Treatment of xerosis with nuclease fusion proteins

文档序号:197097 发布日期:2021-11-02 浏览:27次 中文

阅读说明:本技术 用核酸酶融合蛋白治疗干燥症 (Treatment of xerosis with nuclease fusion proteins ) 是由 J·A·波萨达 D·伯格 于 2020-01-03 设计创作,主要内容包括:本公开提供了通过施用核酸酶融合蛋白来治疗干燥症(Sjogren’s disease)的方法。本公开的方法可用于治疗与干燥症(Sjogren’s disease)相关的症状,包括疲劳。(The present disclosure provides methods of treating Sjogren's disease by administering nuclease fusion proteins. The methods of the present disclosure can be used to treat symptoms associated with Sjogren's disease, including fatigue.)

1. A method of treating xerosis by reducing fatigue in a human patient in need thereof, said method comprising administering to said patient an effective amount of an RNase-Fc fusion protein, thereby treating xerosis by reducing fatigue in said patient.

2. The method of claim 1, wherein the RNase-Fc fusion protein comprises human pancreatic RNase 1.

3. The method of claim 2, wherein the human pancreatic RNase 1 comprises the amino acid sequence shown in SEQ ID NO 2.

4. The method of claim 1, wherein the RNase-Fc fusion protein comprises a wild-type human IgG1 Fc domain or a human IgG1 Fc domain containing one or more mutations.

5. The method of claim 4, wherein the Fc domain comprising one or more mutations binds weakly to Fcyreceptors on human cells.

6. The method of claim 1, wherein the RNase-Fc fusion protein has reduced effector function, optionally selected from opsonization, phagocytosis, complement-dependent cytotoxicity, and antibody-dependent cytotoxicity.

7. The method of claim 4, wherein the human IgG1 Fc domain comprises the P238S mutation and the P331S mutation according to EU numbering.

8. The method of claim 4, wherein the human IgG1 Fc domain comprises a hinge domain, a CH2 domain, and a CH3 domain.

9. The method of claim 4, wherein the human IgG1 Fc domain comprises a substitution of one or more of the three hinge region cysteine residues with serine.

10. The method of claim 9, wherein the Fc domain comprises SCC mutations (residues 220, 226, and 229) according to EU index numbering.

11. The method of claim 1, wherein the human IgGlFc domain comprises the amino acid sequence set forth in SEQ ID No. 22.

12. The method of claim 1, wherein the RNase-Fc fusion protein has the amino acid sequence shown in SEQ ID NO 50.

13. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5-10 mg/kg.

14. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 10 mg/kg.

15. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5 mg/kg.

16. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient by intravenous injection.

17. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5-10mg/kg every two weeks.

18. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5-10mg/kg every two weeks for three months.

19. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient as six infusions once every two weeks over three months.

20. The method of claim 1, wherein the RNase-Fc fusion protein is administered to the patient weekly for three weeks and then every two weeks to achieve or maintain a therapeutic effect.

21. The method of claim 1, wherein treatment reduces fatigue in the patient by at least 1 point in the ESSPRI score relative to the EULAR SS patient reporting index (ESSPRI) score prior to treatment.

22. The method of claim 1, wherein treatment reduces the ESSPRI score by at least 1 point relative to the ESSPRI score prior to treatment.

23. The method of claim 21, wherein fatigue is reduced to a score between 4.5 and 5.5 on an ESSPRI scale of 1 to 10.

24. The method of claim 21, wherein an effective dose of the RNase-Fc is administered to the patient every two weeks.

25. The method of claim 1, wherein treatment improves fatigue of the patient by at least 1 point on the FACIT fatigue scale relative to a chronic disease treatment Functional Assessment (FACIT) score prior to treatment.

26. The method of claim 25, wherein treatment improves fatigue in the patient by at least 2 points on the FACIT fatigue Scale.

27. The method of claim 1, wherein the treatment increases the FACIT fatigue score by at least 1 point relative to a FACIT fatigue score prior to the treatment.

28. The method of claim 27, wherein the treatment increases the FACIT fatigue score by at least 2 points relative to a FACIT fatigue score prior to the treatment.

29. The method of claim 1, wherein treating reduces fatigue in the patient by at least 1 point in the fatigue profile before treatment (ProF) score relative to the ProF score.

30. The method of claim 1, wherein treating reduces fatigue in the patient by at least 1 point in the score for the fatigue profile before treatment (ProF) mental item relative to the score for the ProF mental item.

31. The method of claim 1, wherein treating reduces fatigue of the patient by at least 1 point in the score for a pre-treatment fatigue profile (ProF) physical program relative to the score for a ProF physical program.

32. The method of claim 1, wherein the treatment improves cognitive function of the patient as measured by the DSST test relative to a pre-treatment digital symbol replacement test (DSST) test score.

33. The method of claim 1, wherein treating increases the number of matches the patient completes in 90 seconds in a digital sign replacement test (DSST) test.

34. The method of claim 1, wherein treating reduces the time to complete a DSST test in the patient.

35. A method of treating xerosis by reducing fatigue in a human patient in need thereof, said method comprising administering to said patient a RNase-Fc fusion protein at a dose of about 5-10mg/kg by intravenous injection, thereby treating xerosis by reducing fatigue in said patient.

36. A method of treating xerosis by improving cognitive outcome in a human patient in need thereof, said method comprising administering to said patient an effective amount of an RNase-Fc fusion protein, thereby treating xerosis by improving cognitive outcome in said patient.

37. The method of claim 36, wherein the patient's cognitive effect is improved by at least 1 point in the mental program of ProF relative to the mental program of ProF prior to treatment.

38. A method of treating xerosis by reducing fatigue in a human patient in need thereof, said method comprising administering to said patient an effective amount of an RNase-Fc fusion protein having the amino acid sequence shown in SEQ ID NO:50, thereby treating xerosis by reducing fatigue in said patient.

39. A method of treating xerosis by reducing fatigue in a human patient in need thereof, said method comprising administering to said patient an effective amount of a pharmaceutical composition, wherein said composition comprises an RNase-Fc fusion protein having the amino acid sequence shown in SEQ ID NO: 50; and one or more pharmaceutically acceptable carriers and/or diluents, thereby treating xerosis by reducing fatigue in said patient.

40. The method of claim 35 or 36, wherein the RNase-Fc fusion protein comprises human pancreatic RNase 1.

41. The method of claim 40, wherein said human pancreatic RNase 1 comprises the amino acid sequence shown in SEQ ID NO 2.

42. The method of any one of claims 35-41, wherein the RNase-Fc fusion protein comprises a wild-type human IgG1 Fc domain or a human IgG1 Fc domain containing one or more mutations.

43. The method of claim 42, wherein the Fc domain comprising one or more mutations binds weakly to Fcyreceptors on human cells.

44. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein has reduced effector function, optionally selected from opsonization, phagocytosis, complement-dependent cytotoxicity and antibody-dependent cytotoxicity.

45. The method of claim 42, wherein the human IgG1 Fc domain comprises the P238S mutation and the P331S mutation according to EU numbering.

46. The method of claim 42, wherein the human IgG1 Fc domain comprises a hinge domain, a CH2 domain, and a CH3 domain.

47. The method of claim 42, wherein the human IgG1 Fc domain comprises a substitution of one or more of the three hinge region cysteine residues with serine.

48. The method of claim 47, wherein said Fc domain comprises SCC mutations (residues 220, 226, and 229), numbered according to the EU index.

49. The method of claim 35 or 36, wherein the human IgGlFc domain comprises the amino acid sequence set forth in SEQ ID No. 22.

