Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHOD OF TREATING THE OSTEOARTHRITIC JOINT
Document Type and Number:
WIPO Patent Application WO/2020/257173
Kind Code:
A1
Abstract:
Disclosed herein are methods of treating a patient diagnosed with osteoarthritis in a joint by administering therapeutically effective doses of compositions that decrease synovitis or pain in the joint.

Inventors:
DANANBERG JAMIE (US)
VISICH JENNIFER (US)
HSU BEN (US)
Application Number:
PCT/US2020/037896
Publication Date:
December 24, 2020
Filing Date:
June 16, 2020
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNITY BIOTECHNOLOGY INC (US)
International Classes:
A61K31/496; A61P19/02
Domestic Patent References:
WO2015116740A12015-08-06
WO2017008060A12017-01-12
Foreign References:
KR20120118596A2012-10-29
Other References:
PARK, JIN-SIL ET AL.: "p53 Controls Autoimmune Arthritis via STAT-Mediated Regulation of the Thl7 Cell /Treg Cell Balance in Mice", ARTHRITIS & RHEUMATISM, vol. 65, no. 4, 2013, pages 949 - 959, XP055396804, DOI: 10.1002/art.37841
ZHANG, L ET AL.: "MDM2 promotes rheumatoid arthritis via activation of MAPK and NF-KB", INTERNATIONAL IMMUNOPHARMACOLOGY, vol. 30, 2016, pages 69 - 73, XP029369213, DOI: 10.1016/j.intimp.2015.11.030
ZHANG TING, LI HUIHUA, SHI JUAN, LI SHA, LI MUYUAN, ZHANG LEI, ZHENG LETING, ZHENG DEXIAN, TANG FULIN, ZHANG XUAN, ZHANG FENGCHUN,: "p53 predominantly regulates 1L-6 production and suppresses synovial inflammation in fibroblast-like synoviocytes and adjuvant-induced arthritis", ARTHRITIS RESEARCH & THERAPY, vol. 18, no. 1, 24 November 2016 (2016-11-24), pages 271, XP055775737
Attorney, Agent or Firm:
ROHATGI, Kyanne (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A method of treating a patient diagnosed with osteoarthritis in a j oint comprising administering to the joint a therapeutically effective dose of a compound of Formula I, or a pharmaceutically acceptable salt thereof:

(Formula I)

wherein the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 8 mL of vehicle and the therapeutically effective dose of the compound of Formula I results in a decrease in synovitis in the joint.

2. The method of claim 1, wherein the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 8 mL of vehicle.

3. The method of any one of claims 1-2, wherein the therapeutically effective dose of the compound of Formula I is 4 mg in 8 mL of vehicle.

4. The method of any one of claims 1-3, wherein the compound of Formula I is administered intra-articularly to the joint.

5. The method of any one of claims 1-4, wherein the joint is a fern oro-tibial joint.

6. The method of any one of claims 1-4, wherein the joint is a hip.

7. The method of any one of claims 1-4, wherein the joint is spine, shoulder, ankle, wrist, or finger joint.

8. The method of any one of claims 1-7, wherein the therapeutically effective dose of the compound of Formula I is administered as a single dose.

9. The method of any one of claims 1-8, wherein the therapeutically effective dose of the compound of Formula I is administered to the joint every week for two months, or every 2 weeks for two months, or every month for two months.

10. The method of any one of claims 1-8, wherein the therapeutically effective dose of the compound of Formula I is administered to the joint every 3 months for 6 months, every four months for 8 months, every five months for 10 months, every 6 months for 1 year.

11. The method of any one of claims 1-8, wherein the therapeutically effective dose of the compound of Formula I is not administered to the joint for at least 6 months, for at least 1 year, for at least 18 months, for at least 2 years, for at least 3 years, for at least 4 years, for at least 5 years.

12. The method of any one of claims 1-11, wherein the patient is > 40 years of age.

13. The method of any one of claims 1-11, wherein the patient is < 85 years of age.

14. The method of any one of claims 1-11, wherein the patient is > 40 and < 85 years of age.

15. The method of any one of claims 1-14, wherein a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I.

16. The method of claim 15, wherein the baseline measurement is a baseline pain level.

17. The method of claim 16, wherein the baseline pain level is assessed using a pain assessment protocol.

18. The method of claim 17, wherein the pain assessment protocol is an 11-point Numeric Rating Scale (NRS) or a Western Ontario and McMaster Universities (WOMAC)

Index pain sub scale.

19. The method of any one of claims 15-18, wherein the baseline measurement is a baseline functionality level.

20. The method of claim 19, wherein the baseline functionality level is assessed by WOMAC Index functional sub scale.

21. The method of any one of claims 15-20, wherein the baseline measurement is a baseline radiological categorization.

22. The method of claim 21, wherein the baseline radiological categorization is a Kellgren-Lawrence grade.

23. The method of claim 22, wherein the joint is the fern oro-tibial joint.

24. The method of any one of claims 15-23, wherein the baseline measurement is a baseline synovitis level.

25. The method of claim 24, wherein the baseline synovitis level of the patient is assessed using an 11 point MRI score.

26. The method of any one of claims 1-25, wherein the decrease in synovitis in the joint is measured 12 weeks after the compound of Formula I is administered to the patient.

27. The method of claim 26, wherein the decrease in synovitis is by a decrease of at least 1 point as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient.

28. The method of claim 27, wherein the decrease in synovitis is by a decrease of at least 4 points, at least 5 points, at least 8 points, at least 9 points, at least 10 points, wherein the decrease is between 1-4 points, between 4-8 points, between 8-10 points, between 10-13 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient.

29. The method of claim 28, wherein the decrease in synovitis is by a decrease of between 1-4 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient.

30. The method of any one of claims 15-29, wherein the baseline measurement is a baseline synovial fluid level.

31. The method of claim 30, wherein the baseline synovial fluid level of the patient is measured by physical examination, or by ultrasound, or by MRI.

32. The method of claims 30 or 31, wherein the therapeutically effective dose of the compound of Formula I results in a decrease in synovial fluid in the joint as measured by physical examination, ultrasound, or MRI, as compared to the baseline synovial fluid level.

33. The method of claim 32, wherein the decrease in synovial fluid in the joint is measured 24 weeks, 12 weeks, or 8 weeks, or 4 weeks, or 2 weeks after the compound of Formula I is administered to the patient.

34. The method of any one of claims 16-33, wherein the therapeutically effective dose of the compound of Formula I further results in a decrease in pain in the joint as compared to the baseline pain level.

35. The method of claim 34, wherein the decrease in pain in the joint is measured 12 weeks or measured 24 weeks after the compound of Formula I is administered to the patient.

36. The method of claim 35, wherein the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the 11-point NRS.

37. The method of claim 36, wherein the decrease in pain in the joint is a change from the baseline pain level of between 2 to 4 points as assessed by the 11 -point NRS.

38. The method of claim 35, wherein the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the WOMAC Index pain sub scale.

39. The method of claim 38, wherein the decrease in pain in the joint is a change from the baseline pain level of 0.5 point, of 1 point, or of 2 points as assessed by the WOMAC Index pain sub scale.

40. The method of claim 35, wherein the decrease in pain in the joint is a change from the baseline pain level of about 50%, 40%, 30%, between 30-40%, or between 40-50% as measured by the 11 -point NRS or WOMAC Index pain subscale.

41. The method of any one of claims 34-40, wherein the decrease in pain relative to the baseline pain level lasts 3 months or more, 4 months or more, lasts 5 months or more, lasts 6 months or more, lasts 7 months or more, lasts 8 months or more, lasts 9 months or more, lasts 10 months or more, lasts 11 months or more, lasts 1 year or more, lasts 2 years or more, lasts 3 years or more, lasts 4 years or more, or lasts 5 years or more.

42. The method of any one of claims 19-41, wherein the therapeutically effective dose of the compound of Formula I results in an increase functionality of the joint as measured by a decrease in the WOMAC Index functionality subscale score relative to the baseline functionality level as measured by the WOMAC Index functionality subscale.

43. The method of claim 42, wherein the increase in functionality of the joint is measured 12 weeks or 24 weeks after the compound of Formula I is administered to the patient.

44. The method of claim 42, wherein the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points.

45. The method of claim 44, wherein the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of 0.5 point, of 1 point, of 2 points.

46. The method of any one of claims 15-45, wherein the baseline measurement is a baseline biomarker level.

47. The method of claim 46, wherein the baseline biomarker levels are measured from the patient’s synovial fluid.

48. The method of claims 46 or 47, wherein the therapeutically effective dose of the compound of Formula I results in modulation of a biomarker level relative to the baseline biomarker level.

49. The method of claim 48, wherein the modulation of the biomarker levels relative to the baseline biomarker levels is a decrease.

50. The method of any one of claims 46-49, wherein the biomarker comprises MMP-10, MMP-12, MMP-13, IL-6, IL-10, CCL20, A2M, or VEGF-C.

51. The method of claims 49 or 50, wherein the modulation of the biomarker relative to the baseline biomarker level is measured 4 weeks, measured 8 weeks, measured 12 weeks, or measured 24 weeks after the compound of Formula I is administered to the patient.

52. The method of any one of claims 1-51, wherein the therapeutically effective dose of the compound of Formula I results in the patient’s global impression of change (PGIC) to become much improved or very much improved relative to the patient’s baseline impression.

53. The method of any one of claims 46-52, wherein the biomarker comprises a cytokine or chemokine such as CCL20, CCL19, IL-10, or IL-6.

54. The method of any one of claims 46-53, wherein the biomarker comprises IL-6.

55. The method of any one of claims 46-54, wherein the biomarker comprises a growth factor or adhesion molecule such as ICAM-1 or VEGF-C.

56. The method of any one of claims 46-55, wherein the biomarker comprises a protease or protease inhibitor such as MMP-3, MMP-10, MMP-12, MMP-13, or A2M.

57. The method of any one of claims 1-56, wherein the therapeutically effective dose of the compound of Formula I is at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, at least 10 mg, or more.

58. The method of any one of claims 1-57, wherein the therapeutically effective dose is in at least 8.5 ml, at least 9 mL, at least 10 mL, at least 11 mL, at least 12 mL, or more, of vehicle.

59. The method of any one of claims 1-58, wherein the therapeutically effective dose results in an improved joint architecture measurement.

60. The method of claim 59, wherein a baseline joint architecture measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I, and wherein the improved joint architecture measurement comprises an improvement compared to the baseline joint architecture measurement.

61. The method of claim 59 or 60, wherein the joint architecture measurement is measured using MRI, ultrasound, computed tomography, or X-ray device.

62. The method of claim 60 or 61, wherein the improvement comprises a reversal, slowing, or stopping of an adverse effect on the joint architecture measurement relative to the baseline joint architecture measurement.

63. The method of any one of claims 59-62, wherein the joint architecture measurement comprises a cartilage thickness measurement.

64. The method of any one of claims 60-63, wherein the baseline joint architecture measurement comprises a baseline cartilage thickness measurement.

65. The method of claim 64, wherein the cartilage thickness measurement and the baseline cartilage thickness measurement are measured using X-ray or MRI.

66. The method of any one of claims 63-65, wherein the cartilage thickness measurement comprises a joint space width (JSW) measurement.

67. The method of any one of claims 64-66, wherein the baseline cartilage thickness measurement comprises a baseline JSW measurement.

68. The method of claim 67, wherein the improvement comprises an increase in the JSW measurement relative to the baseline JSW measurement of at least 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0 mm, or more.

69. The method of any one of claims 63-65, wherein the cartilage thickness measurement comprises a joint space narrowing (JSN) measurement.

70. The method of claim 63 or 64, wherein the baseline cartilage thickness measurement comprises a baseline JSN measurement.

71. The method of claim 70, wherein the improvement comprises reversing, stopping or decreasing the JSN measurement relative to the baseline JSN measurement.

72. The method of any one of claims 59-71, wherein the joint architecture measurement comprises a subchondral bone surface area measurement.

73. The method of any one of claims 60-72, wherein the baseline joint architecture measurement comprises a baseline subchondral bone surface area measurement.

74. The method of claim 73, wherein the improvement comprises a 50 Hounsfield Unit (HU), 100 HU, 150 HU, 200 HU, 250 HU, 300 HU, 350 HU, 400 HU, 450 HU, 500 HU, or more, increase in the subchondral bone surface area measurement relative to the baseline subchondral bone surface area measurement.

75. The method of claim 73 or 74, wherein the subchondral bone surface area

measurement and the baseline subchondral bone surface area measurement are measured using X-ray or MRI.

76. The method of any one of claims 59-75, wherein the joint architecture measurement comprises a Kellgren-Lawrence (KL) grade.

77. The method of any one of claims 60-76, wherein the baseline joint architecture measurement comprises a baseline KL grade.

78. The method of claim 77, wherein the improvement comprises a decrease in the KL grade relative to the baseline KL grade by one KL grade or more.

79. The method of any one of claims 59-78, wherein the joint architecture measurement comprises a progression through a KL grading scale.

80. The method of any one of claims 60-79, wherein the baseline joint architecture measurement comprises a baseline progression through the KL grading scale.

81. The method of claim 80, wherein the improvement comprises a decrease in a rate of the progression through the KL grading scale relative to a rate of the baseline progression through the KL grading scale grade, or comprises preventing the progression through the KL grading scale, as assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, or 5 years or more after administration of the therapeutically effective dose of the compound of Formula I.

82. A method of treating a patient diagnosed with osteoarthritis in a joint comprising administering to the joint a therapeutically effective dose of a compound of Formula I, or a pharmaceutically acceptable salt thereof:

(Formula I)

wherein the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 8 mL of vehicle and the therapeutically effective dose of the compound of Formula I results in a decrease in pain in the joint.

83. The method of claim 82, wherein the therapeutically effective dose of the compound of Formula I is at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, at least 10 mg, or more.

84. The method of any one of claims 82 or 83, wherein the therapeutically effective dose is in at least 8.5 ml, at least 9 mL, at least 10 mL, at least 11 mL, at least 12 mL, or more, of vehicle.

85. The method of any one of claims 82-84, wherein the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 8 mL of vehicle.

86. The method of any one of claims 82-85, wherein the therapeutically effective dose of the compound of Formula I is 4 mg in 8 mL of vehicle.

87. The method of any one of claims 82-86, wherein the compound of Formula I is administered intra-articularly to the joint.

88. The method of any one of claims 82-87, wherein the joint is a fern oro-tibial joint.

89. The method of any one of claims 82-87, wherein the joint is a hip.

90. The method of any one of claims 82-87, wherein the joint is spine, shoulder, ankle, wrist, or finger joint.

91. The method of any one of claims 82-90, wherein the therapeutically effective dose of the compound of Formula I is administered as a single dose.

92. The method of any one of claims 82-91, wherein the therapeutically effective dose of the compound of Formula I is administered to the joint every week for two months, or every 2 weeks for two months, or every month for two months.

93. The method of any one of claims 82-91, wherein the therapeutically effective dose of the compound of Formula I is administered to the joint every 3 months for 6 months, every four months for 8 months, every five months for 10 months, every 6 months for 1 year.

