Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
LYOPHILIZED FORMULATION OF A MONOCLONAL ANTIBODY AGAINST TRANSTHYRETIN
Document Type and Number:
WIPO Patent Application WO/2019/108689
Kind Code:
A1
Abstract:
The invention provides antibody formulations and methods useful for prophylaxis or treatment of transthyretin-related amyloidosis.

Inventors:
SOTO JAY (US)
HAWE ANDREA (DE)
TANTIPOLPHAN RUEDEEPORN (DE)
HEINDL STEFAN (AT)
Application Number:
PCT/US2018/062902
Publication Date:
June 06, 2019
Filing Date:
November 28, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PROTHENA BIOSCIENCES LTD (IE)
SOTO JAY (US)
International Classes:
C07K16/18; A61K39/00; A61K51/10
Domestic Patent References:
WO2016120810A12016-08-04
Foreign References:
US20160340420A12016-11-24
Other References:
See also references of EP 3717512A4
Attorney, Agent or Firm:
LIEBESCHUETZ, Joe et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A pharmaceutical formulation comprising:

(a) a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:61 and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Kabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:l and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16, wherein the antibody is present at a concentration within the range from about 25 mg/mL to about 75 mg/mL;

(b) a buffer present at a concentration of about 20 mM, wherein the buffer is citrate, histidine, phosphate or succinate;

(c) either a sugar present at a concentration within the range from about 205 mM to about 260 mM, wherein the sugar is sucrose or trehalose or, if the sugar is absent about 160 mM arginine is present, and

(d) a surfactant present at a concentration within the range from about 0.01% to about 1 % by weight; wherein the formulation is characterized by a pH within the range from about 5.0 to about 6.5; provided that:

(i) if histidine or succinate buffer is present, the pH is about 6.0;

(ii) if phosphate buffer is present, the pH is about 6.5; and

(iii) if histidine and trehalose are present the surfactant is a. PS80; or b. PS20, provided that if PS20 is present, about 25 mM L-arginine is also present.

2. The formulation of claim 1, that is essentially free of mannitol or sorbitol.

3. The formulation of claim 1 , wherein the buffer is histidine.

4. The formulation of claim 3, wherein the sugar is present in a range from about 230 mM to about 250 mM

5. The formulation of claim 4, wherein the sugar is present at about 230-240 mM.

6. The formulation of claim 5, wherein the sugar is sucrose and the surfactant is PS 20 or PX188.

7. The formulation of claim 6, wherein the surfactant is PS20 at a concentration of 0.02% w/w.

8. The formulation of claim 7, wherein the surfactant is PX188 at a concentration of 0.04% w/w.

9. The formulation of claim 5, wherein the sugar is trehalose.

10. The formulation of claim 1, wherein 160 mM arginine is present.

11. The formulation of claim 1, wherein the buffer is citrate.

12. The formulation of claim 11, wherein the sugar is present at 230 mM.

13. The formulation of claim 12, wherein the surfactant is 0.02% PS20.

14. The formulation of claim 1, wherein the buffer is phosphate.

15. The formulation of claim 14, wherein the sugar is present and is sucrose.

16. The formulation of claim 1, wherein the buffer is succinate.

17. The formulation of claim 16, wherein the sugar is present and is sucrose.

18. The formulation of claim 4, wherein the sugar is present and is trehalose.

19. The formulation of claim 18, wherein the trehalose is present at a concentration of 205 mM.

20. The formulation of claim 4, wherein the surfactant is PS20.

21. The formulation of claim 1, comprising:

(a) 20 mM citrate, 230 mM trehalose and 0.02% w/w PS20 at pH 5;

(b) 20 mM histidine, 230 mM sucrose and 0.02% w/w PS20;

(c) 20 mM phosphate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5;

(d) 20 mM citrate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5;

(e) 20 mM histidine, 230 mM trehalose and 0.02% w/w PS80;

(f) 20 mM histidine, 0.02% w/w PS20 and 160 mM L-arginine;

(g) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188;

(h) 20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or

(i) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L- arginine.

22. The formulation of claim 21, wherein the formulation consists essentially of the antibody and about:

(a) 20 mM citrate, 230 mM trehalose and 0.02% w/w PS20 at pH 5;

(b) 20 mM histidine, 230 mM sucrose and 0.02% w/w PS20;

(c) 20 mM phosphate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5;

(d) 20 mM citrate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (e) 20 mM histidine, 230 mM trehalose and 0.02% w/w PS80;

(f) 20 mM histidine, 0.02% w/w PS20 and 160 mM L-arginine;

(g) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188;

(h) 20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or

(i) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L- arginine.

23. The formulation of claim 22, wherein the formulation consists essentially of the antibody and about:

(a) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188;

(b) 20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or

(c) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L- arginine.

24. The formulation of any of the preceding claims, wherein the antibody comprises a mature heavy chain variable region comprising the amino acid sequence of SEQ ID NO:65, and a mature light chain variable region comprising the amino acid sequence of SEQ ID NO:76.

25. The formulation of claim 24, wherein the formulation consists essentially of the antibody at about 50 mg/ml, and about 20 mM histidine, about 240 mM sucrose and about 0.04% w/w PX188.

26. The formulation of any of the preceding claims, wherein the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO:6l, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO:70 except that positions H52 and L26 by Kabat numbering can each be independently N or S.

27. The formulation of any of claims 1-25, wherein the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO: 1, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO: 16.

28. The pharmaceutical formulation of any of the preceding claims, wherein the monoclonal antibody is a humanized, chimeric or veneered antibody.

29. The formulation of claim 28, wherein the monoclonal antibody is humanized.

30. The formulation of claim 29, wherein the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:64-66 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:74- 76.

31. The formulation of claim 29, wherein the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:5-l2 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs: l9- 23.

32. The formulation of claim 30, wherein the mature heavy chain has an amino acid sequence comprising SEQ ID NO:65 and the mature light chain has an amino acid sequence comprising SEQ ID NO:76.

33. The formulation of claim 31, wherein the mature heavy chain has an amino acid sequence comprising SEQ ID NO: 11 and the mature light chain has an amino acid sequence comprising SEQ ID NO: 19.

34. The humanized antibody of any one of claims 28-33, wherein the mature heavy chain variable region is fused to a heavy chain constant region and the mature light chain variable region is fused to a light chain constant region.

35. The humanized antibody of claim 34, wherein the mature heavy chain variable region is fused to a heavy chain constant region having the sequence of SEQ ID NO: 103 provided the C-terminal lysine can be absent and/or the mature light chain variable region is fused to a light chain constant region having the sequence of SEQ ID NO: 104 or 105.

36. The formulation of any preceding claim, wherein the monoclonal antibody is present at a concentration of about 50 mg/mL.

37. The formulation of any preceding claim, wherein the histidine buffer is at a concentration of about 20 mM.

38. The formulation of any preceding claim, wherein the pH is about 5.75 to

6.25.

39. The formulation of any preceding claim, wherein the sugar/polyol is sucrose present at a concentration of about 240 mM.

40. The formulation of any preceding claim, wherein the poloxamer is polo amer 188 at a concentration of about 0.04% by weight.

41. The formulation of any preceding claim, wherein less than about 5% or 3% by weight of the antibody is present as an aggregate in the formulation.

42. The formulation of any preceding clam wherein at least 95% or 97% of antibody by weight runs as a single peak under HP-SEC analysis.

43. The formulation of any one of claims 1-42, which is sterile.

44. The formulation of any one of claims 1-43, which is stable on freezing and thawing.

45. The formulation of any one of claims 1-44, which has an osmolality of from about 270 mOsmol/kg to about 330 mOsmol/kg.

46. A lyophilized formulation of an antibody, comprising

(a) a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:61 and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Kabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:l and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16;

(b) histidine;

(c) a sugar/polyol; and

(d) a poloxamer.

47. The lyophilized formulation of claim 46 prepared by lyophilizing the formulation of any of claims 1-45.

48. The lyophilized formulation of claim 46, which is reconstitutable with water to a pH of between about 5.5 to about 6.5.

49. The lyophilized formulation of claim 46, which is reconstitutable with water to a pH of about 6.0.

50. The lyophilized formulation of claim 46, containing about 10 mg to about 40 mg of the antibody.

51. A lyophilized formulation, which is reconstitutable with water to yield an aqueous solution comprising:

(a) an antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:6l and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Kabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:l and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16 wherein the antibody is present at a concentration within the range from about 25 mg/mL to about 75 mg/mL; (b) buffer present at a concentration within a range from about 10 mM to about 30 mM;

(c) sugar/polyol present at a concentration within a range from about 220 mM to about 260 mM;

(d) surfactant present at a concentration within a range from about 0.01% to about 0.1% by weight; and

(e) a pH within a range from about 5.5 to about 7.0.

52. The lyophilized formulation of claim 51, wherein the antibody comprises a mature heavy chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:64-66 and a mature light chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:74-76, or an antibody comprising a mature heavy chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:5-12 and a mature light chain variable region having an amino acid sequence comprising any one of SEQ ID NOs: 19-23.

53. The lyophilized formulation of claim 52, which is reconstitutable with water so that the antibody is present at a concentration of about 50 mg/mL.

54. The lyophilized formulation of any of claims 51-53, wherein the buffer comprises histidine.

55. The lyophilized formulation of any of claims 51-54, which is reconstitutable with water so that the pH is about 6.0.

56. The lyophilized formulation of any of claims 51-55, wherein the sugar/polyol is sucrose.

57. The lyophilized formulation of any of claims 51-56, wherein the surfactant is poloxomer.

58. The lyophilized formulation of claim 57, wherein the poloxomer is

PX188.

59. The lyophilized formulation of claim 51, wherein:

(a) the antibody comprises a mature heavy chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:64-66 and a mature light chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:74-76 and is reconstitutable to a concentration of about 50 mg/ml;

(b) the buffer is histidine and is reconstitutable to a concentration of about 20 mM;

(c) the sugar/polyol is sucrose and is reconstitutable to a concentration of about 240 mM; and

(d) the surfactant is poloxomer 188 and is reconstitutable to a concentration of about 0.04% by weight.

60. The lyophilized formulation of claim 59, which is reconstitutable with water so that the pH is about 6.0.

61. The lyophilized formulation of claim 60, wherein the antibody comprises a mature heavy chain variable region having an amino acid sequence comprising SEQ ID NO:65 and a mature light chain variable region having an amino acid sequence comprising SEQ ID NO:76.

62. The lyophilized formulation of claim 61, wherein the mature heavy chain variable region is fused to a heavy chain constant region having the sequence of SEQ ID NO: 103 provided the C-terminal lysine can be absent and the mature light chain variable region is fused to a light chain constant region having the sequence of SEQ ID NO: 104

63. The lyophilized formulation of any of the preceding claims, wherein at least 95 or 97 % of the antibody runs as a single peak under HP-SEC after storage for up to 3 months at 2-8 °C.

64. A method of reconstituting the lyophilized formulation of any of claims 46-63 comprising: combining the lyophilized formulation with sterile water to produce a liquid formulation.

65. The method of claim 64, further comprising introducing the reconstituted formulation into a bag of isotonic fluid for infusion.

66. The method of claim 64, wherein the lyophilized formulation is in powder form or in the form of a solid foam in a vial.

67. A sterile lyophilized dosage form of an antibody formulation in a 20 ml vial consisting essentially of:

(i) an antibody within a range of about 225-275 mg;

(ii) histidine within a range of about 15-19 mg;

(iii) poloxamer PX188 within a range of about 2-2.5 mg; and;

(iv) sucrose within a range of about 400-490 mg; wherein the antibody is a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:6l and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Rabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO: 1 and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16.

68. The lyophilized dosage form of claim 67, wherein the vial has contents consisting essentially of:

(i) about 250 mg of the antibody;

(ii) about 16.8 mg of L-histidine;

(iii) about 2.2 mg of poloxomer PX188; and

(iv) about 445.3 mg of sucrose.

69. The lyophilized dosage form of claim 67 or 68, wherein the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO:6l, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO:70 except that positions H52 and L26 by Kabat numbering can each be independently N or S.

70. The lyophilized dosage form of claim 67 or 68, wherein the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO: 1, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO: 16.

71. The lyophilized dosage form of claim 67 or 68, wherein the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:64-66 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:74-76.

72. The lyophilized dosage form of claim 67 or 68, wherein the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:5-l2 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:l9-23.

73. The lyophilized dosage form of claim 67 or 68, wherein the mature heavy chain has an amino acid sequence comprising SEQ ID NO:65 and the mature light chain has an amino acid sequence comprising SEQ ID NO:76.

74. The lyophilized dosage form of claim 67 or 68, wherein the mature heavy chain has an amino acid sequence comprising SEQ ID NO: 11 and the mature light chain has an amino acid sequence comprising SEQ ID NO: 19.

75. The lyophilized dosage form of claim 67 or 68, wherein the mature heavy chain variable region is fused to a heavy chain constant region having the sequence of SEQ ID NO: 103 provided the C-terminal lysine can be absent and/or the mature light chain variable region is fused to a light chain constant region having the sequence of SEQ ID NO: 104 or 105.

76. The lyophilized dosage form of claim 67 or 68, wherein the mature heavy chain has the sequence of SEQ ID NO:82, except the C-terminal lysine may be absent, and the mature light chain has the sequence of SEQ ID NO:86.

77. A method of preparing the lyophilized dosage form of any of claims 67-76 for administration to a subject, comprising:

(i) reconstituting the antibody formulation to a volume of about 5.0 mL with sterile water, and

(ii) diluting the reconstituted antibody formulation of step (i) in normal saline for infusion.

78. The method of claim of 77, wherein the total volume for infusion is 250 mL.

79. The method of claim of 77, wherein the total volume for infusion is 100 mL.

80. The method of claim of 77, wherein the total volume for infusion is 500 mL.

81. A reconstituted formulation resulting from reconstituting the lyophilized formulation of any of claims 46-80.

82. The reconstituted formulation of claim 81, comprising the antibody at a concentration of about 50 mg/mL, the histidine buffer at a concentration of about 20 mM, poloxamer at a concentration of about 0.04% by weight, and at pH of about 6.0.

83. The reconstituted formulation of claim 81 or 82, wherein at least 95, 96, 97, 98 or 99% of the antibody runs as a single peak on high pressure size exclusion

chromatography.

84. A method of treating or effecting prophylaxis of a subject having or at risk of a transthyretin-mediated amyloidosis, comprising administering to the subject an effective regime of the pharmaceutical formulation of any one of claims 1-23 or a reconstituted form of the lyophilized formulation of any one of claims 46-63.

85. The method of claim 84, wherein the subject has been previously treated with a TTR tetramer stabilizer, an antisense oligonucleotide based therapy, an RNA interference (RNAi) based therapy, or an amyloid degrader.

86. The method of claim 85, wherein the subject no longer receives the treatment with the TTR tetramer stabilizer, antisense oligonucleotide based therapy, RNA interference (RNAi) based therapy, or amyloid degrader.

87. The method of claim 84, wherein the subject is concurrently receiving treatment with diflunisal.

88. The method of claim 84, wherein the subject is concurrently receiving treatment with tafamidis.

89. The method of claim 84, wherein the subject is concurrently receiving treatment with a TTR tetramer stabilizer, an antisense oligonucleotide based therapy, an RNA interference (RNAi) based therapy, or an amyloid degrader.

90. The method of claim 89, wherein the subject is concurrently receiving treatment with tafamidis, difluisal, patisiran, inotersen, 4'-iodo-4'-deoxydoxorubicin (IDOX), doxycycline, tauroursodeoxycholic acid (TUDCA), cyclodextrin (CyD), or an anti-SAP antibody.

91. The method of claim 84, wherein the subject has been diagnosed with ATTR amyloidosis.

92. The method of claim 84, wherein the subject has wild-type ATTR- cardiomyopathy.

93. The method of claim 84, wherein the subject has hereditary ATTR- cardiomyopathy.

94. The method of claim 84, wherein the subject has hereditary ATTR- polyneuropathy.

95. The method of claim 84, wherein the subject has ATTR cardiac involvement.

96. The method of claim 84, wherein the subject has ATTR amyloidosis peripheral neuropathy involvement.

97. The method of claim 84, wherein the pharmaceutical formulation, reconstituted form of the lyophilized formulation or the reconstituted form of the lyophilized antibody formulation is admini tered to the subject intravenously in diluted form.

98. The method of claim 97, wherein the diluent for the diluted form is normal saline.

99. The method of claim 98, wherein the total volume of the diluted form administered to the subject is at least about 100 mL.

100. The method of claim 99, wherein the total volume of the diluted form administered to the subject is at least about 250 mL.

101. The method of claim 100, wherein the total volume of the diluted form administered to the subject is at least about 500 mL.

102. The method of claim 100, wherein the total volume is about 250 mL.

103. The method of claim 97, wherein 0.1, 0.2, 0.3, 1, 3, 10 or 30 mg/kg of the antibody is administered to the subject about once every 28 days.

104. The method of claim 103, wherein 0.1, 0.2, 0.3, 1 or 3 mg/kg of the antibody is administered to the subject as an infusion over a range of about 60 to 120 minutes.

105. The method of claim 103, wherein 10 or 30 mg/kg of the antibody is administered to the subject as an infusion over a range of about 90 to 180 minutes.

106. The method of claim 97, wherein the subject is premedicated with an antihistamine.

107. The method of claim 97, wherein the subject is admini tered diphenhydramine within a range of about 30 to 90 minutes prior to the antibody administration.

108. The method of claim 107, wherein the subject is administered acetaminophen within a range of about 30 to 90 minutes prior to the antibody administration.

109. The method of claim 84, wherein the duration of the regime is at least 3 months.

110. The method of claim 109, wherein the duration of the regime is at least 12 months.

111. The method of claim 84, wherein the subject is receiving concomitant tafamidis.

112. A method of treating or effecting prophylaxis of a subject having or at risk of a transthyretin-mediated amyloidosis, comprising administering an effective regime of a TTR tetramer stabilizer, an antisense oligonucleotide based therapy, an RNA interference (RNAi) based therapy, or an amyloid degrader, wherein the subject has previously been treated with the pharmaceutical formulation of any one of claims 1 -23 or a reconstituted form of the lyophilized formulation of any one of claims 46-63.