50. The method of claim 35 or 36, wherein the RNase-Fc fusion protein has the amino acid sequence shown in SEQ ID No. 50.

51. The method of any one of claims 36-39, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5-10 mg/kg.

52. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 10 mg/kg.

53. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5 mg/kg.

54. The method of any one of claims 36-39, wherein the RNase-Fc fusion protein is administered to the patient by intravenous injection.

55. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5-10mg/kg every two weeks.

56. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein is administered to the patient at a dose of about 5-10mg/kg every two weeks for three months.

57. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein is administered to the patient in six infusions once every two weeks over three months.

58. The method of any one of claims 35-39, wherein the RNase-Fc fusion protein is administered to the patient weekly for three weeks and then every two weeks.

59. The method of any one of claims 35-39, wherein treatment reduces fatigue in the patient by at least 1 point in an EULAR SS patient reporting index (ESSPRI) score relative to the ESSPRI score prior to treatment.

60. The method of any one of claims 35-39, wherein treatment reduces the ESSPRI score by at least 1 point relative to the ESSPRI score prior to treatment.

61. The method of claim 59, wherein fatigue is reduced to a score between 4.5 and 5.5 on an ESSPRI scale of 1 to 10.

62. The method of claim 59, wherein an effective dose of the RNase-Fc is administered every two weeks to the patient.

63. The method of any one of claims 35-39, wherein treatment improves fatigue in the patient by at least 1 point on the FACIT fatigue scale relative to a chronic disease treatment Functional Assessment (FACIT) score prior to treatment.

64. The method of claim 63, wherein treatment improves fatigue in the patient by at least 2 points on the FACIT fatigue Scale.

65. The method of any of claims 35-39, wherein treatment increases the FACIT fatigue score by at least 1 point relative to the FACIT fatigue score prior to treatment.

66. The method of claim 65, wherein treatment increases the FACIT fatigue score by at least 2 points relative to a FACIT fatigue score prior to treatment.

67. The method of any one of claims 35-39, wherein treatment reduces fatigue in the patient by at least 1 point in the fatigue Profile before treatment (ProF) score relative to the ProF score.

68. The method of any one of claims 35-39, wherein treatment reduces fatigue in the patient by at least 1 point in the score for the fatigue profile before treatment (ProF) mental item relative to the score for the PROF mental item.

69. The method of any one of claims 35-39, wherein the treatment reduces fatigue in the patient by at least 1 point in the score for the fatigue profile before treatment (ProF) physical program relative to the score for the ProF physical program.

70. The method of any one of claims 35-39, wherein the treatment improves cognitive function of the patient as measured by the DSST test relative to a pre-treatment Digital Symbol Substitution Test (DSST) test score.

71. The method of any one of claims 35-39, wherein the treatment increases the number of matches a patient completes in a digital sign replacement test (DSST) test within 90 seconds.

72. The method of any one of claims 35-39, wherein treatment reduces the time to completion of a DSST test in a patient.

73. A kit comprising a container containing an injectable solution and instructions for treating xerosis by reducing fatigue in a human patient in need thereof, comprising:

An effective amount of RNase-Fc fusion protein shown as SEQ ID NO. 50; and

one or more pharmaceutically acceptable carriers and/or diluents;

wherein the injectable solution is prepared for intravenous administration.

74. An RNase-Fc fusion protein for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein.

Use of an RNase-Fc fusion protein in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof.

76. An RNase-Fc fusion protein for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient by intravenous injection a dose of about 5-10mg/kg of RNase-Fc fusion protein.

77. use of an RNase-Fc fusion protein in the manufacture of a medicament for the treatment of xerosis by reducing fatigue in a human patient in need thereof, said use comprising administering to said patient by intravenous injection a dose of about 5-10mg/kg of the RNase-Fc fusion protein.

78. An RNase-Fc fusion protein for use in a method of treating xerosis by improving cognitive outcome in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein.

79.RNase-Fc fusion protein for the manufacture of a medicament for treating xerosis by improving cognitive effects in a human patient in need thereof, said use comprising administering to said patient an effective amount of the RNase-Fc fusion protein.

80. An RNase-Fc fusion protein for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein having an amino acid sequence set forth as SEQ ID NO: 50.

Use of an RNase-Fc fusion protein in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof, the use comprising administering to said patient an effective amount of an RNase-Fc fusion protein having an amino acid sequence as set forth in SEQ ID NO: 50.

82. An RNase-Fc fusion protein for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of a pharmaceutical composition, wherein the composition comprises an RNase-Fc fusion protein having an amino acid sequence as set forth in SEQ ID NO: 50; and one or more pharmaceutically acceptable carriers and/or diluents.

83. An RNase-Fc fusion protein for use in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof, said use comprising administering to said patient an effective amount of a pharmaceutical composition, wherein said composition comprises an RNase-Fc fusion protein having an amino acid sequence as set forth in SEQ ID NO: 50; and one or more pharmaceutically acceptable carriers and/or diluents.

84. A method of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in a reduction of one or more inflammation-associated genes.

85. The method of claim 84, wherein said one or more inflammation-associated genes are selected from the group consisting of IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and STAT 5B.

86. The method of claim 84, wherein said one or more inflammation-associated genes is selected from IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and STAT 5B.

87. A method of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in an increase in one or more inflammation-associated genes.

88. The method of claim 86, wherein said one or more inflammation-associated genes are selected from CXCL10(IP-10), CD163, RIPK2 and CCR 2.

89. A method of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in an increase in one or more cytokines and an improvement in fatigue.

90. The method of claim 89, wherein said cytokine is CXCL 10.

91. A method of identifying a patient with xerosis as a candidate for treatment with an RNA nuclease agent, the method comprising:

(a) determining an inflammation-associated gene expression profile in a sample obtained from the patient; and

(b) comparing the inflammation-associated gene expression profile determined in step (a) with an inflammation-associated gene expression profile in a sample obtained from a suitable control subject,

wherein the inflammation-associated gene expression profile indicates that the patient is a candidate for treatment with an RNA nuclease agent.

92. The method of claim 91, wherein the inflammation-associated gene is selected from the group consisting of MAP3K8, ACKR3, STAT1, STAT2, TRIM37, and ZNF 606.

93. use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in the reduction of one or more inflammation-associated genes.

94. The use of claim 93, wherein said one or more inflammation-associated genes are selected from the group consisting of IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and STAT 5B.

95. The use of claim 93, wherein said one or more inflammation-associated genes is selected from IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and STAT 5B.

96. An RNA nuclease agent for use in a method of treatment of xerosis, the use comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in the reduction of one or more inflammation-associated genes.

97. The RNA nuclease agent of claim 96, wherein the one or more inflammation-associated genes are selected from IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and STAT 5B.

98. The RNA nuclease agent of claim 96, wherein the one or more inflammation-associated genes are selected from IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and STAT 5B.

99. use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in an increase in one or more inflammation-associated genes.

100. The use of claim 99, wherein said one or more inflammation-associated genes are selected from CXCL10(IP-10), CD163, RIPK2, and CCR 2.

101. An RNA nuclease agent for use in a method of treatment of xerosis, the use comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in an increase in one or more inflammation-associated genes.

102. The RNA nuclease agent of claim 101, wherein the one or more inflammation-associated genes are selected from CXCL10(IP-10), CD163, RIPK2, and CCR 2.

Use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in an increase in one or more cytokines and an improvement in fatigue.

