embodiments, the subject has at least 10% fibrosis of tire lungs as determined by HRCT. In some embodiments, an HRCT scan is further paired with functional respiratory imaging (FRI).

In some embodiments of the present disclosure, serologic tests are performed in the subject. In some embodiments, antibodies (e.g. autoantibodies) are detected in a serologic test. In some embodiments, the subject has an increased level of one or more antibodies selected from the group consisting of antinuclear (ANA), anticentromere (ACA), anti- topoisornerase I (anti-ScI-70), and anti-RNA polymerase III antibodies. In some embodiment, the subject has antinuclear antibodies. In some embodiments, the subject has anti -topoisornerase I antibodies.

In some embodiments of the present disclosure, the subject has a diffusing capacity for carbon monoxide (DLCQ) of greater than or equal to 40% of predicted. In some embodiments of the present disclosure, the subject has a DLCO of between about 40% and about 89% of predicted. In some embodiments of the present disclosure, the subject has a DLCO of between about 90% and about 80% of predicted. In some embodiments, the subject has a DLCO of between about 80% and about 70% of predicted. In some embodiments, the subject has a DLCO of between about 70% and about 60% of predicted. In some embodiments, the subject has a DLCO of between about 60% and about 50% of predicted. In some embodiments, the subject has a DLCO of between about 50% and about 40% of predicted. In some embodiments, the subject has a DLCO of between about 40% and about 30% of predicted. In some embodiments, the subject has a DLCO of between about 30% and about 20% of predicted. In some embodiments, the DLCO is measured by a rapidly responding gas analyzer (RGA).

In some embodiments, the present disclosure provides methods of treating SSc with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor P ( ' TbKP) antagonist to a subject in need thereof, wherein the subject has an improved distance walked in a six minute walk test after the administration. In some embodiments, a subject improves distance walked in a six minute walk test by at least about 30 meters compared to a reference subject. In some embodiments, a reference subject is a healthy person of similar characteristics (e.g,, sex, age, height, weight). In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 10 meters and about 15 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 15 meters and about 20 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 20 meters and about 25 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 25 meters and about 30 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 30 meters and about 35 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 35 meters and about 40 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 40 meters and about 45 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 45 meters and about 50 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 50 meters and about 55 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 55 meters and about 60 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 60 meters and about 65 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 65 meters and about 70 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 70 meters and about 75 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute w¾dk test by at least between about 75 meters and about 80 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 80 meters and about 85 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 85 meters and about 90 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 90 meters and about 95 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 95 meters and about 100 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least more than 100 meters compared to a reference subject.

In some embodiments, the subject is evaluated for skin thickness using a modified rodnan skin score (niRSS). In some embodiments, the administration improves a modified rodnan skin score (rriRSS) of the subject. In some embodiments, tire administration improves an mRSS of the subject by between about 1% and about 3%. In some embodiments, the administration improves an mRSS of the subject by between about 1% and about 5%. In some embodiments, the administration improves an mRSS of the subject by between about 1% and about 8%. In some embodiments, the administration improves an mRSS of the subject by between about 1% and about 10%. In some embodiments, the administration improves an mRSS of the subject by between about 10% and about 15%. In some embodiments, the administration improves the mRSS by between about 10% and about 20%. In some embodiments, the administration improves the mRSS by between about 20% and about 30%. In some embodiments, the administration improves the mRSS by between about 30% and about 40%. In some embodiments, the administration improves the mRSS by between about 40% and about 50%. In some embodiments, the administration improves the mRSS by between about 50% and about 60%. In some embodiments, the administration improves the mRSS by between about 60% and about 70%. In some embodiments, the mRSS is improved after the administering, compared to a baseline measurement. In some embodiments, the mRSS is decreased after the administering, compared to a baseline measurement. In some embodiments, the mRSS is an average of measurements of multiple areas on the subject. In some embodiments, the subject has a total mRSS of greater than or equal to 15 at baseline.

In some embodiments, the administration improves a St. George’s Respiratory' Questionnaire (SGRQ) score of the subject. In some embodiments, the administration improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 1% and about 3%. In some embodiments, the administration improves a St. George’s Respirator}' Questionnaire (SGRQ) score of a subject by between about 1% and about 5%. In some embodiments, the administration improves a St. George’s Respiratory' Questionnaire (SGRQ) score of a subject by between about 1% and about 8%. In some embodiments, the administration improves a St. George’s Respiratory' Questionnaire (SGRQ) score of a subject by between about 1% and about 10%. In some embodiments, the administration improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 10% and about 15%. In some embodiments, the administration improves the SGRQ by between about 10% and about 20%. In some embodiments, the administration improves the SGRQ by between about 20% and about 30%. In some embodiments, the administration improves the SGRQ by between about 30% and about 40%. In some embodiments, the administration improves the SGRQ by between about 40% and about 50%. In some embodiments, the administration improves tire SGRQ by between about 50% and about 60%. In some embodiments, the administration improves the SGRQ by between about 60% and about 70%. In some embodiments, the SGRQ score is improved after the administering, compared to a baseline measurement. In some embodiments, the SGRQ is decreased after the administering, compared to a baseline measurement. In some embodiments, the subject is evaluated for respiratory function using a St. George’s Respiratory Questionnaire (SGRQ). In some embodiments, the administration changes the subject’s SGRQ score by about 4 units.

In some embodiments, the administration changes the subject’s SGRQ score by about 8 units. In some embodiments, the administration changes the subject’s SGRQ score by about 12 units. In some embodiments, the administration changes the subject’s SGRQ score by between about 0 and about 4 units. In some embodiments, the administration changes the subject’s SGRQ score by between about 1 and about 4 units. In some embodiments, the administration changes the subject’s SGRQ score by between about 5 and about 8 units. In some embodiments, the administration changes the subject’s SGRQ score by between about 9 and about 12 units. In some embodiments, the administration changes the subject’s SGRQ score by about more than 12 units.

In some embodiments, the administration improves a Composite Response Index in Systemic Sclerosis (CRISS) score of the subject. In some embodiments, the CR1SS score is improved after the administering, compared to a baseline measurement. In some embodiments, the CRISS score is increased after the administering, compared to a baseline measurement.

In some embodiments, the administration improves a King’s Brief Interstitial Lung Disease (KBILD) score of the subject. In some embodiments, the KBILD score is improved after the administering, compared to a baseline measurement. In some embodiments, the

KBILD score is increased after the administering, compared to a baseline measurement.

In some embodiments, the administration improves a Health Assessment Questionnaire-Diability Index (HAQ-DI) score of the subject. In some embodiments, the HAQ-DI score is improved after the administering, compared to a baseline measurement. In some embodiments, the administration improves a physical/physician global assessment score of the subject. In some embodiments, the physical/physician global assessment score is improved after the administering, compared to a baseline measurement.

In some embodiments, the administration improves a patient global assessment score of the subject. In some embodiments, the patient global assessment score is improved after the administering, compared to a baseline measurement.

In some embodiments of the present disclosure, the administration increases the subject’s length of life. In some embodiments, the administration reduces the subject’s risk of death due to SSc (e.g. SSc-ILD). In some embodiments, the adm inistration reduces the subject’s risk of hospitalization due to SSc (e.g. SSc-ILD). In some embodiments, the administration lengthens the time to clinical worsening of the subject. In some embodiments, the administration prevents a decline in FVC of the subject of greater than 10% of predicted relative to a baseline measurement. In some embodiments, the administration prevents at least a greater than 20% increase in mRSS of the subject. In some embodiments, the administration prevents at least a greater than 5 point increase in total mRSS of the subject. In some embodiments, the subject has an increased level of one or more biomarkers selected from the group consisting of aSMA, ADAM 12, Anti-HSP7G IgG, BAFF, BLyS, C3M, C4M, C6M, Cal5,3, CC16, CCL2, CCL18, Colla!, Col3al, C ' TGF, CXCL4, E- selectin, ET-1, fibronectin, ICAM, IL-6, IL-8, KL-6, MCP-1, MMP7, MMP12, Muc5B, Osteopontin, PAI-1, periostin, pro-C3, pro-C4, SP-A, SP-D, Tnfa, YCAM, YEGF, WFDC2 (HE4) and YKL-40. In some embodiments, the subject has an increased level of one or more biomarkers selected from the group consisting of Cal5.3, CC16, KL-6, SP-A, and SP- D. In some embodiments, the subject has an increased level of one or more biomarkers selected from the group consisting of Collal, ColBal, CTGF, fibronectin, PAI-1, and Tnfa. In some embodiments, the subject has an increased level of one or more biomarkers selected from the group consisting of pro-C3, pro-C4, pro-C6, C3M, C4M, C6M, Collal, and aSMA. In some embodiments, the subject has an increased level of one or more biomarkers selected from the group consisting of WFDC2 (HE4), CCL18, osteopontin, periostin, KL-6, YKL-40, and SP-D. In some embodiments, the subject has an increased level of one or more biomarkers selected from the group consisting of pro~C3, pro~C4, pro-C6, C3M, C4M, C6M, Collal, aSMA, WFDC2 (HE4), CCL18, osteopontin, periostin, KL-6, YKL-40, and SP-D.

In some embodiments, the subject has a C -reactive protein (CRP) level of greater than or equal to 3 mg/1,.

In some embodiments, a subject of the present disclosure has one or more clinical complications of SSc. In some embodiments, the subject has one or more clinical complications of the skin. In some embodiments, one or more clinical complications of the skin is selected from the group consisting of calcinosis cutis, capillary' changes at the nail beds, depigmentation, digital tip ulcers and/or pitting at fingertips, dryness, edema, hyperpigmentation, lipoatrophy, loss of appendicular hair, pruritus, telangiectasia, and/or traumatic skin ulcerations over finger joints. in some embodiments, one or more clinical complications of SSc is selected from the group consisting of fibrosis of the skin and/or other internal organs, production of specific autoantibodies (e.g., antinuclear antibody (ANA), anticentromere, anti -topoisome rase I, anti- RNA polymerase III), and evidence of vasculopathy. In some embodiments, the subject has one or more clinical complications of SSc according to the European League Against

Rheumatism (EULAR) criteria. In some embodiments, one or more clinical complications of SSc is selected from the group consisting of skin thickening of fingers extending proximal to metacarpophalangeal joints. In some embodiments, one or more clinical complications of SSc is selected from the group consisting of skin thickening of the fingers, fingertip lesions, telangiectasia, Raynaud’s phenomenon, abnormal nailfold capillaroscopy, and/or presence of pulmonary arterial hypertension (PAH) and interstitial lung disease (ILD.) In some embodiments, one or more clinical complications of SSc is selected from the group consisting of heartburn, erectile dysfunction in men, and dyspnea. In some embodiments, one or more clinical complications of SSc is selected from tire group consisting of fatigue, breathlessness, dry cough, and bibasilar fine respiratory crackles. In some embodiments, one or more clinical complications of SSc is selected from the group consisting of arrythmia, kidney damage (scleroderma renal crisis), increased risk of cancers, myocardial disease (e.g., myocardial fibrosis, myocardial ischemia), muscle atrophy (e.g., sarcopenia), muscle weakness, myopathy, pericardial disease, and thromboembolic risk.

In some embodimen ts, of the present disclosure, one or more clinical complications of SSc is selected from die group consisting of musculoskeletal complications, gastrointestinal complications, cardiac complications, renal complications, neuromuscular complications (e.g., muscle atrophy, muscle weakness, myopathy), genitourinary 7 complications (e.g., erectile dysfunction in men), increased risk of cancer (e.g., lung cancer), and increased risk of thromboembolic complications.

In some embodiments, the subject has one or more clinical complications of SSc comprising one or more complications of the lungs. In some embodiments, one or more complications of the lungs is selected from the group consisting of alveolitis, interstitial pulmonary fibrosis or interstitial lung disease (ILD), recurrent aspiration, and pulmonary vasciiiopalhy. In some embodiments, one or more complications of the lungs is selected from the group consisting of ground glass opacities on HRCT, a neutrophilic or eosinophilic BAL, and declining FVC and/or DLCO scores.

In some embodiments of the present disclosure, the subject has one or more clinical complications of SSc comprising one or more musculoskeletal complications. In some embodiments, one or more musculoskeletal complications of SSc is selected from the group consisting of arthritis, tendinitis, tendon friction rubs, and/or joint contractures. In some embodiments, one or more musculoskeletal complications of SSc is selected from the group consisting of swelling of the hands, arthralgia, myalgia, and fatigue. In some embodiments of the present disclosure, the subject has one or more clinical complications of SSc comprising one or more gastrointestinal complications. In some embodiments, one or more gastrointestinal complications of SSc is selected from the group consisting of dysphagia, choking, heartburn, hoarseness, cough after swallowing, early satiety, bloating, alternating constipation and/or diarrhea, episodic pseudo-obstruction and/or bacterial small bowel overgrowth with malabsorption, fecal incontinence, chronic gastroesophageal reflux, and recurrent episodes of microaspiration. In some embodiments, the subject has one or more of chronic gastroesophageal reflux and/or recurrent episodes of microaspiration . In some embodiments of the present disclosure, the subject has one or more clinical complications of SSc comprising one or more cardiac complications. In some embodiments, one or more cardiac complications of SSc is selected from the group consisting of complications in myocardium, pericardium, and/or conduction system

In some embodiments, the subject has one or more clinical complications of SSc comprising one or more renal complications. In some embodiments, one or more renal complications of SSc is selected from the group consisting of kidney damage, vascular fibrosis of the kidney, interstitial collagen accumulation of the kidney, glomerulonephritis, impaired renal reserve, microalbuminuria, and scleroderma renal crisis (SRC). In some embodiments, the subject has one a complication of SSc comprising SRC. In some embodiments, SRC comprises one or more complications selected from the group consisting of abrupt onset of marked or malignant hypertension, acute onset of oliguric renal failure, urinalysis that reveals only mild proteinuria with few cells or casts, microangiopathic hemolysis anemia and thrombocytopenia.

In some embodiments, the disclosure provides methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), comprising administering a Transforming Growth Facto r-b Receptor II f ί ' bKϋ) antagonist to a subject in need thereof, wherein a dose of the TbKII antagonist comprises between about 0.75 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, the TbKII antagonist is administered in a dose of about 0.75 mg/kg once every two weeks to a subject in need thereof. In some embodiments, the TpRIl antagonist is administered in a dose of about 2.25 mg/kg once every four weeks to a subject in need thereof. In some embodiments, the TpRH antagonist is administered in a dose of about 1.5 mg/kg once every two weeks to a subject in need thereof. In some embodiments, the TbIIII antagonist is administered in a dose of about 4.5 mg/kg once every four weeks to a subject m need thereof. In some embodiments, the TjlRII antagonist is administered in a dose of about 3.0 rng/kg once every two weeks to a subject in need thereof. In some embodiments, the TpRll antagonist is administered in a dose of about 6.0 mg/kg once every four -weeks to a subject in need thereof. In some embodiments, the TbKII antagonist is administered subcutaneously. In some embodiments, the TbKII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 10 and about 20 ug/mL in the subject. In some embodiments, the TbKII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 30 and about 50 ug/mL in the subject.

In some embodiments, the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, ambrisentan, AVID200, AVID300, azathioprine, BCD-089, belimumab, BG00011, BMS-986020, bortezomib, bosentan, brentuximab, carlumab, CC- 90001, clazakizumab, COR-001, cyclophosphamide (CYC), cyclosporine A, deefrekumab, EHP-101, elzonris/SL-401, etanercept, FCX-013, fresobmumab, GLPG1690,

GASK2126458, GSK2330811, GSK3008348, IBIO-CFB03, ifetroban, IFNy, miatmib, immune globulin, IW001, lanifibranor, lebrikizuniab, levilimab, losartan, macitentan, MED1- 5117, methotrexate, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decan oate, nintedanib (Ofev), oiokizumab, pamrevlumab, pirfenidone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, rituximab, SAR156597, sildenafil, siltuximab, simtuzumab, sirolimus, sirakumab, tacrolimus, tadalafil, tanzisertib, TD139, tetrathiomolybdate, tocilizumab, tralokinumab, treprostinil, vobarilizumab, warfarin, zileuton, and ziltivekimab.

In some embodiments, methods of the present disclosure further comprise administration of one or more of abatacept, abituzumab, ajulemic acid, ambrisentan, AVID200, AVID300, azathioprine, BCD-089, belimumab, BGQ0011, BMS-986020, bortezomib, bosentan, brentuximab, carlumab, CC-90001, clazakizumab, CQR-G01, cyclophosphamide (CYC), cyclosporine A, dectrekumab, EHP-101, elzonris/SL-401, etanercept, FCX-013, fresobmumab, GLPG1690, GASK2I26458, GSK2330811, GSK3008348, IBIO-CFB03, ifetroban, IFNy, imatimb, immune globulin, IWQ01, lanifibranor, lebrikizumab, levilimab, losartan, macitentan, MEDI-5117, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decanoate, oiokizumab, pamrevlumab, pirfenidone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, rituximab, SARI 56597, sildenafil, siituximab, simtuzumab, sirolimus, sirukumab, tacrolimus, tadalafil, tanzisertib, TD139, tetrathiomolybdate, tocilizumab, tralokinumab, treprostinil, vobarilizumab, warfarin, zileuton, and ziltivekimab. In some embodiments of the present disclosure, the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, AVID200, AVID300, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPGI690, GSK233Q811, IBIO-CFB03, ifetroban, imatimb, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SAR156597, tacrolimus, tadalafsl, and tocilizumab.

In some embodiments, methods of the present disclosure further comprise administration of one or more of abatacept, abituzumab, ajulemic acid, AVID20G, AVID300, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IBIO- CFB03, ifetroban, imatinib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomahdomide, privigen, riociguat, rituximab, SAR156597, tacrolimus, tadalafii, and tocilizumab. In some embodiments, the subject has been treated with one or more of azathioprine, cyclophosphamide (CY C), mycophenolate mofetil (MMF), and nintedanib.

In some embodiments, methods of the present disclosure further comprise administration of one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and nintedanib. In some embodiments, methods of the present disclosure further comprise administration of one or more of cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), nintedanib, and rituximab.

In some embodiments, the disclosure provides methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TbKII) polypeptide to a subject in need thereof, wherein the method further comprises administration of one or more of IL-6 and/or IL-6R antibodies.

In some embodiments, IL-6 antibodies comprise one or more of clazakizumab, COR- 001, MEDI-5117, olokizumab, si!tuximab, sirukumab, and zi!tivekimah. In some embodiments, IL-6R antibodies comprise one or more of BCD-089 (levilimab), tocilizumab, and vobarilizumab.

In some embodiments, the subject has been treated with one or more of clazakizumab, COR-001, MEDI-5117, olokizumab, siltuximab, sirukumab, and ziltivekimab. in some embodiments, methods of the present disclosure further comprise administration of one or more of clazakizumab, COR-001, MEDI-5117, olokizumab, siltuximab, sirukumab, and ziltivekimab.

In some embodiments, the subject has been treated with one or more of BCD-089 (levilimab), tocilizumab, and vobarilizumab.

In some embodiments, methods of the present disclosure further comprise administration of one or more of BCD-089 (levilimab), tocilizumab, and vobarilizumab. In some embodiments, die disclosure provides methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), steps comprising: measuring at least one initial point of lung function in a subject; administering a TbKίί fusion protein to the subject; re -measuring the at least one point of lung function in the subject; and based on the measuring, determining a change in the rate of decline in lung function of the subject.

In some embodiments, the rate of decline in lung function is measured by Forced Vital Capacity (FVC) of the subject.

In some embodiments, the rate of decline in lung function is measured as an annual rate of decline in Forced Vital Capacity (FVC) of the subject. In some embodiments, the initial point of lung function comprises an FVC of greater than or equal to 50% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 100% and about 90% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 90% and about 80% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 80% and about 70% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 70% and about 60% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 60% and about 50% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 50% and about 40% of predicted. In some embodiments, the initial point of lung function comprises an FV C of between about 40% and about 30% of predicted. In some embodiments, the initial point of lung function comprises an FVC of between about 30% and about 20% of predicted.

In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 1% and about 10%. In some embodiments, an annual rate of decline m FVC of the subject is reduced by between about 5% and about 10%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10% and about 15%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 15% and about 20%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20% and about 25%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 25% and about 30%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30% and about 35%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 35% and about 40%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40% and about 45%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 45% and about 50%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50% and about 55%. in some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 55% and about 60%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60% and about 65%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 65% and about 70%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10% and about 20%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20% and about 30%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30% and about 40%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40% and about 50%. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50% and about 60%. In some embodiments, an annual rate of decline in F VC of the subject is reduced by between about 60% and about 70%.

In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 1 mL and about 10 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 5 mL and about 10 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10 mL and about 15 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 15 mL and about 20 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20 mL and about 25 mL. In some embodiments, an annual rate of decline in FVC of die subject is reduced by between about 25 mL and about 30 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30 mL and about 35 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 35 mL and about 40 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40 mL and about 45 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 45 mL and about 50 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50 mL and about 55 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 55 mL and about 60 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60 mL and about 65 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 65 mL and about 70 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 70 mL and about 75 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 75 mL and about 80 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 85 mL and about 90 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 90 mL and about 95 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 95 ml, and about 100 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by about 100 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 10 mL and about 20 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 20 mL and about 30 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 30 mL and about 40 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 40 mL and about 50 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 50 mL and about 60 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 60 mL and about 70 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 70 ml, and about 80 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 80 mL and about 90 mL. In some embodiments, an annual rate of decline in FVC of the subject is reduced by between about 90 mL and about 100 ml,. In some embodiments, an annual rate of decline in FVC of the subject is reduced by more than about 100 mL. In some embodiments, an annual rate of decline in forced vital capacity (FVC) of the subject is measured over a time period of at least one year after the administering and is relati ve to a subject treated with standard of care (SOC).

In some embodiments, the disclosure provides methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), comprising administering a Transforming Growth Facto r-b Receptor II ( ' TbKIί) antagonist to a subject in need thereof, wherein a dose of the TbKII antagonist comprises between about 0.75 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, the TbKII antagonist is administered in a dose of about 0.75 mg/kg once every two weeks to a subject in need thereof. In some embodiments, the TbKP antagonist is administered in a dose of about 2.25 mg/kg once every four weeks to a subject in need thereof. In some embodiments, the TpR!l antagonist is administered in a dose of about 1.5 mg/kg once every two weeks to a subject in need thereof. In some embodiments, the TpRH antagonist is administered in a dose of about 4.5 mg/kg once every four weeks to a subject in need thereof. In some embodiments, the TbIIII antagonist is administered in a dose of about 3.0 mg/kg once every two weeks to a subject in need thereof. In some embodiments, the TpRII antagonist is administered in a dose of about 6.0 mg/kg once every' four weeks to a subject in need thereof. In some embodiments, the TpRH antagonist is administered subcutaneously. In some embodiments, the TbίIP antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 10 and about 20 ug/mL in the subject. In some embodiments, the TpRH antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 30 and about 50 ug/mL in the subject.

In some embodiments of the present disclosure, the subject has been treated with one or more of abatacept, abituzumab, ajulemic acid, AVID200, AVID300, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-I01, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IBIO-CFB03, ifetroban, imatimb, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SAR156597, tacrolimus, tadalafil, and tocilizuinab.

In some embodiments, methods of the present disclosure further comprise administration of one or more of abatacept, abituzumab, ajulemic acid, AVID200, AVID300, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IB1Ό- CFB03, ifetroban, imatimb, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SAR156597, tacrolimus, tadalafil, and tocilizumab. In some embodiments, the subject has been treated with one or more of azathioprine, cyclophosphamide (CYC), mycophenolate mofetil (MMF), and nintedanib.

In some embodiments, methods of the present disclosure further comprise administration of one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and nintedanib. In some embodiments, methods of the present disclosure further comprise administration of one or more of cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), nintedanib, and rituximab.

In some embodiments, the disclosure provides methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TpRII) polypeptide to a subject in need thereof, wherein the method further comprises administration of one or more of IL-6 and/or IL-6R antibodies.

In some embodiments, IL-6 antibodies comprise one or more of clazakizumab, COR- 001, MEDI-5117, olokizumab, siltuximab, sirukumab, and ziltivekimab. In some embodiments, IL-6R antibodies comprise one or more of BCD-089 (levilimab), tocilizumab, and vobanlizumab.

In some embodiments, the subject has been treated with one or more of clazakizumab, COR-Q01, MEDI-5117, olokizumab, siltuximab, sirukumab, and ziltivekimab.

In some embodiments, methods of the present disclosure further comprise administration of one or more of clazakizumab, COR-001, MEDI-5117, olokizumab, siltuximab, sirukumab, and ziltivekimab.

In some embodiments, the subject has been treated with one or more of BCD-089 (levilimab), tocilizumab, and vobarilizumab.

In some embodiments, methods of the present disclosure further comprise administration of one or more of BCD-089 (levilimab), tocilizumab, and vobarilizumab.

In some embodiments, tire disclosure provides for a TpRII antagonist comprising a TpRII extracellular domain, wherein the TbKP extracellular domain comprises an amino acid sequence at least 80% identical to: i) a sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1 or ii) a sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2. In some embodiments, the TpRII extracellular domain comprises an amino acid sequence at least 90% identical to a sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1. In some embodiments, the TpRII extracellular domain comprises an amino acid sequence at least 95% identical to a sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1. In some embodiments, the TpRII extracellular domain comprises an amino acid sequence beginning at any of positions 23 to 35 of SEQ ID NO: 1 and ending at any of positions 153 to 159 of SEQ ID NO: 1. In some embodiments, the TpRII extracellular domain comprises an amino acid sequence at least 90% identical to a sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2. In some embodiments, the TpRII extracellular domain comprises an amino acid sequence at least 95% identical to a sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2. In some embodiments, tire TpRII extracellular domain comprises an amino acid sequence beginning at any of positions 23 to 60 of SEQ ID NO: 2 and ending at any of positions 178 to 184 of SEQ ID NO: 2. In some embodiments, the TbίIII extracellular domain comprises an amino acid sequence at least 90% identical to SEQ ID NO: 18. In some embodiments, the TbKII extracellular domain comprises an amino acid sequence at least 95% identical to SEQ ID NO: 18. In some embodiments, the TbKII extracellular domain comprises the amino acid sequence of SEQ ID NO: 18. in some embodiments, the ' TbKII polypeptide is a fusion protein further comprising a heterologous domain. In some embodiments, the heterologous domain comprises an immunoglobulin Fc domain. In some embodiments, the immunoglobulin Fc domain is a human immunoglobulin Fc domain. In some embodiments, the heterologous domain comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 20. In some embodiments, the heterologous domain comprises an amino acid sequence that is at least 95% identical to SEQ ID NO: 20. In some embodiments, the heterologous domain comprises the amino acid sequence of SEQ ID NO: 20. In some embodiments, the C-terminal lysine residue of the Fc domain can be deleted. Tire ammo acid sequence of SEQ ID NO: 20 may optionally be provided with the lysine removed from the ( ' -terminus (SEQ ID NO: 72). In some embodiments, the heterologous domain comprises an amino acid sequence that is at least 90% identical to SEQ ID NO: 72. In some embodiments, the heterologous domain comprises an amino acid sequence that is at least 95% identical to SEQ ID NO: 72. In some embodiments, the heterologous domain comprises the ammo acid sequence of SEQ ID NO: 72.

In some embodiments, the TbKII antagonist or fusion protein further comprises a linker. In some embodiments, the linker comprises (GGGGS)n, wherein n = > 4 (SEQ ID NO: 59). In some embodiments, the linker comprises the amino acid sequence of SEQ ID NO: 6.

In some embodiments, the TbKII antagonist or fusion protein does not include amino acids 185-592 of SEQ ID NO: 2. In some embodiments, the TbKII polypeptide or fusion protein does not include amino acids 1-22 of SEQ ID NO: 2,

In some embodiments, the fusion protein consists of or consists essentially of: a) a TjiR!I polypeptide portion comprising an amino acid sequence that is at least 85%, 90%,

95%, 97%, or 99% identical to the ammo acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids; b) a linker portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or 1 additional amino acids; c) a heterologous portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 20 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids; and d) optionally a leader sequence (e.g., SEQ ID NO: 23). In some embodiments, the fusion protein consists of or consists essentially of: a) a

TpRH polypeptide portion comprising an ammo acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids; b) a linker portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or 1 additional amino acids; c) a heterologous portion comprising an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 72 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids; and d) optionally a leader sequence (e.g., SEQ ID NO: 23). In some embodiments, the fusion protein consists of or consists essentially of: a) a

T|)RJ1 polypeptide portion comprising the amino acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids; b) a linker portion comprising the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or I additional amino acids; c) a heterologous portion comprising the amino acid sequence of SEQ ID NO: 20 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids; and d) optionally a leader sequence (e.g., SEQ ID NO: 23).

In some embodiments, the fusion protein consists of or consists essentially of: a) a T|)RH polypeptide portion comprising the amino acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional ammo acids; b) a linker portion comprising the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or 1 additional amino acids; c) a heterologous portion comprising the amino acid sequence of SEQ ID NO: 72 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids; and d) optionally a leader sequence (e.g., SEQ ID NO: 23).

In some embodiments, the fusion protein comprises: a) an extracellular domain of a T|)RH portion; wherein the extracellular domain comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the sequence of SEQ ID NO: 18; b) a heterologous portion, wherein the heterologous portion comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the sequence of SEQ ID NO: 20; and c) a linker portion connecting the extracellular domain and the heterologous portion; wherein the linker comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 6.

In some embodiments, the fusion protein comprises: a) an extracellular domain of a TpRff portion; wherein the extracellular domain comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the sequence of SEQ ID NO: 18; h) a heterologous portion, wherein the heterologous portion comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the sequence of SEQ ID NO: 72; and c) a linker portion connecting the extracellular domain and the heterologous portion; wherein tire linker comprises an amino acid sequence that is at least 85%, 90%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 6.

In some embodiments, the fusion protein comprises: a) an extracellular domain of a TpRH portion; wherein the extracellular domain comprises the amino acid sequence of SEQ ID NO: 18; b) a heterologous portion, wherein the heterologous portion comprises the amino acid sequence of SEQ ID NO: 20; and c) a linker portion connecting the extracellular domain and the heterologous portion; wherein the linker comprises the ammo acid sequence of SEQ ID NO: 6.

In some embodiments, the fusion protein comprises: a) an extracellular domain of a TpRff portion; wherein the extracellular domain comprises the amino acid sequence of SEQ ID NO: 18; b) a heterologous portion, wherein the heterologous portion comprises the amino acid sequence of SEQ ID NO: 72: and c) a linker portion connecting die extracellular domain and the heterologous portion; wherein the linker comprises the amino acid sequence of SEQ ID NO: 6.

In some embodiments, die TpRII antagonist comprises an amino acid sequence at least 90% identical to SEQ ID NO: 48. In some embodiments, the TpRII antagonist comprises an amino acid sequence at least 95% identical to SEQ ID NO: 48. In some embodiments, TpRII antagonist comprises the amino acid sequence of SEQ ID NO: 48. In some embodiments, the TpRII antagonist consists of the amino acid sequence of SEQ ID NO: 48.

In some embodiments, the TpRII antagonist comprises an amino acid sequence at least 90% identical to SEQ ID NO: 67. In some embodiments, the TbKII antagonist comprises an amino acid sequence at least 95% identical to SEQ ID NO: 67. In some embodiments, the TpRff antagonist comprises the amino acid sequence of SEQ ID NO: 67.

In some embodiments, the TbMI antagonist consists of the amino acid sequence of SEQ ID NO: 67 In some embodiments, the polypeptide includes one or more modified amino acid residues selected from: a glycosylated amino acid, a PEGylated amino acid, a farnesylated amino acid, an acetylated amino acid, a biotinylated amino acid, an amino acid conjugated to a lipid moiety, and an ammo acid conjugated to an organic derivatizing agent. In some embodiments, the polypeptide is glycosylated.

BRIEF DESCRIPTION OF THE DRAWINGS

Figure 1 shows the amino acid sequence of native precursor for the B (short) isoform of human TORb receptor type II (liTpRII) (NP 003233.4) (SEQ ID NO: 1). Solid underline indicates the mature extracellular domain (ECD) (residues 23-159), and double underline indicates valine that is replaced in the A (long) isoform. Dotted underline denotes leader (residues 1-22).

Figure 2 shows the amino acid sequence of native precursor for the A (long) isoform of human TfiRU (NP_001020018.1) (SEQ ID NO: 2). Solid underline indicates the mature ECD (residues 23-184), and double underline indicates the splice-generated isoleucine substitution. Dotted underline denotes leader (residues 1-22).

Figure 3 shows a comparison of the linker sequences of five different TbMII constructs (SEQ ID NOs 62-66, respectively, in order of appearance).

Figures 4A and 4B show in tabular form the binding affinity between TORbI and TORb3 and one of several different TbKII-Ro fusion protein constructs.

Figures 5A and SC graph the results from reporter gene assays testing the affinity ' of TORbI for one of several different TbKII-Fc fusion protein constructs. Figures 5B and 5D graph die results from reporter gene assays testing the affinity of the TGFp3 for one of several different TbKP-Ro fusion protein constructs, figures 5E and 5F provide ICso data from these same experiments in tabular form. figure 6 shows anti-fibrotic effects of mTpRII-mFc, a murine fusion protein that selectively binds to TGF-bI and TOR-b3, in a mouse model of SSc-ILD induced by bleomycin infusion. mTpRII-mFc is administered at different doses under different dosing schedules. Control mice were given subcutaneous injections of PBS (10 uL/g; “Control”) twice per week over a 28 day period. Bleomycin treated mice were given subcutaneous injections of either PBS (10 uL/g; “Vehicle”), Pan TGFpi-3 antibody' (10 mg/kg; “Pan TGFpi-3 Ah”), or a specified dose and frequency of mTpRII-mFc, Both the Vehicle and Pan TGFpl-3 Ab groups were administered PBS and Pan TGFpi-3 Ab, respectively, twice per week over a 28 day period Mice given mTpRII-mFc were subcutaneously injected with either 3 mg/kg mT[3Rli-mFc, 10 mg/kg mTflRII-niFc, or 100 mg/kg mTpRII-mFc on two different dosing schedules. One subset was dosed with hίϊ ' bKίί-ihRo once every two weeks over a 28 day period and a second group was dosed with mTpRII-mFc twice per week over a 28 day period. The two different dosing schedules are denoted by white bars {e.g., twice per week, on day 7, day 10, day 14, day 17, day 21, day 24, and day 28) or grey bars {e.g., once ever} 7 two weeks, on day 7 and day 21). Lung fibrosis was assessed by picrosirius red collagen staining with quantitative image analysis. Bleomycin vehicle-treated mice (“¥6111016”) showed an increasing trend in picrosirius red staining relative to saline-infused controls (“Control”). This increasing trend was significantly inhibited by ihTbKP-htTo at all doses and all dosing frequencies tested. Statistical analysis (One way ANOVA with Dunnett’s post hoc tests): * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001 vs “Vehicle”. DETAILED DESCRIPTION 1. Overview

Proteins described herein are the human forms, unless otherwise specified. NCBI references for the proteins are as follows: human TbKII isoform A (hTpRIIiong),

(NP .. 001020018.1) (SEQ ID NO: 2) and human TpRII isofonn B (hTpRIWt), (NP_003233,4) (SEQ ID NO: 1). Sequences of native TpRII proteins from human are set forth m Figures 1 and 2. In some embodiments, the TpRII proteins are from non-human animals, such as a mouse, rat, cow 7 or monkey.

The TGFP superfamily contains a variety of growth factors that share common sequence elements and structural motifs. These proteins are known to exert biological effects on a large variety of cell types in both vertebrates and invertebrates. Members of the superfamily perform important functions during embryonic development in pattern formation and tissue specification and can influence a variety of differentiation processes, including adipogenesis, myogenesis, chondrogenesis, cardiogenesis, hematopoiesis, neurogenesis, and epithelial cell differentiation. By manipulating the activity of a member of the TORb family, it is often possible to cause significant physiological changes in an organism. For example, the Piedmontese and Belgian Blue cattle breeds cany 7 a loss-of-function mutation in the GDF8 (also called myostatin) gene that causes a marked increase in muscle mass. Grobet et ah, Nat Genet. 1997, 17(l):71-4. Similarly, in humans, inactive alleles of GDF8 are associated with increased muscle mass and, reportedly, exceptional strength. Schuelke et al., N Engl J Med 2004, 350:2682-8.

TORb signals are mediated by heteromeric complexes of type I (e.g. TbMI) and type II (e.g, TbKII) serine/ threonine kinase receptors, which phosphorylate and activate downstream SMAD proteins upon ligand stimulation (Massague, 2000, Nat. Rev. Mol. Cell Biol. 1 : 169- 178). These type I and type II receptors are transmembrane proteins, composed of a ligand- binding extracellular domain with cysteine-rich region, a transmembrane domain, and a cytoplasmic domain with predicted serine/threonine specificity. Type I receptors are essential for signaling; and type II receptors are required for binding ligands and for expression of type I receptors. Type I and II receptors form a stable complex after ligand binding, resulting in phosphorylation of type I receptors by type II receptors. TORb has three mammalian isoforms, TGFpl, TORb2 and TίIRb3, each with distinct functions in vivo. The binding of TGFj!s to TbMI is a crucial step in initiating activation of the TGFj! signaling pathway, leading to phosphorylation of SMAD2, and translocation of the activated SMAD2/SMAD4 complex to the nucleus to modulate gene expression .

In part, the disclosure relates to TBRH antagonists that can be used to treat systemic sclerosis (SSc), particularly clinical complications of SSc including, for example, interstitial lung disease (ILD). In some embodiments, the disclosure provides TbKII polypeptides as antagonists of TGFpl or TORb3 for use in treating SSc or a complication of SSc (e.g., SSc- ILD). As described in greater detail below, the disclosure demonstrates that I]3RII-Fc fusion proteins comprising linkers of certain lengths (e.g., a linker having 21 amino acids) were surprisingly able to bind TORb-l and TORb-3 with stronger affinity than TpRlLFe fusion proteins having a linker of only four amino acids. While TpRH polypeptides, and fusion proteins comprising the same, may affect SSc, including complications of SSc (e.g., SSc- ILD), through a mechanism other that TbKII antagonism, the disclosure nonetheless provides that desirable therapeutic agents may be selected on tire basis of TpRIf antagonism.

Therefore, while not wishing to be bound to a particular mechanism of action, it is expected that other TbKIί antagonists may be useful in the treatment of SSc, particularly complications of SSc such as SSc-TLD. The terms used in this specification generally have their ordinary meanings in the art, within the context of this invention and in the specific context wliere each term is used. Certain terms are discussed below or elsewhere in the specification, to provide additional guidance to the practitioner in d escribing the compositions and methods of the inv ention and how to make and use them. The scope or meaning of any use of a term will be apparent from the specific context in which the term is used.

“Homologous,” in all its grammatical forms and spelling variations, refers to the relationship between two proteins that possess a “common evolutionary origin,”' including proteins from superfamilies in the same species of organism, its well as homologous proteins from different species of organism. Such proteins (and their encoding nucleic acids) have sequence homology, as reflected by their sequence similarity, whether in terms of percent identity or by the presence of specific residues or motifs and conserved positions.

The term “sequence similarity,” in all its grammatical forms, refers to tire degree of identity or correspondence between nucleic acid or amino acid sequences that may or may not share a common evolutionary ' origin. However, in common usage and in the instant application, the term “homologous,” when modified with an adverb such as “highly,” may refer to sequence similarity and may or may not relate to a common evolutionary origin.

“Percent (%) sequence identity” or “percent (%) identical” with respect to a reference polypeptide (or nucleotide) sequence is defined as the percentage of amino acid residues (or nucleic acids) in a candidate sequence that arc identical to the amino acid residues (or nucleic acids) in the reference polypeptide (nucleotide) sequence, after aligning the sequences and introducing gaps, if necessary', to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for aligning sequences, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For purposes herein, however, % amino acid (nucleic acid) sequence identity values are generated using the sequence comparison computer program ALiGN-2. The ALIGN -2 sequence comparison computer program was authored by Genentech, Inc., and the source code has been filed with user documentation in the U.S. Copyright Office,

Washington D.C., 20559, where it is registered under U.S, Copyright Registration No. TXU510087. The ALIGN-2 program is publicly available from Genentech, Inc., South San Francisco, Calif, or may be compiled from the source code. The ALIGN-2 program should be compiled for use on a UNIX operating system, including digital UNIX V4.0D. All sequence comparison parameters are set by the ALIGN-2 program and do not vary'. “Agonize"’, in all its grammatical forms, refers to the process of activating a protein and/or gene (e.g., by activating or amplifying that protein’s gene expression or by inducing an inactive protein to enter an active state) or increasing a protein’s and/or gene’s activity,

“Antagonize”, in all its grammatical forms, refers to the process of inhibiting a protein and/or gene (e.g., by inhibiting or decreasing that protein’s gene expression or by inducing an active protein to enter an inactive state) or decreasing a protein’s and/or gene’s activity 7 .

The terms "about" and "approximately" as used in connection with a numerical value throughout the specification and the claims denotes an interval of accuracy, familiar and acceptable to a person skilled in the art. Numeric ranges disclosed herein are inclusive of the numbers defining the ranges.

The terms "a" and "an" include plural referents unless the context in which the term is used clearly dictates otherwise. The terms "a" (or "an"), as well as the terms "one or more," and "at least one" can be used interchangeably herein. Furthermore, "and/or" where used herein is to be taken as specific disclosure of each of the two or more specified features or components with or without the other. Thus, the term “and/or" as used in a phrase such as "A and/or B" herein is intended to include "A and B," "A or B," "A" (alone), and "B" (alone). Likewise, the term "and/or" as used in a phrase such as "A, B, and/or C" is intended to encompass each of the following aspects: A, B, and C; A, B, or C; A or C; A or B; B or C; A and C; A and B; B and C; A (alone); B (alone); and C (alone). Throughout this specification, tire word “comprise” or variations such as “comprises” or “comprising” will be understood to imply the inclusion of a stated integer or groups of integers but not the exclusion of any other integer or group of integers. As used herein, the term “comprises” also encompasses the use of the narrower terms “consisting” and “consisting essentially of.” The term “consisting essentially of’ is limited to the specified materials or steps and those that do not materially affect the basic and novel characteristics of the invention(s) disclosed herein.

The term “appreciable affinity” as used herein means binding with a dissociation constant (Ko) of less than 50 nM. The terms "polypeptide", "oligopeptide", "peptide" and "protein" are used interchangeably herein to refer to chains of amino acids of any length. The chain may be linear or branched, it may comprise modified amino acids, and/or may be interrupted by nonamino acids. The terms also encompass an ammo acid chain that has been modified naturally or by intervention; for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification, such as conjugation with a labeling component. Also included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. It is understood that the polypeptides can occur as single chains or associated chains.

2. TBRII Antagonists

In part, the disclosure relates to TGFp type II receptor (TbKίί) polypeptides that can he used to treat systemic sclerosis (SSc), particularly clinical complications of SSc including, for example, interstitial lung disease (ILD). While TpRII polypeptides may affect SSc, including complications of SSc (e.g., SSc-iLD), through a mechanism other that inhibiting TpR!l activity, the disclosure nonetheless provides that desirable therapeutic agents may be selected on the basis of TpRII antagonism. Therefore, while not wishing to be bound to a particular mechanism of action, it is expected that additional TpRII antagonists may be useful in the treatment of SSc, particularly complications of SSc (e.g,, SSc-ILD). For example, agents that inhibit the activity and/or expression (e.g., transcription, translation, secretion from a cell, or combinations thereof) of one or more of: i) the TpRII receptor, ii) one or more TpRII-binding ligand (e.g., TGFpl, TGF(12, and/or TGF[33); iii) one or more TpRII- associated type I receptor (e.g., ALK5); iv) one or more TpRIl-associated co-receptor (e.g., betaglycan); and/or v) one or more TpRII downstream signaling component (e.g., Smad proteins), as well as combinations thereof, may be useful in the treatment of SSc, particularly complications of SSc (e.g., SSc-ILD). Such agents are collectively referred to herein as “TpRII antagonists” or “TpRII inhibitors”.

In certain aspects, a TpRII antagonist to be used in accordance with methods and uses described herein is an agent that inhibits activity and/or expression of at least TGFpl (e.g., a TGFpi antagonist). Effects on TGFpl inhibition may be determined, for example, using a cell-based assay including those described herein (e.g., Smad signaling assay). Therefore, in some embodiments, a TpRII antagonist of the disclosure may bind to at least TGFpi. Ligand binding activity may be determined, for example, using a binding affinity assay including those described herein. In some embodiments, a TpRII antagonist of the disclosure binds to at least TGFpi with a KD of at least 1 x 10 "7 M (e.g., at least 1 x 10 “K M, at least 1 x 10 "9 M, at least I x 10 'i0 M, at least 1 x 10 'n M, or at least 1 x 10 "12 M). As described herein, various TpRII antagonists that inhibit TGFpi can be used in accordance with the methods and uses

-2.9- described herein including, for example, ligand traps (e.g., TpRff or betaglycan polypeptides as well as variants thereof), antibodies, small molecules, nucleotide sequences, and combinations thereof. In certain embodiments, a TpRH antagonist that inhibits TORbI may further inhibit one or more of: TORb2, TORb3, TpRff. ALK5, and betaglycan. In some embodiments, a TpRII antagonist that inhibits TORbI further inhibits TGFp3. In some embodiments, a TfJRlI antagonist that inhibits TGFpl does not inhibit or does not substantially inhibit TORb2. In some embodiments, a ' TbKII antagonist that inhibits ' TORbI further inhibits TGFp3 but does not inhibit or does not substantially inhibit TORb2,

In certain aspects, a TbKII antagonist to be used in accordance with methods and uses described herein is an agent that inhibits activity and/or expression of at least TORb2 (e.g., a TGFp2 antagonist). Effects on TORb2 inhibition may be determined, for example, using a cell-based assay including those described herein (e.g., Smad signaling assay). Therefore, in some embodiments, a TbKII antagonist of the disclosure may bind to at least TORb2. Ligand binding activity' may be determined, for example, using a binding affinity' assay including those described herein. In some embodiments, a TbKII antagonist of the disclosure binds to at least TORb2 with a KD of at least 1 x It) "7 M (e.g., at least 1 x 10 ' * M, at least 1 x 10 "9 M, at least 1 x IQ '10 M, at least 1 x 1G 'U M, or at least 1 x 1() "]2 M). As described herein, various ' TbKP antagonists that inhibit TORb2 can be used in accordance with the methods and uses described herein including, for example, ligand traps (e.g., TbEII or betaglycan polypeptides as well as variants thereof), antibodies, small molecules, nucleotide sequences, and combinations thereof. In certain embodiments, a TbKII antagonist that inhibits T(}Rb2 may further inhibit one or more of: TORbI, TORb3, TpRff. ALK5, and betaglycan.

In certain aspects, a TfJRII antagonist to be used in accordance with methods and uses described herein is an agent that inhibits activity' and/or expression of at least TORb3 (e.g., a TGPP>3 antagonist). Effects on TORb3 inhibition may be determined, for example, using a cell-based assay including those described herein (e.g., Smad signaling assay). Therefore, in some embodiments, a TbKII antagonist of the disclosure may bind to at least TORb3. Ligand binding activity' may be determined, for example, using a binding affinity' assay including those described herein. In some embodiments, a TbMI antagonist of the disclosure binds to at least TORb3 with a KD of at least 1 x 10 "7 M (e.g., at least 1 x 10 "8 M, at least 1 x ICr 9 M, at least 1 x 10 ~i0 M, at least 1 x !O '11 M, or at least 1 x 10 "12 M). As described herein, various TbϊIII antagonists that inhibit TORb3 can be used in accordance with the methods and uses described herein including, for example, ligand traps (e.g., TpRff or betaglycan polypeptides as well as variants thereof), antibodies, small molecules, nucleotide sequences, and combinations thereof. In certain embodiments, a TpRII antagonist that inhibits TORb3 may further inhibit one or more of: TORbI, TORb2, TpRff. ALK5, and betaglycan. In some embodiments, a TpRII antagonist that inhibits TORb3 further inhibits TGFpI. In some embodiments, a TfJRlI antagonist that inhibits TGFp3 does not inhibit or does not substantially inhibit TGFp2, In some embodiments, a TpRII antagonist that inhibits TGFp3 further inhibits TGFpI but does not inhibit or does not substantially inhibit TORb2.

In certain aspects, a TpRII antagonist to be used in accordance with methods and uses described herein is an agent that inhibits activity and/or expression of at least a TpR 1 f receptor (e.g., a TpRff receptor antagonist). Effects on TpRff inhibition may be determined, for example, using a cell-based assay including those described herein (e.g., Smad signaling assay). Therefore, in some embodiments, a TbKII antagonist of the disclosure may bind to at least a TBRJf receptor. Ligand binding activity may be determined, for example, using a binding affinity assay including those described herein. In some embodiments, a TbKIΪ antagonist of the disclosure binds to at least a TpRff receptor with a KD of at least 1 x 10 '7 M (e.g., at least 1 x 10 “8 M, at least 1 x 1(T 9 M, at least 1 x 10 ‘to M, at least 1 x 10 "u M, or at least 1 x 10 'i2 M). As described herein, various TpRff antagonists that inhibit a TpRff receptor can be used in accordance with the methods and uses described herein including, for example, ligand traps (e.g,, TpRII or betaglycan polypeptides as well as variants thereof), antibodies, small molecules, nucleotide sequences, and combinations thereof. In certain embodiments, a TpRff antagonist that inhibits the TpRff receptor may further inhibit one or more of: TGFpI, TORb2, TORb3, ALK5, and betaglycan. In some embodiments, a TpRII antagonist that inhibits the TpRII receptor does not inhibit or does not substantially inhibit TGFp2.

In certain aspects, a TbKII antagonist to be used in accordance with methods and uses described herein is an agent that inhibits activity and/or expression of at least ALK5 (e.g., an ALK5 antagonist). Effects on ALK5 inhibition may be determined, for example, using a cell-based assay including those described herein (e.g,, Smad signaling assay). Therefore, in some embodiments, a TpRII antagonist of the disclosure may bind to at least ALK5. Ligand binding activity may be determined, for example, using a binding affinity assay including those described herein. In some embodiments, an ALK5 antagonist of the disclosure binds to at least ALK5 with a KD of at least 1 x 10 '7 M (e.g., at least 1 x IQ '8 M, at least 1 x 1Q ~9 M, at least 1 x 10 ' 10 M, at least 1 x 10 '11 M, or at least 1 x 10 '12 M). As described herein, various TbKII antagonists that inhibit ALK5 can be used in accordance with the methods and uses described herein including, for example, ligand traps (e.g., TbKII or betaglycan polypeptides as well as variants thereof), antibodies, small molecules, nucleotide sequences, and combinations thereof. In certain embodiments, a TbKIί antagonist that inhibits ALK5 may further inhibit one or more of: TORbI, TORb2, TORb3, TbKII, and betaglycan. In some embodiments, a TbKII antagonist that inhibits ALK5 does not inhibit or does not substantially inhibit TORb2. in certain aspects, a TbKII antagonist to be used in accordance with methods and uses described herein is an agent that inhibits activity and/or expression of at least betaglycan (e.g., a betaglycan antagonist). Effects on betaglycan inhibition may be determined, for example, using a cell-based assay including those described herein (e.g., Smad signaling assay). Therefore, in some embodiments, a TbKII antagonist of the disclosure may bind to at least betaglycan. Ligand binding activity may be determined, for example, using a binding affinity assay including those described herein. In some embodiments, a betaglycan antagonist of the disclosure binds to at least betaglycan with a KD of at least 1 x 10 "7 M (e.g., at least 1 x 10 '8 M, at least 1 x 10 '9 M, at least 1 x 10 '10 M, at least 1 x 1G ' !1 M, or at least 1 x 10 "12 M), As described herein, various TbMI antagonists that inhibit betaglycan can be used in accordance with the methods and uses described herein including, for example, ligand traps (e.g., TbKII or betaglycan polypeptides as well as variants thereof), antibodies, small molecules, nucleotide sequences, and combinations thereof. In certain embodiments, a TbKII antagonist that inhibits betaglycan may further inhibit one or more of: TORbI, TGI B2. TORb3, T|)RJL and ALK5. In some embodiments, a TbKII antagonist that inhibits betaglycan does not inhibit or does not substantially inhibit TORb2.

3. TBRIl Polypeptides

In certain aspects, a TpRIi antagonist to be used in accordance with the methods and uses disclosed herein is a TBRIf polypeptide. A TbKII polypeptide may inhibit and/or bind to, for example, one or more TbKII ligands (e.g., TORbI and/or ΊTcRb3). In some embodiments, the ability for a TbKII polypeptide to inhibit activity (e.g., Smad signaling) and/or bind to a target is determined in an in vitro or cell-based assay including, for example, those disclosed herein. As described herein, a TpRII polypeptide may be used alone or in combination with one or more additional active agents or supportive therapies to treat SSc or one or more complications of SSc (e.g,, SSc-ILD).

Naturally occurring TpRII proteins are transmembrane proteins, with a portion of the protein positioned outside the cell (the extracellular portion) and a portion of the protein positioned inside the cell (the intracellular portion). Aspects of the present disclosure encompass variant TpRII polypeptides comprising mutations within the extracellular domain and/or truncated portions of the extracellular domain of Tj lRH. As described above, human TbKII occurs naturally in at least two isoforms - A (long) and B (short) - generated by alternative splicing in the extracellular domain (BCD) (Figures 1 and 2 and SEQ ID NOS: 1 and 2). SEQ ID NO: 27, which corresponds to residues 23-159 of SEQ ID NO: 1, depicts the native full-length extracellular domain of the short isoform of TER! s. SEQ ID NO: 18, which corresponds to residues 23-184 of SEQ ID NO: 2, depicts the native full-length extracellular domain of the long isoform of TbKII, Unless noted otherwise, amino acid position numbering with regard to variants based on the TpRII short and long isoforms refers to the corresponding position in the native precursors, SEQ ID NO: 1 and SEQ ID NO: 2, respectively.