113. The method of claim 112, wherein the subject no longer receives the treatment with the pharmaceutical formulation of any one of claims 1-45.

114. The method of claim 112, wherein the TTR tetramer stabilizer is tafamidis or diflunisal.

115. The method of claim 112, wherein the antisense oligonucleotide based therapy is inotersen.

116. The method of claim 112, wherein the RNA interference (RNAi) based therapy is patisiran or revusiran.

117. The method of claim 112, wherein the amyloid degrader is 4'-iodo-4'- deoxydoxorubicin (IDOX), doxycycline, tauroursodeoxycholic acid (TUDCA), cyclodextrin (CyD), or an anti-SAP antibody.

118. The method of claim 117, wherein the amyloid degraders are doxycycline in combined with tauroursodeoxycholic acid (TUDCA).

119. The method of claim 112, wherein the subject has been diagnosed with ATTR amyloidosis.

120. The method of claim 112, wherein the subject has wild-type ATTR- cardiomyopathy.

121. The method of claim 112, wherein the subject has hereditary ATTR- cardiomyopathy.

122. The method of claim 112 or 121, wherein the subject has hereditary ATTR-polyneuropathy.

123. The method of claim 112, wherein the subject has ATTR cardiac involvement.

124. The method of claim 112, wherein the subject has ATTR amyloidosis peripheral neuropathy involvement.

125. The method of claim 112, wherein the pharmaceutical formulation, reconstituted form of the lyophilized formulation or the reconstituted form of the lyophilized antibody formulation is administered to the subject intravenously in diluted form.

126. The method of claim 125, wherein the diluent for the diluted form is normal saline.

127. The method of claim 126, wherein the total volume of the diluted form administered to the subject is at least about 100 mL.

128. The method of claim 127, wherein the total volume of the diluted form administered to the subject is at least about 250 mL.

129. The method of claim 128, wherein the total volume of the diluted form administered to the subject is at least about 500 mL.

130. The method of claim 128, wherein the total volume is about 250 mL.

131. The method of claim 125, wherein 0.1, 0.2, 0.3, 1, 3, 10 or 30 mg/kg of the antibody is administered to the subject about once every 28 days.

132. The method of claim 131, wherein 0.1, 0.2, 0.3, 1 or 3 mg/kg of the antibody is administered to the subject as an infusion over a range of about 60 to 120 minutes.

133. The method of claim 131, wherein 10 or 30 mg/kg of the antibody is administered to the subject as an infusion over a range of about 90 to 180 minutes.

134. The method of claim 125, wherein the subject is premedicated with an antihistamine.

135. The method of claim 125, wherein the subject is administered diphenhydramine within a range of about 30 to 90 minutes prior to the antibody administration.

136. The method of claim 135, wherein the subject is administered acetaminophen within a range of about 30 to 90 minutes prior to the antibody administration.

137. The method of claim 112, wherein the duration of the regime is at least 3 months.

138. The method of claim 137, wherein the duration of the regime is at least 12 months.

Description:
LYOPHILIZED FORMULATION OF A MONOCLONAL ANTIBODY AGAINST

TRANSTHYRETIN

CROSS-REFERENCE TO RELATED APPLICATION

[0001] This application claims the benefit of US 62/592,294 filed November 29, 2018, which is incorporated by reference in its entirety for all purposes.

REFERENCE TO A SEQUENCE LISTING

[0002] This application includes an electronic sequence listing in a file named

5l9l02_SEQLST.txt, created on November 28, 2018, and containing 140,679 bytes, which is incoporated by reference.

BACKGROUND

[0003] Several diseases are thought to be caused by the abnormal folding and aggregation of disease- specific proteins. These proteins can accumulate into pathologically diagnostic accumulations, known as amyloids, which are visualized by certain histologic stains. Amyloids are thought to elicit inflammatory responses and have multiple negative consequences for the involved tissues. In addition, smaller aggregates of abnormally folded protein may exist and exert cytotoxic effects.

[0004] Transthyretin (TTR) is one of the many proteins that are known to misfold and aggregate (e.g., undergo amyloidogenesis). Transthyretin-related amyloidosis encompasses two forms of disease: familial disease arising from misfolding of a mutated or variant TTR, and a sporadic, non-genetic disease caused by misaggregation of wild-type TTR. The process of TTR amyloidogenesis can cause pathology in the nervous system and/or heart, as well as in other tissues.

SUMMARY OF THE CLAIMED INVENTION

[0005] The invention provides pharmaceutical formulations that comprise (a) a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:6l and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Kabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:l and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16, wherein the antibody is present at a concentration within the range from about 25 mg/mL to about 75 mg/mL; (b) a buffer present at a concentration of about 20 mM, wherein the buffer is citrate, histidine, phosphate or succinate; (c) a sugar present at a concentration within the range from about 205 mM to about 260 mM, wherein the sugar is sucrose or trehalose or, if the sugar is absent about 160 mM arginine is present; and (d) a surfactant present at a concentration within the range from about 0.01% to about 1% by weight; wherein the formulation is characterized by a pH within the range from about 5.0 to about 6.5; provided that: (i) if histidine or succinate buffer is present, the pH is about 6.0; (ii) if phosphate buffer is present, the pH is about 6.5; and (iii) if histidine and trehalose are present the surfactant is a. PS80; or b. PS20, provided that if PS20 is present, about 25 mM L-arginine is also present. Optionally, the formulation is essentially free of mannitol or sorbitol. Optionally, the formulation comprising a monoclonal antiobdy as described in (a) is essentially free of mannitol and sorbitol.

[0006] In some formulations, the buffer is histidine. Optionally, the sugar is present in a range from about 230 mM to about 250 mM, optionally about 230 mM to about 240 mM. Optionally, the sugar is trehalose. Optionally, the sugar is sucrose and the surfactant is PS20 or PX188. Optionally, the surfactant is PS20 at a concentration of 0.02% w/w. Optionally, the surfactant is PX188 at a concentration of 0.04% w/w. In some formulations, 160mM arginine is present. Optionally, the sugar is present and is trehalose. Optionally, the trehalose is present at a concentration of 205 mM. Optionally, the surfactant is PS20.

[0007] In some formulations, the buffer is citrate. Optionally, the sugar is present at 230 mM. Optionally, the surfactant is 0.02% PS20. In some formulations, the buffer is phosphate.

Optionally, the sugar is present and is sucrose. In some formulations, the buffer is succinate. Optionally, the sugar is present and is sucrose.

[0008] Some formulations comprise (a) 20 mM citrate, 230 mM trehalose and 0.02% w/w PS20 at pH 5; (b) 20 mM histidine, 230 mM sucrose and 0.02% w/w PS20; (c) 20 mM phosphate,

230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (d) 20 mM citrate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (e) 20 mM histidine, 230 mM trehalose and 0.02% w/w PS 80; (f) 20 mM histidine, 0.02% w/w PS20 and 160 mM L- arginine; (g) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188; (h) 20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or (i) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L-arginine. Optionally, the formulation consists essentially of the antibody and about: (a) 20 mM citrate, 230 mM trehalose and 0.02% w/w PS20 at pH 5; (b) 20 mM histidine, 230 mM sucrose and 0.02% w/w PS20; (c) 20 mM phosphate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (d) 20 mM citrate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (e) 20 mM histidine, 230 mM trehalose and 0.02% w/w PS80; (f) 20 mM histidine, 0.02% w/w PS20 and 160 mM L-arginine; (g) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188; (h)20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or (i) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L-arginine. Optionally, the formulation consists essentially of the antibody and about: (a) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188; (b) 20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or (c)20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L-arginine.

[0009] In some formulations, the antibody comprises a mature heavy chain variable region comprising the amino acid sequence of SEQ ID NO:65, and a mature light chain variable region comprising the amino acid sequence of SEQ ID NO:76. Optionally, the formulation consists essentially of the antibody at about 50 mg/ml, and about 20 mM histidine, about 240 mM sucrose and about 0.04% w/w PX188.

[0010] In some formualtions, the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO:6l, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO:70 except that positions H52 and L26 by Kabat numbering can each be independently N or S.

[0011] In some formulations, the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO:l, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO: 16.

[0012] In some formulations, the monoclonal antibody is a humanized, chimeric or veneered antibody. Optionally, the monoclonal antibody is humanized. Optionally, the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:64-66 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:74-76. Optionally, the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:5-12 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs: 19-23. Optionally, the mature heavy chain has an amino acid sequence comprising SEQ ID NO: 11 and the mature light chain has an amino acid sequence comprising SEQ ID NO: 19. Optionally, the mature heavy chain has an amino acid sequence comprising SEQ ID NO:65 and the mature light chain has an amino acid sequence comprising SEQ ID NO:76.

[0013] In formulations comprising a humanized antibody, the mature heavy chain variable region is fused to a heavy chain constant region and the mature light chain variable region is fused to a light chain constant region. Optionally, the mature heavy chain variable region is fused to a heavy chain constant region having the sequence of SEQ ID NO: 103 provided the C- terminal lysine can be absent and/or the mature light chain variable region is fused to a light chain constant region having the sequence of SEQ ID NO: 104 or 105.

[0014] In some formulations, the monoclonal antibody is present at a concentration of about 50 mg/mL. In some formulations, the histidine buffer is at a concentration of about 20 mM. In some formulations, the pH is about 5.75 to 6.25. In some formulations, the sugar/polyol is sucrose present at a concentration of about 240 mM. In some formulations, the poloxamer is poloxamer 188 at a concentration of about 0.04% by weight. In some formulations, less than about 5% or 3% by weight of the antibody is present as an aggregate in the formulation. In some

formulations, at least 95% or 97% of antibody by weight runs as a single peak under HP-SEC analysis. Some formulations are sterile. Some formulations are stable on freezing and thawing. Some formulations have an osmolality of from about 270 mOsmol/kg to about 330 mOsmol/kg.

[0015] The invention also provides a lyophilized formulation of an antibody that comprises (a) a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:61 and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Kabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO: 1 and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16; (b) histidine; (c) a sugar/polyol; and (d) a poloxamer. Optionally, the lyophilized formulation is prepared by lyophilizing the above-mentioned formulations. Optionally, the lyophilized formulation is reconstitutable with water to a pH of between about 5.5 to about 6.5. Optionally, the lyophilized formulation is reconstitutable with water to a pH of about 6.0.

Optionally, the lyophilized formulation contains about 10 mg to about 40 mg of the antibody.

[0016] The invention also provides a lyophilized formulation, which is reconstitutable with water to yield an aqueous solution comprising: (a) an antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:6l and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Rabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO: 1 and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16 wherein the antibody is present at a concentration within the range from about 25 mg/mL to about 75 mg/mL; (b) buffer present at a concentration within a range from about 10 mM to about 30 mM; (c) sugar/polyol present at a concentration within a range from about 220 mM to about 260 mM; (d) surfactant present at a concentration within a range from about 0.01% to about 0.1% by weight; and (e) a pH within a range from about 5.5 to about 7.0.

[0017] In some lyophilized formulations, the antibody comprises a mature heavy chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:64-66 and a mature light chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:74-76, or an antibody comprising a mature heavy chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:5-l2 and a mature light chain variable region having an amino acid sequence comprising any one of SEQ ID NOs: 19-23. Optionally, the lyophilized formulation is reconstitutable with water so that the antibody is present at a concentration of about 50 mg/mL. Optionally, the buffer comprises histidine. Optionally, the lyophilized formulation is reconstitutable with water so that the pH is about 6.0. Optionally, the sugar/polyol is sucrose. Optionally, the surfactant is poloxomer. Optionally, the poloxomer is PX188. [0018] In some lyophilized formulations, (a) the antibody comprises a mature heavy chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:64-66 and a mature light chain variable region having an amino acid sequence comprising any one of SEQ ID NOs:74-76 and is reconstitutable to a concentration of about 50 mg/ml; (b) the buffer is histidine and is reconstitutable to a concentration of about 2 c) the sugar/polyol is sucrose and is

reconstitutable to a concentration of about 240 mM; and (d) the surfactant is poloxomer 188 and is reconstitutable to a concentration of about 0.04% by weight. Optionally, the lyophilized formulation is reconstitutable with water so that the pH is about 6.0. Optionally, the antibody comprises a mature heavy chain variable region having an amino acid sequence comprising SEQ ID NO:65 and a mature light chain variable region having an amino acid sequence comprising SEQ ID NO:76. Optionally, the mature heavy chain variable region is fused to a heavy chain constant region having the sequence of SEQ ID NO: 103 and the mature light chain variable region is fused to a light chain constant region having the sequence of SEQ ID NO: 104. In these lyophilized formulations, at least 95 or 97 % of the antibody runs as a single peak under HP-SEC after storage for up to 3 months at 2-8 °C.

[0019] The invention also provides a method of reconstituting the lyophilized formulation that comprises combining the lyophilized formulation with sterile water to produce a liquid formulation. Optionally, it further comprises introducing the reconstituted formulation into a bag of isotonic fluid for infusion. Optionally, the lyophilized formulation is in powder form or in the form of a solid foam in a vial.

[0020] The invention further provides a sterile lyophilized dosage form of an antibody formulation in a 20ml vial consisting essentially of: (i) an antibody within a range of about 225- 275 mg; (ii) histidine within a range of about 15-19 mg; (iii) poloxamer PX188 within a range of about 2-2.5 mg; and; (iv) sucrose within a range of about 400-490 mg; wherein the antibody is a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:6l and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and L26 by Kabat numbering can each be independently N or S, or a monoclonal antibody comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO: 1 and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16. Optionally, the vial has contents consisting essentially of: (i) about 250 mg of the antibody; (ii) about 16.8 mg of L-histidine; (iii) about 2.2 mg of poloxomer PX188; and (iv) about 445.3 mg of sucrose. Optionally, the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO:6l, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO:70 except that positions H52 and L26 by Kabat numbering can each be independently N or S. Optionally, the antibody comprises a mature heavy chain variable region comprising the three Kabat CDRs of SEQ ID NO: 1, and a mature light chain variable region comprising the three Kabat CDRs of SEQ ID NO: 16.

Optionally, the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:64-66 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:74-76. Optionally, the mature heavy chain variable region has an amino acid sequence comprising any one of SEQ ID NOs:5-l2 and the mature light chain variable region has an amino acid sequence comprising any one of SEQ ID NOs: 19-23.

Optionally, the mature heavy chain has an amino acid sequence comprising SEQ ID NO:65 and the mature light chain has an amino acid sequence comprising SEQ ID NO:76. Optionally, the mature heavy chain has an amino acid sequence comprising SEQ ID NO:6 and the mature light chain has an amino acid sequence comprising SEQ ID NO:21. Optionally, the mature heavy chain variable region is fused to a heavy chain constant region having the sequence of SEQ ID NO: 103 provided the C-terminal lysine can be absent and/or the mature light chain variable region is fused to a light chain constant region having the sequence of SEQ ID NO: 104 or 105. Optionally, the mature heavy chain has the sequence of SEQ ID NO:82, except the C-terminal lysine may be absent, and the mature light chain has the sequence of SEQ ID NO:86.

[0021] The invention also provides a method of preparing the lyophilized dosage form of for administration to a subject, comprising: (i) reconstituting the antibody formulation to a volume of about 5.0 mL with sterile water, and (ii) diluting the reconstituted antibody formulation of step (i) in normal saline for infusion. Optionally, the total volume for infusion is 250 mL. Optionally, the total volume for infusion is 100 mL. Optionally, the total volume for infusion is 500 mL.

[0022] The invention also provides a reconstituted formulation resulting from reconstituting lyophilized formulations. Some reconstituted formulations comprise the antibody at a concentration of about 50 mg/mL, the histidine buffer at a concentration of about 20 mM, poloxamer at a concentration of about 0.04% by weight, and at pH of about 6.0. Optionally, at least 95, 96, 97, 98 or 99% of the antibody runs as a single peak on high pressure size exclusion chromatography.

[0023] The invention also provides a method of treating or effecting prophylaxis of a subject having or at risk of a transthyretin- mediated amyloidosis, comprising administering to the subject an effective regime of the pharmaceutical formulation or a reconstituted form of the lyophilized formulation. Optionally, the subject has been previously treated with a TTR tetramer stablizer, an antisense oligonucleotide based therapy, an RNA interference (RNAi) based therapy, or an amyloid degrader. Optionally, the subject no longer receives the treatment with the TTR tetramer stablizer, antisense oligonucleotide based therapy, RNA interference (RNAi) based therapy, or amyloid degrader. Optionally, the subject is concurrently receiving treatment with diflunisal. Optionally, the subject is concurrently receiving treatment with tafamidis. Optionally, the subject is concurrently receiving treatment with a TTR tetramer stablizer, an antisense oligonucleotide based therapy, an RNA interference (RNAi) based therapy, or an amyloid degrader. Optionally, the subject is concurrently reciving treatment with tafamidis, difluisal, patisiran, inotersen, 4'-iodo-4'-deoxydoxorubicin (IDOX), doxycycline, tauroursodeoxycholic acid (TUDCA), cyclodextrin (CyD), or an anti-SAP antibody. Optionally, the subject has been diagnosed with ATTR amyloidosis. Optionally, the subject has wild-type ATTR- cardiomyopathy. Optionally, the subject has hereditary ATTR-cardiomyopathy, or hereditary ATTR-polyneuropathy or both. Optionally, the subject has ATTR cardiac involvement.

Optionally, the subject has ATTR amyloidosis peripheral neuropathy involvement. Optionally, the subject is receiving concomitant tafamidis.