104. The use of claim 103, wherein said cytokine is CXCL 10.

105. An RNA nuclease agent for use in a method of treatment of xerosis, the use comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in an increase in one or more cytokines and an improvement in fatigue.

106. The RNA nuclease agent of claim 105, wherein the cytokine is CXCL 10.

Background

Primary Sjogren's syndrome (pSS) is an autoimmune disease estimated to affect 0.5% to 1% of the general population, of which nine-tenth patients are females (Ramos-Casals,2005, Ann Rheum Dis.64, 347-354; Skopouli,2000, Semin. arthritis Rheum.29: 296-304). Most women with pSS are characterized by mild to moderate disease, manifested by fatigue, joint pain, and dry eyes and/or mouth. The disease is characterized by lymphocytic infiltration of the salivary and lacrimal glands with subsequent inflammation, gland injury and loss of function, resulting in dry eyes and dry mouth. Involvement of the major organ systems of the lung, kidney and liver is a common systemic manifestation of pSS (Malladi,2012, Arthritis Care Res.64: 911-. At the biochemical level, pSS is associated with elevated immunoglobulin levels and the production of antinuclear antibodies to ribonucleoprotein complexes such as SSA/Ro and SSB/La (Bave,2005, Arth. & Rheum.52: 1185-.

Once formed, immune complexes containing RNA are readily internalized into cells of the immune system (e.g., dendritic cells), where the RNA bound to the immune complex is capable of interacting with a Toll-like receptor (TLR), such as the RNA sensor TLR 7. Although TLRs are thought to be key elements of the innate immune system by recognizing pathogen-associated molecular components, it is now clear that host nucleic acids can also activate specific family members, including TLR7, TLR8 and TLR9(Theofilopoulos,2010, nat. rev. rheum.,6: 146-. Cells expressing these receptors do not distribute them to the cell surface; in contrast, TLR7/8/9 was sequestered in endosomes (Theofiloulos, 2010, nat. Rev. Rheum.,6: 146-156). This positioning is assumed to minimize interactions with host nucleic acids. However, when present in an immune complex, the nucleic acid antigen will be actively internalized into the cell by receptor-mediated endocytosis. Effector Fc, complement and B cell receptors all probably promote entry of IC-containing nucleic acids into endosomes (Means,2005, J.Clin.invest.115: 407-1177; Lau,2005, J.Exp.Med.202: 1171-1177; Brkic 2013, Ann.Rheum.Dis.72(5): 728-735). Once internalized, the nucleic acid binds to and activates a resident endosomal TLR. In turn, activated TLRs promote pDC production of type 1 IFN, activation of PMNs, and promotion of B cell proliferation and production of autoantibodies. Thus, nucleic acid-containing antigens contribute to multiple aspects of the pathophysiology of pSS disease.

Fatigue is one of the most common extra-glandular symptoms of sjogren's syndrome, defined as persistent general fatigue. It is estimated that 70% of pSS patients suffer from deep fatigue, which is reported to negatively impact the quality of life. Serologically, about 80% of these patients have anti-Ro/SSA autoantibodies that bind to autoantigens containing small non-coding RNAs. Fatigue can be characterized by strength, duration, and impact on daily function. Notably, fatigue is closely associated with depression in primary care facilities. (Segal et al, Arthritis Rhem. 2008Decumber 15; 59(12): 1780-1778). Therefore, there is a need for a method of improving fatigue in patients with autoimmune diseases (e.g., sjogren's syndrome).

Disclosure of Invention

The present disclosure relates, at least in part, to RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, which are useful for treating sjogren's syndrome in a human patient in need thereof. In some aspects, the disclosure relates to RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, that are useful for treating, reducing, or ameliorating fatigue in sjogren's syndrome patients. In some aspects, the disclosure relates to RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, that are useful for improving, enhancing or increasing cognitive ability, or reducing, reducing or improving cognitive deficits in patients with sjogren's syndrome. In some aspects, the disclosure relates to RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, that are useful for reducing, or ameliorating depression, including fatigue-related depression, in sjogren's syndrome patients. In some aspects, the disclosure relates to compositions comprising an RNase-Fc fusion protein and one or more pharmaceutically acceptable carriers and/or diluents, which are useful in methods of treating or preventing sjogren's syndrome, methods of treating, preventing or reducing fatigue in patients with sjogren's syndrome, and methods of improving cognitive ability in patients with sjogren's syndrome. In some embodiments, the RNase-Fc fusion protein is administered to a human patient by injection (e.g., intravenously) at a dose of about 5-10mg/kg, about 2-8mg/kg, about 3-6mg/kg, about 3mg/kg, about 5mg/kg or about 10 mg/kg.

In some embodiments, the RNase-Fc fusion protein is RSLV-132. RSLV-132 is a nuclease fusion protein comprising a homodimer of two polypeptides, each having the amino acid sequence shown in SEQ ID NO: 50. Each polypeptide of the homodimer has the configuration shown in FIG. 1 from N-terminus to C-terminus of RNase-Fc, in which the wild-type human RNase1 domain (SEQ ID NO:2) is not operably coupled via a linker to the N-terminus of a human IgG Fc domain comprising a mutation of one of the three hinge region cysteine residues to serine (residue 220 or C220S, also referred to herein as the "SCC hinge") and two mutations in the CH2 domain, P238S and P331S. The sequence of the human IgGl Fc domain with these mutations is shown in SEQ ID NO 22.

In some aspects, the disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, the method comprising administering to the patient an effective amount of a nuclease fusion protein comprising an RNase, such as an RNase-Fc fusion protein (e.g., RSLV-132), thereby treating xerosis by reducing fatigue in the patient.

In some aspects, the present disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, the method comprising administering about 5-10mg/kg of an RNase-containing nuclease fusion protein, such as an RNase-Fc fusion protein (e.g., RSLV-132), to the patient by intravenous injection, thereby treating xerosis by reducing fatigue in the patient.

In some aspects, the disclosure provides a method of treating xerosis by improving cognitive outcome in a human patient in need thereof, the method comprising administering to the patient an effective amount of a nuclease fusion protein comprising an RNase, such as an RNase-Fc fusion protein (e.g., RSLV-132), thereby treating xerosis by improving cognitive outcome in the patient. In some aspects, the patient's cognitive effect is improved by at least 1 point in the mental program of the pre-treatment fatigue profile (ProF) mental program relative to the mental program of the ProF. In some aspects, the patient's cognitive effect is improved by greater than 1 point, greater than 2 points, or greater than 3 points in the pro-f mental program relative to the pre-treatment pro-f mental program.

In some aspects, the present disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, the method comprising administering to the patient an effective amount of an RNase-Fc fusion protein having an amino acid sequence set forth as SEQ ID NO:50, thereby treating xerosis by reducing fatigue in the patient. In some aspects, an effective amount of an RNase-Fc fusion protein having the amino acid sequence shown as SEQ ID NO. 50 is a dose of about 5mg/kg to about 10 mg/kg. In some aspects, the RNase-Fc fusion protein is administered in a composition with a pharmaceutically acceptable carrier. In some aspects, compositions comprising an RNase-Fc fusion protein having an amino acid sequence shown in SEQ ID NO:50 are prepared for intravenous injection (e.g., in solution).

In some aspects, the present disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, comprising administering to the patient an effective amount of a pharmaceutical composition, wherein the composition comprises: RNase-Fc fusion protein having an amino acid sequence shown as SEQ ID NO. 50; and one or more pharmaceutically acceptable carriers and/or diluents, thereby treating xerosis by reducing fatigue in the patient. In some aspects, compositions comprising an RNase-Fc fusion protein comprising an amino acid sequence as set forth in SEQ ID NO:50 are prepared for intravenous injection (e.g., in solution).