94. The method of any one of claims 82-91, wherein the therapeutically effective dose of the compound of Formula I is not administered to the joint for at least 6 months, for at least 1 year, for at least 18 months, for at least 2 years, for at least 3 years, for at least 4 years, for at least 5 years.

95. The method of any one of claims 82-94, wherein the patient is > 40 years of age.

96. The method of any one of claims 82-94, wherein the patient is < 85 years of age.

97. The method of any one of claims 82-94, wherein the patient is > 40 and < 85 years of age.

98. The method of any one of claims 82-97, wherein a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I.

99. The method of claim 98, wherein the baseline measurement is a baseline pain level.

100. The method of claim 99, wherein the joint pain and/or baseline pain level is assessed using a pain assessment protocol.

101. The method of claim 100, wherein the pain assessment protocol is an 11-point Numeric Rating Scale (NRS) or a Western Ontario and McMaster Universities (WOMAC)

Index pain sub scale.

102. The method of any one of claims 98-101, wherein the baseline measurement is a baseline functionality level.

103. The method of claim 102, wherein the baseline functionality level is assessed by WOMAC Index functional sub scale.

104. The method of any one of claims 98-103, wherein the baseline measurement is a baseline radiological categorization.

105. The method of claim 104, wherein the baseline radiological categorization is a Kellgren-Lawrence grade.

106. The method of claim 105, wherein the joint is the fern oro-tibial joint.

107. The method of any one of claims 98-106, wherein the baseline measurement is a baseline synovial fluid level.

108. The method of claim 107, wherein the baseline synovial fluid level of the patient is measured by physical examination, or by ultrasound, or by MRI.

109. The method of claims 107 or 108, wherein the therapeutically effective dose of the compound of Formula I results in a decrease in synovial fluid in the joint as measured by physical examination, ultrasound, or MRI, as compared to the baseline synovial fluid level.

110. The method of claim 109, wherein the decrease in synovial fluid in the joint is measured 24 weeks, 12 weeks, or 8 weeks, or 4 weeks, or 2 weeks after the compound of Formula I is administered to the patient.

111. The method of any one of claims 82-111, wherein the decrease in pain in the joint is measured 12 weeks, or 24 weeks after the compound of Formula I is administered to the patient.

112. The method of any one of claims 99-111, wherein the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the 11-point NRS.

113. The method of any one of claims 99-112, wherein the decrease in pain in the joint is a change from the baseline pain level of between 2 to 4 points as assessed by the 11 -point NRS.

114. The method of any one of claims 99-111, wherein the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the WOMAC Index pain sub scale.

115. The method of any one of claims 99-114, wherein the decrease in pain in the joint is a change from the baseline pain level of 0.5 point, of 1 point, or of 2 points as assessed by the WOMAC Index pain subscale.

116. The method of claim 111, wherein the decrease in pain in the joint is a change from the baseline pain level of about 50%, 40%, 30%, between 30-40%, or between 40-50% as measured by the 11 -point NRS or WOMAC Index pain subscale.

117. The method of any one of claims 99-116, wherein the decrease in pain relative to the baseline pain level lasts 3 months or more, 4 months or more, lasts 5 months or more, lasts 6 months or more, lasts 7 months or more, lasts 8 months or more, lasts 9 months or more, lasts 10 months or more, lasts 11 months or more, lasts 1 year or more, lasts 2 years or more, lasts 3 years or more, lasts 4 years or more, or lasts 5 years or more.

118. The method of any one of claims 82-117, wherein the therapeutically effective dose of the compound of Formula I results in an increase functionality of the joint as measured by a decrease in the WOMAC Index functionality subscale score relative to the baseline functionality level as measured by the WOMAC Index functionality subscale.

119. The method of claim 118, wherein the increase in functionality of the joint is measured 12 weeks or 24 weeks after the compound of Formula I is administered to the patient.

120. The method of claim 118, wherein the decrease in the WOMAC Index

functionality subscale score is a change from the baseline functionality level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points.

121. The method of claim 118, wherein the decrease in the WOMAC Index

functionality subscale score is a change from the baseline functionality level of 0.5 point, of 1 point, of 2 points.

122. The method of any one of claims 98-122, wherein the baseline measurement is a baseline biomarker level.

123. The method of claim 122, wherein the baseline biomarker levels are measured from the patient’s synovial fluid.

124. The method of claims 122 or 123, wherein the therapeutically effective dose of the compound of Formula I results in modulation of a biomarker level relative to the baseline biomarker level.

125. The method of claim 124, wherein the modulation of the biomarker levels relative to the baseline biomarker levels is a decrease.

126. The method of any one of claims 122-125, wherein the biomarker comprises MMP-10, MMP-12, MMP-13, IL-6, IL-10, CCL20, A2M, or VEGF-C.

127. The method of claims 125 or 126, wherein the modulation of the biomarker relative to the baseline biomarker level is measured 4 weeks, measured 8 weeks, or measured 12 weeks after the compound of Formula I is administered to the patient.

128. The method of any one of claims 82-127, wherein the therapeutically effective dose of the compound of Formula I results in the patient’s global impression of change (PGIC) to become much improved or very much improved relative to the patient’s baseline impression.

129. The method of any one of claims 122-128, wherein the biomarker comprises a cytokine or chemokine such as CCL20, CCL19, IL-10, or IL-6.

130. The method of any one of claims 122-129, wherein the biomarker comprises IL-

6

131. The method of any one of claims 122-130, wherein the biomarker comprises a growth factor or adhesion molecule such as ICAM-1 or VEGF-C.

132. The method of any one of claims 122-131, wherein the biomarker comprises a protease or protease inhibitor such as MMP-3, MMP-10, MMP-12, MMP-13, or A2M.

133. The method of any one of claims 82-132, wherein the therapeutically effective dose results in an improved joint architecture measurement.

134. The method of claim 133, wherein a baseline joint architecture measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I, and wherein the improved joint architecture measurement comprises an improvement compared to the baseline joint architecture measurement.

135. The method of claim 133 or 134, wherein the joint architecture measurement is measured using MRI, ultrasound, computed tomography, or X-ray device.

136. The method of claim 134 or 135, wherein the improvement comprises a reversal, slowing, or stopping of an adverse effect on the joint architecture measurement relative to the baseline joint architecture measurement.

137. The method of any one of claims 133-136, wherein the joint architecture measurement comprises a cartilage thickness measurement.

138. The method of any one of claims 134-137, wherein the baseline joint architecture measurement comprises a baseline cartilage thickness measurement.

139. The method of claim 138, wherein the cartilage thickness measurement and the baseline cartilage thickness measurement are measured using X-ray or MRI.

140. The method of claim 138 or 139, wherein the cartilage thickness measurement comprises a joint space width (JSW) measurement.

141. The method of claim 138, wherein the baseline cartilage thickness measurement comprises a baseline JSW measurement.

142. The method of claim 141, wherein the improvement comprises an increase in the JSW measurement relative to the baseline JSW measurement of at least 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0 mm, or more.

143. The method of claim 138 or 139, wherein the cartilage thickness measurement comprises a joint space narrowing (JSN) measurement.

144. The method of claim 138, wherein the baseline cartilage thickness measurement comprises a baseline JSN measurement.

145. The method of claim 144, wherein the improvement comprises reversing, stopping or decreasing the JSN measurement relative to the baseline JSN measurement.

146. The method of any one of claims 133-145, wherein the joint architecture measurement comprises a subchondral bone surface area measurement.

147. The method of claim 146, wherein the baseline joint architecture measurement comprises a baseline subchondral bone surface area measurement.

148. The method of claim 147, wherein the improvement comprises a 50 Hounsfield Unit (HU), 100 HU, 150 HU, 200 HU, 250 HU, 300 HU, 350 HU, 400 HU, 450 HU, 500 HU, or more, increase in the subchondral bone surface area measurement relative to the baseline subchondral bone surface area measurement.

149. The method of any one of claims 146-148, wherein the subchondral bone surface area measurement and the baseline subchondral bone surface area measurement are measured using X-ray or MRI.

150. The method of claims 133-149, wherein the joint architecture measurement comprises a Kellgren-Lawrence (KL) grade.

151. The method of claim 150, wherein the baseline j oint architecture measurement comprises a baseline KL grade.

152. The method of claim 151, wherein the improvement comprises a decrease in the KL grade relative to the baseline KL grade by one KL grade or more.

153. The method of claims 133-152, wherein the joint architecture measurement comprises a progression through a KL grading scale.

154. The method of claim 153, wherein the baseline joint architecture measurement comprises a baseline progression through the KL grading scale.

155. The method of claim 154, wherein the improvement comprises a decrease in a rate of the progression through the KL grading scale relative to a rate of the baseline progression through the KL grading scale grade, or comprises preventing the progression through the KL grading scale, as assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, or 5 years or more after administration of the therapeutically effective dose of the compound of Formula I.

Description:
METHOD OF TREATING THE OSTEOARTHRITIC JOINT

CROSS-REFERENCE

[0001] This application claims the benefit of U.S. Provisional Application No. 62/862,672, filed June 17, 2019, and U.S. Provisional Application No. 62/931,719, filed November 6, 2019, which applications are incorporated herein by reference.

BACKGROUND OF THE INVENTION

[0002] Osteoarthritis is a progressive joint disease in which the cartilage between bones in the joint breaks down causing joint pain and stiffness. Osteoarthritis is characterized by breakdown of the cartilage, deterioration of tendons and ligaments, and various degrees of inflammation in the synovium (the joint lining).

SUMMARY OF THE INVENTION

[0003] Disclosed herein are methods of treating a patient diagnosed with osteoarthritis in a joint comprising administering to the joint a therapeutically effective dose of a compound of Formula I, or a pharmaceutically acceptable salt thereof:

(Formula I)

wherein the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 8 mL of vehicle and the therapeutically effective dose of the compound of Formula I results in a decrease in synovitis in the joint. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 8 mL of vehicle. In some embodiments, the compound of Formula I is administered intra-articularly to the joint. In some embodiments, the joint is a fern oro-tibial joint. In some embodiments, the joint is a hip. In some embodiments, the joint is spine, shoulder, ankle, wrist, or finger joint. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered as a single dose. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered to the joint every week for two months, or every 2 weeks for two months, or every month for two months. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered to the joint every 3 months for 6 months, every four months for 8 months, every five months for 10 months, every 6 months for 1 year. In some embodiments, the therapeutically effective dose of the compound of Formula I is not administered to the joint for at least 6 months, for at least 1 year, for at least 18 months, for at least 2 years, for at least 3 years, for at least 4 years, for at least 5 years. In some embodiments, the patient is > 40 years of age. In some embodiments, the patient is < 85 years of age. In some embodiments, the patient is > 40 and < 85 years of age. In some embodiments, a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I. In some embodiments, the baseline measurement is a baseline pain level. In some embodiments, the baseline pain level is assessed using a pain assessment protocol. In some embodiments, the pain assessment protocol is an 11- point Numeric Rating Scale (NRS) or a Western Ontario and McMaster Universities (WOMAC) Index pain sub scale. In some embodiments, the baseline measurement is a baseline functionality level. In some embodiments, the baseline functionality level is assessed by WOMAC Index functional subscale. In some embodiments, the baseline measurement is a baseline radiological categorization. In some embodiments, the baseline radiological categorization is a Kellgren- Lawrence grade. In some embodiments, the joint is the femoro-tibial joint.In some

embodiments, the baseline measurement is a baseline synovitis level. In some embodiments, the baseline synovitis level of the patient is assessed using an 1 lpoint MRI score. In some embodiments, the decrease in synovitis in the joint is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 1 point as measured by the 1 lpoint MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 4 points, at least 5 points, at least 8 points, at least 9 points, at least 10 points, wherein the decrease is between 1-4 points, between 4-8 points, between 8-10 points, between 10-13 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of between 1-4 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the baseline measurement is a baseline synovial fluid level. In some embodiments, the baseline synovial fluid level of the patient is measured by physical examination, or by ultrasound, or by MRI. In some embodiments, the therapeutically effective dose of the compound of Formula I results in a decrease in synovial fluid in the joint as measured by physical examination, ultrasound, or MRI, as compared to the baseline synovial fluid level. In some embodiments, the decrease in synovial fluid in the joint is measured 24 weeks, 12 weeks, or 8 weeks, or 4 weeks, or 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the therapeutically effective dose of the compound of Formula I further results in a decrease in pain in the joint as compared to the baseline pain level. In some embodiments, the decrease in pain in the joint is measured 12 weeks or measured 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of between 2 to 4 points as assessed by the 11- point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of 0.5 point, of 1 point, or of 2 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 50%, 40%, 30%, between 30-40%, or between 40-50% as measured by the 11-point NRS or WOMAC Index pain subscale. In some embodiments, the decrease in pain relative to the baseline pain level lasts 3 months or more, 4 months or more, lasts 5 months or more, lasts 6 months or more, lasts 7 months or more, lasts 8 months or more, lasts 9 months or more, lasts 10 months or more, lasts 11 months or more, lasts 1 year or more, lasts 2 years or more, lasts 3 years or more, lasts 4 years or more, or lasts 5 years or more. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an increase functionality of the joint as measured by a decrease in the WOMAC Index functionality subscale score relative to the baseline functionality level as measured by the WOMAC Index functionality subscale. In some embodiments, the increase in functionality of the joint is measured 12 weeks or 24 weeks after the compound of Formula I is administered to the patient. In some

embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of 0.5 point, of 1 point, of 2 points. In some embodiments, the baseline measurement is a baseline biomarker level. In some embodiments, the baseline biomarker levels are measured from the patient’s synovial fluid. In some embodiments, the therapeutically effective dose of the compound of Formula I results in modulation of a biomarker level relative to the baseline biomarker level. In some embodiments, the modulation of the biomarker levels relative to the baseline biomarker levels is a decrease. In some embodiments, the biomarker comprises MMP- 10, MMP-12, MMP-13, IL-6, IL-10, CCL20, A2M, or VEGF-C. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 4 weeks, measured 8 weeks, measured 12 weeks, or measured 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the therapeutically effective dose of the compound of Formula I results in the patient’s global impression of change (PGIC) to become much improved or very much improved relative to the patient’s baseline impression. In some embodiments, the biomarker comprises a cytokine or chemokine such as CCL20, CCL19, IL-10, or IL-6. In some embodiments, the biomarker comprises IL-6. In some embodiments, the biomarker comprises a growth factor or adhesion molecule such as ICAM-1 or VEGF-C. In some embodiments, the biomarker comprises a protease or protease inhibitor such as MMP-3, MMP-10, MMP-12, MMP-13, or A2M. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, at least 10 mg, or more. In some embodiments, the therapeutically effective dose is in at least 8.5 ml, at least 9 mL, at least 10 mL, at least 11 mL, at least 12 mL, or more, of vehicle. In some embodiments, the therapeutically effective dose results in an improved joint architecture measurement. In some embodiments, a baseline joint architecture measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I, and wherein the improved joint architecture measurement comprises an improvement compared to the baseline joint architecture measurement. In some embodiments, the joint architecture measurement is measured using MRI, ultrasound, computed tomography, or X-ray device. In some embodiments, the improvement comprises a reversal, slowing, or stopping of an adverse effect on the joint architecture measurement relative to the baseline joint architecture measurement. In some embodiments, the joint architecture measurement comprises a cartilage thickness measurement. In some embodiments, the baseline joint architecture measurement comprises a baseline cartilage thickness measurement. In some embodiments, the cartilage thickness measurement and the baseline cartilage thickness measurement are measured using X- ray or MRI. In some embodiments, the cartilage thickness measurement comprises a joint space width (JSW) measurement. In some embodiments, the baseline cartilage thickness measurement comprises a baseline JSW measurement. In some embodiments, the improvement comprises an increase in the JSW measurement relative to the baseline JSW measurement of at least 0.01,

0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0 mm, or more. In some embodiments, the cartilage thickness measurement comprises a joint space narrowing (JSN) measurement. In some embodiments, the baseline cartilage thickness measurement comprises a baseline JSN measurement. In some embodiments, the improvement comprises reversing, stopping or decreasing the JSN measurement relative to the baseline JSN measurement. In some embodiments, the joint architecture measurement comprises a subchondral bone surface area measurement. In some embodiments, the baseline joint architecture measurement comprises a baseline subchondral bone surface area measurement. In some embodiments, the improvement comprises a 50 Hounsfield Unit (HU), 100 HU, 150 HU, 200 HU, 250 HU, 300 HU, 350 HU, 400 HU, 450 HU, 500 HU, or more, increase in the subchondral bone surface area measurement relative to the baseline subchondral bone surface area measurement. In some embodiments, the subchondral bone surface area measurement and the baseline subchondral bone surface area measurement are measured using X-ray or MRI. In some embodiments, the joint architecture measurement comprises a Kellgren-Lawrence (KL) grade. In some embodiments, the baseline joint architecture measurement comprises a baseline KL grade. In some embodiments, the improvement comprises a decrease in the KL grade relative to the baseline KL grade by one KL grade or more. In some embodiments, the joint architecture measurement comprises a progression through a KL grading scale. In some embodiments, the baseline joint architecture measurement comprises a baseline progression through the KL grading scale. In some embodiments, the improvement comprises a decrease in a rate of the progression through the KL grading scale relative to a rate of the baseline progression through the KL grading scale grade, or comprises preventing the progression through the KL grading scale, as assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, or 5 years or more after administration of the therapeutically effective dose of the compound of Formula I.