[0024] The invention also provides that a pharmaceutical formulation, the reconstituted form of the lyophilized formulation or the reconstituted form of the lyophilized antibody formulation is administered to the subject intravenously in diluted form. Optionally, the diluent for the diluted form is normal saline. Optionally, the total volume of the diluted form administered to the subject is at least about 100 mL. Optionally, the total volume of the diluted form administered to the subject is at least about 250 mL. Optionally, the total volume of the diluted form administered to the subject is at least about 500 mL. Optionally, the total volume is about 250 mL. Optionally, 0.1, 0.2, 0.3, 1, 3, 10 or 30 mg/kg of the antibody is administered to the subject about once every 28 days. Optionally, 0.1, 0.2, 0.3, 1 or 3 mg/kg of the antibody is administered to the subject as an infusion over a range of about 60 to 120 minutes. Optionally, 10 or 30 mg/kg of the antibody is administered to the subject as an infusion over a range of about 90 to 180 minutes. Optionally, the subject is premedicated with an antihistamine. Optionally, the subject is adminstered diphenhydramine within a range of about 30 to 90 minutes prior to the antibody administration. Optionally, the subject is administered acetaminophen within a range of about 30 to 90 minutes prior to the antibody administration. Optionally, the duration of the regime is at least 3 months. Optionally, the duration of the regime is at least 12 months.

[0025] The invention further provides a method of treating or effecting prophylaxis of a subject having or at risk of a transthyretin- mediated amyloidosis that comprises administering an effective regime of a TTR tetramer stablizer, an antisense oligonucleotide based therapy, an RNA interference (RNAi) based therapy, or an amyloid degrader, wherein the subject has previously been treated with the pharmaceutical formulation or a reconstituted form of the lyophilized formulation as described above. Optionally, the subject no longer receives the treatment with the pharmaceutical formulation. Optionally, the TTR tetramer stabilizer is tafamidis or diflunisal. Optionally, the antisense oligonucleotide based therapy is inotersen. Optionally, the RNA interference (RNAi) based therapy is patisiran or revusiran. Optionally, the amyloid degraders are 4'-iodo-4'-deoxydoxorubicin (IDOX), doxycycline, tauroursodeoxycholic acid (TUDCA), cyclodextrin (CyD), or an anti-SAP antibody. Optionally, the amyloid degraders are doxycycline in combined with tauroursodeoxycholic acid (TUDCA). Optionally, the subject has been diagnosed with ATTR amyloidosis. Optionally, the subject has wild-type ATTR-cardiomyopathy. Optionally, the subject has hereditary ATTR-cardiomyopathy.

Optionally, the subject has hereditary ATTR-polyneuropathy. Optionally, the subject has ATTR cardiac involvement. Optionally, the subject has ATTR amyloidosis peripheral neuropathy involvement. Optionally, the pharmaceutical formulation, reconstituted form of the lyophilized formulation or the reconstituted form of the lyophilized antibody formulation is administered to the subject intravenously in diluted form. Optionally, the diluent for the diluted form is normal saline. Optionally, the total volume of the diluted form administered to the subject is at least about 100 mL. Optionally, the total volume of the diluted form administered to the subject is at least about 250 mL. Optionally, the total volume of the diluted form administered to the subject is at least about 500 mL. Optionally, the total volume is about 250 mL. Optionally, 0.1, 0.2, 0.3, 1, 3, 10 or 30 mg/kg of the antibody is administered to the subject about once every 28 days. Optionally, 0.1, 0.2, 0.3, 1 or 3 mg/kg of the antibody is administered to the subject as an infusion over a range of about 60 to 120 minutes. Optionally, 10 or 30 mg/kg of the antibody is administered to the subject as an infusion over a range of about 90 to 180 minutes. Optionally, the subject is premedicated with an antihistamine. Optionally, the subject is adminstered diphenhydramine within a range of about 30 to 90 minutes prior to the antibody administration. Optionally, the subject is administered acetaminophen within a range of about 30 to 90 minutes prior to the antibody administration. Optionally, the duration of the regime is at least 3 months. Optionally, the duration of the regime is at least 12 months.

BRIEF DESCRIPTION OF THE SEQUENCES

[0026] SEQ ID NO: 1 sets forth the amino acid sequence of the heavy chain variable region of the mouse 9D5 antibody.

[0027] SEQ ID NO:2 sets forth the amino acid sequence of the mouse heavy chain variable region structure template lSEQ_H.

[0028] SEQ ID NO:3 sets forth the amino acid sequence of the heavy chain variable acceptor ACC# BAC02114.

[0029] SEQ ID NO:4 sets forth the amino acid sequence of the heavy chain variable acceptor ACC#AAX82494.1.

[0030] SEQ ID NO:5 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 1 (Hu9D5VHvl).

[0031] SEQ ID NO:6 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 2 (Hu9D5VHv2).

[0032] SEQ ID NO:7 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 2b (Hu9D5VHv2b). [0033] SEQ ID NO:8 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 3 (Hu9D5VHv3).

[0034] SEQ ID NO:9 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 3b (Hu9D5VHv3b).

[0035] SEQ ID NO: 10 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 4 (Hu9D5VHv4).

[0036] SEQ ID NO: 11 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 4b (Hu9D5VHv4b).

[0037] SEQ ID NO: 12 sets forth the amino acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 5 (Hu9D5VHv5).

[0038] SEQ ID NO: 13 sets forth the amino acid sequence of Kabat CDR-H1 of the mouse 9D5 antibody.

[0039] SEQ ID NO: 14 sets forth the amino acid sequence of Kabat CDR-H2 of the mouse 9D5 antibody.

[0040] SEQ ID NO: 15 sets forth the amino acid sequence of Kabat CDR-H3 of the mouse 9D5 antibody.

[0041] SEQ ID NO: 16 sets forth the amino acid sequence of the light chain variable region of the mouse 9D5 antibody.

[0042] SEQ ID NO: 17 sets forth the amino acid sequence of the mouse light chain variable region structure template 1MJU_L.

[0043] SEQ ID NO: 18 sets forth the amino acid sequence of the light chain variable acceptor ACC# ABC66952.

[0044] SEQ ID NO: 19 sets forth the amino acid sequence of the light chain variable region of the humanized 9D5 antibody version 1 (Hu9D5VLvl).

[0045] SEQ ID NO:20 sets forth the amino acid sequence of the light chain variable region of the humanized 9D5 antibody version 2 (Hu9D5VLv2). [0046] SEQ ID NO:2l sets forth the amino acid sequence of the light chain variable region of the humanized 9D5 antibody version 3 (Hu9D5VLv3).

[0047] SEQ ID NO:22 sets forth the amino acid sequence of the light chain variable region of the humanized 9D5 antibody version 4 (Hu9D5VLv4).

[0048] SEQ ID NO:23 sets forth the amino acid sequence of the light chain variable region of the humanized 9D5 antibody version 5 (Hu9D5VLv5).

[0049] SEQ ID NO:24 sets forth the amino acid sequence of Kabat CDR-L1 of the mouse 9D5 antibody.

[0050] SEQ ID NO:25 sets forth the amino acid sequence of Kabat CDR-L2 of the mouse 9D5 antibody.

[0051] SEQ ID NO:26 sets forth the amino acid sequence of Kabat CDR-L3 of the mouse 9D5 antibody.

[0052] SEQ ID NO:27 sets forth the amino acid sequence of humanized 9D5 heavy chain version 1.

[0053] SEQ ID NO:28 sets forth the amino acid sequence of humanized 9D5 heavy chain version 2.

[0054] SEQ ID NO:29 sets forth the amino acid sequence of humanized 9D5 heavy chain version 2b.

[0055] SEQ ID NO:30 sets forth the amino acid sequence of humanized 9D5 heavy chain version 3.

[0056] SEQ ID NO:31 sets forth the amino acid sequence of humanized 9D5 heavy chain version 3b.

[0057] SEQ ID NO:32 sets forth the amino acid sequence of humanized 9D5 heavy chain version 4.

[0058] SEQ ID NO:33 sets forth the amino acid sequence of humanized 9D5 heavy chain version 4b. [0059] SEQ ID NO:34 sets forth the amino acid sequence humanized 9D5 heavy chain version 5.

[0060] SEQ ID NO:35 sets forth the amino acid sequence of humanized 9D5 light chain version 1.

[0061] SEQ ID NO:36 sets forth the amino acid sequence of humanized 9D5 light chain version 2.

[0062] SEQ ID NO:37 sets forth the amino acid sequence of humanized 9D5 light chain version 3.

[0063] SEQ ID NO:38 sets forth the amino acid sequence of humanized 9D5 light chain version 4.

[0064] SEQ ID NO:39 sets forth the amino acid sequence of humanized 9D5 light chain version 5.

[0065] SEQ ID NO:40 sets forth the nucleic acid sequence of the heavy chain variable region of the mouse 9D5 antibody with signal peptide.

[0066] SEQ ID NO:41 sets forth the amino acid sequence of the heavy chain variable region of the mouse 9D5 antibody with signal peptide.

[0067] SEQ ID NO:42 sets forth the nucleic acid sequence of the light chain variable region of the mouse 9D5 antibody with signal peptide.

[0068] SEQ ID NO:43 sets forth the amino acid sequence of the light chain variable region of the mouse 9D5 antibody with signal peptide.

[0069] SEQ ID NO:44 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 1 (Hu9D5VHvl).

[0070] SEQ ID NO:45 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 2 (Hu9D5VHv2).

[0071] SEQ ID NO:46 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 2b (Hu9D5VHv2b). [0072] SEQ ID NO:47 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 3 (Hu9D5VHv3).

[0073] SEQ ID NO:48 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 3b (Hu9D5VHv3b).

[0074] SEQ ID NO:49 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 4 (Hu9D5VHv4).

[0075] SEQ ID NO:50 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 4b (Hu9D5VHv4b).

[0076] SEQ ID NO:51 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 9D5 antibody version 5 (Hu9D5VHv5).

[0077] SEQ ID NO:52 sets forth the nucleic acid sequence of the light chain variable region of the humanized 9D5 antibody version 1 (Hu9D5VLvl).

[0078] SEQ ID NO:53 sets forth the nucleic acid sequence of the light chain variable region of the humanized 9D5 antibody version 2 (Hu9D5VLv2).

[0079] SEQ ID NO:54 sets forth the nucleic acid sequence of the light chain variable region of the humanized 9D5 antibody version 3 (Hu9D5VLv3).

[0080] SEQ ID NO:55 sets forth the nucleic acid sequence of the light chain variable region of the humanized 9D5 antibody version 4 (Hu9D5VLv4).

[0081] SEQ ID NO:56 sets forth the nucleic acid sequence of the light chain variable region of the humanized 9D5 antibody version 5 (Hu9D5VLv5).

[0082] SEQ ID NO:57 sets forth the amino acid sequence of the mouse 9D5 heavy chain variable region signal peptide.

[0083] SEQ ID NO:58 sets forth the nucleic acid sequence of the mouse 9D5 heavy chain variable region signal peptide.

[0084] SEQ ID NO:59 sets forth the amino acid sequence of the mouse 9D5 light chain variable region signal peptide. [0085] SEQ ID NO:60 sets forth the nucleic acid sequence of the mouse 9D5 light chain variable region signal peptide.

[0086] SEQ ID NO:6l sets forth the amino acid sequence of the heavy chain variable region of the mouse 14G8 antibody.

[0087] SEQ ID NO:62 sets forth the amino acid sequence of the mouse heavy chain variable region structure template 1MQK_H.

[0088] SEQ ID NO:63 sets forth the amino acid sequence of the heavy chain variable acceptor ACC# AAD30410.1.

[0089] SEQ ID NO:64 sets forth the amino acid sequence of the heavy chain variable region of the humanized 14G8 antibody version 1 (Hul4G8VHvl).

[0090] SEQ ID NO:65 sets forth the amino acid sequence of the heavy chain variable region of the humanized 14G8 antibody version 2 (Hul4G8VHv2).

[0091] SEQ ID NO:66 sets forth the amino acid sequence of the heavy chain variable region of the humanized 14G8 antibody version 3 (Hul4G8VHv3).

[0092] SEQ ID NO:67 sets forth the amino acid sequence of Kabat CDR-H1 of the mouse 14G8 antibody.

[0093] SEQ ID NO:68 sets forth the amino acid sequence of Kabat CDR-H2 of the mouse 14G8 antibody.

[0094] SEQ ID NO:69 sets forth the amino acid sequence of Kabat CDR-H3 of the mouse 14G8 antibody.

[0095] SEQ ID NO:70 sets forth the amino acid sequence of the light chain variable region of the mouse 14G8 antibody.

[0096] SEQ ID NO:71 sets forth the amino acid sequence of the mouse light chain variable region structure template 1MJU_L.

[0097] SEQ ID NO:72 sets forth the amino acid sequence of the light chain variable acceptor ACC# ABA71374.1. [0098] SEQ ID NO:73 sets forth the amino acid sequence of the light chain variable acceptor ACC# ABC66952.1.

[0099] SEQ ID NO:74 sets forth the amino acid sequence of the light chain variable region of the humanized 14G8 antibody version 1 (Hul4G8VLvl).

[00100] SEQ ID NO:75 sets forth the amino acid sequence of the light chain variable region of the humanized 14G8 antibody version 2 (Hul4G8VLv2).

[00101] SEQ ID NO:76 sets forth the amino acid sequence of the light chain variable region of the humanized 14G8 antibody version 3 (Hul4G8VLv3).

[00102] SEQ ID NO:77 sets forth the amino acid sequence of Kabat CDR-L1 of the mouse 14G8 antibody.

[00103] SEQ ID NO:78 sets forth the amino acid sequence of Kabat CDR-L2 of the mouse 14G8 antibody.

[00104] SEQ ID NO:79 sets forth the amino acid sequence of Kabat CDR-L3 of the mouse 14G8 antibody.

[00105] SEQ ID NO:80 sets forth the amino acid sequence of Kabat CDR-L1 of the humanized 14G8 antibody version 3 (Hul4G8VLv3).

[00106] SEQ ID NO:81 sets forth the amino acid sequence of humanized 14G8 heavy chain version 1.

[00107] SEQ ID NO:82 sets forth the amino acid sequence of humanized 14G8 heavy chain version 2.

[00108] SEQ ID NO:83 sets forth the amino acid sequence of humanized 14G8 heavy chain version 3.

[00109] SEQ ID NO: 84 sets forth the amino acid sequence of humanized 14G8 light chain version 1.

[00110] SEQ ID NO:85 sets forth the amino acid sequence of humanized 14G8 light chain version 2. [00111] SEQ ID NO:86 sets forth the amino acid sequence of humanized 14G8 light chain version 3.

[00112] SEQ ID NO:87 sets forth the nucleic acid sequence of the heavy chain variable region of the mouse 14G8 antibody with signal peptide.

[00113] SEQ ID NO:88 sets forth the amino acid sequence of the heavy chain variable region of the mouse 14G8 antibody with signal peptide.

[00114] SEQ ID NO:89 sets forth the nucleic acid sequence of the light chain variable region of the mouse 14G8 antibody with signal peptide.

[00115] SEQ ID NO:90 sets forth the amino acid sequence of the light chain variable region of the mouse 14G8 antibody with signal peptide.

[00116] SEQ ID NO:91 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 14G8 antibody version 1 (Hul4G8VHvl).

[00117] SEQ ID NO:92 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 14G8 antibody version 2 (Hul4G8VHv2).

[00118] SEQ ID NO:93 sets forth the nucleic acid sequence of the heavy chain variable region of the humanized 14G8 antibody version 3 (Hul4G8VHv3).

[00119] SEQ ID NO:94 sets forth the nucleic acid sequence of the light chain variable region of the humanized 14G8 antibody version 1 (Hul4G8VLvl).

[00120] SEQ ID NO:95 sets forth the nucleic acid sequence of the light chain variable region of the humanized 14G8 antibody version 2 (Hul4G8VLv2).

[00121] SEQ ID NO:96 sets forth the nucleic acid sequence of the light chain variable region of the humanized 14G8 antibody version 3 (Hul4G8VLv3).

[00122] SEQ ID NO:97 sets forth the amino acid sequence of the mouse 14G8 heavy chain variable region signal peptide.

[00123] SEQ ID NO:98 sets forth the nucleic acid sequence of the mouse 14G8 heavy chain variable region signal peptide. [00124] SEQ ID NO:99 sets forth the amino acid sequence of the mouse 14G8 light chain variable region signal peptide.

[00125] SEQ ID NO: 100 sets forth the nucleic acid sequence of the mouse 14G8 light chain variable region signal peptide.

[00126] SEQ ID NO: 101 sets forth the amino acid sequence of an exemplary human IgGl heavy chain constant region.

[00127] SEQ ID NO: 102 sets forth the amino acid sequence of an exemplary human IgGl heavy chain constant region of the IgGl Glm3 allotype with alanines occupying positions 234 and 235 by EU numbering.

[00128] SEQ ID NO: 103 sets forth the amino acid sequence of an exemplary human IgGl heavy chain constant region of the IgGl Glm3 allotype.

[00129] SEQ ID NO: 104 sets forth the amino acid sequence of an exemplary human kappa light chain constant region having an N-terminal arginine.

[00130] SEQ ID NO: 105 sets forth the amino acid sequence of an exemplary human kappa light chain constant region without an N-terminal arginine.

[00131] SEQ ID NO: 106 sets forth the nucleic acid sequence of an exemplary heavy chain constant region of the Glm3 allotype.

[00132] SEQ ID NO: 107 sets forth the nucleic acid sequence of an exemplary light chain constant region having an N-terminal arginine.

[00133] SEQ ID NO: 108 sets forth the nucleic acid sequence of an exemplary light chain constant region without an N-terminal arginine.

[00134] SEQ ID NO: 109 sets forth the amino acid sequence of human transthyretin set forth in accession number P02766.1 (UniProt).

[00135] SEQ ID NO: 110 sets forth the amino acid sequence of human transthyretin set forth in accession number AAB35639.1 (GenBank). [00136] SEQ ID NO: 111 sets forth the amino acid sequence of human transthyretin set forth in accession number AAB35640.1 (GenBank).

[00137] SEQ ID NO: 112 sets forth the amino acid sequence of human transthyretin set forth in accession number ABI63351.1 (GenBank).

[00138] SEQ ID NO: 113 sets forth the amino acid sequence of residues 89-97 of human transthyretin.

[00139] SEQ ID NO: 114 sets forth the amino acid sequence of a potential transthyretin immunogen.