In any preceding or related embodiment of the methods of the present disclosure, the RNase-Fc fusion protein for use herein comprises human pancreatic RNase 1. In some aspects, human pancreatic RNase 1 comprises the amino acid sequence shown in SEQ ID NO 2. In some aspects, the RNase-Fc fusion proteins of the present disclosure comprise a wild-type human IgG1 Fc domain or a human IgG1 Fc domain containing one or more mutations. In some aspects, the human IgGl Fc domain comprises one or more mutations that reduce Fc γ receptor binding on human cells. In some aspects, the RNase-Fc fusion proteins of the present disclosure have reduced effector function, optionally selected from opsonization, phagocytosis, complement-dependent cytotoxicity, and antibody-dependent cytotoxicity.

In some aspects, the human IgG1 Fc domain comprises a hinge domain, a CH2 domain, and a CH3 domain. In some aspects, the human IgG1 Fc domain comprises a substitution of one or more of the three hinge region cysteine residues with serine. In some aspects, the Fc domain comprises SCC mutations (residues 220, 226, and 229) according to EU index numbering. In some aspects, the human IgGl Fc domain comprises the amino acid sequence set forth as SEQ ID NO: 22.

In any preceding or related embodiment of the methods of the present disclosure, the RNase-Fc fusion protein for use herein comprises a human pancreatic RNase 1 coupled, with or without a linker, to a human IgGl Fc domain, said human pancreatic RNase 1 comprising a C220S mutation, a P238S mutation, and a P331S mutation according to EU numbering.

In any of the preceding or related embodiments of the methods of the present disclosure, the RNase-Fc fusion protein for use herein has the amino acid sequence shown as SEQ ID NO: 50.

In any of the preceding or related embodiments of the methods of the present disclosure, the RNase-Fc fusion protein for use herein, e.g., RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to the patient by intravenous injection. In some aspects, an effective dose of an RNase-Fc fusion protein, such as RSLV-132, or a pharmaceutical composition comprising an RNase-Fc fusion protein is administered to the patient every two weeks.

In some aspects, an RNase-Fc fusion protein of the present disclosure, such as RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to a patient at a dose of about 5-10 mg/kg. In some aspects, an RNase-Fc fusion protein, such as RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to a patient at a dose of about 10 mg/kg. In some aspects, an RNase-Fc fusion protein, such as RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to a patient at a dose of about 5 mg/kg. In some aspects, an RNase-Fc fusion protein of the present disclosure, such as RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to a patient at a dose of about 5-10mg/kg every two weeks. In some aspects, an RNase-Fc fusion protein, such as RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to a patient at a dose of about 5-10mg/kg every two weeks for three months. In some aspects, the RNase-Fc fusion protein, e.g., RSLV-132, or a pharmaceutical composition comprising the RNase-Fc fusion protein, is administered to the patient in six infusions once every two weeks over a three month period. In some aspects, an RNase-Fc fusion protein, e.g., RSLV-132, or a pharmaceutical composition comprising an RNase-Fc fusion protein, is administered to a patient weekly for three weeks, and then once every two weeks to achieve or maintain a therapeutic effect.

In some aspects, the disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, the method comprising administering at least three doses of an RNase-Fc fusion protein (e.g., RSLV-132) or a pharmaceutical composition comprising an RNase-Fc fusion protein, wherein each dose is administered to the patient (e.g., by injection, e.g., intravenous injection) at a dose of about 5-10mg/kg, about 2-8mg/kg, about 3-6mg/kg, about 3mg/kg, about 5mg/kg, or about 10 mg/kg. In some aspects, at least four doses of the RNase-Fc fusion protein are administered to the patient. In some aspects, at least five doses of the RNase-Fc fusion protein are administered to the patient. In some aspects, at least six doses of the RNase-Fc fusion protein are administered to the patient. In some aspects, at least seven doses of the RNase-Fc fusion protein are administered to the patient. In some aspects, at least eight doses of the RNase-Fc fusion protein are administered to the patient.

In some aspects, the disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, the method comprising administering once per week a dosing regimen of a dose of an RNase-Fc fusion protein (e.g., RSLV-132) or a pharmaceutical composition comprising an RNase-Fc fusion protein for at least two weeks, wherein each dose is administered to the patient (e.g., by injection, e.g., intravenous injection) at a dose of about 5-10mg/kg, about 2-8mg/kg, about 3-6mg/kg, about 3mg/kg, about 5mg/kg, or about 10 mg/kg. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for at least 3 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for at least 4 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for at least 5 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for at least 6 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for at least 7 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for at least 8 weeks.

In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient every two weeks for at least 2 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient every two weeks for at least 4 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient every two weeks for at least 6 weeks. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient every two weeks for at least 8 weeks.

In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for three weeks, and then every two weeks for at least 1 month. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for three weeks, and then every two weeks for at least 2 months. In some aspects, one dose of the RNase-Fc fusion protein is administered to the patient weekly for three weeks, and then every two weeks for at least 3 months.

In some aspects, the disclosure provides a method of treating sjogren's disease by reducing fatigue in a human patient in need thereof, the method comprising administering an effective amount of an RNase-Fc fusion protein (e.g., RSLV-132) or a pharmaceutical composition comprising the RNase-Fc fusion protein, wherein treatment reduces fatigue in the patient by at least 1 point in an ESSPRI score relative to a pre-treatment EULAR SS patient reporting index (ESSPRI) score. In some aspects, the treatment reduces the ESSPRI score of the patient by at least 1 point relative to the ESSPRI score prior to treatment. In some aspects, fatigue of the patient is reduced to a score between 4.5 and 5.5 on an esppri scale of 1 to 10.

In some aspects, the disclosure provides a method of treating sjogren's disease by reducing fatigue in a human patient in need thereof, the method comprising administering an effective amount of an RNase-Fc fusion protein (e.g., RSLV-132) or a pharmaceutical composition comprising the RNase-Fc fusion protein, wherein treatment improves fatigue in the patient by at least 1 point in a chronic disease treatment Functional Assessment (FACIT) scale prior to treatment. In some aspects, the treatment improves fatigue in the patient by at least 2 points on the FACIT fatigue scale. In some aspects, the treatment increases the FACIT fatigue score by at least 1 point relative to the FACIT fatigue score prior to treatment. In some aspects, the treatment increases the FACIT fatigue score by at least 2 points relative to the FACIT fatigue score prior to treatment.

In some aspects, the disclosure provides a method of treating xerosis by reducing fatigue in a human patient in need thereof, the method comprising administering an effective amount of an RNase-Fc fusion protein (e.g., RSLV-132) or a pharmaceutical composition comprising the RNase-Fc fusion protein, wherein treatment reduces fatigue in the patient by at least 1 point in the pro f score relative to the pre-treatment fatigue profile (pro f) score. In some aspects, treatment reduces fatigue in the patient by at least 1 point in the score for the pro-f mental item relative to the score for the pre-treatment fatigue profile (pro f) mental item. In some aspects, the treatment reduces fatigue of the patient by at least 1 point in the score for the fatigue profile before treatment (ProF) physical program relative to the score for the ProF physical program.

In some aspects, the present disclosure provides a method of treating xerosis by improving cognitive function in a human patient in need thereof, the method comprising administering an effective amount of an RNase-Fc fusion protein (e.g., RSLV-132) or a pharmaceutical composition comprising the RNase-Fc fusion protein, wherein the treatment improves cognitive function in the patient as measured by a DSST test relative to a pre-treatment Digital Symbol Substitution Test (DSST) test score. In some aspects, the treatment increases the number of matches a patient completes within 90 seconds in a digital sign replacement test (DSST) test. In some aspects, the treatment reduces the time for a patient to complete a DSST test.