In certain embodiments, the disclosure provides variant TpRII polypeptides. A TpRII polypeptide of the disclosure may bind to and inhibit the function of a TGFp superfamily member, such as but not limited to, TORbI or TOEb3. TpRII polypeptides may include a polypeptide consisting of, or comprising, an amino acid sequence at least 80% identical, and optionally at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to a truncated ECD domain of a naturally occurring TpRII polypeptide, whose C- tenninus occurs at any of amino acids 153-159 of SEQ ID NO: 1. TbKIΪ polypeptides may include a polypeptide consisting of, or comprising, an amino acid sequence at least 80% identical, and optionally at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to a truncated PC D domain of a naturally occurring TbKII polypeptide, whose C-terminus occurs at any of amino acids 178-184 of SEQ ID NO: 2, In particular embodiments, the TpRII polypeptides comprise an amino acid sequence at least 80% identical, and optionally at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%,

98%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 18. Optionally, a TpRII polypeptide does not include more than 5 consecutive amino acids, or more than 10, 20, 30, 40, 50, 52, 60, 70, 80, 90, 100, 150 or 200 or more consecutive amino acids from a sequence consisting of amino acids 160-567 of SEQ ID NO: 1 or from a sequence consisting of amino acids 185-592 of SEQ ID NO: 2. In some embodiments, the TbKII polypeptide does not include amino acids 160-567 of SEQ ID NO: 1. In some embodiments, the ' TbKII polypeptide does not include amino acids 1-22 of SEQ ID NO: 1. In some embodiments, the TbίIII polypeptide does not include amino acids 1-22 and 160-567 of SEQ ID NO: 1. In some embodiments, the TfJRII polypeptide does not include amino acids 185-592 of SEQ ID NO: 2, In some embodiments, the TbKIΪ polypeptide does not include amino acids 1-22 of SEQ ID NO: 2. In some embodiments, the TbKII polypeptide does not include amino acids 1-22 and 185-592 of SEQ ID NO: 2. The unprocessed TbKII polypeptide may either include or exclude any signal sequence, as well as any sequence N-terminal to the signal sequence.

As elaborated herein, the N-terminus of the mature (processed) TbKII polypeptide may occur at any of amino acids 23-35 of SEQ ID NO: 1 or 23-60 of SEQ ID NO: 2. Examples of mature TbίIII polypeptides include, but are not limited to, amino acids 23-159 of SEQ ID NO: 1 (set forth in SEQ ID NO: 27), amino acids 29-159 of SEQ ID NO: 1 (set forth in SEQ ID NO: 28), amino acids 35-159 of SEQ ID NO: 1 (set forth in SEQ ID NO: 29), amino acids 23-153 of SEQ ID NO: 1 (set forth in SEQ ID NO: 30), amino acids 29-153 of SEQ ID NO:

1 (set forth in SEQ ID NO: 3 i), amino acids 35-153 of SEQ ID NO: 1 (set forth in SEQ ID NO: 32), amino acids 23-184 of SEQ ID NO: 2 (set forth in SEQ ID NO: 18), amino acids 29-184 of SEQ ID NO: 2 (set forth in SEQ ID NO: 33), ammo acids 60-184 of SEQ ID NO: 2 (set forth in SEQ ID NO: 29), ammo acids 23-178 of SEQ ID NO: 2 (set forth in SEQ ID

NO: 34), amino acids 29-178 of SEQ ID NO: 2 (set forth in SEQ ID NO: 35), and amino acids 60-178 of SEQ ID NO: 2 (set forth in SEQ ID NO: 32). It will be understood by one of skill in the art that corresponding variants based on the long isofomi of TbKIί will include nucleotide sequences encoding the 25-ammo acid insertion along with a conservative Vai-Ile substitution at the flanking position C-terminal to the insertion . The TbKII polypeptides accordingly may include isolated extracellular portions of TpRII polypeptides, including both the short and the long isoforms, variants thereof (including variants that comprise, for example, no more than 2, 3, 4, 5, 10, 15, 20, 25, 30, or 35 amino acid substitutions in the sequence corresponding to amino acids 23-159 of SEQ ID NO: 1 or amino acids 23-184 of SEQ ID NO: 2), fragments thereof, and fusion proteins comprising any of the foregoing, but in each case preferably any of the foregoing TbίIII polypeptides will retain substantial affinity for at least one of, or both of, TORbI or TORb3. Generally, a TbKII polypeptide will be designed to be soluble in aqueous solutions at biologically relevant temperatures, pH levels, and osmolality. In some embodiments, the variant TbKII polypeptides of the disclosure comprise one or more mutations in the extracellular domain that confer an altered ligand binding profile. A TbKII polypeptide may include one, two, five or more alterations in the amino acid sequence relative to the corresponding portion of a naturally occurring TbKII polypeptide. In some embodiments, the mutation results in a substitution, insertion, or deletion at the position corresponding to position 70 of SEQ ID NO: 1. In some embodiments, the mutation results in a substitution, insertion, or deletion at the position corresponding to position 110 of SEQ ID NO: 1. Examples include, but are not limited to, an N to D substitution or a D to K substitution in the positions corresponding to positions 70 and 110, respectively, of SEQ ID NO: 1. Examples of such variant TbKP polypeptides include, but are not limited to, the sequences set forth in SEQ ID NOs: 36-39. A TbKII polypeptide may comprise a polypeptide or portion thereof that is encoded by any one of SEQ ID NOs: 10, 12, 14 or 16, or silent variants thereof or nucleic acids that hybridize to the complement thereof under stringent hybridization conditions. In particular embodiments, a TbKII polypeptide may comprise a polypeptide or portion thereof that is encoded by any one of SEQ ID NO: 12, or silent variants thereof or nucleic acids that hybridize to the complement thereof under stringent hybridization conditions.

In some embodiments, the variant TbKII polypeptides of the disclosure further comprise an insertion of 36 amino acids (SEQ ID NO: 41) between the pair of glutamate residues (positions 151 and 152 of SEQ ID NO: 1, or positions 176 and 177 of SEQ ID NO: 2) located near the C-terminus of the human TbKίί ECD, as occurs naturally in the human TjlRII isoform C (Konrad et ai., BMC Genomics 8:318, 2007).

The disclosure further demonstrates that TbKII polypeptides can be modified to selectively antagonize TfJRII ligands. The N70 residue represents a potential glycosylation site. In some embodiments, the TbKIΐ polypeptides are agiycosylated. In some embodiments, the TbKII polypeptides are agiycosylated or have reduced glycosylation at position Asnl57. In some embodiments, the TbKII polypeptides are agiycosylated or have reduced glycosylation at position Asn73.

In certain embodiments, a TjlRII polypeptide binds to TGFpl, and the TbKII polypeptide does not show substantial binding to TOEb3. In certain embodiments, a TpRII polypeptide binds to TGFp3, and the TpRII polypeptide does not show substantial binding to TGFpl. Binding may be assessed using purified proteins in solution or in a surface plasmon resonance system, such as a Biacore™ system. In certain embodiments, a TpRII polypeptide inhibits TG Gb I cellular signaling, and the TjtRH polypeptide has an intermediate or limited inhibitory effect on TGFfB signaling.

In certain embodiments, a TbKΪI polypeptide inhibits TGFp3 cellular signaling, and the TpRff polypeptide has an intennediate or limited inhibitory effect on TGFpi signaling. Inhibitory 7 effect on cell signaling can be assayed by methods known in the art.

Taken together, an active portion of a TpRff polypeptide may comprise amino acid residues 23-153, 23-154, 23-155, 23-156, 23-157, or 23-158 of SEQ ID NO: 1, as well as variants of these amino acid residues starting at any of amino acids 24-35 of SEQ ID NO: 1. Similarly, an active portion of a TpRff polypeptide may comprise amino acid residues 23- 178, 23-179, 23-180, 23-181, 23-182, or 23-183 of SEQ ID NO: 2, as well as variants of these ammo acid residues starting at any of ammo acids 24-60 of SEQ ID NO: 2, Exemplary TpRH polypeptides comprise amino acid residues 29-159, 35-159, 23-153, 29-153 and 35- 153 of SEQ ID NO: 1 or ammo acid residues 29-184, 60-184, 23-178, 29-178 and 60-178 of SEQ ID NO: 2. Variants within these ranges are also contemplated, particularly those having at least 80%, 85%, 90%, 95%, or 99% identity to the corresponding portion of SEQ ID NO: 1 or SEQ ID NO: 2. A TpRII polypeptide may be selected that does not include the sequence consisting of amino acid residues 160-567 of SEIQ ID NO: 1 or amino acid residues 185-592 of SEQ ID NO: 2. In particular embodiments, the TbKII polypeptides comprise an amino acid sequence at least 80% identical, and optionally at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the amino acid sequence of SEQ ID

NO: 18.

As described above, the disclosure provides TpRII polypeptides sharing a specified degree of sequence identity or similarity to a naturally occurring TbKII polypeptide. To determine the percent identity' of two amino acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). The amino acid residues at corresponding amino acid positions are then compared. When a position in the first sequence is occupied by the same amino acid residue as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid

"identity" is equivalent to amino acid "homology"). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity and similarity between two sequences can be accomplished using a mathematical algorithm (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D, W., ed,, Academic Press, New Y ork, 1993; Computer Analysis of Sequence Data, Part 1, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991),

In one embodiment, the percent identity between two amino acid sequences is determined using the Needleman and Wimscli (J Mol, Biol. (48):444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com). In a specific embodiment, the following parameters are used in the GAP program: either a Blosum 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4 and a length weight of 1, 2, 3, 4, 5, or 6, In yet another embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (Devereux, J., etal, Nucleic Acids Res. 12(1):387 (1984)) (available at http://www.gcg.com). Exemplary parameters include using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80 and a length weight of 1, 2, 3, 4, 5, or 6, Unless otherwise specified, percent identity between two amino acid sequences is to be determined using the GAP program using a Blosum 62 matrix, a GAP weight of 10 and a length weight of 3, and if such algorithm cannot compute the desired percent identity, a suitable alternative disclosed herein should be selected.

In another embodiment, the percent identity between two amino acid sequences is determined using the algorithm of E. Myers and W. Miller (CABIQS, 4: 11-17 (1989)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.

Another embodiment for determining the best overall alignment between two ammo acid sequences can be determined using the FASTDB computer program based on the algorithm of Brutlag etal. (Comp. App. Biosci., 6:237-245 (1990)). In a sequence alignment the query and subject sequences are both amino acid sequences. The result of said global sequence alignment is presented in terms of percent identity. In one embodiment, amino acid sequence identity is performed using the FASTDB computer program based on the algorithm of Brut! ag et al. (Comp. App. Biosci., 6:237-245 (1990)). In a specific embodiment, parameters employed to calculate percent identity and similarity of an amino acid alignment comprise: Matrix=PAM 150, k-tuple : =2, Mismatch Penalty-!, Joining Penalty-20, Randomization Group Length=0, Cutoff Score=T, Gap Penalty=5 and Gap Size Penalty=0.05.

TbKII polypeptides may additionally include any of various leader sequences at the N-terminus. Such a sequence would allow the peptides to be expressed and targeted to the secretion pathway in a eukaryotic system. See, e.g., Ernst et al., U.S. Pat. No. 5,082,783 (1992). Alternatively, a native TpRII signal sequence may he used to effect extrusion from the cell. Possible leader sequences include native leaders, tissue plasminogen activator (TPA) and honeybee meliitin (SEQ ID NOs: 22-24, respectively). Examples ofTpRH-Fc fusion proteins incorporating a TPA leader sequence include SEQ ID NOs: 11, 13, 15, 17, 68, 69, 70, and 71. Processing of signal peptides may vary ' depending on the leader sequence chosen, the cell type used and culture conditions, among other variables, and therefore actual N-terminal start sites for mature TbKII polypeptides may shift by 1, 2, 3, 4 or 5 ammo acids in either the N-terminal or C-terminal direction. Examples of T]3RII-Fc fusion proteins include SEQ ID NOs: 11, 13, 15, 17, 68, 69, 70, and 71 . It will be understood by one of skill in the art that corresponding variants based on the long isoform of TpRII will include the 25- amino acid insertion along with a conservative Val-Ile substitution at the flanking position C- terminal to the insertion.

In some embodiments, any of the TpRII polypeptides disclosed herein are at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, 99% or 100% identical to the amino acid sequence of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49 51, or 67, but lack one or more N-terminal amino acids as compared to the amino acid sequences of SEQ ID NO: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49 51, 67, or 78. In some embodiments, the TbKII polypeptide lacks the ammo acid corresponding to the first amino acid (threonine) of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49, 51, 67, or 78. In some embodiments, the TpRII polypeptide lacks the amino acids corresponding to the first and second amino acids (threonine and isoleucine, respecti vely) of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49, 51, 67, or 78. In some embodiments, the TpRII polypeptide lacks the amino acids corresponding to the first, second and third ammo acids (threonine, isoleucine, and proline, respectively) of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49,

51, 67, or 78. In some embodiments, the TpRII polypeptide lacks the amino acids corresponding to the first, second, third and fourth amino acids (threonine, isoleucine, proline, proline, respectively) of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49, 51, 67, or 78. In some embodiments, any of the TiiRH polypeptides disclosed herein are at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, 99% or 100% identical to the ammo acid sequence of any one of SEQ ID NOs: 18, 51, or 78, but lack the amino acid corresponding to the first amino acid (threonine) of SEQ ID NO: 18, 51 , or 78, In some embodiments, the TflRH polypeptide lacks the amino acids corresponding to the first and second amino acids !threonine and isoleucine, respectively) of SEQ ID NO: 18, 51, or 78. In some embodiments, the TBR I f polypeptide lacks the amino acids corresponding to the first, second and third amino acids (threonine, isoleucine, and proline, respectively) of SEQ ID NO: 18, 51, or 78.

In some embodiments, the TfiRII polypeptide lacks the amino acids corresponding to the first, second, third and fourth amino acids (threonine, isoleucine, proline, proline, respectively) of SEQ ID NO: 18, 51, or 78.

In some embodiments, the disclosure provides for a composition comprising a mixture of TbίIII polypeptides, wherein the TflRII polypeptides in the composition each comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, 99% or 100% identical to the amino acid sequence of any one of SEQ ID NOs:

18, 27, 30, 34, 36, 37, 38, 39, 48, 49, 51, 67, or 78; but wherein at least a portion of the TbίIII polypeptides (e.g., at least 1%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%) m the composition include the amino acids corresponding to the first, second, third and fourth amino acids (threonine, isoleucine, proline and proline, respectively) of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49,

51 , 67, or 78; and wherein at least a portion of the TbKίί polypeptides (e.g. , at least 1 %, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%) in the composition lack one or more of the amino acids corresponding to the first, second, third and fourth ammo acids (threonine, isoleucine, proline and proline, respectively) of any one of SEQ ID NOs: 18, 27, 30, 34, 36, 37, 38, 39, 48, 49, 51, 67, or 78, In some embodiments, the disclosure provides for a composition comprising a mixture of TbKP polypeptides, wherein the TbίIII polypeptides are at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, 99% or 100% identical to the amino acid sequence of any one of SEQ ID NOs: 18, 51 , or 78, but wherein at least 30% to 80% of the TbKII polypeptides in the composition lack the amino acid corresponding to the first amino acid (threonine) of SEQ ID NO: i8, 51, or 78.

In certain embodiments, the present disclosure contemplates specific mutations of the TpR!l polypeptides so as to alter the glycosylation of the polypeptide. Such mutations may be selected so as to introduce or eliminate one or more glycosylation sites, such as Q-iinked orN-linked glycosylation sites. Asparagine -linked glycosylation recognition sites generally comprise a tripeptide sequence, asparagine-X-threonine (or asparagine-X-serine) (where “X” is any amino acid) which is specifically recognized by appropriate cellular glycosylation enzymes. The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the wild-type TbKII polypeptide (for O-linked glycosylation sites). A variety of amino acid substitutions or deletions at one or both of the first or third amino acid positions of a glycosylation recognition site (and/or amino acid deletion at the second position) results in non-glycosylation at the modified tripeptide sequence. Another means of increasing the number of carbohydrate moieties on a TbKP polypeptide is by chemical or enzymatic coupling of glycosides to the TjlRil polypeptide. Depending on the coupling mode used, the sugar(s) may be attached to (a) arginine and histidine; (b) free carboxyl groups; (c) free sulfhydryl groups such as those of cysteine; (d) free hydroxy l groups such as those of serine, threonine, or hydroxyproline; (e) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan; or (t) the amide group of glutamine. These methods are described in WO 87/05330 published Sep. 11, 1987, and in Aplin and Wriston (1981) CRC Crit. Rev. Biochem., pp. 259-306, incorporated by reference herein. Removal of one or more carbohydrate moieties present on a TbKII polypeptide may be accomplished chemically and/or enzymatically. Chemical deg!ycosylation may involve, for example, exposure of the TbίIII polypeptide to the compound trifluoromethanesulfonic acid, or an equivalent compound. This treatment results in the cleavage of most or ail sugars except the linking sugar (N-acetylglueosamine or N-acetylgalactosamine), while leaving the amino acid sequence intact. Chemical deglycosylation is further described by Hakim uddin et al. (1987) Arch. Biochem. Biophys. 259:52 and by Edge et al. (1981) Anal. Biochem. 118:131. Enzymatic cleavage of carbohydrate moieties on TbίIII polypeptides can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al. (1987) Meth. Enzyrnol. 138:350. The sequence of a TjsR!l polypeptide may be adjusted, as appropriate, depending on the ty pe of expression sy stem used, as mammalian, yeast, insect and plant cells may all introduce differing glycosylation patterns that can be affected by the amino acid sequence of the peptide. In general, TbKII polypeptides for use in humans will be expressed in a mammalian cell line that provides proper glycosylation, such as HEK293 or CHO ceil lines, although other mammalian expression cell lines, yeast cell lines with engineered glycosylation enzymes, and insect cells are expected to be useful as well.

This disclosure further contemplates a method of generating mutants, particularly sets of combinatorial mutants of a TbKP polypeptide, as well as truncation mutants; pools of combinatorial mutants arc especially useful for identifying functional variant sequences. The purpose of screening such combinatorial libraries may be to generate, for example, TbίIII polypeptide variants which can act as either agonists or antagonist, or alternatively, which possess novel activities all together. A variety of screening assays are provided below, and such assays may be used to evaluate variants. For example, a TiiRl! polypeptide variant may be screened for ability to bind to a TbKII ligand, to prevent binding of a TbKP ligand to a TjlR!l polypeptide or to interfere with signaling caused by a ' TbKII ligand. The activity of a TjlRII polypeptide or its variants may also be tested in a cell-based or in vivo assay, particularly any of the assays disclosed in the Examples. Combinatorially-derived variants can be generated which have a selective or generally increased potency relative to a TtyRII polypeptide comprising an extracellular domain of a naturally occurring TbKP polypeptide. Likewise, mutagenesis can give rise to variants which have serum half-lives dramatically different than the corresponding wild-type TjlR!! polypeptide. For example, the altered protein can be rendered either more stable or less stable to proteolytic degradation or other processes which result in destruction of, or otherwise elimination or inactivation of, a native TbKII polypeptide. Such variants, and the genes which encode them, can be utilized to alter TbKII polypeptide levels by modulating the half-life of the TpRII polypeptides. For instance, a short, half-life can give rise to more transient biological effects and can allow tighter control of recombinant Tf!RII polypeptide levels within the patient. In an Fc fusion protein, mutations may be made in the linker (if any) and/or the Fc portion to alter the half-life of the protein.

A combinatorial library' may be produced by way of a degenerate library' of genes encoding a library of polypeptides which each include at least a portion of potential TbMί polypeptide sequences. For instance, a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential TbKII polypeptide nucleotide sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display ).

There are many ways by which the library ' of potential TjlR!l polypeptide variants can be generated from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic genes then be ligated into an appropriate vector for expression. The synthesis of degenerate oligonucleotides is well known in the art (see for example, Narang, SA (1983) Tetrahedron 39:3; Itakura et a!,, (198!) Recombinant DNA, Proc. 3rd Cleveland Sympos. Macromolecules, ed. AG Walton, Amsterdam: Elsevier pp273-289: Itakura et ah, (1984) Anmi Rev. Biochern. 53:323; Itakura et al., (1984) Science 198:1056; Ike el al., (1983) Nucleic Acid Res. 11:477). Such techniques have been employed in the directed evolution of other proteins (see, for example, Scott et al., (1990) Science 249:386-390; Roberts et al., (1992) PNAS USA 89:2429-2433; Devlin et al., (1990) Science 249: 404-406; Cwirla et al,, (1990) PNAS USA 87: 6378-6382; as well as U.S. Patent Nos: 5,223,409, 5,198,346, and

5,096,815).

Alternatively, other forms of mutagenesis can be utilized to generate a combinatorial library'. For example, TbKII polypeptide variants can be generated and isolated from a library by screening using, for example, alanine scanning mutagenesis and the like (Ruf et al, (1994) Biochemistry- 33: 1565-1572; Wang et al, (1994) J. Biol. ( ' hem. 269:3095-3099; Balint et al, (1993) Gene 137: 109-118; Grodberg et al, (1993) Eur. I, Biochem. 218:597-601 ; Nagashima et al, (1993) I. Biol. Chem. 268:2888-2892; Low man et al, (1991) Biochemistry 30: 10832- 10838; and Cunningham et al, (1989) Science 244:1081-1085), by linker scanning mutagenesis (Gustin et al, (1993) Virology 193:653-660; Brown et al, (1992) Mol. Cell Biol. 12:2644-2652; McKnight et al, (1982) Science 232:316); by saturation mutagenesis (Meyers et al, (1986) Science 232:613); by PCR mutagenesis (Leung et al, (1989) Method Cell Mol Biol 1: 11-19); or by random mutagenesis, including chemical mutagenesis, etc. (Miller et al, (1992) A Short Course in Bacterial Genetics, CSHL Press, Cold Spring Harbor, NY; and Greener et al, (1994) Strategies in Mol Biol 7:32-34). Linker scanning mutagenesis, particularly in a combinatorial setting, is an attractive method for identify ing truncated (bioactive) forms of ' TbKII polypeptides.

A wide range of techniques are known in the art for screening gene products of combinatorial libraries made by point mutations and truncations, and, for that matter, for screening cDNA libraries for gene products having a certain property. Such techniques will be generally adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of TbKίί polypeptides. Hie most widely used techniques for screening large gene libraries typically comprises cloning tire gene library into replicable expression vectors, transforming appropri ate cells with the resulting library' of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates relatively easy isolation of the vector encoding the gene whose product was detected. Preferred assays include TpRH ligand binding assays and ligand-mediated cell signaling assays.

In certain embodiments, the Tb R 11 polypeptides of the disclosure may further comprise post-translational modifications in addition to any that are naturally present in the TflRH polypeptides. Such modifications include, but are not limited to, acetylation, carboxylation, giycosylation, phosphorylation, lipidation, pegylation (polyethylene glycol) and acylation. As a result, the modified TbKII polypeptides may contain non-amino acid elements, such as polyethylene glycols, lipids, mono- or poly-saccharides, and phosphates. Effects of such non-amino acid elements on the functionality of a TflRH polypeptide may be tested as described herein for other TbίIII polypeptide variants. When a TfJRll polypeptide is produced in cells by cleaving a nascent form of the TbKIί polypeptide, post-translational processing may also be important for correct folding and/or function of the protein. Different ceils (such as CHQ, HeLa, MDCK, 293, W138, NIH-3T3 or HEK-293) have specific cellular machine!·}' and characteristic mechanisms for such post-translational activities and may be chosen to ensure the correct modification and processing of the TbKII polypeptides,

4. Linkers

The disclosure provides for TbKIί fusion proteins, and in these embodiments, the TjlRII portion is connected to the heterologous portion (e.g. , Fc portion) by means of a linker. In some embodiments, the linkers are glycine and serine rich linkers. Oilier near neutral amino acids, such as, but not limited to, Thr, Asn, Pro and Ala, may also be used m the linker sequence. In some embodiments, the linker comprises various permutations of ammo acid sequences containing Gly and Ser. In some embodiments, the linker is greater than 10 amino acids in length. In further embodiments, the linkers have a length of at least 12, 15, 20, 21, 25, 30, 35, 40, 45 or 50 amino acids. In some embodiments, the linker is less than 40, 35, 30, 25, 22 or 20 amino acids. In some embodiments, the linker is 10-50, 10-40, 10-30, 10-25, 10- 21, 10-15, 10, 15-25, 17-22, 20, or 21 amino acids in length. In preferred embodiments, the linker comprises the amino acid sequence GlyGlyGiyGlySer (GGGGS) (SEQ ID NO: 19), or repetitions thereof (GGGGS)n, where n > 2 (SEQ ID NO: 57). In particular embodiments n > 3, or n = 3-10. The application teaches the surprising finding that proteins comprising a TpRll portion and a heterologous portion fused together by means of a (GGGGS)4 linker (SEQ ID NO: 59) were associated with a stronger affinity for TΌEbI and TOEb3 as compared to a TbKII fusion protein where n < 4. As such, in preferred embodiments, n > 4, or n = 4-10. The application also teaches that proteins comprising (GGGGS)n linkers

( ' GGGGS’ disclosed as SEQ ID NO: 19) in which n > 4 had similar inhibitory properties as proteins having the (GGGGSfi linker (SEQ ID NO: 59). As such, in some embodiments, n is not greater than 4 in a (GGGGS)n linker (SEQ ID NO: 19). In some embodiments, n = 4-10, 4-9, 4-8, 4-7, 4-6, 4-5, 5-8, 5-7, or 5-6. In some embodiments, n :=: 3, 4, 5, 6, or 7. In particular embodiments, n = 4. In some embodiments, a linker comprising a (GGGGS)n sequence (SEQ ID NO: 19) also comprises an N “terminal threonine. In some embodiments, the linker is any one of the following:

GGGGSGGGGS (SEQ ID NO: 21) TGGGGSGGGGS (SEQ ID NO: 4)

TGGGGSGGGGSGGGGS (SEQ ID NO: 5)

TGGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 6) TGGGGSGGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 25) TGGGGSGGGGSGGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 26) or TGGGGSGGGG SGGGGSGGGGSGGGGSGGGGSGGGGS (SEQ ID NO : 40) .

In some embodiments, the linker comprises the amino acid sequence of TGGGPKSCDK (SEQ ID NO: 7). In some embodiments, the linker is any one of SEQ ID NOs: 21, 4-7, 25-26 or 40 lacking the N-temnnal threonine. In some embodiments, the linker does not comprise the amino acid sequence of SEQ ID NO: 26 or 40.

5. Heterologous Domains

In certain aspects, functional variants or modified forms of the TjfRli polypeptides include fusion proteins having at least a portion of the TBRli polypeptides and one or more heterologous portions. Well-known examples of such heterologous portions include, but are not limited to, polyhistidme, Glu-Glu, glutathione S transferase (GST), thioredoxm, protein A, protein G, an immunoglobulin heavy chain constant region (Fc), maltose binding protein (MBP), or human serum albumin. A heterologous portion may be selected so as to confer a desired property. For example, some heterologous portions are particularly useful for isolation of the fusion proteins by affinity chromatography. For tire purpose of affinity purification, relevant matrices for affinity chromatography, such as glutathione-, amylase-, and nickel- or cobalt-conjugated resins are used. Many of such matrices are available in “kit” form, such as the Pharmacia GST purification system and the QIAexpress™ sy stem (Qiagen) useful with (HISe (SEQ ID NO: 61)) fusion partners. As another example, a heterologous portion may be selected so as to facili tate detection of the TbKII polypeptides. Examples of such detection domains include the various fluorescent proteins (e.g., GFP) as well as

“epitope tags,” which are usually short peptide sequences for which a specific antibody is available. Well known epitope tags for which specific monoclonal antibodies are readily available include FLAG, influenza vinis haemagglutinin (HA), and c-myc tags. In some cases, the heterologous portions have a protease cleavage site, such as for Factor Xa or Thrombin, which allows the relevant protease to partially digest the fusion proteins and thereby liberate the recombinant proteins therefrom. The liberated proteins can then be isolated from the heterologous portion by subsequent chromatographic separation. In certain preferred embodiments, a TpRff polypeptide is fused with a domain that stabilizes the TbKII polypeptide in vivo (a “stabilizer” domain). By “stabilizing” is meant anything that increases serum half life, regardless of whether this is because of decreased destruction, decreased clearance by the kidney, or other pharmacokinetic effect. Fusions with the Fc portion of an immunoglobulin are known to confer desirable pharmacokinetic properties on a wide range of proteins. Likewise, fusions to human serum albumin can confer desirable properties. Other types of heterologous portions that may he selected include multimerizing (e.g., dimerizing, tetramerizing) domains and functional domains.

As specific examples, the present disclosure provides fusion proteins comprising variants of TbKII polypeptides fused to an Fc domain sequence of SEQ ID NO: 20. Optionally, the Fc domain has one or more mutations at residues such as Asp-265, Lys-322, and Asn-434 (numbered in accordance with the corresponding full-length IgG). In certain cases, the mutant Fc domain having one or more of these mutations (e.g., Asp-265 mutation) has reduced ability of binding to the Fey receptor relative to a wildtype Fc domain. In other cases, the mutant Fc domain having one or more of these mutations (e.g., Asn-434 mutation) has increased ability of binding to the MHC class T-related Fc-receptor (FcRN) relative to awildtype Fc domain. In some embodiments, the C-terminal lysine residue of the Fc domain can be deleted. The ammo acid sequence of SEQ ID NO: 20 may optionally be provided with the lysine removed from the C-terminus (SEQ ID NO: 72).

It is understood that different elements of the fusion proteins may be arranged in any manner that is consistent with the desired functionality. For example, a TbKII polypeptide may be placed C-terminal to a heterologous domain, or, alternatively, a heterologous domain may be placed C-terminal to a TbKIΪ polypeptide. The TbKίί polypeptide domain and the heterologous domain need not be adjacent in a fusion protein, and additional domains or amino acid sequences may be included C- or N-terminal to either domain or between the domains. As used herein, the term "immunoglobulin Fc domain" or simply “Fc” is understood to mean the carboxyl-terminal portion of an immunoglobulin chain constant region, preferably an immunoglobulin heavy chain constant region, or a portion thereof. For example, an immunoglobulin Fc region may comprise 1) a CHI domain, a CH2 domain, and a CH3 domain, 2) a CHI domain and a CH2 domain, 3) a CHI domain and a CH3 domain, 4) a CH2 domain and a CHS domain, or 5 ) a combination of two or more domains and an immunoglobulin hinge region. In a preferred embodiment the immunoglobulin Fc region comprises at least an immunoglobulin hinge region a CH2 domain and a CHS domain, and preferably lacks the CHI domain. In some embodiments, the immunoglobulin Fc region is a human immunoglobulin Fc region.

In one embodiment, the class of immunoglobulin from which the heavy chain constant region is derived is IgG (Igy) (g subclasses 1, 2, 3, or 4). Other classes of immunoglobulin, IgA (Iga), IgD (Igb), IgE (Igs) and IgM (Igp), may be used. The choice of appropriate immunoglobulin heavy chain constant region is discussed in detail in U.S. Pat. Nos. 5,541,087 and 5,726,044. The choice of particular immunoglobulin heavy chain constant region sequences from certain immunoglobulin classes and subclasses to achieve a particular result is considered to be within the level of skill in the art. Hie portion of the DMA construct encoding the immunoglobulin Fc region preferably comprises at least a portion of a hinge domain, and preferably at least a portion of a CHb domain of Fc gamma or the homologous domains in any of IgA, IgD, IgE, or IgM.

Furthermore, it is contemplated that substitution or deletion of amino acids within the immunoglobulin heavy chain constant regions may be useful m the practice of the methods and compositions disclosed herein. One example would be to introduce amino acid substitutions in the upper CH2 region to create an Fc variant with reduced affinity for Fc receptors (Cole et al. (1997) J. Immunol. 159:3613).

Antibodies and Fc fusion proteins with reduced effector function may be produced by introducing changes in the amino acid sequence, including, but are not limited to, the Ala-Ala mutation described by Bluestone et al. (see WO 94/28027 and WO 98/47531; also see Xu et al. 2000 Cell Immunol 200; 16-26). Thus, in certain embodiments, Fc fusion proteins of the disclosure with mutations within the constant region including the Ala-Ala mutation may be used to reduce or abolish effector function. According to these embodiments, antibodies and Fc fusion proteins may comprise a mutation to an alanine at position 234 or a mutation to an alanine at position 235, or a combination thereof. In one embodiment, the antibody or Fc fusion protein comprises an IgG 4 framework, wherein the Ala-Ala mutation would describe a mutation(s) from phenylalanine to alanine at position 234 and/or a mutation from leucine to alanine at position 235. In another embodiment, the antibody or Fc fusion protein comprises an IgG! framework, wherein the Ala-Ala mutation would describe a mutation(s) from leucine to alanine at position 234 and/or a mutation from leucine to alanine at position 235. While alanine substitutions at these sites are effective in reducing ADCC in both human and murine antibodies, these substitutions are less effective at reducing CDC activity. Another single variant P329A, identified by a random mutagenesis approach to map the Clq binding site of the Fc, is highly effective at reducing CDC activity while retaining ADCC activity. A combination of L234A, L235A, and P329A (LALA-PG, Kabat positions) substitutions have been shown to effectively silence the effector function of human IgGl antibodies. For a detailed discussion ofLALA, LALA-PG, and other mutations, see Lo et al. (2017) 1 Biol. Chem. 292:3900-3908, the contents of which are hereby incorporated herein by reference in their entirety ' . In some embodiments, Fc fusion proteins of the disclosure comprise L234A, L235 A, and P329G mutations (LALA-PG; Kabat positions) in the Fc region of the heavy chain. The antibody or Fc fusion protein may alternatively or additionally carry other mutations, including the point mutation K322A in the CH2 domain (Hezareh et ai. 2001 J Virol. 75: 12161-8). In particular embodiments, the antibody or Fc fusion protein may be modified to either enhance or inhibit complement dependent cytotoxicity (CDC). Modulated CDC activity' may be achieved by introducing one or more amino acid substitutions, insertions, or deletions in an Fc region (see, e.g., U.S. Pat. No. 6,194,551). Alternatively, or additionally, cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody or Fc fusion protein thus generated may have improved or reduced internalization capability and/or increased or decreased complement-mediated cell killing. See Caron et al., I. Exp Med. 176:1191-1195 (1992) and Shopes, B. J. Immunol. 148:2918-2922 (1992), W099/51642, Duncan & Winter Nature 322: 738-40 (1988); U.S. Pat No. 5,648,260; U.S. Pat. No. 5,624,821; and W094/29351. In some embodiments, the disclosure provides for TbKII polypeptides fusion proteins comprising an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 11, 13, 15, 17, 68, 69, 70, and 71, or biologically active fragments thereof. In some embodiments, the TbKIί polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs: 11, 13, 15, 17, 68, 69, 70, and 71 or biologically active fragments thereof. In some embodiments, the C-temiinal lysine residue of an Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 11 may optionally' be provided with the lysine removed from the C-terminus (SEQ ID NO: 68). The amino acid sequence of SEQ ID NO: 13 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 69). The amino acid sequence of SEQ ID NO: 15 may optionally be provided with the lysine removed from the C-terminus (SEQ ID NO: 70). The amino acid sequence of SEQ ID NO: 17 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 71).

In some embodiments, the ' TbKII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 13, or a biologically active fragment thereof. In some embodiments, the ' TbKII polypeptides fusion proteins comprise an ammo acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%,

95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 69, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 50, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion proteins compri se an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 77, or a biologically active fragment thereof. In some embodiments, the TbίIP polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 51, or a biologically active fragment thereof. In some embodiments, the TjsRJI polypeptides fusion proteins comprise an ammo acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID

NO: 78, or a biologically active fragment thereof In some embodiments, the TbKII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%,

90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 52, or a biologically active fragment thereof. In some embodiments, the TfyRII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 79, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion proteins comprise an ammo acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 53, or a biologically active fragment thereof. In some embodiments, the TjlRll polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 80, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%,

90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 54, or a biologically active fragment thereof. In some embodiments, the TbKIί polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 81, or a biologically active fragment thereof. In some embodiments, the TfiRII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 55, or a biologically active fragment thereof. In some embodiments, the TpRII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 82, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the ammo acid sequence of any one of SEQ ID NO: 56, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 83, or a biologically active fragment thereof. In some embodiments, the TbKII polypeptides fusion protein comprises an ammo acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of SEQ ID NO: 20, or a biologically active fragment thereof. In some embodiments, the TbKIΪ polypeptides fusion proteins comprise an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NO: 72, or a biologically active fragment thereof.

In some embodiments, the fusion proteins described herein have improved binding affinity for TOEbί and TOEb3. In some embodiments, a fusion protein comprising a linker at least 10 ammo acids m length (e.g., a fusion protein having the ammo acid sequence of any one of SEQ ID NOs: 11, 13, 15, 50-56, 68-70, and 77-83) has improved binding affinity for TGFpl and TGFfl3 as compared to a reference fusion protein (e.g., a fusion protein having the amino acid sequence of SEQ ID NO: 9 or SEQ ID NO: 88), In some embodiments, the fusion protein binds to TGFpl with a KD of less than 200 pM, less than 150 pM, less than 100 pM, less than 75 pM, less than 50 pM or less than 25 pM. In some embodiments, the fusion protein binds to TGFfB with a KD of less than 75 pM, less than 70 pM, less than 60 pM, less than 50 pM, less than 40 pM, less than 35 pM, less than 25 pM, less than 15, less than 10, or less than 5 pM.

In some embodiments any of the polypeptides disclosed herein inhibits TORbI and/or TGFp3 in a measurable assay. In some embodiments, the polypeptide inhibits TGF ' pi with an ICso of less than 1.0, 0.9, 0.8. 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.08, 0.09, 0.07, 0,06, 0.05, 0.04, 0.03, or 0.02 nM, as determined using a reporter gene assay. In some embodiments, the polypeptide inhibits TGFfB with an ICso of less than 1.0, 0.9, 0.8. 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.09, 0.08, 0.07, 0.06, 0.05, 0.04, 0.03, or 0.02 nM, as determined using a reporter gene assay. In some embodiments, the reporter gene assay is a CAGA reporter assay. In some embodiments, the CAGA assay is based on a human lung carcinoma cell line transfected with a pGL3(CAGA)12 reporter plasmid (Dennler et al, 1998, EMBO 17: 3091-3100) as well as a Renilla reporter plasmid (pRLCMV) to control for transfection efficiency. The CAGA motif is present in tire promoters of TGFp-responsive genes (for example, PAI-1), so this vector is of general use for factors signaling through SMAD2 and SMAD3. See, e.g.. Example 2.

6, Fusion Polypeptides

In some embodiments, die disclosure provides for TfIRII-containing fusion polypeptides. The fusion polypeptides may be prepared according to any of die methods disclosed herein or that are known in the art.

In some embodiments, any of the fusion polypeptides disclosed herein comprises the following components: a) any of the TbIIII polypeptides disclosed herein (“A”), b) any of die linkers disclosed herein (“B”), c) any of the heterologous portions disclosed herein (“C”), and optionally a linker (“X”). In such embodiments, the fusion polypeptide may be arranged in a manner as follows (N-terminus to C-terminus): A-B-C or C-B-A. In such embodiments, the fusion polypeptide may be arranged in a manner as follow's (N-terminus to C-terminus): X-A- B-C or X-C-B-A. In some embodiments, the fusion polypeptide comprises each of A, B and C (and optionally a leader sequence such as the amino acid sequence of SEQ ID NO: 23), and comprises no more than 100, 90, 80, 70, 60, 50, 40, 30, 20, 10, 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGyiation).

In some embodiments, the fusion polypeptide comprises a leader sequence (e.g,, SEQ ID NO: 23) positioned in a manner as follows (N-terminus to C-terminus): X-A-B-C, and the fusion polypeptide comprises 1, 2, 3, 4, or 5 amino acids between X and A. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g, SEQ ID NO: 23) positioned in a manner as follows (N-terminus to C-terminus): X-C-B-A, and the fusion polypeptide comprises 1, 2, 3, 4, or 5 amino acids between X and C. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g., SEQ ID NO: 23) positioned in a manner as follows (N-terminus to C-terminus): X-A-B-C, and the fusion polypeptide comprises an alanine between X and A. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g., SEQ ID NO: 23) positioned in a manner as follows (N- terminus to C-terminus): X-C-B-A, and the fusion polypeptide comprises an alanine between X and C. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g., SEQ ID NO: 23) positioned in a manner as follows (N-terminus to C-terminus): X-A-B-C, and the fusion polypeptide comprises a glycine and an alanine between X and A. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g., SEQ ID NO: 23) positioned in a manner as follows (N-terminus to C-terminus): X-C-B-A, and the fusion polypeptide comprises a glycine and an alanine between X and C. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g., SEQ ID NO: 23) positioned m a manner as follows (N-terminus to C-temiinus): X-A-B-C, and the fusion polypeptide comprises a threonine between X and A. In some embodiments, the fusion polypeptide comprises a leader sequence (e.g., SEQ ID NO: 23) positioned in a manner as follows (N- terminus to C-terminus): X-C-B-A, and the fusion polypeptide comprises a threonine between X and C.

In some embodiments, the fusion polypeptide comprises an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to any of the TbKP polypeptide amino acid sequences disclosed herein (e.g., SEQ ID NO: 18), -wherein the ' TbKII polypeptide portion of the fusion polypeptide comprises no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGyiation). In some embodiments, the fusion polypeptide comprises an ammo acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to any of the linker sequences disclosed herein (e.g., SEQ ID NO: 6), wherein the linker portion of the fusion polypeptide comprises no more than 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation). In some embodiments, the fusion polypeptide comprises an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to any of the heterologous portion sequences disclosed herein (e.g., SEQ ID NO: 20), wherein the heterologous portion of the fusion polypeptide comprises no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation).

In some embodiments, the fusion polypeptide comprises any of the ' TbKII polypeptide amino acid sequences disclosed herein (e.g., SEQ ID NO: 18), wherein the TbKII polypeptide portion of the fusion polypeptide comprises no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation). In some embodiments, the fusion polypeptide comprises any of the linker sequences disclosed herein (e.g., SEQ ID NO: 6), wherein the linker portion of the fusion polypeptide comprises no more than 5, 4, 3, 2 or 1 additional ammo acids (but which may include further post-translational modifications, such as PEGylation). In some embodiments, the fusion polypeptide comprises any of the heterologous portion sequences disclosed herein (e.g., SEQ ID NO: 20 or 72), wherein the heterologous portion of the fusion polypeptide comprises no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation).

In some embodiments, the disclosure provides for a fusion polypeptide, wherein the fusion polypeptide consists or consists essentially of (and not necessarily in the following order): a) an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%,

97%, or 99% identical to any of the TbKII polypeptide ammo acid sequences disclosed herein (e.g., SEQ ID NO: 18), wherein the TbKII polypeptide portion of the fusion polypeptide comprises no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or I additional amino acids (but which may include further post-translational modifications, such as PEGylation); b) an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to any of the linker sequences disclosed herein (e.g., SEQ ID NO: 6), wherein the linker portion of the fusion polypeptide comprises no more than 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and c) an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to any of the heterologous portion sequences disclosed herein (e.g., SEQ ID NO: 20 or 72), wherein the heterologous portion of the fusion polypeptide comprises no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids (but which may include further post- translational modifications, such as PEGylation); and d) optionally a leader sequence (e.g., SEQ ID NO: 23). In some embodiments, the disclosure provides for a fusion polypeptide, wherein the fusion polypeptide consists or consists essentially of (and not necessarily in the following order): a) any of the THRU polypeptide amino acid sequences disclosed herein (e.g., SEQ ID NO: 18), wherein the TbMί polypeptide portion of the fusion polypeptide comprises no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); b) any of the linker sequences disclosed herein (e.g,, SEQ ID NO: 6), wherein the linker portion of the fusion polypeptide comprises no more than 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and c) any of the heterologous portion sequences disclosed herein (e.g. , SEQ ID NO: 20 or 72), wherein the heterologous portion of the fusion polypeptide comprises no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and d) optionally a leader sequence (e.g., SEQ ID NO: 23). In some embodiments, the disclosure provides for a fusion polypeptide consisting of or consisting essentially of (and not necessarily in the following order): a) a TfiRH polypeptide portion consisting of an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); b) a linker portion consisting of an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 97%, or 99% identical to the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and c) a heterologous portion consisting of an amino acid sequence that is at least 85%, 90%, 91 %, 92%, 93%, 94%, 95%, 97%, or 99% identical to the ammo acid sequence of SEQ ID NO: 20 or SEQ ID NO: 72 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and d) optionally a leader sequence (e.g., SEQ ID NO: 23). In some embodiments, the disclosure provides for a fusion polypeptide consisting or consisting essentially of (and not necessarily in the following order): a) a TbKII polypeptide portion consisting of the ammo acid sequence of SEQ ID NO: 18 and no more than 10, 9, 8, 7, 6, 5,

4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); b) a linker portion consisting of the amino acid sequence of SEQ ID NO: 6 and no more than 5, 4, 3, 2 or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and c) a heterologous portion consisting of the ammo acid sequence of SEQ ID NO: 20 or SEQ ID NO: 72 and no more than 25, 20, 15, 10, 5, 4, 3, 2, or 1 additional amino acids (but which may include further post-translational modifications, such as PEGylation); and d) optionally a leader sequence (e.g., SEQ ID NO: 23).

In some embodiments, the fusion protein does not comprise a leader sequence. In some embodiments, the fusion protein comprises an amino acid sequence that is at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 99%, or 100% identical to the amino acid sequence of SEQ ID NO: 48.

TIPPHVQKSDVEMEAQKDEIICPSCNRTAHPLRHINNDMIVTDNNGAVKFPQLCKFC DVRFSTCDNQK SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCIMKE KKKPGETF FMCSCSSDECNDNIIFSEEYNTSNPDTGGGGSGGGGSGGGGSGGGGSTHTCPPCPAPELL GGPSVFLF PPKPKDTLMISRTPEVTCVW DVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRW SVLTVLHQ DWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKG FYPSDIAV EWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQK SLSLSPGK (SEQ ID NO: 48).

In some embodiments, the C -terminal lysine residue of the Fc domain can be deleted.

In some embodiments, the amino add sequence of SEQ ID NO: 48 may optionally be provided with the lysine removed from the C -terminus (SEQ ID NO: 67):

TIPPHVQKSDVEMEAQKDEIICPSCNRTAHPLRHINNDMIVTDNNGAVKFPQLCKFC DVRFSTCDNQK

SCMSNCSITSICEKPQEVCVAVWRKNDENITLETVCHDPKLPYHDFILEDAASPKCI MKEKKKPGETF

FMCSCSSDECNDNIIFSEEYNTSNPDTGGGGSGGGGSGGGGSGGGGSTHTCPPCPAP ELLGGPSVFLF

PPKPKDTLMISRTPEVTCVWDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYR W SVLTVLHQ

DWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKKQVSLTCL VKGFYPSDIAV

EWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHY TQKSLSLSPG

(SEQ ID NO: (57).

In some embodiments, the disclosure provides for a TbKIί fusion polypeptide wherein the polypeptide does not comprise an antibody or antigen-binding portion thereof. In some embodiments, the polypeptide does not bind with appreciable affinity to a cytokine other than a transforming growth factor beta superfamily ligand (e,g., TORbI, TOEb2 and/or ΊΌRb3),

In some embodiments, the polypeptide does not bind with appreciable affinity' to a cytokine other than TORbI, TOEb2 and/or T(3Rb3. In some embodiments, the polypeptide does not bind with appreciable affinity to a cytokine other than TGFpl and/or TGFp3. In some embodiments, the polypeptide does not hind with appreciable affinity to CD4, CD8, CD25, CTLA-4, TL-10, TORb Receptor, PD- 1, PD-L1, PD-L2, RANK, RANKL, HER2/neu,

EGFR1, CD20, VEGF, TNF-a, TNFR2, FoxP3, CD80, CD86, IFN-a, IFN-b, IFN-g, GITR, 4-1BB, OX-40, TLRl-10, ErbB-1, HER1, ErbB-3/HER3, ErbB-4/HER4, IGFR, IGFBP, IGF- 1R, PDGFR, FGFR, VEGFR, HGFR, TRK receptor, ephrin receptors, AXL receptors, LTK receptors, TIE receptors, angiopoietinl, 2, ROR receptor, DDR receptor, RET receptor, KLG receptor, RYK receptor, MuSK receptor, II,bK, IlaR, TNTRSF, TRAIL receptor, ARTC1, alpha-actinin-4, Bcr-abl, B-RAF, caspases, beta-catenin, fihronectin, GPNMB, GDP-L, LDLR, I II.L-L2. MLA-A 11 , HSP70, Kϊ AA205 , MART2, MUM- 1, 2, 3, PAP, neo-PAP,

NFYC, OGT, OS-9, pml-RARalpha fusion protein, PRDX5, PTPRK, KRAS2, NRAS,

HRAS, RBAF600, SIRT2. SNRPDl, SYT-SSX1 or -SSX2 fusion protein, Triosephosphate Isomerase, BAGE, BAGE-1 . BAGE-2, 3, 4, 5, GAGE-1, 2, 3, 4, 5, 6, 7, 8, GnT-V, HERV-K MEL, KK-LC, KM-HN-1, LAGE, LAGE-1, CAMEL, MAGE-1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-AS, MAGE-A6, MAGE-A8, MAGE-A9, MAGE-A 10 , MAGE-A 11 ,

MAGE-A12, MAGE-3, MAGE-B 1 , MAGE-B2, MAGE-B5 , MAGE-B6, MAGE-C1, MAGE-C2, rnucin 1 (MUC1), MART - 1 /Melan- A (MLANA), gplOO, gp 100/Pme 117 (SILV), tyrosinase (TYR), TRP-1, HAGE, NA-88, NY-ESO-1, NY-ESO-l/LAGE-2, SAGE, Spl7. SSX-1, 2, 3, 4, TRP2-1NT2, carcino-embryonic antigen (CEA), Kallikfein 4, mammaglobm-A, OAI, prostate specific antigen (PSA), prostate specific membrane antigen, TRP-1/, 75. TRP-2, AIM-2. BING-4, CPSF, cyelin Dl, Ep-CAM, EpbA3, FGF-5, gp250, iCE), AFP, M-CSF, mdm-2, MUCI, p53 (TP53), PBF, FRAME, PSMA, RAGE-1. RNF43, RU2AS, SOX 10, STEAP1, survivin (BIRCS), hTERT, telomerase, WT1, SYCP1, BRDT, SPANX, XAGE, ADAM2, PAGE-5, LIP1, CTAGE-1, CSAGE, MMAl, CAGE, BORIS, HOM-TES-85, AF15q 14, HCA66I, LDHC, MORC, SGY-1, SPOl 1, TPX1, NY-SAR-35,

FTHLI7, NXF2 TDRD1, TEX 15, FATE, TPTE, estrogen receptors (UR), androgen receptors (AR), CD40, CD30, CD20, CD 19, CD33, CD4, CD25, CD3, CA 72-4, CA 15-3, CA 27-29, CA 125, CA 19-9, beta-human chorionic gonadotropin, 1-2 microglobulin, squamous cell carcinoma antigen, neuron-specific enoJase, heat shock protein gp96, GM2, sargramostim, CTLA-4, 707-AP, ART-4, CAP-1, CLCA2, Cyp-B, HST-2, HPV proteins, EBV proteins. Hepatitis B or C vims proteins, and/or HIV proteins.

In some embodiments, the disclosure provides for a TbKII fusion polypeptide wherein the polypeptide does not comprise an additional ligand binding domain in addition to the TjlRII domain. In some embodiments, the polypeptide comprises a linear amino acid sequence comprising a TbRP domain and a heterologous portion (e.g. , an Fc portion), but the linear amino acid sequence does not comprise any additional ligand binding domains. In some embodiments, the polypeptide comprises a linear amino acid sequence comprising a TpRII domain and an Fc portion, but the linear amino acid sequence does not comprise any additional ligand binding domains. In some embodiments, the disclosure provides for a I|)RJI fusion polypeptide wherein the polypeptide does not comprise multiple ligand binding domains in a single linear ammo acid sequence. In some embodiments, the disclosure provides for a TbKίI fusion polypeptide wherein the polypeptide does not comprise more than one continuous linker sequence in a single linear amino acid sequence. In some embodiments, the polypeptide does not comprise multiple continuous glycine and/or serine linkers {e.g., a linker comprising (GGGGS)n, wherein n = > 4 (SEQ ID NO: 59)) in a single linear amino acid sequence. In some embodiments, the disclosure provides for a ' TbKII fusion polypeptide wherein the heterologous portion is an Fc domain, and wherein only one continuous linker is covalently bound to the Fc domain. In some embodiments, the only one continuous linker comprises or consists of a (GGGGS)n linker, wherein n = > 4 (SEQ ID NO: 59).

7. Nucleic Adds and Methods of Manufacture

In certain embodiments, the present disclosure makes available isolated and/or purified forms of the TbKII polypeptides fusion proteins, which are isolated from, or otherwise substantially free of (e.g., at least 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% free of), other proteins and/or other TbίIII polypeptide species. TbKII polypeptides will generally be produced by expression from recombinant nucleic acids.

In certain embodiments, the disclosure includes nucleic acids encoding soluble TbMI polypeptides comprising the coding sequence for an extracellular portion of a TbίIII protein. In further embodiments, this disclosure also pertains to a host ceil comprising such nucleic acids. The host cell may be any prokaryotic or eukaryotic cell. For example, a polypeptide of the present disclosure may be expressed in bacterial cells such as E. coli, insect cells (e.g., using a baculovirus expression system), yeast, or mammalian cells. Other suitable host ceils are known to those skilled in the art. Accordingly, some embodiments of the present disclosure further pertain to methods of producing the TbKIί polypeptides.

In certain aspects, the disclosure provides isolated and/or recombinant nucleic acids encoding any of the TbKII polypeptides, including fragments, functional vari ants and fusion proteins disclosed herein. SEQ ID NOs: 10, 12 and 14 encode variants of TbKIί extracellular domain fused to an IgG Fc domain. The subject nucleic acids may be single-stranded or double stranded. Such nucleic acids may be DNA or RNA molecules. These nucleic acids may be used, for example, in methods for making TbKTί polypeptides or as direct therapeutic agents {e.g,, in an antisense, RNAi or gene therapy approach). In certain aspects, the subject nucleic acids encoding TbKII polypeptides are further understood to include nucleic acids that are variants of SEQ ID NOs: 10, 12 and 14. Variant nucleotide sequences include sequences that differ by one or more nucleotide substitutions, additions or deletions, such as allelic variants, In certain embodiments, the disclosure provides isolated or recombinant nucleic acid sequences that are at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NOs: 10, 12 and 14. In particular embodiments, the disclosure provides isolated or recombinant nucleic acid sequences that are at least 80%,

85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to SEQ ID NO: 12, or fragments thereof. One of ordinary skill in the art will appreciate that nucleic acid sequences complementary to SEQ ID NOs: 10, 12 and 14, and variants of SEQ ID NOs: 10, 12 and 14 are also within the scope of this disclosure. In further embodiments, the nucleic acid sequences of the disclosure can be isolated, recombinant, and/or fused with a heterologous nucleotide sequence, or in a DNA library. In other embodiments, nucleic acids of the disclosure also include nucleotide sequences that hybridize under highly stringent conditions to the nucleotide sequences designated in SEQ ID NOs: 10, 12 and 14 complement sequences of SEQ ID NOs: 10, 12 and 14, or fragments thereof. As discussed above, one of ordinary skill in the art will understand readily that appropriate stringency conditions which promote DNA hybridization can be varied. For example, one could perform the hybridization at 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50°C to a high stringency of about 0.2 x SSC at 50°C. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions at about 65°C. Both temperature and salt may be varied, or temperature or salt concentration may be held constant while the oilier variable is changed. In some embodiments, the disclosure provides nucleic acids which hybridize under low stringency conditions of 6 x SSC at room temperature followed by a wash at 2 x SSC at room temperature, Isolated nucleic acids which differ from the nucleic acids as set forth in SEQ ID NOs:

10, 12 and 14 due to degeneracy in the genetic code are also within the scope of the disclosure. For example, a number of ammo acids are designated by more than one triplet. Codons that specify the same amino acid, or synonyms (for example, CAU and CAC are synonyms for histidine) may result in “silent"’ mutations which do not affect the amino acid sequence of the protein. However, it is expected that DNA sequence polymorphisms that do lead to changes in the amino acid sequences of the subject proteins will exist among mammalian cells. One skilled in the art will appreciate that these variations in one or more nucleotides (up to about 3-5% of the nucleotides) of the nucleic acids encoding a particular protein may exist among individuals of a given species due to natural allelic variation. Any and all such nucleotide variations and resulting amino acid polymorphisms are within the scope of this disclosure.