[00140] SEQ ID NO: 115 sets forth the amino acid sequence of a potential transthyretin immunogen.

[00141] SEQ ID NO: 116 sets forth the amino acid sequence of a potential transthyretin immunogen.

[00142] SEQ ID NO: 117 sets forth the amino acid sequence of composite Chothia-Kabat CDR-H1 of the mouse 9D5 antibody.

[00143] SEQ ID NO: 118 sets forth the amino acid sequence of composite Chothia-Kabat CDR-H1 of the mouse 14G8 antibody.

DEFINITIONS

[00144] The term“antibody” includes intact antibodies and binding fragments thereof. Typically, fragments compete with the intact antibody from which they were derived for specific binding to the target. Fragments include separate heavy chains, separate light chains, Fab, Fab', F(ab')2, F(ab)c, Fv, single chain antibodies, and single domain antibodies The term“antibody” also includes a bispecific antibody. A bispecific or bifunctional antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites (see, e.g., Songsivilai and Lachmann, Clin. Exp. Immunol., 79:315-321 (1990); Kostelny et al., J.

Immunol., 148:1547-53 (1992)). [00145] The basic antibody structural unit is a tetramer of subunits. Each tetramer includes two identical pairs of polypeptide chains, each pair having one“light” chain (about 25 kDa) and one“heavy” chain (about 50-70 kDa). The amino -terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. When initially expressed, this variable region is typically linked to a cleavable signal peptide. The variable region without the signal peptide is sometimes referred to as a mature variable region. Thus, for example, a light chain mature variable region means a light chain variable region without the light chain signal peptide. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. A constant region can include any or all of a CHI region, hinge region, CH2 region, and CH3 region.

[00146] Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a“J” region of about 12 or more amino acids, with the heavy chain also including a “D” region of about 10 or more amino acids. ( See generally, Fundamental Immunology (Paul, W., ed., 2nd ed. Raven Press, N.Y., 1989), Ch. 7) (incorporated by reference in its entirety for all purposes).

[00147] The mature variable regions of each light/heavy chain pair form the antibody binding site. Thus, an intact antibody has two binding sites. Except for bifunctional or bispecific antibodies, the two binding sites are the same. The chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDRs from the two chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N- terminal to C-terminal, both light and heavy chains comprise the regions FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each region is in accordance with the definitions of Rabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD, 1987 and 1991), or Chothia & Lesk, J. Mol. Biol. 196:901- 917 (1987); Chothia et al, Nature 342:878-883 (1989) or CDRs can be defined by the alternative definitions in Table 1 below. Rabat also provides a widely used numbering convention (Rabat numbering) in which corresponding residues between different heavy chains or between different light chains are assigned the same number.

[00148] Table 1: Conventional Definitions of CDRs Using Kabat Numbering

Table 1

*CDR-H1 by Chothia can end at H32, H33, or H34 (depending on the length of the loop). This is because the Kabat numbering scheme places insertions of extra residues at 35A and 35B, whereas Chothia numbering places them at 31A and 3 IB. If neither H35A nor H35B (Kabat numbering) is present, the Chothia CDR- Hl loop ends at H32. If only H35A is present, it ends at H33. If both H35A and H35B are present, it ends at H34.

[00149] The mature variable region of a heavy or light chain is being compared with the same region of a reference antibody, the percentage sequence identity between the subject and reference antibody regions is the number of positions occupied by the same amino acid in both the subject and reference antibody region divided by the total number of aligned positions of the two regions (with gaps not counted) multiplied by 100 to convert to percentage.

[00150] For purposes of classifying amino acids substitutions as conservative or non conservative, amino acids are grouped as follows: Group I (hydrophobic sidechains): Norleucine, Met, Ala, Val, Leu, lie; Group II (neutral hydrophilic side chains): Cys, Ser, Thr; Group III (acidic side chains): Asp, Glu; Group IV (basic side chains): Asn, Gln, His, Lys, Arg; Group V (residues influencing chain orientation): Gly, Pro; and Group VI (aromatic side chains): Trp, Tyr, Phe. Conservative substitutions involve substitutions between amino acids in the same class. [00151] Non-conservative substitutions constitute exchanging a member of one of these classes for a member of another.

[00152] Antibodies of the invention typically bind to their designated target with an affinity constant of at least 10 6 , 10 7 , 10 8 , 10 9 , or 10 10 M 1 . Such binding is specific binding in that it is detectably higher in magnitude and distinguishable from non-specific binding occurring to at least one unrelated target. Specific binding can be the result of formation of bonds between particular functional groups or particular spatial fit (e.g., lock and key type) whereas nonspecific binding is usually the result of van der Waals forces. Specific binding does not however necessarily imply that a monoclonal antibody binds one and only one target.

[00153] The term“symptom” refers to subjective evidence of a disease, such as altered gait, as perceived by a subject. A“sign” refers to objective evidence of a disease as observed by a physician.

[00154] An individual is at increased risk of a disease if the subject has at least one known risk-factor (e.g., genetic, biochemical, family history, situational exposure) placing individuals with that risk factor at a statistically significant greater risk of developing the disease than individuals without the risk factor. Statistical significance means p<0.05.

[00155] Unless otherwise apparent from the context, the term“about” encompasses values within ±5 or ±10% of a stated value.

[00156] Unless otherwise apparent from the context, reference to a range includes any integer within the range.

[00157] The term“native” with respect to the structure transthyretin (TTR) refers to the normal folded structure of TTR in its properly functioning state (i.e., a TTR tetramer). As TTR is a tetramer in its natively folded form, non-native forms of TTR include, for example, misfolded TTR tetramers, TTR monomers, aggregated forms of TTR, and fibril forms of TTR. Non-native forms of TTR can include molecules comprising wild-type TTR amino acid sequences or mutations.

[00158] The term“misfolded” with respect to TTR refers to the secondary and tertiary structure of a TTR polypeptide monomer or multimer, and indicates that the polypeptide has adopted a conformation that is not normal for that protein in its properly functioning state.

Although TTR misfolding can be caused by mutations in the protein (e.g., deletion, substitution, or addition), wild-type TTR proteins can also be misfolded in diseases, exposing specific epitopes.

[00159] The term“pharmaceutically acceptable” means that the carrier, diluent, excipient, or auxiliary is compatible with the other ingredients of the formulation and not substantially deleterious to the recipient thereof.

[00160] The term“patient” includes human and other mammalian subjects that receive either prophylactic or therapeutic treatment.

[00161] An individual is at increased risk of a disease if the subject has at least one known risk- factor (e.g., genetic, biochemical, family history, and situational exposure) placing individuals with that risk factor at a statistically significant greater risk of developing the disease than individuals without the risk factor.

[00162] The term“disease” refers to any abnormal condition that impairs physiological function. The term is used broadly to encompass any disorder, illness, abnormality, pathology, sickness, condition, or syndrome in which physiological function is impaired, irrespective of the nature of the etiology.

DETAILED DESCRIPTION

L GENERAL

[00163] 14G8 and 9G5 are antibodies binding to transthyretin (TTR). Humanized forms of the antibodies are described in WO2016/120810, incorporated by reference in its entirety for all purposes. The present application provides liquid and lyophilized formulations incorporating antibodies having the CDRs of the 14G8 or 9G5 antibody, particularly chimeric, veneered or humanized forms of 14G8 or 9G5. The formulations include combinations of pharmaceutically acceptable carriers conferring stability on the antibody as further described below. II. Target molecules

[00164] Transthyretin (TTR) is a 127-amino acid, 55 kDa serum and cerebrospinal fluid transport protein primarily synthesized by the liver. It has also been referred to as prealbumin, thyroxine binding prealbumin, ATTR, and TBPA. In its native state, TTR exists as a tetramer. In homozygotes, the tetramers comprise identical 127-amino-acid beta- sheet-rich subunits. In heterozygotes, the TTR tetramers are made up of variant and/or wild-type subunits, typically combined in a statistical fashion.

[00165] The established function of TTR in the blood is to transport holo- retinol binding protein. Although TTR is the major carrier of thyroxine (T 4 ) in the blood of rodents, utilizing binding sites that are orthogonal to those used for /zo/o-retinol binding protein, the T 4 binding sites are effectively unoccupied in humans.

[00166] TTR is one of at least thirty different human proteins whose extracellular misfolding and/or misassembly (amyloidogenesis) into a spectrum of aggregate structures is thought to cause degenerative diseases referred to as amyloid diseases. TTR undergoes conformational changes in order to become amyloidogenic. Partial unfolding exposes stretches of largely uncharged hydrophobic residues in an extended conformation that efficiently misassemble into largely unstructured spherical aggregates that ultimately undergo conformation conversion into cross-beta sheet amyloid structures.

[00167] Unless otherwise apparent from context, reference to transthyretin (TTR) or its fragments or domains includes the natural human amino acid sequences including isoforms, mutants, and allelic variants thereof. Exemplary TTR polypeptide sequences are designated by Accession Numbers P02766.1 (UniProt), AAB35639.1 (GenBank), AAB35640.1

(GenBank), and ABI63351.1 (GenBank) (SEQ ID NOS: 109- 112, respectively). Residues are numbered according to Swiss Prot P02766.1, with the first amino acid of the mature protein (i.e., not including the 20 amino acid signal sequence) designated residue 1. In any other TTR protein, residues are numbered according to the corresponding residues in P02766.1 on maximum alignment. III. Transthyretin Amyloidosis

[00168] Transthyretin (TTR) amyloidosis is a systemic disorder characterized by pathogenic, misfolded TTR and the extracellular deposition of amyloid fibrils composed of TTR. TTR amyloidosis is generally caused by destabilization of the native TTR tetramer form (due to environmental or genetic conditions), leading to dissociation, misfolding, and aggregation of TTR into amyloid fibrils that accumulate in various organs and tissues, causing progressive dysfunction. See, e.g., Almeida and Saraiva, FEBS Letters 586:2891-2896 (2012); Ando el al, Orphanet Journal of Rare Diseases 8:31 (2013).

[00169] In humans, both wild-type TTR tetramers and mixed tetramers comprised of mutant and wild-type subunits can dissociate, misfold, and aggregate, with the process of amyloidogenesis leading to the degeneration of post-mitotic tissue. Thus, TTR amyloidoses encompass diseases caused by pathogenic misfolded TTR resulting from mutations in TTR or resulting from non-mutated, misfolded TTR.

[00170] For example, senile systemic amyloidosis (SSA) and senile cardiac amyloidosis (SCA) are age-related types of amyloidosis that result from the deposition of wild-type TTR amyloid outside and within the cardiomyocytes of the heart. TTR amyloidosis is also the most common form of hereditary (familial) amyloidosis, which is caused by mutations that destabilize the TTR protein. The TTR amyloidoses associated with point mutations in the TTR gene include familial amyloid polyneuropathy (FAP), familial amyloid cardiomyopathy (FAC), and the rare central nervous system selective amyloidosis (CNSA). Patients with hereditary (familial) TTR amyloidosis are almost always heterozygotes, meaning that the TTR tetramers are composed of mutant and/or wild-type TTR subunits, generally statistically distributed. Hereditary (familial) versions of TTR amyloidosis are generally autosomal dominant and are typically earlier onset than the sporadic diseases (SSA and SCA).

[00171] There are over 100 mutations in the gene encoding TTR that have been implicated in the autosomal dominant disorders FAP and FAC. See, e.g., US 2014/0056904; Saraiva, Hum. Mutat. l7(6):493-503 (2001); Damas and Saraiva, J. Struct. Biol. 130:290-299; Dwulet and Benson, Biochem. Biophys. Res. Commun. 114:657-662 (1983). These amyloid-causing mutations are distributed throughout the entire molecule of TTR. Generally, the more destabilizing the mutant subunits are to the TTR tetramer structure, the earlier the onset of amyloid disease. The pathogenic potential of a TTR variant is generally determined by a combination of its instability and its cellular secretion efficiency. The initial pathology caused by some TTR variants comes from their selective destruction of cardiac tissue, whereas that from other TTR variants comes from compromising the peripheral and autonomic nervous system.

The tissue damage caused by TTR amyloidogenesis appear to stem largely from the toxicity of small, diffusible TTR aggregates, although accumulation of extracellular amyloid may contribute and almost certainly compromises organ structure in the late stages of the TTR amyloidosis.

[00172] TTR amyloidosis presents in many different forms, with considerable phenotypic variation across individuals and geographic locations. For example, TTR amyloidosis can present as a progressive, axonal sensory autonomic and motor neuropathy. TTR amyloidosis can also present as an infiltrative cardiomyopathy.

[00173] The age at onset of disease-related symptoms varies between the second and ninth decades of life, with great variations across different populations. The multisystem involvement of TTR amyloidosis is a clue to its diagnosis. For example, TTR amyloidosis diagnosis is considered when one or several of the following are present: (1) family history of neuropathic disease, especially associated with heart failure; (2) neuropathic pain or progressive sensory disturbances of unknown etiology; (3) carpal tunnel syndrome without obvious cause, particularly if it is bilateral and requires surgical release; (4) gastrointestinal motility

disturbances or autonomic nerve dysfunction of unknown etiology (e.g., erectile dysfunction, orthostatic hypotension, neurogenic gladder); (5) cardiac disease characterized by thickened ventricular walls in the absence of hypertension; (6) advanced atrio-ventricular block of unknown origin, particularly when accompanied by a thickened heart; and (6) vitreous body inclusions of the cotton- wool type. See Ando et al., Orphanet Journal of Rare Diseases 8:31 (2013). Other symptoms can include, for example, polyneuropathy, sensory loss, pain, weakness in lower limbs, dyshidrosis, diarrhea, constipation, weight loss, and urinary

incontinence/retention.

[00174] Peripheral neuropathy can be detected and quantified by various clinical scales. For example, Clinical Neuropathy Assessment (CAN), (Dyck PJ, Hughes RAC, O’Brien PC. Quantitating overall neuropathic symptoms, impairments and outcomes. In: Dyck PJ, Thomas PK,, editors. Peripheral neuropathy. 4 th ed. Philadelphia, PA: Elsevier Saunders; 2005.

P 1031-51), provides a scale comprising a Lower Limb Lunction score (LLL), and ability (scored 0) or inability (scored 1) to walk on the toes, walk on the heels, and arise from a kneeling position; items are scored separately for each side The Neuropathy Impairment Score (NIS) provides a clinical assessment that tests muscle strength, reflex activity, and sensation of toes and fingers. Lor subjects with hATTR with peripheral neuropathy, neurologic function can be assessed over time using NIS. NIS involves a neurologic exam of lower limbs upper limbs and cranial nerves with total score of 244 (weakness 192, sensation 32, reflexes 20). The Neuropathy Symptoms and Change (NSC) is a 38-item questionnaire that assesses the severity and change of symptoms (weakness, sensory, autonomic) of peripheral neuropathy with 3 scales: number of symptoms, severity of symptoms (mild=l , moderated, severed) which can be used to assess change over time, and a change category (comparing a symptom to baseline). Other scales are the Lamilial Amyloidotic Polyneuropathy (PAP) stage and the Polyneuropathy Disability (PND) score.

[00175] Diagnosis of TTR amyloidosis typically relies on target organ biopsies, followed by histological staining of the excised tissue with the amyloid-specific dye, Congo red. If a positive test for amyloid is observed, immunohistochemical staining for TTR is subsequently performed to ensure that the precursor protein responsible for amyloid formation is indeed TTR. Lor familial forms of the diseases, demonstration of a mutation in the gene encoding TTR is then needed before diagnosis can be made. This can be accomplished, for example, through isoelectric focusing electrophoresis, polymerase chain reaction, or laser dissection/liquid chromatography-tandem mass spectrometry. See, e.g., US 2014/0056904; Ruberg and Berk, Circulation 126: 1286-1300 (2012); Ando et al., Orphanet Journal of Rare Diseases 8:31 (2013).

IV. ANTIBODIES

A. Binding Specificity and Punctional Properties

[00176] The antibody 14G8 was originally isolated as a mouse antibody having a mature heavy chain variable region defined by SEQ ID NO:6l and a mature light chain variable defined by SEQ ID NO: 70. Rabat CDRH1, H2 and H3 have SEQ ID NOs:67-69 and CDRL1, L2 and L3 have SEQ ID NOs:77-79. A composite Chothia-Kabat CDR-H1 is provided as SEQ ID NO: l l8.

[00177] The antibody 9G5 was also originally isolated as a mouse antibody having a mature heavy chain variable region defined by SEQ ID NO:6l and a mature light chain variable defined by SEQ ID NO:70. Kabat CDRH1, H2 and H3 have SEQ ID NOs:l3-l5 and Rabat CDRL1, L2 and L3 have SEQ ID NOs:24-26. The Kabat CDRs of 9G5 are the same as those of 14G8 except that in 9G5 positions H52 and L26 by Kabat numbering are occupied by N and in 9G5 by S. A composite Chothia-Kabat CDR-H1 is provided as SEQ ID NO: 117.

[00178] CDRs can alternatively be defined by any of the following conventions described above.

[00179] The formuations of the invention include an antibody comprising a mature heavy chain comprising CDRs Hl, H2 and H3 of 14G8 and CDRs Ll, L2 and L3 of 14G8 except that positions H52 and L26 by Kabat numbering can each be independently N or S. Some antibodies include CDRs Hl, H2 and H3 of 14G8 and CDRs Ll, L2 and L3 of 14G8 wherein positions H52 and L26 are both occupied by N. Some antibodies include a mature heavy chain comprising CDRs HI, H2, H3 of 9G5 and a mature light chain comprising CDRs Ll, L2 and L3 of 9G5, wherein positions H52 and L26 are S.

[00180] Unless otherwise apparent from the context reference to 14G8 or 9G5 should be understood as referrring to any of the mouse, chimeric, veneered, and humanized forms of the mouse antibody these antibodies. These antibodies specifically bind within approximately amino acid residues 89-97 (SEQ ID NO: 113) of TTR. Such epitopes are buried in the native TTR tetramer and exposed in monomeric, misfolded, aggregated, or fibril forms of TTR.