In other aspects, the present disclosure provides kits comprising a container comprising an injectable solution comprising an effective amount of an RNase-Fc fusion protein (e.g., RSLV-132) of the present disclosure or an RNase-Fc fusion protein having an amino acid sequence set forth in SEQ ID NO:50 or a pharmaceutical composition comprising an RNase-Fc fusion protein; and one or more pharmaceutically acceptable carriers and/or diluents; and instructions for treating xerosis by reducing fatigue in a human patient in need thereof, wherein the injectable solution is prepared for intravenous administration.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient by intravenous injection a dose of about 5-10mg/kg of RNase-Fc fusion protein.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof, the use comprising administering to the patient by intravenous injection a dose of about 5-10mg/kg of the RNase-Fc fusion protein.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in a method of treating xerosis by improving cognitive effects in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for treating xerosis by improving cognitive effects in a human patient in need thereof, the use comprising administering to the patient an effective amount of the RNase-Fc fusion protein.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein having an amino acid sequence set forth in SEQ ID NO: 50.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of an RNase-Fc fusion protein having an amino acid sequence set forth as SEQ ID NO: 50.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in a method of treating xerosis by reducing fatigue in a human patient in need thereof, the treatment comprising administering to the patient an effective amount of the pharmaceutical composition, wherein the composition comprises: RNase-Fc fusion protein, which has an amino acid sequence shown as SEQ ID NO. 50; and one or more pharmaceutically acceptable carriers and/or diluents.

In some aspects, the present disclosure provides an RNase-Fc fusion protein (e.g., RSLV-132), or a pharmaceutical composition comprising the same, for use in the manufacture of a medicament for treating xerosis by reducing fatigue in a human patient in need thereof, the use comprising administering to the patient an effective amount of a pharmaceutical composition, wherein the composition comprises: RNase-Fc fusion protein, which has an amino acid sequence shown as SEQ ID NO. 50; and one or more pharmaceutically acceptable carriers and/or diluents.

In some aspects, the present disclosure provides methods of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from the group consisting of IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and STAT 5B.

In some aspects, the present disclosure provides methods of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein the treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from the group comprising IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and STAT 5B.

In some aspects, the present disclosure provides a use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to a patient an effective amount of the RNA nuclease agent, wherein the treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and STAT 5B.

In some aspects, the present disclosure provides a use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to a patient an effective amount of the RNA nuclease agent, wherein the treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and STAT 5B.

In some aspects, the present disclosure provides an RNA nuclease agent for use in a method of treating xerosis, the method for comprising administering to a patient an effective amount of the RNA nuclease agent, wherein treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and STAT 5B.

In some aspects, the present disclosure provides an RNA nuclease agent for use in a method of treating xerosis, the method for comprising administering to a patient an effective amount of the RNA nuclease agent, wherein treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and STAT 5B.

In some aspects, the disclosure provides a method of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein treatment results in an increase in one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from CXCL10(IP-10), CD163, RIPK2, and CCR 2.

In some aspects, the present disclosure provides a use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to a patient an effective amount of the RNA nuclease agent, wherein treatment results in an increase in one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from CXCL10(IP-10), CD163, RIPK2, and CCR 2.

In some aspects, the present disclosure provides an RNA nuclease agent for use in a method of treating xerosis, the method for comprising administering to a patient an effective amount of the RNA nuclease agent, wherein treatment results in an increase in one or more inflammation-associated genes. In some aspects, the one or more inflammation-associated genes are selected from CXCL10(IP-10), CD163, RIPK2, and CCR 2.

In some aspects, the disclosure provides methods of treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent, wherein treatment results in an increase in one or more cytokines and an improvement in fatigue. In some aspects, the cytokine is CXCL 10.

In some aspects, the present disclosure provides a use of an RNA nuclease agent in the manufacture of a medicament for treating xerosis, the use comprising administering to a patient an effective amount of the RNA nuclease agent, wherein treatment results in an increase in one or more cytokines and an improvement in fatigue. In some aspects, the cytokine is CXCL 10.

In some aspects, the disclosure provides an RNA nuclease agent for use in a method of treating xerosis, the use comprising administering to a patient an effective amount of the RNA nuclease agent, wherein treatment results in an increase in one or more cytokines and an improvement in fatigue. In some aspects, the cytokine is CXCL 10.

In some aspects, the disclosure provides methods of identifying a patient with xerosis as a candidate for treatment with an RNA nuclease agent, comprising: (a) determining an inflammation-associated gene expression profile in a sample obtained from the patient; and (b) comparing the inflammation-associated gene expression profile determined in step (a) with an inflammation-associated gene expression profile in a sample obtained from a suitable control subject, wherein the inflammation-associated gene expression profile indicates that the patient is a candidate for treatment with an RNA nuclease agent. In some aspects, the inflammation-associated gene is selected from the group consisting of MAP3K8, ACKR3, STAT1, STAT2, TRIM37, and ZNF 606.

Drawings

This patent or this application document contains at least one drawing executed in color. Copies of this patent or this patent application publication with color drawings will be provided by the office upon request and payment of the necessary fee.

These and other features, aspects, and advantages of the present invention will become better understood with regard to the following description and accompanying drawings where:

FIG. 1 depicts the configuration of RSLV-132, RSLV-132 being a homodimeric RNase-Fc fusion protein comprising two polypeptides. Each polypeptide of the homodimer has an RNase-Fc configuration, with the wild-type human RNase1 domain not operably coupled via a linker to the N-terminus of the human IgG1 Fc domain, which human IgG1 Fc domain comprises the SCC hinge and CH2 mutations P238S and P331S.

FIG. 2 graphically depicts the improvement in ESSPRI score for pSS patients treated with RSLV-132 compared to placebo-treated patients. ESSPRI scores were evaluated for pSS patients receiving RSLV-132 and placebo treatment on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. A reduction in the espri score of at least 1 point is clinically significant.

Figure 3 graphically depicts the improvement in ESSPRI score for patients treated with RSLV-132 compared to placebo-treated patients. The change in ESSPRI score from baseline over time is provided on the y-axis. ESSPRI scores were assessed for patients receiving RSLV-132 and placebo treatment on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. A reduction in the espri score of at least 1 point is clinically significant.

FIG. 4 illustratesMean change from baseline in fatigue terms for RSLV-132 and the espsri of the placebo group are plotted (p ═ 0.136). The fatigue program score is shown to be improved for the ESSPRI score for patients receiving RSLV-132 treatment compared to patients receiving placebo treatment. The fatigue item score for ESSPRI is provided on the y-axis as a function of time from baseline. ESSPRI scores were assessed for patients receiving RSLV-132 and placebo treatment on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. Results from each visit were analyzed using separate one-way analysis of variance (1-way ANOVA) models, each of which examined a null hypothesis (H) with zero true mean difference between treatment groups0). Unadjusted alpha is 0.05. All standard error bars used the standard error at day 99.

Figure 5A graphically depicts the improvement in fatigue program score for espsri in patients treated with RSLV-132 compared to patients treated with placebo. The fatigue item score for ESSPRI is provided on the y-axis. ESSPRI scores were assessed for patients receiving RSLV-132 and placebo treatment on study day 1 (baseline) and on study day 99.