It will be appreciated by one of skill in the art that corresponding variants based on the long isoform of ' TbKII will include nucleotide sequences encoding the 25-amino acid insertion along with a conservative Val-Ile substitution at the flanking position C-terminal to the insertion. It will also be appreciated that corresponding variants based on either the long (A) or short (B) isoforms of TbKII will include variant nucleotide sequences comprising an insertion of 108 nucleotides, encoding a 36-amino-acid insertion (SEQ ID NO: 41), at the same location described for naturally occurring TbKII isofomi C.

In certain embodiments, the recombinant nucleic acids of the disclosure may be operabiy linked to one or more regulatory nucleotide sequences in an expression construct. Regulator} ' nucleotide sequences will generally be appropriate to the host cell used for expression. Numerous types of appropriate expression vectors and suitable regulator}' sequences are known in the art for a variety of host cells. Typically, said one or more regulatory nucleotide sequences may include, but are not limited to, promoter sequences, leader or signal sequences, ribosomal binding sites, transcriptional start and termination sequences, translational start and termination sequences, and enhancer or activator sequences. Constitutive or inducible promoters as known in the art are contemplated by the disclosure. Tire promoters may be either naturally occurring promoters, or hybrid promoters that combine elements of more than one promoter. An expression construct may be present in a cell on an episome, such as a plasmid, or the expression construct may be inserted in a chromosome. In a preferred embodiment, the expression vector contains a selectable marker gene to allow' the selection of transformed host cells. Selectable marker genes are well known in the art and will vary' with the host cell used.

In certain aspects disclosed herein, the subject nucleic acid is provided in an expression vector comprising a nucleotide sequence encoding a TbEIί polypeptide and operabiy linked to at least one regulatory sequence. Regulatory' sequences are art-recognized and are selected to direct expression of the TbKII polypeptide. Accordingly, the term regulatory sequence includes promoters, enhancers, and other expression control elements. Exemplary regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enzyrnology, Academic Press, San Diego, CA (1990). For instance, any of a wide variety of expression control sequences that control the expression of a DNA sequence when operatively linked to it rnay be used in these vectors to express DNA sequences encoding a TpRll polypeptide. Such useful expression control sequences, include, for example, the early and late promoters of SV40, tet promoter, adenovirus or cytomegalovirus immediate early promoter, RSV promoters, the lac system, the trp system, the TAG or TRC system, T7 promoter whose expression is directed by T7 RNA polymerase, the major operator and promoter regions of phage lambda , the control regions for fd coat protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., Pho5, the promoters of the yeast a-mating factors, the polyhedron promoter of the baculovirus system and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof. It should be understood that the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed. Moreover, the vector's copy number, the ability to control that copy number and the expression of any other protein encoded by the vector, such as antibiotic markers, should also be considered. A recombinant nucleic acid included in the disclosure can be produced by ligating the cloned gene, or a portion thereof, into a vector suitable for expression in either prokaryotic cells, eukaryotic cells (yeast, avian, insect or mammalian), or both. Expression vehicles for production of a recombinant Tp-RH polypeptide include plasmids and other vectors. For instance, suitable vectors include plasmids of the types: pBR322-derived plasmids, pEMBL- derived plasmids, pEX-derived plasmids, pBTac -derived plasmids and pUC-derived plasmids for expression in prokaryotic cells, such as E. coll.

Some mammalian expression vectors contain both prokaryotic sequences to facilitate the propagation of the vector in bacteria, and one or more eukaryotic transcription units that are expressed in eukaryotic cells. The pcDNAI/amp, pcDNAI/neo, pRc/CMV, pSV2gpt, pSV2neo, pSV2-dhfr, pTk2, pRSYneo, pMSG, pSVT7, pko-neo and pHyg derived vectors are examples of mammalian expression vectors suitable for transfection of eukaryotic cells. Some of these vectors are modified with sequences from bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells. Alternatively, derivatives of viruses such as the bovine papilloma virus (BPV-1), or Epstein- Barr vims (pHEBo, pREP -derived and p205) can be used for transient expression of proteins in eukaryotic cells. Examples of other viral (including retroviral) expression systems can be found below in the description of gene therapy delivery systems. The various methods employed in the preparation of the plasmids and in transformation of host organisms are well known in die art. For other suitable expression systems for both prokaiyotic and eukaryotic cells, as well as general recombinant procedures, see Molecular Cloning A Laboratory Manual, 3rd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory' Press, 2001). In some instances, it may be desirable to express the recombinant polypeptides by the use of a bacu!ovirus expression system. Examples of such baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941), pAcUW-derived vectors (such as pAcUWl), and pBlueBac -derived vectors (such as the B-gal containing pBlueBac III).

In certain embodiments, a vector will be designed for production of the subject TbKίί polypeptides in CHO cells, such as a Pcmv-Script vector (Stratagene, La Jolla, Calif), pcDN4 vectors (Invitrogen, Carlsbad, Calif.) and pCI-neo vectors (Promega, Madison,

Wise.). In a preferred embodiment, a vector will be designed for production of the subject TbKίϊ polypeptides in HEK-293 cells. As will be apparent, the subject gene constructs can be used to cause expression of the subject TbKP polypeptides in cells propagated in culture, e.g., to produce proteins, including fusion proteins or variant proteins, for purification. This disclosure also pertains to a host cell transfected with a recombinant gene including a coding sequence (e.g., SEQ ID NOs: 10, 12, or 14) for one or more of the subject TbKP polypeptides. The host cell may be any prokaiyotic or eukaryotic cell. For example, a TpRII polypeptide disclosed herein may be expressed in bacterial cells such as E. coll, insect ceils (e.g., using a baculovirus expression system), yeast, or mammalian ceils. Other suitable host cells are known to those skilled in the art.

Accordingly, the present disclosure further pertains to methods of producing the subject TbKIί polypeptides. For example, a host cell transfected with an expression vector encoding a T BRI! polypeptide can be cultured under appropriate conditions to allow expression of the T ' pRll polypeptide to occur. The TbKII polypeptide may be secreted and isolated from a mixture of cells and medium containing the TpRII polypeptide.

Alternatively, the TpRII polypeptide may be retained cytoplasmically or in a membrane fraction and the cells harvested, lysed and the protein isolated. A cell culture includes host cells, and media. Suitable media for cell culture are well known in the art. The subject TpRII polypeptides can be isolated from cell culture medium, host ceils, or both, using techniques known in the art for purifying proteins, including ion-exchange chromatography, gel filtration chromatography, ultrafiltration, electrophoresis, immunoaffmity purification with antibodies specific for particular epitopes of the TbKIί polypeptides and affinity purification with an agent that binds to a domain fused to the TbKII polypeptide (e.g., a protein A column may be used to purify an TpiR!i- Fc fusion). In a preferred embodiment, the TpRll polypeptide is a fusion protein containing a domain which facilitates its purification. As an example, purification may be achieved by a series of column chromatography steps, including, for example, three or more of the following, in any order; protein A chromatography, Q sepharose chromatography, phenylsepharose chromatography, size exclusion chromatography, and cation exchange chromatography. The purification could be completed with viral filtration and buffer exchange.

In another embodiment, a fusion gene coding for a purification leader sequence, such as a poly-(His)/enterokmase cleavage site sequence at the N-terminus of the desired portion of the recombinant TbKίί polypeptide, can allow purification of the expressed fusion protein by affinity chromatography using a Ni 2+ metal resin. The purification leader sequence can then be subsequently removed by treatment with enterokinase to provide the purified TbKII polypeptide (e.g., see Hochuli et ah, (1987) J. Chromatography 411; 177: and Janknecht et al., PNAS USA 88:8972).

Techniques for making fusion genes are well known. Essentially, the joining of various DNA fragments coding for different polypeptide sequences is performed in accordance with conventional techniques, employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fiision gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers wiiich give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al., John Wiley & Sons; 1992).

8. Alterations in Fc-fusion proteins

The application further provides TbKII-Fc fusion proteins with engineered or variant Fc regions. Such antibodies and Fc fusion proteins may be useful, for example, in modulating effector functions, such as, antigen-dependent cytotoxicity (ADCC) and complement -dependent cytotoxicity (CDC). Additionally, the modifications may improve the stability of the antibodies and Fc fusion proteins. Amino acid sequence variants of the antibodies and Fc fusion proteins are prepared by introducing appropriate nucleotide changes into the DNA, or by peptide synthesis. Such variants include, for example, deletions from, and/or insertions into and/or substitutions of, residues within the amino acid sequences of the antibodies and Fc fusion proteins disclosed herein. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics. The amino acid changes also may alter post- translational processes of the antibodies and Fc fusion proteins, such as changing the number or position of glycosylation sites.

Antibodies and Fc fusion proteins with reduced effector function may be produced by introducing changes in the amino acid sequence, including, but are not limited to, the Ala- Ala mutation described by Bluestone et al. (see WO 94/28027 and WO 98/47531 ; also see Xu et al. 2000 Cell Immunol 200; 16-26). Thus, in certain embodiments, Fc fusion proteins of the disclosure with mutations within the constant region including the Ala- Ala mutation may be used to reduce or abolish effector function. According to these embodiments, antibodies and Fc fusion proteins may comprise a mutation to an alanine at position 234 or a mutation to an alanine at position 235, or a combination thereof. In one embodiment, the antibody or Fc fusion protein comprises an IgG4 framework, wherein the Ala-Ala mutation would describe a mutation(s) from phenylalanine to alanine at position 234 and/or a mutation from leucine to alanine at position 235. In another embodiment, the antibody or Fc fusion protein comprises an IgGl framework, wherein the Ala-Ala mutation would describe a mutation(s) from leucine to alanine at position 234 and/or a mutation from leucine to alanine at position 235. The antibody or Fc fusion protein may alternatively or additionally carry other mutations, including the point mutation K322A in the CH2 domain (Hezareh et al. 2001 I Virol. 75; 12161-8).

In particular embodiments, the antibody or Fc fusion protein may be modified to either enhance or inhibit complement dependent cytotoxicity (CDC). Modulated CDC activity' may be achieved by introducing one or more amino acid substitutions, insertions, or deletions in an Fc region (see, e.g., U.S. Pat. No. 6,194,551). Alternatively, or additionally, cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodime ric antibody thus generated may have improved or reduced internalization capability and/or increased or decreased complement-mediated cell killing. See Caron et ah, J. Exp Med. 176: 1191-1195 (1992) and Shopes, B. I. Immunol. 148:2918-2922 (1992), W099/51642, Duncan & Winter Nature 322: 738-40 (1988); U.S. Pat. No. 5,648,260; U.S. Pat. No. 5,624,821; and W094/29351.

9. Antibody Antagonists In certain aspects, a TpR [ f antagonist to be used in accordance with the methods and uses disclosed herein is an antibody, or combination of antibodies. An antibody TbKP antagonist may inhibit and/or bind to, for example, one or more ' TbKII ligands (e.g., TOR ' bI, TORb2, and/or TORb3), the TbKII receptor, TpRII-associated type I receptor (e.g., ALK5), and/or TpR.ff co-receptor (e.g., betaglycan). In some embodiments, the ability for an antibody TjlR!l antagonist antibody to inhibit activity (e.g., Smad signaling) and/or bind to a target is determined in an in vitro or cell-based assay including, for example, those disclosed herein. As described herein, an antibody TbKII antagonist may be used alone or in combination with one or more additional active agents or supportive therapies to treat SSc or one or more complications of SSc (e.g., SSc-ILD). In certain embodiments, a TpRII antagonist is an antibody that inhibits at least

TGFpl. Therefore, m some embodiments, an antibody TbKIί antagonist binds to at least TGFpi. As used herein, a TORbI antibody (anti-TGFpi antibody) generally refers to an antibody that is capable of binding to TORbI with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting TORbI. In certain embodiments, the extent of binding of an anti-TGFpl antibody to an unrelated, non-TGFpl protein is less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% of the binding of the antibody to TORbI as measured, for example, by a radioimmunoassay (RIA). In certain embodiments, an anti-TGPpi antibody binds to an epitope of TORbI that is conserved among TGFpl from different species. In certain preferred embodiments, an anti-TGFpl antibody binds to human TORbI. In some embodiments, a TORbI antibody may inhibit TORbI from binding to a type I, type 11, and/or co-receptor (e.g., TbKII, ALK5, and/or betaglycan) and thus inhibit TGFpl -mediated signaling (e.g., Smad signaling). It should be noted that ' TORbI shares some sequence homology to TORb2 and TORb3. Therefore, antibodies that bind TORbI, in some embodiments, may also bind to TORb2 and/or TORb3. In some embodiments, the disclosure relates to a multispecific antibody (e.g., bi-specific antibody), and uses thereof, that binds to TOR ' bI and further binds to, for example, one or more additional TpRII ligands (e.g., TORb2, TGFp3, or TORb2 and TORb3), one or more type I and/or type II receptors (e.g., TpRH and ALK5), and/or one or more co-receptors (e.g., betaglycan). In some embodiments, a multispecific antibody that binds to TGFpi does not bind or does not substantially bind to TGFp2 (e.g., binds to TGF[32 with a KD of greater than 1 x 10 "7 M or has relatively modest binding, e.g,, about 1 x IGr 8 M or about i x !0 "9 M). In some embodiments, a multispecific antibody that binds to TORbI further binds to TORb3 but does not bind or does not substantially bind to TGFB2 (e.g,, binds to TORb2 with a KD of greater than 1 x 10 “7 M or has relatively modest binding, e.g., about 1 x I0 ~8 M or about 1 x !O "9 M). In some embodiments, the disclosure relates to combinations of antibodies, and uses thereof, wherein the combination of antibodies comprises a TORb I antibody and one or more additional antibodies that bind to, for example, one or more additional TbKII ligands (e.g., TORb2, TORb3, or TGFp2 and TORb3), one or more type I and/or type II receptors (e.g., TbKII and ALK5), and/or one or more co-receptors (e.g., betaglycan). In some embodiments, a combination of antibodies that comprises a TORbI antibody does not comprise a TORb2 antibody . In some embodiments, a combination of antibodies that comprises a TGFpi antibody further comprises a TGFp3 antibody but does not comprise a TORb2 antibody.

In certain embodiments, an antibody TbKII antagonist is an antibody that inhibits at least TGFp2. Therefore, in some embodiments, an antibody TbKII antagonist antibody binds to at least TGFp2. As used herein, a TGFp2 antibody (anti-TGFp2 antibody) generally refers to an antibody that is capable of binding to TGFp2 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting TGFp2. In certain embodiments, the extent of binding of an anti-TGFp2 antibody to an unrelated, hoh-TORb2 protein is less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% of the binding of the antibody to TORb2 as measured, for example, by a radioimmunoassay (KIA).

In certain embodiments, an amti-TGFp2 antibody binds to an epitope of TORb2 that is conserved among TORb2 from different species. In certain preferred embodiments, an anti- TORb2 antibody binds to human TORb2. In some embodiments, a TGFp2 antibody may inhibit TGFp2 from binding to a type I, type P, and/or co-receptor (e.g., TpRII, ALK5, and/or betaglycan) and thus inhibit TORb2 activity (e.g., Smad signaling). It should be noted that TORb2 shares some sequence homology to TGFp I and TGFp3. Therefore, antibodies that bind TORb2, m some embodiments, may also bind to TG Rb ! and/or TGFB3. In some embodiments, the disclosure relates to a multispecific antibody (e.g., hi-speeific antibody), and uses thereof, that binds to TORb2 and further binds to, for example, one or more additional TpRII ligands (e.g., TGFpi, TGFp3, or TGFpi and TORb3), one or more type I and/or type II receptors (e.g., TpRII and ALK5), and/or one or more co-receptors (e.g., betaglycan) In some embodiments, the disclosure relates to combinations of antibodies, and uses thereof, wherein the combination of antibodies comprises a TGFp2 antibody and one or more additional antibodies that bind to, for example, one or more additional THRU ligands (e.g., TGFpl, TORb3, or TGFpl and TGFp3), one or more type I and/or type II receptors

(e.g., TpRII and ALK5), and/or one or more co-receptors (e.g., betaglycan).

In certain embodiments, an antibody TbKP antagonist is an antibody that inhibits at least TORb3. Therefore, in some embodiments, an antibody TpRII antagonist antibody binds to at least TGFp3, As used herein, a TGFp3 antibody (anti-TGFp3 antibody) generally refers to an antibody that is capable of binding to TORb3 with sufficient affinity' such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting TORb3. In certain embodiments, the extent of binding of an anti-TGFpS antibody to an unrelated, non-TGFp3 protein is less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than 1% of the binding of the antibody to TORb3 as measured, for example, by a radioimmunoassay (RIA). In certain embodiments, an anti-TGFp3 antibody binds to an epitope of TGFp3 that is conserved among TORb3 from different species. In certain preferred embodiments, an anti- TGFp3 antibody binds to human TORb3. In some embodiments, a TGFp3 antibody 7 may inhibit TORb3 from binding to a type I, type II, and/or co-receptor (e.g., TbKII, ALK5, and/or betaglycan) and thus inhibit TORb3 activity (e.g., Smad signaling). It should be noted that TORb3 shares some sequence homology to TORb2 and TGFpl . Therefore, antibodies that bind TGFpS, in some embodiments, may also bind to TORb2 and/or TORbI. In some embodiments, the disclosure relates to a multispecific antibody (e.g., bi-specific antibody), and uses thereof, that binds to TGFp3 and further binds to, for example, one or more additional TpRII ligands (e.g., TORb2, TORbI, or TORb2 and TORbI), one or more type I and/or type II receptors (e.g., TpRII and ALK5), and/or one or more co-receptors (e.g., betaglycan). In some embodiments, a multispecific antibody that binds to TGFp3 does not bind or does not substantially bind to TORb2 (e.g., binds to TORb2 with a KD of greater than 1 x 10 "7 M or has relatively modest binding, e.g., about 1 x 10 "8 M or about 1 x 10 "y M). In some embodiments, a multispecific antibody that binds to TGFp3 further binds to TGFp i but does not bind or does not substantially bind to TORb2 (e.g., binds to TORb2 with a KD of greater than 1 x 10 '7 M or has relatively modest binding, e.g., about 1 x 1 O '8 M or about 1 x 10 "y M). In some embodiments, the disclosure relates to combinations of antibodies, and uses thereof, wherein the combination of antibodies comprises a TGFp3 antibody and one or more additional antibodies that bind to, for example, one or more additional TBRfi ligands (e.g., TGFp2, TGFfll, or TGFp2 and TGFpl), one or more type 1 and/or type II receptors (e.g., TbKίϊ and ALK5), and/or one or more co-receptors (e.g., betaglycam). In some embodiments, a combination of antibodies that comprises a TORb3 antibody does not comprise a TORb2 antibody. In some embodiments, a combination of antibodies that comprises a TGFp3 antibody further comprises a TORbI antibody but does not comprise a TORb2 antibody.

In certain aspects, an antibody TbίIII antagonist is an antibody that inhibits at least the TbKίϊ receptor. Therefore, in some embodiments, an antibody TbKIί antagonist binds to at least the TiiRH receptor. As used herein, a TpR. II receptor antibody generally refers to an antibody that binds to a TbKII receptor with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent m targeting a TbίIΪI receptor. In certain embodiments, the extent of binding of an an!·- 1 bKP receptor antibody to an unrelated, non- TpRH receptor protein is less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than about 1 % of the binding of the antibody to the TPRH receptor as measured, for example, by a radioimmunoassay (KIA), Biacore, or other protein-protein interaction or binding affinity assay. In certain embodiments, an hhίί-TbKII receptor antibody binds to an epitope of a TbKII receptor that is conserved among TpR [ f receptors from different species. In certain preferred embodiments, an anti-TBRfi receptor antibody binds to a human TbKII receptor. In some embodiments, an amti-TpRII receptor antibody may inhibit one or more TpRII ligands [e.g., TGFpl; TORb2; TGFp3; TORbI and TORb3; TGFpl and TORb2;

TORb2 and TGFp3; or TORbI, TORb2, and TORb3] from binding to a TpR if receptor. In some embodiments, an anu-TpRH receptor antibody is a multispecific antibody (e.g., bi- specific antibody) that binds to a TpRII receptor and one or more TpR 11 ligands [e.g., TGFpl, TGFp2, and TGFp3 ], type I receptor (e.g., ALK5), and/or co-receptor (e.g., hetaglycan). In some embodiments, the disclosure relates to combinations of antibodies, and uses thereof, wherein the combination of antibodies compri ses an anti-TpRII receptor antibody and one or more additional antibodies that bind to, for example, one or more TtyRH ligands [e.g., TORbI , TORb2, and TORb3], type I receptors (e.g., ALK5), and/or co-receptor (e.g., betaglycan). In certain aspects, an antibody TbKIΪ antagonist is an antibody that inhibits at least

ALK5. Therefore, in some embodiments, an antibody TbKII antagonist antibody binds to at least ALK5. As used herein, an ALK5 antibody (anti- ALK5 antibody) generally refers to an antibody that binds to ALK5 with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting ALK5. In certain embodiments, the extent of binding of an anti-ALKS antibody to an unrelated, non-ALK5 protein is less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than about 1% of the binding of the antibody to ALK5 as measured, for example, by a radioimmunoassay (RIA), Biacore, or other protein- protein interaction or binding affinity assay. In certain embodiments, an anti-ALK5 antibody binds to an epitope of ALK5 that is conserved among ALK5 from different species. In certain preferred embodiments, an anti-ALK5 antibody binds to human ALK5. In some embodiments, an anti~ALK5 antibody may inhibit one or more TbR.P ligands [e.g., TGFpl; TGFp2; TGFfi3; TGFpl and TGF(}3; TGFfil and TGFp2: TGFfl2 and TGFfiB; or TGFpl, TGFp2, and TORb3] from binding to ALK5. In some embodiments, an anti-ALK5 antibody is a multispecific antibody (e.g,, bi-specific antibody) that binds to ALK5 and one or more TpRII ligands [e.g., TORbI, TORb2, and TGF[13], type II receptor (e.g., TpRH ·. and/or co- receptor (e.g., betaglycan). In some embodiments, the disclosure relates to combinations of antibodies, and uses thereof, wherein the combination of antibodies comprises an anti-ALK5 antibody and one or more additional antibodies that bind to, for example, one or more TbKII ligands [e.g., TORbI, TGFB2. and TGFp3 ], type II receptors (e.g., TfiRII), and/or co-reeeptor (e.g., betaglycan).

In certain aspects, an antibody TbϊIII antagonist is an antibody that inhibits at least betaglycan. Therefore, in some embodiments, an antibody TbKP antagonist binds to at least betaglycan. As used herein, a betaglycan antibody (anti-betaglycan antibody) generally refers to an antibody that binds to betaglycan with sufficient affinity such that the antibody is useful as a diagnostic and/or therapeutic agent in targeting betaglycan. In certain embodiments, the extent of binding of an anti-betaglycan antibody to an unrelated, non- betaglycan protein is less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or less than about 1% of the binding of die antibody to betaglycan as measured, for example, by a radioimmunoassay (RIA), Biacore, or other protein-protein interaction or binding affinity assay. In certain embodiments, an anti-betaglycan antibody binds to an epitope of betaglycan that is conserved among betaglycan from different species. In certain preferred embodiments, an anti-betaglycan antibody binds to human betaglycan. In some embodiments, an anti-betaglycan antibody may inhibit one or more TbKII ligands [e.g., TGFpl; TGFP2; TGFp3; TGFpl and TORb3; TGFpl and TGFp2; TGFp2 and TGFp3; or TGFpl, TORb2, and TORb3] from binding to betaglycan. In some embodiments, an anti- betaglycan antibody is a multispecific antibody (e.g., bi-specific antibody) that binds to betaglycan and one or more TpRJI ligands [e.g., TOEbI, TGF[¾2, and TORb3], type I receptor (e.g., ALK5), and/or type II receptors (e.g., TpRIl). In some embodiments, the disclosure relates to combinations of antibodies, and uses thereof, wherein the combination of antibodies comprises an anti-betaglycan antibody and one or more additional antibodies that bind to, for example, one or more TpRII ligands [e.g., TORbI, TGFp2, and TGFp3 |, type I receptors (e.g., ALK5), and/or type II receptors (e.g., TbίIP).

The term antibody is used herein in the broadest sense and encompasses various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, miiitispecifie antibodies (e.g., bispeeific antibodies), and antibody fragments so long as they exhibit the desired antigen-binding activity'. An antibody fragment refers to a molecule other than an intact antibody that comprises a portion of an intact antibody that binds the antigen to which the intact antibody binds. Examples of antibody fragments include but are not limited to Fv, Fab, Fab', Fab'-SH, F(ab')2; diabodies; linear antibodies; single-chain antibody molecules (e.g. , scFv); and multispecific antibodies formed from antibody fragments. See, e.g., Hudson et al. (2003) Nat. Med. 9: 129-134; Pliickthun, in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., (Springer- Verlag, New York), pp. 269-315 (1994); WO 93/16185; and U.S. Pat Nos, 5,571,894, 5,587,458, and 5,869,046. Antibodies disclosed herein may be polyclonal antibodies or monoclonal antibodies. In certain embodiments, the antibodies of the present disclosure comprise a label attached thereto and able to be detected (e.g., the label can be a radioisotope, fluorescent compound, enzyme, or enzyme co-factor). In preferred embodiments, the antibodies of the present disclosure are isolated antibodies. Diabodies are antibody fragments with two antigen-binding sites that may be bivalent or bispeeific. See, e.g., EP 404,097; WO 1993/01161; Hudson et al. (2003) Nat. Med. 9: 129-134 (2003); and Hollinger etal. (1993) Proc. Natl. Acad. Sci. USA 90: 6444-6448. Triabodies and tetrabodies are also described in Hudson et al. (2003) Nat. Med. 9: 129-134. Single-domain antibodies are antibody fragments comprising all or a portion of the heavy-chain variable domain or all or a portion of the light- chain variable domain of an antibody. In certain embodiments, a single-domain antibody is a human single-domain antibody. See, e.g., U.S. Pat. No. 6,248,516. Antibody fragments can be made by various techniques, including but not limited to proteolytic digestion of an intact antibody as well as production by recombinant host cells (e.g., E. eoli or phage), as described herein. The antibodies herein may be of any class. The class of an antibody refers to the type of constant domain or constant region possessed by its heavy chain. There are five major classes of antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may he further divided into subclasses (isotypes), for example, IgGi, IgG2, IgGs, IgGy IgAi, and IgA2. The heavy -chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu.

In general, an antibody for use in the methods disclosed herein specifically binds to its target antigen, preferably with high binding affinity. Affinity may be expressed as a KD value and reflects the intrinsic binding affinity (e.g., with minimized avidity effects). Typically, binding affinity is measured in vitro, whether m a cell-free or cell -associated setting. Any of a number of assays known in tire art, including those disclosed herein, can be used to obtain binding affinity measurements including, for example, surface plasmon resonance (Biacore™ assay), radiolabeled antigen binding assay (RIA), and ELISA. In some embodiments, antibodies of the present disclosure bind to their target antigens (e.g. TOTbI, TOTb2, TOEb2, ALK5, betaglycan, and TpRII.) with at least a KD of lx 1G '7 or stronger, IxlQ '8 or stronger, lxlO "9 or stronger, lxlO "10 or stronger, lxI0 '!1 or stronger, lxlO '12 or stronger, 1X10 "IJ or stronger, or lx 10 ' 14 or stronger.

In certain embodiments, KD is measured by RIA performed with the Fab version of an antibody of interest and its target antigen as described by the following assay. Solution binding affinity of Fabs for the antigen is measured by equilibrating Fab with a minimal concentration of radiolabeled antigen (e.g. , U3 I-labeled) in the presence of a titration series of unlabeled antigen, then capturing bound antigen with an anti-Fab antibody-coated plate [see, e.g., Chen et al. (1999) I. Mol, Biol. 293:865-881], To establish conditions for the assay, multi-well plates (e.g., MICROTITER ® from Thermo Scientific) are coated (e.g., overnight) with a capturing anti-Fab antibody (e.g., from Cappel Labs) and subsequently blocked with bovine scram albumin, preferably at room temperature (e.g., approximately 23°C). In a nonadsorbent plate, radiolabeled antigen are mixed with serial dilutions of a Fab of interest [e.g., consistent with assessment of the anti-VEGF antibody, Fab- 12, in Presta et al, (1997) Cancer Res. 57:4593-4599] . Idle Fab of interest is then incubated, preferably overnight but the incubation may continue for a longer period (e.g. , about 65 hours) to ensure that equilibrium is reached. Thereafter, the mixtures are transferred to the capture plate for incubation, preferably at room temperature for about one hour. The solution is then removed and the plate is washed times several times, preferably with polysorbate 20 and PBS mixture. When the plates have dried, scintillant (e.g., MICROSCINT ® from Packard) is added, and the plates are counted on a gamma counter (e.g., TOPCOUNT ® from Packard).

According to another embodiment, KD is measured using surface plasm on resonance assays using, for example a BIACORE ® 2000 or a BIACORE ® 3000 (Biacore, Inc., Piscataway, N.J.) with immobilized antigen CMS chips at about 10 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CMS, Biacore, Inc.) are activated with N-ethy ] -N'-(3-dimethy ] aminopropyl)-carbodiimide hydrochloride (EDC) and N- hydroxysuccinimide (NHS) according to the supplier's instructions. For example, an antigen can be diluted with 10 inM sodium acetate, pH 4.8, to 5 pg/ml (about 0.2 pM) before injection at a flow rate of 5 mΐ/minute to achieve approximately 10 response units (RU) of coupled protein. Following tire injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of Fab (0.78 nM to 500 nM) are injected in PBS with 0.05% polysorbate 20 (TWEEN-20 ® ) surfactant (PBST) at a flow rate of approximately 25 mΐ/min. Association rates (k 0n ) and dissociation rates (k 0ff ) are calculated using, for example, a simple one-to-one Langmuir binding model (BIACORE ®

Evaluation Software version 3.2) by simultaneously fitting the association and dissociation sensorgrams. The equilibrium dissociation constant (KD) is calculated as the ratio koff / kon [see, e.g., Chen et al., (1999) I. Mol. Biol. 293:865-881], If the on-rate exceeds, for example, 10 6 M '1 s '1 by the surface piasmon resonance assay above, then the on-rate can be determined by using a fluorescent quenching technique that measures the increase or decrease in fluorescence emission intensity (e.g., exci†ation=295 nm; emission=340 nm, 16 nm bandpass) of a 20 nM anti-antigen antibody (Fab form) in P BS in the presence of increasing concentrations of antigen as measured in a spectrometer, such as a stop-flow equipped spectrophotometer (Aviv Instruments) or a 8000-series SLM-AMINCO ® spectrophotometer (ThermoSpectronic) with a stirred cuvette.

The nucleic acid and amino acid sequences of TbKίί, LEK5. betaglycan, TΌRbI, TOEb2, and TGFf>3, particularly human sequences, are well known in the art and thus antibody antagonists for use in accordance with this disclosure may be routinely made by the skilled artisan based on the knowledge in the art and teachings pro vided herein. In certain embodiments, an antibody provided herein is a chimeric antibody. A chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while the remainder of the heavy and/or light chain is derived from a different source or species. Certain chimeric antibodies are described, for example, in U.S. Pat. No. 4,816,567; and Morrison et a /., (1984) Proc. Natl. Acad. Sci. USA, 81:6851-6855. In some embodiments, a chimeric antibody comprises a non-lmman variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or nonhuman primate, such as a monkey) and a human constant region. In some embodiments, a chimeric antibody is a "class switched" antibody in which the class or subclass has been changed from that of the parent antibody. In general, chimeric antibodies include antigenbinding fragments thereof.

In certain embodiments, a chimeric antibody provided herein is a humanized antibody. A humanized antibody refers to a chimeric antibody comprising amino acid residues from non-human hypervariable regions (HVRs) and amino acid residues from human framework regions (FRs). In certain embodiments, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which ail or substantially all of the HVRs (e.g., CDRs) correspond to those of a non-human antibody, and all or substantially all of the FRs correspond to those of a human antibody. A humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. A "humanized form" of an antibody, e.g., a non-human antibody, refers to an antibody that has undergone humanization. Humanized antibodies and methods of making them are reviewed, for example, in Almagro and Fransson (2008) Front. Biosci.

13: 1619-1633 and are further described, for example, m Riechmann et al., (1988) Nature 332:323-329; Queen et al. (1989) Proc. Nat'3 Acad. Sci. USA 86: 10029-10033; U.S, Pat. Nos.

5,821,337, 7,527,791 , 6,982,321, and 7,087,409; Kashmiri et al., (2005) Methods 36:25-34 [describing SDR (a-CDR) grafting]; Padlan, Mol. Immunol. (1991) 28:489-498 (describing "resurfacing"); Dall’Acqua etal. (2005) Methods 36:43-60 (describing "FR shuffling"); Osbourn etal. (2005) Methods 36:61-68; and Niunka a al Br. J. Cancer (2000) 83:252-260 (describing the "guided selection" approach to FR shuffling).

Human framework regions that may be used for humanization include but are not limited to: framework regions selected using the "best-fit" method [see, e.g., Sims etal.

(1993) J. Immunol. 151:2296 ]; framework regions derived from the consensus sequence of human antibodies of a particular subgroup of light-chain or heavy-chain variable regions [see, e.g., Carter etal. (1992) Proc. Natl. Acad. Sci. USA, 89:4285; and Presta etal. (1993) I.

Immunol., 151:2623]; human mature (somatically mutated) framework regions or human germline framework regions [see, e.g., Almagro and Fransson (2008) Front. Biosci. 13:1619- 1633]; and framework regions derived from screening FR libraries [see, e.g., Baca et cd., (1997) J. Biol. Chem. 272:10678-10684; and Rosok et cd, (1996) J. Biol. Chem. 271:22611- 22618],

In certain embodiments, an antibody provided herein is a human antibody. Human antibodies can be produced using various techniques known in the art. Human antibodies are described generally in van Dijk and van de Winkel (2001) Cisrr. Opm. Pharmacol. 5: 368-74 and Lonberg (2008) Curr. Opin. Immunol. 20:450-459.

In some embodiments, human antibodies may be prepared by administering an immunogen (e.g., a TbKIί, ALK5, betag!ycan, TORbI, TORb2, or TORb3 polypeptide) to a transgenic animal that has been modified to produce intact human antibodies or intact antibodies with human variable regions in response to antigenic challenge. Such animals typically contain all or a portion of the human immunoglobulin loci, which replace the endogenous immunoglobulin loci, or which are present extrachromosomally or integrated randomly into the animal's chromosomes. In such transgenic animals, the endogenous immunoglobulin loci have generally been inactivated. For a review of methods for obtaining human antibodies from transgenic animals, see, for example, Lonberg (2005) Nat.

Biotechnol. 23: 1117-1125; U.S, Pat. Nos. 6,075,181 and 6,150,584 (describing XENOMOUSE™ technology); U.S. Pat. No. 5,770,429 (describing HuMab ® technology); U.S. Pat. No. 7,041,870 (describing K-M MOUSE ® technology); and U.S. Patent Application Publication No. 2007/0061900 (describing VelociMouse ® technology). Human variable regions from intact antibodies generated by such animals may be further modified, for example, by combining with a different human constant region.

Human antibodies provided herein can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines for the production of human monoclonal antibodies have been described [see, e.g., Kozbor J. Immunol., (1984) 133: 3001; Brodeur etal. (1987) Monoclonal Antibody Production Techniques and Applications, pp. 51- 63, Marcel Dekker, Inc., New York; and Boemer et at. (1991) J. Immunol., 147: 86], Human antibodies generated via human B-cell hybridoma technology are also described in Li et al. , (2006) Proc. Natl. Acad. Sci. USA, 103:3557-3562. Additional methods include those described, for example, in U.S. Pat. No. 7,189,826 (describing production of monoclonal human TgM antibodies from hybridoma cell lines) and Ni, Xiandai Mianyixue (2006) 26(4):265-268 (2006) (describing human-human hybridomas). Human hybridoma technology (Trioma technology) is also described in Vollmers and Brandlein (2005) Histol. HistopathoL, 20(3): 927-937 (2005) and Vollmers and Brandlein (2005) Methods Find Exp. Clm. Pharmacol., 27(3): 185-91.

Human antibodies provided herein may also be generated by isolating Fv done variable -domain sequences selected from human-derived phage display libraries. Such variable-domain sequences may then be combined with a desired human constant domain. Techniques for selecting human antibodies from antibody libraries are described herein.

For example, antibodies of the present disclosure may be isolated by screening combinatorial libraries for antibodies with the desired activity or activities. A variety of methods are known in the art for generating phage-display libraries and screening such libraries for antibodies possessing the desired binding characteristics. Such methods are reviewed, for example, in Hoogenboom et al. (2001) in Methods in Molecular Biology 178: 1- 37, O'Brien et al, ed., Human Press, Totowa, N.J. and further described, for example, in the McCafferty et al. (1991) Nature 348:552-554; Clackson etal., (1991) Nature 352: 624-628; Marks etal. (1992) J. Mol. Biol. 222:581-597; Marks and Bradbury 7 (2003) in Methods in Molecular Biology 248: 161-175, Lo, ed., Human Press, Totowa, N.J.; Sidhu etal. (2004) j. Mol. Biol. 338(2):299-3!0; Lee etal. (2004) I, Mol. Biol, 340(5): 1073-1093; Feilouse (2004) Proc. Natl. Acad. Sci. USA 101(34): 12467-12472; and Lee et al. (2004) J. Immunol.

Methods 284(1-2): 119-132.

In certain phage display methods, repertoires of VH and VL genes are separately cloned by polymerase chain reaction (PCR) and recombined randomly in phage libraries, which can then be screened for antigen-binding phage as described in Winter et al. (1994) Ann Rev. Immunol., 12: 433-455. Phage typically display antibody fragments, either as single-chain Fv (scFv) fragments or as Fab fragments. Libraries from immunized sources pro vide high-affinity antibodies to the immunogen (e.g, abetaglycan, TbKII, TGFpl,

TORb2, or TGFP3 polypeptide) without the requirement of constructing hybridomas. Alternatively, the naive repertoire can be cloned (e.g., from human) to provide a single source of antibodies directed against a wide range of non-self and also self-antigens without any immunization as described by Griffiths etal. (1993) EMBO J, 12: 725-734. Finally, naive libraries can also be made synthetically by cloning un-rearranged V-gene segments from stem cells and using PCR primers containing random sequence to encode the highly variable CDR3 regions and to accomplish rearrangement in vitro, as described by Hoogenboom and Whiter (1992) J. Mol. Biol., 227: 381-388. Patent publications describing human antibody phage libraries include, for example: U.S. Pat, No. 5,750,373, and U.S, Patent Publication Nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936, and 2009/0002360.

In certain embodiments, an antibody provided herein is a multispecific antibody, for example, a bispecific antibody. Multispecific antibodies (typically monoclonal antibodies) have binding specificities for at least two different epitopes {e.g., two, three, four, five, or six or more) on one or more (e.g., two, three, four, five, six or more) antigens. Engineered antibodies with three or more functional antigen binding sites, including "octopus antibodies," are also included herein (see, e.g., US 2006/0025576A1).

In certain embodiments, the antibodies disclosed herein are monoclonal antibodies. Monoclonal antibody refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variant antibodies, e.g., containing naturally occurring mutations or arising during production of a monoclonal antibody preparation, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different epi topes, each monoclonal antibody of a monoclonal antibody preparation is directed against a single epitope on an antigen. Tims, the modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies and is not to be construed as requiring production of the antibody by any particular method. For example, the monoclonal antibodies to be used in accordance with the present methods may be made by a variety of techniques, including but not limited to the hybridoma method, recombinant DNA methods, phage-display methods, and methods utilizing transgenic animals containing all or part of the human immunoglobulin loci, such methods and other exemplary 7 methods for making monoclonal antibodies being described herein.

For example, by using immunogens derived from a TbKII receptor polypeptide, anti- protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols [see, e.g., Antibodies: A Laboratory Manual (1988) ed. by Harlow and Lane, Cold Spring Harbor Press], A mammal, such as a mouse, hamster, or rabbit can be immunized with an immunogenic form of the TbKII polypeptide, an antigenic fragment which is capable of eliciting an antibody response, or a fusion protein. Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers or other techniques well known in the art. An immunogenic portion of a TfiRfl polypeptide can be administered in the presence of adjuvant. The progress of immunization can be monitored by detection of antibody titers in piasma or serum. Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibody production and/or level of binding affinity. Following immunization of an animal with an antigenic preparation of TbKII polypeptide, anti sera can be obtained and, if desired, polyclonal antibodies can be isolated from the serum. To produce monoclonal antibodies, antibody-producing cells (lymphocytes) can be harvested from an immunized animal and fused by standard somatic cell fusion procedures with immortalizing cells such as myeloma cells to yield hybridotna ceils. Such techniques are well known in the art, and include, for example, the hybridoma technique [see, e.g., Kohler and Milstein (1975) Nature, 256: 495-497], the human B cell hybridoma technique [see, e.g., Kozbar etal. (1983) Immunology Today, 4:72], and the EBV-hybridoma technique to produce human monoclonal antibodies [Cole et al. (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc. pp. 77-96], Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with a TbKP polypeptide, and monoclonal antibodies isolated from a culture comprising such hybridoma cells.

In certain embodiments, one or more amino acid modifications may be introduced into the Fc region of an antibody provided herein thereby generating an Fc-region variant. The Fc-region variant may comprise a human Fc-region sequence (e.g., a human IgGl , TgG2, IgG3 or IgG4 Fc region) comprising an amino acid modification (e.g., a substitution, deletion, and/or addition) at one or more amino acid positions.

For example, the present disclosure contemplates an antibody variant that possesses some but not ail effector functions, which make it a desirable candidate for applications in which the half-life of the antibody m vivo is important yet for which certain effector functions [e.g., complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC)] are unnecessary or deleterious. In vitro and/or in vivo cytotoxicity assays can be conducted to confirm the reduction/depletion of CDC and/or ADCC activities. For example, Fc receptor (FcR) binding assays can be conducted to ensure that the antibody lacks FcyR binding (hence likely lacking ADCC activity), but retains FcRn binding ability. The primary cells for mediating ADCC, NK ceils, express FcyRIII only, whereas monocytes express FcyRI, FcyRII and FcyRIII. FcR expression on hematopoietic cells is summarized in, for example, Ravetch and Kinet (1991) Annu. Rev. Immunol. 9:457-492. Non-limiting examples of in vitro assays to assess ADCC activity of a molecule of interest are described in U.S. Pat. No. 5,500,362; HeUstrom, 1. etal. (1986) Proc, Nat'l Acad. Sci. USA 83:7059-7063; Hell strom, I etal. (1985) Proc. Nat’l Acad. Sci. USA 82: 1499-1502; U.S. Pat. No. 5,821,337; and Bruggemann, M. etal. (1987) J. Exp. Med. 166: 1351-1361. Alternatively, non- radioactive assay methods may be employed (e.g, ACT!™, non-radioactive cytotoxicity- assay for flow cytometry; CellTechnoiogy, Inc. Mountain View, Calif; and CytoTox 96 ® non-radioactive cytotoxicity assay, Promega, Madison, Wis.). Useful effector ceils for such assays include peripheral blood mononuclear cells (PBMC) and natural killer (NK) cells. Alternatively, or additionally, ADCC activity of the molecule of interest may be assessed in vivo, for example, in an animal model such as that disclosed in dynes et al. (1998) Proc.

Nat'l Acad. Sci. USA 95:652-656. Clq binding assays may also be carried out to confirm that the antibody is unable to bind Clq and hence lacks CDC activity [see, e.g., Clq and C3c binding ELISA in WO 2006/029879 and WO 2005/100402], To assess complement activation, a CDC assay may be performed [see, e.g,, Gazzano-Santoro etal. (1996) j. Immunol. Methods 202: 163; Cragg, M. S. etal. (2003) Blood 101:1045-1052; and Cragg, M. S, and M. J. Glennie (2004) Blood 103:2738-2743], FcRn binding and in vivo clearance/halflife determinations can also be performed using methods known in the art [see, e.g., Petkova, S. B, et al (2006) Int. Immunol. 18(12): 1759-1769].

Antibodies of the present disclosure with reduced effector function include those with substitution of one or more of Fc region residues 238, 265, 269, 270, 297, 327 and 329 (U.S. Pat. No. 6,737,056). Such Fc mutants include Fc mutants with substitutions at two or more of amino acid positions 265, 269, 270, 297 and 327, including the so-called "DANA" Fc mutant with substitution of residues 265 and 297 to alanine (U.S. Pat. No. 7,332,581).

In certain embodiments, it may be desirable to create cysteine-engineered antibodies, e.g., "thioMAbs," in which one or more residues of an antibody are substituted with cy steine residues. In particular embodiments, the substituted residues occur at accessible sites of the antibody. By substituting those residues with cysteine, reactive thiol groups are thereby positioned at accessible sites of the antibody and may be used to conjugate the antibody to other moieties, such as drug moieties or linker-drug moieties, to create an immunoconjugate, as described further herein. In certain embodiments, any one or more of the following residues may be substituted with cysteine: V205 (Kabat numbering) of the light chain; Al 18 (EU numbering) of the heavy chain; and S400 (EU numbering) of the heavy-chain Fc region. Cysteine engineered antibodies may be generated as described, for example., in U.S. Pat. No. 7,521,541.

In addition, the techniques used to screen antibodies in order to identify a desirable antibody may influence the properties of the antibody obtained. For example, if an antibody is to be used for binding an antigen in solution, it may be desirable to test solution binding. A variety of different techniques are available fortesting interaction between antibodies and antigens to identify particularly desirable antibodies. Such techniques include ELISA s, surface plasmon resonance binding assays (e.g., the Biacore™ binding assay, Biacore AB, Uppsala, Sweden), sandwich assays (e.g., the paramagnetic bead system of IGEN International, Inc., Gaithersburg, Maryland), western blots, immunoprecipitation assays, and immunohistochemistry.

In certain embodiments, amino acid sequence variants of the antibodies and/or the binding polypeptides provided herein are contemplated. For example, it may be desirable to improve die binding affini ty and/or other biological properties of the antibody and/or binding polypeptide. Amino acid sequence variants of an antibody and/or binding polypeptides may be prepared by introducing appropriate modifications into the nucleotide sequence encoding the antibody' and/or binding polypeptide, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into, and/or substitutions of residues within, the amino acid sequences of the antibody and/or binding polypeptide. Any combination of deletion, insertion, and substitution can be made to arrive at the final construct, provided that the final construct possesses the desired characteristics, e.g., target- binding (TbKII, ALK5, betagiycan, TGF(31, TGFp2, and/or TGF(}3).

Alterations (e.g., substitutions) may be made in HVRs, for example, to improve antibody affinity. Such alterations may be made in HVR "hotspots," i.e., residues encoded by codons that undergo mutation at high frequency during the somatic maturation process (see, e.g., Chowdhury (2008) Methods Mol, Biol. 207: 179-196 (2008)), and/or SDRs (a-CDRs), with the resulting variant VH or VL being tested for binding affinity. Affinity maturation by- constructing and reselecting from secondary libraries has been described in the art [see, e.g., Hoogenboom etaL, in Methods in Molecular Biology 178: 1-37, O'Brien etal., ed,, Human Press, Totowa, N.J., (2001)]. In some embodiments of affinity maturation, diversity is introduced into the variable genes chosen for maturation by any of a variety of methods (e.g., error-prone PCR, chain shuffling, or oligonucleotide-directed mutagenesis). A secondary- library' is then created. The library is then screened to identify any antibody variants with the desired affinity. Another method to introduce diversity involves HVR-directed approaches, in which se veral HVR residues {e.g., 4-6 residues at a time) are randomized. HVR residues involved in antigen binding may be specifically identified, e.g., using alanine scanning mutagenesis or modeling. CDR-H3 and CDR-L3 in particular are often targeted, In certain embodiments, substitutions, insertions, or deletions may occur within one or more HVRs so long as such alterations do not substantially reduce the ability of the antibody to bind to the antigen. For example, conservative alterations (e.g., conservative substitutions as provided herein) that do not substantially reduce binding affinity may be made in HVRs. Such alterations may be outside of HVR "hotspots" or SDRs. In certain embodiments of the variant VH and VL sequences provided above, each HVR either is unaltered, or contains no more than one, two, or three amino acid substitutions.

A useful method for identification of residues or regions of the antibody and/or the binding polypeptide that may be targeted for mutagenesis is called "alanine scanning mutagenesis", as described by Cunningham and Wells (1989) Science, 244:1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced by a neutral or negatively charged amino acid (e.g., alanine or polyalanine) to determine whether the interaction of the antibody or binding polypeptide with antigen is affected. Further substitutions may be introduced at the ammo acid locations demonstrating functional sensitivity to the initial substitutions. Alternatively, or additionally, a crystal structure of an antigen -antibody complex can be used to identify contact points between the antibody and antigen. Such contact residues and neighboring residues may be targeted or eliminated as candidates for substitution. Variants may be screened to determine whether they contain the desired properties.

Ammo-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. Other insertional variants of the antibody molecule include fission of tire N- or C-terminus of the antibody to an enzyme (e.g., for ADEPT) or a polypeptide which increases the serum half-life of the antibody.

In certain embodiments, an antibody and/or binding polypeptide provided herein may be further modified to contain additional non-proteinaceous moieties that are known in the art and readily available. The moieties suitable for derivatization of the antibody and/or binding polypeptide include but are not limited to water-soluble polymers. Non-limiting examples of water-soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethy!cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone, poly-1, 3-dioxolane, poly-l,3,6-trioxane, ethylene/maleic anhydride copolymer, polyaminoacids (either homopolymers or random copolymers), and dextran or poly(n-vinyl pyrrolidone)poiyethyiene glycol, propropylene glycol homopolymers, prolypropylene oxide/ethylene oxide co-polymers, po!yoxyethy!ated polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof. Polyethylene glycol propionaldehyde may have advantages in manufacturing due to its stability in water. The polymer may be of any molecular weight, and may be branched or unbranched . The number of polymers attached to the antibody and/or binding polypeptide may vary, and if more than one polymer are attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the antibody and/or binding polypeptide to be improved, whether the antibody derivative and/or binding polypeptide derivative will he used in a therapy under defined conditions.

10. Small Molecule Antagonists In certain aspects, a TbKII antagonist to be used in accordance with the methods and uses disclosed herein is a small molecule (a small molecule TbKII antagonist), or combination of small molecules. A small molecule TfiRII antagonist may inhibit, for example, one or more TbKII ligands (e.g., TORbI, TORb2, and TGFP3), TjlRII receptor, TpRII-associated type I receptor (e.g., ALK5), TpRII-associated co-receptor (e.g., betaglycan), and/or downstream signaling component (e.g., Smad proteins). In some embodiments, the ability for a small molecule THRU antagonist to inhibit signaling (e.g., Smad signaling) is determined in a cell -based assay including, for example, those described herein. A small molecule TbKIί antagonist may be used alone or in combination with one or more additional active agents and/or supportive therapies to treat S Sc or a clinical complication of SSc (e.g., SSc-ILD).