[00181] Other antibodies can be obtained by mutagenesis of cDNA encoding the heavy and light chains of an exemplary antibody, such as 14G8 or 9G5. Monoclonal antibodies that are at least 70%, 80%, 90%, 95%, 96%, 97%, 98%, or 99% identical to 14G8 in amino acid sequence of the mature heavy and/or light chain variable regions and maintain its functional properties, and/or which differ from the respective antibody by a small number of functionally inconsequential amino acid substitutions (e.g., conservative substitutions), deletions, or insertions are also included in the invention. [00182] Antibodies including the CDRs of 14G8 or 9G5 as generally described above are characterized by their ability to bind to monomeric, misfolded, aggregated, or fibril forms of TTR preferentially over native tetrameric forms of TTR. In addition, these antibodies are characterized by their preferential immunoreactivity on TTR-mediated amyloidosis cardiac tissue but not on healthy cardiac tissue. Preferential binding or immunoreactivity can be relative e.g., at least 2 fold better or absolute (i.e., no binding or immunoreactivity to native TTR, or health cardiac tissue).

[00183] Some antibodies can inhibit or reduce aggregation of TTR, inhibit or reduce TTR fibril formation, reduce or clear TTR deposits or aggregated TTR, or stabilize non toxic conformations of TTR in an animal model or clinical trial. Some antibodies can treat, effect prophylaxis of, or delay the onset of a TTR amyloidosis as shown in an animal model or clinical trial. Exemplary animal models for testing activity against a TTR amyloidosis include those described in Kohno et al., Am. J. Path. 150(4): 1497- 1508 (1997); Teng et al., Laboratory Investigations 81 :385-396 (2001); Wakasugi et al., Proc. Japan Acad. 63B :344-347 (1987); Shimada et al., Mol. Biol. Med. 6:333-343 (1989);

Nagata et al., J. Biochem. 117: 169- 175 (1995); Sousa et al., Am. J. Path. 161 : 1935- 1948 (2002); and Santos et al., Neurobiology of Aging 31:280-289 (2010).

B. Humanized Antibodies

[00184] A humanized antibody is a genetically engineered antibody in which CDRs from a non-human“donor” antibody are grafted into human“acceptor” antibody sequences (see, e.g., Queen, US 5,530,101 and 5,585,089; Winter, US 5,225,539; Carter, US 6,407,213; Adair, US 5,859,205; and Foote, US 6,881,557). The acceptor antibody sequences can be, for example, a mature human antibody sequence, a composite of such sequences, a consensus sequence of human antibody sequences, or a germline region sequence. Thus, a humanized antibody is an antibody having at least three, four, five or all CDRs entirely or substantially from a donor antibody and variable region framework sequences and constant regions, if present, entirely or substantially from human antibody sequences. Similarly a humanized heavy chain has at least one, two and usually all three CDRs entirely or substantially from a donor antibody heavy chain, and a heavy chain variable region framework sequence and heavy chain constant region, if present, substantially from human heavy chain variable region framework and constant region sequences. Similarly a humanized light chain has at least one, two and usually all three CDRs entirely or substantially from a donor antibody light chain, and a light chain variable region framework sequence and light chain constant region, if present, substantially from human light chain variable region framework and constant region sequences. Other than nanobodies and dAbs, a humanized antibody comprises a humanized heavy chain and a humanized light chain. A CDR in a humanized antibody is substantially from a corresponding CDR in a non- human antibody when at least 85%, 90%, 95% or 100% of corresponding residues (as defined by any conventional definition, such as, for example, by Kabat) are identical between the respective CDRs. The variable region framework sequences of an antibody chain or the constant region of an antibody chain are substantially from a human variable region framework sequence or human constant region respectively when at least 85%, 90%, 95% or 100% of corresponding residues defined by any conventional definition, such as, for example, by Kabat are identical.

[00185] Although humanized antibodies often incorporate all six CDRs (defined by any conventional definition, such as, for example, by Kabat) from a mouse antibody, they can also be made with less than all CDRs (e.g., at least 3, 4, or 5 CDRs) from a mouse antibody (e.g., Pascalis et al., J. Immunol. 169:3076, 2002; Vajdos et al, J. of Mol. Biol., 320: 415-428, 2002; Iwahashi et al.. Mol. Immunol. 36:1079-1091, 1999; Tamura et al, J. Immunol., 164:1432-1441, 2000).

[00186] In some antibodies only part of the CDRs, namely the subset of CDR residues required for binding, termed the SDRs, are needed to retain binding in a humanized antibody. CDR residues not contacting antigen and not in the SDRs can be identified based on previous studies (for example residues H60-H65 in CDR H2 are often not required), from regions of Kabat CDRs lying outside Chothia hypervariable loops (Chothia, J. Mol. Biol. 196:901, 1987), by molecular modeling and/or empirically, or as described in Gonzales et al., Mol. Immunol. 41: 863, 2004. In such humanized antibodies at positions in which one or more donor CDR residues is absent or in which an entire donor CDR is omitted, the amino acid occupying the position can be an amino acid occupying the corresponding position (by Kabat numbering) in the acceptor antibody sequence. The number of such substitutions of acceptor for donor amino acids in the CDRs to include reflects a balance of competing considerations. Such substitutions are potentially advantageous in decreasing the number of mouse amino acids in a humanized antibody and consequently decreasing potential immunogenicity. However, substitutions can also cause changes of affinity, and substantial reductions in affinity are preferably avoided. Positions for substitution within CDRs and amino acids to substitute can also be selected empirically.

[00187] The human acceptor antibody sequences can optionally be selected from among the many known human antibody sequences to provide a high degree of sequence identity (e.g., 65-85% identity) between a human acceptor sequence variable region frameworks and corresponding variable region frameworks of a donor antibody chain.

[00188] Examples of acceptor sequences for the heavy chain are the human mature heavy chain variable regions with NCBI accession codes BAC02114 and AAX82494.1 (SEQ ID NOs:3 and 4) and heavy chain variable regions of human Kabat subgroup 3. BAC02114 shares the same canonical form as mouse 9D5 heavy chain. Other examples of acceptor sequences for the heavy chain are the human mature heavy chain variable regions with NCBI accession codes AAD30410.1 and AAX82494.1 (SEQ ID NOs:63 and 4, respectively) and heavy chain variable regions of human Kabat subgroup 1. AAD30410.1 and AAX82494.1 include two CDRs having the same canonical form as mouse 14G8 heavy chain. Examples of acceptor sequences for the light chain are the human mature light chain variable region with NCBI accession code

ABC66952 (SEQ ID NO:l8) and light chain variable regions of human Kabat subgroup 3. ABC66952 includes two CDRs having the same canonical form as mouse 9D5 light chain. Other examples of acceptor sequences for the light chain are the human mature light chain variable regions with NCBI accession codes ABA71374.1 and ABC66952.1 (SEQ ID NOs:72 and 73, respectively) and light chain variable regions of human Kabat subgroup 2. ABA71374.1 and ABC66952.1 have the same canonical form as mouse 14G8 light chain.

[00189] If more than one human acceptor antibody sequence is selected, a composite or hybrid of those acceptors can be used, and the amino acids used at different positions in the humanized light chain and heavy chain variable regions can be taken from any of the human acceptor antibody sequences used. For example, the human mature heavy chain variable regions with NCBI accession codes BAC02114 and AAX82494.1 (SEQ ID NOs:3 and 4) were used as acceptor sequences for humanization of the 9D5 mature heavy chain variable region. Examples of positions in which these two acceptors differ include positions H19 (R or K), H40 (A or T), H44 (G or R), H49 (S or A), H77 (S or T), H82a (N or S), H83 (R or K), H84 (A or S), and H89 (V or M). Humanized versions of the 9D5 heavy chain variable region can include either amino acid at any of these positions. Similarly, the human mature heavy chain variable regions with NCBI accession codes AAD30410.1 and AAX82494.1 (SEQ ID NOs:63 and 4, respectively) were used as acceptor sequences for humanization of the 14G8 mature heavy chain variable region. Examples of positions in which these two acceptors differ include positions H82a (N or S), H83 (R or K), H84 (A or S), and H89 (V or M). Humanized versions of the 14G8 heavy chain variable region can include either amino acid at any of these positions. Similarly, the human mature light chain variable regions with NCBI accession codes ABA71374.1 and ABC66952.1 (SEQ ID NOs:72 and 73, respectively) were used as acceptor sequences for humanization of the 14G8 mature light chain variable region. An example of a position in which these two acceptors differ is position L18 (S or P). Humanized versions of the 14G8 light chain variable region can include either amino acid at this position.

[00190] Certain amino acids from the human variable region framework residues can be selected for substitution based on their possible influence on CDR conformation and/or binding to antigen. Investigation of such possible influences is by modeling, examination of the characteristics of the amino acids at particular locations, or empirical observation of the effects of substitution or mutagenesis of particular amino acids.

[00191] For example, when an amino acid differs between a murine mature variable region framework residue and a selected human mature variable region framework residue, the human framework amino acid can be substituted by the equivalent framework amino acid from the mouse antibody when it is reasonably expected that the amino acid:

(1) noncovalently binds antigen directly,

(2) is adjacent to a CDR region,

(3) otherwise interacts with a CDR region (e.g. is within about 6 A of a CDR region)

(4) mediates interaction between the heavy and light chains. [00192] Framework residues from classes (1) through (3) as defined by Queen, US 5,530,101, are sometimes alternately referred to as canonical and vernier residues. Framework residues that help define the conformation of a CDR loop are sometimes referred to as canonical residues (Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987); Thornton & Martin, J. Mol. Biol. 263:800815 (1996)). Framework residues that support antigen-binding loop conformations and play a role in fine-tuning the fit of an antibody to antigen are sometimes referred to as vernier residues (Foote & Winter, J. Mol. Biol 224:487-499 (1992)).

[00193] Other framework residues that are candidates for substitution are residues creating a potential glycosylation site. Still other candidates for substitution are acceptor human framework amino acids that are unusual for a human immunoglobulin at that position. These amino acids can be substituted with amino acids from the equivalent position of the mouse donor antibody or from the equivalent positions of more typical human immunoglobulins.

[00194] For reasons such as possible influence on CDR conformation and/or binding to antigen, mediating interaction between heavy and light chains, interaction with the constant region, being a site for desired or undesired post-translational modification, being an unusual residue for its position in a human variable region sequence and therefore potentially immunogenic, getting aggregation potential, and other reasons, the following 15 variable region framework positions were considered as candidates for substitutions in the eight exemplified Hu9D5 mature heavy chain variable regions and the five exemplified Hu9D5 mature light chain variable regions, as further specified in the examples: H42 (G42E), FI47 (W47L), H69 (I69F), H82 (M82S), H82b (S82(b)L), H108 (T108L), L8 (P8A), L9 (L9P), L18 (P18S), L19 (A19V), L36 (Y36F), L39 (K39R), L60 (D60S), L70 (D70A), and L74 (K74R). Likewise, the following 11 variable region framework positions were considered as candidates for substitutions in the three exemplified Hul4G8 mature heavy chain variable regions and the three exemplified Hul4G8 mature light chain variable regions, as further specified in the examples: Hl (Q1E), H3 (Q3K), H47 (W47L), H105 (Q105T), L8 (P8A), L9 (L9P), L19 (A19V), L26 (N26S), L36 (Y36F), L60 (D60S), and L70 (D70A). [00195] Here, as elsewhere, the first-mentioned residue is the residue of a humanized antibody formed by grafting Kabat CDRs or a composite Chothia-Kabat CDR in the case of CDR-H1 into a human acceptor framework (e.g., a composite or hybrid human acceptor framework), and the second-mentioned residue is a residue being considered for replacing such residue. Thus, within variable region frameworks, the first mentioned residue is human, and within CDRs, the first mentioned residue is mouse.

[00196] Exemplified Hu9D5 antibodies include any permutations or combinations of the exemplified mature heavy and light chain variable regions (e.g., VHvl/VLvl or H1L1,

VHvi/VLv2 or H1L2, VHvi/VLv3 or H1L3, VHvi/VLv4 or H1L4, VHvi/VLv5 or H1L5, VHv2/VLvl or H2L1, VHv2/VLv2 or H2L2, VHv2/VLv3 or H2L3, VHv2/VLv4 or H2L4, VHv2/VLv5 or H2L5, VHv2b/VLvi or H2bLl, VHv2b/VLv2 or H2bL2, VHv2b/VLv3 or H2bL3, VHv2b/VLv4 or H2bL4, VHv2b/VLv5 or H2bL5, VHv3/VLvl or H3L1, VHv3/VLv2 or H3L2, VHv3/VLv3 or H3L3, VHv3/VLv4 or H3L4, VHv3/VLv5 or H3L5, VHv3bNLvl or H3bLl, VHv3b/VLv2 or H3bL2, VHv3b/VLv3 or H3bL3, VHv3b/VLv4 or H3bL4,

VHv3b/VLv5 or H3bL5, VHv4/VLvl or H4L1, VHv4/VLv2 or H4L2, VHv4/VLv3 or H4L3, VHv4/VLv4 or H4L4, VHv4/VLv5 or H4L5, VHv4b/VLvl or H4bLl, VHv4b/VLv2 or H4bL2, VHv4b/VLv3 or H4bL3, VHv4b/VLv4 or H4bL4, VHv4b/VLv5 or H4bL5, VHv5/VLvi or H5L1, VHv5/VLv2 or H5L2, VHv5/VLv3 or H5L3, VHv5/VLv4 or H5L4, and VHv5/VLv5 or H5L5).

[00197] The invention provides formulations of variants of humanized 9D5 antibodies in which the humanized mature heavy chain variable region shows at least 90%, 95%, 96%, 97%, 98%, or 99% identity to a humanized Hu9D5VHv4b (SEQ ID NO: 11) and the humanized mature light chain variable region shows at least 90%, 95%, 96%, 97%, 98%, or 99% identity to a Hu9D5VLvl (SEQ ID NO: 19). In some such antibodies, at least 1, 2, or all 3 of the backmutations or other mutations in Hu9D5 H4bLl are retained. The invention also provides variants of the other exemplified humanized 9D5 antibodies. Such variants have mature light and heavy chain variable regions showing at least 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to the mature light and heavy chain variable regions of the exemplified humanized 9D5 H1L1, H1L2, H1L3, H1L4, H1L5, H2L1, H2L2, H2L3, H2L4, H2L5, H2bLl, H2bL2, H2bL3, H2bL4, H2bL5, H3L1, H3L2, H3L3, H3L4, H3L5, H3bLl, Hb3L2, H3bL3, Hb3L4, H3bL5, H4L1, H4L2, H4L3, H4L4, H4L5, H4bLl, H4bL2, H4bL3, H4bL4, H4bL5, H5L1, H5L2, H5L3, H5L4, or H5L5 antibodies.

[00198] Variable regions framework positions are in accordance with Kabat numbering unless otherwise stated. Other such variants typically differ from the sequences of the exemplified Hu9D5 heavy and light chains by a small number (e.g., typically no more than 1, 2, 3, 5, 10, or 15) of replacements, deletions or insertions. Such differences are usually in the framework but can also occur in the CDRs.

[00199] Exemplified Hul4G8 antibodies include any permutations or combinations of the exemplified mature heavy and light chain variable regions (e.g., VHvl/VLvl or H1L1, VHvl/VLv2 or H1L2, VHvl/VLv3 or H1L3, VHv2NLvl or H2L1, VHv2/VLv2 or H2L2, VHv2/VLv3 or H2L3, VHv3/VLvl or H3L1, VHv3/VLv2 or H3L2, and VHv3/VLv3 or H3L3).

[00200] The invention provides formulations of variants of humanized 14G8 antibodies in which the humanized mature heavy chain variable region shows at least 90%, 95%, 96%, 97%, 98%, or 99% identity to Hul4G8VHv2 (Hul4G8 H2) (SEQ ID NO:65) and the humanized mature light chain variable region shows at least 90%, 95%, 96%, 97%, 98%, or 99% identity to Hul4G8VLv3 (Hul4G8 L3) (SEQ ID NO:76). In some such antibodies, at least 1, 2, 3, 4, or all 5 of the backmutations or other mutations in Hul4G8 H2L3 are retained. The invention also provides variants of the other exemplified humanized 14G8 antibodies. Such variants have mature light and heavy chain variable regions showing at least 90%, 95%, 96%, 97%, 98%, or 99% sequence identity to the mature light and heavy chain variable regions of the exemplified humanized 14G8 H1L1, H1L2, H1L3, H2L1, H2L2, H2L3, H3L1, H3L2, or H3L3 antibodies.

[00201] In some antibodies, at least one of positions Hl and H47 in the Vh region is occupied by E and L, respectively. In some antibodies, positions HI and H47 in the Vh region are occupied by E and L, respectively, as in Hul4G8VHv2 and Hul4G8VHv3. In some antibodies, at least one of positions H3 and H105 in the Vh region is occupied by K and T, respectively. In some antibodies, positions H3 and H105 in the Vh region are occupied by K and T, respectively, as in Hul4G8VHvl. In some antibodies, position L36 in the Vk region is occupied by F, as in Hul4G8VLv2. In some antibodies, at least one of positions L8, L9, L19, L26, L60, and L70 in the Vk region is occupied by A, P, V, S, S, and A, respectively. In some antibodies, positions L8, L9, L19, and L70 in the Vk region are occupied by A, P, V, and A, respectively, as in Hul4G8VLvl. In some antibodies, positions L26 and L60 in the Vk region are each occupied by S, as in Hul4G8VLv3. The CDR regions of such humanized antibodies can be identical or substantially identical to the CDR regions of the 14G8 mouse donor antibody .

The CDR regions can be defined by any conventional definition (e.g., Chothia, or composite of Chothia and Kabat), for example, as defined by Rabat.

[00202] Variable regions framework positions are in accordance with Kabat numbering unless otherwise stated. Other such variants typically differ from the sequences of the exemplified Hul4G8 heavy and light chains by a small number (e.g., typically no more than 1, 2, 3, 5, 10, or 15) of replacements, deletions or insertions. Such differences are usually in the framework but can also occur in the CDRs.