[0004] Disclosed herein are methods of treating a patient diagnosed with osteoarthritis in a joint comprising administering to the joint a therapeutically effective dose of a compound of Formula I, or a pharmaceutically acceptable salt thereof: (Formula I)

wherein the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 8 mL of vehicle and the therapeutically effective dose of the compound of Formula I results in a decrease in pain in the joint. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2.5 mg, at least 3 mg, at least 3.5 mg, at least 4 mg, at least 4.5 mg, at least 5 mg, at least 6 mg, at least 7 mg, at least 8 mg, at least 9 mg, at least 10 mg, or more. In some embodiments, the therapeutically effective dose is in at least 8.5 ml, at least 9 mL, at least 10 mL, at least 11 mL, at least 12 mL, or more, of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 8 mL of vehicle. In some embodiments, the compound of Formula I is administered intra-articularly to the joint. In some embodiments, the joint is a femoro-tibial joint. In some embodiments, the joint is a hip. In some embodiments, the joint is spine, shoulder, ankle, wrist, or finger joint. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered as a single dose. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered to the joint every week for two months, or every 2 weeks for two months, or every month for two months. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered to the joint every 3 months for 6 months, every four months for 8 months, every five months for 10 months, every 6 months for 1 year. In some embodiments, the therapeutically effective dose of the compound of Formula I is not administered to the joint for at least 6 months, for at least 1 year, for at least 18 months, for at least 2 years, for at least 3 years, for at least 4 years, for at least 5 years. In some embodiments, the patient is > 40 years of age. In some embodiments, the patient is < 85 years of age. In some embodiments, the patient is > 40 and < 85 years of age. In some embodiments, a baseline measurement is obtained from the patient prior to

administering to the joint a therapeutically effective dose of the compound of Formula I. In some embodiments, the baseline measurement is a baseline pain level. In some embodiments, the joint pain and/or baseline pain level is assessed using a pain assessment protocol. In some embodiments, the pain assessment protocol is an 11 -point Numeric Rating Scale (NRS) or a Western Ontario and McMaster Universities (WOMAC) Index pain subscale. In some embodiments, the baseline measurement is a baseline functionality level. In some embodiments, the baseline functionality level is assessed by WOMAC Index functional subscale. In some embodiments, the baseline measurement is a baseline radiological categorization. In some embodiments, the baseline radiological categorization is a Kellgren-Lawrence grade. In some embodiments, the joint is the fern oro-tibial joint. In some embodiments, the baseline

measurement is a baseline synovial fluid level. In some embodiments, the baseline synovial fluid level of the patient is measured by physical examination, or by ultrasound, or by MRI. In some embodiments, the therapeutically effective dose of the compound of Formula I results in a decrease in synovial fluid in the joint as measured by physical examination, ultrasound, or MRI, as compared to the baseline synovial fluid level. In some embodiments, the decrease in synovial fluid in the joint is measured 24 weeks, 12 weeks, or 8 weeks, or 4 weeks, or 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain in the joint is measured 12 weeks, or 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of between 2 to 4 points as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of 0.5 point, of 1 point, or of 2 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 50%, 40%, 30%, between 30-40%, or between 40- 50% as measured by the 11 -point NRS or WOMAC Index pain subscale. In some embodiments, the decrease in pain relative to the baseline pain level lasts 3 months or more, 4 months or more, lasts 5 months or more, lasts 6 months or more, lasts 7 months or more, lasts 8 months or more, lasts 9 months or more, lasts 10 months or more, lasts 11 months or more, lasts 1 year or more, lasts 2 years or more, lasts 3 years or more, lasts 4 years or more, or lasts 5 years or more. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an increase functionality of the joint as measured by a decrease in the WOMAC Index functionality subscale score relative to the baseline functionality level as measured by the WOMAC Index functionality subscale. In some embodiments, the increase in functionality of the joint is measured 12 weeks or 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of 0.5 point, of 1 point, of 2 points. In some embodiments, the baseline measurement is a baseline biomarker level. In some embodiments, the baseline biomarker levels are measured from the patient’s synovial fluid. In some embodiments, the therapeutically effective dose of the compound of Formula I results in modulation of a biomarker level relative to the baseline biomarker level.

In some embodiments, the modulation of the biomarker levels relative to the baseline biomarker levels is a decrease. In some embodiments, the biomarker comprises MMP-10, MMP-12, MMP-13, IL-6, IL-10, CCL20, A2M, or VEGF-C. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 4 weeks, measured 8 weeks, or measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the therapeutically effective dose of the compound of Formula I results in the patient’s global impression of change (PGIC) to become much improved or very much improved relative to the patient’s baseline impression. In some embodiments, the biomarker comprises a cytokine or chemokine such as CCL20, CCL19, IL-10, or IL-6. In some embodiments, the biomarker comprises IL-6. In some embodiments, the biomarker comprises a growth factor or adhesion molecule such as ICAM-1 or VEGF-C. In some embodiments, the biomarker comprises a protease or protease inhibitor such as MMP-3, MMP-10, MMP-12, MMP-13, or A2M. In some embodiments, the therapeutically effective dose results in an improved joint architecture measurement. In some embodiments, a baseline joint architecture measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I, and wherein the improved joint architecture measurement comprises an improvement compared to the baseline joint architecture measurement. In some embodiments, the joint architecture measurement is measured using MRI, ultrasound, computed tomography, or X-ray device. In some embodiments, the improvement comprises a reversal, slowing, or stopping of an adverse effect on the joint architecture measurement relative to the baseline joint architecture measurement. In some embodiments, the joint architecture measurement comprises a cartilage thickness measurement. In some embodiments, the baseline joint architecture measurement comprises a baseline cartilage thickness measurement. In some embodiments, the cartilage thickness measurement and the baseline cartilage thickness measurement are measured using X-ray or MRI. In some embodiments, the cartilage thickness measurement comprises a joint space width (JSW) measurement. In some embodiments, the baseline cartilage thickness measurement comprises a baseline JSW measurement. In some embodiments, the improvement comprises an increase in the JSW measurement relative to the baseline JSW measurement of at least 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10,

0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2,

1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0 mm, or more. In some embodiments, the cartilage thickness measurement comprises a joint space narrowing (JSN) measurement. In some embodiments, the baseline cartilage thickness measurement comprises a baseline JSN measurement. In some embodiments, the improvement comprises reversing, stopping or decreasing the JSN

measurement relative to the baseline JSN measurement. In some embodiments, the joint architecture measurement comprises a subchondral bone surface area measurement. In some embodiments, the baseline joint architecture measurement comprises a baseline subchondral bone surface area measurement. In some embodiments, the improvement comprises a 50 Hounsfield Unit (HU), 100 HU, 150 HU, 200 HU, 250 HU, 300 HU, 350 HU, 400 HU, 450 HU, 500 HU, or more, increase in the subchondral bone surface area measurement relative to the baseline subchondral bone surface area measurement. In some embodiments, the subchondral bone surface area measurement and the baseline subchondral bone surface area measurement are measured using X-ray or MRI. In some embodiments, the joint architecture measurement comprises a Kellgren-Lawrence (KL) grade. In some embodiments, the baseline joint architecture measurement comprises a baseline KL grade. In some embodiments, the

improvement comprises a decrease in the KL grade relative to the baseline KL grade by one KL grade or more. In some embodiments, the joint architecture measurement comprises a progression through a KL grading scale. In some embodiments, the baseline joint architecture measurement comprises a baseline progression through the KL grading scale. In some embodiments, the improvement comprises a decrease in a rate of the progression through the KL grading scale relative to a rate of the baseline progression through the KL grading scale grade, or comprises preventing the progression through the KL grading scale, as assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, or 5 years or more after administration of the therapeutically effective dose of the compound of Formula I.

BRIEF DESCRIPTION OF THE DRAWINGS

[0005] The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative

embodiments, in which the principles of the disclosure are utilized, and the accompanying drawings of which:

[0006] FIG. 1 illustrates a Phase I clinical trial single ascending dose study.

[0007] FIG. 2 illustrates that no serious adverse events were observed in a Phase I clinical trial single ascending dose study.

[0008] FIG. 3 illustrates a dose dependent effect of the compound of Formula I in decreasing osteoarthritis pain as assessed by Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC) sub scores.

[0009] FIG. 4 illustrates a grouped dosage dependent effect of the compound of Formula I in decreasing osteoarthritis pain as assessed by WOMAC subscores.

[0010] FIG. 5 illustrates a dose dependent effect of the compound of Formula I in decreasing osteoarthritis pain as assessed by a numerical rating scale (NRS).

[0011] FIG. 6 illustrates a grouped dosage dependent effect of the compound of Formula I in decreasing osteoarthritis pain as assessed by an NRS.

[0012] FIG. 7 illustrates a dose dependent effect of the compound of Formula I in improving physical function in subjects with osteoarthritis, as assessed by WOMAC subscores.

[0013] FIG. 8 illustrates a grouped dosage dependent effect of the compound of Formula I in improving physical function in subjects with osteoarthritis, as assessed by WOMAC subscores.

[0014] FIG. 9A illustrates a beneficial effect on pain, as assessed by an NRS, of a single dose of the compound of Formula I relative to placebo.

[0015] FIG. 9B illustrates a beneficial effect on pain, as assessed by WOMAC subscores, of a single dose of the compound of Formula I relative to placebo.

[0016] FIG. 9C illustrates a beneficial effect on physical function, as assessed by WOMAC subscores, of a single dose of the compound of Formula I relative to placebo.

[0017] FIG. 10 illustrates effects of the compound of Formula I relative to placebo on synovial fluid biomarkers levels.

[0018] FIG. 11 illustrates a design for a Phase II clinical trial.

[0019] FIG. 12 illustrates patient demographics in a clinical study. [0020] FIG. 13 illustrates baseline patient characteristics in a clinical study.

[0021] FIG. 14 illustrates beneficial effects of the compound of Formula I in subjects with osteoarthritis, as assessed by Patient Global Impression of Change (PGIC) scores.

[0022] FIG. 15 illustrates effects of the compound of Formula I on pain and physical function in subjects with osteoarthritis, as assessed by WOMAC subscores.

INCORPORATION BY REFERENCE

[0023] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.

PET ATT, ED DESCRIPTION OF THE INVENTION

[0024] In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the embodiments provided may be practiced without these details. Unless the context requires otherwise, throughout the specification and claims which follow, the word“comprise” and variations thereof, such as,“comprises” and“comprising” are to be construed in an open, inclusive sense, that is, as“including, but not limited to.” As used in this specification and the appended claims, the singular forms“a,”“an,” and“the” include plural referents unless the content clearly dictates otherwise. It should also be noted that the term“or” is generally employed in its sense including“and/or” unless the content clearly dictates otherwise. Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed embodiments.

[0025] Osteoarthritis is a degenerative joint disease characterized by fibrillation of the cartilage at sites of high mechanical stress, bone sclerosis, and thickening of the synovium and the joint capsule. Fibrillation is a local surface disorganization involving splitting of the superficial layers of the cartilage. The early splitting is tangential with the cartilage surface, following the axes of the predominant collagen bundles. Collagen within the cartilage becomes disorganized, and proteoglycans are lost from the cartilage surface. In the absence of protective and lubricating effects of proteoglycans in a joint, collagen fibers become susceptible to degradation, and mechanical destruction ensues. Predisposing risk factors for developing osteoarthritis include increasing age, obesity, previous joint injury, overuse of the joint, weak thigh muscles, and genetics. It is a common cause of chronic disability in the elderly.

Symptoms of osteoarthritis include sore or stiff joints, particularly the hips, knees, and lower back, after inactivity or overuse; stiffness after resting that goes away after movement; and pain that is worse after activity or toward the end of the day. Chronic inflammation is thought to be the main age-related factor that contributes to osteoarthritis.

[0026] Senescent cells accumulate in tissues and organs of individuals as they age and are found at sites of age-related pathologies, including osteoarthritic joints. Senescent cells are believed important to inhibiting proliferation of dysfunctional or damaged cells and particularly to constraining development of malignancy (see, e.g., Campisi, Curr. Opin. Genet. Dev. 21 : 107-12 (2011); Campisi, Trends Cell Biol. 11 : S27-31 (2001); Prieur et al., Curr. Opin. Cell Biol.

20: 150-55 (2008)); nevertheless, cellular senescence is a molecular mechanism that drives age- associated osteoarthritis and also osteoarthritis caused by joint trauma.

[0027] A senolytic agent is an agent that selectively (preferentially or to a greater degree) destroys, kills, removes, or facilitates selective destruction of senescent cells over non-senescent cells of the same cell type. A senolytic agent is used in an amount and for a time sufficient that selectively kills senescent cells but is insufficient to cause the death of a non-senescent cell.