In certain aspects, a small molecule TbKII antagonist inhibits at least TGFpl (e.g., inhibition of Smad signaling). Therefore, in some embodiments, a small molecule inhibitor ofTGPpi binds to TORbI . In some embodiments, a small molecule inhibitor of TGFpI inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of TGFpi. In some embodiments, a small molecule inhibitor of TGFpi further inhibits one or more of TORb2, TΌRb3, TpRII, ALK5, and betaglycan. In some embodiments, a small molecule inhibitor of TORbI does not inhibit or does not substantially inhibit TORb2, In some embodiments, a small molecule inhibitor of TORbI further inhibits TGFp3 but does not inhibit or does not substantially inhibit TORb2. In certain aspects, a small molecule TbKII antagonist inhibits at least TORb2. Therefore, in some embodiments, a small molecule inhibitor of TORb2 binds to TORb2. In some embodiments, a small molecule inhibitor of TORb2 inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of TGFp2, In some embodiments, a small molecule inhibitor of TGFp2 further inhibits one or more of TORb3, TGFpi, TbϊIII, ALK5, and betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits at least TORb3. Therefore, in some embodiments, a small molecule inhibitor of TGFjG binds to TGFp3. In some embodiments, a small molecule inhibitor of TΌR ' b3 inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of TORb3. In some embodiments, a small molecule inhibitor of TORb3 further inhibits one or more of TGFp2, TORbI, TbKII, ALK5, and betaglycan. In some embodiments, a small molecule inhibitor of TORb3 does not inhibit or does not substantially inhibit TORb2. In some embodiments, a small molecule inhibitor of TORb3 further inhibits TORbI but does not inhibit or does not substantially inhibit TORb2. In certain aspects, a small molecule TbKII antagonist inhibits at least a TpRII receptor. Therefore, in some embodiments, a small molecule inhibitor of TpRII binds to a TpR!l receptor. In some embodiments, a small molecule inhibitor of TpRII inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of a TbKIί receptor. In some embodiments, a small molecule inhibitor of a TpRII receptor further inhibits one or more of TORbI, TORb2, TORb3, ALK5, and betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits TORbI from binding to a TpRU receptor. In certain aspects, a small molecule TpRff antagonist small molecule inhibits TGFp2 from binding to a TpRII receptor. In certain aspects, a small molecule TbKII antagonist inhibits TORb3 from binding to a TbKII receptor. In certain aspects, a small molecule TpRII antagonist inhibits TGFpi and TGFp3 from binding to a TpRII receptor. In certain aspects, a small molecule TpRff antagonist inhibits TGFpi, TORb2, and TORb3 from binding to a TpRII receptor. In certain aspects, a small molecule TpRII antagonist inhibits TGFpi from binding to a TbKII receptor but does not inhibit or does not substantially inhibit TORb2 from binding to a TbKP receptor. In certain aspects, a small molecule TbKP antagonist inhibits TGFp3 from binding to a TpRII receptor but does not inhibit or does not substantially inhibit TGFp2 from binding to a TbKII receptor. In certain aspects, a small molecule TpRff antagonist inhibits TGFpi and TORb3 from binding to a TpRII receptor but does not inhibit or does not substantially inhibit TORb2 from binding to a TbKII receptor. In certain aspects, a small molecule TPRH antagonist inhibits at least ALK5. Therefore, in some embodiments, a small molecule inhibitor of ALK5 binds to ALK5. In some embodiments, a small molecule inhibitor of ALK5 inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of ALK5. In some embodiments, a small molecule inhibitor of ALK5 further inhibits one or more of TGFpi, TGFp2, TORb3, TbKII, and betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits TGFpl from binding to ALK5. In certain aspects, a small molecule

TpRII antagonist inhibits TGFp2 from binding to ALK5. In certain aspects, a small molecule

TpRII antagonist inhibits TGFp3 from binding to ALK5. In certain aspects, a small molecule

TpRII antagonist inhibits TORbI and TGFjG from binding to ALK5. In certain aspects, a small molecule TbKIΪ antagonist inhibits TORbί , TORb2, and TGFp3 from binding to ALK5. In certain aspects, a small molecule TbKII antagonist inhibits TORbI from binding to ALK5 but does not inhibit or does not substantially inhibit TGFp2 from binding to ALK5. In certain aspects, a small molecule TpRII antagonist inhibits TORb3 from binding to ALK5 but does not inhibit or does not substantially inhibit TORb2 from binding to ALK5. In certain aspects, a small molecule TpRII antagonist inhibits TORbI and TGFp3 from binding to ALK5 but does not inhibit or does not substantially inhibit TGFp2 from binding to ALK5. In certain aspects, a small molecule TbMΪ antagonist inhibits at least betaglycan. Therefore, in some embodiments, a small molecule inhibitor of betaglycan binds to betaglycan. In some embodiments, a small molecule inhibitor of betaglycan inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of betaglycan. In some embodiments, a small molecule inhibitor of betaglycan further inhibits one or more of TGFpl, TORb2, TORb3, TpRII, and ALK5. In certain aspects, a small molecule TpRII antagonist inhibits TGFpl from binding to betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits TORb2 from binding to betaglycan. In certain aspects, a small molecule TpRII antagonist small molecule inhibits TGFp3 from binding to betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits TGFpi and TORb3 from binding to betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits TGFpi, TGFp2, and TGFp<3 from binding to betaglycan. In certain aspects, a small molecule TpRII antagonist inhibits TGFpi from binding to betaglycan but does not inhibit or does not substantially inhibit TGFp2 from binding to betaglycan. In certain aspects, a small molecule I|)RJI antagonist inhibits TORb3 from binding to betaglycan but does not inhibit or does not substantially inhibit TORb2 from binding to betaglycan. In certain aspects, a small molecule TbKP antagonist inhibits TOR ' bI and TORb3 from binding to betaglycan but does not inhibit or does not substantially inhibit TORb2 from binding to betaglycan. Small molecule TbKII antagonist can be direct or indirect inhibitors. For example, a small molecule TbKII antagonist, or combination of small molecules, may inhibit the expression (e.g,, transcription, translation, cellular secretion, or combinations thereof) of at least one or more of TbKII, ALK5, betaglycan, TORbI , TORb2, TORb3, and/or one or more downstream TbMί signaling factors (Smads). Alternatively, a direct small molecule TbKII antagonist, or combination of small molecules, may directly bind to, for example, one or more of TbKII, ALK5, betaglycan, TORbI, TORb2, and TORb3 or one or more downstream TbίIII signaling factors. Combinations of one or more indirect and one or more direct small molecule TbίIII antagonist may be used in accordance with the methods and uses disclosed herein . Binding organic small molecule antagonists of the present disclosure may be identified and chemically synthesized using known methodology (see, e.g., PCX Publication Nos. WO 00/00823 and WO 00/39585). In general, small molecule antagonists of the disclosure are usually less than about 2000 daltons in size, alternatively less than about 1500, 750, 500, 250 or 200 daltons in size, wherein such organic small molecules that are capable of binding, preferably specifically, to a polypeptide as described herein (e.g., TbKII, ALK5, betaglycan, TORbI, TORb2, and TORb3). Such small molecule antagonists may be identified without undue experimentation using well-known techniques. In this regard, it is noted that techniques for screening organic small molecule libraries for molecules that are capable of binding to a polypeptide target are well-known in the art (see, e.g., international patent publication Nos, WO00/00823 and WOQO/39585),

Binding organic small molecules of the present disclosure may be, for example, aldehydes, ketones, oximes, hydrazones, semicarbazones, carbazides, primary amines, secondary' amines, tertiary amines, N-substituted hydrazines, hydrazides, alcohols, ethers, thiols, thioethers, disulfides, carboxylic acids, esters, amides, ureas, carbamates, carbonates, ketals, thioketals, acetals, thioacetals, aryl halides, aryl sulfonates, alkyl halides, alkyl sulfonates, aromatic compounds, heterocyclic compounds, anilines, alkenes, alkynes, diols, amino alcohols, oxazolidines, oxazolines, thiazolidines, thiazohnes, enamines, sulfonamides, epoxides, aziridines, isocyanates, sulfonyl chlorides, diazo compounds, and acid chlorides. 11. Antagonist Polynucleotides

In certain aspects, a TpRIi antagonist to be used in accordance with the methods and uses disclosed herein is a polynucleotide (a polynucleotide TbR.II antagonist), or combination of polynucleotides. A polynucleotide TbKII antagonist may inhibit, for example, one or more TpRII ligands (e.g., TORbI, TGFp2, and TORb3), TbKII receptor, TpRII-assoaated type I receptor (e.g., ALK5), TpRII-associated co-receptor (e.g., betaglycan), and/or downstream signaling component (e.g., Smad proteins). In some embodiments, the ability for a polynucleotide Tj/Rll antagonist to inhibit signaling (e.g., Smad signaling) is determined in a cell-based assay including, for example, those described herein. A polynucleotide TpRIi antagonist may be used alone or in combination with one or more additional active agents and/or supportive therapies to treat SSc or a clinical complication of SSc (e.g., SSc-ILD).

In certain aspects, a polynucleotide TpRIl antagonist inhibits at least TΌRbI (e.g., inhibition of Smad signaling). Therefore, in some embodiments, a polynucleotide inhibitor ofTGFpi binds to TORbI. In some embodiments, a polynucleotide inhibitor of TGFpl inhibits expression (e.g, , transcription, translation, secretion, or combinations thereof) of TGFpi. In some embodiments, a polynucleotide inhibitor of TORbί further inhibits one or more of TORb2, TORb3, TbKII, ALK5, and betaglycan. In some embodiments, a polynucleotide inhibitor of TORbI does not inhibit or does not substantially inhibit TORb2.

In some embodiments, a polynucleotide inhibitor of TGFB 1 further inhibits TORb3 but does not inhibit or does not substantially inhibit TORb2, In certain aspects, a polynucleotide

T|)RH antagonist inhibits at least TORb2. Therefore, in some embodiments, a polynucleotide inhibitor of TORb2 binds to TGFp2. In some embodiments, a polynucleotide inhibitor of TORb2 inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of TGFp2. In some embodiments, a polynucleotide inhibitor of TORb2 further inhibits one or more of TGFp3, TORbI, TbIIII, ALK5, and betaglycan. In certain aspects, a polynucleotide TBRif antagonist inhibits at least TORb3. Therefore, in some embodiments, a polynucleotide inhibitor of TORb3 binds to TORb3. In some embodiments, a polynucleotide inhibitor of TORb3 inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of TGFp3. In some embodiments, a polynucleotide inhibitor of TORb3 further inhibits one or more of TORb2, ' TORbI, TbRII, ALK5, and betaglycan. In some embodiments, a polynucleotide inhibitor of TORb3 does not inhibit or does not substantially inhibit TORb2. In some embodiments, a polynucleotide inhibitor of TGFp3 further inhibits TORbI but does not inhibit or does not substantially inhibit TORb2. In certain aspects, a polynucleotide TpRII antagonist inhibits at least a TpRII receptor. Therefore, in some embodiments, a polynucleotide inhibitor of TpRI! binds to a TbKII receptor. In some embodiments, a polynucleotide inhibitor of TpRII inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of a TpRII receptor. In some embodiments, a polynucleotide inhibitor of a TpRII receptor further inhibits one or more of TGFpl, TGF[32, TGFB3. ALK5, and betaglycan. In some embodiments, a polynucleotide TpRU antagonist inhibits TORbI from binding to a TbKII receptor. In some embodiments, a polynucleotide TPRH antagonist inhibits TORb2 from binding to a TpRII receptor. In some embodiments, a polynucleotide TpRII antagonist inhibits TORb3 from binding to a TbKII receptor. In some embodiments, a polynucleotide TbKΪI antagonist inhibits TORbI and TGFp3 from binding to a TpRII receptor. In some embodiments, a polynucleotide TpRII antagonist inhibits TORbI, TORb2, and TGFp3 from binding to a TpRII receptor. In some embodiments, a polynucleotide TpRII antagonist inhibits TGFpl from binding to a TpRII receptor but does not inhibit or does not substantially inhibit TGFp2 from binding to a TpRII receptor. In some embodiments, a polynucleotide TpRII antagonist inhibits TORb3 from binding to a TpRII receptor but does not inhibit or does not substantially inhibit TGFp2 from binding to a TpRII receptor. In some embodiments, a polynucleotide TpRII antagonist inhibits TGFpi and TGFp3 from binding to a TpRII receptor but does not inhibit or does not substantially inhibit TORb2 from binding to a TpRII receptor. In certain aspects, a polynucleotide TpRII antagonist inhibits at least ALK5. Therefore, in some embodiments, a polynucleotide inhibitor of ALK5 binds to ALK5. In some embodiments, a polynucleotide inhibitor of ALK5 inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of ALK5. In some embodiments, a polynucleotide inhibitor of ALK5 further inhibits one or more of TORbI, TGFp2, TGFp3, TpRII, and betaglycan. In some embodiments, a polynucleotide TpRII antagonist inhibits TOR ' bI from binding to ALK5. In some embodiments, a polynucleotide TpRII antagonist inhibits TORb2 from binding to ALK5. In some embodiments, a polynucleotide TpRII antagonist inhibits TGFp3 from binding to ALK5, In some embodiments, a polynucleotide TpRII antagonist inhibits TGFpl and TGFp3 from binding to ALK5. In some embodiments, a polynucleotide TpRII antagonist inhibits TGFpl, TORb2, and TORb3 from binding to ALK5. In some embodiments, a polynucleotide TpRII antagonist inhibits TGFp I from binding to ALK5 but does not inhibit or does not substantially inhibit TORb2 from binding to ALK5. In some embodiments, a polynucleotide TpRII antagonist inhibits TORb3 from binding to ALK5 but does not inhibit or does not substantially inhibit TGFp2 from binding to ALK5. In some embodiments, a polynucleotide TpRff antagonist inhibits TORbI and TGFp3 from binding to ALK5 but does not inhibit or does not substantially inhibit TGFp2 from binding to ALK5. In certain aspects, a polynucleotide TbKII antagonist inhibits at least betagiycan. Therefore, in some embodiments, a polynucleotide inhibitor of betagiycan binds to betagiycan. In some embodiments, a polynucleotide inhibitor of betagiycan inhibits expression (e.g., transcription, translation, secretion, or combinations thereof) of betagiycan. In some embodiments, a polynucleotide inhibitor of betagiycan further inhibits one or more of TGFpl, TORb2, TORb3, TbKII, and ALK5. In some embodiments, a polynucleotide TbKII antagonist inhibits TGFpi from binding to betagiycan. In some embodiments, a polynucleotide TbKII antagonist inhibits TOTb2 from binding to betagiycan. In some embodiments, a polynucleotide Tb R 11 antagonist inhibits TORb3 from binding to betagiycan. In some embodiments, a polynucleotide TbKII antagonist inhibits TGFpl and TORb3 from binding to betagiycan. In some embodiments, a polynucleotide TbKII antagonist inhibits TORbI, TOTb2, and TORb3 from binding to betagiycan. In some embodiments, a polynucleotide TpRIl antagonist inhibits TORbI from binding to betagiycan but does not inhibit or does not substantially inhibit TGFp2 from binding to betagiycan. In some embodiments, a polynucleotide TpRli antagonist inhibits TORb3 from binding to betagiycan but does not inhibit or does not substantially inhibit TGFp2 from binding to betagiycan. In some embodiments, a polynucleotide TbKII antagonist inhibits TGFfll and TORb3 from binding to betagiycan but does not inhibit or does not substantially inhibit TORb2 from binding to betagiycan.

The polynucleotide antagonists of the present disclosure may be an antisense nucleic acid, an RNAi molecule [e.g., small interfering RNA (siRNA), small-hairpin RNA (shRNA), microRNA (miRNA)], an aptanier and/or a ribozynie. The nucleic acid and amino acid sequences of human TbEII, ALK5, betagiycan, TORbI, TGF[)2, and TGFp3 are known in the art and thus polynucleotide antagonists for use m accordance with methods of the present disclosure may be routinely made by the skilled artisan based on the knowledge in the art and teachings provided herein.

For example, antisense technology can be used to control gene expression through antisense DNA or RNA, or through triple-helix formation. Antisense techniques are discussed, for example, in Okano (1991) J. Neurochem. 56:560; Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988). Triple helix formation is discussed in, for instance, Cooney etal. (1988) Science 241:456; and Dervan et al., (1991)Science 251 : 1300. The methods are based on binding of a polynucleotide to a complementary DMA or RNA. In some embodiments, tire antisense nucleic acids comprise a single-stranded RNA or DNA sequence that is complementary to at least a portion of an RNA transcript of a desired gene. However, absolute complementarity, although preferred, is not required.

A sequence "complementary to at least a portion of an RNA," referred to herein, means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; m the case of double -stranded antisense nucleic acids of a gene disclosed herein, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed. The ability' to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid. Generally, the larger the hybridizing nucleic acid, the more base mismatches with an RNA it may contain, and still form a stable duplex (or triplex as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex. Polynucleotides that are complementary to the 5' end of the message, for example, the

5 '-untranslated sequence up to and including the AUG initiation codon, should work most efficiently at inhibiting translation. However, sequences complementary to the 3'- untranslated sequences of mRNAs have been shown to be effective at inhibiting translation of mRNAs as well [see, e.g., Wagner, R., (1994) Nature 372:333-335], Thus, oligonucleotides complementary to either the 5'- or 3 '-untranslated, noncoding regions of a gene of the disclosure, could be used in an antisense approach to inhibit translation of an endogenous mRNA. Polynucleotides complementary to the 5 '-untranslated region of the mRNA should include the complement of the AUG start codon. Antisense polynucleotides complementary to mRNA coding regions are less efficient inhibitors of translation but could be used in accordance with the methods of tire present disclosure. Whether designed to hybridize to the 5 '-untranslated, 3 '-untranslated, or coding regions of an mRNA of the disclosure, antisense nucleic acids should be at least six nucleotides in length, and are preferably oligonucleotides ranging from 6 to about 50 nucleotides in length. In specific aspects, the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides, or at least 50 nucleotides. In one embodiment, the antisense nucleic acid of the present disclosure is produced intraceliularly by transcription from an exogenous sequence. For example, a vector or a portion thereof, is transcribed, producing an antisense nucleic acid (RNA) of a gene of the disclosure. Such a vector would contain a sequence encoding the desired antisense nucleic acid. Such a vector can remain episomai or become chromosomaily integrated, as long as it can be transcribed to produce the desired antisense RNA. Such vectors can be constructed by recombinant DNA technology methods standard in the art. Vectors can be plasmid, viral, or others known in the art, used for replication and expression in vertebrate cells. Expression of the sequence encoding desired genes of the instant disclosure, or fragments thereof, can be by any promoter known in the art to act in vertebrate, preferably human cells. Such promoters can be inducible or constitutive. Such promoters include, but are not limited to, the SV40 early promoter region [see, e.g., Benoist and Chambon (1981) Nature 29:304-310], the promoter contained in the 3' long terminal repeat of Rous sarcoma virus [see, e.g., Yamamoto et al. (1980) Cell 22:787-797], the herpes thymidine promoter [see, e.g., Wagner etal. (1981) Proc. Natl. Acad. Sci. U.S.A. 78: 1441-1445], and the regulatory' sequences of the metallothionein gene [see, e.g., Brinster, et al. (1982) Nature 296:39-42] .

In some embodiments, the polynucleotide antagonists are interfering KNA or RNAi molecules that target the expression of one or more genes. RNAi refers to the expression of an RNA which interferes with the expression of the targeted mRNA. Specifically, RNAi silences a targeted gene via interacting with the specific mRN A through a siRNA (small interfering RNA). The ds RNA complex is then targeted for degradation by the cell. An siRNA molecule is a double -stranded RNA duplex of 10 to 50 nucleotides in length, which interferes with the expression of a target gene which is sufficiently complementary {e.g. at least 80% identity' to the gene). In some embodiments, the siRNA molecule comprises a nucleotide sequence that is at least 85, 90, 95, 96, 97, 98, 99, or 100% identical to the nucleotide sequence of the target gene.

Additional RNAi molecules include short-hairpin RNA (shRNA); also short- interfering hairpin and micro RNA (miRNA). The shRNA molecule contains sense and antisense sequences from a target gene connected by a loop. The shRNA is transported from the nucleus into the cytoplasm, and it is degraded along with the mRNA. Pol III or U6 promoters can be used to express RNAs for RNAi. Paddison et al. [Genes & Dev, (2002) 16:948-958, 2002] have used small RNA molecules folded into hairpins as a means to effect RNAi. Accordingly, such short hairpin RNA (shRNA) molecules are also advantageously used in the methods described herein. The length of the stem and loop of functional shRNAs varies; stem lengths can range anywhere from about 25 to about 30 nt, and loop size can range between 4 to about 25 nt without affecting silencing activity. While not wishing to be bound by any particular theory, it is believed that these shRNAs resemble the double- stranded RNA (dsRNA) products of the DICER RNase and, in any event, have the same capacity for inhibiting expression of a specific gene. The shRNA can be expressed from a ientiviral vector. An miRNA is a single-stranded RNA of about 10 to 70 nucleotides in length that are initially transcribed as pre-miRNA characterized by a ‘"stem-loop” structure and which are subsequently processed into mature miRNA after further processing through the RISC.

Molecules that mediate RNAi, including without limitation siRNA, can be produced in vitro by chemical synthesis (Hohjoh, FEES Lett 521:195-199, 2002), hydrolysis of dsRNA (Yang etaL, Proc Natl Acad Sci USA 99:9942-9947, 2002), by in vitro transcription with T7 RNA polymerase (Donzeet ei al Nucleic Acids Res 30:e46, 2002; Yu et at., Proc Natl Acad Sci USA 99:6047-6052, 2002), and by hydrolysis of double-stranded RNA using a nuclease such as E. cob RNase 111 (Yang etaL, Proc Natl Acad Sci USA 99:9942-9947, 2002).

According to another aspect, the disclosure provides polynucleotide antagonists including but not limited to, a decoy DNA, a double-stranded DNA, a single-stranded DNA, a complexed DNA, an encapsulated DNA, a viral DNA, a plasmid DNA, a naked RNA, an encapsulated RNA, a viral RNA, a double-stranded RNA, a molecule capable of generating RNA interference, or combinations thereof.

In some embodiments, the polynucleotide antagonists of the disclosure are aptamers. Aptamers are nucleic acid molecules, including double-stranded DNA and single-stranded RNA molecules, which bind to and form tertiary' structures that specifically bind to a target molecule, such as a TpRII, betaglycan, TGFii I . TGF[¾2, and TORb3 polypeptide. The generation and therapeutic use of aptamers are well established in the art. See, e.g., U.S. Pat. No. 5,475,096, Additional information on aptamers can be found in U.S, Patent Application Publication No. 20060148748. Nucleic acid aptamers are selected using methods known in the art, for example via the Systematic Evolution of Ligands by Exponential Enrichment (SELEX) process, SELEX is a method for the in vitro evolution of nucleic acid molecules with highly specific binding to target molecules as described in, e.g,, U.S, Pat. Nos. 5,475,096, 5,580,737, 5,567,588, 5,707,796, 5,763,177, 6,011,577, and 6,699,843. Another screening method to identify aptamers is described in U.S. Pat. No. 5,270,163. The SELEX process is based on the capacity of nucleic acids for forming a variety ' of two- and three- dimensional structures, as well as the chemical versatility available within the nucleotide monomers to act as ligands (form specific binding pairs) with virtually any chemical compound, whether monomeric or polymeric, including other nucleic acid molecules and polypeptides. Molecules of any size or composition can sen/e as targets. The SELEX method involves selection from a mixture of candidate oligonucleotides and step-wise iterations of binding, partitioning and amplification, using the same general selection scheme, to achieve desired binding affinity and selectivity. Starting from a mixture of nucleic acids, which can comprise a segment of randomized sequence, the SELEX method includes steps of contacting the mixture with the target under conditions favorable for binding; partitioning unbound nucleic acids from those nucleic acids which have bound specifically to target molecules; dissociating the nucleic acid-target complexes; amplifying the nucleic acids dissociated from the nucleic acid-target complexes to yield a ligand enriched mixture of nucleic acids. The steps of binding, partitioning, dissociating and amplifying are repeated through as many cycles as desired to yield highly specific high affinity nucleic acid ligands to the target molecule.

Typically, such binding molecules are separately administered to the animal [see, e.g., O'Connor (1991) J. Neurochem. 56:560], but such binding molecules can also be expressed in vivo from polynucleotides taken up by a host cell and expressed in vivo [see, e.g. ,

Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression, CRC Press, Boca Raton, Fla. (1988)].

12. Screening Assays In certain aspects, the present invention relates to the use of ' TbKII polypeptides (e.g., soluble TpRH polypeptides) to identify 7 compounds (agents) which are agonist or antagonists of the TGFpi, TORb3 and TiiRlf signaling pathway. Compounds identified through this screening can be tested to assess their ability to modulate TGFp 1 and TGFp3 signaling activity in vitro. Specifically, compounds identified through this screening can be tested to assess their ability to treat SSc, including clinical complications of SSc (e.g., SSc-ILD), in a subject in need thereof. Accordingly, these compounds can further be tested in animal models to assess their ability to treat SSc, including complications of SSc (e.g., SSc-ILD).

There are numerous approaches to screening for therapeutic agents for treating SSc, including complications of SSc (e.g., SSc-ILD), by targeting TORbI, TGFp3 and TbKII polypeptides. In certain embodiments, high-throughput screening of compounds can be carried out to identify’ agents that perturb TGFpi.. TGPpS or TpRII-mediated cell signaling.

In certain embodiments, the assay is earned out to screen and identify compounds that specifically inhibit or reduce binding of a TbKίί polypeptide to TGFpi or TGFp3. Altematively, the assay can be used to identify compounds that enhance binding of a TbίIίί polypeptide to TGFpl or TGFfB. in a further embodiment, the compounds can be identified by their ability to interact with a TG!- ' bί.. TORb3 or TbKII polypeptide.

A variety of assay formats will suffice, and, in light of the present disclosure, those not expressly described herein will nevertheless be comprehended by one of ordinary skill in the art. As described herein, the test compounds (agents) of the in vention may be created by any combinatorial chemical method. Alternatively, the subject compounds may be naturally occurring biomolecules synthesized in vivo or in vitro. Compounds (agents) to be tested for their ability to act as therapeutic agents to treat SSc, including complications of SSc (e.g., SSc-ILD), can be produced, for example, by bacteria, yeast, plants or other organisms (e.g., natural products), produced chemically (e.g,, small molecules, including peptidomimetics), or produced recombinantly. Test compounds contemplated by the present invention include non-peptidyl organic molecules, peptides, polypeptides, peptidomimetics, sugars, hormones, and nucleic acid molecules. In a specific embodiment, the test agent is a small organic molecule having a molecular weight of less than about 2,000 daltons.

The test compounds of the invention can be provided as single, discrete entities, or provided in libraries of greater complexity-, such as made by combinatorial chemistry. These libraries can comprise, for example, alcohols, alkyl halides, amines, amides, esters, aldehydes, ethers and other classes of organic compounds. Presentation of test compounds to the test system can be in either an isolated form or as mixtures of compounds, especially in initial screening steps. Optionally, the compounds may be optionally derivatized with other compounds and have derivatizing groups that facilitate isolation of the compounds. Nonlimiting examples of derivatizing groups include biotin, fluorescein, digoxygenin, green fluorescent protein, isotopes, polyhistidine, magnetic beads, glutathione S transferase (GST), photoactivatable crosslinkers or any combinations thereof.

In many drug screening programs, which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with purified or semi-purified proteins, are often preferred as “primary'” screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound. Moreover, the effects of cellular toxicity or bioavailability ' of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity between a TbKII polypeptide and TGFpl or TGFfB.

Merely to illustrate, in an exemplary' screening assay of the present invention, the compound of interest is contacted with an isolated and purified TpRII polypeptide which is ordinarily capable of binding to TG Gb i or TORb3. To the mixture of the compound and TbKH polypeptide is then added a composition containing a TbKIί ligand. Detection and quantification oίTbKP/TORbί or TbKII7TORb3 complexes provides a means for determining the compound's efficacy at inhibiting (or potentiating) complex formation between the TbIIII polypeptide and TORbI or TGF(33. The efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound. Moreover, a control assay can also be performed to provide a baseline for comparison. For example, in a control assay, isolated and a purified TORbI or TORb3 is added to a composition containing the TfiRl! polypeptide, and the formation of TbKII/TORbI or TbKII/TORb3 complex is quantitated in the absence of the test compound. It will be understood that, in general, the order in which the reactants may be admixed can be varied, and can be admixed simultaneously. Moreover, in place of purified proteins, cellular extracts and lysates may be used to render a suitable cell-free assay system.

Complex formation between the TbKII polypeptide and TORb I or TORb3 may be detected by a variety of techniques. For instance, modulation of the formation of complexes can be quantitated using, for example, detectably labeled proteins such as radiolabeled (e.g., 32 P, 35 S, 14 C or 3 H), fluorescently labeled (e.g., FITC), or enzymatically labeled TbKIί polypeptide or TORb! or TORb3, by immunoassay, or by chromatographic detection.

In certain embodiments, the present invention contemplates the use of fluorescence polarization assays and fluorescence resonance energy transfer (FRET) assays m measuring, either directly or indirectly, the degree of interaction between a TbKP polypeptide and its binding protein. Further, other modes of detection, such as those based on optical waveguides (PCT Publication WO 96/26432 and U.S. Pat. No. 5,677,196), surface plasmon resonance (SPR), surface charge sensors, and surface force sensors, are compatible with many embodiments of the invention. Moreover, the present invention contemplates the use of an interaction trap assay, also known as the “two hybrid assay, ’ for identifying agents that disrupt or potentiate interaction between a TbKIΊ polypeptide and its binding protein. See for example, U.S. Pat. No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) I Biol Chem 268:12046-12054; Bartel et al. (1993) Biotechniques 14:920-924; and Iwabuchi et al. (1993) Oncogene 8: 1693-1696). In a specific embodiment, the present invention contemplates the use of reverse two hybrid systems to identify compounds (e.g., small molecules or peptides) that dissociate interactions between a TbMI polypeptide and its binding protein. See for example, Vidal and Legrain, (1999) Nucleic Acids Res 27:919-29; Vida! and Legrain, (1999) Trends Biotechnol 17:374-81; and U.S. Pat. Nos. 5,525,490; 5,955,280; and 5,965,368.

In certain embodiments, the subject compounds are identified by their ability to interact w 7 ith a TfiRii or TGFpl or TORb3 polypeptide of the invention. The interaction between the compound and the TbKII or TORbI or TORb3 polypeptide may be covalent or non-covalent. For example, such interaction can be identified at the protein level using in vitro biochemical methods, including photo-crosslinking, radiolabeled ligand binding, and affinity chromatography (JaJkoby WB et al., 1974, Methods in Enzymoiogy 46: 1). In certain cases, the compounds may be screened in a mechanism based assay, such as an assay to detect compounds which bind to a TGFpl or TGFf)3 or TbKII polypeptide. This may include a solid-phase or fluid-phase binding event. Alternatively, the gene encoding a TΌRbI or TORb3 or TbR.P polypeptide can be transfected with a reporter system (e.g., b-galactosidase, luciferase, or green fluorescent protein) into a ceil and screened against the library preferably by a high-throughput screening or with individual members of the library'. Other mechanism- based binding assays may be used, for example, binding assays which detect changes in free energy. Binding assays can be performed with the target fixed to a well, bead or chip or captured by an immobilized antibody or resolved by capillary electrophoresis. Hie bound compounds may be detected usually using colorimetric or fluorescence or surface plasmon resonance.

In certain aspects, the present invention provides methods and agents for modulating (stimulating or inhibiting) TGF[31- or TΌRb3 -mediated cell signaling. Therefore, any compound identified can be tested in whole cells or tissues, in vitro or in vivo, to confirm their ability' to modulate TORbI or T(}Rb3 signaling. Various methods known in the art can be utilized for this purpose.

13. Systemic Sclerosis associated Complications In part, the disclosure relates to TpRli antagonists that can be used to treat systemic sclerosis (SSc), particularly clinical complications of SSc including, for example, interstitial lung disease (ILD). Accordingly, the disclosure provides methods for treating SSc, or a complication thereof (e.g,, SSc-ILD) comprising administering to a patient in need thereof one or more TpRI! antagonist. In some embodiments, the disclosure provides methods of treating diffuse systemic sclerosis. In some embodiments, the disclosure provides methods of treating limited systemic sclerosis. Optionally, such methods further comprise administering to the patient one or more additional active agents and/or supportive therapies for treating SSc or a complication of SSc (e.g., SSc-ILD). The terms "treatment", "treating", “alleviation” and the like are used herein to generally mean obtaining a desired pharmacologic and/or physiologic effect, and may also be used to refer to improving, alleviating, and/or decreasing the severity of one or more clinical complication of a condition being treated. The effect may be prophylactic in terms of completely or partially delaying the onset or recurrence of a disease, condition, or complications thereof, and/or may be therapeutic in terms of a partial or complete cure for a disease or condition and/or adverse effect atributable to the disease or condition.

"Treatment" as used herein covers any treatment of a disease or condition of a mammal, particularly a human. As used herein, a therapeutic that “prevents” a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in a treated sample relative to an untreated control sample, or delays the onset of the disease or condition, relative to an untreated control sample.

The terms “patient”, “subject”, or “individual” are used interchangeably herein and refer to either a human or a non-human animal. These terms include mammals, such as humans, non-human primates, laboratory animals, livestock animals (including bovines, porcines, camels, etc.), companion animals (e.g., canines, felines, other domesticated animals, etc.) and rodents (e.g., mice and rats). In particular embodiments, the patient, subject or individual is a human.

Overview of Systemic Sclerosis Systemic sclerosis (SSc) is a chronic autoimmune disease usually marked by abnormal growth of fibrous connective tissue in skin and often in the internal organs. There are approximately 5,000 to 10,000 cases of SSc diagnosed every year in the United States, with an annual incidence of approximately 20 to 24 new eases per 1 million adults. Globally, the overall incidence rates range from 8 to 56 new cases per I million persons per year, and the prevalence rates fall between 38 and 341 cases per 1 million persons per year. SSc is typically characterized by immune dysfunction, vasculopathy, cellular inflammation, and fibrosis of the skin and internal organs.

The disclosure provides methods of treating or preventing a disease or condition associated with a TΌRb superfamily member by administering to a subject one or more TbKH antagonists, including a TbKII polypeptides and fusion proteins comprising the same as described herein. In some embodiments, the disease or condition is associated with dysregulated TGPpi or TGFp3 signaling. In some embodiments, the disease or condition to be treated is systemic sclerosis. In some embodiments, a TbMI antagonist for use in treating systemic sclerosis is administered to a subject in need thereof. In some embodiments, systemic sclerosis is characterized as diffuse cutaneous systemic sclerosis (dcSSc) (e.g., diffuse systemic sclerosis). In some embodiments, systemic sclerosis is characterized as limited cutaneous systemic sclerosis (IcSSc) (eg., limited systemic sclerosis). In some embodiments the disease or condition to be treated is diffuse systemic sclerosis. In some embodiments, a TpRII antagonist for use in treating diffuse systemic sclerosis is administered to a subject in need thereof. In some embodiments, the disease or condition to be treated is limited systemic sclerosis. In some embodiments, a TpRII antagonist for use in treating limited systemic sclerosis is administered to a subject in need thereof In some embodiments, the disease or condition to be treated is systemic sclerosis associated with interstitial lung disease (SSc-ILD). In some embodiments, a TbKII antagonist for use in treating SSc-ILD is administered to a subject in need thereof. In some embodiments, the disclosure provides a method of treating SSc-ILD, comprising administering a TbKII antagonist to a subject in need thereof In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a TpRH antagonist to a subject in need thereof, wherein the systemic sclerosis is diffuse systemic sclerosis.

In some embodiments, the disclosure provides methods of treating SSc (eg., SSc- ILD), comprising administering a TbKP antagonist to a subject in need thereof, wherein the treatment increases the subject’s length of life when compared to a reference subject that is not receiving treatment. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the treatment reduces the subject’s risk of death due to SSc (e.g., SSc-ILD) when compared to a reference subject that is not receiving treatment. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the treatment reduces the subject’s risk of hospitalization due to SSc (e.g., SSc-ILD) when compared to a reference subject that is not receiving treatment. In some embodiments, a reference subject that is not receiving treatment has similar characteristics to the subject (height, sex, age, race, weight) and is not receiving one or more of TpRII antagonists of the present disclosure. In some embodiments, a subject of the present disclosure with SSc-ILD will have one or more additional complications of SSc. A common feature of SSc comprises presence of skin thickening in a subject, though there are subjects with SSc that have no detectable skin involvement (e.g,, systemic sclerosis sine scleroderma). Subjects with systemic sclerosis sme scleroderma typically have vascular and/or fibrotic features of systemic disease. In some embodiments, a subject with SSc (e.g., SSc-ILD) has no observed clinical complications of the skin. In some embodiments, a subject has no observed clinical complication of the skin but does have presence of one or more of Raynaud’s phenomenon, nailfold capillary' alterations, gastrointestinal involvement, renal crisis, PAH, and/or ILL). In some embodiments, a subject has systemic sclerosis sine scleroderma. In some embodiments, the disease or condition to he treated with one or more TjlRII antagonists of the present disclosure is systemic sclerosis sine scleroderma.

In some embodiments, a subject with SSc (e.g., SSc-ILD) has one or more clinical complications of the skin. In some embodiments, a clinical complication of the skin of a subject with SSc is selected from the group consisting of calcinosis cutis, capillary ' changes at the nail beds, depigmentation, digital tip ulcers and/or pitting at fingertips, dryness, edema, hyperpigmentation, lipoatrophy, loss of appendicular hair, pruritus, telangiectasia, and traumatic skm ulcerations over finger joints. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a TbKII antagonist to a subject in need thereof, wherein the subject has one or more clinical complications of the skin selected from the group consisting of calcinosis cutis, capillary ' changes at the nail beds, depigmentation, digital tip ulcers and/or pitting at fingertips, dryness, edema, hyperpigmentation, lipoatrophy, loss of appendicular hair, pruritus, telangiectasia, and traumatic skm ulcerations over finger joints. Subjects with clinical complications of the skin are divided into two major classifications of systemic sclerosis, which are defined by areas and extensiveness of the skin involved. Limited cutaneous systemic sclerosis (lcSSc), or limited systemic sclerosis, is typically limited to hands, forearms, feet, and the face: while diffuse cutaneous systemic sclerosis (dcSSc), or diffuse systemic sclerosis, extends beyond these regions of the body. In some embodiments, a subject has limited systemic sclerosis (lcSSc). In some embodiments, the disease or condition to be treated is limited systemic sclerosis (lcSSc). In some embodiments, a subject has diffuse systemic sclerosis (dcSSc). In some embodiments, the disease or condition to be treated is diffuse systemic sclerosis (dcSSc). In some embodiments, a subject has diffuse systemic sclerosis (dcSSc) without interstitial lung disease (ILL)). In some embodiments, the disease or condition to be treated is diffuse systemic sclerosis (dcSSc) without ILD. In some embodiments, a subject has diffuse systemic sclerosis (dcSSc) with interstitial lung disease (ILD). In some embodiments, tire disease or condition to be treated is diffuse systemic sclerosis (deSSe) with ILD.

In some embodiments, a subject with SSc (e.g,, SSc-ILD) may have features of one or more other systemic diseases. In some embodiments, a subject with SSc may have features of systemic lupus erythematosus (SLE), rheumatoid arthritis, polymyositis, and/or Sjogren’s syndrome. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc- ILD) comprising administering a Tb R 11 antagonist to a subject in need thereof, wherein the subject has one or more features of other systemic diseases selected from the group consisting of systemic lupus erythematosus (SLE), rheumatoid arthritis, polymyositis, and Sjogren’s syndrome. In some embodiments, a subject has systemic sclerosis with overlap syndrome.

In some embodiments, the disease or condition to be treated is systemic sclerosis with overlap syndrome.

‘The 2013 Classification Criteria” or “the Criteria”, developed by the American College of Rheumatology (ACR) and the European League Against Rheumatism (EULAR), may be implemented to identify patients with SSc (e.g., SSc-ILD). In some embodiments, the disclosure provides methods of treating a subject that has SSc (e.g., SSc-ILD) according to the EULAR criteria. The Criteria comprises three hallmarks of SSc: fibrosis of the skin and/or other internal organs, production of specific autoantibodies (e.g., antinuclear antibody (ANA), anticentromere, anti-topoisomerase I, anti-RNA polymerase III), and/or evidence of vascuiopafhy. Presence of skin thickening of fingers extending proximal to metacarpophalangeal joints is sufficient for a patient to be classified as having SSc (e.g., SSc- ILD). If the Criteria are not present, additive items with varying weights may be used to identify SSc (e.g., SSc-ILD), including skin thickening of the fingers, fingertip lesions, telangiectasia, Raynaud’s phenomenon, abnormal nailfold capillaroscopy, and/or presence of pulmonary arterial hypertension (PAH) and/or interstitial lung disease (ILD). In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a ' TbKII antagonist to a subject in need thereof, wherein the subject has one or more of fibrosis of the skin and/or other internal organs, production of specific autoantibodies (e.g., antinuclear antibody (ANA), anticentromere, anti-topoisomerase I, anti-RNA polymerase III), and/or evidence of vasculopathy. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering hTbKP antagonist to a subject in need thereof, wherein the subject has one or more of skin thickening of the fingers, fingertip lesions, telangiectasia, Raynaud’s phenomenon, abnormal nailfold capiliaroscopy, and/or presence of pulmonary arterial hypertension (PAH) and/or interstitial lung disease (1LD).

Other clinical complications of SSc (e.g., SSc-ILD) comprise heartburn, erectile dysfunction in men, and/or dyspnea, which is generally defined as dyspnea on exertion associated with restrictive change sin pulmonary function, evidence of pulmonary changes on radiography or HRCT scans, and/or dyspnea on exertion associate with evidence of PAH on Dopper echocardiography. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a TilRII antagonist to a subject in need thereof, wherein the sub j ect has one or more of heartburn, erectile dysfunction, and/or dyspnea.

Vasculopathy includes Raynaud’s phenomenon and/or ischemic digital ulcers, which are both common in subjects with SSc (e.g., SSc-ILD). Raynaud’s phenomenon is classically viewed as re versible vasospasm due to functional changes in the digital arteries of the hands and/or feet. Over time, many subjects with SSc (e.g., SSc-ILD) develop progressive structural changes in small blood vessels, with permanently impaired blood flow. In some embodiments, a subject has Raynaud’s syndrome. In some embodiments, a subject has structural changes in the small blood vessels. In some embodiments, a subject with Raynaud’s phenomenon has one or more of ischemic pain, digital ulceration, tophic changes, refractory or progressive ischemia and/or infarction. Early occurrence of digital ulcers can indicate diffuse systemic sclerosis and/or positive detection of antitopoisomerase I.

Approximately 80% of subjects diagnosed with SSc (e.g., SSc-ILD) will eventually develop some degree of a complication in the lungs. Examples of major types of lung complications associated with SSc (e.g., SSc-ILD) include alveolitis, interstitial pulmonary- fibrosis or interstitial lung disease (ILD), recurrent aspiration, and/or pulmonary vasculopathy. In some embodiments, a subject with SSc (e.g., SSc-ILD) has one or more additional lung complications, in some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a TjlRH antagonist to a subject m need thereof, wherein the subject has one or more additional lung complications selected from the group consisting of alveolitis, recurrent aspiration, and/or pulmonary vasculopathy. In subjects with diffuse SSc (e.g., SSc-ILD) there is a high risk of having an inflammatory pulmonary process that resembles nonspecific interstitial pneumonitis and/or the early onset of interstitial fibrosis. Interstitial lung disease (ILD) is a leading cause of morbidity and/or mortality in subjects with SSc (e.g,, SSc-ILD). Subjects with dcSSc are more likely to have rapid progression of skin thickening and/or early development of lung fibrosis, as well as an increased risk of renal and/or cardiac involvement. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a lj!RII antagonist to a subject in need thereof, wherein the subject has rapid progression of skin thickening and/or early development of lung fibrosis, as well as an increased risk of renal and/or cardiac involvement.

Risk factors that can indicate a possibly higher likelihood of developing ILL) in SSc subjects includes, but is not limited to, presence of diffuse SSc, African-American ethnicity, older age at disease onset, and/or presence of SSc-related autoantibodies. However, all subjects suspected of having SSc (e.g., SSc-ILD) typically receive a comprehensive assessment which includes, but is not limited to, a physical assessment, assessment of respiratory' symptoms, chest imaging with high resolution computed tomography (HRCT), and/or pulmonary function tests including spirometry.

Clinical complications that are most common in SSc-ILD are fatigue, breathlessness (e.g., exertional dyspnea), and/or dry cough. One of the most characteristic complications of interstitial lung disease (ILD) comprises hibasilar fine inspiratory crackles (e.g., “Velcro” rales). In some embodiments, a subject with SSc-ILD has bibasilar fine inspiratory crackles (e.g., “Velcro” rales) upon physical examination. In some embodiments, a subject has one or more symptoms including fatigue, breathlessness (e.g,, exertional dyspnea), and/or dry cough. In some embodiments, a subject with SSc is suspected to have SSc-ILD when presenting with complications comprising one or more of dyspnea, cough, auscultatory crackles, and/or abnormalities on pulmonary function tests and/or a chest radiograph. In some embodiments, the disclosure provides methods of treating SSc-ILD comprising administering a TpRII antagonist to a subject in need thereof, wherein the subject has one or more clinical complications of ILD including fatigue, breathlessness, dry cough, and/or bibasilar fine respiratory crackles.

A subset of subjects with SSc (e.g., SSc-ILD) can develop rapidly progressive ILD during the first few years (e.g., approximately 2 years) of their diagnosis of ILD. This subset typically presents ground-glass opacities on high resolution computed tomography (HRCT), a neutrophilic or eosinophilic bronchioa!veolar lavage (BAL), and/or declining spirometry (e.g., FVC) or diffusion capacity (DLCO) on pulmonary function tests (PFTs). Forced vital capacity (FVC) correlates well with tidal volumes and/or other resting lung volumes in ILD, which are typically reduced. Among static lung volume tests, FVC is reduced to a greater extent than the functional residual capacity. In some embodiments, the disclosure provides methods of treating SSc-ILD comprising administering a TpRII antagonist to a subject in need thereof, wherein the subject has one or more of ground glass opacities on HRCT, a neutrophilic or eosinophilic BAL, and/or declining FVC and/or DLCO scores.

Pulmonary vascular disease, namely Pulmonary Arterial Hypertension (PAH), can occur in a subject with SSc (e.g., SSc-ILD). Generally, PAH is a late complication of SSc (e.g., SSc-ILD). Approximately 10-40% of subjects with SSc (e.g., SSc-ILD) will develop

PAH, and/or PAH is typically more commonly found in IcSSc subjects. PAH can occur with or without the presence of ILD in the subject. In some embodiments, a subject that has SSc- ILD does not have PAH. In some embodiments, a subject that has SSc-ILD has PAH. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a TbKII antagonist to a subject in need thereof, wherein the subject also has PAH.

Other major contributors to a decrease in mortality in SSc (e.g., SSc-ILD) comprise arrythmia, kidney damage (scleroderma renal crisis), increased risk of cancers, myocardial disease (e.g., myocardial fibrosis, myocardial ischemia), muscle atrophy (e.g., sarcopenia), muscle weakness, myopathy, pericardial disease, and/or thromboembolic risk. In some embodiments, a subject with SSc (e.g., SSc-ILD) also has one or more of arrythmia, kidney damage (scleroderma renal crisis), increased risk of cancers, myocardial disease (e.g., myocardial fibrosis, myocardial ischemia), muscle atrophy (e.g., sarcopenia), muscle weakness, myopathy, pericardial disease, and/or thromboembolic risk. In some embodiments, the disclosure provides methods of treating SSc (e.g., SSc-ILD) comprising administering a TbϊϋI antagonist to a subject in need thereof, wherein the subject has one or more of arrythmia, kidney damage (scleroderma renal crisis), increased risk of cancers, myocardial disease (e.g., myocardial fibrosis, myocardial ischemia), muscle atrophy (e.g., sarcopenia), muscle weakness, myopathy, pericardial disease, and/or thromboembolic risk. Aside from vasculopathy and/or pulmonary' complications, other clinical complications can be present in a subject with SSc(e.g., SSc-ILD). Musculoskeletal complications, gastrointestinal complications, cardiac complications, renal complications, neuromuscular complications (e.g., muscle atrophy, muscle weakness, myopathy), genitourinary complications (e.g., erectile dysfunction in men), increased risk of cancer (e.g., lung cancer), and/or increased risk of thromboembolic complications are possible, among other complications. In some embodiments, a subject has one or more clinical complications of SSc (e.g., SSc-ILD) selected from the group consisting of musculoskeletal complications, gastrointestinal complications, cardiac complications, renal complications, neuromuscular complications (e.g., muscle atrophy, muscle weakness, myopathy), genitourinary complications (e.g., erectile dysfunction in men), increased risk of cancer (e.g., lung cancer), and/or increased risk of thromboembolic complications. Musculoskeletal complications can include, but are not limited to arthritis, tendinitis, tendon friction rubs, and/or joint contractures. In some embodiments, a subject with SSc (e.g., SSc-ILD) has one or more musculoskeletal complications selected from the group consisting of arthritis, tendinitis, tendon friction mbs, and joint contractures. For subjects with dcSSc specifically, early musculoskeletal complications can include, but are not limited to, swelling of the hands, arthralgia, myalgia, and/or fatigue. In some embodiments, a subject with SSc (e.g., SSc-ILD) also has one or more of swelling of the hands, arthralgia, myalgia, and/or fatigue. Gastrointestinal complications can include, but are not limited to, dysphagia, choking, heartburn, hoarseness, cough after swallowing, early satiety', bloating, alternating constipation and/or diarrhea, episodic pseudo-obstruction and/or bacterial small bowel overgrowth with malabsorption, fecal incontinence, chronic gastroesophageal reflux, and/or recurrent episodes of microaspiration. In some embodiments, a subject with SSc (e.g., SSc-ILD) has one or more of gastrointestinal complications selected from the group consisting of dysphagia, choking, heartburn, hoarseness, cough after swallowing, early satiety, bloating, alternating constipation and/or diarrhea, episodic pseudo-obstruction and/or bacterial small bowel overgrowth with malabsorption, fecal incontinence, chronic gastroesophageal reflux, and/or recurrent episodes of microaspiration. Chronic gastroesophageal reflux and/or recurrent episodes of microaspiration may contribute to development of 1LD in subjects with SSc (e.g., SSc-ILD). In some embodiments, a subject with SSC (e.g., SSc-ILD) also has one or more of chronic gastroesophageal reflux and/or recurrent episodes of microaspiration. Cardiac complications can include all domains of the heart, including myocardium, pericardium, and/or conduction system. In some embodiments, a subject with SSc (e.g., SSc-ILD) has one or more cardiac complications selected from the group consisting of complications in myocardium, pericardium, and conduction system. Cardiac complications can occur secondary to PAH and/or ILD or scleroderma renal crisis (SRC). In some embodiments, a subject with SSc (e.g., SSc-ILD) also has pericardial disease. In some embodiments, a subject with SSc (e.g,, SSc-ILD) has myocardial disease. In some embodiments, a subject with SSc (e.g., SSe-ILD) also has arrythmia. Renal complications can include, but are not limited to, kidney damage, vascular fibrosis of the kidney, interstitial collagen accumulation of the kidney, glomerulonephritis, impaired renal reserve, microalbuminuria, and/or scleroderma renal crisis (SRC). In some embodiments, a subject with SSc (e.g., SSc-ILD) has one or more renal complications selected from the group consisting of kidney damage, vascular fibrosis of the kidney, interstitial collagen accumulation of the kidney, glomerulonephritis, impaired renal reserve, microalbuminuria, and SRC. In some embodiments, a subject with SSc (e.g., SSc-ILD) also has SRC. In some embodiments, a subject with diffuse systemic sclerosis has SRC. Scleroderma renal crisis is typically characterized by one or more of abrupt onset of marked or malignant hypertension, acute onset of oliguric renal failure, urinalysis that re veals only mild proteinuria with few cells or casts, and/or microangiopathic hemolysis anemia and/or thrombocytopenia.

In some embodiments, certain drugs can be linked to development of SSc (e.g., SSc- ILD) in subject. In some embodiments, a subject with SSc (e.g., SSc-ILD) has been administered one or more of cancer chemotherapeutic drugs, bleomycin, and/or docetaxei. In some embodiments, a subject has developed SSc-like complications at an injection site when administered an injection of vitamin K, vitamin B12, and/or analgesic pentazocine.

Pulmonary Function Tests Pulmonary' function tests (PFTs) are typically perfonned in SSc subjects at an initial time of diagnosis to determine if SSc has affected the lungs of the subject (e.g., SSc-ILD), and/or also to what extent. PFTs may be performed at least annually to evaluate any new onset of dyspnea, cough, or abnormalities found in radiography. A small portion of subjects with positive HRCT scans for SSc-ILD can have normal pulmonary function test results. Spirometry and Forced Vital Capacity (FVC)

Spirometry, or measuring of breath, is a major pulmonary' function test that can determine volume and/or speed (flow) of air that is inhaled and exhaled by a subject. A spirometer is used to measure forced vital capacity (FVC) (measured in liters, milliliters, and/or percentage of predicted) in a forced expiratory volume (FEV) test, among other characteristics. In an FEV test, a subject takes a deep breath, and exhales into a sensor as hard and for as long as possible (e.g., at least 6 seconds). Inhalation can also be tested using spirometry. An FEV test is typically repeated at least three times to ensure accuracy. "‘Normal” ranges for FVC are typically considered to be between 80% and 100% of predicted, “Of predicted” refers to reporting the subject’s results as a percentage of the known predicted values for a healthy subject of similar characteristics (e.g., height, sex, age, race, weight). Other measurements that can be taken include, but are not limited to, FEVi, wherein the FVC is measured within the first second of forced exhalation, and/or forced expiratory' flow (FEF), which measures the flow of air coming out of the lung during the middle portion of forced expiration. An FEV I/FVC ratio is also typically calculated. In some embodiments, a subject of the present disclosure has an FYC of between about 100% and about 90% of predicted. In some embodiments, a subject of the present disclosure has an FVC of between about 90% and about 80% of predicted. In some embodiments, a subject of the presen t disclosure has an FVC of between about 80% and about 70% of predicted. In some embodiments, a subject of the present disclosure has an FVC of between about 70% and about 60% of predicted. In some embodiments, a subject of the present disclosure has an FVC of between about 60% and about 50% of predicted. In some embodiments, a subject of the present disclosure has an FVC of between about 50% and about 40% of predicted. In some embodiments, a subject of the present disclosure has an FVC of between about 40% and about 30% of predicted. In some embodiments, a subject of the present disclosure has an FVC of between about 30% and about 20% of predicted. In some embodiments, a subject of the present disclosure has an FVC of greater than or equal to 50% of predicted.

A general measurement of disease progression can be an annual rate of decline in FVC. In some embodiments, an annual rate of decline in forced vital capacity (FVC) of a subject is measured over a time period of at least one year after administration of one or more TbKII antagonists of the present disclosure, in some embodiments, an annual rate of decline in forced vital capacity (FVC) of a subject is measured over a time period of at least 52 weeks after administration of one or more TflR!I antagonists of the present disclosure. In some embodiments, an annual rate of decline in forced vital capacity (FVC) of a subject is measured over a time period of at least one year after administration of one or more TbKIί antagonists of the present disclosure and is compared to a baseline FVC measurement. In some embodiments, an annual rate of decline in forced vital capacity (FVC) of a subject is measured over a time period of at least 52 weeks after administration of one or more TbKII antagonists of the presen t disclosure and is compared to a baseline FVC measurement. In some embodiments, administration of one or more TbKII antagonists of the present disclosure results in a decline m an annual rate of forced vital capacity (FVC). In some embodim ents, administration of one or m ore TbKII antagonists of the present disclosure results in a reduction in annual rate of decline of forced vital capacity (FVC). In some embodiments, an annual rate of decline in FVC has been slowed. In some embodiments, a subject is determined to have a slowing in the rate of decline in pulmonary function after administration of one or more TbKII antagonists of the present disclosure.

In some embodimen ts, administration of one or more TbKII antagonists of the present disclosure slows the annual rate of decline in FVC compared to a baseline measurement. In some embodiments, administration of one or more TpRII antagonists of the present disclosure slows the annual rate of decline in FVC compared to a subject administered standard of care (SOC), in some embodiments, standard of care comprises administration of an immunosuppressive therapy. In some embodiments, standard of care comprises administration of mycophenolate mofetil (MMF). In some embodiments, standard of care comprises administration of methotrexate. In some embodiments, standard of care comprises administration of cyclophosphamide. In some embodiments, standard of care comprises administration of nintedanib (Ofev). In some embodiments, standard of care comprises administration of rituximab. In some embodiments, standard of care comprises administration of one or more of mycophenolate mofetil (MMF), methotrexate, cyclophosphamide, nintedanib (Ofev), and rituximab. In some embodiments, standard of care comprises administration of at least one therapy selected from the group consisting of mycophenolate mofetil (MMF), methotrexate, cyclophosphamide, nintedanib (Ofev), and rituximab. In some embodiments, administration of one or more TbKIΪ antagonists of the present disclosure slows the annual rate of decline in FVC compared to a subject who has bad a lung transplant.