[00203] A possibility for additional variation in humanized 14G8 or 9G5 variants is additional backmutations in the variable region frameworks. Many of the framework residues not in contact with the CDRs in the humanized mAb can accommodate

substitutions of amino acids from the corresponding positions of the donor mouse mAh or other mouse or human antibodies, and even many potential CDR-contact residues are also amenable to substitution. Even amino acids within the CDRs may be altered, for example, with residues found at the corresponding position of the human acceptor sequence used to supply variable region frameworks. In addition, alternate human acceptor sequences can be used, for example, for the heavy and/or light chain. If different acceptor sequences are used, one or more of the backmutations recommended above may not be performed because the corresponding donor and acceptor residues are already the same without backmutations.

[00204] Preferably, replacements or backmutations in humanized 14G8 or 9G5 variants (whether or not conservative) have no substantial deleterious effect on the binding affinity or potency of the humanized mAb, that is, its ability to bind to monomeric TTR (e.g., the potency in some or all of the assays described in the present examples of the variant humanized 14G8 or 9G5 antibody is essentially the same or at least 90% of, i.e., within experimental error, as that of murine 14G8 or 9G5 antibody). c. Selection of Constant Region

[00205] The heavy and light chain variable regions of humanized antibodies can be linked to at least a portion of a human constant region. The choice of constant region depends, in part, whether antibody-dependent cell-mediated cytotoxicity, antibody dependent cellular

phagocytosis and/or complement dependent cytotoxicity are desired. For example, human isotopes IgGl and IgG3 have complement-dependent cytotoxicity and human isotypes IgG2 and IgG4 do not. Human IgGl and IgG3 also induce stronger cell mediated effector functions than human IgG2 and IgG4. Light chain constant regions can be lambda or kappa.

[00206] One or several amino acids at the amino or carboxy terminus of the light and/or heavy chain, such as the C-terminal lysine of the heavy chain, may be missing or derivatized in a proportion or all of the molecules. Substitutions can be made in the constant regions to reduce or increase effector function such as complement-mediated cytotoxicity or ADCC (see, e.g., Winter et al., US Patent No. 5,624,821; Tso et al., US Patent No. 5,834,597; and Lazar et al., Proc. Natl. Acad. Sci. USA 103:4005, 2006), or to prolong half-life in humans (see, e.g., Hinton et al., I. Biol. Chem. 279:6213, 2004). Exemplary substitutions include a Gln at position 250 and/or a Leu at position 428 (EU numbering is used in this paragraph for the constant region) for increasing the half-life of an antibody. Substitution at any or all of positions 234, 235, 236 and/or 237 reduce affinity for Fey receptors, particularly FcyRI receptor (see, e.g., US 6,624,821). An alanine substitution at positions 234, 235, and 237 of human IgGl can be used for reducing effector functions. Some antibodies have alanine substitution at positions 234, 235 and 237 of human IgGl for reducing effector functions. Optionally, positions 234, 236 and/or 237 in human IgG2 are substituted with alanine and position 235 with glutamine (see, e.g., US 5,624,821). In some antibodies, a mutation at one or more of positions 241, 264, 265, 270, 296, 297, 322, 329, and 331 by EU numbering of human IgGl is used. In some antibodies, a mutation at one or more of positions 318, 320, and 322 by EU numbering of human IgGl is used. In some antibodies, positions 234 and/or 235 are substituted with alanine and/or position 329 is substituted with glycine. In some antibodies, positions 234 and 235 are substituted with alanine, such as in SEQ ID NO: 102. In some antibodies, the isotype is human IgG2 or IgG4. [00207] An exemplary human light chain kappa constant region has the amino acid sequence of SEQ ID NO: 104. The N-terminal arginine of SEQ ID NO: 104 can be omitted, in which case light chain kappa constant region has the amino acid sequence of SEQ ID NO: 105. Antibodies can be expressed as tetramers containing two light and two heavy chains, as separate heavy chains, light chains, as Fab, Fab', F(ab')2, and Fv, or as single chain antibodies in which heavy and light chain mature variable domains are linked through a spacer.

[00208] Human constant regions show allotypic variation and isoallotypic variation between different individuals, that is, the constant regions can differ in different individuals at one or more polymorphic positions. Isoallotypes differ from allotypes in that sera recognizing an isoallotype bind to a non-polymorphic region of a one or more other isotypes. Reference to a human constant region includes a constant region with any natural allotype or any permutation of residues occupying positions in natural allotypes. Examplary heavy chain sequences including those having the amino acid sequences SEQ ID NOs: l0l-l03 with SEQ ID NO: 103, which is of IgGl Glm3 allotyp, preferred. Reference to a human constant region includes a constant region with any natural allotype or any permutation of residues occupying positions in natural allotypes.

D. Expression of Recombinant Antibodies

[00209] Antibodies can be produced by recombinant expression. Nucleic acids encoding the antibodies can be codon-optimized for expression in the desired cell-type (e.g., CHO or Sp2/0). Recombinant nucleic acid constructs typically include an expression control sequence operably linked to the coding sequences of antibody chains, including naturally-associated or heterologous promoter regions. The expression control sequences can be eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and the collection and purification of the crossreacting antibodies. The vector or vectors encoding the antibody chains can also contain a selectable gene, such as dihydrofolate reductase, to allow amplification of copy number of the nucleic acids encoding the antibody chains.

[00210] E. coli is a prokaryotic host particularly useful for expressing antibodies, particularly antibody fragments. Microbes, such as yeast are also useful for expression. Saccharomyces is a preferred yeast host, with suitable vectors having expression control sequences, an origin of replication, termination sequences and the like as desired. Typical promoters include 3-phosphoglycerate kinase and other glycolytic enzymes. Inducible yeast promoters include, among others, promoters from alcohol dehydrogenase, isocytochrome C, and enzymes responsible for maltose and galactose utilizations.

[00211] Mammalian cells can be used for expressing nucleotide segments encoding immunoglobulins or fragments thereof. See Winnacker, From Genes to Clones, (VCH

Publishers, NY, 1987). A number of suitable host cell lines capable of secreting intact heterologous proteins have been developed in the art, and include CHO cell lines, various COS cell lines, HeLa cells, HEK293 cells, L cells, and non-antibody-producing myelomas including Sp2/0 and NSO. It can be advantageous to use nonhuman cells. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen et al., Immunol. Rev. 89:49 (1986)), and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Suitable expression control sequences are promoters derived from endogenous genes, cytomegalovirus, SV40, adenovirus, bovine papillomavirus, and the like. See Co et ah, J. Immunol. 148:1149 (1992).

[00212] Having introduced vector(s) encoding antibody heavy and light chains into cell culture, cell pools can be screened for growth productivity and product quality in serum-free media. Top-producing cell pools can then be subjected ot FACS-based single-cell cloning to generate monoclonal lines. Specific productivities above 50 pg or 100 pg per cell per day, which correspond to product titers of greater than 7.5 g/L culture, can be advantageous.

Antibodies produced by single cell clones can also be tested for turbidity, filtration properties, PAGE, IEF, UV scan, HP-SEC, carboydrate-oligosaccharide mapping, mass spectrometery, and bining assay, such as ELISA or Biacore. A selected clone can then be banked in multiple vials and stored frozen for subsequent use.

[00213] Methodology for commercial production of antibodies including codon optimization, selection of promoters, transcription elements, and terminators, serum- free single cell cloning, cell banking, use of selection markers for amplification of copy number, CHO terminator, serum free single cell cloning, improvement of protein titers (see, e.g., US

5,786,464, US 6,114,148, US 6,063,598, US 7,569,339, W02004/050884, W02008/012142, W02008/012142, W02005/019442, W02008/107388, and W02009/027471 , and US

5,888,809).

[00214] Once expressed, antibodies can be purified according to standard procedures of the art, including protein A capture, column chromatography (e.g., hydrophobic interaction or ion exchange), low-pH for viral inactivation and the like (see generally, Scopes, Protein

Purification (Springer-Verlag, NY, 1982)).

[00215] Antibodies used to prepare the disclosed formulations are typically isolated or purified, i.e., substantially free of cellular material or other contaminating proteins from the cells in which they are produced, or substantially free of chemical precursors or other chemicals when chemically synthesized. For example, an antibody that is substantially free of cellular material includes preparations of the antibody having less than about 30%, 25%, 20%, 15%, 10%, 8%, 5%, 2%, 1%, 0.5%, 0.1%, or less (by dry weight) of contaminating protein. When an antibody is recombinantly produced, it is also substantially free of culture medium such that culture medium represents less than about 30%, 25%, 20%, 15%, 10%, 8%, 5%, 2%, 1%, 0.5%, 0.1%, or less, of the volume of the protein preparation. When an antibody is produced by chemical synthesis, it is preferably substantially free of or separated from chemical precursors or other chemicals involved in the synthesis of the protein. Accordingly, such antibody preparations have less than about 30%, 25%, 20%, 15%, 10%, 8%, 5%, 2%, 1%, 0.5%, 0.1%, or less (by dry weight) of chemical precursors or compounds other than the antibody drug substance. Recombinantly expressed antibody can be purified by methods, , such as, for example, affinity chromatography, acid treatment, depth filtration, anion exchange chromatography, cation exchange

chromatography, nanofiltration, ultrafiltration, dialysis and diafiltration.

[00216] The purified antibody drug substance can be adjusted to a solution comprising any of the formulations described herein, diluted to the desired concentration and stored until ready for use. Optionally, the formulation can be stored in concentrated form until ready for use. E. Conjugates

[00217] Antibodies used in the disclosed formulations can be coupled with a therapeutic moiety, such as a cytotoxic agent, a radiotherapeutic agent, an immunomodulator, a second antibody (e.g., to form an antibody heteroconjugate), or any other biologically active agent that facilitates or enhances the activity of a chimeric, veneered or humanized 14G8 or 9G5.

Representative therapeutic moieties include drugs that reduce levels of TTR, stabilize the native tetrameric structure of TTR, inhibit aggregation of TTR, disrupt TTR fibril or amyloid formation, or counteract cellular toxicity.

[00218] The antibodies disclosed herein can also be coupled or conjugated to one or more other antibodies (e.g., to form antibody heteroconjugates). Such other antibodies can bind to different epitopes within TTR or a portion thereof or can bind to a different target antigen.

[00219] Antibodies can also be coupled with a detectable label. Such antibodies can be used, for example, for diagnosing a TTR amyloidosis, for monitoring progression of a TTR amyloidosis, and/or for assessing efficacy of treatment. Such antibodies are particularly useful for performing such determinations in subjects having or being susceptible to a TTR amyloidosis, or in appropriate biological samples obtained from such subjects. Representative detectable labels that may be coupled or linked to an antibody include various enzymes, such as horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;

prosthetic groups, such streptavidin/biotin and avidin biotin; fluorescent materials, such as umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; luminescent materials, such as luminol;

bioluminescent materials, such as luciferase, luciferin, and aequorin; radioactive materials, such as yttrium 90 (90Y), radiosilver- l l l, radiosilver- 199, Bismuth 213 , iodine ( 131 1, 125 1, 123 1, 121 I), carbon ( 14 C), sulfur ( 5 S), tritium ( 3 H), indium ( 115 In, 113 In, 112 In, m In), technetium ( 99 Tc), thallium ( 201 Ti), gallium ( 68 Ga, 67 Ga), palladium ( 103 Pd), molybdenum ( 99 Mo), xenon ( 133 Xe), fluorine ( 18 F) , 153 Sm, 177 Lu , 159 Gd , 149 Pm, 140 La , 175 Yb , 166 Ho , 90 Y , 47 Sc , 186 Re , 188 Re , 142 Pr, 105 Rn , 97 RU, 68 Ge, 57 Co, 65 Zn, 85 Sr, 32 P, 153 Gd, 169 Yb, 51 Cr, 54 Mn, 75 Se, 113 Sn, and 117 Tin; positron emitting metals using various positron emission tomographies; nonradioactive paramagnetic metal ions; and molecules that are radiolabelled or conjugated to specific radioisotopes. [00220] Linkage of radioisotopes to antibodies may be performed with conventional bifunction chelates. For radio silver- 111 and radio silver- 199 linkage, sulfur-based linkers may be used. See Hazra et al, Cell Biophys. 24-25:1-7 (1994). Linkage of silver radioisotopes may involve reducing the immunoglobulin with ascorbic acid. For radioisotopes such as 11 lln and 90Y, ibritumomab tiuxetan can be used and will react with such isotopes to form 11 lln- ibritumomab tiuxetan and 90Y-ibritumomab tiuxetan, respectively. See Witzig, Cancer

Chemother. Phannacol., 48 Suppl LS91-S95 (2001).

[00221] Therapeutic moieties, other proteins, other antibodies, and/or detectable labels may be coupled or conjugated, directly or indirectly through an intermediate (e.g., a linker), to an antibody of the invention. See e.g., Arnon et al, "Monoclonal Antibodies For

Immunotargeting Of Drugs In Cancer Therapy," in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Flellstrom et al, "Antibodies For Drug Delivery," in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, "Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review," in Monoclonal Antibodies 84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475506 (1985); "Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy," in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303- 16 (Academic Press 1985); and Thorpe et al, Immunol. Rev., 62:119-58 (1982). Suitable linkers include, for example, cleavable and non-cleavable linkers. Different linkers that release the coupled therapeutic moieties, proteins, antibodies, and/or detectable labels under acidic or reducing conditions, on exposure to specific proteases, or under other defined conditions can be employed.

Y. Formulations

[00222] Formulations (also known as pharmaceutical compositions) of the invention comprise any of the monoclonal antibodies described in this application including chimeric, veneered or humanized version of antibody 14G8 or 9G5, a buffer, one or more sugars and/or polyols and a surfactant, and have a pFI within the range from about 4.5 to about 7.5. Other components (besides water in liquid formulations), such as, for example, arginine, lysine, NaCl, sorbitol or mannitol may or may not be present. The formulations can be in liquid or in lyophilized form. Liquid formulations can refer to a formulation before lyophilization or after reconstitution of a lyophilized formulation. In general, components of a formulation other than water occur in the same relative proportions by weight or moles in a lyophilized formulation as in a liquid formulation prior to lyophilization. Likewise, components of the formulation after reconsitution with water are in general in the same relative proportions as in the formulation prelyophilization or the lyophilized formulation but the absolute concentrations can change in proportion to the relative volumes of the formulation pre and post reconstitution. The volume post reconstitution can be the same, less or more than the volume prelyophilization. Usually the volume post reconstitution is the same within a factor of 5, 3, 2, 1.5, 1.2 or 1.1 of the volume prelyophilization. If for example, the volume post reconstitution is twice the volume prelyophilization the concentrations of components are approximately half that post

reconstitution as prelyophilization.

[00223] In liquid formulations, the antibody can be present at a concentration within a range from about 10-100, 15-80, 20-65, 25-75, 40-65, 45-65 mg/mL, among others. In some formulations, the antibody is present at 55-65 mg/ml prelyophilization and 45-55 mg/ml after reconstitution. In some formulations, the antibody is present at 50 mg/ml after reconstitution.

[00224] Formulations include a buffer, such as, for example, citrate, histidine, phosphate or succinate, to confer a pH range of from about 4.5 to about 7.5, for example, a pH of 5.5, 5.6, 5.7, 5.8, 5.9, 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7.0. Some formulations have a pH of between about 5.5 to about 7.0, or about 5.5 to about 6.5, or about 5.5 to about 6.0, or about 6.0 to about 6.5, or about 6.0 to about 7.0, or about 5.75-6.25. Some formulations have a pH of about 6.0 and some formulations have a pH of about 6.5. In some formulations, histidine is present at a concentration within a range of about 10-30 mM, or 15-25 mM, for example at a concentration of about 10 mM, 20 mM or 25 mM. In some formulations, citrate is present at a concentration within a range of about 10-30 mM, for example at a concentration of about 10 mM or 20 mM. In some formulations, phosphate is present at a concentration of about 20 mM. In some formulations, succinate is present at a concentration of about 20 mM. [00225] Formulations include a sugar/polyol, such as, for example, trehalose or sucrose. The sugar/polyol can be present at about 30 mM to about 260 mM, or about 150-350 mM, or about 200-300 mM, or about 220-260 mM, or about 230-250 mM, or about 205-240 mM, or about 205-250 mM or about 205-260 mM, or, or about 230-250 mM, or about 230-240 mM, or about 30 mM, about 205 mM or about 240 mM. In some formulations, trehalose is present at a concentration within a range of about 205-260 mM, about 205-250 mM, or about 205-240 mM, such as, for example, about 205 mM, about 230 mM or about 240 mM. In some formulations, sucrose is present at a concentration within a range of about 30-260 mM, about 30-250 mM, or about 30-240 mM, such as, for example, about 30 mM, about 230 mM or about 240 mM.

[00226] Formulations include a surfactant, such as, for example, polysorbate 20 (PS20), polysorbate 80 (PS 80) or a poloxamer, for example, poloxamer 188 (also known as PX188, PLURONIC F68 or FLOCOR). The surfactant can be present at a concentration within the range from about 0.01%-0.1%, 0.02%-0.04%, or 0.03%-0.05% by weight. For example, the concentration can be 0.005%, 0.01%, 0.015%, 0.02%, 0.025%, 0.03%, 0.035%, 0.04%, 0.045%, or 0.05% by weight. Poloxamers are nonionic triblock copolymers composed of a central hydrophobic chain of polyoxypropylene (poly(propylene oxide)) flanked by two hydrophilic chains of polyoxyethylene (poly(ethylene oxide)). Some formulations include about 0.02 % w/w PS20, about 0.02% w/w PS80 or about 0.04% poloxamer, for example, 0.04% poloxamer PX188 by weight.

[00227] Some such formulations are characterized by an osmolality in the range of about 270 mOsm/kg to about 330 mOsm/kg, such as, for example, about 335 mOsm/kg.