[0028] Senolytic agents, such as the compound of Formula I, as shown below, prevent (i.e., reduces the likelihood of occurrence), reduces or inhibits loss or erosion of proteoglycan layers in a joint, reduces inflammation in the affected joint, and promotes (i.e., stimulates, enhances, induces) production of collagen (e.g., type 2 collagen). Removal of senescent cells causes a reduction in the amount (i.e., level) of inflammatory cytokines such as, for example, IL-6, produced in a joint and inflammation is reduced. Methods are provided herein for treating osteoarthritis, for selectively killing senescent cells in an osteoarthritic joint of a subject, and/or inducing, for example, collagen (such as Type 2 collagen) production in the joint of a subject in need thereof by administering at least one senolytic agent (which may be combined with at least one pharmaceutically acceptable excipient to form a pharmaceutical composition) to the subject. A senolytic agent also may be used for decreasing (inhibiting, reducing) production of, for example, metalloproteinase 13 (MMP-13), which degrades collagen in a joint, and for restoring proteoglycan layer or inhibiting loss and/or degradation of the proteoglycan layer. Treatment with the senolytic agent thereby also prevents (i.e., reduces likelihood of occurrence of), inhibits, or decreases erosion, or slows (i.e., decreases rate) erosion of the bone. In addition, the methods comprising administering a senolytic agent can reduce joint pain and are therefore useful for pain management of osteoarthritic joints.

[0029] Disclosed herein are methods of treating a patient diagnosed with osteoarthritis in a joint comprising administering to the joint a therapeutically effective dose of a compound of Formula I, or a pharmaceutically acceptable salt thereof: (Formula I)

wherein the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 8 mL of vehicle and the therapeutically effective dose of the compound results in a decrease in synovitis in the joint. The compound of Formula I is shown in the structure above and is also known as 4-[[(4S,5R)-4,5-bis(4-chlorophenyl)-4,5-dihydro-2-[4-methoxy -2-(l- methylethoxy)phenyl]-lH-imidazol-l-yl]carbonyl]-2-piperazino ne. The present disclosure further contemplates that therapeutically effective doses of cytotoxins, or cytostatic agents of which the compound of Formula I is an example, may be efficacious in treating an osteoarthritic joint as described herein.

Therapeutically effective dose of the compound of Formula I

[0030] In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 12 mL of vehicle.

In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 2 mg in at least 16 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula

I is 2 mg in 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 9 mL of vehicle. In some embodiments, the

therapeutically effective dose of the compound of Formula I is 2 mg in 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in

I I mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 2 mg in 16 mL of vehicle.

[0031] In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 3 mg in at least 16 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 3 mg in 16 mL of vehicle.

[0032] In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 4 mg in at least 16 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 4 mg in 16 mL of vehicle.

[0033] In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 6 mg in at least 16 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 6 mg in 16 mL of vehicle.

[0034] In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is at least 8 mg in at least 16 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 1 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 2 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 3 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 4 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 5 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 6 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 7 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 8 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 9 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 10 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 11 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 12 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 13 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 14 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 15 mL of vehicle. In some embodiments, the therapeutically effective dose of the compound of Formula I is 8 mg in 16 mL of vehicle.

Administration

[0035] In some embodiments, the compound of Formula I is administered locally to the joint. In some embodiments, the compound of Formula I is administered intra-articularly to the joint.

In some embodiments, the joint is a fern oro-tibial joint. In some embodiments, the joint is a hip. In some embodiments, the joint is spine, shoulder, ankle, wrist, or finger joint. In some embodiments, the joint is the spine. In some embodiments, the joint is the shoulder. In some embodiments, the joint is the ankle. In some embodiments, the joint is the wrist. In some embodiments, the joint is the finger.

[0036] In some embodiments, the therapeutically effective dose of the compound of Formula I is administered as a single dose. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered to the joint every week for two months, or every 2 weeks for two months, or every month for two months. In some embodiments, the

therapeutically effective dose of the compound of Formula I is administered to the joint every 3 months for 6 months, every four months for 8 months, every five months for 10 months, every 6 months for 1 year. In some embodiments, the therapeutically effective dose of the compound of Formula I is administered to the joint every 3 months for 6 months, every four months for 8 months, every five months for 10 months, every 6 months for 1 year. In some embodiments, the therapeutically effective dose of the compound of Formula I is not administered to the joint for at least 6 months, for at least 1 year, for at least 18 months, for at least 2 years, for at least 3 years, for at least 4 years, for at least 5 years.

[0037] In some embodiments, the joint has senescent cells. In some embodiments, the senescent cells are senescent chondrocytes or senescent epithelial cells, or senescent synoviocytes. In some embodiments, the senescent cells are senescent chondrocytes. In some embodiments, the senescent cells are senescent epithelial cells. In some embodiments, the senescent cells are senescent synoviocytes.

[0038] Senescent cells may be detected by morphology (as viewed by microscopy, for example); production of senescence associated markers such as, for example,

senescence-associated b-galactosidase (SA-P-gal), rΐό 11 ^ 43 , p21, PAI-1, or any one or more SASP factors ( e.g IL-6, MMP3).The presence of the senescent cell-associated polypeptide pl6 can be determined by standard immunochemistry, such as immunoblotting analysis. Expression of pl6 mRNA in a cell can be measured by a variety of techniques including quantitative PCR.

Patient

[0039] As used herein the term“individual,”“patient,” or“subject” refers to individuals diagnosed with, suspected of being afflicted with, or at-risk of developing at least one disease for which the described compositions and method are useful for treating. In certain

embodiments, the individual is a mammal. In certain embodiments, the mammal is a human.

[0040] In some embodiments, the patient is > 40 years of age. In some embodiments, the patient is > 45 years of age. In some embodiments, the patient is > 50 years of age. In some embodiments, the patient is > 55 years of age. In some embodiments, the patient is > 60 years of age. In some embodiments, the patient is < 85 years of age. In some embodiments, the patient is < 80 years of age. In some embodiments, the patient is < 75 years of age. In some embodiments, the patient is < 70 years of age. In some embodiments, the patient is < 75 years of age. In some embodiments, the patient is > 40 and less than < 85 years of age. In some embodiments, the patient is > 45 and less than < 80 years of age. In some embodiments, the patient is > 50 and less than < 75 years of age. In some embodiments, the patient is > 55 and less than < 70 years of age.

[0041] In some embodiments, a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I.

[0042] In some embodiments, the baseline measurement is a baseline pain level. In some embodiments, the baseline pain level is assessed using a pain assessment protocol. In some embodiments, the pain assessment protocol is an 11 -point Numeric Rating Scale (NRS) or a Western Ontario and McMaster Universities (WOMAC) Index pain subscale.

[0043] In some embodiments, the pain assessment protocol is the 11 -point Numeric Rating Scale (NRS). In some embodiments, the 11 -point Numeric Rating Scale (NRS) comprises asking the patient to rate their pain on a scale from 0 to 10 during, for example but not limited to, a 24 hour period, in which 0 is no pain, and 10 represents the worst possible pain. In some embodiments, the baseline pain level of the patient is a mean of > 3 and < 9 points on the 11- point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 4 and < 9 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 5 and < 9 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 3 and < 8 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 4 and < 8 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 5 and < 8 points on the 11- point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 3 and < 7 points on the 11 -point NRS In some embodiments, the baseline pain level of the patient is a mean of > 4 and < 7 points on the 11 -point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 5 and < 7 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 4 and < 5 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 5 and < 6 points on the 11- point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 6 and < 7 points on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 7 and < 8 points on the 11 -point NRS. In some embodiments, the baseline pain level of the patient is a mean of > 8 and < 9 points on the 11 -point NRS. As used herein the term “about” refers to an amount that is near the stated amount by 10% or less. In some embodiments, the baseline pain level of the patient is a mean of about 4 on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of about 5 on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of about 6 on the 11 -point NRS. In some embodiments, the baseline pain level of the patient is a mean of about 7 on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of about 8 on the 11-point NRS. In some embodiments, the baseline pain level of the patient is a mean of about 9 on the 11 -point NRS.

[0044] In some embodiments, the pain assessment protocol is the WOMAC pain subscale. In some embodiments, the WOMAC Index pain subscale comprises a set of questionnaires in which a health professional evaluates the condition of the patient. In some embodiments, the questionnaire contains 5 questions pertaining to the amount of pain that the patient feels during walking, using stairs, in bed, sitting, and standing. Each question is scored on a scale of 0 to 4 which correspond to: none (0), mild (1), moderate (2), severe (3), and extreme (4). The scores are summed up, with a total possible score of 0-20 for the pain subscale. In some embodiments, the baseline pain level of the patient is > 6 on the WOMAC pain subscale. In some

embodiments, the baseline pain level of the patient is > 6 and less than <_17 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is > 6 and less than < 16 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is > 6 and less than <_15 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is > 6 and less than <_14 on the WOMAC pain subscale. In some

embodiments, the baseline pain level of the patient is > 6 and less than <_13 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is > 6 and less than < 12 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is > 6 and less than <_11 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is > 6 and less than <_10 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 6. In some embodiments, the baseline pain level of the patient is 7 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 8 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 9 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 10 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 11 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 12 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 13 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 14 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 15 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 16 on the WOMAC pain subscale. In some embodiments, the baseline pain level of the patient is 17 on the WOMAC pain sub scale. In some embodiments, the baseline pain level of the patient is 18 on the WOMAC pain sub scale. In some

embodiments, the baseline pain level of the patient is 19 on the WOMAC pain subscale.

[0045] In some embodiments, the baseline measurement is a baseline functionality level. In some embodiments, the baseline functionality level is assessed by WOMAC Index functionality subscale. In some embodiments, the baseline functionality level is assessed by WOMAC Index functional subscale. In some embodiments, the WOMAC Index functional subscale comprises a set of questionnaires in which a health professional evaluates the condition of the patient. In some embodiments, the questionnaire contains 17 questions such as, for example: the degree of difficulty with performing the following: A. Descending stairs, B. Ascending stairs, C. Rising from sitting, D. Standing, E. Bending to floor, F. Walking on a flat surface, G. Getting in or out of a car, H. Going shopping, I. Putting on socks/stockings, J. Rising from bed, K. Taking off socks/stockings, L. Lying in bed, M. Getting in/out of the bath, N. Sitting, O. Getting on/off the toilet, P. Heavy domestic duties (such as mowing the lawn, lifting heavy grocery bags), Q. Light domestic duties (such as tidying a room, dusting, or cooking).

[0046] Each question is scored on a scale of 0 to 4 which correspond to: none (0), mild (1), moderate (2), severe (3), and extreme (4). The scores are summed up, with a total possible score of 0-68 for the WOMAC functional subscale. In some embodiments, the baseline functionality level of the patient is > 17 and < 68. In some embodiments, the baseline functionality level of the patient is > 18 and < 68. In some embodiments, the baseline functionality level of the patient is > 19 and < 68. In some embodiments, the baseline functionality level of the patient is > 20 and < 68. In some embodiments, the baseline functionality level of the patient is > 21 and < 68. In some embodiments, the baseline functionality level of the patient is > 22 and < 68. In some embodiments, the baseline functionality level of the patient is > 23 and < 68. In some embodiments, the baseline functionality level of the patient is > 24 and < 68. In some embodiments, the baseline functionality level of the patient is > 25 and < 68. In some embodiments, the baseline functionality level of the patient is > 26 and < 68. In some embodiments, the baseline functionality level of the patient is > 27 and < 68. In some embodiments, the baseline functionality level of the patient is > 28 and < 68. In some embodiments, the baseline functionality level of the patient is > 29 and < 68. In some embodiments, the baseline functionality level of the patient is > 30 and < 68. In some embodiments, the baseline functionality level of the patient is > 31 and < 68. In some embodiments, the baseline functionality level of the patient is > 32 and < 68. In some embodiments, the baseline functionality level of the patient is > 33 and < 68. In some embodiments, the baseline functionality level of the patient is > 34 and < 68. In some embodiments, the baseline functionality level of the patient is > 35 and < 68. In some embodiments, the baseline functionality level of the patient is > 36 and < 68. In some embodiments, the baseline functionality level of the patient is > 37 and < 68. In some embodiments, the baseline functionality level of the patient is > 38 and < 68. In some embodiments, the baseline functionality level of the patient is > 39 and < 68. In some embodiments, the baseline functionality level of the patient is > 40 and < 68. In some embodiments, the baseline functionality level of the patient is > 41 and < 68. In some embodiments, the baseline functionality level of the patient is > 42 and < 68. In some embodiments, the baseline functionality level of the patient is > 43 and < 68. In some embodiments, the baseline functionality level of the patient is > 44 and < 68. In some embodiments, the baseline functionality level of the patient is > 45 and < 68. In some embodiments, the baseline functionality level of the patient is > 46 and < 68. In some embodiments, the baseline functionality level of the patient is > 47 and < 68. In some embodiments, the baseline functionality level of the patient is > 48 and < 68. In some embodiments, the baseline functionality level of the patient is > 49 and < 68. In some embodiments, the baseline functionality level of the patient is > 30 and < 68. In some embodiments, the baseline functionality level of the patient is > 31 and < 68. In some embodiments, the baseline functionality level of the patient is > 32 and < 68. In some embodiments, the baseline functionality level of the patient is > 33 and < 68. In some embodiments, the baseline functionality level of the patient is > 34 and < 68. In some embodiments, the baseline functionality level of the patient is > 35 and < 68. In some embodiments, the baseline functionality level of the patient is > 36 and < 68. In some embodiments, the baseline functionality level of the patient is > 37 and < 68. In some embodiments, the baseline functionality level of the patient is > 38 and < 68. In some embodiments, the baseline functionality level of the patient is > 39 and < 68. In some embodiments, the baseline functionality level of the patient is > 50 and < 68. In some embodiments, the baseline functionality level of the patient is > 51 and < 68. In some embodiments, the baseline functionality level of the patient is > 52 and < 68. In some embodiments, the baseline functionality level of the patient is > 53 and < 68. In some embodiments, the baseline functionality level of the patient is > 54 and < 68. In some embodiments, the baseline functionality level of the patient is > 55 and < 68. In some embodiments, the baseline functionality level of the patient is > 56 and < 68. In some embodiments, the baseline functionality level of the patient is > 57 and < 68. In some embodiments, the baseline functionality level of the patient is > 58 and < 68. In some embodiments, the baseline functionality level of the patient is > 59 and < 68. In some embodiments, the baseline functionality level of the patient is > 60 and < 68.

[0047] In some embodiments, the baseline measurement is a baseline stiffness level. In some embodiments, the baseline stiffness level is assessed by WOMAC Index stiffness subscale. In some embodiments, the WOMAC Index stiffness subscale comprises a set of questionnaires in which a health professional evaluates the condition of the patient. In some embodiments, the questionnaire contains 2 questions, such as, for example: How severe is your stiffness after first awakening in the morning? How severe is your stiffness after sitting, lying, or resting later in the day? Each question is scored on a scale of 0 to 4 which correspond to: none (0), mild (1), moderate (2), severe (3), and extreme (4). The scores are summed up, with a total possible score of 0-8 for the WOMAC stiffness subscale. In some embodiments, the baseline stiffness level of the patient is > 1 and < 8. In some embodiments, the baseline stiffness level of the patient is > 1 and < 7. In some embodiments, the baseline stiffness level of the patient is > 1 and < 6. In some embodiments, the baseline stiffness level of the patient is > 1 and < 5. In some embodiments, the baseline stiffness level of the patient is > 1 and < 4. In some embodiments, the baseline stiffness level of the patient is > 1 and < 3. In some embodiments, the baseline stiffness level of the patient is 1. In some embodiments, the baseline stiffness level of the patient is 2. In some embodiments, the baseline stiffness level of the patient is 3. In some embodiments, the baseline stiffness level of the patient is 4. In some embodiments, the baseline stiffness level of the patient is 5. In some embodiments, the baseline stiffness level of the patient is 6. In some embodiments, the baseline stiffness level of the patient is 7. In some embodiments, the baseline stiffness level of the patient is 8.