In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), steps comprising measuring at least one initial point of lung function in a subject; administering a Transforming Growth Factor-b Receptor II (TpRII) fusion polypeptide to the subject; remeasuring the at least one point of lung function in the subject; and based on the measuring, determining a change in the rate of decline in lung function of the subject.

In some embodiments the present disclosure provides methods of treating systemic sclerosis associated with interstitial lung disease (SSc-ILD), steps comprising measuring at least one initial point of lung function in a subject; administering a Transforming Growth Factor-b Receptor II (TpRII) fusion polypeptide to the subject; re-measuring the at least one point of lung function in the sub j ect; and based on the measuring, determining a change in the rate of decline in lung function of the subject.

In some embodiments, the rate of decline in lung function is measured by Forced Vital Capacity (FVC) of the subject. In some embodiments, the rate of decline in lung function is measured as an annual rate of decline in Forced Vital Capacity (FVC) of the subject.

In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FVC of between about 100% and about 90% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FVC of between about 90% and about 80% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FVC of between about 80% and about 70% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FV C of between about 70% and about 60% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FVC of between about 60% and about 50% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FVC of between about 50% and about 40% of predicted. In some embodiments, a subject of the presen t disclosure has an initial point of lung function comprising an FVC of between about 40% and about 30% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FV C of between about 30% and about 20% of predicted. In some embodiments, a subject of the present disclosure has an initial point of lung function comprising an FVC of greater than or equal to 50% of predicted. In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TbKIί) fusion polypeptide to the subject, wherein the subject has an F V C of greater than or equal to 50% of predicted at baseline.

In some embodiments, an annual rate of decline in FVC of a subject is between about 1 % and about 10%. In some embodiments, an annual rate of decline in FVC of a subject is between about 5% and about 10%. In some embodiments, an annual rate of decline in FVC of a subject is between about 10% and about 15%. In some embodiments, an annual rate of decline in FV C of a subject is between about 15% and about 20%. In some embodiments, an annual rate of decline in FV C of a subject is between about 20% and about 25%. In some embodiments, an annual rate of decline in FVC of a subject is between about 25% and about 30%. In some embodiments, an annual rate of decline in FVC of a subject is between about 30% and about 35%. In some embodiments, an annual rate of decline in FVC of a subject is between about 35% and about 40%. In some embodiments, an annual rate of decline in FVC of a subject is between about 40% and about 45%. In some embodiments, an annual rate of decline in FVC of a subject is between about 45% and about 50%. In some embodiments, an annual rate of decline in FVC of a subject is between about 50% and about 55%. In some embodiments, an annual rate of decline in FVC of a subject is between about 55% and about 60%. In some embodiments, an annual rate of decline in FVC of a subject is between about 60% and about 65%. In some embodiments, an annual rate of decline in FVC of a subject is between about 65% and about 70%. In some embodiments, an annual rate of decline in FVC of a subject is between about 10% and about 20%. In some embodiments, an annual rate of decline in FV C of a subject is between about 20% and about 30%. In some embodiments, an annual rate of decline in FVC of a subject is between about 30% and about 40%. In some embodiments, an annual rate of decline in FVC of a subject is between about 40% and about 50%. In some embodiments, an annual rate of decline in FVC of a subject is between about 50% and about 60%. In some embodiments, an annual rate of decline in FVC of a subject is between about 60% and about 70%.

In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a TfiRH antagonist to a subject in need thereof, wherein an annual rate of decline of the subject is reduced. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 1% and about 10%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 5% and about 10%.

In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10% and about 15%. In some embodiments, an annual rate of decline in F VC of a subject is reduced by between about 15% and about 20%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20% and about 25%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 25% and about 30%. in some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30% and about 35%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 35% and about 40%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40% and about 45%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 45% and about 50%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50% and about 55%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 55% and about 60%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 60% and about 65%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 65% and about 70%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10% and about 20%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20% and about 30%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30% and about 40%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40% and about 50%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50% and about 60%. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 60% and about 70%.

In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a TbKIΊ antagonist to a subject in need thereof, wherein an annual rate of decline of the subject is reduced relative to a subject treated with standard of care (SQC). In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 1% and about 10% relative to a subject treated with SQC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 5% and about 10% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10% and about 15% relative to a subject treated with SQC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 15% and about 20% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 20% and about 25% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 25% and about 30% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30% and about 35% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 35% and about 40% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 40% and about 45% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 45% and about 50% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50% and about 55% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 55% and about 60% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 60% and about 65% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 65% and about 70% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10% and about 20% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20% and about 30% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30% and about 40% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40% and about 50% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50% and about 60% relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 60% and about 70% relative to a subject treated with SOC. In some embodiments, annual rate of decline is measured in milliliters (mL). In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 1 mL and about 10 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 5 mL and about 10 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10 mL and about 15 mL. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 15 mL and about 20 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20 ml, and about 25 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 25 mL and about 30 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30 mL and about 35 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 35 mL and about 40 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40 mL and about 45 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 45 mL and about 50 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50 mL and about 55 mL, In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 55 mL and about 60 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 60 ml, and about 65 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 65 mL and about 70 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 70 rnL and about 75 mL. In some embodiments, an annual rate of decline m FVC of a subject is reduced by between about 75 mL and about 80 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 85 mL and about 90 mL, In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 90 mL and about 95 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 95 mL and about 100 inL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10 mL and about 20 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20 mL and about 30 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30 mL and about 40 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40 mL and about 50 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50 mL and about 60 mL. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 60 mL and about 70 mL. In some embodiments, an annual rate of decline in F VC of a subject is reduced by between about 70 mL and about 80 mL, In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 80 mL and about 90 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 90 ml, and about 100 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by about 100 mL. In some embodiments, an annual rate of decline in FVC of a subject is reduced by more than about 100 mL.

In some embodiments, annual rate of decline is measured in milliliters (ml.) relative to a subject treated with standard of care (SOC). In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 1 mL and about 10 rnL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 5 mL and about 10 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 10 rnL and about 15 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FV C of a subject is reduced by between about 15 mL and about 20 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20 mL and about 25 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 25 mL and about 30 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30 mL and about 35 rnL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 35 mL and about 40 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40 mL and about 45 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 45 rnL and about 50 mL relative to a subject treated with SQC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50 mL and about 55 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 55 mL and about 60 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 60 ml, and about 65 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 65 mL and about 70 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 70 mL and about 75 mL relative to a subject treated wi th SOC, In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 75 mL and about 80 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 85 mL and about 90 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 90 mL and about 95 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 95 mL and about 100 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 10 rnL and about 20 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 20 mL and about 30 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 30 mL and about 40 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 40 mL and about 50 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 50 mL and about 60 rnL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 60 mL and about 70 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 70 mL and about 80 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 80 mL and about 90 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by between about 90 mL and about 100 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline in FVC of a subject is reduced by about 100 mL relative to a subject treated with SOC. In some embodiments, an annual rate of decline m FVC of a subject is reduced by more than about 100 mL relative to a subject treated with SOC.

In some embodimen ts, administration of one or more TbKII antagonists of the present disclosure results in a reduction in annual rate of decline of forced vital capacity (FVC) relative to a subject treated with standard of care (SOC). In some embodiments, an annual rate of decline in FVC has been slowed relative to a subject treated with standard of care (SOC). In some embodiments, administration of one or more TbKP antagonists of the present disclosure slows the annual rate of decline in FVC relative to a subject treated with standard of care (SOC). In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary' complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TbKII) fusion polypeptide to the subject; wherein an annual rate of decline in FVC of a subject is reduced by about 100 mL relative to a subject treated with SOC. ). In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (Ί ' bKP) fusion polypeptide to the subject; wherein an annual rate of decline in FVC of a subject is reduced by 100 mL relative to a subject treated with SOC. In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Facto r-b Receptor II ( ' TbKII) fusion polypeptide to the sub j ect; wherein an annual rate of decline in FVC of a subject is reduced by more than about 100 mL relative to a subject treated with SOC. Diffusing Capacity for Carbon Monoxide (DLCO)

Measurement of diffusing capacity of the lungs for carbon monoxide (DLCO) is a medical test that determines how much oxygen travels from the alveoli of the lungs to the blood stream. Testing for DLCO involves measuring the partial pressure difference between inspired and expired carbon monoxide. A DLCO measurement relies on the strong affinity' and/or large absorption capacity of red blood cells for carbon monoxide and thus demonstrates gas uptake by the capillaries that are less dependent on cardiac output. A measurement of DLCO can be affected by atmospheric pressure and/or altitude and correction factors can be calculated using the method recommended by the American Thoracic Society'. Expected DLCO is also affected by the amount of hemoglobin, carboxyhernoglobin, age and/or sex of the subject. Generally, DLCO is measured in "mL/min/kPa" and can also be presented as a percentage of predicted. Of predicted ’ refers to reporting the subject’s results as a percentage of the known predicted values for a healthy subject of similar characteristics (e,g., height, sex, age, race, and weight). DLCO can be measured by one or more of a rapidly responding gas analyzer (RGA), flow volume analyzer, and/or gas analyzer. Several factors can lead to a decrease in DLCO, which includes, but is not limited to, hindrance in the alveolar wall (e.g., fibrosis, alveolitis, vasculitis), decrease of total lung area (e.g., restrictive lung disease or lung resection (partial or total)), chronic obstructive pulmonary disease (COPD) due to decreased surface area in the alveoli as well as damage to the capillary bed, pulmonary embolism, cardiac insufficiency, pulmonary hypertension (PH), exposure to bleomycin (upon administration of at least about 200 units), chronic heart failure, anemia due to a decrease in blood volume, and/or amiodarone high cumulative dose (e.g., at least about 400 milligrams per day).

In some embodiments, DLCO is measured using a rapidly responding gas analyzer (RGA). In some embodiments, a subject has a DLCO of between about 90% and about 80% of predicted. In some embodiments, a subject has a DLCO of between about 80% and about 70% of predicted. In some embodiments, a subject has a DLCO of between about 70% and about 60% of predicted. In some embodiments, a subject has a DLCO of between about 60% and about 50% of predicted. In some embodiments, a subject has a DLCO of between about 50% and about 40% of predicted. In some embodiments, a subject has a DLCO of between about 40% and about 30% of predicted. In some embodiments, a subject has a DLCO of between about 30% and about 20% of predicted.

In some embodiments, a subject of the present disclosure has a DLCO of greater than or equal to 40% of predicted. In some embodiments, a subject of the present disclosure has a DLCO of greater than or equal to 40% of predicted, corrected by Hgb. In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Facto r-b Receptor II (TbMII) fusion polypeptide to the subject; wherein the subject has a DLCO of greater than or equal to 40% of predicted at baseline. In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TbHIP fusion polypeptide to the subject; wherein the subject has a DLCO of greater than or equal to 40% of predicted (corrected by Hgb) at baseline. In some embodiments, a subject of the present disclosure has a DLCO of between about 40% and about 89% of predicted. In some embodiments, a subject of the present disclosure has a DLCO of between about 40% and about 89% of predicted, corrected by Hgb. In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TbKP) fusion polypeptide to the subject; wherein the subject has a DLCO of between about 40% and about 89% of predicted at baseline. In some embodiments the present disclosure provides methods of treating systemic sclerosis with one or more pulmonary complications (e.g., SSc-ILD), comprising administering a Transforming Growth Factor-b Receptor II (TpRII) fusion polypeptide to the subject; wherein the subject has a DLCO of between about 40% and about 89% of predicted (corrected by Hgb) at baseline.

In some embodiments, an annual rate of decline in diffusing capacity of the lungs (DLCO) of a subject is measured over a time period of at least one year after administration of one or more THRU antagonists of the present disclosure. In some embodiments, an annual rate of decline in diffusing capacity of the lungs (DLCO) of a subject is measured over a time period of at least one year after administration of one or more TpRII antagonists of the present disclosure and is compared to a baseline DLCO measurement. In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a TbίOI antagonist to a subject m need thereof, wherein an annual rate of DLCO of the subject is reduced.

In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 1% and about 10%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 5% and about 10%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 10% and abou t 15%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 15% and about 20%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 20% and about 25%. In some embodiments, an annual rate of decline m DLCO of a subject is reduced by between about 25% and about 30%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 30% and about 35%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 35% and about 40%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 40% and about 45%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 45% and about 50%. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 50% and about 55%. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 55% and about 60%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 60% and about 65%. In some embodiments, an annual rate of decline in

DLCQ of a subject is reduced by between about 65% and about 70%. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 10% and about 20%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 20% and about 30%. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 30% and about 40%. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 40% and about 50%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 50% and about 60%. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 60% and about 70%. In some embodiments, annual rate of decline is measured in mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 1 mL/min/kPa and about 10 mL/min/kPa per year. In some embodiments, an annual rate of decline m DLCO of a subject is reduced by between about 5 mL/min/kPa and about 10 mL/min/kPa per year. In some embodiments, an annual rate of decline in D LCO of a subject is reduced by between about 10 mL/min/kPa and about 15 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 15 mL/min/kPa and about 20 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 20 mL/min/kPa and about 25 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 25 mL/min/kPa and about 30 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 30 mL/min/kPa and about 35 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 35 mL/min/kPa and about 40 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 40 mL/min/kPa and about 45 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 45 mL/min/kPa and about 50 mL/min/kPa per year. In some embodiments, an annual rate of decline m DLCO of a subject is reduced by between about 50 mL/min/kPa and about 55 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 55 mL/min/kPa and about 60 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCQ of a subject is reduced by between about 60 mL/min/kPa and about 65 mL/min/kPa per year, in some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 65 mL/min/kPa and about 70 mL/min/kPa per year. In some embodiments, an annual rate of decline in D LCO of a subject is reduced by between about 70 mL/min/kPa and about 75 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 75 mL/min/kPa and about 80 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 85 mL/min/kPa and about 90 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 90 mL/min/kPa and about 95 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 95 mL/min/kPa and about 100 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 10 mL/min/kPa and about 20 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 20 mL/min/kPa and about 30 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 30 mL/min/kPa and about 40 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 40 mL/min/kPa and about 50 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 50 mL/min/kPa and about 60 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 60 mL/min/kPa and about 70 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 70 mL/min/kPa and about 80 mL/mm/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 80 mL/min/kPa and about 90 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by between about 90 mL/min/kPa and about 100 mL/min/kPa per year. In some embodiments, an annual rate of decline in DLCO of a subject is reduced by more than about 100 mL/min/kPa per year.

Lung volume

Measuremen t of lung volumes may be important when spirometry' shows a decreased forced vital capacity. Body plethysmography is the typical way to measure lung volumes, particularly in the setting of significant airflow obstruction. Alternative methods to measure lung function include, but are not limited to, helium dilution, nitrogen washout, and/or measurements based on chest imaging.

Measurements of total lung capacity (TLC) using chest radiograph and/or high resolution computed tomography (HRCT) generally correlate within 15 percent of those obtained by body plethysmography.

Common lung volume measurements include vital capacity, total lung capacity, functional residual capacity', and/or residual volume. Vital capacity (VC) comprises maximum volume exhaled after maximum inspiration, and can be measured during forced exhalation (i· VC) or slow exhalation (SVC). Functional residual capacity (FRC) comprises volume of air remaining in chest at the end of a tidal volume breath. Residual volume (RV) comprises volume of air remaining in chest after maximal exhalation. Expiratory reserve volume (ERV) comprises volume of air exhaled from end-tidal volume (FRC) to point of maximal exhalation (RV), therefore RV plus ERV = FRC. inspiratory capacity (IC) comprises maximum inspiration from end-tidal volume (FRC) to total lung capacity. Total lung capacity (TLC) comprises volume of air in lungs at end of maximal inspiration (usually calculated by RV plus VC or FRC plus IC)

Combination of an FEV1/FVC and TLC both less than the fifth percentile lower limit of normal is considered a mixed defect. Combination of a normal FEV 1/FVC and a normal TLC, and a low FEV1 or FVC, is considered a nonspecific pattern. A pattern in which an FVC is disproportionately reduced relative to TLC has been described as a complex restrictive pattern.

In some embodiments, a subject with SSc (e.g., SSc-ILD) has a decreased lung volume. In some embodiments, a subject with SSc (e.g., SSc-ILD) has a decreased total lung capacity (TLC),

In some embodiments, a subject has a TLC of between about 100% and about 90% of predicted. In some embodiments, a subject has a TLC of between about 90% and about 80% of predicted. In some embodiments, a subject has a TLC of between about 80% and about 70% of predicted. In some embodiments, a subject has a TLC of between about 70% and about 60% of predicted. In some embodiments, a subject has a TLC of between about 60% and about 50% of predicted. In some embodiments, a subject has a TLC of between about 50% and about 40% of predicted. In some embodiments, a subject has a TLC of between about 40% and about 30% of predicted. In some embodiments, a subject has a TLC of between about 30% and about 20% of predicted. Six Minute Walk Test

A six-minute walk test (6MWT) assesses both distance walked (meters) and/or degree of oxygen desaturation of a subject. It can be used as a measure of submaximal exercise performance in a variety of pulmonary' diseases (e.g,, SSc-ILD, idiopathic pulmonary' fibrosis (IPF) and/or pulmonary' arterial hypertension (PAH)). Assessment of pulse oxygen desaturation can be problematic in patients with SSc due to Raynaud phenomenon and/or poor finger perfusion. If the appropri ate attachment is avail able, measurement of pulse oxygen saturation using an earlobe clip may be more effective for subjects with SSc with a pulmonary complication (e.g., SSc-ILD). Desaturation on ambulation is common to both ILD and/or pulmonary hypertension. A healthy subject (e.g., a subject without SSc-ILD or any other respiratory' disease) typically walks between about 400 to about 700 meters in a six minute walk test. Average distance walked may vary by characteristics such as sex, age, weight, or height, among others. A reference subject for a six minute walk test may be a healthy subject (e.g., no respiratory' disease) of similar age, height, weight, and/or same sex. An improvement of at least about 30 meters in distance walked during a six minute walk test typically' indicates an improvement in respiratory function.

In some embodiments, a subject of the present disclosure has a decrease in distance walked during a six minute walk test. In some embodiments, a subject has a decrease in oxygen desaturation. In some embodiments, administration of one or more TflRII antagonists of the present disclosure results in an increase in distance walked during a six minute walk test. In some embodiments, administration of one or more TbKP antagonists of the present disclosure results in an increase in oxygen desaturation during a six minute walk test.

In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbίIP) antagonist to a subject in need thereof, wherein the subject has an improved distance walked in a six minute walk test after the treatment. In some embodiments, a subject improves distance walked in a six minute walk test by at least about 30 meters compared to a reference subject. In some embodiments, a reference subject is a healthy person of similar characteristics (e.g., sex, age, height, weight). In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 10 meters and about 15 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 15 meters and about 20 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 20 meters and about 25 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 25 meters and about 30 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 30 meters and about 35 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 35 meters and about 40 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 40 meters and about 45 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 45 meters and about 50 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 50 meters and about 55 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 55 meters and about 60 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 60 meters and about 65 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 65 meters and about 70 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 70 meters and about 75 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 75 meters and about 80 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 80 meters and about 85 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 85 meters and about 90 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 90 meters and about 95 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least between about 95 meters and about 100 meters compared to a reference subject. In some embodiments, a subject improves distance walked in a six minute walk test by at least more than 100 meters compared to a reference subject. In some embodiments, treatment of a subject with one or more of a TpR [ f antagonist of the present disclosure improves distance walked in a six minute walk test of a subject. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 1% and about 3%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 1% and about 5%. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 1% and about 8%. In some embodiments, treatment of a subject with one or more of a ' TbKII antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 1% and about 10%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 10% and about 15%. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 15% and about 20%. In some embodiments, treatment of a subject with one or more of a ' TbKII antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 20% and about 25%. In some embodiments, treatment of a subject with one or more of a ' TbKH antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 25% and about 30%. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 30% and about 35%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 35% and about 40%. In some embodiments, treatment of a subject with one or more of a ' TbKH antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 40% and about 45%. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure improves distance walked in a six minute walk test of a subject by between about 45% and about 50%. Pulmonary Imaging

Chest Radiography

Chest radiography can be used together with other methods of detection described herein to determine if a subject has SSc-ILD, Tire most common radiographic abnormality on a routine chest radiograph is a reticular pattern (e.g., fine or ground glass, medium or irregular, coarse or honeycomb), however, nodular or mixed paterns (e.g., alveolar filling and/or increased interstitial markings) are not unusual. Basie patterns of diffuse lung disease include, but are not limited to, nodular (including micronodular and/or military), reticular (fine or ground glass, medium or irregular, coarse or honeycomb), linear (interlobular septal or Kerley lines and/or intralobular septal lines), combined reticular and/or nodular, destructive, alveolar, bronchial, and/or vascular. Although a chest radiograph is useful in suggesting presence of interstitial lung disease (ILD), the correlation between die radiographic pattern and the stage of disease (clinical or histopathologic) is generally poor. A radiographic finding of honeycombing (small cystic spaces) may correlate with pathologi c findings and, when present, can portend a poor prognosis. In evaluation of ILD, it is important to review' all previous chest films to assess die rate of change in disease activity.

Honeycombing can be a feature of endstage interstitial lung disease. It represents restructuring of the lung parenchyma, with simplification of the lung architecture and subsequent small cyst formation surrounded by fibrotic tissue. Accompanying bronchiolectasis is regularly present. The small subpleural spaces, which usually measure between 3 and 10 tnm in diameter may connect with small airway and some collapse on expiratory imaging. Honeycombs are usually stacked and are not separated by intervening normal lung. Bronchiolectases w'hich can mimic honeycombing are usually separated by- interposed lung parenchyma. The expiratory collapse of some of these spaces distinguishes them from the subpleural spaces formed by paraseptal emphysema, which do not decrease in size with expiration. In addition, honeycombs have slightly thicker walls than emphysematous spaces. Honeycombing forms an array of multilayered, stacked spaces, while paraseptal emphysematous spaces are single-layered (i.e., single-tiered). The differentiation of honeycombing, bronchiolectasis and paraseptal emphysema is typically achieved with additional analysis using high resolution computed tomography. Honeycombing can be seen in subjects suffering from, but not limited to IPF/UIP, other interstitial pneumonias, Langerhans cell histiocytosis (previously eosinophilic granuloma), collagen vascular diseases, healed necrotizing infections, endstage pneumoconiosis, and endstage hypersensitivity pneumonitis (i.e., extrinsic allergic alveolitis). Classic radiographic features of established SSe-ILD comprise symmetric, reticular (e.g., fine or ground glass, medium or irregular, coarse or honeycomb), opacities. Radiographic features are usually most pronounced at the lung bases.

In some embodiments, a subject has a chest radiograph with one or more radiographic abnormalities. In some embodiments, a subject has a chest radiograph with one or more of a reticular pattern, a modular patern, or a mixed pattern. In some embodiments, a sub j ect has a chest radiograph with one or more of a nodular pattern, reticular pattern, a linear pattern, a combined reticular and nodular pattern, a destructive pattern, an alveolar pattern, a bronchial pattern, and/or a vascular patern. In some embodiments, a subject has a chest radiograph with a reticular patern. In some embodiments, a subject has a chest radiograph with a honeycombing pattern.

A chest radiograph can be normal in as many as 10 percent of patients with some forms of ILL), particularly those with hypersensitivity pneumonitis. Thus, a complete evaluation is generally undertaken even if a patient has a normal chest radiograph or radiographic evidence of ILD .

High Resolution Computed Topography (HRCT)

A more sensitive alternative to chest radiography for diagnosing and/or evaluating SSc with a pulmonary complication (e.g., SSe-ILD) is High Resolution-Computer Topography (HRCT). HRCT scans thin sections (3 mm or less) of the lung. HRCT can demonstrate character and/or distribution of fine structural abnormalities that are not visible on chest radiographs. Prone (e.g., face down) images are often obtained in addition to the usual supine (e.g., face up) images to differentiate between increased density due to ILD and/or dependent atelectasis at the posterior lung bases. HRCT is generally performed using a conventional CT scanner. However, imaging parameters are chosen to maximize spatial resolution, which include, but are not limited to, a narrow slice width (usually 1-2 mm), a high spatial resolution image reconstruction algorithm, field of view is minimization (e.g., to minimize the size of each pixel), and/or other scan factors (e.g., focal spot). Depending on the suspected diagnosis, an HRCT scan may be performed in both inspiration and expiration. HRCT is conventionally performed by taking thin sections (e.g., less than 3 mm) that are about 10-40 mm apart. The resulting scan is generally a few images that should be representative of the lungs in general, but that cover only approximately one tenth of the lungs. HRCT patterns found in SSe-ILD, for example, may mirror common histopathological patterns of idiopathic interstitial pneumonias, which include but are not limited to fibrotic nonspecific interstitial pneumonia (NSIP), usual interstitial pneumonia (UIP), and/or centrilobular fibrosis. Among patients with SSc- ILD, the most common pathologic pattern is typically NSIP, which can be associated with an HRCT finding of ground glass opacities in a peripheral distribution and/or a lower proportion of coarse reticulation, but is not typically associated with a honeycomb pattern. Early HRCT changes are typically a narrow, often ill-defined, subpleural crescent of increased density in the posterior (dependent) segments of the lower lobes. As SSc-ILD progresses, there is usually volume loss associated with a reticular appearance and/or traction bronchiectasis. In a subset of patients with a histologic UIP pattern, the HRCT appearance is usually similar to that of fibrotic NSIP. However, sometimes the HRCT pattern is more consistent with that of UIP with bibasilar reticular opacities, associated with traction bronchiectasis and/or the development of subpleural honeycomb air spaces, which ultimately coalesce into large cystic air spaces. A honeycomb pattern can be indicative of a subject that is in advanced stages of disease and will not respond to immunosuppressive therapy. Centrilobular fibrosis is typically a rare pattern that may be associated with patchy ground glass or consolidative opacities with a central distribution on HRCT scanning. Centrilobular nodules and/or “tree- in-bud” patterns are other features of recurrent aspiration. In some embodiments, a subject with SSc-ILD has an HRCT scan that reveals lung parenchymal changes including any round glass opacity 7 and fibrotic peripheral reticulations and honey combing without abnormalities. In some embodiments, a subject has undergone a High Resolution Computed Topography scan (HRCT) of the lungs prior to treatment. In some embodiments, a subject has undergone a HRCT scan of the lungs prior to treatment with one or more TbKII antagonists of the present disclosure. In some embodiments, a subject receives a HRCT scan of the lungs during treatment with one or more TbϋII antagonists of the present disclosure. In some embodiments, a subject receives a HRCT scan of the lungs after treatment with one or more TpRll antagonists of the present disclosure. In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a TbKϊΐ antagonist to a subject in need thereof, wherein an amount of fibrosis or pattern of interstitial pneumonia is determined by HRCT scan of the kings of the subject.

In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 1 % and about 10% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 5% and about 10% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 10% and about 15% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 15% and about 20% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 20% and about 25% fibrosis of the lungs.

In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 25% and about 30% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 30% and about 35% fibrosis of the kings. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 35% and about 40% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 40% and about 45% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 45% and about 50% fibrosis of tire kings. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 1% and about 10% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 10% and about 20% fibrosis of the kings. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 20% and about 30% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 30% and about 40% fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 40% and about 50% fibrosis of the kings. In some embodiments, an HRCT scan of a subject has determined a presence of at least 10% fibrosis of the kings. In some embodiments, an HRCT scan of a subject has determined a presence of greater than or equal to 10% fibrosis of the kings. In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a TflRH antagonist to a subject in need thereof, wherein an amount of abnormalities is determined by HRCT scan of the lungs of the subject. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 1 % and about 10% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 5% and about 10% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 10% and about 15% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 15% and about 20% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 20% and about 25% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 25% and about 30% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 30% and about 35% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 35% and about 40% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 40% and about 45% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 45% and about 50% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 1% and about 10% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 10% and about 20% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 20% and about 30% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 30% and about 40% abnormalities in the lungs. In some embodiments, an HRCT scan of a subject has determined a presence of at least between about 40% and about 50% abnormalities in the lungs. in some embodiments, the present disclosure provides methods of treating SSe-ILD, comprising administering a TjiRH antagonist to a subject in need thereof, wherein an improvement in the amount of fibrosis in the lungs is determined by HRCT scan of the lungs of the subject. In some embodiments, an improvement in the amount of fibrosis in the lungs of a subject comprises a decrease in the amount of fibrosis in the lungs.

In some embodiments, an HRCT scan of a subject has determined an impro vement of at least between about 1% and about 10% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 5% and about 10% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 10% and about 15% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 15% and about 20% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 20% and about 25% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 25% and about 30% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an impro vement of at least between about 30% and about 35% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 35% and about 40% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 40% and about 45% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 45% and about 50% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 1% and about 10% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 10% and about 20% of fibrosis of tire lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 20% and about 30% of fibrosis of the lungs, in some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 30% and about 40% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined an improvement of at least between about 40% and about 50% of fibrosis of the lungs.

In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 1% and about 10% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 5% and about 10% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 10% and about 15% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 15% and about 20% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 20% and about 25% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 25% and about 30% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 30% and about 35% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 35% and about 40% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 40% and about 45% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 45% and about 50% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 1 % and about 10% of fibrosis of the lungs.

In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 10% and about 20% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 20% and about 30% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 30% and about 40% of fibrosis of the lungs. In some embodiments, an HRCT scan of a subject has determined a decrease of at least between about 40% and about 50% of fibrosis of the lungs.

In some embodiments, an HRCT scan of a subject with SSc-ILD show ' s a pattern of nonspecific interstitial pneumonia (NSTP) in the lungs of the subject. In some embodiments, an HRCT scan of a subject with SSc-ILD show's a pattern of usual interstitial pneumonia (UIP) in the lungs of the subject. In some embodiments, an HRCT scan of a subject with SSc-ILD show's a honeycomb pattern. In some embodiments, a subject with an HRCT scan showing a honeycomb pattern does not respond to immunosuppressive therapy.

HRCT can be paired with a technology called Functional Respiratory Imaging (FRI). FRI is a non-invasive measurement of the patient-specific respirator)' system. A set of distinct biomarkers analyzes exposure, structure and function of the lungs and airways. Phase i comprises medical imagine, typically using HRCT scans. Phase 2 comprises image processing and measurements. Phase 3 comprises flow' simulation, where Computational Fluid Dynamics (CFD) is used to quantify airflow and exposure to inhaled particles. FRI provides regional information on airway resistance, air trapping, ventilation mapping, ventilation/perfusion reserve, lung and lobar volume, emphysema, internal airflow distribution, blood vessel volume, nodule volume, airway (wall) volume, and aerosol deposition. In some embodiments, a subject has undergone FRI of the lungs prior to treatment. In some embodiments, a subject has undergone an FRI analysis of the lungs prior to treatment with one or more TpRII antagonists of the present disclosure. In some embodiments, a subject receives an FRI analysis of the lungs during treatment with one or more Tb Ri I antagonists of the present disclosure. In some embodiments, a subject receives an FRI analysis of the lungs after treatment with one or more TpRII antagonists of the present disclosure. In some embodiments, the present disclosure provides methods of treating SSc- ILD, comprising administering a THRU antagonist to a sub j ect in need thereof, wherein an amount of fibrosis or pattern of interstitial pneumonia is determined by FRI analysis of the lungs of the subject.

In some embodiments, the present disclosure provides methods of treating SSc-ILD, comprising administering a TpRlf antagonist to a subject in need thereof, wherein an improvement in the amount of fibrosis in the lungs is determined by FRI analysis of the lungs of the subject. In some embodiments, an improvement in the amount of fibrosis in the lungs of a subject comprises a decrease in the amount of fibrosis in the lungs.

In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 1 % and about 10% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 5% and about 10% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 10% and about 15% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 15% and about 20% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 20% and about 25% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 25% and about 30% of fibrosis of the lungs, hi some embodiments, FRI analysis of a subject has determined an improvement of at least between about 30% and about 35% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 35% and about 40% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 40% and about 45% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 45% and about 50% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 1% and about

10% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 10% and about 20% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 20% and about 30% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improvement of at least between about 30% and about 40% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined an improv ement of at least between about 40% and about 50% of fibrosis of the lungs.

In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 1% and about 10% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 5% and about 10% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 10% and about 15% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 15% and about 20% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 20% and about 25% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 25% and about 30% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 30% and about 35% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 35% and about 40% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 40% and about 45% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 45% and about 50% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 1% and about 10% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 10% and about 20% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 20% and about 30% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 30% and about 40% of fibrosis of the lungs. In some embodiments, FRI analysis of a subject has determined a decrease of at least between about 40% and about 50% of fibrosis of the lungs.

Laboratory Tests

Routine laboratory' tests are typically run for subjects suspected of having SSc (e.g., SSc-ILD), as well as throughout treatment after diagnosis. In some embodiments, a routine laboratory test is selected from the group consisting of a complete blood count test, measurement of serum creatinine levels, measurement of creatine kinase levels, and/or urinalysis. In some embodiments, a complete blood count test of a subject with SSc (e.g., SSc-ILD) reveals one or more of anemia, iron deficiency, and/or gastrointestinal blood loss, among other complications . In some embodiments, serum creatinine levels in a subject with SSc (e.g., SSc-ILD) reveals renal dysfunction. In some embodiments, a test for creatine kinase levels in a subject with SSc (e.g., SSc-ILD) reveals myopathy and/or myositis. In some embodiments, a urinalysis test m a subject with SSc (e.g., SSc-ILD) reveals proteinuria and/or cellular casts.

Characteristic A utoantihodi.es In some embodiments, serologic tests are performed in a subject that has SSc (e.g., SSc-ILD) or is suspected of having SSc (e.g., SSc-ILD). In some embodiments, autoantibodies (also referred to as antibodies herein) are detected in a serologic test. In some embodiments, detection of one or more antibodies indicates presence of SSc-ILD. In some embodiments, levels of one or more antibodies in a subject sample are increased relative to a reference sample. In some embodiments, levels of one or more antibodies in a subject sample are compared to reference levels of antibodies in an otherwise healthy population of similar characteristics to the subject (e.g., age, gender, etc,). In some embodiments, detection of antibodies indicates an increased percent chance of clinical complication. In some embodiments, levels of antibodies indicate an increased percent chance of clinical complication of ILD in a subject with SSc. In some embodiments, a subject has one or more of antinuclear antibody, anticentromere antibody, antitopoisomerase I antibody, and/or anti- RNA polymerase III antibody. In some embodiments, a subject with SSc-ILD has one or more of antinuclear antibody, anticentromere antibody, antitopoisomerase I antibody, and/or anti-RNA polymerase III antibody. In some embodiments, a subject with SSc-ILD has increased levels of one or more of antinuclear antibody, anticentromere antibody, antitopoisomerase I antibody, and/or anti-RNA polymerase ΪP antibody. In some embodiments, a subject with SSc-ILD has increased levels of one or more of antinuclear antibody, anticentromere antibody, antitopoisomerase I antibody, and/or anti-RNA polymerase III antibody when compared to a subject that does not have SSc-ILD. In some embodiments, a subject without SSc-ILD does not have one or more of antinuclear antibody, anticentromere antibody, antitopoisomerase I antibody, and/or anti-RNA polymerase III antibody. In some embodiments, a subject has antinuclear antibody (ANA). In some embodiments, a subject has detectable antitopoisomerase I (anti-ScI-70). In some embodiments, a subject has detectable anticentromere antibody (ACA). In some embodiments, a subject has detectable anti-RNA polymerase III antibody. In some embodiments, a subject has an increased level of antinuclear antibody (ANA). In some embodiments, a subject has an increased level of antitopoisomerase I (anti-ScI-70). In some embodiments, a subject has an increased level of anticentromere antibody (ACA). In some embodiments, a subject has an increased level of anti-RNA polymerase III antibody.

Approximately 95% of patients with SSc-ILD test positive for antinuclear antibody (ANA). Antitopoisomerase I antibodies are generally associated with diffuse SSc (dcSSc) as well as a higher risk of the subject to develop ILD. Anticentromere antibodies are generally associated with limited SSc (lcSSc). In contrast, approximately 5% of subjects with dcSSc typically have detectable anticentromere antibodies. Anti-RNA polymerase III antibodies are generally found in subjects with dcSSc, and/or may be associated with rapid skin complication, as well as increased risk of scleroderma renal crisis (SRC), and/or cancers. In subjects with Raynaud’s phenomenon, presence of anti-RNA polymerase III and/or antitopoisornerase I antibodies can present an increased risk of the subject developing SSc. Serum antibodies may also indicate probability of skin thickness. Subjects with anti-RNA polymerase III may have more rapid progression of skin thickness than subjects with anti- Sc 1-70 (topoisomerase I) antibody.

In some embodiments, a laboratory test is used to confirm that a subject has SSc (e.g., SSc-ILD). In some embodiments, a laboratory test is used to differentiate SSc (e.g., SSc-

TLD) from other possible diagnoses. In some embodiments, a subject is administered a test to detect a rheumatoid factor. In some embodiments, a subject is administered a test to detect antibodies to citrullinated peptides (anti-CCP). In some embodiments, a subject is administered a test to detect systemic lupus erythematosus (SLE)-associated antibodies. In some embodiments, a subject is administered a test to detect antibodies associated with overlap connective tissue diseases (e.g., RNP antibodies).

Fibrosis Biomarkers

Biomarkers disclosed herein may he defined as characteristics that are objectively measured and/or evaluated as an indicator of normal biologic processes, pathogenic processes (e.g., SSc-ILD), or pharmacologic responses to a therapeutic intervention (e.g., treatment of SSc-ILD with one or more TpRff antagonists of the present disclosure). Biomarkers of respiratory disease (e.g., SSc-ILD) may be measured by way of one or more of imaging, lung fimction, genetic polymorphisms and/or biochemical molecules identifiable in king tissue, BAL fluid and/or blood. Serum biomarkers of the present disclosure for SSc-ILD can be useful in all stages of clinical treatment, including, but not limited to, evaluation for predisposition (e.g., identify risk of a subject developing ILD or detect early disease), diagnosis SSc-ILD (e.g., identify presence of ILD and/or aide in classification, prognosis of SSc-ILD (e.g., predict disease progression and/or clinical events), therapeutic treatment of SSc-ILD (e.g., predict and/or quantify response to therapy), and/or research/clinical studies of SSc-ILD (e.g., act as a surrogate endpoint in research or clinical practice and/or identify novel treatment targets). In some embodiments, the disclosure provides a method of treating SSc-ILD, comprising administering a TpRH antagonist to a subject in need thereof, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of otSMA, ADAM 12, Arrti-HSP70 IgG, BAFF, BLyS, C3M, C4M, C6M, Cal5.3, CC16, CCL2, CCL18, Collal, Col3al, CTGF, CXCL4, E-selectm, ET-1, fibronectm, 1CAM, IL-6, IL-8, KL-6, MCP-I, MMP7, MMP12, Muc5B, Osteopontin, PAI-i, penostin, pro-C3, pro-C4, pro-C6, SP-A, SP-D, Tnfa, VCAM, VEGF, WFDC2(HE4), and YKL-40. In some embodiments, a biomarker for SSc-ILD is selected from the group consisting of KL-6/MUC1, surface protein A (SP-A), surface protein D (SP-D), CC 16, and/or Cal 5.3.

In some embodiments, a biomarker for SSc-ILD may be involved in one or more alveolar epithelial cell damage pathways. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of Cal5.3, CC16, KL-6, SP-A, and SP-D. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of Cal5.3, CC16, KL-6, SP-A, and SP-D, when compared with a subject that does not have SSc-ILD. In some embodiments, the disclosure provides methods of treating SSc- ILD, comprising administering a TBRH antagonist to a sub j ect in need thereof, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of Cal5.3, CC16, KL-6, SP-A, and SP-D.

In some aspects, lev els of KL-6 (encoded by MUC1), a glycoprotein found predominantly on type II pneumocytes and/or alveolar macrophages, may be elevated in the serum of subjects with SSc-ILD. Levels of KL-6 rnay correlate with the presence of pneumonitis and/or a radiological fibrosis score in subjects with SSc. In some embodiments, levels of KL-6 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of KL-6 when compared with a subject that does not have SSc-ILD.

Surfactant proteins A and/or D (SP-A and/or SP-D, respectively) are generally elevated in serum of subjects with SSc-ILD, Surfactant proteins comprise lipoprotein complexes contained within pulmonary surfactant, and/or are produced by type II AECs and/or Clara cells, which are generally important for imiate immune defense mechanisms and/or modulating inflammatory response at the alveolar air-liquid interface. In SSc-ILD, studies have typically found serum SP-D to be more sensitive but less specific than KL-6 for detecting disease. Several studies have also demonstrated negative correlation between serum SP-D and/or lung function, and/or worsening complications and/or lung function m longitudinal measures. In some embodiments, levels of SP-A and/or SP-D are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of SP-A and/or SP-D when compared with a subject that does not have SSc- ILD.

Clara cells are typically multifunctional cells that are predominantly localized at terminal bronchioles that can secrete 16kDa Clara cell protein (CC16). Clara cell protein (CC16) generally has important protective, immunosuppressive and/or anti-inflammatory functions. Serum CC16 has been shown to be elevated across several pulmonary conditions including SSc-ILD, compared with healthy controls and/or subjects with SSc but without ILD. In some embodiments, levels of CC16 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of CC16 when compared with a subject that does not have SSc-ILD.

Cancer associated antigen carbohydrate antigen 15.3 (Cal 5.3) is produced from the same MUC1 gene that encodes for KL-6 and/or is expressed on various epithelial ceils, including type II AECs. Elevated Cal5.3 has been demonstrated in IPF and/or SSc-ILD. In a study comprising over 200 subjects, CaJ5.3 level correlated strongly with HRC ' T scores. Furthermore, Cal 5.3 has been shown to outperform FVC in predicting survival. In some embodiments, levels of Cal5.3 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level Cal 15.3 when compared with a subject that does not have SSc-ILD.

In some embodiments, a biomarker for SSc-ILD is selected from the group consisting of MMP7, osteopontin, and/or ADAM12. In some embodiments, a biomarker for SSc-ILD may be involved in one or more aberrant fibrogenesis and/or matrix remodeling pathways. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of MMP7, osteopontin, and/or ADAM 12. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of MMP7, osteopontin, and/or ADAM12, when compared with a subject that does not have SSc-ILD.

Matrix metalloproteinases (MMPs) and/or their inhibitors (tissue inhibitors of metalloproteinases, TLMPs) are proteases important in mediating ECM degradation, activity of inflammatory' mediators and/or growth factors in the lung. In SSc-ILD, serum MMP7, MMP12 and/or TIMP1 have been shown to be elevated when compared with healthy controls and/or SSc without ILD, and/or have been shown to correlate inversely with pulmonary function. In some embodiments, levels of any one of MM7, MMP12, and/or TIMP1 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of any one of MM7, MMP12, and/or TIMPl when compared with a subject that does not have SSc-ILD.

ADAM proteins (a disintegrin and/or metalloproteinases) are a group of multifunctional proteins that share a metalloprotease domain with MMPs, and/or have a unique, multifunctional domain with both proteolytic and/or adhesive functions, and/or are known to play an important role in ceil binding, migration and/or signaling. They are implicated in a variety of diseases including CTD, malignancy, Alzheimer’s, Crohn’s disease and/or possibly pulmonary' fibrosis. Serum levels of AD AMI 2 have been shown to be elevated in subjects with SSc-ILD, compared to subjects that do not have SSc-ILD, and/or correlate positively with FVC and/or extent of ground glass opacities on HRC ' T, and/or correlate negatively with HRCT fibrosis score. In some embodiments, levels of ADAM! 2 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of ADAM12 when compared with a subject that does not have SSc-ILD. Osteopontin (OPN) is a multifunctional protein that is known to regulate inflammation, cellular immune response and/or T cell function, with a pro-fibrotic effect in ILD through mechanisms that are not well known. Serum OPN levels are elevated in IPF and/or SSc compared with subjects that do not have SSc-ILD, but may not differentiate well between ILD subtypes. In some embodiments, levels of OPN are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of OPN when compared with a subject that does not have SSc-ILD.

In some embodiments, a biomarker for SSc-ILD is selected from the group consisting of CCL18, YKL-40, ICAM, VCAM, E-selectin, Anti-HSP70 IgG, BLyS/BAFF, CCL2/MCP- 1, IL-6, and/or CXCL4. In some embodiments, a biomarker for SSc-ILD may be involved in one or more immune dysregulation and/or inflammation pathways. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of CCL18, YKL-40, ICAM, VCAM, E-selectin, Anti-HSP70 IgG, BLyS/BAFF, CCL2/MCP-I, IL-6, and/or CXCL4. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of CCL18, YKL-40, ICAM, VCAM, E-selectin, Anti-HSP70 IgG, BLyS/BAFF, CCL2/MCP-1, IL-6, and/or CXCL4, when compared with a subject that does not have SSc- ILD.

C-C motif chemokine ligand 18 (CCL18) is primarily produced by alveolar macrophages with an important role in stimulating fibroblasts to synthesize collagen in fibrotic lung diseases. Although unable to differentiate between ILD subtypes, longitudinal CCL18 levels have demonstrated correlation with lung function and/or mortality in 1PF and/or SSc-ILD subjects. In some embodiments, levels of CCL18 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of CCL18 when compared with a subject that does not have SSc-ILD.

YKL-40 is a chitmase-like glycoprotein, thought to have a role in regulating connective tissue cell proliferation and/or angiogenesis. Seram YKL-40 may not be able to distinguish between ILD subtypes, however, elevated levels have been identified across a variety of inflammatory and/or fibrotic diseases including, but not limited to, ILD, liver fibrosis, inflammatory arthropathies, asthma and/or chronic obstructive pulmonary disease (COPD). In some embodiments, levels of YKL-40 are elevated in a subject that has SSc- ILD. In some embodiments, a subject of the present disclosure has an increased level of YKL-40 when compared with a subject that does not have SSc-ILD.

Adhesion molecules, including intercellular adhesion molecule (ICAM1), vascular cell adhesion molecule 1 (VCAM1), and/or E-selectin, are typically expressed on leucocytes and/or vascular endothelial cells, and are important in mediating adhesion and/or interaction of these cells. Elevated levels of ICAM!, VCAM1, and/or E-selectin are detectable across a number of inflammatory and/or fibrovascular conditions including SSc. Integrins interact with adhesion molecules such as V CAM and/or ICAM, and/or other growth-factor receptors and/or ECM components to activate downstream signaling pathways and/or TGF-b mediated pulmonary 7 fibrosis. Elevated levels of ICAM, VC AM and/or E-selectin have been demonstrated in SSc, and ICAM has been associated with the combined presence of diffuse, rapidly progressive SSc, digital contractures, ILD and/or joint involvement. In some embodiments, levels of one or more of ICAM 1, CAM1, and/or E-selectin are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of one or more of ICAM 1, CAM1, and/or E-selectin when compared with a subject that does not have SSc-ILD.

Heat shock protein 70 (HSP70) typically induces T-cell proliferation and/or pro- fibrotic cytokine production. Autoantibodies to HSP70 may have pathogenic potential in ILD by augmenting neutrophil recruitment, complement activation and/or production of inflammatory mediators in target organs.

B lymphocyte stimulator (BLyS), also known as B-ceil activating factor (BAFF), is a cytokine belonging to the tumor necrosis factor family, critical to B cell maturation and/or antibody production with a possible pathogenic role in several autoimmune diseases and/or IPF. In some embodiments, levels ofBLyS are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of BLyS when compared with a subject that does not have SSc-ILD.

C-C motif chemokine 2 (CCL2), previously known as monocyte chemoattractant protein- 1 (MCP-1), plays an important role in innate immunity and/or inflammation, with a potential pro-fibrotic effect in SSc, among other conditions. A study of SSc-ILD patients found that CCL2 was an independent predictor of progression-free survival in IPF. In some embodiments, levels of CCL2 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of CCL2 when compared with a subject that does not have SSc-ILD.

Cytokine IL-6 is involved in the differentiation of CD4+ T cells to pro-fibrotic Th2 type cells, and implicated in the activation of fibroblasts. In SSc patients, a study has shown that IL-6 was associated with shorter survival and/or time to decline in FVC adjusted for age, smoking, composite physiologic index, pulmonary hypertension and/or skin disease. In some embodiments, levels of IL-6 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of IL-6 when compared with a subject that does not have SSc-ILD.

CXCL4, also known as platelet factor 4, is a potent anti-angiogenic chemokine. One study determined that serum CXCL4 levels can be 270-times higher in SSc patients compared with healthy controls and/or an association with decline in DLCO. However, CXCL4 levels were noted to decline at 12 and/or 24 months after immunosuppressive therapy (e.g,, cyclophosphamide and/or mycophenolate), and/or a fall in CXCL4 from baseline-to-12 months was associated with improvement of FV C from 12-to-24 months. In some embodiments, levels of CXCL4 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the presen t disclosure has an increased level of CXCL4 when compared with a subject that does not have SSc-ILD.

In some embodiments, a biomarker for SSc-ILD is selected from the group consisting of VEGF, ET-1, and/or IL-8. In some embodiments, a biomarker for SSc-ILD may be involved in one or more vascular and/or endothelium pathways. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of VEGF, ET-1, and IL-8. In some embodiments, a subject of the present disclosure has an increased level of one or more biomarkers selected from the group consisting of VEGF, ET-1 , and/or IL-8, when compared with a subject that does not have SSc-ILD. Aberrant angiogenesis is generally implicated in the pathogenesis of pulmonary- fibrosis and/or fundamental mediators of this process include vascular endothelial growth factor (VEGF), endothelin 1 (ET-1) and/or interleukin-8 (EL-8). VEGF is a central cytokine and growth factor for endothelial and/or type II AECs. Nintedanib, a tyrosine kinase inhibitor that targets VEGF signaling slows disease progression in IPF. In a recent study of SSc patients, elevated VEGF correlated with disease seventy measured by DLCO and/or HRCT involvement, DL-8 (also known as CXCL8) is produced by phagocytes when exposed to inflammatory stimuli and/or attracts neutrophils and/or promotes angiogenesis.

Endothelin- 1 (ET-1) is a potent vasoactive peptide with diverse properties including vasoconstriction, bronchoconstriction, cell growth, turnover and/or fibroblast activation. Both IL-8 and/or ET-1 are elevated in idiopathic ILD compared with controls and/or have demonstrated correlation with HRCT fibrosis scores, pulmonary function decline and/or reduced overall, transplant-free and/or progression-free survival. In SSc, studies have demonstrated correlation of ET-1 with vascular markers of disease severity including digital ulcers, renal vascular disease and/or pulmonary hypertension. In some embodiments, levels of one or more of VEGF, IL-8, and/or ET-1 are elevated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of one or more of VEGF, IL-8, and/or ET-1 when compared with a subject that does not have SSc-ILD.

Other biomarkers used to determine if a subject has SSc-ILD include, but are not limited to Collal, ColBal, CTGF, fibronectin, PAl-1, and/or Tnfa. in some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a THRU antagonist to a subject in need thereof, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of ADAM 12, Anti-HSP70 IgG, BAFF, BLyS, CaI5.3, CC16, CCL2, CCL18, Collal, Col3al, CTGF, CXCL4, E-selectin, ET-1, fibronectin, ICAM, IE-6, IL-8, KL-6, MCP-1, MMP7, Osteopontin, PAI-1, SP-A, SP-D, Tnfa, VCAM, VEGF, and YKL-40 when compared with a subject that does not have SSc-ILD.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a THRU antagonist to a subject in need thereof, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of Cal5.3, CC16, KL-6, SP-A, and SP-D when compared with a subject that does not have SSc-ILD.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TbίIII antagonist to a subject in need thereof, wherein the subject has an increased level of one or more biomarkers selected from the group consisting of Collal, Co!3al, CTGF, fibronectm, PAI-1, and Tnfa when compared with a subject that does not have SSc-ILD.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TiiRJl antagonist to a subject in need thereof wherein tire subject has a change in level of one or more collagen biomarkers compared to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TbR.II antagonist to a subject in need thereof, wherein the subject has an increase in levels of one or more collagen biomarkers compared to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a sub j ect in need thereof, wherein the subject has an increase in levels of one or more collagen biomarkers when compared with a subject that does not have SSc-ILD. In some embodiments, a collagen biomarker is selected from the group consisting of type I (C1M), type III (C3 A, C3M), type IV (C4M), type V (C5M) and type VI (C6M) collagen degradation biomarkers. In some embodiments, a collagen biomarker is selected from the group consisting of type I (PRO-C1), II (PRG-C2), III (PRO-C3), IV (PRQ-C4), V (PRQ-C5) and VI (PRO-C6) collagen formation biomarkers. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TbKII antagonist to a subject in need thereof, wherein the subject has an increased level of one or more collagen degradation biomarkers selected from the group consisting of type I (C1M), type III (C3A, C3M), type IV (C4M), type V (C5M) and type VI (C6M), when compared with a subject that does not have SSc-ILD. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TbIIII antagonist to a subject in need thereof, wherein the subject has an increased level of one or more collagen formation biomarkers selected from the group consisting of type I (PRO-C1), II (PRO-C2), III (PRO-C3), IV (PRO-C4), V (PRO-C5) and VI (PRQ-C6), when compared with a subject that does not have SSc-ILD.

Alpha-smooth muscle actin (aSMA) is the aetin isoform that predominates within vascular smooth-muscle cells and plays an important role in fibrogenesis. Myofibroblasts are metabolically and morphologically distinctive fibroblasts expressing aSMA, and their activation plays a key role in development of the fibrotic response. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TbRIΐ antagonist to a subject in need thereof, wherein the subject has an increased level of aSMA when compared with a subject that does not have SSc-ILD. The Muc5B promoter polymorphism is a known genetic risk factor for Idopathic Pulmonary Fibrosis (IPF), and is likely involved in disease pathogenesis through an increase in Muc5B expression in terminal bronchi and honeycombed cysts. Expression of MUC5B is also highly correlated with expression of cilium genes in IPF lung. Mucociliary' dysfunction in the distal airway may play a role in the development of progressive fibroproliferative lung disease (Yang, l.V. et al., Ann Am Thorac Soc., 2015, S193-S199). In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TbϊIII antagonist to a subject in need thereof, wherein the subject has an increased level of Muc5B when compared with a subject that does not have SSc-ILD. Similarly, periostin has been shown to be a biomarker for IPF and related diseases.

Monomeric periostin has been identified in diseases such as atopic dermatitis, systemic scleroderma, and asthma. Both monomeric and total periostin have been shown to be correlated with decline of %VC and % DLCO (Qhta, S. et al. PLOS One, 2017, 1-17). In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TjlRII antagonist to a subject in need thereof, wherein the subject has an increased level of periostin when compared with a subject that does not have SSc-ILD.