[00228] An exemplary formulation characterized by a pH within the range from about 5.0 to about 6.5 includes (a) a chimeric, veneered or humanized version of antibody 14G8 comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:61 and a mature light chain variable region comprising three CDRs of SEQ ID NO:70, except that positions H52 and/or L26 by Rabat numbering can be N or S, or a chimeric, veneered or humanized version of anti-TTR antibody 9G5 comprising a mature heavy chain variable region comprising three CDRs of SEQ ID NO:l and a mature light chain variable region comprising three CDRs of SEQ ID NO: 16, wherein the antibody is present at a concentration within the range from about 25 mg/mL to about 75 mg/mL; (b) histidine, citrate, phosphate or succinate present at a concentration of about 20 mM; (c) sucrose or trehalose present at a concentration within the range from about 295 mM to about 240 mM or if the sugar is absent 160 mM arginine is present; and (d) a surfactant present at a concentration within the range from about 0.01% to about 0.1% by weight; provided that (i) if histidine or succinate buffer is present, the pH is about 6.0, (ii) if phosphate buffer is present, the pH is about 6.5, and if histidine and trehalose are present the surfactant is PS 80 or PS20, provided that if PS20 is present, 25 mM L- arginine is also present.

[00229] Some formulations are essentially free of mannitol or sorbitol or both mannitol and sorbitol. In some formulations, the buffer comprises histidine, such as, for example, a histidine buffer. In some such formulations, the sugar can be present in a range from about 230 mM to about 240 mM. In some formulations, the sugar is sucrose and the surfactant is PS20 or PX188, for example PS20 at a concentration of 0.02% w/w or PX188 at a concentration of 0.04% w/w. In other formulations, the sugar is trehalose. In some formulations l60mM arginine is present. The buffer of some formulations can be citrate. In some such formulations, the sugar is present at 230mM. In some such formulations the surfactant is 0.02% PS20. Alternatively, the buffer can be phosphate. In some such formulations, sucrose is present. In other formulations, the buffer is succinate. In some such formulations sucrose is present. In some formulations comprising histidine, trehalose is present, for example at a concentration of 205 mM.

[00230] Exemplary formulations include about (a) 20 mM citrate, 230 mM trehalose and 0.02% w/w PS20 at pH5; (b) 20 mM histidine, 230 mM sucrose and 0.02% w/w PS20; (c) 20 mM phosphate, 230 mM sucrose and 0.02% w/w PS20 at pH6.5; (d) 20 mM citrate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (e) 20 mM histidine, 230 mM trehalose and 0.02% w/w PS80; (f) 20 mM histidine, 0.02% w/w PS20 and 160 mM L-arginine; (g) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188; (h) 20 mM succinate, 240 mM sucrose and 0.022% w/w PS20 at a pH of 6.0 and (i) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L- arginine. Some of such formulations consist essentially of the antibody and (a) 20 mM citrate, 230 mM trehalose and 0.02% w/w PS20 at pH5; (b) 20 mM histidine, 230 mM sucrose and 0.02% w/w PS20; (c) 20 mM phosphate, 230 mM sucrose and 0.02% w/w PS20 at pH6.5; (d) 20 mM citrate, 230 mM sucrose and 0.02% w/w PS20 at pH 6.5; (e) 20 mM histidine, 230 mM trehalose and 0.02% w/w PS80; (f) 20 mM histidine, 0.02% w/w PS20 and 160 mM L-arginine; (g) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188; (h) 20 mM succinate, 240 mM sucrose and 0.022% w/w PS20 at a pH of 6.0 or (i) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L-arginine. For example, the formulation can consist essentially of the antibody and about (a) 20 mM histidine, 240 mM sucrose and 0.04% w/w PX188; (b) 20 mM succinate, 240 mM sucrose and 0.02% w/w PS20 at a pH of 6.0; or (c) 20 mM histidine, 205 mM trehalose, 0.02% w/w PS20 and 25 mM L-arginine.

[00231] For example, the formulation can include (a) an antibody comprising a mature light chain having the amino acid sequence set forth as SEQ ID NO:65 and a mature heavy chain comprising an amino acid sequence set forth as SEQ ID NO:76, which is present at a

concentration of about 45-65 mg/mL; (b) a histidine buffer at a concentration of about 15-25 mM; (c) sucrose at a concentration of about 220-260 mM; (d) poloxamer at a concentration of about 0.03-0.05%; and a pH of about 5.75-6.25. The formulation can include (a) an antibody comprising a mature light chain having the amino acid sequence set forth as SEQ ID NO:65 and a mature heavy chain comprising an amino acid sequence set forth as SEQ ID NO:76, which is present at a concentration of about 45-65 mg/mL; (b) a histidine buffer at a concentration of about 20 mM; (c) sucrose at a concentration of about 240 mM; (d) poloxamer at a concentration of about 0.04%; and a pH of about 6. For example, the formulation can consist essentially of an antibody comprising a mature light chain having the amino acid sequence set forth as SEQ ID NO:65 and a mature heavy chain comprising an amino acid sequence set forth as SEQ ID NO:76, and about 20 mM histidine, about 240 mM sucrose and about 0.04% w/w PX188.

[00232] Lyophilized formulations include any of the antibodies described herein, (b) a buffer, such as histidine; (c) a surgar/polyol such as sucrose; and (d) a surfactant, such as a poloxamer. In some lyophilized formulations, any residual water constitutes less than 5% by weight and in some such formulations less than 2 or 1% by weight of the formulation. The amounts of components depend on the volume lyophilized but can be for example about 100- 300, 150-250 or 225-275 mg of antibody, about 15-35 or 15-19 mg buffer, and about 0.2 - 2.5 or 2.0-2.5 mg surfactant and about 400-490 mg of sugar or polyol. An exemplary lyophilized formulation includes 225-275 mg of a humanized 14G8 antibody, about 15-19 mg of L- histidine, about 400-490 mg sucrose and about 2.0-2.5 mg poloxamer, or the same components present in the same proportions but in different amounts. Lyophilized formulations can be prepared from any of the liquid formulations described above. One such lyophilized formulation consists essentially of about 250 mg of a humanized 14G8 antibody, about 16.8 mg of L-histidine, about 2.2 mg of poloxomer PX188 and about 445.3 mg of sucrose or different amounts of the same components in the same proportions. Lyophilized formulations can be stored frozen (e.g., -20 °C), in the cold (e.g., 4 °C) or at room temperature (e.g., 22 °C). An exemplary vial size for a lyophilized formulation is 20 ml.

[00233] Lyophilized formulations can be reconstituted by combining with suitable liquid, for example, sterile water. Lyophilized formulations can be reconstitutable with sterile water to a particle-free solution by eye within less than 5, 4, 3, 2 minutes. Reconstitution can be measured to the same volume (+/-20%) as that of the liquid formulation before lyophilization.

Reconstitution to a certain desired final volume, for example, about 5 ml, can be achieved by adding a certain amount of liquid taking into account the volume occupied by the dry components. For example, some lyophilized formulations can be reconstituted to a total volume of about 5 ml by adding about 4.9 ml sterile water. The reconstitution can result in the components having approximately the same concentrations, relative and absolute, as before lyophilization or can result in approximately the same relative concentrations as before lyophilization but deceased or increased absolute concentrations. Some lyophilized formulations are constituted in a volume of about 1.2 fold the volume of the prelyophilized formulation resulting in a decrease of concentrations of about 17%. Some reconstituted formulations include an antibody concentration of about 40-60 mg/mL, for example, about 50 mg/mL; (b) a histidine buffer present at a concentration of about 15-25 mM, for example, about 20 mM; (c) sucrose present at a concentration of about 200-300 mM, for example, about 240 mM; (d) poloxamer present at a concentration of about 0.01% to about 0.1% by weight, for example, about 0.04% by weight; and (e) a pH of about 5.5-6.5, for example, about 6.0.

[00234] Liquid or reconstituted lyophilized formulations can be substantially isotonic implying an osmolality of about 250-350 mOsm/kg water. Some formulations have an osmolality of about 335 mOsm/kg. Some formulations have an osmolality of 270-300 mOsm/kg. Liquid or reconstituted lyophilized formulations can also be hypertonic > 350 mOsm/kg water or hypotonic (<250 mOsm/kg water).

[00235] Liquid formulations (typically after reconsitution) can be added to infusion bag containing a diluent such as normal saline or Ringer’s solution before administration to the patient. The volume of the infusion bag is usually relatively large (e.g., 50 ml to 1 L, or 100-500 ml) compared with the volume of the liquid formulation or constituted lyophilized formulation (e.g., 1-10 ml). Several liquids can be used in the infusion bag, such as normal saline, lactated Ringers solution, or 5% dextrose solution, each of which is substantially isotonic. In an exemplary regime about 5 ml of liquid or reconstituted lyophilized formulation is injected through the port of a lOO-ml bag of normal saline and administered by iv infusion over a period of about an hour at a flow rate of about 1.75 ml/min.

[00236] Formulations intended for administration to humans are preferably made under good manufacturing practices (GMP) approved or approvable by the FDA or a regulatory agency for a country other than the United States, for example, the European Medicines Agency, for preparation of drugs for administration to humans. Typically, the formulations are sterile, for example, as accomplished by sterile filtration using a 0.2 pm or a 0.22 pm filter.

[00237] Stability of formulation can be assessed after storage in the lyophilized form followed by reconstitution. Examplary formulation are stable at 38°C-42°C (e.g., as assessed by high performance size exclusion chromatography (HPSEC)) after storage in lyophiliized form for at least about 30 days, formulations having stability at 20°C-24°C after storage for at least about 1 year, and formulations having stability at 2°C-4°C after storage for at least about 3 years. A formulation is considered stable if, after incubation at one or more of these specified combinations of time and temperature, it meets the below definition for low to undetectable fragmentation and/or low to undetectable aggregation. More particularly, the disclosed formulations exhibit low to undetectable levels of antibody aggregation and/or fragmentation, or a low or undetectable increase in antibody fragmentation and/or aggregation above an initial level (e.g., less than about 10% aggregation). Some formulations exhibit < about 5% combined aggregation and/or fragmentation. A formulation having low to undetectable levels of fragmentation contains at least about 80%, 85%, 90%, 95%, 97%, 98%, or 99%, of the total protein, for example, in a single peak as determined by high performance size exclusion chromatography (HPSEC), or in two peaks (one corresponding to each of the antibody heavy chains and antibody light chains) by reduced Capillary Gel Electrophoresis (rCGE), representing the non-degraded antibody, and containing no other single peaks having more than 5%, more than 4%, more than 3%, more than 2%, more than 1%, or more than 0.5% of the total protein each. A formulation having low to undetectable levels of aggregation contains no more than about 15%, no more than about 10%, no more that about 5%, no more than about 4%, no more than about 3%, no more than about 2%, no more than about 1%, or no more than about 0.5% aggregation by weight protein, as measured by high performance size exclusion chromatography (HPSEC). For example, in some formulations, less than about 10% of the anti-synuclein antibody is present as an aggregate. Stable formulations of the invention also show little or no loss of biological activity(ies) of antibody having, for example, binding affinity measurable by ELISAs and/or additional functional assay, that is at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% of an initial measurable value. Some formulations have a binding affinity that is from about 60% to about 140% of an initial measurable value of the reference material.

VI. Methods of Treatment

[00238] Formulations of the invention can be used for treating or effecting prophylaxis of a disease in a patient having or at risk for the disease mediated at least in part by transthyretin (TTR), and particularly by monomeric, misfolded, aggregated, or fibril forms of TTR. . In some methods of treatment, the patient has been diagnosed with ATTR amyloidosis. Some such patients may have ATTR cardiac involvement and/or peripheral neurpathy involvement. Some patients have wild-type ATTR-cardiomyopathy in which normal, 'wild type' TTR proteins clump together and form amyloid deposits. Some patients have hereditary ATTR-cardiomyopathy. Some patients have hereditary polyneuropathy.

[00239] Formulations are administered in an effective regime meaning a dosage, route of administration and frequency of administration that delays the onset, reduces the severity, inhibits further deterioration, and/or ameliorates at least one sign or symptom of a disorder being treated. If a patient is already suffering from a disorder, the regime can be referred to as a therapeutically effective regime. If the patient is at elevated risk of the disorder relative to the general population but is not yet experiencing symptoms, the regime can be referred to as a prophylactically effective regime. In some instances, therapeutic or prophylactic efficacy can be observed in an individual patient relative to historical controls or past experience in the same patient. In other instances, therapeutic or prophylactic efficacy can be demonstrated in a preclinical or clinical trial in a population of treated patients relative to a control population of untreated patients.

[00240] The frequency of administration depends on the half-life of the antibody in the circulation, the condition of the patient and the route of administration among other factors. The frequency can be daily, weekly, monthly, quarterly, or at irregular intervals in response to changes in the patient's condition or progression of the disorder being treated. An exemplary frequency for intravenous administration is between weekly and quarterly over a continuous cause of treatment, for example, once every four weeks, although more or less frequent dosing is also possible. For subcutaneous administration, an exemplary dosing frequency is daily to monthly, although more or less frequent dosing is also possible.

[00241] The number of dosages administered depends on whether the disorder is acute or chronic and the response of the disorder to the treatment. For acute disorders or acute exacerbations of a chronic disorder, between 1 and 10 doses are often sufficient. Sometimes a single bolus dose, optionally in divided form, is sufficient for an acute disorder or acute exacerbation of a chronic disorder. Treatment can be repeated for recurrence of an acute disorder or acute exacerbation. For chronic disorders, an antibody can be administered at regular intervals, e.g., weekly, fortnightly, monthly, quarterly, every six months for at least 3 months, 12 months, 5 years, 10 years, or the life of the patient.

[00242] A regime is considered therapeutically or prophylactically effective if an individual treated patient achieves an outcome more favorable than the mean outcome in a control population of comparable patients not treated by methods of the invention, or if a more favorable outcome is demonstrated in treated patients versus control patients in a controlled clinical trial (e.g., a phase II, phase II/III or phase III trial) at the p < 0.05 or 0.01 or even 0.001 level. [00243] Effective doses vary depending on many different factors, such as means of administration, target site, physiological state of the subject, whether the subject is human or an animal, other medications administered, and whether treatment is prophylactic or therapeutic.

[00244] An exemplary dose range for antibodies can be from about 0.1 to 80 mg/kg, 0.1- 30, 0.5-5, or 1-10 mg/kg body weight (e.g., 0.1, 0.2, 0.3, 1.0, 3.0, 10.0 or 30.0 mg/kg) or 10- 5000, e.g., 10-1500 mg as a fixed dosage. Some methods increase the dose with time. The dose can be increased in increments of one (i.e., one change of dose), two, three or more. There can be 1, 2, 3 or more dosages at each level prior to the final level. The final level can be continued for e.g., at least 15 doages. For example, an initial dose can be in the range 0.1 mg/kg to 3 mg/kg for three infusions one every 28 days, then three infusions at an intermediate level also every 28 days, followed by dosing at a higher level of e.g., 10-30 mg/kg every 38 hours, optionally at least 15 times. The dosage depends on the condition of the patient and response to prior treatment, if any, whether the treatment is prophylactic or therapeutic and whether the disorder is acute or chronic, among other factors.

[00245] Formulations can be administered via a peripheral route. Routes of administration include topical, intravenous, oral, subcutaneous, intraarterial, intracranial, intrathecal, intraperitoneal, intranasal or intramuscular. For example, routes for administration of the formulations provided herein can be intravenous or subcutaneous. Intravenous administration can be, for example, by infusion over a period within a range of 30-180 minutes, such as 30-90 minutes, 60-120 minutes, or 90-180 minutes. This type of injection is most typically performed in the arm or leg muscles. In some such methods, the patient may be premedicated with a pain reliever or an antihistamine or diphenhydramine. For example, diphenhydramine can be administered to the patient within a range of about 30 to 90 minutes prior to the antibody administration. Some patients may be premedicated with acetominophen within a range of about 30 to 90 minutes prior to the antibody administration. In some methods, agents are injected directly into a particular tissue where deposits have accumulated, for example, intracranial injection.

[00246] Fiquid formulations can be administered directly to the patient, for example, by subcutaneous injection or by infusion. Lyophilized formulations are reconstituted in a liquid, for example, sterile water prior to administration to the patient. For infusion, the liquid formulation is introduced into a bag of isotonic fluid, for example, normal saline. For example, a lyophilized dosage form of the formulation can be reconstituted to a volume of about 5.0 mL with sterile water, and diluted in normal saline for infusion, for example, for a total infusion volume of 100 mL, 250 mL, or 500 mL.

[00247] The regimes comprising the formulated antibodies herein can be administered in combination with other therapies to a subject having or at risk of a transthyretin- mediated amyloidosis. Optionally, in addition to administering the formulated antibody herein, such therapies are administered sequentially or separately to a subject in need thereof. Optionally, the subject no longer receives the treatment with the pharmaceutical formulation comprising the formulated antibodies.

[00248] These therapies include TTR stabilizers, such as tafamidis and diflunisal, gene therapies to suppress TTR expression and subsequent TTR protein production including use of small interfering RNAs, such as patisiran and inotersen, and antisense oligonucleotides, as well as amyloid degraders including 4'-iodo-4'-deoxydoxorubicin (IDOX), doxycycline,

tauroursodeoxycholic acid (TUDCA), and cyclodextrin (CyD) and anti-SAP antibodies.

[00249] TTR stabilizers, such as tafamidis (Pfizer’s Vyndaquel) (see, e.g.,

W02G1 1 116123, US 9,150,489) and diflunisal (generic), can be used to maintain TTR’s normal soluble tetrameric structure and to limit the number of TTR monomers in the circulation. For example, tafamidis is under active investigation as a novel compound that binds to the thyroxine- binding sites of the TTR tetramer, inhibiting its dissociation into monomers and blocking the rate-limiting step in the TTR amyloidogenests cascade. Diflunisal binds and stabilizes common familial TTR variants against acid-mediate fibril formation.