Fig. 5B graphically depicts the pain item score for ESSPRI for patients treated with RSLV-132 compared to placebo-treated patients. The pain item score for ESSPRI is provided on the y-axis. ESSPRI scores were assessed for patients receiving RSLV-132 and placebo treatment on study day 1 (baseline) and on study day 99.

Figure 5C graphically depicts the dry item score of ESSPRI for patients treated with RSLV-132 compared to placebo-treated patients. The dry item score of ESSPRI is provided on the y-axis. ESSPRI scores were assessed for patients receiving RSLV-132 and placebo treatment on study day 1 (baseline) and on study day 99.

Figure 6 graphically depicts the mean change from baseline in FACIT for RSLV-132 and placebo (p 0.92). Showing an improvement in FACIT fatigue score in patients receiving RSLV-132 treatment compared to patients receiving placebo treatment. Patients receiving RSLV-132 and placebo treatment were evaluated for FACIT fatigue score on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. An increase in the FACIT fatigue score indicates an improvement in fatigue. The results of each visit were analyzed using separate one-way analysis of variance (1-way ANOVA) models, each testing a null hypothesis (H0) with zero true mean difference between treatment groups. Unadjusted alpha is 0.05. All standard error bars used the standard error at day 99.

Fig. 7 graphically depicts the improvement in ProF for patients treated with RSLV-132 compared to patients treated with placebo. ProF score changes from baseline over time are provided on the y-axis. The ProF score was assessed in patients receiving RSLV-132 and placebo treatment on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. A decrease in the ProF score indicates an improvement in fatigue.

Fig. 8 graphically depicts the mean change in the ProF mental fatigue program for the RSLV-132 and placebo groups (p ═ 0.046). Shows an improvement in the ProF mental program in patients treated with RSLV-132 compared to patients treated with placebo. The change in the scores of the ProF mental items from baseline over time is provided on the y-axis. Patients receiving RSLV-132 and placebo treatment were evaluated for their scores for the ProF mental program on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. A decrease in the ProF score indicates an improvement in fatigue. The results of each visit were analyzed using separate one-way analysis of variance (1-way ANOVA) models, each testing a null hypothesis (H0) with zero true mean difference between treatment groups. Unadjusted alpha is 0.05. All standard error bars used the standard error at day 99.

Fig. 9 graphically depicts the improvement of the ProF somatic program in patients treated with RSLV-132 compared to patients treated with placebo. The change in the scores of the ProF body items from baseline over time is provided on the y-axis. The scores of the ProF somatic items were assessed for patients receiving RSLV-132 and placebo treatment on study day 1 (baseline), days 29, 57, 85 and 99/end of treatment. A decrease in the ProF score indicates an improvement in fatigue.

Figures 10A and 10B provide the results of a numerical symbol replacement test (DSST) in patients treated with RSLV-132 and placebo. Patients were tested for DSST at study baseline (day 1) and day 99. As shown in fig. 10A, "Total of 90 seconds (Total 90 s)" means the Total number of symbols that match a number within 90 seconds. "Completion" refers to the time in seconds to complete the test. Statistically significant improvements in the time to completion of the DSST test were observed in patients treated with RSLV-132 from the initial baseline test (day 1) to the follow-up (day 99). Figure 10B graphically depicts an increase in completion time for the placebo group and a decrease in completion time for RSLV-132 treated patients.

Figure 11 depicts changes in gene expression at day 99 compared to day 1 (baseline) relative to RSLV-132 subjects with or without achieved clinical response. The genes shown in the heatmap are those highly correlated with FACIT tool results (R)2>0.6)。

Figures 12A-C depict gene expression patterns at baseline (prior to study drug administration) for subjects subsequently undergoing RSLV-132 treatment with MCII on day 99 compared to baseline (prior to study drug administration) for subjects not undergoing MCII. The genes with the highest relevance to a given tool are shown. FIG. 12A depicts the FACIT-associated genes (R) 2>0.6). FIG. 12B depicts ProF-associated genes (R)2>0.6). FIG. 12C depicts ESSPRI-associated genes (R)2>0.6)。

Detailed Description

The present disclosure provides RNase-containing nuclease fusion proteins comprising an RNase-Fc fusion protein that digests circulating RNA and RNA complexed with autoantibodies and immune complexes, thereby treating patients with primary sjogren's syndrome (pSS). The disclosure also provides methods of treating diseases characterized by elevated levels of circulating RNA and/or RNA-containing autoantibodies (such as sjogren's syndrome), and methods of treating disease symptoms characterized by elevated levels of circulating RNA and RNA-containing autoantibodies (such as fatigue associated with sjogren's syndrome in a human patient in need thereof). The present disclosure also provides effective treatments and dosing regimens for administering RNase-containing nuclease fusion proteins (including RNase-Fc fusion proteins) to human patients in need thereof, including patients with sjogren's syndrome.

The present disclosure is based, at least in part, on the surprising discovery that treatment of pSS patients by administering a nuclease fusion protein comprising an RNase (e.g., RSLV-132) reduces fatigue associated with pSS in the patients. Without being bound by theory, it is believed that the RNase-containing nuclease fusion proteins of the present disclosure are capable of digesting circulating RNA, as well as RNA complexed with autoantibodies and immune complexes in autoimmune patients, thereby alleviating symptoms of autoimmune diseases (e.g., fatigue associated with sjogren's syndrome).

It is also believed, without being bound by theory, that by treating sjogren's syndrome patients with RNase-containing nuclease fusion proteins (such as RSLV-132), circulating RNA (whether associated with autoantibodies or free in circulation) is reduced, thereby reducing TLR activation and activation of several downstream inflammatory pathways. Thus, by treating sjogren's syndrome patients, including pSS patients, with RNase-containing nuclease fusion proteins (e.g., RSLV-132), activation of the proinflammatory cascade can be reduced, reduced or inhibited, thereby reducing or diminishing the overall inflammatory profile of sjogren's syndrome patients, thereby reducing or diminishing symptoms associated with sjogren's syndrome, including fatigue, pain, dryness, depression and/or cognitive impairment.

Unexpectedly, it was found that treatment of pSS patients by administration of a nuclease fusion protein comprising an RNase (e.g., RSLV-132) resulted in an improvement in patient fatigue which consistently showed an improvement in post-treatment fatigue by three separate tests. In particular, when the EULAR SS patient reporting index (ESSPRI) was performed on pSS patients after treatment with RSLV-132, the patients' ESSPRI score showed a clinically significant drop of 1 point or more in the fatigue term of ESSPRI score. A decrease in the espsri score is associated with an improvement in fatigue. Chronic disease test Functional Assessment (FACIT) of these patients following treatment with RSLV-132 resulted in an increase in the FACIT fatigue score, which was associated with decreased fatigue. Also, treatment with RSLV-132 followed by fatigue profile (ProF) testing resulted in a decrease in ProF scores, which correlates with decreased fatigue.

Furthermore, it was surprisingly found that after treatment of pSS patients with RLSV-132, the patients exhibited improved cognitive ability as measured by the digital symbol replacement test (DSST). Accordingly, the present disclosure provides nuclease fusion proteins comprising the fusion protein RNase, including RNase-Fc, that are useful in methods of treating sjogren's syndrome, methods of reducing fatigue associated with sjogren's syndrome, and methods of improving cognition in patients with sjogren's syndrome. The present disclosure also provides compositions comprising an RNase-Fc fusion protein and one or more pharmaceutically acceptable carriers and/or diluents, which are useful in methods of treating sjogren's syndrome, methods of reducing fatigue associated with sjogren's syndrome, and methods of improving cognition in patients with sjogren's syndrome. In some embodiments, the RNase-Fc fusion protein is RSLV-132. In some embodiments, the RNase-Fc fusion protein is administered to a human patient at about 5-10mg/kg, about 2-8mg/kg, about 3-6mg/kg, about 3mg/kg, about 5mg/kg, or about 10 mg/kg.