[0048] In some embodiments, the baseline measurement is a baseline radiological

categorization. In some embodiments, the baseline radiological categorization is a Kellgren- Lawrence grade. The Kellgren-Lawrence grading system is a radiological classification system for assessing severity of osteoarthritis. The grading system categorizes osteoarthritis severity according to the following grades: grade 0 - no radiographic features of OA are present, grade 1- doubtful joint space narrowing (JSN) and possible osteophytic lipping, grade 2 - definite osteophytes and possible JSN on anteroposterior weight-bearing radiograph, grade 3 - multiple osteophytes, definite JSN, sclerosis, possible bony deformity, grade 4 - large osteophytes, marked JSN, severe sclerosis and definite bony deformity. In some embodiments, the baseline radiological categorization of the patient is 1 through 4 on the Kellgren-Lawrence grading system. In some embodiments, the baseline radiological categorization of the patient is 1 on the Kellgren-Lawrence grading system. In some embodiments, the baseline radiological categorization of the patient is 2 on the Kellgren-Lawrence grading system. In some

embodiments, the baseline radiological categorization of the patient is 3 on the Kellgren- Lawrence grading system. In some embodiments, the baseline radiological categorization of the patient is 4 on the Kellgren-Lawrence grading system. In some embodiments, the baseline radiological categorization is of the fern oro-tibial joint.

[0049] In some embodiments, the baseline measurement is a baseline synovitis level. In some embodiments, the baseline synovitis level is assessed using an 11 point MRI score based on Guermazi et al. [assessment of synovitis with contrast-enhanced MRI using a while-joint semiquantitiative scoring system in people with, or at high risk of, knee osteoarthritis: the MOST study.” Ann Rheum Dis 2011;70:805-811. In the 11 point MRI reading, 9 anatomical sites as outlined in Table 1 are evaluated for synovitis. If present, Baker’s cysts or loose bodies, are scored in addition to the 9 anatomical locations. Synovitis comprises an inflammation of the synovial membrane and is characterized on MRI by thickening and enhancement after administration of intravenous contrast agent. Each site as outlined in Table 1 is scored based on the maximal thickness in any slice at each site as follows: grade 0 if <2 mm, grade 1 if 2-4 mm and grade 2 if >4 mm (as shown in Table 2) with a maximal score of 22. The scores are summed to calculate a synovitis level (as shown in Table 3).

Table 1. Locations for 11 point MRI score

Table 2. Synovial Thickness Scoring

Table 3. Summed Score Categories

[0050] In some embodiments, the baseline synovitis level of the patient is at least 7 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 8 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 9 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 10 as measured by the 11 point MRI score.

In some embodiments, the baseline synovitis level of the patient is at least 11 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 12 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 13 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 14 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 15 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is at least 16 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is > 7 and < 18 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is > 8 and < 17 as measured by the 11 point MRI score. In some embodiments, the baseline synovitis level of the patient is > 8 and < 16 as measured by the 11 point MRI score.

[0051] In some embodiments, the baseline measurement is a baseline joint architecture measurement. In some embodiments, joint architecture refers to bone structure, cartilage structure, joint space width (JSW), or joint space narrowing (JSN), in the joint. In some embodiments, the baseline joint architecture measurement is a baseline cartilage thickness measurement, a baseline subchondral bone surface area measurement, a baseline Kellgren- Lawrence (KL) grade, or a baseline progression through a KL grading scale. In some

embodiments, the baseline joint architecture measurement is baseline bone, tendon, or meniscus measurement. In some embodiments, the baseline joint architecture measurement is measured using MRI, ultrasound, computed tomography, or an X-ray device. In some embodiments, the joint of the baseline joint architecture measurement is a knee. In some embodiments, the knee is the patient’s left knee. In some embodiments, the knee is the patient’s right knee.

[0052] In some embodiments, the baseline joint architecture measurement is a baseline cartilage thickness measurement. In some embodiments, the baseline cartilage thickness measurement is measured using X-ray or MRI. In some embodiments, the baseline cartilage thickness measurement is a baseline joint space width (JSW) measurement or a baseline joint space narrowing (JSN) measurement.

[0053] In some embodiments, the baseline cartilage thickness measurement is a baseline joint space width (JSW) measurement. In some embodiments, the baseline JSW measurement is a baseline medial JSW measurement. In some embodiments, the baseline JSW measurement is a baseline lateral JSW measurement. In some embodiments, the baseline JSW is a baseline minimum JSW. In some embodiments, the baseline minimum JSW is the shortest visible distance between the femoral condyle and the tibial plateau.

[0054] In some embodiments, the baseline JSW measurement is 2 mm. In some embodiments, the baseline JSW measurement is 2.5 mm. In some embodiments, the baseline JSW measurement is 3 mm. In some embodiments, the baseline JSW measurement is 3.5 mm. In some embodiments, the baseline JSW measurement is 4 mm. In some embodiments, the baseline JSW measurement is 4.5 mm. In some embodiments, the baseline JSW measurement is 5 mm. In some embodiments, the baseline JSW measurement is 5.5 mm. In some embodiments, the baseline JSW measurement is 6 mm. In some embodiments, the baseline JSW measurement is 6.5 mm. In some embodiments, the baseline JSW measurement is 7 mm.

[0055] In some embodiments, the baseline cartilage thickness measurement is a baseline joint space narrowing (JSN) measurement. When JSN occurs, cartilage in the joint may wear away, and the existing cartilage may be prevented from keeping bones of the joint a normal distance apart. In some embodiments, the baseline JSN measurement is a baseline change in JSW. In some embodiments, the baseline JSW measurement is measured based on longitudinal changes in a minimum JSW measurement. In some embodiments, the baseline change in JSW of the baseline JSN measurement is 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12,

0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4,

1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 mm, or more. In some embodiments, the baseline JSN measurement is a baseline change in JSW over time. In some embodiments, the time of the baseline change in JSW over time is 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, or 10 years. For example, the baseline JSN measurement may be 0.02 to 0.05 mm per year.

[0056] In some embodiments, the baseline joint architecture measurement is a baseline subchondral bone surface area measurement. In some embodiments, the baseline subchondral bone surface area measurement is measured using X-ray or MRI. In some embodiments, the baseline subchondral bone surface area measurement is a baseline bone density. In some embodiments, the baseline bone density is 200 Hounsfield Units (HU), 250 HU, 300 HU, 350

HU, 400 HU, 450 HU, 500 HU, 550 HU, 600 HU, 650 HU, 700 HU, 750 HU, or 800 HU. In some embodiments, the baseline subchondral bone surface area measurement is a baseline number of cystic areas. In some embodiments, the baseline subchondral bone surface area measurement is a baseline number of cystic areas is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more. In some embodiments, the cystic areas comprise low-density bone areas.

[0057] In some embodiments, the baseline joint architecture measurement is a baseline

Kellgren-Lawrence (KL) grade, or a grade using another ordinal scoring system. In some embodiments, the baseline KL grade is 1. In some embodiments, the baseline KL grade is 2. In some embodiments, the baseline KL grade is 3. In some embodiments, the baseline KL grade is 4. In some embodiments, the baseline KL grade is 2 or 3. In some embodiments, the baseline KL grade is 3 or 4. In some embodiments, the baseline KL grade is 2, 3 or 4.

[0058] In some embodiments, the baseline joint architecture measurement is a baseline progression through a KL grading scale. In some embodiments, the baseline progression through the KL grading scale comprises a KL grade increase. In some embodiments, the KL grade increase is 0.5. In some embodiments, the KL grade increase is 1. In some embodiments, the KL grade increase is 2. In some embodiments, the KL grade increase is 3. In some embodiments, the KL grade increase of the baseline progression through the KL grading scale is over time. In some embodiments, time of the KL grade increase is 1 month, 2 months, 3 months, 4 months,

5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, or 10 years. For example, the baseline progression through a KL grading scale may be a 0.5 grade increase per year.

Efficacy of Therapeutically effective dose of the compound of Formula I

[0059] In some embodiments, efficacy of the therapeutically effective dose of the compound of Formula I is assessed. In some embodiments, a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I.

[0060] In some embodiments, the baseline measurement is a baseline synovitis level. In some embodiments, the baseline synovitis level is assessed using an 11 point MRI score. In some embodiments, the decrease in synovitis in the joint is measured by the 11-point MRI score.

[0061] In some embodiments, the decrease in synovitis in the joint is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 1 point as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient.

In some embodiments, the decrease in synovitis is by a decrease of at least 4 points, at least 5 points, at least 8 points, at least 9 points, at least 10 points, wherein the decrease is between 1-4 points, between 4-8 points, between 8-10 points, between 10-13 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some

embodiments, the decrease in synovitis is by a decrease of between 1-4 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient.

[0062] In some embodiments, the decrease in synovitis is by a decrease of at least 2 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 3 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 4 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 5 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 6 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 7 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 8 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 9 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 10 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 11 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 12 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 13 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 14 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 15 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 16 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 17 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 18 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 19 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient. In some embodiments, the decrease in synovitis is by a decrease of at least 20 points as measured by the 11 point MRI score as compared to the baseline synovitis level of the patient.

[0063] In some embodiments, the decrease in synovitis in the joint is measured 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 4 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 6 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 8 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 6 months after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovitis in the joint is measured 1 year after the compound of Formula I is administered to the patient.

[0064] In some embodiments, the baseline measurement is a baseline synovial fluid level. In some embodiments, the baseline synovial fluid level of the patient is measured by physical examination, or by ultrasound, or by MRF In some embodiments, the therapeutically effective dose of the compound of Formula I results in a decrease in synovial fluid in the joint as measured by physical examination, ultrasound, or MRI, as compared to the baseline synovial fluid level. In some embodiments, the decrease in synovial fluid in the joint is measured 24 weeks, 12 weeks, or 8 weeks, or 4 weeks, or 2 weeks after the compound of Formula I is administered to the patient.

[0065] In some embodiments, the decrease in synovial fluid in the joint is measured 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 4 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 6 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 8 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 10 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 6 months after the compound of Formula I is administered to the patient. In some embodiments, the decrease in synovial fluid in the joint is measured 1 year after the compound of Formula I is administered to the patient.

[0066] In some embodiments, efficacy of the therapeutically effective dose of the compound of Formula I is assessed. In some embodiments, a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula I.

[0067] In some embodiments, the baseline measurement is a pain level. In some embodiments, the therapeutically effective dose of the compound of Formula I further results in a decrease in pain in the joint as compared to the baseline pain level. In some embodiments, the decrease in pain in the joint is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain in the joint is measured 24 weeks after the compound of Formula I is administered to the patient.

[0068] In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of between 2 to 4 points as assessed by the 11-point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 1 point as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 2 points as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 3 points as assessed by the 11-point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 4 points as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 5 points as assessed by the 11-point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 6 points as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 7 points as assessed by the 11- point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 8 points as assessed by the 11-point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 9 points as assessed by the 11 -point NRS. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 10 points as assessed by the 11 -point NRS.

[0069] In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points, as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of 0.5 point, of 1 point, or of 2 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 0.5 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 1 point as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 2 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 3 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 4 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 5 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 6 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 7 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 8 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 9 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 10 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 11 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 12 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 13 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 14 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 15 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 16 points as assessed by the WOMAC Index pain sub scale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 17 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 18 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 19 points as assessed by the WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 20 points as assessed by the WOMAC Index pain sub scale.

[0070] In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 50%, 40%, 30%, between 30-40%, or between 40-50% as measured by the 11 -point NRS or WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 50% as measured by the 11 -point NRS or WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 40% as measured by the 11 -point NRS or

WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 30% as measured by the 11 -point NRS or

WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 20% as measured by the 11 -point NRS or

WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 10% as measured by the 11 -point NRS or

WOMAC Index pain subscale. In some embodiments, the decrease in pain in the joint is a change from the baseline pain level of about 5% as measured by the 11 -point NRS or WOMAC Index pain subscale. In some embodiments, the decrease in pain relative to the baseline pain level lasts 3 months or more, 4 months or more, lasts 5 months or more, lasts 6 months or more, lasts 7 months or more, lasts 8 months or more, lasts 9 months or more, lasts 10 months or more, lasts 11 months or more, lasts 1 year or more, lasts 2 years or more, lasts 3 years or more, lasts 4 years or more, or lasts 5 years or more.

[0071] In some embodiments, the decrease in pain is measured 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain is measured 4 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain is measured 6 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain is measured 8 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain is measured 6 months after the compound of Formula I is administered to the patient. In some embodiments, the decrease in pain is measured 1 year after the compound of Formula I is administered to the patient.

[0072] In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts 3 months or more, 4 months or more, lasts 5 months or more, lasts 6 months or more, lasts 7 months or more, lasts 8 months or more, lasts 9 months or more, lasts 10 months or more, lasts 11 months or more, lasts 1 year or more, lasts 2 years or more, lasts 3 years or more, lasts 4 years or more, or lasts 5 years or more. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 3 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 4 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 5 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 6 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 7 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 8 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 9 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 10 months. In some

embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 11 months. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 1 year. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 2 years. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 3 years. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 4 years. In some embodiments, the decrease the decrease in pain relative to the baseline pain level lasts about 5 years.

[0073] In some embodiments, the therapeutically effective dose of the compound of Formula I results in an increase functionality of the joint as measured by a decrease in the WOMAC Index functionality subscale score relative to the baseline functionality level as measured by the WOMAC Index functionality subscale. In some embodiments, the increase in functionality of the joint is measured 12 weeks after the compound of Formula I is administered to the patient.

In some embodiments, the increase in functionality of the joint is measured 24 weeks after the compound of Formula I is administered to the patient.

[0074] In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of at least 0.5 point, at least one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points, at least nine points, or at least ten points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of 0.5 point, of 1 point, of 2 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 0.5 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 1 point. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 2 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 3 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 4 points.

In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 5 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 6 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 7 points.

In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 8 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 9 points. In some embodiments, the decrease in the WOMAC Index functionality subscale score is a change from the baseline functionality level of about 10 points.

[0075] In some embodiments, the increase in functionality of the joint is measured 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the increase in functionality of the joint is measured 4 weeks after the compound of Formula I is administered to the patient. In some embodiments, the increase in functionality of the joint is measured 6 weeks after the compound of Formula I is administered to the patient. In some embodiments, the increase in functionality of the joint is measured 8 weeks after the compound of Formula I is administered to the patient. In some embodiments, the increase in functionality of the joint is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the increase in functionality of the joint is measured 6 months after the compound of Formula I is administered to the patient. In some embodiments, the increase in functionality of the joint is measured 1 year after the compound of Formula I is administered to the patient.