The Whey Acidic Protein domain (WFDC2 (HEA)) is an evolutionarily conserved motif found in a number of proteins, the best studied of which are antiproteinases involved in the innate immune defense of multiple epithelia. The WFDC2 gene encodes a two WAP domain-containing protein, initially suggested as a marker for epididymis, and it has been shown that it is highly expressed in the lung and salivary gland. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TflRII antagonist to a subject in need thereof, wherein the subject has an increased level of WFDC2 (HEA) when compared with a subject that does not have SSc-ILD.

C-reactive Protein (CRP)

C-reactive protein (CRP) is an annular (ring-shaped), pentamenc protein found in blood plasma, whose circulating concentrations rise in response to inflammation. CRP is an acute -phase protein of hepatic origin that increases following interleukin-6 secretion by macrophages and T cells. Its physiological role is to bind to lysophosphatidylcholine expressed on the surface of dead or dying cells (and some types of bacteria) in order to activate the complement system via Clq. CRP is synthesized by the liver in response to factors released by macrophages and fat cells (adipocytes). In some embodiments, levels of CRP are elevated in a subject that has SSc. In some embodiments, levels of CRP are ele vated in a subject that has SSc-ILD. In some embodiments, a subject of the present disclosure has an increased level of CRP when compared with a subject that does not have SSc (e.g.. SSc-ILD). In some embodiments, a subject of the present disclosure has a CRP level of greater than or equal to 3 mg/L.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TbR. Gί antagonist to a subject in need thereof, wherein the subject has an increased level of CRP.

In some embodiments, a subject of the present disclosure has a CRP level of greater than or equal to 3 mg/L at baseline. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TbKII antagonist to a subject in need thereof, wherein the subject has a CRP level of greater than or equal to 3 mg/L at baseline. Bronchoalveolar lavage (BAL)

Bronchoalveolar lavage (BAL) is performed during flexible bronchoscopy to obtain samples of cells and/or fluid from distal airways and/or alveoli. Lavage fluid is sent for cell counts; cultures for mycobacterial, viral, and/or fungal pathogens; and/or cytologic analysis. BAL is particularly useful in the evaluation of patients with interstitial lung disease (ILD) that is associated with hemoptysis, is acute or rapidly progressive, or is likely caused by one or more of the following diseases: sarcoidosis, hypersensitivity' pneumonitis, pulmonary Langerhans histiocytosis, or infection. Virtually all patients presenting with hemoptysis and/or radiographic ILD are recommended to undergo BAL with sequential lavages to confirm an alveolar source of bleeding and/or identify any infectious etiologies. The majority of patients with an acute onset of ILD will undergo BAL to evaluate for acute eosinophilic pneumonia, alveolar hemorrhage, malignancy, and/or opportunistic or atypical infection, which can often be diagnosed on the basis of BAL findings. For patients with a subacute or chronic presentation of ILD, BAL is often performed when sarcoidosis, hypersensitivity pneumonitis, pulmonary ' Langerhans cell histiocytosis (PLCH), and/or infection are suspected based on the radiographic pattern (eg., upper lobe predominance of reticular opacities, hilar iymphadenopathy, irregular cystic airspaces), history' of exposure (e.g., bird keeping, farming), or concomitant clinical findings (e.g., hemoptysis, renal insufficiency). In these patients, the results of BAL analysis may be used to narrow' the differential diagnostic possibilities between various types of ILD, but tissue confirmation is usually required.

One of the main roles of bronchoscopy and/or broncfaoalveolar lavage (BAL) in subjects with SSc and/or suspected ILD is to exclude other causes of ILD. Thus, for most subjects, these tests are not necessary or ordered. However, for patients with atypical features as mentioned above, such as fever, productive cough, hemoptysis, peripheral blood eosinophilia, or focal consolidation on chest imaging, bronchoscopy with BAL may be indicated.

Patients with SSc-ILD typically have elevated numbers of granulocytes in their BAL fluid, particularly neutrophils and/or eosinophils, and/or may sometimes have an increase in lymphocytes and/or mast cells. BAL in SSc-ILD has sometimes been used to assess the degree of alveolitis and/or predict response to immunosuppressive therapy.

In some embodiments, a subject with SSc-ILD is tested using BAL. In some embodiments, SSc-ILD in a subject is confirmed using BAL. in some embodiments, a BAL exam of a subject is used to assess the degree of alveolitis and/or predict response to immunosuppressi ve therapy .

Lung Biopsy and Pathology

Skin biopsies of subjects suspected to have SSc (e.g. SSc-ILD) are not commonly utilized. However, skin biopsies can be useful in differentiating SSc (e.g. SSc-ILD) from other skin-related diseases or changes. SSc-ILD is characterized by excessive disposition of compact bundles of collagen in the dermal layers. Dermal expansion, edema, perivascular mononuclear inflammatory 7 cell infiltration, and/or fibrosis can be seen histologically in a skm biopsy sample. Loss of intradermal fat can be seen, as well as displacement of white adipose by fibrotic tissue. Lung biopsies are typically obtained via video-assisted thoracoscopic surgery.

Radiologic pattern on HRCT is an important consideration when deciding whether a patient should undergo a surgical lung biopsy. Subjects with a UIP pattern on HRCT generally demonstrates basal predominant (occasionally diffuse) and/or subpleural predominant, often heterogeneous reticular opacities with peripheral traction bronchiectasis or bronchiolectasis, honeycombing, and/or absence of features to suggest an alternative diagnosis. Consistently, a UIP pattern on HRCT is very 7 specific in predicting a UIP pattern on histopathology, and therefore a surgical lung biopsy is not recommended in subjects with a UIP pattern on HRCT. A “probable” UIP patern on HRCT is similar to that of a UIP pattern except honeycombing is absent; i.e., reticular opacities are predominantly basal and/or subpleural, with peripheral traction bronchiectasis or bronchiolectasis and/or a heterogeneous distribution. In this case, obtaining a surgical lung biopsy in subjects with a probable UIP radiologic pattern is recommended to further assist diagnosis.

Fibrotic N SIP is typically the most frequently noted histopathologic finding on lung biopsy in SSc-ILD. Within the cl assification of NSIP, variable amounts of mononuclear cell infiltration and/or alveolar septal fibrosis can be seen. In S Sc-associated NSIP, the pattern is more fibrotic and less inflammatory, thus leading to the designation fibrotic NSIP, rather than cellular NSIP. Additional characteristics of NSIP are temporal homogeneity and/or absence of fibroblast foci.

A pattern consistent with UIP is occasionally seen in SSc-ILD. When compared with lung biopsies from patients with idiopathic pulmonary fibrosis, biopsies of SSc-associated UIP have a lower number of fibroblast foci and/or a higher number of germinal centers. In some embodiments, a subject with SSc-ILD has a histologic pattern of fibrotic

NSIP in a lung biopsy. In some embodiments, a subject with SSc-ILD has a histologic pattern of UIP in a lung biopsy. In some embodiments, a subject with a probable UIP patern on HRCT is a candidate for a lung biopsy. Composite Response Index in Systemic Sclerosis (CRISS)

The Composite Response Index in Systemic Sclerosis (CRISS) is a 2-step process for measuring the probability of improvement of patients typically with diffuse cutaneous systemic sclerosis (dcSSc), with values ranging from 0.0 (no improvement) to 1.0.

In step 1, patients who develop one of the following attributable to systemic sclerosis that require treatment: (1) new onset of renal crisis, (2) new 7 onset or worsening of lung fibrosis, (3) new' onset of pulmonary arterial hypertension (measured by right sided heart cathertization), or (4) new' onset of left ventricular failure (e.g., a left ventricular ejection fraction of <45%); during a clinical trial, are considered as not improved and assigned a probability of improving equal to 0.0. Specifically regarding lung fibrosis, if a subject has an FVC of less than 80% predicted and develops an annual rate of decline in FVC of >15% of predicted, confirmed by a subsequent FVC test within a month, and further confirmed to have ILD by HRCT, then a subject is deemed to have no improvement and will have a CRISS score of 0.0. For the remaining patients with complete data after a clinical trial is complete, step 2 involves computing the predicted probability of improving for each individual, using an equation that involves change in modified rodnan skin score (mRSS) from baseline, change in FVC % predicted from baseline, change in patient global assessment, change in physical/physician global assessment, and change in health assessment questionnaire- disability index (HAQ-DI). Subjects in step 2 for whom the predicted probability (CR1SS score) is >0.60 are considered improved, while subjects for whom the predicted probability (CRISS score) is <0.60 are considered not improved.

In some embodiments, a subject is evaluated using a patient global assessment. In some embodiments, a subject is evaluated using a physical/physician global assessment. In some embodiments, a patient is evaluated using a health assessment questionnaire -disability' index (HAQ-DI). In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TflRlI antagonist to a subject in need thereof, wherein the antagonist improves the subject’s patient global assessment score compared to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TflRII antagonist to a subject in need thereof, wherein the antagonist improves the subject’s physical/physician global assessment score compared to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpiRli antagonist to a subject in need thereof, wherein the antagonist improves the subject’s HAQ-DI score compared to a baseline measurement.

The 2 groups (study drug versus placebo or active comparator) can then be compared in a 2 V 2 table using appropriate significance tests. The predicted probabilities obtained using CRISS can also be assessed as a continuous variable, and the distributions of the probability of improving for patients receiving study drug versus placebo can be compared using nonparametric tests.

In some embodiments, a subject with SSc has a CRISS score of 0.0. In some embodiments, a subject with a CRISS score of 0.0 is considered not improved. In some embodiments, a subject with SSc has a CRISS score of 1.0. In some embodiments, a subject with a CRISS score of 1.0 compared to a baseline measurement is considered improved. In some embodiments, a subject with a predicted probability (CRISS score) in step 2 of >0.60 compared to a baseline measurement is considered improved. In some embodiments, a subject with a CRISS score of about 0.60 to about 1.0 compared to a baseline measurement is considered improved. In some embodiments, a subject with a predicted probability (CRISS score) in step 2 of <0.60 compared to a baseline measurement is not improved. In some embodiments, a subject with a CRISS score of about 0.0 to about 0.60 compared to a baseline measurement is considered not improved.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TflRH antagonist to a subject in need thereof, wherein the antagonist increases tire subject’s CRISS score compared to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TjlRII antagonist to a subject in need thereof, wherein the antagonist increases the subject’s CRISS score to between about 0.60 and about 1.0, compared to a baseline measurement.

St. George ’s Respiratory Questionnaire (SGRQ)

In some embodiments, a subject of the present disclosure may be evaluated using a respiratory questionnaire. In some embodiments, a subject of the present disclosure may be evaluated using a St. George’s Respiratory Questionnaire (SGRQ). In some embodiments, an SGRQ evaluates one or more of overall health, daily life, and/or perceived well-being of a subject with one or more obstructive airway diseases (e.g., SSc-ILD). An SGRQ score ranges from 0 to 100 units, with higher scores indicating more limited respirator} 7 function. Questions in an SGRQ are typically sorted into two categories comprising complications and activities that can cause and/or are limited by breathlessness. Categories can be split into multiple parts. Questions can have one or more methods of scoring, including but not limited to scaling, dichotomous true/false, and/or four-point Likert scale. In some embodiments, a subject’s SGRQ score positively correlates with presence of cough, sputum and/or wheeze in a subject. In some embodiments, a subject’s SGRQ score correlates with one or more measures of disease acti vity including but not limited to FEV1, FVC, Sa02 at rest, 6-MWD, MRC dyspnea grade, anxiety score, depression score. Sickness Impact Profile (SIP) total score, and SIP physical and/or psychosocial domains. Generally, a mean change in score of about 4 units indicates slightly efficacious treatment, a mean change in score of about 8 units indicates moderately efficacious treatment, and a mean change in score of about 12 units indicates very efficacious treatment. In some embodiments, efficacious treatment comprises a decrease in an SGRQ score of a subject. In some embodiments, treatment of a subject with one or more of a T[1RII antagonist of the present disclosure changes a subject’s SGRQ score by about 4 units. In some embodiments, treatment of a subject with one or more of a TBRH antagonist of the present disclosure changes a subject’s SGRQ score by about 8 units. In some embodiments, treatment of a subject with one or more of a T[1R1I antagonist of tire present disclosure changes a subject’s SGRQ score by about 12 units. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure changes a subject’s SGRQ score by between about 0 and about 4 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s SGRQ score by between about 1 and about 4 units. In some embodiments, treatment of a subject with one or more of a TbίIII antagonist of the present disclosure changes a subject’s SGRQ score by between about 5 and about 8 units. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure changes a subject’s SGRQ score by between about 9 and about 12 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s SGRQ score by about more than 12 units.

In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject. In some embodiments, treatment of a subject w 7 ith one or more of a TbH Gί antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 1% and about 3%. In some embodiments, treatment of a subject with one or more of a TpRH antagonist of the present disclosure improves a St. George’s Respiratory 7 Questionnaire (SGRQ) score of a subject by between about 1% and about 5%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a St. George ' s Respiratory 7 Questionnaire

(SGRQ) score of a subject by between about 1% and about 8%. In some embodiments, treatment of a subject with one or more of a TbRII antagonist of the present disclosure improves a St. George’s Respiratory 7 Questionnaire (SGRQ) score of a subject by between about 1% and about 10%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a St. George’s Respiratory 7

Questionnaire (SGRQ) score of a subject by between about 10% and about 15%. In some embodiments, treatment of a subject with one or more of a TbίIII antagonist of the present disclosure improves a St. George’s Respiratory 7 Questionnaire (SGRQ) score of a subject by between about 15% and about 20%. In some embodiments, treatment of a subject with one or more of a TfiRH antagonist of the present disclosure improves a St. George’s Respiratory 7 Questionnaire (SGRQ) score of a subject by between about 20% and about 25%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 25% and about 30%. In some embodiments, treatment of a subject with one or more of a TbKίί antagonist of the present disclosure improves a St. George’s Respiratory- Questionnaire (SGRQ) score of a subject by between about 30% and about 35%. In some embodiments, treatment of a subject with one or more of a ' TbKII antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 35% and about 40%. In some embodiments, treatment of a subject with one or more of a T ' bKII antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 40% and about 45%. In some embodiments, treatment of a subject with one or more of a TOR If antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 45% and about 50%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a St. George’s Respiratory ' Questionnaire (SGRQ) score of a subject by between about 55% and about 60%. In some embodiments, treatment of a subject with one or more of a TbίIII antagonist of the present di sclosure improves a St. George’s Respiratory ' Questionnaire (SGRQ) score of a subject by between about 60% and about 65%. In some embodiments, treatment of a subject with one or more of a TbKίί antagonist of the present disclosure improves a St. George’s Respiratory' Questionnaire (SGRQ) score of a subject by between about 65% and about 70%. In some embodiments, treatment of a subject with one or more of a TbRII antagonist of the present disclosure improves a St. George’s Respiratory- Questionnaire (SGRQ) score of a subject by between about 10% and about 20%. In some embodiments, treatment of a subject with one or more of a TbKίϊ antagonist of the present disclosure improves a St. George’s Respiratory- Questionnaire (SGRQ) score of a subject by between about 20% and about 30%. In some embodiments, treatment of a subject with one or more of a TpiRli antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 30% and about 40%. In some embodiments, treatment of a subject with one or more of a TpR.fl antagonist of the present disclosure improves a St. George’s Respiratory Questionnaire (SGRQ) score of a subject by between about 40% and about 50%. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure improves a St. George’s Respiratory' Questionnaire (SGRQ) score of a subject by between about 60% and about 70%. In some embodiments, an SGRQ score is improved after the administering of one or more of a TbKII antagonists of the present disclosure, compared to a baseline measurement. In some embodiments, an SGRQ score is decreased after the administering of one or more of a TbRII antagonists of the present disclosure, compared to a baseline measurement. King ’s Brief Interstitial Lung Disease (KBJLD) Questionnaire

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a ' TbKIί antagonist to a subject in need thereof, wherein the subject is evaluated using the King’s Brief Interstitial Lung Disease (KBILD) questionnaire, In some embodiments, the disclosure provides a method of treating SSc (e.g. SSc-ILD), comprising administering a TjlR!l antagonist to a subject in need thereof, wherein the administration improves subject’s KBILD score.

The King’s Brief Interstitial Lung Disease (KBILD) is a 15 -item validated health- related quality of life (HRQOL) questionnaire. The KBILD questionnaire consists of three domains (breathlessness and activities, chest symptoms and psychological). The method of scoring the KBILD has recently changed to incorporate a logit-scale transformation from one that used raw' item responses, as this is potentially a more linear scale. The KBILD questionnaire help to measure the impact ILD has on a subject ' s wellbeing and daily life. KBILDs can be used to monitor and assess how well treatment for interstitial lung disease associated conditions (e.g., SSc-ILD) is working. Each question in the KBILD can be answered with a rank from 1 to 7, with 1 being ‘ all of the time” and 7 being “none of the time”. The lowest score a subject can obtain is 15 points. The highest score a subject can obtain is 105 points.

In some embodiments, a subject of the present disclosure may be evaluated using a respiratory questionnaire. In some embodiments, a subject of the present disclosure may be evaluated using a King’s Brief Interstitial Lung Disease (KBILD) questionnaire. In some embodiments, an KBILD evaluates one or more of overall heal th, daily life, response to treatment and/or perceived well-being of a subject with interstitial lung disease (e.g., SSc- ILD). In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 1 unit. In some embodiments, treatment of a subject with one or more of a TbίIII antagonist of the present disclosure changes a subject’s KBILD score by about 3 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 7 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 10 units, in some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 14 units. In some embodiments, treatment of a subject with one or more of a TpR.H antagonist of the present disclosure changes a subject’s KBILD score by about 21 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 28 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 35 units. In some embodiments, treatment of a subject with one or more of a TfiRII antagonist of tire present disclosure changes a subject’s KBILD score by about 42 units. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure changes a subject’s KBILD score by about 49 units. In some embodiments, treatment of a subject with one or more of a ' TbKIί antagonist of the present disclosure changes a subject’s KBILD score by about 56 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 63 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of tire present disclosure changes a subject’s KBILD score by about 70 units. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure changes a subject’s KBILD score by about 77 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 84 units. In some embodiments, treatment of a subject with one or more of a TBRJl antagonist of the present disclosure changes a subject’s KBILD score by about 91 units. In some embodiments, treatment of a subject with one or more of a THRU antagonist of the present disclosure changes a subject’s KBILD score by about 98 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject’s KBILD score by about 105 units.

In some embodiments, treatment of a subject with one or more of a TbKIί antagonist of the present disclosure changes a subject’s KBILD score by between about 0 and about 3 units. In some embodiments, treatment of a subject with one or more of a TBRJl antagonist of the present disclosure changes a subject’s KBILD score by between about I and about 7 units. In some embodiments, treatment of a subject with one or more of a TBRJl antagonist of the present disclosure changes a subject’s KBILD score by between about 7 and about 14 units. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure changes a subject ' s KBILD score by between about 14 and about 21 units. In some embodiments, treatment of a subject with one or more of a TbKP antagonist of the present disclosure changes a subject’s KBILD score by about more than 21 units. In some embodiments, treatment of a subject with one or more of a TpR [ f antagonist of the present disclosure improves a KBILD score of a subject. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 1% and about 3%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 1% and about 5%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 1% and about 8%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 1% and about

10%. In some embodiments, treatment of a subject with one or more of a TbKP an tagonist of the present disclosure improves a KBILD score of a subject by between about 10% and about 15%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a KBILD score of a subject by between about 15% and about 20%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a KBILD score of a subject by between about 20% and about 25%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a KBILD score of a subject by between about 25% and about 30%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a KBILD score of a subject by between about 30% and about 35%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 35% and about 40%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 40% and about 45%. In some embodiments, treatment of a subject with one or more of a TbKP an tagonist of the present disclosure improves a KBILD score of a subject by between about 45% and about 50%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 55% and about 60%. In some embodiments, treatment of a subject with one or more of a TpR 11 antagonist of the present disclosure improves a KBILD score of a subject by between about 60% and about 65%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves a KBILD score of a subject by between about 65% and about 70%. In some embodiments, treatment of a subject with one or more of a TbKP an tagonist of the present disclosure improves a KBILD score of a subject by between about 10% and about 20%. In some embodiments, treatment of a subject with one or more of a TiiRI I antagonist of the present disclosure improves a KBILD score of a subject by between about 20% and about 30%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a KBILD score of a subject by between about 30% and about 40%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a KBILD score of a subject by between about 40% and about 50%. In some embodiments, treatment of a subject with one or more of a TbKIί antagonist of the present disclosure improves a KBILD score of a subject by between about 60% and about 70%. In some embodiments, a KBILD score is improved after the administering of one or more of a ' TbKII antagonists of the present disclosure, compared to a baseline measurement.

In some embodiments, an KBILD score is increased after the administering of one or more of a TpRH antagonists of the present disclosure, compared to a baseline measurement.

Modified Rodnan Skin Score (mRSS) In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-

ILD), comprising administering a TbKIί antagonist to a subject in need thereof, wherein the subject is evaluated for skin thickness using a modified rodnan skin score (mRSS). In some embodiments, the disclosure provides a method of treating SSc (e.g. SSc-ILD), comprising administering a TbKIί antagonist to a subject in need thereof, wherein the mRSS is an average of measurements of multiple areas on the subject.

A modified Rodnan Skin Score (mRSS) evaluates a subject for extent of skin thickness. In some embodiments, mRSS is measured by skin palpitation. In some embodiments, skin palpitation is performed by pinching skin with fingers. In some embodiments, mRSS is measured by a scale of 0 to 3 for each area of palpitation. In some embodiments, an mRSS of 0 comprises an indication of normal skin comprising appearance of fine wrinkles and not skin thickness. In some embodiments, an mRSS of 1 comprises an indication of mild skin thickness comprising skin folds easily made between two fingers. In some embodiments, an mRSS of 2 comprises an indication of moderate skin thickness comprising difficulty in making skin folds and no wrinkles are present. In some embodiments, an mRSS of 3 comprises an indication of severe thickness comprising an inability to make skm folds between 2 examining fingers. In some embodiments, several areas are evaluated for an mRSS. In some embodiments, 17 surface anatomic areas are evaluated for an mRSS. In some embodiments, a representative area (e.g., most representative score of a specific area) of a subject is evaluated for an mRSS. In some embodiments, a global average (e.g., score individual areas and take average of the area as a whole) is calculated for an mRSS.

In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 5. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 10. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 15. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 20. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 25. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 30. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 35. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 40. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 45. In some embodiments, a subject of the present disclosure has a total mRSS baseline measurement of greater than or equal to 50. In some embodiments, a subject of the present disclosure has a total mRSS of greater than or equal to 15 at baseline. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the subject has an mRSS of greater than or equal to 15 at baseline.

In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves a modified rodnan skin score (mRSS) of the subject. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves an mRSS of the subject. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves an mRSS of the subject by between about 1% and about 3%. In some embodiments, treatment of a subject with one or more of a TbKP antagonist of the present disclosure improves an mRSS of the subject by between about 1 % and about 5%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves an mRSS of the subject by between about 1% and about 8%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves an mRSS of the subject by between about 1% and about 10%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure impro ves an mRSS of the subject by between about 10% and about 15%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure improves an mRSS of the subject by between about 15% and about 20%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves an mRSS of the subject by between about 20% and about 25%. In some embodiments, treatment of a subject with one or more of a TbMI antagonist of the present disclosure impro ves an mRSS of the subject by between about 25% and about 30%. In some embodiments, treatment of a subject with one or more of a TbKP antagonist of the present disclosure improves an mRSS of the subject by between about 30% and about 35%. In some embodiments, treatment of a subject with one or more of a ' TbKII antagonist of the present disclosure improves an mRSS of the subject by between about 35% and about 40%. In some embodiments, treatment of a subject with one or more of a T ' BRH antagonist of the present disclosure improves an mRSS of the subject by between about 40% and about 45%. In some embodiments, treatment of a subject with one or more of a TbKP antagonist of the present disclosure improves an mRSS of the subject by between about 45% and about 50%. In some embodiments, treatment of a subject with one or more of a TPRH antagonist of the present disclosure improves an mRSS of the subject by between about 55% and about 60%. In some embodiments, treatment of a subject with one or more of a TpRII antagonist of the present disclosure improves an mRSS of the subject by between about 60% and about 65%. In some embodiments, treatment of a subject with one or more of a TbKII antagonist of the present disclosure impro ves an mRSS of the subject by between about 65% and about 70%. In some embodiments, treatment of a subject with one or more of a TbKP antagonist of the present disclosure improves an mRSS of the subject by between about 10% and about 20%. In some embodiments, treatment of a subject with one or more of a TpRil antagonist of the present disclosure improves an mRSS of the subject by between about 20% and about 30%. In some embodiments, treatment of a subject with one or more of a T ' BRH antagonist of the present disclosure improves an mRSS of the subject by between about 30% and about 40%. In some embodiments, treatment of a subject with one or more of a TbKP antagonist of the present disclosure improves an mRSS of the subject by between about 40% and about 50%, In some embodiments, treatment of a subject with one or more of a TPRH antagonist of the present disclosure improves an mRSS of the subject by between about 60% and about 70%. In some embodiments, a mRSS is improved after the administering of one or m ore of a TbKΪΪ antagonists of the present disclosure, compared to a baseline measurement. In some embodiments, an mRSS is decreased after the administering of one or more of a TbKII antagonists of the present disclosure, compared to a baseline measurement.

Health Assessment Questionnaire Disability Index (HAQ-DI) In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-

1LD), comprising administering a TbίIII antagonist to a subject in need thereof, wherein the subject is evaluated using a Health Assessment Questionnaire Disability Index (HAQ-DI).

The Health Assessment Questionnaire-Disability (HAQ-DI) consists of 20 questions referring to eight component sets: dressing/grooming, arising, eating, walking, hygiene, reach, grip and activities. The total score indicates the patient’s self-assessed level of disability. A negative change from baseline indicates improvement.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- 1LD), comprising administering a TbίIII antagonist to a subject in need thereof, wherein the antagonist improves the subject’s HAQ-DI score compared to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g, SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist imparts a negative change in HAQ-DI score of a subject compared to a baseline measurement. Physical/Physician Global Assessment and Patient Global Assessment

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TbMΪ antagonist to a subject in need thereof, wherein the subject is evaluated using a physical/physician global assessment.

The physician’s global assessment is to be completed on tire basis of examination and overall assessment of the patient after all other trial procedures have been completed. The physician’s assessment of the patient’s SSc status will be scored on a 100-mrn horizontal visual analog scale (VAS).

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TbMΪ antagonist to a subject in need thereof, wherein the antagonist improves the subject’s physical/physician global assessment score compared to a baseline measurement.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TbKII antagonist to a subject in need thereof, wherein the subject is evaluated using a patient global assessment. The patient’s global assessment represents the patient’s (e.g., subject’s) overall assessment of his or her current SSc status on a 100-mm horizontal visual analog scale (VAS). The patient’s global assessment is self-administered by the patient.

In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist improves the subject’s patient global assessment score compared to a baseline measurement.

Clinical W r jrs erring In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-

TLD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist improves the time to clinical worsening of the subject. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpR!l antagonist to a subject in need thereof, wherein the antagonist lengthens the time to clinical worsening of the subject. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist slows the rate of decline of pulmonary function of the subject. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist extends the subject’s length of life compared to a baseline measurement prior to the administering. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist prevents a decline in FVC of the subject of greater than 10% of predicted relative to a baseline measurement. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist prevents at least a greater than 20% increase in mRSS of the subject. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc-ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist prevents at least a greater than 5 point increase in total mRSS of the subject. In some embodiments, the disclosure provides methods of treating SSc (e.g. SSc- ILD), comprising administering a TpRII antagonist to a subject in need thereof, wherein the antagonist prevents an occurrence of an SSc-related complication in the subject. Combination Therapies

In certain aspects, the disclosure contemplates the use of a TbIIΪI antagonist in combination with one or more additional active agents or other supportive therapy for treating systemic sclerosis or a complication of SSc (e.g., SSc-ILD).

As used herein, "‘in combination with”, "‘combinations of”, “combined with”, or “conjoint” administration refers to any form of administration such that additional active agents or supportive therapies (e.g., second, third, fourth, etc.) are still effective in the body (e.g., multiple compounds are simultaneously effective in the patient for some period of time, which may include synergistic effects of those compounds). Effectiveness may not correlate to measurable concentration of the agent in blood, serum, or plasma. For example, the different therapeutic compounds can be administered either in tire same formulation or in separate formulations, either concomitantly or sequentially, and on different schedules. Thus, a subject who receives such treatment can benefit from a combined effect of different active agents or therapies. One or more TflRH antagonists of the disclosure can be administered concurrently with, prior to, or subsequent to, one or more other additional agents or supportive therapies, such as those disclosed herein. In general, each active agent or therapy will be administered at a dose and/or on a time schedule determined for that particular agent. The particular combination to employ in a regimen will take into account compatibility of the TbMIΊ antagonist of the present disclosure with the additional active agent or therapy and/or the desired effect,

In some embodiments, a patient has been treated with one or more active agents, which is not a ' TbKII antagonist, or other supportive therapy for treating SSc, or a complication of SSc (e.g., SSc-ILD) prior to administration of a Tj!R!l antagonist. In some embodiments, a patient was not previously taking one or more other acti ve agents or supportive therapies for treating SSc, or a complication of SSc (e.g., SSc-ILD) prior to administration of a TbKII antagonist. In some embodiments a patient is administered a Tb R 11 antagonist in combination with one or more additional active agents and/or supportive therapies for treating SSc, or a complication of SSc (e.g., SSc-ILD).

In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises abatacept (e.g., orencia). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises abituzumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc- ILD) comprises ajuiemic acid (e.g,, anahasum or lenabasum or resunab). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises ambrisentan. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises AVID200. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises AVID300. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises azathioprine (e.g., imuran). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises BCD- 089. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises belimumab (e.g., benlysta). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises BG0001I. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc- ILD) comprises BMS-986020. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises bortezomib (e.g., velcade). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises bosentan. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises brentuximab (e.g., adcetris). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises carlumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises CC-90001. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises clazakizumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g,, SSc-ILD) comprises COR-001. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises cyclophosphamide or CYC. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises cyclosporine A. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises dectrekumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises EHP-101. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises elzonris or SL-401. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises etanercept. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises FCX-013. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises fresohmumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises GLPG1690. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises GSK2I26458. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises GSK2330811 . In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises GSK3008348. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises 1BIO-CFB03. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises ifetroban (e.g., vasculaii). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises IFNy. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises imatmib. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises immune globulin (e.g., pri vigen). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises IWOOL In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises lanifibranor. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises lebrikizumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises ievilimab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises losartan. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises macitentan. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (eg., SSc- ILD) comprises MEDI-5117. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises methotrexate. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises MSCs. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises mycophenolate mofetil or MMF. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises MAC. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises nandroione decanoate. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises nintedanib (e.g., ofev). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises olokizumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises pamrevlumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises pirfenidone (e.g., esbriei). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises pirfenidone and vismodegib. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises pomahdomide (e.g., pomalyst). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises PRM-151. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises riociguat (e.g., adempas). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises rituximab (e.g., rituxan). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises SAR156597. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises sildenafil. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises siltuximab (e.g., sylvant). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises simtuzumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises sirolirnus. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises sirukumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises tacrolimus. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises tadaiafi!. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises tanzisertib. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises TD139. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises tetrathiomolybdate. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises tocilizumab (e.g., actemra). In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises tralokmumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises treprostinii. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises vobarilizumab. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises warfarin. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises ziieuton. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g,, SSc-ILD) comprises ziltivekimab. In some embodiments, a subject has been treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of abatacept, abituzumab, ajulemic acid, ambiisentan, AVID20Q, AVID300, azathioprine, BCD-089, behmumab, BG00011, BMS-986020, bortezomib, bosentan, brentuximab, carlumab, CC- 90001, clazakizumab, COR-001, cyclophosphamide (CYC), cyclosporine A, dectrekumab,

EHP-101, elzonris/SL-401, etanercept, FCX-013, fresolimumab, GLPG1690,

GASK2126458, GSK2330811, GSK3008348, IB1G-CFB03, ifetroban, IFNy, imatinib, immune globulin, IWOOl, lanifibranor, lebrikizumab, levilimab, losartan, macitentan, MEDI- 5117, methotrexate, MSCs, mycophenolate mofetil (MMF), NAC, nandrolone decanoate, nintedanib, olokizumab, pamrevlumab, pirfenidone, pirfenidone and vismodegib, pomalidomide, PRM-151. riociguat, rituximab, SAR156597, sildenafil, siltuximab, simtuzumab, sirolimus, sirukurnab, tacrolimus, tadalafd, tanzisertib, TD139, tetrathiomolybdate, tocilizumab, tralokinumab, treprostinil, vobarilizumab, warfarin, zileuton, and ziltivekimab. In some embodiments, a subject is further treated with a therapeutic agent for treating

SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of abatacept, abituzumab, ajulemic acid, AVID200, AVID300, azathioprine, BCD-089, belimumab, bortezomib, bosentan, brentuximab, clazakizumab, COR-001, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL-401, FCX-013, GLPG1690, GSK2330811, IBIO- CFB03, ifetroban, imatinib, immune globulin, lanifibranor, levilimab, MEDI-5117, mycophenolate mofetil (MMF), olokizumab, pirfenidone, pomalidomide, riociguat, rituximab, SAR156597, siltuximab, sirukurnab, tacrolimus, tadalafd, tocilizumab, vobarilizumab, and/or ziltivekimab.

In some embodiments, a subject has been treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of abatacept, abituzumab, ajulemic acid, AVID200, AYID300. azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL- 401, FCX-013, GLPG1690, GSK2330811, IBIO-CFB03, ifetroban, imatinib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, pri vigen, riociguat, rituximab, SAR156597, tacrolimus, tadalafd, and/or tocilizumab.

In some embodiments, a subject is further treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of abatacept, abituzumab, ajulemic acid, AVID200, AVID300, azathioprine, belimumab, bortezomib, bosentan, brentuximab, cyclophosphamide (CYC), cyclosporine A, EHP-101, elzonris/SL- 401, FCX-013, GLPG1690, GSK2330811, IBIO-CFBQ3, ifetroban, miatraib, lanifibranor, methotrexate, mycophenolate mofetil (MMF), nintedanib, pirfenidone, pomalidomide, privigen, riociguat, rituximab, SARI 56597, tacrolimus, tadalafil, and/or tocilizumab.

In some embodiments, a subject has been treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and/or mntedanib. In some embodiments, the disclosure provi des methods of treating SSc-ILD, comprising administering a TbKII antagonist to a subject in need thereof, wherein the method further comprises administration of one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and/or nintedanib.

In some embodiments, a subject is further treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and/or nintedanib. In some embodiments, the disclosure provi des methods of treating SSc-ILD, comprising administering a TbKII antagonist to a subject in need thereof, wherein the method further comprises administration of one or more of azathioprine, cyclophosphamide (CYC), methotrexate, mycophenolate mofetil (MMF), and/or nintedanib.

In some embodimen ts, administration of one or more TbKII antagonists of the present disclosure slows tire annual rate of decline in FVC compared to a subject administered standard of care (SOC). In some embodiments, standard of care comprises administration of an immunosuppressive therapy. In some embodiments, standard of care comprises administration of mycophenolate mofetil (MMF), In some embodiments, standard of care comprises administration of methotrexate. In some embodiments, standard of care comprises administration of cyclophosphamide. In some embodiments, standard of care comprises administration of nintedanib (Ofev). In some embodiments, standard of care comprises administration of rituximab. In some embodiments, standard of care comprises administration of one or more of mycophenolate mofetil (MMF), methotrexate, cyclophosphamide, nintedanib (Ofev), and rituximab. In some embodiments, standard of care comprises administration of at least one therapy selected from the group consisting of mycophenolate mofetil (MMF), methotrexate, cyclophosphamide, nintedanib (Ofev), and rituximab. In some embodiments, administration of one or more TbKII antagonists of the present disclosure slows the annual rate of decline in FVC compared to a subject who has had a lung transplant. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises one or more of 1L-6 and/or/or 1L-6R antibodies. In some embodiments, IL-6 antibodies comprise one or more of clazakizumab, CQR-OOi, MEDI- 5117, olokizumab, siltuximab, sirukumab, and/or ziltivekimab. In some embodiments, IL-6R antibodies comprise one or more of BCD-089 (levilimab), tocilizumab, and/or vobarilizumab.

In some embodiments, a subject has been treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of clazakizumab, COR-OOI, MEDI-51I7, olokizumab, siltuximab, sirukumab, and/or ziltivekimab.

In some embodiments, a subject is further treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of clazakizumab, COR-OOI, MEDI-5117, olokizumab, siltuximab, sirukumab, and/or ziltivekimab

In some embodiments, a subject has been treated with a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of BCD-089 (levilimab), tocilizumab, and/or vobarilizumab. In some embodiments, a subject is further treated with a therapeutic agent for treating

SSc, or a complication of SSc (e.g., SSc-ILD) comprising one or more of BCD-089 (levilimab), tocilizumab, and/or vobarilizumab.

In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD)) targets IL-3R. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) comprises an endostatin. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets PPARy/CB2. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets MMP-1 cells. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets autotaxin. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g,, SSc-ILD) targets sGC. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets IL-4/IL-13. In some embodiments, a therapeutic agent for treating SSe, or a complication of SSc (e.g., SSc-ILD) targets IL-6/IL-6R. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets TPR. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets oncostatin M. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSe (e.g., SSc-ILD) targets p38. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets FGFR/PDGFR. In some embodiments, a therapeutic agent for treating SSc, or a complication of SSc (e.g., SSc-ILD) targets CB1/CB2.

The present disclosure contemplates the use of TbKII antagonists in combination with one or more supportive therapies for treating scleroderma and/or/or SSc-ILD, In some embodiments, a supportive therapy for treating SSc, or a complication of SSc (e.g., SSc-ILD) is a hematopoietic stem cell transplantation (HSCT). In some embodiments, one or more TjlR!l antagonists of the present disclosure are administered prior to a hematopoietic stem cell transplantation. In some embodiments, one or more TbMΪ antagonists of the present disclosure are administered during a hematopoietic stem cell transplantation. In some embodiments, one or more TbKIί antagonists of the present disclosure are administered after a hematopoietic stem cell transplantation.

In some embodiments, a supportive therapy for treating SSc, or a complication of SSc (e.g., SSc-ILD) is a lung transplant. In some embodiments, one or more TbKII antagonists of the present disclosure are administered prior to a lung transplant. In some embodiments, one or more TbKII antagonists of the present disclosure are administered during a lung transplant. In some embodiments, one or more TbMί antagonists of the present disclosure are administered after a lung transplant.

In some embodiments, a subject with SSc (e.g,, SSc-ILD) may have one or more additional fibrotic diseases or conditions. As used herein, the term "fibrosis" or “fibrotic” refers to tire aberrant formation or de velopment of excess fibrous connecti ve tissue by ceils in an organ or tissue. Although processes related to fibrosis can occur as part of normal tissue formation or repair, dysreguiation of these processes can lead to altered cellular composition and/or excess connective tissue deposition that progressively impairs to tissue or organ function. The formation of fibrous tissue can result from a reparative or reactive process. As used herein, the terms "fibrotic disorder", "fibrotic condition," and/or "fibrotic disease," are used interchangeably to refer to a disorder, condition or disease characterized by fibrosis. Additional fibrotic diseases or conditions include, but are not limited to, fibroproliferative disorders associated with vascular diseases, such as cardiac disease, cerebral disease, and/or peripheral vascular disease, as well as tissues and/or organ systems including the heart, skin, kidney, peritoneum, gut, and/or liver (as disclosed in, e.g., Wynn, 2004, Nat Rev 4:583-594, incorporated herein by reference). Exemplary additional fibrotic diseases or conditions that can be treated include, but are not limited to, renal fibrosis, including nephropathies associated with injury/fibrosis, e.g., chronic nephropathies associated with diabetes (e.g., diabetic nephropathy), lupus, glomerular nephritis, focal segmental glomerular sclerosis, and/or IgA nephropathy; gut fibrosis, e.g., radiation-induced gut fibrosis; liver fibrosis, e.g., cirrhosis, alcohol-induced liver fibrosis, biliary duct injury, primary biliary cirrhosis, infection or viral-induced liver fibrosis, congenital hepatic fibrosis and/or autoimmune hepatitis; and/or other fibrotic conditions, such as cystic fibrosis, endomyocardial fibrosis, mediastinal fibrosis, sarcoidosis, spinal cord injury /fibrosis, myelofibrosis, vascular restenosis, atherosclerosis, , injection fibrosis (which can occur as a complication of intramuscular injections, especially in children), endomyocardial fibrosis, retroperitoneal fibrosis, nephrogenic systemic fibrosis, vascular fibrosis, pancreatic fibrosis, liver fibrosis (e.g., cirrhosis), renal fibrosis, musculoskeletal fibrosis, cardiac fibrosis (e.g., endomyocardial fibrosis, idiopathic myocardiopathy), skin fibrosis (e.g., , post-traumatic, operative cutaneous scarring, keloids and/or cutaneous keloid formation), eye fibrosis (e.g., glaucoma, sclerosis of the eyes, conjunctival and/or comeal scarring, and/or pterygium), myelofibrosis, chronic graft-versus-host disease, Peyronie’s disease, post-cystoscopic urethral stenosis, idiopathic and/or pharmacologically induced retroperitoneal fibrosis, mediastinal fibrosis, proliferative fibrosis, neoplastic fibrosis, Dupuytren's disease, strictures, neural scarring, dermal scarring, idiopathic pulmonary fibrosis and/or radiation induced fibrosis.

In certain aspects, any of the TbKίϊ antagonists disclosed herein may be used, alone or in combination with one or more supportive therapies or active agents, treat one or more additional diseases or conditions occurring in a subject with SSc (e.g. SSc-ILD), comprising myelofibrosis (e.g., primary myelofibrosis, post-polycythemia vera myelofibrosis, and/or post-essential thrombocythemia myelofibrosis). In particular, ActRIIB antagonists may be used, alone or in combination with one or more supportive therapies or active agents, to treat one or more complications of an additional diseases or conditions occurring in a subject with SSc (e.g SSc-ILD), myelofibrosis, wherein complications include, for example, ineffective hematopoiesis, anemia, inflammation, fibrosis (e.g., bone marrow fibrosis, spleen fibrosis, and/or liver fibrosis), pancytopenia, thrombocytopenia, extramedullary hematopoiesis (e.g., splenic extramedullary hematopoiesis, hepatic extramedullary hematopoiesis, pulmonary extramedullary hematopoiesis, and/or lymphatic extramedullary hematopoiesis), hepatomegaly, splenomegaly, osteosclerosis, osteomyelofibrosis, poikilocytosis, fatigue, weight loss, night sweats, fever, pruritus, bone pain, early satiety, abdominal pain or discomfort, arthralgias, myalgias, parasthesias, cachexia, splenic infarct, and/or bleeding.

As used herein, inhibition of the fibrotic response of a cell, includes, but is not limited to the inhibition of the fibrotic response of one or more cells within the liver (or liver tissue); one or more cells within the kidney (or renal tissue); one or more cells within muscle tissue; one or more cells within the heart (or cardiac tissue); one or more cells within the pancreas; one or more ceils within the skin; one or more ceils within the bone, one or more cells within the vasculature, one or more stem cells, or one or more cells within the eye.

In some embodiments, any of the TbKII antagonists of the disclosure may be used for treating one or more additional diseases or conditions occurring in a subject with SSc (e.g SSc-ILD), comprising chronic obstructive pulmonary disease (CQPD), chronic obstructive airway disorder, idiopathic pulmonary fibrosis and/or asthma. In part, the disclosure also relates to methods of treating pulmonary hypertension (e.g., pulmonary arterial hypertension) comprising administering to a patient in need thereof an effecti ve amount of a TpRII antagonist (e.g. , a antagonist comprising the amino acid sequence of any one of SEQ ID NOs: 13, 50-56, 69, and 77-83). In some embodiments, the disclosure contemplates methods of treating one or more complications of pulmonary hypertension in a subject with SSc (e.g., SSc-ILD) (e.g., smooth muscle and/or endothelial cell proliferation in the pulmonary artery, angiogenesis in the pulmonary artery, dyspnea, chest pain, pulmonary' vascular remodeling, right ventricular hypertrophy, and/or pulmonary' fibrosis) comprising administering to a patient in need thereof an effective amount of a TbKII antagonist. In some embodiments, the disclosure contemplates methods of treating one or more complications of pulmonary hypertension in a subject with SSc (e.g., SSc-ILD) comprising administering to a patient in need thereof an effective amount of a TbKII antagonist. The present invention contemplates the use of TbKII antagonists in combination with one or more other therapeutic modalities in a subject with SSc (e.g SSc-ILD). Thus, in addition to the use of TbKII antagonists, one may also administer to the subject one or more "standard" therapies for treating fibrotic disorders in a subject with SSc (e.g., SSc-ILD). For example, the TbKII antagonists can be administered to a subject with SSc (e.g., SSc-ILD) in combination with (i.e., together with) cytotoxins, immunosuppressive agents, radiotoxic agents, and/or therapeutic antibodies. Particular co-therapeutics contemplated by the present invention include, but are not limited to, steroids (e.g., corticosteroids, such as Prednisone), immune-suppressing and/or anti-inflammatory agents (e.g., gamma-interferon, cyclophosphamide, azathioprine, methotrexate, penicillamine, cyclosporine, colchicine, antithymocyte globulin, mycophenolate mofetil, and/or hydroxychloroquine), cytotoxic drugs, calcium channel blockers (e.g., nifedipine), angiotensin converting enzyme inhibitors (ACE) inhibitors, para-aminobenzoic acid (PABA), dimethyl sulfoxide, transforming growth factor beta (TORb) inhibitors, interleukin-5 (IL-5) inhibitors, and/or pan caspase inhibitors. Additional anti-fibrotic agents that may be used in combination with TbKII antagonists and administered to a subject with SSc (e.g., SSc-ILD) include, but are not limited to, lectins (as described in, for example, U.S. Patent No.: 7,026,283, the entire contents of which is incorporated herein by reference), as well as the anti-fibrotic agents described by Wynn et al (2007, J Clin Invest 117:524-529, the entire contents of which is incorporated herein by reference). For example, additional anti-fibrotic agents and/or therapies include, but are not limited to, various anti- inflammatory/immunosuppressive/cytotoxic drugs (including colchicine, azathioprine, cyclophosphamide, prednisone, thalidomide, pentoxify lline and/or theophylline), TGFfi signaling modifiers (including relaxin, SMAD7, HGF, and/or BMP7, as well as TGFfi!,

TpR!. TpR!l. EGR-I, and/or CTGF inhibitors), cytokine and/or cytokine receptor antagonists (inhibitors of IL-ϊb, 1L-5, 1L-6, IL- 13, IL-21, IL-4R, IL-13Ral, GM-CSF, TNF-ot, oncostatm M, W1SP-I, and/or PDGFs), cytokines and/or chemokines (IFN-g, IFN-a/b, IL-12, IL-1G, HGF, CXCL10, and/or CXCL11), chemokine antagonists (inhibitors of CXCL1, CXCL2, CXCL12, CCL2, CCL3, CCL6, CCL17, and/or CCL18), chemokine receptor antagonists (inhibitors of CCR2, CCR3, CCR5, CCR7, CXCR2, and/or CXCR4), TLR antagonists (inhibitors of TLR3, TLR4, and/or TLR9), angiogenesis antagonists (VEGF-specific antibodies and/or adenosine deaminase replacement therapy), antihypertensive drugs (beta blockers and/or inhibitors of ANG 11, ACE, and/or aldosterone), vasoactive substances (ET- 1 receptor antagonists and/or bosentan), inhibitors of the enzymes that synthesize and/or process collagen (inhibitors of prolyl hydroxylase), B cell antagonists (rituximab), integrin/adhesion molecule antagonists (molecules that block aΐb! and/or anb6 integrins, as w'ell as inhibitors of integrin-linked kinase, and/or antibodies specific for ICAM-I and/or VCAM-I), proapoptotic drugs that target myofibroblasts, MMP inhibitors (inhibitors of MMP2, MMP9, and/or MMP12), and/or T1MP inhibitors (antibodies specific for TIMP-i ).

14. Pharmaceutical Compositions

The therapeutic agents described herein (e.g., THRU fusion antagonists including but not limited to TbBP polypeptides) may be formulated into pharmaceutical compositions. Pharmaceutical compositions for use in accordance with the present disclosure may be formulated in conventional manner using one or more physiologically acceptable carriers or excipients. Such formulations will generally be substantially pyrogen-free, in compliance with most regulatory requirements.

In some embodiments, the therapeutic method of the disclosure includes reconstituting a lyophilized powder of a TiiRH antagonist of the present disclosure. In some embodiments, the lyophilized TbKII antagonist is reconstituted in sterile water. In some embodiments, a single use vial of lyophilized TbKII antagonist comprises 50 mg of the TfVRH antagonist. In some embodiments, the lyophilized TbKII antagonist is reconstituted for administration by subcutaneous injection.

In certain embodiments, the therapeutic method of the disclosure includes administering the composition systemicaily, or locally as an implant or device. When administered, the therapeutic composition for use in this disclosure is in a pyrogen-free, physiologically acceptable form. Therapeutically useful agents other than tire TbKII antagonists which may also optionally be included in the composition as described above, may be administered simultaneously or sequentially with the subject compounds (e.g., TbRIΪ antagonists) in the methods disclosed herein.

Typically, protein therapeutic agents disclosed herein will be administered parentally, and/or particularly intravenously or subcutaneously. In some embodiments, TbKP antagonists of the present disclosure are administered subcutaneously. Pharmaceutical compositions suitable for parenteral administration may comprise one or more TbKII antagonists in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. Examples of suitable aqueous and/or nonaqueous carriers which may be employed in the pharmaceutical compositions of the disclosure include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and tire like), and suitable mixtures thereof, vegetable oils, such as olive oil, and/or injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the req uired particle size m the case of dispersions, and by the use of surfactants.

The compositions and/or formulations may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration.

Further, the composition may be encapsulated or injected in a form for delivery 7 to a target tissue site. In certain embodiments, compositions of the present invention may include a matrix capable of delivering one or more therapeutic compounds (e.g., TiiRI I antagonists) to a target tissue site, providing a structure for the developing tissue and/or optimally capable of being resorbed into the body. For example, the matrix may provide slow release of the TpRff antagonists. Such matrices may be formed of materials presently in use for other implanted medical applications.

The choice of matrix material is based on biocompatibility, biodegradability, mechanical properties, cosmetic appearance and/or interface properties. The particular application of the subject compositions will define the appropriate formulation. Potential matrices for the compositions may be biodegradable and/or chemically defined calcium sulfate, tricalcium phosphate, hydroxyapatite, poiylactic acid and/or polyanhydrides. Other potential materials are biodegradable and/or biologically well defined, such as bone or dermal collagen. Further matrices are comprised of pure proteins or extracellular matrix components. Other potential matrices are non-biodegradable and/or chemically defined, such as sintered hydroxyapatite, bioglass, aluminates, or other ceramics. Matrices may be comprised of combinations of any of the above mentioned types of material, such as poiylactic acid and/or hydroxyapatite or collagen and/or tricalcium phosphate. The bioceramics may be altered in composition, such as in calcium-aluminate-phosphate and processing to alter pore size, particle size, particle shape, and/or biodegradability.

In certain embodiments, methods of the disclosure can be administered for orally, e.g., in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and/or acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and/or glycerin, or sucrose and/or acacia) and/or as mouth washes and the like, each containing a predetermined amount of an agent as an active ingredient. An agent may also be administered as a bolus, electuary or paste.

In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules, and the like), one or more therapeutic compounds of the present disclosure may be mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or/or any of tire following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or/or silicic acid: (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose, and/or/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar- agar, calcium carbonate, potato or tapioca starch, algmic acid, certain silicates, and/or sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and/or glycerol monostearate; (8) absorbents, such as kaolin and/or bentonite clay; (9) lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and/or mixtures thereof; and/or (10) coloring agents. In the case of capsules, tablets and/or pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and/or hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and/or emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor, and/or sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and/or fatty acid esters of sorbitan, and/or mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and/or suspending agents, sweetening, flavoring, coloring, perfuming, and/or preservative agents. Suspensions, in addition to the active compounds, may contain suspending agents such as ethoxylated isostearyl alcohols, polyoxyethylene sorbitol, and/or sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and/or tragacanth, and/or mixtures thereof.

The compositions of the invention may also contain adjuvants, such as preserv atives, wetting agents, emulsifying agents and/or dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and/or antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and/or gelatin. ft is understood that the dosage regimen will be determined by the attending physician considering various factors which modify the action of the subject compounds of the disclosure (e.g., TfJRll antagonists). The various factors include, but are not limited to, the patient's age, sex, and/or diet, the severity disease, time of administration, and/or other clinical factors. Optionally, the dosage may vary with the type of matrix used in the reconstitution and/or the types of compounds in the composition. The addition of other known growth factors to the final composition, may also affect the dosage. Progress can be monitored by periodic assessment of bone growth and/or repair, for example, X-rays (including DEXA), histomorphometric determinations, and/or tetracycline labeling.

In some embodiments, one or more TbKII antagonists of the present disclosure are administered in one or more doses. In some embodiments, a dose of one or more THRU antagonists comprises 0.05 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 0.10 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 0.15 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 0.20 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKIί antagonists comprises 0.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbRP antagonists comprises 0.50 mg/kg of the antagonists. In some embodiments, a dose of one or more

T|)RH antagonists comprises 0.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 1.00 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 1.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbR.P antagonists comprises 1.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRII antagonists comprises 1.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbϊIII antagonists comprises 2.00 mg/kg of the antagonists. In some embodiments, a dose of one or more TpR.ff antagonists comprises 2.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 2.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbR.II antagonists comprises 2.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 3.00 mg/kg of the antagonists. In some embodiments, a dose of one or more TbMII antagonists comprises 3.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbR.P antagonists comprises 3.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 3.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 4.00 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKίί antagonists comprises 4.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRII antagonists comprises 4.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises 4.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 5.00 mg/kg of the antagonists. In some embodiments, a dose of one or more TbEII antagonists comprises 5.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 5.50 mg/kg of the antagonists. In some embodiments, a dose of one or more

TpRff antagonists comprises 5.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRIi antagonists comprises 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises 6.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRIi antagonists comprises 6.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKίί antagonists comprises 6.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises 7.0 mg/kg of the antagonists.