In addition, it was found that patients who achieved a clinical response after treatment of patients with primary sjogren syndrome (pSS) with an RNA nuclease agent (e.g., RSLV-132) exhibited reduced expression of inflammation-associated genes. For example, expression of IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, and STAT5B is reduced in patients treated with an RNA nuclease agent (e.g., RSLV-132) and exhibiting a clinical response. It was also found that patients achieving clinical response showed increased expression of inflammation-associated genes following treatment with RSLV-132. For example, the expression of CXCL10(IP-10), CD163, RIPK2 and CCR2 is increased in patients receiving RNA nuclease agent (e.g., RSLV-132) treatment and exhibiting a clinical response.

It has further been found that prior to administration of an RNA nuclease agent (e.g., RSLV-132), a unique gene expression profile exists in patients with autoimmune diseases (e.g., primary sjogren syndrome (pSS)) that subsequently have a positive clinical response to the RNA nuclease agent (e.g., RSLV-132). For example, when baseline gene expression is correlated with FACIT, ProF, or ESSPRI, specific characteristics are revealed in RSLV-132 responders. STAT1 and STAT2 expression decreased was associated with the FACIT test, ZNF606 expression increased and TRIM37 expression decreased was associated with the ProF test, ACKR3 expression increased and MAPK3K8 expression decreased was associated with the espri test.

Without being bound by theory, it is believed that certain patients may have circulating RNA molecules that promote chronic activation of these patient inflammatory pathways, and that removal of circulating non-coding RNA and removal of pro-inflammatory RNA by RNA nuclease agents (e.g., RSLV-132) is often helpful in treating patients with autoimmune diseases (e.g., pSS). Thus, the use of a "gene expression fingerprint" allows the identification of patients who would most benefit from treatment with an RNA nuclease agent (e.g., RSLV-132).

Definition of

Unless otherwise indicated, the terms used in the claims and the specification are defined as follows.

"amino acid" refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, such as hydroxyproline, gamma-carboxyglutamic acid, and O-phosphoserine. Amino acid analogs refer to compounds having the same basic chemical structure as a naturally occurring amino acid, i.e., a carbon, carboxyl, amino, and R group bound to a hydrogen, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Amino acid mimetics refer to compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.

Amino acids may be referred to herein by their commonly known three-letter symbols or one-letter symbols as recommended by the IUPAC-IUB biochemical nomenclature commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.

"amino acid substitution" refers to the replacement of at least one existing amino acid residue in a predetermined amino acid sequence (the amino acid sequence of the starting polypeptide) with a second, different "replacement" amino acid residue. "amino acid insertion" refers to the incorporation of at least one additional amino acid into a predetermined amino acid sequence. Although insertions typically consist of the insertion of one or two amino acid residues, larger "peptide insertions" may be made, for example, insertions of about three to about five or even up to about ten, fifteen or twenty amino acid residues. The inserted residues may be naturally occurring or non-naturally occurring as disclosed above. "amino acid deletion" refers to the removal of at least one amino acid residue from a predetermined amino acid sequence.

"polypeptide," "peptide," and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residues is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers.

"nucleic acid" refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single-or double-stranded form. Unless specifically limited, the term includes nucleic acids containing known analogs of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly includes conservatively modified variants thereof (e.g., degenerate codon substitutions) and complementary sequences, as well as the sequence explicitly indicated. In particular, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is replaced by mixed base and/or deoxyinosine residues (Batzer et al, Nucleic Acid Res 1991; 19: 5081; Ohtsuka et al, JBC 1985; 260: 2605-8); rossolini et al, mu omicl Cell Probes 1994; 8:91-8). For arginine and leucine, the modification of the second base may also be conservative. The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene.

The polynucleotide of the present invention may be composed of any polyribonucleotide or polydeoxyribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. For example, a polynucleotide may be composed of single-and double-stranded DNA, DNA comprising a mixture of single-and double-stranded regions, single-and double-stranded RNA, and RNA comprising a mixture of single-and double-stranded regions, hybrid molecules comprising DNA and RNA, which may be single-stranded, or more typically double-stranded, or a mixture of single-and double-stranded regions. In addition, a polynucleotide may be composed of a triple-stranded region comprising RNA or DNA or both RNA and DNA. Polynucleotides may also comprise one or more modified bases or DNA or RNA backbones modified for stability or other reasons. "modified" bases include, for example, tritylated bases and unusual bases such as inosine. Various modifications can be made to DNA and RNA; thus, "polynucleotide" includes chemically, enzymatically or metabolically modified forms.

As used herein, the terms "operably connected" or "operably coupled" refer to a juxtaposition (juxtaposition) in which the components described are in a relationship permitting them to function in their intended manner.

As used herein, the term "glycosylation" or "glycosylated" refers to the process or result of adding a sugar moiety to a molecule.

As used herein, the term "altered glycosylation" refers to a molecule that is aglycosylated, deglycosylated, or under-glycosylated.

As used herein, "glycosylation site" refers to a site that is likely to accept a carbohydrate moiety, as well as a site within a protein to which the carbohydrate moiety is actually attached, and includes any amino acid sequence that can act as an acceptor for oligosaccharides and/or carbohydrates.

As used herein, the term "aglycosylated" or "aglycosylated" refers to a molecule that produces an unglycosylated form (e.g., by engineering a protein or polypeptide to lack amino acid residues that serve as glycosylation receptors). Alternatively, the protein or polypeptide may be expressed in, for example, E.coli, to produce an aglycosylated protein or polypeptide.

As used herein, the term "deglycosylation" or "deglycosylated" refers to the process or result of enzymatic removal of the sugar moiety on a molecule.

As used herein, the term "under-glycosylation" or "under-glycosylation" refers to a molecule in which one or more carbohydrate structures that would normally be present if produced in a mammalian cell have been omitted, removed, modified or masked.

As used herein, the terms "Fc region" and "Fc domain" are the portions formed by the respective Fc domains (or Fc portions) of the two heavy chains of a native immunoglobulin, without the antigen-binding variable region. In some embodiments, the Fc domain begins at the hinge region upstream of the papain cleavage site and ends at the C-terminus of the antibody. Thus, a complete Fc domain comprises at least a hinge domain, a CH2 domain, and a CH3 domain. In certain embodiments, the Fc domain comprises at least one of: a hinge (e.g., upper, middle, and/or lower hinge region) domain, a CH2 domain, a CH3 domain, a CH4 domain, or a variant, portion, or fragment thereof. In other embodiments, the Fc domain comprises a complete Fc domain (i.e., the hinge domain, CH2 domain, and CH3 domain). In one embodiment, the Fc domain comprises a hinge domain (or portion thereof) fused to a CH3 domain (or portion thereof). In another embodiment, the Fc domain comprises a CH2 domain (or portion thereof) fused to a CH3 domain (or portion thereof). In another embodiment, the Fc domain consists of a CH3 domain or portion thereof. In another embodiment, the Fc domain consists of a hinge domain (or portion thereof) and a CH3 domain (or portion thereof). In another embodiment, the Fc domain consists of a CH2 domain (or portion thereof) and a CH3 domain. In another embodiment, the Fc domain consists of a hinge domain (or portion thereof) and a CH2 domain (or portion thereof). In one embodiment, the Fc domain lacks at least a portion of a CH2 domain (e.g., all or part of a CH2 domain). In one embodiment, the Fc domain of the invention comprises at least a portion of an Fc molecule required for FcRn binding as known in the art. In one embodiment, the Fc domain of the present invention comprises at least a portion of an Fc molecule required for protein a binding as known in the art. In one embodiment, the Fc domain of the present invention comprises at least a portion of an Fc molecule required for protein G binding as known in the art. An Fc domain herein generally refers to a polypeptide comprising all or part of the Fc domain of an immunoglobulin heavy chain. This includes, but is not limited to, polypeptides comprising the entire CH1, hinge, CH2, and/or CH3 domains, as well as fragments of such peptides comprising only, for example, the hinge, CH2, and CH3 domains. The Fc domain may be derived from any species and/or any subtype of immunoglobulin, including but not limited to human IgG1, IgG2, IgG3, IgG4, IgD, IgA, IgE, or IgM antibodies. Fc domains include native Fc and Fc variant molecules. As with Fc variants and native Fc, the term Fc domain includes molecules in monomeric or multimeric form, whether digested from the whole antibody or otherwise produced.