[0076] In some embodiments, the baseline measurement is a baseline stiffness level. In some embodiments, the baseline stiffness level is assessed by WOMAC Index stiffness subscale. In some embodiments, the therapeutically effective dose of the compound of Formula I results in a decrease stiffness in the joint as assessed by WOMAC Index stiffness subscale relative to the baseline stiffness level. In some embodiments, the decrease in stiffness in the joint is a change from the baseline stiffness level of at least 0.5 point, one point, at least two points, at least three points, at least four points, at least five points, at least six points, at least seven points, at least eight points as assessed by WOMAC Index stiffness subscale. In some embodiments, the decrease in stiffness in the joint is measured 24 weeks, or 12 weeks, or 8 weeks, or 4 weeks, or 2 weeks after the compound of Formula I is administered to the patient.

[0077] In some embodiments, efficacy of the therapeutically effective dose of the compound of Formula I is assessed. In some embodiments, a baseline measurement is obtained from the patient prior to administering to the joint a therapeutically effective dose of the compound of Formula F In some embodiments, the baseline measurement is a baseline biomarker level. In some embodiments, the baseline biomarker levels are measured from the patient’s synovial fluid. In some embodiments, the baseline biomarker levels are measured from the patient’s plasma. In some embodiments, the therapeutically effective dose of the compound of Formula I results in modulation of a biomarker level relative to the baseline biomarker level. In some embodiments, the modulation of the biomarker levels relative to the baseline biomarker levels is a decrease. In some embodiments, the modulation of the biomarker levels relative to the baseline biomarker levels is an increase. In some embodiments, the biomarker comprises MMP- 10, MMP-12, MMP-13, IL-6, IL-10, CCL20, A2M, or VEGF-C. In some embodiments, the biomarker comprises CXCL10, CCL20, CCL19, CXCL1, CD14, Complement protein C3, GDF15, IL-10, IL-6, fibronectin, ICAM-1, VEGF-C, MMP-1, MMP-3, MMP-10, MMP-12, MMP-13, TIMP-1, or A2M. In some embodiments, the biomarker comprises CCL20, CCL19, IL-10, IL-6, ICAM-1, VEGF-C, MMP-3, MMP-10, MMP-12, MMP-13, or A2M. In some embodiments, the biomarker comprises IL-6. However, the biomarker is not limited to these examples.

[0078] In some embodiments, the biomarker comprises a cytokine or chemokine. In some embodiments, the cytokine or chemokine comprises C-X-C motif chemokine 10 (CXCL10; Perrott et al. Geroscience 2017; Kisand et al. Osteoarthritis Cartilage 2018), Chemokine (C-C motif) ligand 20 (CCL20; Freund et al. Trends Mol Med. 2010; Alaaeddine et al. Inflamm Res. 2015), Chemokine (C-C motif) ligand 19 (CCL19; Carter et al. Genome Biol. 2005; Robinson et al. Nat Rev Rheumatol. 2017), Interleukin 11 (IL-11; Freund et al. Trends Mol Med. 2010; Kokebie et al. Arthritis Res Ther. 2011), S100 calcium-binding protein A8 (S100A8; Ruan et al. Osteoarthritis Cartilage 2019), chemokine (C-X-C motif) ligand 1 (CXCL1; Freund et al. Trends Mol Med. 2010; Moradi et al. Clin Exp Immunol. 2015), cluster of differentiation 14 (CD 14; Gomez -Aristizabal et al. Arthritis Res Ther. 2019), Interleukin 1 beta (IL-Ib; Freund et al.

Trends Mol Med. 2010; De Bakker et al. Veterinary Record 2017; Fujita et al. Am J Vet Res. 2005), Complement protein C3 (Cardoso et al. Ageing Res Rev. 2018), Growth Differentiation Factor 15 (GDF15; Kim et al. Genes Dev. 2018; Cevidanes et al. Osteoarthritis Cartilage 2014), Granulocyte-macrophage colony-stimulating factor (GM-CSF; Freund et al. Trends Mol Med. 2010; Lee et al. Arthritis Res Ther. 2018), Interleukin 10 (IL-10; Greenhill et al. Arthritis Res Ther. 2014), Chemokine (C-X-C motif) ligand 6 (CXCL6; Freund et al. Trends Mol Med. 2010; Scanzello J Orthop Res. 2017), Interleukin 6 (IL-6; Freund et al. Trends Mol Med. 2010; Guan et al. Discov Med. 2019), Tumor necrosis factor receptor 2 (TNFR2; Freund et al. Trends Mol Med. 2010; Simao et al. Rheumatol Int. 2014), or a combination thereof. In some embodiments, the cytokine or chemokine comprises CXCL10, CCL20, CCL19, CXCL1, CD14, C3, GDF15, IL-10, IL-6, or a combination thereof. In some embodiments, the cytokine or chemokine comprises CCL20, CCL19, IL-10, IL-6, or a combination thereof. In some embodiments, the cytokine or chemokine comprises IL-6. In some embodiments, the cytokine or chemokine is selected from the group consisting of CXCL10, CCL20, CCL19, IL-11, S100A8, CXCL1,

CD 14, IL-Ib, C3, GDF15, GM-CSF, IL-10, CXCL6, IL-6, and TNFR2. In some embodiments, the cytokine or chemokine is selected from the group consisting of CXCL10, CCL20, CCL19, CXCL1, CD14, C3, GDF15, IL-10, and IL-6. In some embodiments, the cytokine or chemokine is selected from the group consisting of CCL20, CCL19, IL-10, and IL-6. In some embodiments, the cytokine or chemokine is IL-6.

[0079] In some embodiments, the biomarker comprises a growth factor or adhesion molecule. In some embodiments, the growth factor or adhesion molecule comprises fibronectin (Scuderi et al. J Bone Joint Surg Am. 2011), intercellular adhesion molecule 1 (ICAM-1; Freund et al.

Trends Mol Med. 2010; Karatay et al. Ann Clin Lab Sci. 2004), vascular endothelial growth factor C (VEGF-C; Paavonen et al. J Rheumatol. 2002; Wauke et al. J Rheumatol. 2002), serpin family F member 1 (SERPINFl; Ritter et al. Arthritis Res Ther. 2014), Pigment epithelium- derived factor (PEDF), or a combination thereof. In some embodiments, the growth factor or adhesion molecule comprises fibronectin, ICAM-1, VEGF-C, or a combination thereof. In some embodiments, the growth factor or adhesion molecule comprises ICAM-1, VEGF-C, or a combination thereof. In some embodiments, the growth factor or adhesion molecule is selected from the group consisting of ICAM-1, VEGF-C, SERPINFl, and PEDF. In some embodiments, the growth factor or adhesion molecule is selected from the group consisting of fibronectin, ICAM-1, and VEGF-C. In some embodiments, the growth factor or adhesion molecule is selected from the group consisting of ICAM-1 and VEGF-C.

[0080] In some embodiments, the biomarker comprises a protease or a protease inhibitor. In some embodiments, the protease or protease inhibitor comprises matrix metalloproteinase- 1 (MMP-1; Nagase et al. Matrix Suppl. 1992; Freund et al. Trends Mol Med. 2010; Wyatt et al. Osteoarthritis Cartilage 2018), matrix metalloproteinase-3 (MMP-3; Sellers and Murphy Int Rev Connect Tissue Res. 1981; Freund et al. Trends Mol Med. 2010; Cevidanes et al. Osteoarthritis Cartilage 2014), matrix metalloproteinase- 13 (MMP-13; Mitchell et al. J Clin Invest. 1996; Reboul et al. J Clin Invest. 1996; Freund et al. Trends Mol Med. 2010; Ma et al. Connect Tissue Res. 2019), TEMP metallopeptidase inhibitor 1 (TEMPI; Freund et al. Trends Mol Med. 2010; Cevidanes et al. Osteoarthritis Cartilage 2014), A2M (Chou et al. Osteoarthritis Cartilage 2018; Zhao et al. J Cell Biochem. 2015) or a combination thereof. In some embodiments, the protease or protease inhibitor comprises MMP-1, MMP-3, MMP-10, MMP-12, MMP-13, TEMP-1, A2M, or a combination thereof. In some embodiments, the protease or protease inhibitor comprises MMP-3, MMP-10, MMP-12, MMP-13, A2M, or a combination thereof. In some embodiments, the protease or protease inhibitor is selected from the group consisting of MMP-1, MMP-3, MMP-13, and TEMPI. In some embodiments, the protease or protease inhibitor is selected from the group consisting of MMP-1, MMP-3, MMP-10, MMP-12, MMP-13, TEMP-1, and A2M. In some embodiments, the protease or protease inhibitor is selected from the group consisting of MMP-3, MMP-10, MMP-12, MMP-13, and A2M.

[0081] In some embodiments, the biomarker comprises a cytokine or chemokine, a growth factor or adhesion molecule, or a protease or a protease inhibitor. Examples of cytokines, chemokines, growth factors, adhesion molecules, proteases, and protease inhibitors include but are not limited to those provided above. In some embodiments, the biomarker comprises a biomarker other than a cytokine, chemokine, growth factor, adhesion molecule, protease, or protease inhibitor.

[0082] In some embodiments, the biomarker comprises a cytokine or chemokine, or a growth factor or adhesion molecule. For example, the biomarker may comprise CXCL10, CCL20, CCL19, CXCL1, CD 14, C3, GDF15, IL-10, IL-6, fibronectin, ICAM-1, or VEGF-C. In some embodiments, the biomarker comprises CCL20, CCL19, IL-10, IL-6, ICAM-1, or VEGF-C. In some embodiments, the biomarker comprises IL-6, fibronectin, ICAM-1, or VEGF-C. In some embodiments, the biomarker comprises IL-6, ICAM-1, or VEGF-C.

[0083] In some embodiments, the biomarker comprises a cytokine or chemokine, or a protease or a protease inhibitor. For example, the biomarker may comprise CXCL10, CCL20, CCL19, CXCL1, CD 14, C3, GDF15, IL-10, IL-6, MMP-1, MMP-3, MMP-10, MMP-12, MMP-13, TIMP-1, or A2M. In some embodiments, the biomarker comprises CCL20, CCL19, IL-10, IL- 6, MMP-3, MMP-10, MMP-12, MMP-13, or A2M. In some embodiments, the biomarker comprises IL-6, MMP-1, MMP-3, MMP-10, MMP-12, MMP-13, TIMP-1, or A2M. In some embodiments, the biomarker comprises IL-6, MMP-3, MMP-10, MMP-12, MMP-13, or A2M.

[0084] In some embodiments, the biomarker comprises a growth factor or adhesion molecule, or a protease or a protease inhibitor. For example, the biomarker may comprise fibronectin, ICAM-1, VEGF-C, MMP-1, MMP-3, MMP-10, MMP-12, MMP-13, TIMP-1, or A2M. In some embodiments, the biomarker comprises ICAM-1, VEGF-C, MMP-3, MMP-10, MMP-12, MMP- 13, or A2M.

[0085] In some embodiments, the level of CCL20 is decreased relative to a baseline CCL20 level. In some embodiments, the level of CCL19 is increased relative to a baseline CCL19 level. In some embodiments, the level of IL-10 is decreased relative to a baseline IL-10 level. In some embodiments, the level of IL-6 is decreased relative to a baseline IL-6 level. In some

embodiments, the level of MMP-3 is increased relative to a baseline MMP-3 level. In some embodiments, the level of MMP-10 is decreased relative to a baseline MMP-10 level. In some embodiments, the level of MMP-12 is decreased relative to a baseline MMP-12 level. In some embodiments, the level of MMP-13 is decreased relative to a baseline MMP-13 level. In some embodiments, the level of A2M is increased relative to a baseline A2M level. In some embodiments, the level of ICAM-1 is increased relative to a baseline ICAM-1 level. In some embodiments, the level of VEGF-C is decreased relative to a baseline VEGF-C level.

[0086] In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 4 weeks, measured 8 weeks, measured 12 weeks, or measured 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 2 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 4 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 6 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 8 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 10 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 24 weeks after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 6 months after the compound of Formula I is administered to the patient. In some embodiments, the modulation of the biomarker relative to the baseline biomarker level is measured 1 year after the compound of Formula I is administered to the patient.

[0087] In some embodiments, the contemplated treatment methods result in a "much improved" or "very much improved" patient global impression of change (PGIC) for the patient. PGIC, as used herein, is a patient-reported outcome of how a treatment is progressing, and can be used as an indicator of meaningful change in treatments, such as for example, osteoarthritic pain. As used herein, the PGIC is scored on a 5 level scale, ranging from (5): minimally worse; (4): no change; (3): minimally improved; (2) much improved; and (1) very much improved.

[0088] In some embodiments, the therapeutically effective dose of the compound of Formula I results in the patient’s global impression of change (PGIC) to become much improved or very much improved relative to the patient’s baseline impression.

[0089] In some embodiments, the baseline measurement is a joint architecture measurement.

In some embodiments, the therapeutically effective dose of the compound of Formula I further results in an improvement in the joint architecture measurement compared to the baseline joint architecture measurement. In some embodiments, the improvement comprises a reversal, slowing, or stopping of an adverse effect on the joint architecture measurement relative to the baseline joint architecture measurement. In some embodiments, the improvement in the joint architecture measurement is measured 24 weeks, or 12 weeks after the compound of Formula I is administered to the patient. In some embodiments, the joint architecture measurement is a cartilage thickness measurement, a subchondral bone surface area measurement, a Kellgren- Lawrence (KL) grade, or a progression through a KL grading scale. In some embodiments, the joint architecture measurement is bone, tendon, or meniscus measurement. In some

embodiments, the joint architecture measurement is measured using MRI, ultrasound, computed tomography, or an X-ray device. In some embodiments, the joint of the joint architecture measurement is a knee. In some embodiments, the knee is the patient’s left knee. In some embodiments, the knee is the patient’s right knee.

[0090] In some embodiments, the baseline joint architecture measurement is a baseline cartilage thickness measurement. In some embodiments, the joint architecture measurement is a cartilage thickness measurement. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an improvement in the cartilage thickness measurement compared to the baseline cartilage thickness measurement. In some embodiments, the cartilage thickness measurement is measured using X-ray or MRI.

[0091] In some embodiments, the baseline cartilage thickness measurement is a baseline joint space width (JSW) measurement. In some embodiments, the cartilage thickness measurement is a JSW measurement. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an improvement in the JSW measurement compared to the baseline JSW measurement. In some embodiments, the JSW measurement is a medial JSW measurement. In some embodiments, the JSW measurement is a lateral JSW measurement. In some

embodiments, the JSW measurement is a minimum JSW. In some embodiments, the JSW is the shortest visible distance between the femoral condyle and the tibial plateau.

[0092] In some embodiments, the improvement is an increase in the JSW measurement relative to the baseline JSW measurement of at least 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1,

1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, 2.0 mm, or more. For example, the improvement may be an increase in the JSW measurement relative to the baseline JSW measurement of 0.01-2.00 mm, 0.02-1.00 mm, or 0.05-0.50 mm, or 0.10-0.20 mm. In some embodiments, the improvement is an increase in the JSW measurement relative to the baseline JSW measurement of 0.1-0.6 mm. In some embodiments, the increase in the JSW measurement is at least 0.01 mm. In some embodiments, the increase in the JSW measurement is at least 0.05 mm. In some embodiments, the increase in the JSW measurement is at least 0.10 mm. In some embodiments, the increase in the JSW measurement is at least 0.25 mm. In some embodiments, the increase in the JSW measurement is at least 0.50 mm. In some embodiments, the increase in the JSW measurement is at least 1.00 mm. In some embodiments, the increase in the JSW measurement is at least 1.50 mm. In some embodiments, the increase in the JSW measurement is at least 2.00 mm. In some embodiments, the increase in the JSW measurement is at least 2.50 mm.