In some embodiments, a dose of one or more TbKIί antagonists comprises no more than 0.05 mg/kg of the antagonists. In some embodiments, a dose of one or more ' TbKII antagonists comprises no more than 0.10 mg/kg of the antagonists. In some embodiments, a dose of one or more TpR [ f antagonists comprises no more than 0.15 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRIi antagonists comprises no more than 0.20 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises no more than 0.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises no more than 0.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises no more than 0.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises no more than 1.00 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises no more than 1.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TpR II antagonists comprises no more than 1.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises no more than 1.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRIi antagonists comprises no more than 2.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRff antagonists comprises no more than 2.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TpR f f antagonists comprises no more than 2.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRIi antagonists comprises no more than 2.75 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises no more than 3.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRIi antagonists comprises no more than 3.5 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises no more than 4.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKίί antagonists comprises no more than 4.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbR.II antagonists comprises no more than 5.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises no more than 5.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbEIΪ antagonists comprises no more than 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises no more than 6.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TfJRII antagonists comprises no more than 7.0 nig/kg of the antagonists.

In some embodiments, a dose of one or more TbKII antagonists comprises at least 0.05 mg/kg of the antagonists. In some embodiments, a dose of one or more TpRII antagonists comprises at least 0.10 mg/kg of the antagonists. In some embodiments, a dose of one or more THRU antagonists comprises at least 0.15 nig/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises at least 0.20 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII antagonists comprises at least 0.25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKIΊ fusion antagonists comprises between about 0.25 mg/to about 4.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII fusion antagonists comprises between about 0.05 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII fusion antagonists comprises between about 0.25 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII fusion antagonists comprises between about 0.75 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbίIII fusion antagonists comprises between about 0.75 mg/to about 2,25 mg/kg of the antagonists. In some embodiments, a dose of one or more TbίIII fusion antagonists comprises between about 1.25 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKIί fusion antagonists comprises between about 1.25 mg/to about 2.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbKII fusion antagonists comprises between about 1.50 mg/to about 4.50 mg/kg of the antagonists. In some embodiments, a dose of one or more TbίIII fusion antagonists comprises between about 2.5 mg/to about 4.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TbEII fusion antagonists comprises between about 3.0 mg/to about 6.0 mg/kg of the antagonists. In some embodiments, a dose of one or more TTiR!f fusion antagonists comprises between about 4.0 mg/to about 6.0 mg/kg of the antagonists.

In some embodiments, a dose of one or more TpRIl antagonists of the present disclosure is administered once every day. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every' two days. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every ' three days. In some embodiments, a dose of one or more TbKIΪ antagonists of the present disclosure is administered once every' four days. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every five days. In some embodiments, a dose of one or more TbKP antagonists of the present disclosure is administered once every ' six days. In some embodiments, a dose of one or more TbKII an tagonists of the present disclosure is administered once every' week. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every two weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every three weeks. In some embodiments, a dose of one or more TbMI antagonists of the present disclosure is administered once every ' four weeks. In some embodiments, a dose of one or more TbKIΐ antagonists of the present disclosure is administered once every' five weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every six weeks. In some embodiments, a dose of one or more TbKIί antagonists of the present disclosure is administered once every' seven weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every' eight weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every' other week. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every' month. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every ' two months. In some embodiments, a dose of one or more TbKIΊ antagonists of the present disclosure is administered once every' three months. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered once every four months. In some embodiments, a dose of one or more TER! I antagonists of the present disclosure is administered once every ' five months. In some embodiments, a dose of one or more TbKIΐ antagonists of the present disclosure is administered once eveiy six months. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered once even 7 year.

In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every' day. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered twice ever}' two days. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every three days. In some embodiments, a dose of one or snore TbKII antagonists of the present disclosure is administered twice ever}' four days. In some embodiments, a dose of one or more TbίIP antagonists of tire present disclosure is administered twice every ' five days. In some embodiments, a dose of one or snore TbKII antagonists of the present disclosure is administered twice every six days. In some embodiments, a dose of one or more TfiRII an tagonists of the present disclosure is administered twice every week. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every ' two weeks. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every three weeks. In some embodiments, a dose of one or more TpRII antagonists of the present disclosure is administered twice eveiy' four weeks. In some embodiments, a dose of one or more THRU antagonists of the present disclosure is administered twice every' five weeks. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every six weeks. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every seven weeks. In some embodiments, a dose of one or more THR U antagonists of the present disclosure is administered twice every' eight weeks. In some embodiments, a dose of one or more TfiRII antagonists of tire present disclosure is administered twice every' other week. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every month. In some embodiments, a dose of one or more TbKP antagonists of the present disclosure is administered twice eveiy' two months. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every' three months. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice every four months. In some embodiments, a dose of one or more TfiRII antagonists of the present disclosure is administered twice eveiy' five months. In some embodiments, a dose of one or more TfiR II antagonists of the present disclosure is administered twice every six months. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered twice every year.

In some embodiments, a dose of one or more TpRIl antagonists of the present disclosure is administered three times every day. In some embodiments, a dose of one or more TbKII antagonists of die present disclosure is administered three times every' two days. In some embodiments, a dose of one or more TbIIII antagonists of the present disclosure is administered three times every three days. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every' four days. In some embodiments, a dose of one or more TbKP antagonists of the present disclosure is administered three times every' five days. In some embodiments, a dose of one or more TbKIΐ antagonists of the present disclosure is administered three times every ' six days. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every week. In some embodiments, a dose of one or more TbKIί antagonists of the present disclosure is administered three times every ' two weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every three weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every ' four weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every five weeks. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every six weeks. In som e embodiments, a dose of one or m ore TbKII antagonists of the present disclosure is administered three times every' seven weeks. In some embodiments, a dose of one or more TjlR.H antagonists of the present disclosure is administered three times every' eight weeks. In some embodiments, a dose of one or more TbKP antagonists of the present disclosure is administered three times every ' other week. In some embodiments, a dose of one or more

TbKII antagonists of the present disclosure is administered three times every month. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every two months. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every' three months. In some embodiments, a dose of one or more TbίIII antagonists of the present disclosure is administered three times every' four months. In some embodiments, a dose of one or more TbKIΪ antagonists of the present disclosure is administered three times every ' five months. In some embodiments, a dose of one or more TbKII antagonists of the present disclosure is administered three times every' six months. In some embodiments, a dose of one or more TjlRII antagonists of the present disclosure is administered three times every' year.

In some embodiments, a dose of 0.05 mg/kg of one or more ' TbKII antagonists is administered once every' day. In some embodiments, a dose of 0.05 mg/kg of one or more TbKII antagonists is administered once every week. In some embodiments, a dose of 0.05 mg/kg of one or more TbίIP antagonists is administered once every two weeks. In some embodiments, a dose of 0.05 mg/kg of one or more TbKίϊ antagonists is administered once every' three weeks. In some embodiments, a dose of 0.05 mg/kg of one or more TbKII antagonists is administered once every four weeks. In some embodiments, a dose of 0.05 mg/kg of one or more TbKII antagonists is administered once every' five weeks. In some embodiments, a dose of 0.05 mg/kg of one or more TjlRII antagonists is administered once every six weeks.

In some embodiments, a dose of 0.10 mg/kg of one or more TbίIII antagonists is administered once every ' day. In some embodiments, a dose of 0.10 mg/kg of one or more TbKII antagonists is administered once every' week. In some embodiments, a dose of 0.10 mg/kg of one or more TbKII antagonists is administered once every' two weeks. In some embodiments, a dose of 0.10 mg/kg of one or more TbKIΊ antagonists is administered once every' three weeks. In some embodiments, a dose of 0.10 mg/kg of one or more TbKII antagonists is administered once every four weeks. In some embodiments, a dose of 0.10 mg/kg of one or more TbίIP antagonists is administered once every five weeks. In some embodiments, a dose of 0.10 mg/kg of one or more TbKII antagonists is administered once every' six weeks.

In some embodiments, a dose of 0.15 mg/kg of one or more TbKII antagonists is administered once every day. In some embodiments, a dose of 0.15 mg/kg of one or more TbKII antagonists is administered once every' week. In some embodiments, a dose of 0.15 mg/kg of one or more TbKII antagonists is administered once every two w'eeks. In some embodiments, a dose of 0.15 mg/kg of one or more TbIIII antagonists is administered once every' three weeks. In some embodiments, a dose of 0.15 mg/kg of one or more TbKIί antagonists is administered once every' four weeks. In some embodiments, a dose of 0.15 mg/kg of one or more TbKII antagonists is administered once every' five weeks. In some embodiments, a dose of 0.15 mg/kg of one or more TbI¾I1 antagonists is administered once every' six weeks.

In some embodiments, a dose of 0.20 mg/kg of one or more TbKII antagonists is administered once every' day. In some embodiments, a dose of 0.20 mg/kg of one or more TfiRII antagonists is administered once every week. In some embodiments, a dose of 0.20 mg/kg of one or more TbKP antagonists is administered once every two weeks. In some embodiments, a dose of 0.20 mg/kg of one or more TfiRII antagonists is administered once every' three weeks. In some embodiments, a dose of 0.20 mg/kg of one or more TfiRII antagonists is administered once every four weeks. In some embodiments, a dose of 0.20 mg/kg of one or more TfiRII antagonists is administered once every five weeks. In some embodiments, a dose of 0.20 mg/kg of one or more TfiRII antagonists is administered once every/ six weeks.

In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every day. In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every' week. In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every two weeks. In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every 7 three weeks. In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every' four weeks. In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every' five weeks. In some embodiments, a dose of 0.25 mg/kg of one or more TfiRII antagonists is administered once every' six weeks.

In some embodiments, a dose of 0.50 mg/kg of one or more TBRJI antagonists is administered once every' day. In some embodiments, a dose of 0.50 mg/kg of one or more TfiRII antagonists is administered once every' week. In some embodiments, a dose of 0.50 mg/kg of one or more TfiRII antagonists is administered once every' two weeks. In some embodiments, a dose of 0.50 mg/kg of one or more TfiRII antagonists is administered once every three weeks. In some embodiments, a dose of 0.50 mg/kg of one or more TfiRII antagonists is administered once every' four weeks. In some embodiments, a dose of 0.50 mg/kg of one or more TBRJI antagonists is administered once every five weeks. In some embodiments, a dose of 0.50 mg/kg of one or more TfiRII antagonists is administered once every' six weeks.

In some embodiments, a dose of 0.75 mg/kg of one or more TfiRII antagonists is administered once every' day. In some embodiments, a dose of 0.75 mg/kg of one or more TfiRII antagonists is administered once every week. In some embodiments, a dose of 0.75 mg/kg of one or more TfiRII antagonists is administered once every two weeks. In some embodiments, a dose of 0.75 mg/kg of one or more TfiRII antagonists is administered once every three weeks. In some embodiments, a dose of 0.75 mg/kg of one or more TfiRII antagonists is administered once every four weeks. In some embodiments, a dose of 0.75 mg/kg of one or more TbKP antagonists is administered once every five weeks. In some embodiments, a dose of 0.75 mg/kg of one or more TpRII antagonists is administered once every' six weeks. In some embodiments, a dose of 1.00 mg/kg of one or more TpRII antagonists is administered once even day. In some embodiments, a dose of 1.00 mg/kg of one or more TbMI antagonists is administered once ever} 7 week. In some embodiments, a dose of 1.00 mg/kg of one or more TbKP antagonists is administered once even, 7 two weeks. In some embodiments, a dose of 1.00 mg/kg of one or more TpRII antagonists is administered once every' three weeks. In some embodiments, a dose of 1.00 mg/kg of one or more TbKIΪ antagonists is administered once every 7 four weeks. In some embodiments, a dose of 1.00 mg/kg of one or more TpRII antagonists is administered once every five weeks. In some embodiments, a dose of 1.00 mg/kg of one or more TpRff antagonists is administered once every 7 six weeks. In some embodiments, a dose of 1.25 mg/kg of one or more THRU antagonists is administered once every day. In some embodiments, a dose of 1.25 mg/kg of one or more TbMI antagonists is administered once every 7 week. In some embodiments, a dose of 1.25 mg/kg of one or more TpRII antagonists is administered once every two weeks. In some embodiments, a dose of 1.25 mg/kg of one or more TpRII antagonists is administered once every three weeks. In some embodiments, a dose of 1.25 mg/kg of one or more TbKII antagonists is administered once every 7 four weeks. In some embodiments, a dose of 1.25 mg/kg of one or more TpRII antagonists is administered once every 7 five w 7 eeks. In some embodiments, a dose of 1.25 mg/kg of one or more TpRII antagonists is administered once every six weeks. In some embodiments, a dose of 1.50 mg/kg of one or more TpRII antagonists is administered once every 7 day. In some embodiments, a dose of 1.50 mg/kg of one or more TpRII antagonists is administered once every week. In some embodiments, a dose of 1.50 mg/kg of one or more TbKP antagonists is administered once every 7 two weeks. In some embodiments, a dose of 1.50 mg/kg of one or more TpRII antagonists is administered once every three weeks. In some embodiments, a dose of 1.50 mg/kg of one or more TpRII antagonists is administered once every four weeks. In some embodiments, a dose of 1.50 mg/kg of one or more TpRII antagonists is admini stered once every five weeks. In some embodiments, a dose of 1.50 mg/kg of one or more TpRII antagonists is administered once every six weeks. In some embodiments, a dose of 1.75 mg/kg of one or more TbKII antagonists is administered once every day. In some embodiments, a dose of 1.75 mg/kg of one or more TbKP antagonists is administered once every' week. In some embodiments, a dose of 1.75 mg/kg of one or more TbKII antagonists is administered once every two weeks. In some embodiments, a dose of 1.75 mg/kg of one or more Tf!RII antagonists is administered once every three weeks. In some embodiments, a dose of 1.75 mg/kg of one or more TbKII antagonists is administered once every four ’weeks. In some embodiments, a dose of 1.75 mg/kg of one or more TbMΪ antagonists is administered once even,' five weeks. In some embodiments, a dose of 1.75 mg/kg of one or more TjlRll antagonists is administered once every' six weeks.

In some embodiments, a dose of 2.00 mg/kg of one or more TbKII antagonists is administered once every' day. In some embodiments, a dose of 2.00 mg/kg of one or more TbKII antagonists is administered once every week. In some embodiments, a dose of 2.00 mg/kg of one or more TbKIί antagonists is administered once every ' two weeks. In some embodiments, a dose of 2.00 mg/kg of one or more TbKII antagonists is administered once every three w'eeks. In some embodiments, a dose of 2.00 mg/kg of one or more TbKII antagonists is administered once every' four weeks. In some embodiments, a dose of 2.00 mg/kg of one or more TbKII antagonists is administered once every five weeks. In some embodiments, a dose of 2.00 mg/kg of one or more TpRil antagonists is administered once every six weeks.

In some embodiments, a dose of 2.25 mg/kg of one or more TbKP antagonists is administered once every ' day. In some embodiments, a dose of 2.25 mg/kg of one or more T|)RH antagonists is administered once every week. In some embodiments, a dose of 2.25 mg/kg of one or more TbKP antagonists is administered once every two weeks. In some embodiments, a dose of 2.25 mg/kg of one or more TjlRII antagonists is administered once every three weeks. In some embodiments, a dose of 2.25 mg/kg of one or more TbKII antagonists is administered once every four weeks. In some embodiments, a dose of 2.25 mg/kg of one or more TbKIΪ antagonists is administered once every ' five weeks. In some embodiments, a dose of 2.25 mg/kg of one or more TbKII antagonists is administered once every six weeks.

In some embodiments, a dose of 2.50 mg/kg of one or more TbKII antagonists is administered once every day. In some embodiments, a dose of 2.50 mg/kg of one or more TbKIΐ antagonists is administered once every' week. In some embodiments, a dose of 2.50 mg/kg of one or more TbίIP antagonists is administered once every two w'eeks. In some embodiments, a dose of 2.50 mg/kg of one or more TfiRH antagonists is administered once every three weeks. In some embodiments, a dose of 2.50 mg/kg of one or more TfIRII antagonists is administered once every 7 four weeks. In some embodiments, a dose of 2.50 mg/kg of one or more TbKII antagonists is administered once every five weeks. In some embodiments, a dose of 2.50 mg/kg of one or more TjiR!I antagonists is administered once every six weeks.

In some embodiments, a dose of 2.75 mg/kg of one or more TbKP antagonists is administered once every 7 day. In some embodiments, a dose of 2.75 mg/kg of one or more TbKII antagonists is administered once every week. In some embodiments, a dose of 2.75 mg/kg of one or more TbKII antagonists is administered once every two weeks. In some embodiments, a dose of 2.75 mg/kg of one or more TbR2I antagonists is administered once every three weeks. In some embodiments, a dose of 2.75 mg/kg of one or more TbKII antagonists is administered once every four weeks. In some embodiments, a dose of 2.75 mg/kg of one or more TbMΪ antagonists is administered once every 7 five weeks. In some embodiments, a dose of 2.75 mg/kg of one or more TbKII antagonists is administered once every six weeks.

In some embodiments, a dose of 3.00 mg/kg of one or more ' TbKII antagonists is administered once every 7 day. In some embodiments, a dose of 3.00 mg/kg of one or more TbKII antagonists is administered once every week. In some embodiments, a dose of 3.00 mg/kg of one or more TbίIII antagonists is administered once every two weeks. In some embodiments, a dose of 3.00 mg/kg of one or more TbKIΪ antagonists is administered once every 7 three weeks. In some embodiments, a dose of 3.00 mg/kg of one or more TbKII antagonists is administered once every four weeks. In some embodiments, a dose of 3.00 mg/kg of one or more TbKP antagonists is administered once every five weeks. In some embodiments, a dose of 3.00 mg/kg of one or more TjrRII antagonists is administered once every six weeks.

In some embodiments, a dose of 3.50 mg/kg of one or more TbίIII antagonists is administered once every 7 day. In some embodiments, a dose of 3.50 mg/kg of one or more TbKII antagonists is administered once every 7 week. In some embodiments, a dose of 3.50 mg/kg of one or more TbKII antagonists is administered once every 7 two weeks. In some embodiments, a dose of 3.50 mg/kg of one or more TpRII antagonists is administered once every 7 three weeks. In some embodiments, a dose of 3.50 mg/kg of one or more TbKII antagonists is administered once every 7 four weeks. In some embodiments, a dose of 3.50 mg/kg of one or more TbίIP antagonists is administered once every five weeks. In some embodiments, a dose of 3.50 mg/kg of one or more TfiRH antagonists is administered once every six weeks.

In some embodiments, a dose of 4.00 mg/kg of one or more ' TbKII antagonists is administered once every' day. In some embodiments, a dose of 4.00 mg/kg of one or more TbKII antagonists is administered once every week. In some embodiments, a dose of 4.00 mg/kg of one or more TbίIP antagonists is administered once every two weeks. In some embodiments, a dose of 4.00 mg/kg of one or more TbKίϊ antagonists is administered once every' three weeks. In some embodiments, a dose of 4.00 mg/kg of one or more TbKII antagonists is administered once every four weeks. In some embodiments, a dose of 4.00 mg/kg of one or more TbKII antagonists is administered once eveiy five weeks. In some embodiments, a dose of 4.00 mg/kg of one or more TjlRII antagonists is administered once every six weeks.

In some embodiments, a dose of no more than 4.50 mg/kg of one or more TbKII antagonists is administered once every' day. In some embodiments, a dose of no more than 4.50 mg/kg of one or more TbKII antagonists is administered once every week. In some embodiments, a dose of no more than 4.50 mg/kg of one or more TpRII antagonists is administered once every two weeks. In some embodiments, a dose of no more than 4.50 mg/kg of one or more TbKII antagonists is administered once every three weeks. In some embodiments, a dose of 4.50 mg/kg of one or more TpRII antagonists is administered once every four weeks. In some embodiments, a dose of 4.50 mg/kg of one or more TbKP antagonists is administered once every five weeks. In some embodiments, a dose of 4.50 mg/kg of one or more TbKP antagonists is administered once every' six weeks.

In some embodiments, a dose of 5.00 mg/kg of one or more TbKII antagonists is administered once eveiy day. In some embodiments, a dose of 5.00 mg/kg of one or more TbKII antagonists is administered once every' week. In some embodiments, a dose of 5.00 mg/kg of one or more TbKII antagonists is administered once every two weeks. In some embodiments, a dose of 5.00 mg/kg of one or more TbIIII antagonists is administered once every' three weeks. In some embodiments, a dose of 5.00 mg/kg of one or more TbKIί antagonists is administered once every' four weeks. In some embodiments, a dose of 5.00 mg/kg of one or more TbKII antagonists is administered once every' five weeks. In some embodiments, a dose of 5.00 mg/kg of one or more TjtR!! antagonists is administered once every' six weeks.

In some embodiments, a dose of no more than 5.50 mg/kg of one or more TOR If antagonists is administered once every day. In some embodiments, a dose of no more than 5.50 mg/kg of one or more TiiRH antagonists is administered once every week. In some embodiments, a dose of no more than 5.50 mg/kg of one or more TbIIII antagonists is administered once every two weeks. In some embodiments, a dose of no more than 5.50 mg/kg of one or more TiiRH antagonists is administered once every three weeks. In some embodiments, a dose of 5.50 mg/kg of one or more Tf!RII antagonists is administered once every four weeks. In some embodiments, a dose of 5.50 mg/kg of one or more TbKP antagonists is administered once every five weeks. In some embodiments, a dose of 5.50 mg/kg of one or more THRU antagonists is administered once every' six weeks.

In some embodiments, a dose of 6.00 mg/kg of one or more TbKII antagonists is administered once every day. In some embodiments, a dose of 6.00 mg/kg of one or more TbKIΐ antagonists is administered once every' week. In some embodiments, a dose of 6,00 mg/kg of one or more TbKII antagonists is administered once every two weeks. In some embodiments, a dose of 6.00 mg/kg of one or more TbίIII antagonists is administered once every 7 three weeks. In some embodiments, a dose of 6.00 mg/kg of one or more TbKIί antagonists is administered once every' four weeks. In some embodiments, a dose of 6.00 mg/kg of one or more TbKII antagonists is administered once every' five weeks. In some embodiments, a dose of 6.00 mg/kg of one or more TbKIΊ antagonists is administered once every' six weeks.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKP) antagonist to a subject in need thereof, wherein the TbKIΊ antagonist is admini stered in a dose of between about 0.05 mg/kg and about 6,00 mg/kg to a subject in need thereof. In some embodiments, the TpRII antagonist is administered at least once every' week. In some embodiments, the Tj!RII antagonist is administered at least once every three weeks. In some embodiments, the TpRII antagonist is administered at least once every four weeks. In some embodiments, the TbίIII antagonist is administered at least once every five weeks. In some embodiments, the TbKH antagonist is administered at least once every six w'eeks. In some embodiments, the TfiR!l antagonist is administered subcutaneously.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II fl ' BRH) antagonist to a subject in need thereof, wherein the TbIIII antagonist is administered in a dose of between about 0.25 mg/kg and about 4.50 mg/kg to a subject in need thereof In some embodiments, the TiiRH antagonist is administered at least once every ' week. In some embodiments, the TpRII antagonist is administered at least once every three weeks. In some embodiments, the T|)RH antagonist is administered at least once ever}' four weeks. In some embodiments, the

TbIIII antagonist is administered at least once every five weeks. In some embodiments, the

TbKII antagonist is administered at least once every six weeks. In some embodiments, the TpRII antagonist is administered subcutaneously. In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKP) antagonist to a subject in need thereof, wherein the TbMI antagonist is admini stered in a dose of between about 0.75 mg/kg and about 6,00 mg/kg to a subject in need thereof. In some embodiments, the TpRH antagonist is administered at least once every week. In some embodiments, the TbKII antagonist is administered at least once every three weeks. In some embodiments, the TpR!l antagonist is administered at least once every four weeks. In some embodiments, the

TpRH antagonist is administered at least once every five weeks. In some embodiments, the

' TbKH antagonist is administered at least once every six weeks. In some embodiments, the TfiR!l antagonist is administered subcutaneously. In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKP antagonist is administered in a dose of between about 0.75 mg/kg and about 2.25 mg/kg to a subject in need thereof. In some embodiments, the THRU antagonist is administered at least once every week. In some embodiments, the ' TbKH antagonist is administered at least once every three weeks. In some embodiments, the TfiR!l antagonist is administered at least once every ' four weeks. In some embodiments, the

TbϊIII antagonist is administered at least once every' five weeks. In some embodiments, the

TflRII antagonist is administered at least once every' six weeks. In some embodiments, tire Tj!RII antagonist is administered subcutaneously. In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKH antagonist is administered in a dose of between about 1.25 mg/kg and about 6.00 mg/kg to a subject in need thereof. In some embodiments, the THRU antagonist is administered at least once every' week. In some embodiments, the TflRII antagonist is administered at least once every' three weeks. In some embodiments, the Tj!RII antagonist is administered at least once every four weeks. In some embodiments, the

TbKP antagonist is administered at least once every five weeks. In some embodiments, the

TpRII antagonist is administered at least once every six weeks. In some embodiments, the Tb!¾II antagonist is administered subcutaneously. In some embodiments, die disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbίIP) antagonist to a subject in need thereof, wherein the TbKP antagonist is administered in a dose of between about 1.25 mg/kg and about 2.00 mg/kg to a subject in need thereof. In some embodiments, the TbIPI antagonist is administered at least once every week. In some embodiments, the TjlR!! antagonist is administered at least once every three weeks. In some embodiments, the TjlR!l antagonist is administered at least once every four weeks. In some embodiments, the TbϊIII antagonist is administered at least once every' five weeks. In some embodiments, the TbίIII antagonist is administered at least once every six weeks. In some embodiments, the TbKίϊ antagonist is administered subcutaneously.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered in a dose of between about 1.50 mg/kg and about 4.50 mg/kg to a subject in need thereof. In some embodiments, the THRU antagonist is administered at least once every' week. In some embodiments, the TpRII antagonist is administered at least once every' three weeks. In some embodiments, the TbKίϊ antagonist is administered at least once every four weeks. In some embodiments, the

TpRII antagonist is administered at least once every five weeks. In some embodiments, the

TpRH antagonist is administered at least once every six w'eeks. In some embodiments, the TbKII antagonist is administered subcutaneously.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbMI antagonist is administered in a dose of between about 2.50 mg/kg and about 4.00 mg/kg to a subject in need thereof. In some embodiments, the Tb R 11 antagonist is administered at least once every ' week. In some embodiments, the

TpRH antagonist is administered at least once every three weeks. In some embodiments, the TjlR!! antagonist is administered at least once every four weeks. In some embodiments, the

TbMII antagonist is administered at least once every ' five weeks. In some embodiments, the

TjlRII antagonist is administered at least once every' six weeks. In some embodiments, the TbKII antagonist is administered subcutaneously.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transform ing Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered in a dose of between about 3.00 mg/kg and about 6.00 mg/kg to a subject in need thereof. In some embodiments, the TbίIίI antagonist is administered at least once every' week. In some embodiments, the TbIIII antagonist is administered at least once every three weeks. In some embodiments, the TbKII antagonist is administered at least once every four weeks. In some embodiments, the TpRII antagonist is administered at least once every five weeks. In some embodiments, the TbίIII antagonist is administered at least once every six weeks. In some embodiments, the TpRII antagonist is administered subcutaneously.

In some embodiments, the disclosure provides methods of treating SSe-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the Tj!RII antagonist is administered in a dose of between about 4.00 mg/kg and about 6.0 mg/kg to a subject in need thereof. In some embodiments, the TbKP antagonist is administered at least once every ' week. In some embodiments, the TpRH antagonist is administered at least once every three weeks. In some embodiments, the TpRII antagonist is administered at least once every four weeks. In some embodiments, the TbMII antagonist is administered at least once every ' five weeks. In some embodiments, the TjlRII antagonist is administered at least once every' six weeks. In some embodiments, the TjsRfl antagonist is administered subcutaneously .

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered in a dose of between about 0.75 mg/kg once every two weeks to a subject in need thereof.

In some embodiments, the disclosure provides methods of treating SSe-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbMI antagonist is administered in a dose of between about 2.25 mg/kg once every four weeks to a subject in need thereof. In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered in a dose of between about 1.50 mg/kg once every two weeks to a subject in need thereof.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, w'herein the TbίIII antagonist is administered in a dose of between about 4.50 mg/kg once every four weeks to a subject in need thereof.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKP) antagonist to a subject in need thereof, wherein the TpRII antagonist is administered in a dose of between about 3.00 mg/kg once every two w'eeks to a subject in need thereof.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TpRII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered in a dose of between about 6.00 mg/kg once every four weeks to a subject in need thereof.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor IT (TpRII) antagonist to a subject in need thereof, wtierein the TpRII antagonist is administered in a dose of between about 1.25 mg/kg once every two weeks to a subject in need thereof

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TpRII) antagonist to a subject in need thereof, wherein the TpRII antagonist is administered in a dose of between about 2.00 mg/kg once every four weeks to a subject in need thereof. In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TpRII) antagonist to a subject in need thereof, wherein the TpRII antagonist is administered in a dose of between about 2.50 mg/kg once every' two weeks to a subject in need thereof.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKP) antagonist to a subject in need thereof, wherein the TpRII antagonist is administered in a dose of between about 4.00 mg/kg once every-· four weeks to a subject in need thereof.

In some embodiments, die disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TpRII) antagonist to a subject in need thereof, wherein the TpRII antagonist is administered in a dose of between about 4.00 mg/kg once every' two weeks to a subject in need thereof.

In some embodiments, the TpRII antagonist is administered by a parenteral route of administration. In some embodiments, a parenteral route of administration is selected from the group consisting of intramuscular, intraperitoneal, intradermal, intravitreal, epidural, intracerebral, intra-arterial, intraarticular, intra-cavemous, intra-lesional, intraosseous, intraocular, intrathecal, intravenous, transdermal, trans-mucosal, extra-amniotic administration, subcutaneous, and combinations thereof. In some embodiments, a parenteral route of administration is subcutaneous. In some embodiments, a parenteral route of administration is a subcutaneous injection. In some embodiments, compositions of the present disclosure are administered by subcutaneous injection.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKΪΪ) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a serum concentration of antagonist of at least between about 10 and about 20 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbίIP) antagonist to a subject in need thereof, wherein the Tj!Rfl antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 10 and about 20 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor 11 (TbKP) antagonist to a subject in need thereof, wherein the TjlR!l antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 20 and about 30 ug/mL m the subject.

In some embodiments, die disclosure provides methods of treating SSc-ILD, comprising administering a Transform ing Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbRII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 30 and about 40 ug/mL in the subject. In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKP) antagonist to a subject m need thereof, wtierein the TbKII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 40 and about 50 ug/mL in the subject.

In some embodiments, die disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TpRH antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 10 and about 50 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TpRII) antagonist to a subject in need thereof, wtierein the TfyRII antagonist is administered at a dose that achieves a seram concentration of antagonist of between about 30 and about 50 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TfiRII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 50 and about 60 ug/mL in the subject.

In some embodiments, the disclosure provides m ethods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 60 and about 70 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor IT (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 70 and about 80 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a serum concentration of antagonist of between about 10 and about 80 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transform ing Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbRII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of at least between about 30 and about 50 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor IT (TpRII) antagonist to a subject in need thereof, wherein the TfiRII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 30 and about 50 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKP) antagonist to a subject in need thereof, wherein the Tj!R!l antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 10 and about 20 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transform ing Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbRII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 20 and about 30 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-il.l ) . comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 30 and about 40 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbίIP) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 40 and about 50 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II ( ' TbKP) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 50 and about 60 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TbKII antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 60 and about 70 ug/mL in the subject.

In some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbίIP) antagonist to a subject in need thereof, wherein the TbH,P antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 70 and about 80 ug/mL in the subject. in some embodiments, the disclosure provides methods of treating SSc-ILD, comprising administering a Transforming Growth Factor-b Receptor II (TbKII) antagonist to a subject in need thereof, wherein the TjtRfl antagonist is administered at a dose that achieves a concentration of antagonist in the lung of between about 10 and about 80 ug/mL in the subject.

In certain embodiments, the present invention also provides gene therapy for the in vivo production of TbKII antagonists. Such therapy would achieve its therapeutic effect by introduction of the TpRJI antagonist polynucleotide sequences into cells or tissues having the disorders as listed above. Delivery of TbIIII antagonist polynucleotide sequences can be achieved using a recombinant expression vector such as a chimeric virus or a colloidal dispersion system. Preferred for therapeutic delivery of TbKII antagonist polynucleotide sequences is the use of targeted liposomes.

Various viral vectors which can be utilized for gene therapy as taught herein include adenovirus, herpes virus, vaccinia, or, preferably, an RNA virus such as a retrovirus. Preferably, the retroviral vector is a derivative of a murine or avian retrovirus. Examples of retroviral vectors in which a single foreign gene can be inserted include but are not limited to: Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary' tumor virus (MuMTV), and/or Rous Sarcoma Virus (RSV). A number of additional retroviral vectors can incorporate multiple genes. All of these vectors can transfer or incorporate a gene for a selectable marker so that transduced cells can be identified and/or generated. Retroviral vectors can be made target-specific by attaching, for example, a sugar, a glyco!ipid, or a protein . Preferred targeting is accomplished by using an antibody. Those of skill in the art will recognize that specific polynucleotide sequences can be inserted into the retroviral genome or attached to a viral envelope to allow? target specific delivery ' of the retroviral vector containing the Tb R 11 antagonist polynucleotide. In a preferred embodiment, the vector is targeted to bone or cartilage. Alternatively, tissue culture cells can be directly transfected with plasmids encoding the retroviral structural genes gag, pol and/or env, by conventional calcium phosphate transfection. These cells are then transfected with the vector plasmid containing the genes of interest. The resulting cells release the retroviral vector into the culture medium.

Another targeted delivery system for TbKII antagonist polynucleotides is a colloidal dispersion system. Colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and/or lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and/or liposomes. The preferred colloidal system of this invention is a liposome. Liposomes are artificial membrane vesicles which are useful as delivery ? vehicles in vitro and/or in vivo. RNA, DNA and/or intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (see e.g., Fraley, et al., Trends Biochem. Sen, 6:77, 1981). Methods for efficient gene transfer using a liposome vehicle, are known in the art, see e.g., Mannino, et al., Biotechniques, 6:682, 1988. The composition of the liposome is usually a combination of phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used. The physical characteristics of liposomes depend on pH, ionic strength, and/or the presence of divalent cations.

Examples of lipids useful in liposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatdylserine, phosphatidylethanolamine, sphingolipids, cerebrosides, and/or gangliosides. Illustrative phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine, and/or distearoylphosphatidylcholine. The targeting of liposomes is also possible based on, for example, organ-specificity, cell-specificity, and/or organelle-specificity and is known in the art. The disclosure provides formulations that may be varied to include acids and/or bases to adjust the pH; and/or buffering agents to keep the pH within a narrow 7 range.

EXEMPLIFICATION

The invention now being generally described, it will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain embodiments of the present invention, and are not intended to limit the invention.

Example 1. Generation of receptor fusion protein variants TbMI ECD variants TpRII fusion proteins comprising a soluble extracellular portion of human TbίIII and a human Fc portion were generated. For each fusion protein, a TbEP amino acid sequence having the amino acid sequence of SEQ ID NO: 18 w 7 as fused to an IgG Fc portion having the amino acid sequence of S EQ ID NO: 20 by means of one of several different linkers. Each of the fusion proteins also included a TPA leader sequence having tire ammo acid sequence of SEQ ID NO : 23 (bel ow) ,

Tissue plasminogen activator (TPA): MDAMKRGLCCVLLLCGAVFVSP ( SEQ ID NO : 23 )

An illustration summary of several of the constructs designed is provided as Figure 3. A table detailing the sequences for the different constructs tested in the Exemplification section is provided below':

The amino acid sequences for the construct components and each of the constructs, along with the nucleic acid sequence used to express these constructs, are provided below.

TbKII Portion: Amino Acid Sequence

1 TI PPHVQKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF

51 PQLCKFCDVR FSTCDNQKSC MSNCSITSIC EKPQEVCVAV WRKNDENITL

101 ETVCHDPKLP YHDFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI 151 I FSEEYNTSN PD ( SEQ I D NO : 18 )

Fc Portion: Amino Acid Sequence

1 THTCPPCPAP ELLGGPSVFL FPPKPKDTLM I SRTPEVTCV WDVSHEDPE

51 VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK

101 VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSREEMTKNQ VSLTCLVKGF 151 YP S D I AVEWE SNGQPENNYK TTPPVLDSDG S FFLYSKLTV DKSRWQQGNV 201 FSCSVMHEAL HNHYTQKSLS LSPGK ( SEQ ID NO : 20 )

The C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 20 may optionally be provided with the lysine removed from the C- termmus (SEQ ID NO: 72)

1 THTCPPCPAP ELLGGPSVFL FPPKPKDTLM I SRTPEVTCV WDVSHEDPE

51 VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK

101 VSNKALPAPI EKTI SKAKGQ PREPQVYTLP PSREEMTKNQ VSLTCLVKGF

151 Y P S D I AVEWE SNGQPENNYK TTPPVLDSDG S FFLYSKLTV DKSRWQQGNV

201 FSCSVMHEAL HNHYTQKSLS LSPG ( SEQ I D NO : 72 )

IiΊbRII-IiRo: Nucleic Acid Sequence

1 AT G GAT Ci C AA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC

51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCCAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT 151 AGGACTGCCC AT C CACT GAG ACATATTAAT AACGACATGA TAGTCACTGA

201 CAACAACGGT GCAGT CAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA

251 GATTTTCCAC CTGTGACAAC CAGAAATCCT G C A T GAG C AA CT GCAGCAT C

301 AC C T C CAT C T GTGAGAAGCC ACAGGAAGT C TGTGTGGCTG TAT G GAGAAA 351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC 401 CCTACCATGA CTTTATTCTG GAAGATGCTG CTTCTCCAAA GTGCATTATG 451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC 501 TGATGAGTGC AATGACAACA TCA!CTTCTC AGAAGAATAT AACACCAGCA 551 ATCCTGACAC CGGTGGTGGA ACTCACACAT GCCCACCGTG CCCAGCACCT 601 GAACTCCTGG GGGGACCGTC AGTCTTCCTC TTCCCCCCAA AACCCAAGGA 651 CACCCTCATG ATCTCCCGGA CCCCTGAGGT CACATGCGTG GTGGTGGACG 701 TGAGCCACGA AGACCCTGAG GTCAAGTTCA ACTGGTACGT GGACGGCGTG

751 GAGGTGCATA ATGCCAAGAC AAAGCCGCGG GAGGAGCAGT ACAACAGCAC

801 GTACCGTGTG GTCAGCGTCC TCACCGTCCT GCACCAGGAC TGGCTGAATG

851 GCAAGGAGTA CAAGTGCAAG GTCTCCAACA AAGCCCTCCC AGCCCCCATC

901 GAGAAAACCA TCTCCAAAGC CAAAGGGCAG CCCCGAGAAC CACAGGTGTA

951 CACCCTGCCC CCATCCCGGG AGGAGATGAC CAAGAACCAG GTCAGCCTGA

1001 CCTGCCTGGT CAAAGGCTTC TATCCCAGCG ACATCGCCGT GGAGTGGGAG

1051 AGCAATGGGC AGCCGGAGAA CAACTACAAG ACCACGCCTC CCGTGCTGGA

1101 CTCCGACGGC TCCTTCTTCC TCTATAGCAA GCTCACCGTG GACAAGAGCA

1151 GGTGGCAGCA GGGGAACGTC TTCTCATGCT CCGTGATGCA TGAGGCTCTG

1201 CACAACCACT ACACGCAGAA GAGCCTCTCC CTGTCTCCGG GTA.AAs.TGA (SEQ NO: 8) hT ' pRlI-hFc: Amino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN 51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDYRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAW RKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG THTCPPCPAP 201 ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV W DVSHEDPE VKFNWYVDGV 251 EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI 301 EKTISKAKGQ PREPQVYTLP PSREEMTKNQ VSLTCLVKGF YPSDIAVEWE 351 SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL 401 HNHYTQKSLS LSPGK (SEQ ID NO: 9)

The C -terminal lysine residue of the Fc domain can be deleted, Tire amino acid sequence of SEQ ID NO: 9 may optionally be provided with the lysine removed from the C- temiinus (SEQ ID NO: 88):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN 51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI 101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM 151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG THTCPPCPAP 201 ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV W DVSHEDPE VKFNWYVDGV 251 EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI 301 EKTISKAKGQ PREPQVYTLP PSREEMTKNQ VSLTCLVKGF YPSDIAVEWE

351 SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL

401 HNHYTQKSLS LSPG (SEQ ID NO: 88) hT/jRII (G4S)3-hFc: Nucleic Acid Sequence

1 ATGGATGCAA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC

51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCCAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT

151 AGGACTGCCC ATCCACTGAG ACATATTAAT AACGACATGA TAGTCACTGA 201 CAACAACGGT GCAGTCAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA 251 GATTTTCCAC CTGTGACAAC CAGAAATCCT GCATGAGCAA CTGCAGCATC 301 ACCTCCATCT GTGAGAAGCC ACAGGAAGTC TGTGTGGCTG TATGGAGAAA 351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC 401 CCTACCATGA C!TTATTC!G GAAGATGCTG CTTCTCCAAA GTGCATTATG 451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC 501 TGATGAGTGC AATGACAACA TCATCTTCTC AGAAGAATAT AACACCAGCA 551 ATCCTGACAC CGGTGGTGGA GGAAGTGGTG GAGGTGGTTC TGGAGGTGGT 601 GGAAGTACTC ACACATGCCC ACCGTGCCCA GCACCTGAAC TCCTGGGGGG 651 ACCGTCAGTC TTCCTCTTCC CCCCAAAACC CAAGGACACC CTCATGATCT 701 CCCGGACCCC TGAGGTCACA TGCGTGGTGG TGGACGTGAG CCACGAAGAC 751 CCTGAGGTCA AGTTCAACTG GTACGTGGAC GGCGTGGAGG TGCATAATGC 801 CAAGACAAAG CCGCGGGAGG AGCAGTACAA CAGCACGTAC CGTGTGGTCA 851 GCGTCCTCAC CGTCCTGCAC CAGGACTGGC TGAATGGCAA GGAGTACAAG 901 TGCAAGGTCT CCAACAAAGC CCTCCCAGCC CCCATCGAGA AAACCATCTC 951 CAAAGCCAAA GGGGAGCCCC GAGAACCACA GGTGTACACC CTGCCCCCAT 1001 CCCGGGAGGA GATGACCAAG AACCAGGTCA GCCTGACCTG CCTGGTCAAA 1051 GGCTTCTATC CCAGCGACAT CGCCGTGGAG TGGGAGAGCA ATGGGCAGCC 1101 GGAGAACAAC TACAAGACCA CGCCTCCCGT GCTGGACTCC GACGGCTCCT 1151 TCTTCCTCTA TAGCAAGCTC ACCGTGGACA AGAGCAGGTG GCAGCAGGGG 1201 AACGTCTTCT CATGCTCCGT GATGCATGAG GCTCTGCACA ACCACTACAC 1251 GCAGAAGAGC CTCTCCCTGT CTCCGGGTAA ATGA (SEQ :D NO: 10) hTfiRJJ (G4S)3-hFc: Amino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVW DVSHED

251 PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RW SVLTVLH QDWLNGKEYK

301 CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSREEMTK NQVSLTCLVK

351 GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG

401 NVFSCSVMHE ALHNHYTQKS LSLSPGK (SEQ ID NO: 11)

The C-terminal lysine residue of the Fc domain can be deleted, Tire amino acid sequence of SEQ ID NO: 11 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 68):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVW DVSHED

251 PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RW SVLTVLH QDWLNGKEYK

301 CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSREEMTK NQVSLTCLVK

351 GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG

401 NVFSCSVMHE ALHNHYTQKS LSLSPG (SEQ ID NO: 68) hT/jRII (G4S)4-hFc: Nucleic Acid Sequence

1 ATGGATGCAA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC 51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCCAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT

151 AGGACTGCCC ATCCACTGAG ACATATTAAT AACGACATGA TAGTCACTGA

201 CAACAACGGT GCAGTCAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA 251 GATTTTCCAC CTGTGACAAC CAGAAATCCT GCATGAGCAA CTGCAGCATC

301 ACCTCCATCT GTGAGAAGCC ACAGGAAGTC TGTGTGGCTG TATGGAGAAA

351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC

401 CCTAGCATGA CTTTATTCTG GAAGATGCTG CTTCTCCAAA GTGCATTATG

451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC

501 TGATGAGTGC AATGACAACA TCATCTTCTC AGAAGAATAT AACACCAGCA

551 ATCCTGACAC CGGTGGTGGA GGTTCTGGAG GTGGAGGAAG TGGTGGAGGT

601 GGTTCTGGAG GTGGTGGAAG TACTCACACA TGCCCACCGT GCCCAGCACC

651 TGAACTCCTG GGGGGACCGT CAGTCTTCCT CTTCCCCCCA AAACCCAAGG

701 ACACCCTCAT GATCTCCCGG ACCCCTGAGG TCACATGCGT GGTGGTGGAC

751 GTGAGCCACG AAGACCCTGA GGTCAAGTTC AACTGGTACG TGGACGGCGT

801 GGAGGTGCAT AATGCCAAGA CAAAGCCGCG GGAGGAGCAG TACAACAGCA

851 CGTACCGTGT GGTCAGCGTC CTCACCGTCC TGCACCAGGA CTGGCTGAAT

901 GGCAAGGAGT ACAAGTGCAA GGTC!CCAAC AAAGCCCTCC CAGCCCCCAT

951 CGAGAAAACC ATCTCCAAAG CCAAAGGGCA GCCCCGAGAA CCACAGGTGT

1001 ACACCCTGCC CCCATCCCGG GAGGAGATGA CCAAGAACCA GGTCAGCCTG

1051 ACCTGCCTGG TCAAAGGCTT CTATCCCAGC GACATCGCCG TGGAGTGGGA

1101 GAGCAATGGG CAGCCGGAGA ACAACTACAA GACCACGCCT CCCGTGCTGG

1151 ACTCCGACGG CTCCTTCTTC CTCTATAGCA AGCTCACCGT GGACAAGAGC

1201 AGGTGGCAGC AGGGGAACGT CTTCTCATGC TCCGTGATGC ATGAGGCTCT

1251 GCACAACCAC TACACGCAGA AGAGCCTCTC CCTGTCTCCG GGTAAATGA

(SEQ ID NO: 12) hTjjRJJ (G4S)4-hFc: Amino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDETICPSCN 51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAW RKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSGGGGSTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVW D

251 VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRW SV LTVLHQDWLN

301 GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR EEMTKNQVSL

351 TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS

401 RWOOGNVFSC SVMHEALHNH YTQKSLSLSP GK (SEQ ID NO: 13)

The C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 13 may optionally be provided with the lysine removed from the C- temiinus (SEQ ID NO: 69):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSGGGGSTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVW D

251 VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRW SV LTVLHQDWLN

301 GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR EEMTKNQVSL

351 TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS

401 RWQQGNVFSC SVMHEALHNH YTQKSLSLSP G (SEQ ID NO: 69)

HTbKII ( G4S)4-hFc : Ammo Acid Sequence lacking leader sequence

1 GATIPPHVQK SDVEMEAQKD EIICP3CNRT AHPLRHINND MIVTDNNGAV

51 KFPQLCKFCD VRFSTCDNQK SCMSNCSITS ICEKPQEVCV AVWRKNDENI

101 TLETVCHDPK LPYHDFILED AASPKCIMKE KKKPGETFFM CSCSSDECND

151 NIIFSEEYNT SNPDTGGGGS GGGGSGGGGS GGGGSTHTCP PCPAPELLGG 201 PSVFLFPPKP KDTLMISRTP EVTCWVDVS HEDPEVKFNW YVDGVEVHNA

251 KTKPREBQYN STYRW SVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS

301 KAKGQPREPQ VYTLPPSREE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP

351 ENNYKTTPPV LDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYT

401 QKSLSLSPGK (SEQ ID NO: 50)

The C-terminal lysine residue of the Fc domain can be deleted, Tire amino acid sequence of SEQ ID NO: 50 may optionally be provided with the lysine removed from the C- termmus (SEQ ID NO: 77):

1 GATIPPHVQK SDVEMEAQKD EIICPSCNRT AHPLRHINND MIVTDNNGAV

51 KFPQLCKFCD VRFSTCDNQK SCMSNCSITS ICEKPQEVCV AVWRKNDENI

101 TLETVCHDPK LPYHDFILED AASPKCIMKE KKKPGETFFM CSCSSDECND

151 NIIFSEEYNT SNPDTGGGGS GGGGSGGGGS GGGGSTHTCP PCPAPELLGG

201 PSVFLFPPKP KDTLMISRTP EVTCWVDVS HEDPEVKFNW YVDGVEVHNA

251 KTKPREEQYN STYRW SVLT VLHQDWLNGK EYKCKVSNKA,LPAPIEKTIS

301 KAKGQPREPQ VYTLPPSREE MTKNQVSLTC LVKGFYPSDI AVEWESNGQP

351 ENNYKTTPPV LDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYT

401 QKSLSLSPG (SEQ ID NO: 77)

ItίbKII (G4S)4-hFc: Amino Acid Sequence lacking leader sequence and lacking glycine prior to hTbHII portion

1 ATIPPHVQKS DVEMEAQKDE IICPSCNRTA HPLRHINNDM IVTDNNGAVK

51 FPQLCKFCDV RFSTCDNQKS CMSNCSITSI CEKPQEVCVA W RKNDENIT

101 LETVCHDPKL PYHDFILEDA ASPKCIMKEK KKPGETFFMC SCSSDECNDN

151 IIFSEEYNTS NPDTGGGGSG GGGSGGGGSG GGGSTHTCPP CPAPELLGGP

201 SVFLFPPKPK DTLMISRTPE VTCVWDVSH EDPEVKFKWY VDGVEVHNAK

251 TKPREEQYNS TYRW SVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK

301 AKGQPREPQV YTLPPSREEM TKNQVSLTCL VKGFYPSDIA VEWESNGQPE

351 NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ

401 KSLSLSPGK (SEQ ID NO: 52)

The C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 52 may optionally be provided with the lysine removed from the €- terminus (SEQ ID NO: 79):

1 ATIPPHVQKS DVEMEAQKDE IICPSCNRTA HPLRHINNDM IVTDNNGAVK

51 FPQLCKFCDV RFSTCDNQKS CMSNCSITSI CEKPQEVCVA W RKNDENIT

101 LETVCHDPKL PYHDFILEDA ASPKCIMKEK KKPGETFFMC SCSSDECNDN

151 IIFSEEYNTS NPDTGGGGSG GGGSGGGGSG GGGSTHTCPP CPAPELLGGP

201 SVFLFPPKPK DTLMISRTPE VTCVW DVSH EDPEVKFNWY VDGVEVHNAK

251 TKPREEQYNS TYRW SVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK

301 AKGQPREPQV YTLPPSREEM TKNQVSLTCL VKGFYPSDIA VEWESNGQPE

351 NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ

401 KSLSLSPG (SEQ ID NO: 79)

IiTbKII (G4S)4-hFc: Amino Acid Sequence lacking leader sequence and lacking glycine and alanine prior to IiTbKP portion

1 TIPPHVQKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF

51 PQLCKFCDVR FSTCDNQKSC MSNCSITSIC EKPQEVCVAV WRKNDENIT!

101 ETVCHDPKLP YHDFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI 151 IFSEEYNTSN PDTGGGGSGG GGSGGGGSGG GGSTHTCPPC PAPELLGGPS 201 VFLFPPKPKD TLMISRTPEV TCVW DVSHE DPEVKFNWYV DGVEVHNAKT 251 KPREEQYNST YRW SVLTVL HQDWLNGKEY KCKVSNKALP APIEKTISKA 301 KGOPREPQVY TLPPSREEMT KNQVSLTCLV KGFYPSDIAV EWE3NGOPEN 351 NYKTTPPVLD SDGSFFLYSK LTVDKSRWQQ GNVFSCSVMH EALHNHYTQK 401 SLSLSPGK (SEQ ID NO: 51)

The C-terminal lysine residue of the Fc domain can be deleted, Tire amino acid sequence of SEQ ID NO: 51 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 78):

1 TIPPHVOKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF

51 PQLCKFCDVR FSTCDNQKSC MSNCSITSIC EKPQEVCVAV WRKNDENITL

101 ETVCHDPKLP YHDFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI

151 IFSEEYNTSN PDTGGGGSGG GGSGGGGSGG GGSTHTCPPC PAPELLGGPS

201 VFLFPPKPKD TLMISRTPEV TCWVDVSHE DPEVKFNWYV DGVEVHNAKT 251 KPREEQYNST YRW SVLTVL HQDWLNGKEY KCKVSNKALP APIEKTISKA 301 KGOPREPQVY TLPPSREEMT KNQVSLTCLV KGFYPSDIAV EWE3NGOPEN 351 NYKTTPPVLD SDGSFFLYSK LTVDKSRWQQ GNVFSCSVMH EALHNHYTQK 401 SLSLSPG (SEQ ID NO: 78)

HTbKII (G4S)4~hFc: A mino Acid Sequence lacking leader sequence and lacking glycine, alanine, and threonine prior to Ii GbEII portion

1 IPPHVOKSDV EMEAQKDEII CPSCNRTAHP LRHINNDMIV TDNNGAVKFP

51 QLCKFCDVRF STCDNQKSCM SNCSITSICE KPOEVCVAVW RKNDENITLE

101 TVCHDPKLPY HDFILEDAAS PKCIMKEKKK PGETFFMCSC SSDECNDNII

151 FSEEYNTSNP DTGGGGSGGG GSGGGGSGGG GSTHTCPPCP APELLGGPSV

201 FLFPPKPKDT LMISRTPEVT CVW DVSHED PEVKFNWYVD GVEVHNAKTK

251 PREEQYNSTY RW SVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK

301 GQPREPQVYT LPPSREEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN

351 YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS

401 LSLSPGK (SEQ ID NO: 53)

The C-terminal lysine residue of the Fc domain can be deleted, Tire amino acid sequence of SEQ ID NO: 53 may optionally be provided with the lysine removed from the C- temiinus (SEQ ID NO: 80):

1 IPPHVQKSDV EMEAQKDEII CPSCNRTAHP LRHINNDMIV TDNNGAVKFP

51 QLCKFCDVRF STCDNQKSCM SNCSITSICE KPOEVCVAVW RKNDENITLE

101 TVCHDPKLPY HDFILEDAAS PKCIMKEKKK PGETFFMCSC SSDECNDNII

151 FSEEYNTSNP DTGGGGSGGG GSGGGGSGGG GSTHTCPPCP APELLGGPSV

201 FLFPPKPKDT LMISRTPEVT CVW DVSHED PEVKFNWYVD GVEVHNAKTK

251 PREEQYNSTY RW SVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK

301 GQPREPQVYT LPPSREEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN

351 YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS

401 LSLSPG (SEQ ID NO: 80) hTbKP (G4S)4~hFc: A mino Acid Sequence lacking leader sequence and lacking glycine, alanine, threonine, and isoleucine prior to IίTbEII portion