[00250] RNA inhibiting therapies bind the targeted mRNA and thereby suppress mRNA expression and prevent translation of the corresponding protein. The clinical results so far have indicated that small interfering RNA, e.g , patisiran or revusiran (see, e.g., WO 2016033326), and antisense oligonucleotide, e.g., inotersen (see., e.g., US 8,101,743 and 9,061,044), are potent approaches to eliminate TTR protein production by triggering the degradation of TTR mRNA or inhibiting translation. [00251] Patisiran, a siRNA therapy is being investigated in a Phase III multicenter clinical trial for hereditary ATT amyloidosis patients with polyneuropathy (hATTR-PN). Also under investigation, is inotersen (IONIS-TTRRx), a subcutaneously administered antisense oligonucleotide targeting the same patient group as patisiran.

[00252] Antisense oligonucleotides (AS Os) are under clinical investigation for their ability to inhibit hepatic expression of amyloidogenic TTR protein. Currently, the ASO compound, ISIS-TTR Rx , is under investigation in a phase 3 multicenter, randomized, double blind, placebo-controlled clinical trial in patients with familial amyloid polyneuropathy (FAP).

[00253] Amyloid degraders such as the doxycycline/tauroursodeoxychoiie acid combination therapy have the potential to remove TTR amyloid deposited in the organs. For example, combined doxycycline and tauroursodeoxycholic acid (TUDCA) disrupt TTR amyloid fibrils and appeared to have an acceptable safety profile in a small phase 2 open- label study among 20 TTR patients.

[00254] Another example of TTR degraders are anti-SAP antibodies. Anti-SAP antibodies are antibodies against a normal non- fibrillar glycoprotein SAP, which promotes a giant cell reaction that removes visceral amyloid deposits.

X. Examples

Example 1. Pre-Formulation Testing at Low Concentration

[00255] Pre-formulation development studies were conducted on a humanized 14G8 antibody having the mature heavy chain sequence of SEQ ID NO:82 (except that the C -terminal lysine can be absent) and the mature light chain sequence of SEQ ID NO:86. Pre-formulation testing was performed with the humanized 14G8 antibody in 20 formulations containing combinations of various buffers in a pH range of from about 4.5 to about 7.0 with and without certain sugars, surfactants and other excipients.

[00256] The properties of the formulations were tested by methods including visual inspection, dynamic light scattering (DLS) micro-flow imaging (MFI), differential scanning fluorimetry (DSF), differential scanning calorimetry (DSC), and high performance size exclusion chromatography (HP-SEC). [00257] The buffers tested were citrate, histidine, succinate and phosphate in a

concentration ranging from about 10 mM to about 20 mM. The sugars tested were trehalose and sucrose in a concentration ranging from about 205 mM to about 230 mM. Formulations having trehalose in a citrate or histidine buffer were tested, as were formulations having sucrose in any of the four buffers.

[00258] The surfactants tested were PS20, PS 80 and poloxamer 188 in a concentration ranging from about 0.02% w/w to about 0.04% w/w. PS20 was tested in various formulations. Poloxomer and PS80 were tested only in one formulation, which contained trehalose in a histidine buffer.

[00259] Additional excipients tested were L- arginine, L-lysine and NaCl in

concentrations ranging from about 25 mM to about 160 mM in histidine formulations with and without trehalose or PS20.

[00260] The results of the formulations under each of these tests (F1-F20) were assigned values according to the relative extent of change compared to TO. Most favorable were circumstances under which no changes were observed. Unacceptable were those circumstances under which large changes were observed. Additional values were assigned to circumstances under which small changes were observed and under which intermediate changes were observed. Tables 2-3 report the results of subjecting the samples to freeze-thawing stress (T-FT) and storage for 1 week at 50° C (T-50) as determined by visual inspection, MFI, DLS and HP-SEC. Table 2 provides a list of common properties of formulations for which no large changes were observed in any of the tests. Overall, formulations F13 and F20, both of which lacked surfactant, showed the worst performance upon stressing. Formulations with a pH of 4.5 or less or 7.0 or greater generally did not perform as well as other formulations. 10 mM buffer concentrations did not perform as well as 20 mM buffer concentrations. The presence of sugars was beneficial, although formulations with no sugars and a high concentration of L-arginine performed better than formulations lacking both sugars and high concentrations of L-arginine. [00261] Table 2 provides alist of conclusions from formulations exhibiting respectable performance in all tests (i.e., no large changes).

Table 2

[00262] Table 3 provides a list of specific excipient combinations that did not show large changes under any particular test.

Table 3

[00263] However, because all of the formulations exhibited intermediate changes in at least one of the tests, further modifications of the formulations were performed, as discussed in Example 2, to create formulations that would provide the most stabilizing conditions for the antibody.

Example 2: Formulation Screening at 50 mg/ml

[00264] Based on the results from the pre-formulation screening, formulations F21-F31 listed in Table 4 were subjected to testing at a concentration of 50 mg/ml antibody. [00265] Table 4: List of Formulations Tested at 50 mg/ml

Table 4

[00266] The samples were subjected to visual inspection, glass transition temperature, MFI, HP-SEC and cIEF tests as appropriate after preparation (T-liquid), lyophilization (TO), two weeks storage in liquid state at 25°C and one month and three months storage of lyophilized forms at 2-8°C, 25°C and 40°C. The results of the formulations under each of these tests were assigned values of“bad”,“acceptable”,“good” and“very good”. Acceptable formulations had 4000-6000 particles > l0pm/ml (F27), greater than 94.0% monomer and less than 3% high molecular weight species (F22-F27 and F30), and greater than 74% main isoforms after storage for three months at 40°C/75% r.h.. Table 5 provides a list of common properties of formulations that did not receive a“bad” score in any of the tests performed.

[00267] Table 5: List of conclusions from formulations exhibiting acceptable to very good performance in all tests.

Table 5

[00268] Table 6: List of formulations exhibiting acceptable to very good performance in all tests.

Table 6

[00269] Formulation F25 was found to be superior to all formulations tested, exhibiting less than 4000 particles/ml, greater than 96.0% monomer with less than 2.5% high molecular weight species, and 77% main isoforms (determined by cIEF) in the liquid formulation stored for 2 weeks at 25°C and in the lyophilized forms stored for 1 month and 3 months at temperatures tested (40°C). Based on these studies, Formulation 25 (20 mM histidine-HCl, 240 mM sucrose, 0.04% poloxamer, pH 6.0) was identified as the lead candidate for further development as described in Example 3.

Example 3: Preparation and Characterization of Formulation F25

[00270] Sample Preparation: The liquid formulation prelyophilization was 61 mg/ml humanized 14G8, 20 mM histidine, 240 mM sucrose and 0.04% poloxamer. After lyophilization, the formulation was reconstituted to an antibody concentration of about 50 mg/ml.

[00271] The frozen starting material as described above was thawed in a water bath at 20 °C. After thawing, material of two batches was mixed and protein concentration was determined. The aliquot for the second batch was frozen again to -80°C until further usage. The sample concentration was adjusted by dilution with formulation buffer to obtain a target concentration of 50 mg/ml. Surfactant was added by spiking an aliquot of a concentrated stock solution in water directly into the formulation. Then , the sample was filtered through 0.22-pm PVDF membrane filters under a laminar flow hood. Finally, the filtered formulation was filled into pre-cleaned glass type I, 20R vials; each vial with a filling volume of 5 ml prior to loading onto freeze-dryer shelves.

[00272] Lyophilization: Lyophilization of the samples was performed by using an Epsilon 2-12D pilot scale freeze-dryer (Martin Christ, Osterode, Germany). The lyophilization processes were based on the critical product temperatures of the chosen formulation (Tg' of -25.7 °C and a Tconset of -25.3 °C) and were developed for the active DP. The starting point for selected conditions to perform freeze drying was based on published guidelines (see e.g., Carpenter JF et ah, Pharm Res. 1997 August, 4(8):969-75; Jameel F et ah, Book chapter 30 in Formulation and Process Development Strategies for Manufacturing Biopharmaceuticals, published online:

August 2010; and Remmele RL et al, Curr Pharm Bio techno 1., 2012 March, l3(3):471-96). A standard freezing protocol was applied that is used for sucrose-based formulations by cooling to - 45 °C with a rate of 0.5 °C/minute. Initial guess for the shelf temperature during primary drying was maintained such that resulting product temperature is at or below the measured Tg' and measured collapse temperature of the formulation, in order to avoid collapse during primary drying. A chamber pressure of 0.13 mbar was chosen so that vacuum was within a practical range for drying at both a laboratory, pilot and commercial scale (recommended range of chamber pressure is 0.07-0.20 mbar), also considering the solids content of the formulation.

Thus, the chamber pressure selected would ensure optimum sublimation and completion of primary drying within a reasonable period of time (on a lab scale, approximately 25 h). Shelf temperature during secondary drying was maintained at 25 °C under a chamber pressure of 0.13 mbar. For sucrose-based formulations with a high solids content. A shelf temperature of 25-45 °C during secondary drying should be appropriate for water desorption within in a reasonable time to achieve a low moisture content without an adverse impact on product quality (Pikal MJ, International Journal of Pharmaceutics, 1990, 60:203-217; Startzel P et al., Journal of

Pharmaceutical Sciences, 2015, 104:2345-2358; and Davis JM et al, Pharm Dev Technol. 2013, July- August, 18(4): 883-96.)· Ramp rates from primary to secondary drying were kept low (conservative) to avoid product temperature rise above shelf temperature (and possibly above Tg) during initial phases of secondary drying when moisture content in the product are high. The vacuum during the freeze-drying process was controlled by a capacitance (MKS) gauge. During the freeze-drying process, the Plexiglas door of the freeze-dryer was covered with a stainless steel shield to reduce heat radiation. In order to produce samples of different residual moistures and to characterize the drying behavior of the formulation in the course of the processes, single shelf closure was performed at the following sampling time points: 1) T-IPC1: at the end of primary drying; 2) T-IPC2: after the ramp to the secondary drying temperature (25 °C); 3) T- IPC3: after 5 h (DevRun#l) and 6 h (DevRun#2) of secondary drying at 25 °C; 4) T-final (=T0): at the end of secondary drying.

Analytical Charaterization of Lyophilized Formulation 25 :

[00273] Optical Appearance: In general, freeze-dried formulations in the course of drying (T-IPC1, T-IPC2, T-IPC3) showed excellent retention of the cake structure with off-white cakes.

[00274] Karl Fischer titration: The water content of the lyophilized cakes was determined using the coulometric Karl Fischer titrator Aqua 40.00 (Analytik Jena GmbH, Jena, Germany), which is equipped with a headspace module. For the measurement, about 15 mg sample were weighed into 2R glass vials in a glove box under humidity controlled conditions (rel. humidity < 5%) and heated to 120 °C in the oven connected to the reaction vessel via a tubing system. The evaporated water was transferred into the titration solution and the amount of water was determined. The measurement was performed until no more water evaporation was detectable. Water content of the sample was calculated considering environmental moisture as determined in three blanks. At the end of both cycles, all samples had a residual moisture of <1.0%.

[00275] Differential Scanning Calorimetry (DSC): Differential scanning calorimetry

(DSC) in a Mettler Toledo DSC1_943 (Mettler Toledo, Giessen, Germany) was used to determine thermal events of the frozen formulation, e.g. the glass transition temperature of the lyophilized products (Tg). For the physico-chemical analysis of the dried product 10 mg of the freeze-dried product were analyzed in crimped Al-crucibles (Mettler Toledo, Giessen, Germany). The samples were cooled to 0 °C with 10 K/min and reheated to 120 °C with a scanning rate of 10 K/min. This temperature profile was repeated in a second cycle. The midpoint of the endothermic shift of the baseline during the heating scan was taken as Tg. The glass transition temperature varied invesersely with residual moisture content.

[00276] X-ray powder diffraction (XRD): Wide angle X-ray powder diffraction (XRD) was used to study the morphology of lyophilized products. The X-ray diffractometer Empyrean (Panalytical, Almelo, The Netherlands) equipped with a copper anode (45 kV, 40 mA, Kal emission at a wavelength of 0.154 nm) and a PIXceBD detector was used. Approximately 100 mg of the freeze-dried samples were analyzed in reflection mode in the angular range from 5-45° 2Q, with a step size of 0.04° 2Q and a counting time of 100 seconds per step. The

morphology/crystallinity of the freeze-dried cakes of samples generated in the course of freeze drying (T-IPC1, T-IPC2, T-IPC3, TO) in both lyophilization process were determined by XRD. All samples exhibited a fully amorphous cake structure and no distinct peaks referring to crystallization of buffer salts or other excipients were detected. In all the samples, only one broad peak with a maximum at roughly 20 degrees 2Q was observed, which is characterisitic of amorphous samples (Liu W et al, AAPS Pharmscitech, 2005, Vol. 6(2)).

[00277] Reconstitution of the Lypophilized Formulations: The reconstitution volume was determined by weighing of four vials per shelf pre and post lyo for both cycles to determine the mass of the removed water. Samples of the lyophilized products (placebo and active vials) were reconstituted under a laminar flow hood according to the following procedure: the required amount of ultrapure water (Milli-Q water) was added to the lyophilized product (into the center of the vial) by using a pipette. The vial was carefully slewed (shaking was avoided). The reconstitution time was measured as the time to achieve a full reconstitution of the lyophilized product after addition of the liquid. The reconstitution behavior was judged, mainly with respect to foaming. The reconstitution time (dissolution of all visible solids) of samples generated in the course of freeze drying (T-IPC1, T-IPC2, T-IPC3, TO) in both lyophilization processes as determined after addition of ultra-pure water to the lyophilizates is shown in Table 7. After addition of 4.5 ml of ultra-pure water the reconstitution of the end products was completed in less than 4 minutes.

[00278] Table 7: Reconstitution times of samples generated at different time points during lyophilization

Table 7

[00279] pH: Formulation pH was measured with a calibrated pH meter (SevenEasy®, Mettler Toledo AG, Schwerzenbach, Switzerland) using a low ionic strength electrode (InLab® Pure Pro) or a high/normal ionic strength electrode (InLab® Micro). The pH-value of all samples generated in the course of freeze drying (T-IPC1, T-IPC2, T-IPC3, TO) in both lyophilization processes after reconstitution is shown in Table 8. No change in pH was observed before and after lyophilization. After preparation (T-liquid), the target pH-value of 6.0 was achieved in both cycles and the pH-values remained within the target range of 6.0 ± 0.1 in the course and after the two lyophilization processes.

[00280] Table 8: pH-values of samples generated at different time points during lyophilization.

Table 8

[00281] Osmolality: Osmolality of the samples was measured by method of freezing-point depression using a Knauer Automatic Semi-Micro Osmometer No. A0300 (Knauer, Berlin, Germany). The osmolality of all samples generated in the course of freeze drying (T-IPC1, T- IPC2, T-IPC3, TO) in both lyophilization processes after reconstitution is shown in Table 9. The osmolality after preparation (T-liquid) was within the physiological range (270 - 330 mOsmol/kg) and remained basically unchanged in the course and after lyophilization.

[00282] Table 9: Osmolality of samples generated at different time points during lyophilization.

Table 9

[00283] UV-Vis spectroscopy: A Tecan Safire 2 plate reader (Tecan Austria GmbH, Grodig, Austria) was used for concentration determination and turbidity assessment. Triplicates of 200 pl of the samples were prepared in 96 well plates (Corning Incorporation, NY, USA). After the measurement, the obtained absorption values were corrected for the pathlength and subtracted with corresponding blank. All samples were diluted to 0.5 1 mg/ml in placebo buffer prior to the measurement. An extinction coefficient based on a concentration of 1 mg/ml at a pathlength of 1 cm of 1.404 ml mg 1 cm 1 at 280 nm was used for concentration calculation (information from Rentschler). Furthermore, an increase in optical density at 350 nm from light scattering of particles was used to evaluate the turbidity of the samples. To calculate the aggregation index (A.I.) the following equation was used: A.I. = 100 A A350 / (A280 - A350)). The protein concentration of all samples (n=3) generated in the course of freeze drying (T-IPC1, T-IPC2, T-IPC3, TO) in both lyophilization processes after reconstitution is shown in Table 10. After preparation (T-liquid), the target concentration of 50 ± 2.5 mg/ml was achieved for all samples and remained within the specification in the course and after lyophilization. [00284] Table 10: Protein concentration of samples generated at different time points during lyophilization.

Table 10

[00285] High Performance Size Exclusion Chromatography (HP-SEC): HP-SEC was performed according to the information obtained from Rentschler. Of the Bio-Rad gel filtration standard (GFS) and the antibody samples, an amount of 20 LI g was loaded on the column (concentration of 1 mg/ml was used). As shown in Table 11, no change in low molecular weight solids was observed and with lyophilization, and change in high molecular weight solids was slight or none. Thus, lyophilization retains the vast majority of antibody in monomer form.

[00286] Table 11: Relative content of different species in samples generated at different time points during lyophilization.

Table 11

[00287] Various changes in form and details can be made therein without departing from the spirit and scope of the invention. Unless otherwise apparent from the context, any embodiment, aspect, element, feature, step or the like can be used in combination with any other. Insofar as information associated with a citation may change with time, the information associated with the citation at the earliest effective filing date is meant, the earliest effective filing date for a citation meaning the filing date of the present application or earlier priority application disclosing the citation. All references, issued patents and patent applications cited within the body of the instant specification are hereby incorporated by reference in their entirety, for all purposes. Any embodiment, aspect, feature, element, step or the like can be combined with any other unless the context indicates otherwise. When a composition is said to comprise certain specified components, the application should be read unless the context requires otherwise as disclosing that in the alternative, the composition may consist of or consist essentially of the specified components. For example, when an antibody chain is said to have an amino acid sequence comprising a specified SEQ ID NO., it should be understood unless the context requires otherwise that alternatively the antibody chain can consist of or consist essentially of the SEQ ID NO. “Consisting essentially of’ is used in accordance with convention to designate the basic and novel components of a composition. Unless otherwise apparent from the context, water can also be present in any amount. Other components may be present in minor amounts having no significant effect on the activity or stability of the composition.

“Essentially free of’ is likewise defined to indicate a component may be present, if at all, only in such minor amounts. When a component of a composition is said to be present at a

concentration of“about” a designated value or range of values, the application should be read as disclosing in the alternative that the component can be present at the designated value or range of values.