[0093] In some embodiments, the baseline cartilage thickness measurement is a baseline joint space narrowing (JSN) measurement. In some embodiments, the cartilage thickness

measurement is a JSN measurement. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an improvement in the JSN measurement compared to the baseline JSN measurement. In some embodiments, the improvement comprises reversing, stopping or decreasing the JSN measurement relative to the baseline JSN measurement. [0094] In some embodiments, the JSW measurement is measured based on longitudinal changes in a minimum JSW measurement. In some embodiments, the change in JSW of the JSN measurement is 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 mm, less than a change in a JSW of a baseline JSN measurement. In some embodiments, the therapeutically effective dose of the compound of Formula I decreases JSN by 0.01, 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.10, 0.11, 0.12, 0.13, 0.14, 0.15, 0.16, 0.17, 0.18, 0.19, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 mm, or more. For example, the therapeutically effective dose of the compound of Formula I may decrease JSN by 0.05-2.00 mm, 0.1-0.6 mm, 0.10-1.50 mm, or 0.20-1.00 mm. In some embodiments, the JSN measurement is a change in JSW over time. In some embodiments, the time of the change in JSW over time is 1 week, 2 weeks, 3 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months, 10 months, 11 months, 1 year, 2 years, 3 years, 4 years, 5 years, 6 years, 7 years, 8 years, 9 years, or 10 years. For example, the improvement may be a JSN measurement of 0.02 to 0.05 mm per year less than the baseline JSN measurement.

[0095] In some embodiments, the baseline joint architecture measurement is a baseline subchondral bone surface area measurement. In some embodiments, the joint architecture measurement is a subchondral bone surface area measurement. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an improvement in the subchondral bone surface area measurement compared to the baseline subchondral bone surface area measurement. In some embodiments, the subchondral bone surface area measurement and the baseline subchondral bone surface area measurement are measured using X-ray or MRI.

[0096] In some embodiments, the subchondral bone surface area measurement is a bone density measurement, and the improvement is an increase in the bone density measurement compared to a baseline bone density measurement. In some embodiments, the improvement is a 50

Hounsfield Unit (HU), 100 HU, 150 HU, 200 HU, 250 HU, 300 HU, 350 HU, 400 HU, 450 HU, 500 HU, or more, increase in the bone density measurement relative to the baseline bone density measurement. In some embodiments, the subchondral bone surface area measurement is a number of cystic areas compared to a baseline number of cystic areas. In some embodiments, the improvement is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, less cystic areas than the baseline number of cystic areas. In some embodiments, the cystic areas comprise low-density bone areas.

[0097] In some embodiments, the baseline joint architecture measurement is a baseline KL grade. In some embodiments, the joint architecture measurement is a Kellgren-Lawrence (KL) grade. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an improvement in the KL grade compared to the baseline KL grade. In some embodiments, the improvement comprises a decrease in the KL grade relative to the baseline KL grade by one KL grade or more. In some embodiments, the improvement is a decrease in the KL grade relative to the baseline KL grade by half a KL grade. In some embodiments, the improvement is a decrease in the KL grade relative to the baseline KL grade by one KL grade.

In some embodiments, the improvement is a decrease in the KL grade relative to the baseline KL grade by two KL grades. In some embodiments, the improvement is a decrease in the KL grade relative to the baseline KL grade by three KL grades. In some embodiments, the improvement is a decrease in the KL grade relative to the baseline KL grade by four KL grades. In some embodiments, the improvement is a decrease in the KL grade relative to the baseline KL grade by 0.5-4 KL grades.

[0098] In some embodiments, the baseline joint architecture measurement is a baseline progression through the KL grading scale. In some embodiments, the joint architecture measurement is a progression through a KL grading scale. In some embodiments, the therapeutically effective dose of the compound of Formula I results in an improvement in the progression through the KL grading scale compared to the baseline progression through the KL grading scale. In some embodiments, the improvement is a decrease in a rate of the progression through the KL grading scale relative to a rate of the baseline progression through the KL grading scale grade. In some embodiments, the improvement is a prevention of the progression through the KL grading scale. In some embodiments, the improvement is assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, or 5 years or more after administration of the therapeutically effective dose of the compound of Formula I. In some embodiments, the improvement a decrease in the rate of progression through the KL grading scale, relative to the baseline rate of progression through the KL grading scale, by 0.1 KL grades per year, 0.25 KL grades per year, 0.5 KL grades per year, 1 KL grade per year, 1.5 KL grades per year, or 2 KL grades per year.

[0099] In some embodiments, efficacy of the therapeutically effective dose of the compound of Formula I is measured by an inhibition of progression of a patient’s Kellgren-Lawrence (KL) grade, or an improvement of a patient’s KL grade as compared to a patient’s baseline KL grade as assessed prior to administering to the joint a therapeutically effective dose of the compound of Formula I over a period of time. In some embodiments, a therapeutically effective dose of the compound of Formula I to the joint of a patient diagnosed with osteoarthritis inhibits

progression of a patient’s KL grade from KL grade 1 to 2, or from KL grade 2 to 3, or from KL grade 3 to 4 as assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, 5 years or more after administration of the therapeutically effective dose of the compound of Formula I. In some embodiments, a therapeutically effective dose of the compound of Formula I to the joint of a patient diagnosed with osteoarthritis improves or reverses a patient’s KL grade by one KL grade, by two KL grades, by three KL grades, by four KL grades as assessed 3 months or more, 4 months or more, 5 months or more, 6 months or more, 7 months or more, 8 months or more, 9 months or more, 10 months or more, 11 months or more, 12 months or more, 13 months or more, 14 months or more, 15 months or more 16 months or more, 17 months or more, 18 months or more, 2 years or more, 2.5 years or more, 3 years or more, 3.5 years or more, 4 years or more, 4.5 years or more, 5 years or more after administration of the therapeutically effective dose of the compound of Formula I.

Pharmaceutical Compositions

[00100] In some embodiments, the compound of Formula I is formulated into one or more pharmaceutical compositions. Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that are used pharmaceutically. A summary of pharmaceutical compositions described herein is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H.A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkins 1999), herein incorporated by reference for such disclosure.

[00101] In general, the compound of Formula I is synthesized according to organic synthesis techniques known to those skilled in this art, starting from commercially available chemicals and/or from compounds described in the chemical literature. Specific and analogous reactants may also be identified through the indices of known chemicals prepared by the Chemical Abstract Service of the American Chemical Society, which are available in most public and university libraries, as well as through on-line databases (the American Chemical Society, Washington, D.C., may be contacted for more details). Chemicals that are known but not commercially available in catalogs may be prepared by custom chemical synthesis houses, where many of the standard chemical supply houses ( e.g ., those listed above) provide custom synthesis services. A reference for the preparation and selection of pharmaceutical salts of the present disclosure is P. H. Stahl & C. G. Wermuth“Handbook of Pharmaceutical Salts,” Verlag Helvetica Chimica Acta, Zurich, 2002. Methods known to one of ordinary skill in the art may be identified through various reference books and databases. Suitable reference books and treatises detail the synthesis of reactants useful in the preparation of compounds described herein, or provide references to articles that describe the preparation.

EXAMPLES

[00102] The following illustrative examples are representative of embodiments of compositions and methods described herein and are not meant to be limiting in any way.

Example 1: Clinical Study

[00103] The following describes a clinical study to evaluate safety, tolerability, and pharmacokinetics of a single intra-articular injection of a senolytic in patients diagnosed with painful osteoarthritis of the knee.

[00104] The study is a randomized, double-blind, placebo-controlled single ascending dose study to evaluate safety, tolerability, and pharmacokinetics of the compound of Formula I, in patients diagnosed with painful femoro-tibial osteoarthritis. The study consists of 2 parts: the first part (Part A) is a single ascending dose study, whereas the second part (Part B) is a single dose study at a dose that has been determined to be safe and tolerable in first part of the study (see FIG. 1).

[00105] In Part A, 48 subjects were randomly allocated to receive the compound of Formula I (between 0.1 mg to 4 mg) or placebo in 3 : 1 randomization by dose level (cohort), whereas in Part B, 30 subjects were randomly allocated to receive the compound of Formula I (4 mg dose) or placebo in a 2: 1 randomization (see FIG. 12 and FIG. 13).

[00106] The primary objective was to establish the safety and tolerability of the compound of Formula I given as a single intra-articular injection into the femoro-tibial joint of patients with osteoarthritis as evaluated by the incidence of serious and non-serious adverse events within a 12 week timeframe.

[00107] For Part A: Plasma concentration of the compound of Formula I over 24 hrs following a single intra-articular injection was monitored. Primary endpoints were safety and tolerability. Secondary and exploratory endpoints included plasma pharmacokinetics, synovitis as measured by MRI, pain, and SASP factors in synovial fluid and plasma. The change from baseline to Week 12 of the weekly mean of the average daily pain intensity score as obtained using the 11 -point numerical rating scale, as measured from 0-10, where 0 represents no pain and 10 represents worst pain imaginable. In addition, the change from baseline to Week 12 for the Western Ontario and McMaster Universities (WOMAC) index total score and the pain and function subscales using the 5-point Likert scale were also measured.

[00108] For Part B: The change from baseline to Week 4 of selected senescence- associated secretory phenotype (SASP) factors in plasma samples and in synovial fluid aspirates in patients receiving a single intra-articular injection of the compound of Formula I versus those receiving placebo, were measured. Primary endpoints were safety and tolerability. Secondary endpoints included changes in the levels of SASP factors present in synovial fluid and plasma, and pain. Synovial fluid samples were obtained at baseline and four weeks post-treatment.

Further, the change from baseline to Week 4 for the WOMAC index pain subscale using as derived from the Knee injury and Osteoarthritis Outcome Score (KOOS) were also measured. Finally, patient plasma concentration of the compound of Formula I following a single intra- articular injection was measured within 24 hours of treatment.

[00109] Patient Inclusion Criteria: Patients must be > 40 and < 85 years of age. Patients using non-steroidal anti-inflammatory agent must be on a consistent regimen, dose, and medication for at least 8 weeks prior to receiving treatment.

1. For the first part of the study only: Ambulatory persons with osteoarthritis of the knee and baseline pain with a mean of > 4 and < 9 points on the 24-hour mean pain score (on the 11 -point Numeric Rating Scale) for at least five of the seven days during the screening period.

2. For the second part of the study only: Ambulatory persons with painful osteoarthritis of the knee for at least six months. Patients being studies in this second part with OA must have Kellgren-Lawrence (K-L) scores 1 through 4 in the target knee. Patients must also have a baseline pain >6 on the WOMAC Index pain subscale as derived from the KOOS.

[00110] Patient Exclusion Criteria: Any condition, including laboratory findings and findings in the medical history or in the pre-study assessments, that in the opinion of the health care professional constitutes a risk or contraindication for treatment. Also excluded are any patients with clinically significant co-existing conditions of the cardiovascular, renal, gastrointestinal, respiratory, nervous, metabolic, or hematologic/ immune systems, as judged by their health care professional. Further, any patients with a history of diabetes mellitus according to the American Diabetes Association criteria or patients previously diagnosed by a qualified physician as having diabetes (American Diabetes Association Standards of Medical Care in Diabetes 2016) are also excluded from treatment. Any patients with a history of cardiac rhythm disturbances, abnormal ECG intervals, or use of medications known to impact ECG intervals are excluded. Patients who have undergone diagnostic arthroscopy of the target knee in the previous six months and those patients who have undergone arthroscopic surgery in the last two years prior to the screening visit (including microfracture and menisectomy) on the target knee are both also excluded from treatment. Finally, any patient anticipated to have arthroscopic surgery on either knee at any time or a history of previous total or partial knee arthroplasty in either knee are excluded from treatment.

[00111] Exclusion criteria for the first part only: Patients with an effusion at the screening visit which, in the opinion of the health care professional following examination and discussions with the patient, requires drainage for symptom relief are excluded from treatment.

[00112] Also, patients who have received intra-articular treatment with steroids or hyaluronic acid derivatives within the last 16 weeks prior to screening are excluded from treatment. Also excluded are those patients who use, or have used in the 8 weeks prior to screening, opioid analgesics (with exception of mild opioid analgesics). Patients who have had regenerative joint procedures on any joint including, but not limited to, platelet-rich plasma injections, mesenchymal stem cell transplantation, autologous chondrocyte transplantation, or mosaicplasty are also excluded from treatment. Further, patients with a current or history of other joint diseases such as joint dysplasia, crystal -induced arthropathy (such as gout, or calcium pyrophosphate deposition disease evidenced by clinical and/or radiographic means), aseptic osteonecrosis, acromegaly, Paget's disease, Ehlers-Danlos Syndrome, Gaucher's disease, Stickler's syndrome, joint infection, hemophilia, hemochromatosis, or neuropathic arthropathy of any cause, are also excluded from treatment.

[00113] Any active known or suspected systemic autoimmune disease (except for vitiligo, residual auto-immune hypothyroidism only requiring hormone replacement, psoriasis not requiring systemic treatment for two years, conditions not expected to recur in the absence of an external trigger) or any history of a systemic inflammatory arthritis such as psoriatic, rheumatoid, ankylosing spondylitis or reactive arthritis will prevent a patient from receiving treatment. Further, patients diagnosed presently and symptomatic with fibromyalgia based on American College of Radiology (ACR) Criteria are also excluded from treatment. Finally, patients with a BMI > 40 kg/m2, or whose size exceed the limits of the of the MRI equipment (coil and gantry) will be excluded from treatment.

Results

[00114] Safety Tolerability and PK In Part A, the compound of Formula I was well tolerated up to the maximum administered dose of 4 mg (See FIG. 2). There were no serious adverse events and no patients discontinued because of an adverse event. There were no dose- dependent adverse events or clinical laboratory findings. The majority (66%) of adverse events were mild. In Part B, the compound of Formula I was well tolerated at the 4 mg dose. There were no serious adverse events and no patients discontinued because of an adverse event. The majority (75%) of adverse events were mild and there were no relevant clinical laboratory findings. The compound of Formula I demonstrated dose-proportional plasma pharmacokinetics. Model-based estimates suggested that doses at or above 1 mg may be pharmacologically active. This informed the low dose (0.1, 0.2, and 0.4 mg) and high dose (1, 2, and 4 mg) groupings for analyses.

[00115] Pain Measures. Numerical Rating Scale (NRS): In Part A, evaluation of pain by NRS (0-10 point scale) measured at 12 weeks demonstrated a dose-dependent and clinically meaningful reduction. The range of mean baseline values was between 5.90 to 6.76.