1 PPHVQKSDVE MEAQKDEIIC PSCNRTAHPL RHINNDMIVT DNNGAVKFPQ 51 LCKFCDVRFS TCDNQKSCM3 NCSITSICEK PQEVCVAVWR KNDENITLET

101 VCHDPKLPYH DFILEDAASP KCIMKEKKKP GETFFMCSCS SDECNDNIIF

151 SEEYNTSNPD TGGGGSGGGG SGGGGSGGGG STHTCPPCPA PELLGGPSVF

201 LFPPKPKDTL MISRTPEVTC WVDVSHEDP EVKFNWYVDG VEVHNAKTKP

251 REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG

301 QPREPQVYTL PPSREEMTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY

351 KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL

401 SLSPGK (SEQ ID NO: 54)

The C-terminai lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 54 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 81):

1 PPHVQKSDVE MEAQKDEIIC PSCNRTAHPL RHINNDMIVT DNNGAVKFPQ

51 LCKFCDVRFS TCDNQKSCMS NCSITSICEK PQEVCVAVWR KNDENITLET

101 VCHDPKLPYH DFILEDAASP KCIMKEKKKP GETFFMCSCS SDECNDNIIF

151 SEEYNTSNPD TGGGGSGGGG SGGGGSGGGG STHTCPPCPA PELLGGPSVF

201 LFPPKPKDTL MISRTPEVTC WVDVSHEDP EVKFNWYVDG VEVHNAKTKP

251 REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC K.VSNKALPAP IEKTISKAKG

301 QPREPQVYTL PPSREEMTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY

351 KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL

401 SL3PG (SEQ ID NO: 81)

HTbKII (G4S)4-hFc: Amino Acid Sequence lacking leader sequence and lacking glycine, alanine, threonine, isoleucine, and proline prior to hTbKII portion

1 PHVQKSDVEM EAQKDEIICP SCNRTAHPLR HINNDMIVTD NNGAVKFPQL

51 CKFCDVRFST CDNQKSCMSN CSITSICEKP QEVCVAVWRK NDENITLETV

101 CHDPKLPYHD FILEDAASPK CIMKEKKKPG ETFFMCSCSS DECNDNIIFS

151 EEYNTSNPDT GGGGSGGGGS GGGGSGGGGS THTCPPCPAP ELLGGPSVFL

201 FPPKPKDTLM ISRTPEVTCV W DVSHEDPE VKFNWYVDGV EVHNAKTKPR

251 EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ

301 PREPQVYTLP PSREEMTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK

351 TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV F3CSVMHEAL HNHYTQKSLS

401 LSPGK (SEQ ID NO: 55)

The C-terminai lysine residue of the Fc domain can be deleted. The ammo acid sequence of SEQ ID NO: 55 may optionally be provided with the lysine removed from the C- temiinus (SEQ ID NO: 82):

1 PHVQKSDVEM EAQKDEIICP SCNRTAHPLR HINNDMIVTD NNGAVKFPQL

51 CKFCDVRFST CDNQKSCMSN CSITSICEKP QEVCVAVWRK NDENITLETV

101 CHDPKLPYHD FILEDAASPK CIMKEKKKPG ETFFMCSCSS DECNDNIIFS

151 EEYNTSNPDT GGGGSGGGGS GGGGSGGGGS THTCPPCPAP ELLGGPSVFL

201 FPPKPKDTLM ISRTPEVTCV W DVSHEDPE VKFNWYVDGV EVHNAKTKPR

251 EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ

301 PREPQVYTLP PSREEMTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK

351 TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL HNHYTQKSLS

401 LSPG (SEQ ID NO: 82) hTfiRll (G4S)4-hFc: Amino Acid Sequence lacking leader sequence and lacking glycine, alanine, threonine, isoleucine, proline, and proline prior to hTfiRll portion

1 HVOKSDVEME AQKDEIICPS CNRTAHPLRH INNDMIVTDN NGAVKFPQLC

51 KFCDVRFSTC DNQKSCMSNC SITSICEKPQ EVCVAVWRKN DENITLETVC

101 HDPKLPYHDF ILEDAASPKC IMKEKKKPGE TFFMCSCSSD ECNDNIIFSE

151 EYNTSNPDTG GGG5GGGGSG GGGSGGGGST HTCPPCPAPE LLGGPSVFLF

201 PPKPKDTLMI SRTPEVTCW VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE

251 EQYNSTYRW SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP

301 REPQVYTLPP SREEMTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT

351 TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL

401 SPGK (SEQ ID NO: 56)

The C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 56 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 83):

1 HVOKSDVEME AQKDEIICPS CNRTAHPLRH INNDMIVTDN NGAVKFPQLC

51 KFCDVRFSTC DNQKSCMSNC SITSICEKPQ EVCVAVWRKN DENITLETVC

101 HDPKLPYHDF ILEDAASPKC IMKEKKKPGE TFFMCSCSSD ECNDNIIFSE

151 EYNTSNPDTG GGGSGGGGSG GGGSGGGGST HTCPPCPAPE LLGGPSVFLF

201 PPKPKDTLMI SRTPEVTCW VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE

251 EQYNSTYRW SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP

301 REPQVYTLPP SREEMTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT

351 TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL

401 SPG (SEQ ID NO: 83} hTfiRll (G4S)2-hFc: Nucleic Acid Sequence

1 ATGGATGCAA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC

51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCCAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT 151 AGGACTGCCC ATCCACTGAG ACATATTAAT AACGACATGA TAGTCACTGA 201 CAACAACGGT GCAGTCAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA

251 GATTTTCCAC CTGTGACAAC CAGAAATCCT GCATGAGCAA CTGCAGCATC

301 ACCTCCATCT GTGAGAAGCC ACAGGAAGTC TGTGTGGCTG TATGGAGAAA

351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC 401 CCTACCATGA CTTTATTCTG GAAGATGCTG CTTCTCCAAA GTGCATTATG 451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC

501 TGATGAGTGC AATGACAACA TCATCTTCTC AGAAGAATAT AACACCAGCA 551 ATCCTGACAC CGGTGGAGGT GGTTCTGGAG GTGGTGGAAG TACTCACACA

601 TGCCCACCGT GCCCAGCACC TGAACTCCTG GGGGGACCGT CAGTCTTCCT

651 CTTCCCCCCA AAACCCAAGG ACACCCTCAT GATCTCCCGG ACCCCTGAGG

701 TCACATGCGT GGTGGTGGAC GTGAGCCACG AAGACCCTGA GGTCAAGTTC 751 AACTGGTACG TGGACGGCGT GGAGGTGCAT AATGCCAAGA CAAAGCCGCG

801 GGAGGAGCAG TACAACAGCA CGTACCGTGT GGTCAGCGTC CTCACCGTCC

851 TGCACCAGGA CTGGCTGAAT GGCAAGGAGT ACAAGTGCAA GGTCTCCAAC

901 AAAGCCCTCC CAGCCCCCAT CGAGAAAACC ATCTCCAAAG CCAAAGGGCA

951 GCCCCGAGAA CCACAGGTGT ACACCCTGCC CCCATCCCGG GAGGAGATGA

1001 CCAAGAACCA GGTCAGCCTG ACCTGCCTGG TCAAAGGCTT CTATCCCAGC 1051 GACATCGCCG TGGAGTGGGA GAGCAATGGG CAGCCGGAGA ACAACTACAA 1101 GACCACGCCT CCCGTGCTGG ACTCCGACGG CTCCTTCTTC CTCTATAGCA

1151 AGCTCACCGT GGACAAGAGC AGGTGGCAGC AGGGGAACGT CTTCTCATGC

1201 TCCGTGATGC ATGAGGCTCT GCACAACCAC TACACGCAGA AGAGCCTCTC 1251 CCTGTCTCCG GGTAAATGA (SEQ ID NO: 14) hTfjRlI (G4S)2-hFc: Amino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAW RKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSTHT

201 CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCW VD VSHEDPEVKF

251 NWYVDGVEVH NAKTKPREEQ YNSTYRW SV LTYLHQDWLN GKEYKCKVSN

301 KALPAPIEKT ISKAKGQPRE PQVYTLPPSR EEMTKNQVSL TCLVKGFYPS

351 DIAVEWESKG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC

401 SVMHEALHNH YTQKSLSLSP GK (SEQ ID NO: 15)

The C-terminai lysine residue of the Fc domain can be deleted. The ammo acid sequence of SEQ ID NO: 15 may optionally he provided with the lysine removed from the C- terminus (SEQ ID NO: 70):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAW RKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSTHT

201 CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCW VD VSHEDPEVKF

251 NWYVDGVEVH NAKTKPREEQ YNSTYRW SV LTVLHQDWLN GKEYKCKVSN

301 KALPAPIEKT ISKAKGQPRE PQVYTLPPSR EEMTKNQVSL TCLVKGFYPS

351 DIAVEWBSNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC

401 SVMHEALHNH YTQKSLSLSP G (SEQ ID NO: 70) hT ' bKίϊ extended hinge-hFc: Nucleic Acid Sequence 1 ATGGATGCAA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC 51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCCAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT

151 AGGACTGCCC ATCCACTGAG ACATATTAAT AAGGAGATGA TAGTCACTGA

201 CAACAACGGT GCAGTCAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA 251 GATTTTCCAC CTGTGACAAC CAGAAATCCT GCATGAGCAA CTGCAGCATC 301 ACCTCCATCT GTGAGAAGCC ACAGGAAGTC TGTGTGGCTG TATGGAGAAA

351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC

401 CCTAGCATGA CTTTATTCTG GAAGATGCTG CTTCTCCAAA GTGCATTATG

451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC

501 TGATGAGTGC AATGACAACA TCATCTTCTC AGAAGAATAT AACACCAGCA 551 ATCCTGACAC CGGTGGTGGA CCCAAATCTT GTGACAAAAC TCACACATGC

601 CCACCGTGCC CAGCACCTGA ACTCCTGGGG GGACCGTCAG TCTTCCTCTT 651 CCCCCCAAAA CCCAAGGACA CCCTCATGAT CTCCCGGACC CCTGAGGTCA 701 CATGCGTGGT GGTGGACGTG AGCCACGAAG ACCCTGAGGT CAAGTTCAAC 751 TGGTACGTGG ACGGCGTGGA GGTGCATAAT GCCAAGACAA AGCCGCGGGA 801 GGAGCAGTAC AACAGCACGT ACCGTGTGGT CAGCGTCCTC ACCGTCCTGC 851 ACCAGGACTG GCTGAATGGC AAGGAGTAGA AGTGCAAGGT CTCCAACAAA

901 GCCCTCCCAG CCCCCATCGA GAAAACCATC TCCAAAGCCA AAGGGCAGCC 951 CCGAGAACCA CAGGTGTACA CCCTGCCCCC ATCCCGGGAG GAGATGACCA

1001 AGAACCAGGT CAGCCTGACC TGCCTGGTCA AAGGCTTCTA TCCCAGCGAC

1051 ATCGCCGTGG AGTGGGAGAG CAATGGGCAG CCGGAGAACA ACTACAAGAC

1101 CACGCCTCCC GTGCTGGACT CCGACGGCTC CTTCTTCCTC TATAGCAAGC

1151 TCACCGTGGA CAAGAGCAGG TGGCAGCAGG GGAACGTCTT CTCATGCTCC 1201 GTGATGCATG AGGCTCTGCA CAACCACTAC ACGCAGAAGA GCCTCTCCCT 1251 GTCCCCGGGT AAATGA (SEQ ID NO: 16}

MbKII extended hinge-hFc: Amino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG PKSCDKTHTC

201 PPCPAPELLG GPSYFLFPPK PKDTLMISRT PEVTCVW DV SHEDPEVKFN

251 WYVDGVEVHN AKTKPREEQY NSTYRW SVL TVLHQDWLNG KEYKCKVSNK

301 ALPAPIEKTI 5KAKGQPREP QVYTLPPSRE EMTKNQVSLT CLVKGFYPSD

351 IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS

401 VMHEALHNHY TQKSLSLSPG K (SEQ ID NO: 17)

The C-terminai lysine residue of the Fc domain can be deleted. The ammo acid sequence of SEQ ID NO: 17 may optionally he provided with the lysine removed from the C- terminus (SEQ ID NO: 71):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG PKSCDKTHTC

201 PPCPAPELLG GPSVFLFPPK PKDTLMISRT PEVTCVW DV SHEDPEVKFN

251 WYVDGVEVHN AKTKPREEQY NSTYRW SVL TVLHQDWLNG KEYKCKVSNK

301 ALPAPIEKTI SKAKGQPREP QVYTLPPSRE EMTKNQVSLT CLVKGFYPSD

351 IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS

401 VMHEALHNHY TQKSLSLSPG (SEQ ID NO: 71) hT’ /]RlI (G4S)5-hFc: Amino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSGGGGSGGG GSTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT

251 CWVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RW SVLTVLH

301 QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSREEMTK

351 NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL

401 TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGK (SEQ ID NO: 44)

The C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 44 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 75):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSGGGGSGGG GSTHTCPPCP APELLGGPSV- FLFPPKPKDT LMISRTPEVT

251 CWVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RW SVLTVLH

301 QDWLNGKEYK CKVSNKALPA,PIEKTISKAK GQPREPQVYT LPPSREEMTK

351 NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL

401 TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPG (SEQ ID NO: 75) ^iTbEII (G4S)6-hFc: A mino Acid Sequence

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAHPLRHIN NDMIVTDNKG AVKFPQLCKF CDVRFSTCDN QKSCMSNCSI

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF EMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSGGGGSGGG GSGGGGSTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR

251 TPEVTCW VD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRW SV

301 LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR

351 EEMTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF

401 LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK (SEQ ID

NO: 45)

The C-terminai lysine residue of the Fc domain can be deleted. The ammo acid sequence of SEQ ID NO: 45 may optionally he provided with the lysine removed from the C- terminus (SEQ ID NO: 76):

1 MDAMKRGLCC VLLLCGAVFV SPGATIPPHV QKSDVEMEAQ KDEIICPSCN

51 RTAPIPLRHIN NDMIVTDNNG AVKFPQLCKF CDVRF3TCDN QKSCMSNC31

101 TSICEKPQEV CVAVWRKNDE NITLETVCHD PKLPYHDFIL EDAASPKCIM

151 KEKKKPGETF FMCSCSSDEC NDNIIFSEEY NTSNPDTGGG GSGGGGSGGG

201 GSGGGGSGGG GSGGGGSTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR

251 TPEVTCWVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRW SV

301 LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR

351 EEMTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF

401 LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP G

(SEQ ID NO: 76) hTbKP (G4S)5-hFc: Nucleotide Sequence

1 ATGGATGCAA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC

51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCCAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT

151 AGGACTGCCC ATCCACTGAG ACATATTAAT AACGACATGA TAGTCACTGA

201 CAACAACGGT GCAGTCAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA

251 GATTTTCCAC CTGTGACAAC CAGAAATCCT GCATGAGCAA CTGCAGCATC

301 ACCTCCATCT GTGAGAAGCC ACAGGAAGTC TGTGTGGCTG TATGGAGAAA

351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC

401 CCTACCATGA CTTTATTCTG GAAGATGCTG CTTCTCCAAA GTGCATTATG

451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC

501 TGATGAGTGC AATGACAACA TCATCTTCTC AGAAGAATAT AACACCAGCA

551 ATCCTGACAC CGGTGGAGGA GGTTCTGGTG GTGGAGGTTC TGGAGGTGGA

601 GGAAGTGGTG GAGGTGGTTC TGGAGGTGGT GGAAGTAGTC ACACATGCCC

651 ACCGTGCCCA GCACCTGAAC TCCTGGGGGG ACCGTCAGTC TTCCTCTTCC

701 CCCCAAAACC CAAGGACACC CTCATGATCT CCCGGACCCC TGAGGTCACA

751 TGCGTGGTGG TGGACGTGAG CCACGAAGAC CCTGAGGTCA AGTTCAACTG

801 GTACGTGGAC GGCGTGGAGG TGCATAATGC CAAGACAAAG CCGCGGGAGG

851 AGCAGTACAA CAGCACGTAC CGTGTGGTCA GCGTCCTCAC CGTCCTGCAC

901 CAGGACTGGC TGAATGGCAA GGAGTACAAG TGCAAGGTCT CCAACAAAGC

951 CCTCCCAGCC CCCATCGAGA AAACCATCTC CAAAGCCAAA GGGCAGCCCC

1001 GAGAACCACA GGTGTACACC CTGCCCCCAT CCCGGGAGGA GATGACCAAG

1051 AACCAGGTCA GCCTGACCTG CCTGGTCAAA GGCTTCTATC CCAGCGACAT

1101 CGCCGTGGAG TGGGAGAGCA ATGGGCAGCC GGAGAACAAC TACAAGACCA

1151 CGCCTCCCGT GCTGGACTCC GACGGCTCCT TCTTCCTCTA TAGCAAGCTC

1201 ACCGTGGACA AGAGCAGGTG GCAGCAGGGG AACGTCTTCT CATGCTCCGT 1251 GATGCATGAG GCTCTGCACA ACCACTACAC GCAGAAGAGC CTCTCCCTGT

1301 CTCCGGGTAA ATGA (SEQ ID NO: 46) ίiTbIϋI (G4S)6-hFc: Nucleotide Sequence

1 ATGGATGCAA TGAAGAGAGG GCTCTGCTGT GTGCTGCTGC TGTGTGGAGC

51 AGTCTTCGTT TCGCCCGGCG CCACGATCCC ACCGCACGTT CAGAAGTCGG

101 ATGTGGAAAT GGAGGCCGAG AAAGATGAAA TCATCTGCCC CAGCTGTAAT

151 AGGACTGCCC ATCCACTGAG ACATATTAAT AACGACATGA TAGTCACTGA

201 CAACAACGGT GCAGTCAAGT TTCCACAACT GTGTAAATTT TGTGATGTGA

251 GATTTTCCAC CTGTGACAAC CAGAAATCCT GCATGAGCAA CTGCAGCATC

301 ACCTCCATCT GTGAGAAGCC ACAGGAAGTC TGTGTGGCTG TATGGAGAAA

351 GAATGACGAG AACATAACAC TAGAGACAGT TTGCCATGAC CCCAAGCTCC

401 CCTACCATGA CTTTATTCTG GAAGATGCTG CTTCTCCAAA GTGCATTATG

451 AAGGAAAAAA AAAAGCCTGG TGAGACTTTC TTCATGTGTT CCTGTAGCTC

501 TGATGAGTGC AATGACAACA TCATCTTCTC AGAAGAATAT AACACCAGCA

551 ATCCTGACAC CGGTGGAGGT GGAAGTGGTG GAGGAGGTTC TGGTGGTGGA

601 GGTTCTGGAG GTGGAGGAAG TGGTGGAGGT GGTTCTGGAG GTGGTGGAAG

651 TACTCACACA TGCCCACCGT GCCCAGCACC TGAACTCCTG GGGGGACCGT

701 CAGTCTTCCT CTTCCCCCCA AAACCCAAGG ACACCCTCAT GATCTCCCGG

751 ACCCCTGAGG TCACATGCGT GGTGGTGGAC GTGAGCCACG AAGACCCTGA

801 GGTCAAGTTC AACTGGTACG TGGACGGCGT GGAGGTGCAT AATGCCAAGA

851 CAAAGCCGCG GGAGGAGCAG TACAACAGCA CGTACCGTGT GGTCAGCGTC

901 CTCACCGTCC TGCACCAGGA CTGGCTGAAT GGCAAGGAGT ACAAGTGCAA

951 GGTCTCCAAC AAAGCCCTCC CAGCCCCCAT CGAGAAAACC ATCTCCAAAG

1001 CCAAAGGGCA GCCCCGAGAA CCACAGGTGT ACACCCTGCC CCCATCCCGG

1051 GAGGAGATGA CCAAGAACCA GGTCAGCCTG ACCTGCCTGG TCAAAGGCTT

1101 CTATCCCAGC GACATCGCCG TGGAGTGGGA GAGCAATGGG CAGCCGGAGA

1151 ACAACTACAA GACCACGCCT CCCGTGCTGG ACTCCGACGG CTCCTTCTTC

1201 CTCTATAGCA AGCTCACCGT GGACAAGAGC AGGTGGCAGC AGGGGAACGT

1251 CTTCTCATGC TCCGTGATGC ATGAGGCTCT GCACAACCAC TACACGCAGA

1301 AGAGCCTCTC CCTGTCTCCG GGTAAATGA (SEQ ID NO: 47)

The various constructs were successfully expressed in CHO cells and were purified to a high degree of purity as determined by analytical size-exclusion chromatography and SDS-PAGE. The IiTbKII (G4S)2-hFc, nTfiRIf (G4S)3-hFc, IiTbKII (G4S)4-hFc, IiTbMI (G4S)5-hFc and IiTbKII (G4S)6-hFc proteins displayed similarly strong stability as determined by SDS-PAGE analysis when maintained in PBS for 13 days at 37°C. The hTBRli (G4S)2-hFc, hT ' BRJl (G4S)3-hFc, IiTbKII (G4S)4-hFc proteins were also maintained in rat, mouse or human serum and displayed similarly strong stability.

WR1I ECD variants

In addition to the TbίIP domains included in the fusion proteins described above (e.g., SEQ ID NO: 18), the disclosure also contemplates fusion proteins comprising alternative TbίIίί domains. For example, the fusion protein may comprise the wild-type 1iTbKII £ίioh (23- 159) sequence shown below (SEQ ID NO: 27) or any of the other TbKII polypeptides disclosed below:

1 TIPPHVQKSV NNDMIVTDNN GAVKFPQLCK FCDVRFSTCD NQKSCMSNCS

51 ITSICEKPQE VCVAVWRKND ENITLETVCH DPKLPYHDFI LEDAASPKCI 101 MKEKKKPGET FFMCSCSSDE CNDNIIFSEE YNTSNPD (SEQ ID NO:

27)

(1) The hT[)RlIshori(23 - 159/D 11 OK) amino acid sequence shown below (SEQ ID NO: 36), in which the substituted residue is underlined.

1 TIPPHVQKSV NNDMIVTDNN GAVKFPQLCK FCDVRFSTCD NQKSCMSNCS 51 ITSICEKPQE VCVAVWRKND ENITLETVCH DPKLPYHKFI LEDAASPKCI

101 MKEKKKPGET FFMCSCSSDE CNDNIIFSEE YNTSNPD (SEQ ID NO:

36)

(2) The N-terminally truncated hTpRII Short (29-!59) amino acid sequence shown below (SEQ ID NO: 28). 1 QKSVNNDMIV TDNNGAVKFP QLCKFCDVRF STCDNQKSCM SNCSITSICE

51 KPQEVCVAVW RKNDENITLE TVCHDPKLPY HDFILEDAAS PKCIMKEKKK 101 PGETFFMCSC SSDECNDNII FSEEYNTSNP D (SEQ ID NO: 28)

(3) The N-terminally truncated hTpRIIshort(35-159) amino acid sequence shown below (SEQ ID NO: 29). 1 DMIVTDNNGA VKFPQLCKFC DVRFSTCDNQ KSCMSNCSIT SICEKPQEVC

51 VAVWRKNDEN ITLETVCHDP KLPYHDFILE DAASPKCIMK EKKKPGETFF 101 MCSCSSDECN DNIIFSEEYN TSNPD (SEQ ID NO: 29)

(4) The C-terminally truncated hTpRiishort(23-153) amino acid sequence shown below (SEQ ID NO: 30). 1 TIPPHVQKSV NNDMIVTDNN GAVKFPQLCK FCDVRFSTCD NQKSCMSNCS

51 ITSICEKPQE VCVAVWRKND ENITLETVCH DPKLPYHDFI LEDAASPKCI

101 MKEKKKPGET FFMCSCSSDE CNDNIIFSEE Y (SEQ ID NO: 30)

(5) The C-terminally truncated hTpRIIsbon(23-153/N70D) ammo acid sequence shown below (SEQ ID NO: 38), in which the substituted residue is underlined. 1 TIPPHVQKSV NNDMIVTDNN GAVKFPQLCK FCDVRFSTCD NQKSCMSDCS 51 ITSICEKPQE VCVAVWRKND ENITLETVCH DPKLPYHDFI LEDAASPKCI

101 MKEKKKPGET FFMCSCSSDE CNDNIIFSEE Y (SEQ ID NO: 38)

Applicants also envision five corresponding variants (SEQ ID NOs: 37, 33, 34, 39) based on the wild-type hTpRIIiong(23-184) sequence shown above and below (SEQ ID NO: 49), in which the 25 amino-acid insertion is underlined. Note that splicing results in a conservative ammo acid substitution (Val®Ile) at the flanking position C-terminal to the insertion.

1 TIPPHVQKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF

51 PQLCKFCDVR FSTCDNQKSC MSNCSITSIC EKPQEVCVAV WRKNDENITL

101 ETVCHDPKLP YHDFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI

151 IFSEEYNTSN PD (SEQ ID NO: 49)

(1) The liTpRIIiong(23-184/D135K) amino acid sequence shown below (SEQ ID NO: 37), in which the substituted residue is double underlined.

1 TIPPHVQKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF

51 PQLCKFCDVR FSTCDNQKSC MSNCSITSIC EKPQEVCVAV WRKNDENITL

101 ETVCHDPKLP YHKFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI

151 IFSEEYNTSN PD (SEQ ID NO: 37)

(2) The N-terminally truncated hTpRIIiong(29-184) amino acid sequence shown below (SEQ ID NO: 33).

1 QKSDVEMEAQ KDEIICPSCN RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF

51 CDVRFSTCDN QKSCMSNCSI TSICEKPQEV CVAVWRKNDE NITLETVCHD

101 PKLPYHDFIL EDAASPKCIM KEKKKPGETF FMCSCSSDEC NDNIIFSEEY

151 NTSNPD (SEQ ID NO: 33)

(3) The N-terminally truncated hTpRIii 0ng (6(M 84) amino acid sequence shown below (same as SEQ ID NO: 29),

1 DMIVTDNNGA VKFPQLCKFC DVRFSTCDNQ KSCMSNCSIT SICEKPQEVC

51 VAVWRKNDEN ITLETVCHDP KLPYHDFILE DAASPKCIMK EKKKPGETFF

101 MCSCSSDECN DNIIFSEEYN TSNPD (same as SEQ ID NO: 29)

(4) The C-terminaily truncated hTpRIIi 0ng (23-178) amino acid sequence shown below (SEQ ID NO: 34),

1 TIPPHVQKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF 51 PQLCKFCDVR FSTCDNQKSC MSNCSITSIC EKPQEVCVAV WRKNDENITL

101 ETVCHDPKLP YHDFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI 151 IFSEEY (SEQ ID NO: 34)

(5) The C-terminally truncated hTpRIIiong(23-178/N95D) amino acid sequence shown below (SEQ ID NO: 39), in which the substituted residue is double underlined.

1 TIPPHVQKSD VEMEAQKDEI ICPSCNRTAH PLRHINNDMI VTDNNGAVKF

51 PQLCKFCDVR FSTCDNQKSC MSDCSITSIC EKPQEVCVAV WRKNDENITL

101 ETVCHDPKLP YHDFILEDAA SPKCIMKEKK KPGETFFMCS CSSDECNDNI

151 IFSEEY (SEQ ID NO: 39)

Additional TbKIί ECD variants include:

(A) The N- and C-terminally truncated hTpRIL >hort (35-153) or 1iTbKPΐoi ¾ (60-178) amino acid sequence shown below (SEQ ID NO: 32).

1 DMIVTDNNGA VKFPQLCKFC DVRFSTCDNQ KSCMSNCSIT SICEKPQEVC 51 VAVWRKNDEN ITLETVCHDP KLPYHDFILE DAASPKCIMK EKKKPGETFF 101 MCSCSSDECN DNIIFSEEY (SEQ ID NO: 32)

(B) The N- and C-terminally truncated 1ϊTbKII 3ΐioi( (29-153) amino acid sequence shown below (SEQ ID NO: 31).

1 QKSVNNDMIV TDNNGAVKFP QLCKFCDVRF STCDNQKSCM SNCSITSICE

51 KPQEVCVAVW RKNDENITLE TVCHDPKLPY HDFILEDAAS PKCIMKEKKK

101 PGETFFMCSC SSDECNDNII FSEEY (SEQ ID NO: 31)

(C) The N- and C-terminally truncated hTpRJLong(29-178) amino acid sequence shown below (SEQ ID NO: 35),

1 QKSDVEMEAQ KDEIICPSCN RTAHPLRHIN NDMIVTDNNG AVKFPQLCKF

51 CDVRFSTCDN QKSCMSNCSI TSICEKPQEV CVAVWRKNDE NITLETVCHD

101 PKLPYHDFIL EDAASPKCIM KEKKKPGETF FMCSCSSDEC NDNIIFSEEY

(SEQ ID NO: 35)

Any of the above variants (SEQ ID NO: 36, 28, 29, 30, 38, 37, 33, 34, 39, 32, 31, and 35) could incorporate an insertion of 36 amino acids (SEQ ID NO: 41) between the pair of glutamate residues (positions 151 and 152 of SEQ ID NO: 1, or positions 176 and 177 of SEQ ID NO: 2) located near the C-terminus of the hTpRH ECD, as occurs naturally in the IiTbKII isofomi C (Konrad et al., BMC Genomics 8:318, 2007).

GRCKIRHIGS NNRLQRSTCQ NTGWESAHVM KTPGFR ( SEQ ID NO : 41 )

As an example, the paired glutamate residues flanking the optional insertion site are denoted below (underlined) for the hTpRII Short (29-159) variant (SEQ ID NO: 28).

1 QKSVNNDMIV TDNNGAVKFP QLCKFCDVRF STCDNQKSCM SNCSITSICE 51 KPQEVCVAVW RKNDENITLE TVCHDPKLPY HDFILEDAAS PKCIMKEKKK 101 PGETFFMCSC SSDECNDNI I FSEEYNTSNP D ( SEQ ID NO : 28 )

Fc domain variants

While the constructs described above were generated with an Fc domain having the amino acid sequence of SEQ ID NO: 20, the disclosure contemplates liTfiRII-hFc fusion proteins comprising alternative Fc domains, including a human lgG2 Fc domain (SEQ ID NO: 42, below) or full-length human IgGl Fc (hGIFc) (SEQ ID NO: 43, below). Optionally, a polypeptide unrelated to an Fc domain could be attached in place of the Fc domain.

1 VECPPCPAPP VAGPSVFLFP PKPKDTLMIS RTPEVTCVW DVSHEDPEVQ

51 FNWYVDGVEV HNAKTKPREE QFNST FRVVS VLTVVHQDWL NGKEYKCKVS

101 NKGLPAPIEK TISKTKGQPR EPQVYTLPPS REEMTKNQVS LTCLVKGFYP

151 SDIAVEWESN GQPENNYKTT PPMLDSDGSF FLYSKLTVDK SRWQQGNVFS

201 CSVMHEALHN HYTQKSLSLS PGK ( SEQ ID NO : 42 )

The C-temimal lvsine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 42 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 73):

1 VECPPCPAPP VAGPSVFLFP PKPKDTLMIS RT PEVTCVW DVSHEDPEVQ

51 FNWYVDGVEV HNAKTKPREE QFNSTFRVVS VLTVVHQDWL NGKEYKCKVS

101 NKGLPAPIEK TISKTKGQPR EPQVYTLPPS REEMTKNQVS LTCLVKGFYP

151 SDIAVEWESN GQPENNYKTT PPMLDSDGSF FLYSKLTVDK SRWQQGNVFS

201 CSVMHEALHN HYTQKSLSLS PG ( SEQ ID NO : 73 ) i GGPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVW

51 DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRVVS VLTVLHQDWL

101 NGKEYKCKVS NKALPAPIEK TISKAKGQPR EPQVYTLPPS REEMTKNQVS 151 LTCLVKGFYP SDIAVEWESN GQPENNYKTT PPVLDSDGSF FLYSKLTVDK

201 SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PGK ( SEQ ID NO : 43 )

The C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 43 may optionally be provided with the lysine removed from the C- terminus (SEQ ID NO: 74):

1 GGPKSCDKTH TCPPCPAPEL LGGPSVFLFP PKPKDTLMIS RTPEVTCVW 51 DVSHEDPEVK FNWYVDGVEV HNAKTKPREE QYNSTYRW S VLTVLHQDWL 101 NGKEYKCKVS NKALPAPIEK T ISKAKGQPR EPQVYTLPPS REEMTKNQVS 151 LTCLVKGFYP SDIAVEWESN GQ PENNY KT T PPVLDSDGSF FLYSKLTVDK 201 SRWQQGNVFS CSVMHEALHN HYTQKSLSLS PG (SEQ ID NO: 74)

Leader sequence variants

While the generated constructs described above included the TP A leader sequence, alternative leader sequences may be used, such as the native leader sequence (SEQ ID NO: 22- below) or the honey bee melittin (SEQ ID NO: 24- below) leader sequences.

Native: MGRGLLRGLWPLHIVLWTRIAS ( SEQ ID NO : 22 )

Honey bee melittin (HBML): MKFLVNVALVFMVVYI SYIYA ( SEQ ID NO : 24 ) mT / SRll-mFc mXpRII-mFc comprises murine TbKII extracellular domain and murine IgG2a Fc.

The signal sequence is underlined. The linker in ihTbKII-piRo is TGGG (SEQ ID NO: 9), bolded and underlined below. The human version (SEQ ID NO: 48) has a longer linker (SEQ ID NO: 6). The unprocessed sequence (SEQ ID NO: 84) is below:

1 MDAMKRGLCC VLLLCGAVFV SPGAIPPHVP KSDVEMEAQK DASIHLSCNR 51 TIHPLKHFNS DVMASDNGGA VKLPQLCKFC DVRLSTCDNQ KSCMSNCSIT 101 AICEKPHEVC VAVWRKNDKN ITLETVCHDP KLTYHGFTLE DAASPKCVMK 151 EKKRAGETFF MCACNMEECN DYIIFSEEYT TSSPDTGGGE PRVPITQNPC 201 PPLKECPPCA APDLLGGPSV FIFPPKIKDV LMISLSPMVT CWVDVSEDD 251 PDVQISWFVN NVEVHTAQTQ THREDYNSTL RVV3ALPIQH QDWMSGKEFK 301 CKVNNRALPS PIEKTISKPR GPVRAPQVYV LPPPAEEMTK KEFSLTCMIT 351 GFLPAEIAVD WTSNGRTEQN YKNTATVLDS DGSYFMYSKL RVQKSTWERG 401 SLFACSVVHE GLHNHLTTKT ISRSLGK* (SEQ ID NO: 84)

Die mature version of mTpRII-mFc is shown as SEQ ID NO: 85:

1 IPPHVPKSDV EMEAQKDASI HLSCNRTIHP LKHFNSDVMA SDNGGAVKLP 51 QLCKFCDVRL STCDNQKSCM SNCSITAICE KPHEVCVAVW RKNDKNITLE 101 TVCHDPKLTY HGFTLEDAAS PKCVMKEKKR AGETFFMCAC NMEECNDYI I 151 FSEEYTTS3P DTGGGEPRVP ITQNPCPPLK ECPPCAAPDL LGGPSVFIFP 201 PKIKDVLMIS LSPMVTCVW DVSEDDPDVQ ISWFVNNVEV HTAQTQTHRE 251 DYNSTLRWS ALPIQHQDWM 3GKEFKCKVN NRALPSPIEK TISKPRGPVR 301 APQVYVLPPP AEEMTKKEFS LTCMITGFLP AEIAVDWTSN GRTEQNYKNT 351 ATVLDSDGSY FMYSKLRVQK STWERGSLFA CSWHEGLHN HLTTKTISRS 401 LGK (SEQ ID NO: 85)

Furthermore, the C-terminal lysine residue of the Fc domain can be deleted. The amino acid sequence of SEQ ID NO: 84 may optionally be provided with the lysine removed from the C-terminus (SEQ ID NO: 86)

1 IPPHVPKSDV EMEAQKDAS I HLSCNRTIHP LKHFNSDVMA SDNGGAVKLP 51 QLCKFCDVRL STCDNQKSCM SNCSITAICE KPHEVCVAVW RKNDKNITLE 101 TVCHDPKLTY HGFTLEDAAS PKCVMKEKKR AGETFFMCAC NMEECNDYI I 151 FSEEYTTSSP DTGGGEPRVP ITQNPCPPLK ECPPCAAPDL LGGPSVFIFP 201 PKIKDVLMIS LSPMVTCVW DVSEDDPDVQ ISWFVNNVEV HTAQTQTHRE 251 DYNSTLRWS ALPIQHQDWM SGKEFKCKVN NRALPSPIEK TISKPRGPVR 301 APQVYVLPPP AEEMTKKEFS LTCMITGFLP AEIAVDWTSN GRTEQNYKNT 351 ATVLDSDGSY FMYSKLRVQK STWERGSLFA CSWHEGLHN HLTTKTISRS 401 LG (SEQ ID NO: 86) mTpRII-mFe nucleic acid sequence (SEQ ID NO: 87): AT G GAT G C A A TGAAGAGAGG G C T C T G C T GT GTGCTGCTGC TGTGTGGAGC AGTCTTCGTT TCGCCCGGCG C C AT C C C G C C G C AC G T T C C C AAGTCGGATG

10 TGGAAATGGA AGCCCAGAAA GAT G CAT C C A TCCACCTAAG CT GTAATAGG 15 AC C AT C CAT C C AC T GAAACA TTTTAACAGT GAT GT C AT GG C C AG C G AC AA 20 TGGCGGTGCG GTCAAGCTTC CACAGCTGTG CAAGTTTTGC GATGTGAGAC 25 TGTCCACTTG C GACAAC C AG AAGTCCTGCA T GAG C AAC T G CAGCAT CAC G 30 G C C AT C T G T G AGAAGCCGCA TGAAGTCTGC GTGGCCGTGT GGAGGAAGAA 35 CGACAAGAAC ATTACTCTGG AG AC G GT T T G C C AC G AC C C C AAGCTCACCT 40 AC C AC G G C T T CACTCTGGAA GAT G C C G C T T C T C C C A AG T G T GT CAT GAAG 45 GAAAAGAAAA G G G C G G G C G A GACTTTCTTC AT G T G T G C C T GT AAC AT G GA 50 AGAGTGCAAC GATTACATCA T C T T T T C G GA AGAAT AC AC C AC CAGCAGT C 55 C C G AC AC C G G T G G G G G T GAG CCCAGAGT GC C CAT AAC AC A GAACCCCTGT 60 CCTC CACT CA AAGAGT GT CC C C CAT G C G C A GCTCCAGACC T C T T G G G T G G 65 AC CAT C C G T C T T CAT C T T C C CT C CAAAGAT CAAGGAT GTA CT CAT GAT C T 70 CCCTGAGCCC CATGGTCACA TGTGTGGTGG TGGATGTGAG C GAG GAT G AC 751 CCAGACGTCC AGATCAGCTG GTTTGTGAAG AACGTGGAAG TACACACAGC 801 TCAGACACAA AC C CAT AGAG AG GAT T AC AA CAGTACTCTC CGGGTGGTCA 851 GTGCCCTCCC CAT C C AG C AC CAGGACTGGA TGAGTGGCAA GGAGTTCAAA

901 TGCAAGGTCA ACAACAGAGC CCTCCCATCC CC CATC GAGA AAAC CAT C T C

951 AAAACCCAGA GGGCCAGTAA GAGCTCCACA GGTATATGTC T T G C C T C CAC

1001 CAGCAGAAGA GATGACTAAG AAAGAGTT CA GTCTGACCTG CATGATCACA 1051 GGCTTCTTAC CTGCCGAAAT TGCTGTGGAC TGGACCAGCA AT G G G C GT AC 1101 AGAGCAAAAC TACAAGAACA CC GCAACAGT CCTGGACTCT GATGGTTCTT 1151 ACTTCATGTA CAGCAAGCTC AGAGTACAAA AGAGCACTTG GGAAAGAGGA 1201 AGTCTTTTCG CCTGCTCAGT GGTCCACGAG GGTCTGCACA AT C AC C T T AC 1251 GACTAAGACC AT C T C C C G G T CTCTGGGTAA AT G A ( SEQ I D NO : 37 )

Example 2. Differential ligand inhibition by receptor fusion protein variants in cell- based assay

Affinities of TORbI, TGFp2 and TORb3 for hTpRII (G4S)2-hFc; Ii ' TbMI (G4S)3- hFc; hTpRII (G4S)4-hFc; hTpRff-hFc: and IiTbKII extended hinge-hFc proteins were evaluated in vitro with a Biacore™ instrument, and the results are summarized in Figures 4A and 4B. Each of the fusion proteins was capable of binding TORbI and TGFp3 with high affinity, but the constructs having linker lengths longer than or equal to (G4S)4 (SEQ ID NO: 59) w'ere surprisingly capable of binding to both TGFpl and TGFp3 with higher affinity than constructs having linker lengths shorter than (G4S)4 (SEQ ID NO: 59). Binding between TGFp2 and any of the constructs was low or transient . Deglycosylation of the constructs did not change binding.

A reporter gene assay in A549 cells was used to determine the ability of hXpRH-hFc variants to inhibit activity of TGFpl, TORb2 and TGFp3. This assay is based on a human lung carcinoma cell line transfected with a pGL3(CAGA)12 reporter plasmid (Dennier et al, 1998, EMBO 17: 3091-3100) as well as a Renilia reporter plasmid (pRLCMV) to control for transfection efficiency. The CAGA motif is present in the promoters of TGFp-responsive genes (for example, PAI-1), so this vector is of general use for factors signaling through SMAD2 and SMAD3. On the first day of the assay, A549 cells (ATCC ® : CCL-185™) were distributed in

48-well plates. On the second day, a solution containing pGL3(CAGA)12, pRLCMV, X- tremeGENE 9 (Roche Applied Science), and OptiMEM (Invitrogen) was preincubated, then added to Eagle’s minimum essential medium (EMEM, ATCC ® ) supplemented with 0.1 % BSA, which was applied to the plated cells for incubation overnight at37°C , 5% CO?.. On the third day, medium was removed, and ceils were incubated overnight at 37°C, 5% CO2 with a mixture of ligands and inhibitors prepared as described below.

Serial dilutions of test articles were made in a 48-well plate in assay buffer (EMEM + 0.1 % BSA). An equal volume of assay buffer containing the test ligand was added to obtain a final ligand concentration equal to the EC50 determined previously. Human TΌRbI, human 101 02. and human TORb3 were obtained from PeproTech. Test solutions were incubated at 37°C for 30 minutes, then a portion of the mixture was added to all wells. After incubation with test solutions overnight, cells were rinsed with phosphate-buffered saline, then lysed with passive lysis buffer (Promega E1941) and stored overnight at -70°C. On the fourth and final day, plates were warmed to room temperature with gentle shaking. Cell lysates were transferred in duplicate to a chemiluminescence plate (96-well) and analyzed in a luminometer with reagents from a Duai-Luciferase Reporter Assay system (Promega E1980) to determine normalized luciferase activity.

As illustrated in Figures 5A-5F, the IiTbKII (G4S)2-hFc; hTftRII (G4S)3-hFc; hTpRH (G4S)4-hFc; hTpRH (G4S)5-hFc; hTpRII (G4S)6-hFc; hTpRII-hFc; and hTpRII extended hinge-hFc proteins all were capable of inhibiting both TGFpl and TGFp3. Interestingly, while there was a correlation between improved TGFpi and TORb3 inhibition and linker length for the hTpRII (G4S)2-hFc; hTpRII (G4S)3-hFc and hXpRil (G4S)4-hFc constructs (Figure 5E), this improvement trend appeared to have piateaued for hXpRil (G4S)5-hFc and hTPRII (G4S)6~iiFe constructs (Figure 5F).

Example 3. Clinical Testing and Evaluation in SSc-ILD

TGF-p Receptor Type II (TpRIl) polypeptides and/or pharmaceutical compositions comprising TGF-p Receptor Type II (TbKII) polypeptides of the present disclosure may be tested clinically for success in treatment of the disease, tolerability, safety, and efficacy, among other possible endpoints.

Study Design I Inclusion criteria

A study comprising healthy volunteers, male or female, aged 18 and older is conducted.

Study Design

In this study, single ascending doses (SAD) of one or more pharmaceutical compositions comprising TpRIl polypeptides of the present disclosure are administered to approximately 40 subjects. The subjects are divided into five cohorts of eight subjects each. Within each cohort, six subjects receive active drug (e.g. compositions comprising one or more TpRIl polypeptides of the present disclosure), and two subjects receive a placebo. Cohorts are distinguished by dose level, with a six week interval between each cohort, assessing for safety between each cohort. There is an eight week interval for cohort 3, giving time to review and assess pharmacokinetics (PK) data. At the 4 th cohort level, a decision is made to determine if multiple ascending doses (MAD) should be considered.

Study Endpoints

Primary^ endpoints comprise safety and tolerability evaluations of the administered drug.

Secondary endpoints comprise pharmacokinetics (PK) parameters and/or serum TGF- b measurement.

Analysis

Pharmacokinetics (PK) is the determination of tire fate of a substance in a liv ing organism. Specifically, a substance is fol lowed through liberation (release of a drag from a pharmaceutical formulation), absorption (process of a substance entering blood circulation), distribution (dispersion/dissemination of substances through fluids and/or tissues in the body), metabolism (irreversible transformation of parent compounds into daughter metabolites), and excretion (removal of substances from a body). Common pharmacokinetic metrics are shown in Table 1 below. All or some of these metrics, among others, are determined and analyzed as a secondary' endpoint of the study.

Table 1. Common Pharmacokinetic Metrics

Characteristic Description Symbol Unit Formula

Dose Amount of drug D mol Study design parameter administered

Dosing interval Time between drug t s Study design parameter dose administrations

Cmax Peak plasma C max M Direct measurement concentration of a drag after administration tmax Time to reach Cmax tmax s Direct measurement

Cmin Lowest concentration Cmin, ss M Direct measurement that a drug reaches before the next dose is administered

Volume of Apparent volume in Vd m 3 D distribution which a drug is Co distributed

Concentration Amount of drag in a Co, Css M D given volume of Vcl plasma

Elimination Time required for t½ s ln(2) half-life concentration of drug K e to reach half of its original value Elimination Rate at which a drag is rate constant removed from the body Infusion rate Rate of infusion required to balance elimination

Area under the Integral of curve concentration-time curve

Clearance Volume of plasma cleared of drag per unit of time

Bioavailability Systemical!y available fraction of a drag

Fluctuation Peak trough fluctuation within one dosing interval at steady state

Bioanalyticai methods known to one of skill in the art can be used to determine and map a concentration vs. time profile. Concentrations of a substance are typically measured in a patient's plasma sample. For complicated biomatrices such as plasma, for example, analysis may be done using mass spectrometry.

Other patient characteristics may also be monitored to determine any correlation to drag performance. Patient demographics, body weight, excretory and metabolic system functions, presence/absence of other medications, among other aspects, may be determined and monitored throughout a study.

Study Design 2 Inclusion criteria

A study comprising male or female patients who have been diagnosed with systemic sclerosis, and are aged 18 and older, is conducted. Patient forced vital capacity (FVC) should be between about 40% and about 80% at baseline, and the patient should have at least 10% fibrosis present on a high-resolution computed topography (HRCT) scan of the lungs to be eligible for the study.

Exclusion Criteria Patients with systemic sclerosis and pulmonary arterial hypertension (PAH) are excluded from the study.

Study Design

In this study, one or more pharmaceutical compositions comprising TbMI polypeptides of the present disclosure are administered to approximately 132 subjects, accounting for an approximate 15% drop out rate. The subjects are divided into three cohorts. Cohort 1 comprises 8 patients in a 3: 1 ratio - 6 patients receive active drug (e.g. compositions comprising one or more TbKII polypeptides of the present disclosure), and two patients receive a placebo. Cohort 2 comprises 62 patients in a 1 : 1 ratio - 31 patients receive active drug (e.g. compositions comprising one or more TbKII polypeptides of the present disclosure) and 31 patients receive a placebo. A total of 12 patients with 3 complete cycles will then escalate to the next dose level. In this group, 6 patients receive active drug (e.g. compositions comprising one or more TbKII polypeptides of the present disclosure), and 6 patients receive a placebo. Cohort 3 comprises 62 patients in a 1 : 1 ratio - 31 patients receive active drug (e.g. compositions comprising one or more TbKII polypeptides of the present disclosure) and 31 patients receive a placebo.

Study Endpoints

Primary endpoints comprise safety and tolerability evaluations of the administered drug. Further, a primary endpoint is a predicted percentage of FVC at 52 weeks post- treatment (e.g. annual rate of decline in FVC): a treatment effect of 4.2%, SD= 6%, over placebo.

Secondary endpoints comprise pharmacokinetics (PK) parameters as described above, as well as Modified Rodnan Skin Score (mRSS), fibrosis biomarkers (e.g. serum levels of KL-6, SP-D, CCL18, etc.), time to event of clinical worsening or death, and quality of life.

Analysis

Spirometry, or the measuring of breath, is a major pulmonary function test that can determine the volume and/or speed (flow) of air that is inhaled and exhaled. A spirometer is used to measure forced vital capacity (FVC) (measured in liters) m a forced expiratory volume (FEV) test. In the test, a patient takes a deep breath, and exhales into the sensor as hard and as long as possible (at least 6 seconds). Inhalation can also be tested. An FEV test is typically repeated three times to ensure accuracy. Normal ranges for FVC are between 80% and 100% predicted. Other typical measurements are FEV1, wherein the FV C is measured within the first second of forced exhalation, and also forced expiratory flow (FEF), which is the flow of air coming out of the lung during the middle portion of forced expiration.

Modified Rodnan Skin Score (mRSS) is a measure of skin thickness and is commonly used to analyze systemic sclerosis outcomes.

Study Design 3 inclusion criteria

In this study, one or more pharmaceutical compositions comprising TbKII polypeptides of the present disclosure are administered to approximately 600 subjects, with approximately 300 subjects per arm (one arm is active drug, and the second ami is a placebo). Treatment will last for approximately 48 weeks.

Study Endpoints

Primary endpoints comprise an improved percentage of FVC at 52 weeks post- treatment (e.g. annual rate of decline in FVC).

Secondary endpoints comprise Modified Rodnan Skin Score (mRSS), fibrosis biomarkers (e.g. serum levels of KL-6, SP-D, CCL18, etc.), and time to event of clinical worsening or death .

Analysis FVC, mRSS, and/or biomarker levels can be analyzed using methods as described herein.

Example 4. Dose Schedule Testing of mTpRII-mFc in a mouse model of SSc-ILD

Anti-fibrotie effects of mTpRII-niFc, a murine version of a fusion protein that selectively binds to TGF-bI and TGF-p3, were assessed using different dosing regimens in a mouse bleomycin model of SSc-ILD. Lung fibrosis in this model was induced by bleomycin infused through subcutaneously implanted mini-pumps (Liang, M. etal, Laboratory' Investigation, 2015, 95, pp. 342-350).

Male 6-8-week old C57BL/6 mice were obtained from Jackson Laboratories and divided into study groups (Table 2), Mice were administered saline (Group 1, Control) or bleomycin (Groups 2-9) by osmotic mini-pumps implanted subcutaneously (s.c.) under the back skin. Bleomycin was dosed at 100 U/kg and infused at 0.25 uL/hr over a period of 28 days. Control mice were given subcutaneous injections of PBS (lO uL/g; “Control”) twice per week over a 28 day period. Bleomycin treated mice were given subcutaneous injections of either PBS (10 uL/g; “Vehicle”), Pan TORb1-3 antibody (10 mg/kg; “Pan TGFpi-3 Ab”), or a specified dose and frequency of mTpRJI-mFc. Both the Vehicle and Pan TORb1-3 Ab 5 groups were administered PBS and Pan TORb1-3 Ab, respectively, twice per week over a 28 day period

Mice given mTpRII-mFc were subcutaneously injected with either 3 mg/kg mTpRII- mFc, 10 mg/kg mTpRII-niFc, or 100 mg/kg hiTbKP-ihRo on two different dosing schedules. One subset w?as dosed with mTpRII-mFc once every 7 two weeks over a 28 day period and a 0 second subset was dosed with hiTbKII-hiRo twice per week over a 28 day period. Tire two different dosing schedules are denoted in Figure 6 by white bars (e.g., twice per week, on day 7, day 10, day 14, day 17, day 21, day 24, and day 28) or grey bars (e.g., once every two weeks, on day 7 and day 21). At the completion of the study, mice were euthanized for blood and lung tissue collection. 5

Table 2: Mouse SSc-ILD Study Design

The extent of lung fibrosis was assessed using picrosirius red collagen stain (PSR) with quantitative image analysis using HALO software (Indica Lab Albuquerque, NM), Bleomycin vehicle -treated mice (“Vehicle”) showed an increasing trend in picrosirius red staining relative to saline-infused controls (“Control”). This increasing trend was significantly inhibited by mTpRll-mFc at all doses and all dosing frequencies tested. Bleomycin infusion resulted in an increase in the percent of lung tissue area staining with picrosirius red (e.g., more collagen was present). Administration of inTflRH-mFc prevented an increase in picrosirius red staining in bleomycin treated mice at all doses of inTpRII-mFc tested (e.g., 3 mg/kg, 10 mg/kg, and 100 mg/kg of mTfsRil-mbc) at all frequencies tested

(Figure 6, white bars and grey bars) in comparison to the Vehicle group. Dosing of mTpRII- mFc on a schedule of day 7, day 10, day 14, day 17, day 21, day 24, and day 28 (Figure 6, white bars) reduced picrosirius red staining to a similar extent as dosing of mTpRll-mFc on a dosing schedule of only day 7 and day 21 (Figure 6, grey bars). This data demonstrates that mTpRJI-mFc has anti -fibro tic effects in a bleomycin mouse model of SSc-ILD with using multiple dosing schedules. This data suggests that other TpRll antagonists, particularly those that inhibit at least TGFf!I and TGFp3, may be useful m the treatment of SSc and specific pulmonary complications associated with SSc-ILD. INCORPORATION BY REFERENCE

All publications and patents mentioned herein are hereby incorporated by reference in their entirety as if each individual publication or patent was specifically and individually indicated to be incorporated by reference, While specific embodiments of the subject matter have been discussed, the above specification is illustrative and not restrictive. Many variations will become apparent to those skilled in the art upon review 7 of this specification and the claim s below. The full scope of the invention should be determined by reference to the claims, along w 7 ith their full scope of equivalents, and the specification, along with such variations.