Table 4. Pain Measure as Assessed by Numerical Rating Scale

CFBL=change from baseline; Pbo-Adj=placebo adjusted; low doses=0.1, 0.2, and 0.4 mg; high doses=l, 2, and 4 mg; Tx=treated

WOMAC-A: In Part A, evaluation of pain by WOMAC-A mean item score (0-4 scale) measured at 12 weeks demonstrated a dose-dependent and clinically meaningful reduction. The range of mean baseline values was between 1.80 to 2.36. Part A WOMAC-A pain data are shown in FIG. 3, FIG. 4 and FIG. 9B. Part A NRS pain data are shown in FIG. 5, FIG. 6 and FIG. 9A. The data indicate that a single dose of the compound of Formula I decreased pain, and that the decrease was durable, dose-dependent, and substantial.

Table 5. Pain Measure as Assessed by WOMAC-A

CFBL=change from baseline; Pbo-Adj=placebo adjusted; low doses=0.1, 0.2, and 0.4 mg; high doses=l, 2, and 4 mg; Tx=treated

[00116] In Part B, pain, as measured by WOMAC-A mean item score measured at 4 weeks, showed a numerical reduction that was not statistically different from placebo (FIG. 15).

[00117] Other Measures. Evaluation of function by WOMAC-C mean item score (0-4 scale) demonstrated a dose-dependent and clinically meaningful improvement in patients in the high-dose group. Part A WOMAC-C physical function data are shown in FIG. 7, FIG. 8 and FIG. 9C. The data indicate durable, dose-dependent, and substantial improved effects of a single dose of the compound of Formula I on physical function. There were no observed changes in stiffness as measured by WOMAC-B. Evaluation of Patient Global Impression of Change (PGIC) with treatment demonstrated a higher proportion of patients being“much improved” or “very much improved” versus placebo (FIG. 14).

[00118] Exploratory Measures. Synovial Inflammation by MRI: Evaluation of synovial inflammation by MRI showed a variable response across the dose groups. The two lowest doses showed no significant changes. The two intermediate doses showed significant increases (0.4 mg = +1.80, p < 0.01 and 1 mg = +3.69, p < 0.01), whereas the two highest doses showed significant decreases in synovial inflammation (2 mg = -1.18, p < 0.01 and 4 mg = -2.49, p < 0.01).

[00119] SASP Factors: In Part A, an insufficient number of matched samples were collected due to a lack of withdrawable synovial fluid from patients. Therefore, an analysis of change in SASP from factors from baseline to 12 weeks was not performed. In Part B, 19 biomarkers were analyzed across 20 matched pair samples (FIG. 10). Trends in changes to the levels of particular SASP factors present in synovial fluid (treatment versus placebo) were consistent with senescent cell removal. The SASP factors that changed their prevalence in synovial fluid after the compound of Formula I treatment were MMP-10, MMP-12, MMP-13, IL-6, IL-10, CCL20, A2M, and VEGF-C.

Example 2: Phase 2 Trial

[00120] A second clinical study will be conducted to assess effects of the compound of Formula I on moderate to severe painful knee osteoarthritis (OA). This Phase 2 study builds on the favorable clinical responses, pharmacokinetic (PK) and safety results obtained in Example 1, and assesses the efficacy, safety, and tolerability of a single-dose IA administration of the compound of Formula I in symptomatic knee OA.

[00121] The study includes a randomized, double-blind, placebo-controlled, single-dose, parallel-group study to assess the efficacy, safety, and tolerability of a single-dose intra-articular (IA) administration of the compound of Formula I in patients with moderate to severe painful knee OA (for example, established femoro-tibial OA).

[00122] The study includes approximately 180 total patients > 40 and < 85 years of age with moderate to severe painful knee OA. Approximately 45 patients are randomly (1 : 1 : 1 : 1) included in each of the following 4 groups:

Group 1 : Placebo

Group 2: 0.5 mg of the compound of Formula I

Group 3: 2.0 mg of the compound of Formula I

Group 4: 4.0 mg of the compound of Formula I

[00123] The 4 groups are enrolled concurrently. Approximately 40 patients per group complete a Week 12 visit, assuming a 10% dropout rate. A Week 24 visit is also included. Either the compound of Formula I or placebo is administered as a single 8 mL IA injection on Day 1 of Week 0. The endpoint analyses include changes from baseline to Weeks 12 and 24 in the patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo (see FIG. 11).

[00124] Inclusion Criteria: each patient satisfies the following criteria:

1. Patients who are ambulatory with a diagnosis of OA of the knee, as defined by the

American College of Rheumatology (ACR) Criteria (based on clinical and radiologic ACR criteria for at least 6 months) and who have baseline pain with a mean of > 4 and < 9 on the 11 -point (0-10) average daily pain NRS for at least five of seven days during the Screening period.

2. Patients with a KL grade of 1-4 based on central reading of a weight-bearing radiograph of target knee.

3. Patients aged > 40 and < 85 years.

4. Patients are permitted but not required to use an oral NSAID, serotonin and

norepinephrine reuptake inhibitors (SNRIs) such as Cymbalta ® (duloxetine), tramadol, or acetaminophen, provided that they have been taking a stable dose and regimen of medication for at least 4 weeks prior to Screening.

5. Patients with type 2 diabetes mellitus can be included as long as they are under adequate control (screening hemoglobin Ale < 8.0) and have no evidence or history of diabetic neuropathy.

6. Patients who have the capacity to give informed consent and who are willing and able to comply with all study-related procedures and assessments. Patients who do not have the legal capacity or medical competency to give written informed consent are ineligible for this study; consent via legally authorized representative is not accepted.

[00125] Exclusion Criteria: Patients are ineligible if they meet any of the following criteria:

1. Patients with any condition, including laboratory or imaging findings and findings in the medical history or in the pre-study assessments, that in the opinion of the Investigator or the Medical Monitor constitutes a risk or contraindication for participation in the study or that could interfere with the study objectives, conduct, or evaluation or prevent the patient from fully participating in all aspects of the study.

2. Patients with a body mass index (BMI) > 40 kg/m 2 or a body habitus that precludes the MRI.

3. Patients with clinically significant co-existing conditions of the cardiovascular, renal, gastrointestinal, respiratory, nervous (e.g., neuropathy), metabolic, hematologic, or immune system.

4. Patients with fibromyalgia based on ACR criteria.

5. Patients with any active, known, or suspected systemic autoimmune disease with

musculoskeletal involvement (except for vitiligo, residual autoimmune hypothyroidism requiring only hormone replacement, psoriasis not requiring systemic treatment for 2 years, or conditions not expected to recur in the absence of an external trigger) or any history of a systemic inflammatory arthritis, such as rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis or spondylitis, or reactive arthritis.

Patients who have received IA treatment in the target knee with steroids or hyaluronic acid derivatives within the last 16 weeks prior to Screening, or with extended-release corticosteroid (e.g., Zilretta®) within the last 20 weeks prior to Screening.

Patients who are using a topical NSAID or topical analgesics on the target knee.

Patients who are receiving anticoagulants (e.g., low molecular weight heparin or warfarin), factor Xa inhibitors (e.g., Eliquis®/apixaban), or P2Y 12 inhibitors (e.g., Plavix®/clopidogrel or ticagrelor).

Patients who are receiving chronic oral corticosteroids.

Patients who have used opioid analgesics, marijuana or marijuana-derived products (e.g., cannabidiol), and topical capsaicin on the target knee within 8 weeks prior to Screening and at all times during the study.

Patients with a history of traumatic knee injury to the target knee, including, but not limited to, patients with meniscal root tear, within 2 years of study entry.

Patients who have undergone diagnostic arthroscopy to the target knee in the previous 6 months.

Patients who have undergone arthroscopic surgery (including microfracture and meniscectomy) on the target knee in the last 2 years prior to the Screening visit or are anticipated to have arthroscopic surgery on either knee at any time during the study period.

Patients with a history of previous total or partial knee arthroplasty.

Patients with an effusion at the Screening visit, which, in the opinion of the Investigator following examination and discussions with the patient, requires drainage for symptom relief.

Patients who have had regenerative joint procedures on any joint, including, but not limited to, platelet-rich plasma injections, stem cell transplantation, autologous chondrocyte transplantation, or mosaicplasty.

Patients with abnormal alanine aminotransferase or aspartate aminotransferase > 2 c upper limit of normal (ULN) at Screening.

Patients with bilirubin > 2 x ULN at Screening.

Patients with renal dysfunction as defined by a glomerular filtration rate < 30

mL/min/1.73 m 2 by laboratory testing (Chronic Kidney Disease Epidemiology

Collaboration method). Patients with active or untreated hepatitis B or C or any history of human

immunodeficiency virus infection at Screening.

Patients with any known active infections, including suspicion of IA infection and/or predisposition to infections due to immune system compromise.

Patients with a history of malignancy within 5 years and/or current evidence of active malignancy, except basal cell carcinoma, squamous cell carcinoma of the skin, and completely excised cervical intraepithelial neoplasia.

Patients with a history of drug or alcohol dependence within the last 3 years.

Patients with secondary arthritis that involves the target knee or would confound assessments of knee OA, e.g., joint dysplasia, chronic or recurrent crystal-induced arthritis (gout, calcium pyrophosphate deposition disease, podagra, etc.), aseptic osteonecrosis, acromegaly, Paget’s disease, Ehlers-Danlos syndrome, Gaucher’s disease, Stickler’s syndrome, joint infection, hemophilia, hemochromatosis, or neuropathic arthropathy of any cause.

Patients with radiographic (plain X-ray or MRI) evidence of disease that would impact the ability of the study to meet its primary or secondary objectives; examples include the following:

o Patients with a history of osteonecrosis or who are deemed to have or have risk factors for rapid progression of OA, including sub-chondral insufficiency fracture or spontaneous osteonecrosis of the knee

o Patients with recent fractures (stress, pathologic, or traumatic) defined by the presence of a visible fracture line

o Patients with cystic lesion 0.5 c the femoral or tibial subchondral plate o Patients with a history of pigmented villonodular synovitis

Patients who previously received treatment in study of the compound of Formula I. Patients who have received another investigational drug or investigational vaccine within the last 3 months prior to Screening.

Patients who are participating in or are planning to participate concurrently in another investigational drug or vaccine study.

Female patients who are pregnant, lactating, or of childbearing potential who do not agree to use highly effective methods of birth control (e.g., progesterone-only hormonal contraception, double barrier, or intrauterine device) for 3 months following the treatment. Postmenopausal females (> 45 years old and without menses for more than 1 year) and surgically sterilized females are exempt from these requirements. 30. Male patients who do not agree to use a highly effective method of contraception for 3 months following the treatment, if sexually active with a female partner of childbearing potential.

Results

[00126] A primary objective in this study is to evaluate the effect of IA administration of the compound of Formula I on the change from baseline to Week 12 of pain in the target knee. An endpoint for this objective is change from baseline to Week 12 of the Western Ontario and McMaster Universities Osteoarthritis Index pain subscale (WOMAC-A) score in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have improvements in their WOMAC-A scores that are greater than in the placebo group. Thus, each treatment with the compound of Formula I is expected to decrease knee pain within 12 weeks.

[00127] An additional objective is to evaluate the effect of IA administration of the compound of Formula I on the change from baseline to Week 12 in physical function. An endpoint for this objective is change from baseline to Week 12 of the Western Ontario and McMaster Universities Osteoarthritis Index function subscale (WOMAC-C) score in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have improvements in their WOMAC-C scores that are greater than in the placebo group. Thus, each treatment with the compound of Formula I is expected to increase knee function within 12 weeks.

[00128] An additional objective is to evaluate the effect of IA administration of the compound of Formula I on the change from baseline to Week 12 in pain reported by patients daily over the 12-week study period. An endpoint for this objective is change from baseline to Week 12 of the weekly mean of the average daily pain intensity scores on the 11 -point numeric rating scale (NRS) in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have improvements in their NRS that are greater than in the placebo group. Thus, each treatment with the compound of Formula I is expected to decrease knee pain within 12 weeks.

[00129] An additional objective is to evaluate the effect of IA administration of the compound of Formula I on the duration of symptom improvement (pain and function) following IA administration of the compound of Formula I out to 24 weeks. Endpoints for this objective include change from baseline (over the entire 24-week period, including both the primary study period and the 12-week follow-up period) out to Week 24 for the WOMAC-A, NRS, and WOMAC-C scores in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have improvements in each of their WOMAC-A, NRS, and WOMAC-C scores that are greater than in the placebo group. Thus, each treatment with the compound of Formula I is expected to decrease knee pain and increase knee function until at least 24 weeks after the treatment.

[00130] An additional objective is to evaluate the safety and tolerability of single-dose IA administration of the compound of Formula I. Endpoints for this objective include incidence of adverse events (AEs) as well as change from baseline through Weeks 12 and 24 in vital signs and selected laboratory safety parameters (as deemed clinically appropriate) in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. An AE for purposes of this study is any untoward medical occurrence associated with the treatment whether or not it is considered treatment related. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have a low incidence of AEs, and not to show serious adverse affects on their vital signs throughout the study period. Thus, each treatment with the compound of Formula I is expected to be safe and tolerable.

[00131] An additional objective is to explore the effects of single-dose IA administration of the compound of Formula I on the change from baseline in patient reported assessment of their osteoarthritis at the time of inquiry. Endpoints for this objective include change from baseline to Week 12 and to Week 24 in Patient Global Assessment score in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have improvements in their Patient Global Assessment scores that are greater than in the placebo group at Weeks 12 and 24. Thus, each treatment with the compound of Formula I is expected to improve osteoarthritis within 12 weeks and until at least 24 weeks after the treatment.

[00132] An additional objective is to explore the effects of single-dose IA administration of the compound of Formula I on the change from baseline in patient reported impression of how their osteoarthritis has changed over the course of the study. Endpoints for this objective include change from baseline to Week 12 and to Week 24 in Patient Global Impression of Change score in patients receiving a single 0.5, 2.0, or 4.0 mg dose of the compound of Formula I versus those receiving placebo. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have improvements in their reported impressions that are greater than in the placebo group. Thus, each treatment with the compound of Formula I is expected to improve a patient’s impression of how the patient’s osteoarthritis changes within 12 weeks and until at least 24 weeks after the treatment.

[00133] An additional objective is to explore the effects of single-dose IA administration of the compound of Formula I on soluble biomarkers and imaging biomarkers of joint degeneration and repair. An endpoint for this objective includes change from baseline out to Week 24 in serum/plasma and urine biomarkers of synovium, cartilage, and bone degeneration. Another endpoint for this objective is change from baseline to Week 24 in magnetic resonance imaging-based markers of joint structure. Patients receiving the 0.5, 2.0, and 4.0 mg doses are expected to have beneficial changes in serum/plasma and urine biomarkers of synovium, cartilage, and bone degeneration, and in magnetic resonance imaging-based markers of joint structure, that are greater than in the placebo group at Weeks 12 and 24. Thus, each treatment with the compound of Formula I is expected to improve soluble biomarker levels within and until at least 24 weeks after the treatment. Further, each treatment with the compound of Formula I is expected to improve imaging biomarkers of joint degeneration and repair within 12 weeks and until at least 24 weeks after the treatment.

[00134] Overall, the results are expected to indicate that the compound of Formula I is effective, safe, and tolerable in patients with moderate to severe painful knee OA. A single AI dose of the compound of Formula I at, for example, 0.5, 2, or 4 mg, is expected to be effective for achieving each of these results. Each of these results are expected within 12 weeks of treatment, and are expected to last until at least 24 weeks after treatment.