As described herein, one of ordinary skill in the art will appreciate that any Fc domain can be modified such that its amino acid sequence differs from the native Fc domain of a naturally occurring immunoglobulin molecule.

The Fc domain of the RNase-Fc fusion proteins of the present disclosure may be derived from different immunoglobulin molecules. For example, the Fc domain of an RNase-Fc fusion protein may comprise a CH2 and/or CH3 domain derived from an IgG1 molecule and a hinge region derived from an IgG3 molecule. In another example, the Fc domain may comprise a chimeric hinge region derived in part from an IgG1 molecule and in part from an IgG3 molecule. In another example, the Fc domain may comprise a chimeric hinge derived in part from an IgG1 molecule and in part from an IgG4 molecule. The wild-type human IgGl Fc domain has an amino acid sequence shown as SEQ ID NO. 20.

As used herein, the term "serum half-life" refers to the time required for the serum RNase-Fc fusion protein concentration to decrease by 50% in vivo. The shorter the serum half-life of the RNase-Fc fusion protein, the shorter the time to exert therapeutic effect.

As used herein, the term "RNA nuclease agent" refers to an agent comprising an RNase domain. In some embodiments, the RNA nuclease agent is a nuclease fusion protein comprising an RNase. In some embodiments, the RNA nuclease agent is an RNase-Fc fusion protein. In some embodiments, the RNase domain of the RNA nuclease agent is human pancreatic RNase 1. In some embodiments, the RNA nuclease agent is a polypeptide. In some embodiments, the RNA nuclease agent is RSLV-132.

As used herein, the term "RNase-containing nuclease fusion protein" refers to a polypeptide comprising at least one nuclease domain operably linked, with or without a linker, to a PK moiety (pharmacokinetic moiety), such as an Fc domain or variant or fragment thereof, and nucleic acids encoding such polypeptides. In some embodiments, the RNase-containing nuclease fusion protein is an "RNase-Fc fusion protein," which refers to a polypeptide comprising at least one nuclease domain operably linked, with or without a linker, to an Fc domain, or a variant or fragment thereof, and nucleic acids encoding such polypeptides. In some embodiments, the RNase-Fc fusion protein is a polypeptide comprising at least two nuclease domains operably linked, with or without a linker, to an Fc domain, or a variant or fragment thereof, and nucleic acids encoding such polypeptides. In some embodiments, the nuclease domain is human RNase 1. In some embodiments, the RNase-Fc fusion protein comprises one or more RNase domains and one or more Fc domains. In some embodiments, the RNase-Fc fusion protein comprises one or more RNase domains, one or more Fc domains, and one or more DNase domains. In some embodiments, the RNase-Fc fusion protein comprises an RNase 1 domain operably linked to the N-or C-terminus of an Fc domain and a DNase domain operably linked to the N-or C-terminus of an Fc domain. In some embodiments, the RNase-Fc fusion protein is a tandem RNase-Fc fusion protein, e.g., one or more RNase 1 domains and/or one or more DNase domains are joined in tandem to the N-or C-terminus of one or more Fc domains. In some embodiments, the RNase-Fc fusion protein is a homodimeric RNase-Fc fusion protein (two identical polypeptides). In some embodiments, the RNase-Fc fusion protein is a heterodimeric RNase-Fc fusion protein (two different polypeptides). In some embodiments, the domain of the RNase-Fc fusion protein is operably linked to a linker domain. In some embodiments, the domains of the RNase-Fc fusion protein are operably linked without a linker domain.

As used herein, the term "tandem RNase-Fc fusion protein" refers to a polypeptide comprising at least two nuclease domains (from N-terminus to C-terminus) and an Fc domain, or variants or fragments thereof, linked in tandem, as well as nucleic acids encoding such polypeptides. In some embodiments, the tandem RNase-Fc fusion protein is a polypeptide comprising at least two RNase1 domains operably linked in tandem to at least one Fc domain. In some embodiments, the tandem RNase-Fc fusion protein is a polypeptide comprising at least one DNase1 domain and at least one RNase1 domain operably linked in tandem to the at least one Fc domain. In some embodiments, the tandem RNase-Fc fusion protein comprises, from N-terminus to C-terminus, a DNase1 domain, a first linker, an RNase1 domain, a second linker, and an Fc domain, or a variant or fragment thereof.

As used herein, the term "heterodimeric RNase-Fc fusion protein" refers to a heterodimer comprising a first and a second polypeptide, which together comprise at least two nuclease domains and two Fc domains, variants or fragments thereof, and nucleic acids encoding such polypeptides. In some embodiments, the heterodimer comprises a first RNase1 domain operably linked, with or without a linker, to the N-or C-terminus of the first Fc domain, and a second RNase1 domain operably linked, with or without a linker, to the N-or C-terminus of the second Fc domain, such that the first RNase1 and the second RNase1 domains are at the same end (N-or C-terminus) of the heterodimer. In some embodiments, the heterodimer comprises a first RNase1 domain operably linked, with or without a linker, to the N-terminus of a first Fc domain, and a second RNase1 domain operably linked, with or without a linker, to the C-terminus of a second Fc domain. In some embodiments, the first RNase1 domain is operably linked to the C-terminus of the first Fc domain, with or without a linker, and the first RNase1 domain is operably linked to the N-terminus of the second Fc domain, with or without a linker. In some embodiments, the first and second RNase1 domains of the heterodimer are different. In some embodiments, the heterodimeric RNase-Fc fusion protein is a heterodimer comprising at least one DNase1 domain and at least one RNase1 domain, said DNase1 domain and RNase1 domain being operably linked to at least one Fc domain, wherein DNase1 domain is operably linked to the N-or C-terminus of a first Fc domain with or without a linker, and RNase1 domain is operably linked to the N-or C-terminus of the same (first Fc domain) or a different Fc domain (second Fc domain) with or without a linker, such that DNase1 domain and RNase1 domain are located at opposite ends (N-or C-termini) of the same (first Fc domain) or different Fc domain (second Fc domain). In some embodiments, the heterodimer comprises a DNase.

As used herein, the term "homodimeric RNase-Fc fusion protein" refers to a homodimer comprising first and second polypeptides which together comprise at least two nuclease domains and two Fc domains, variants or fragments thereof, and nucleic acids encoding such polypeptides. In some embodiments, the homodimer comprises a first RNase1 domain operably linked, with or without a linker, to the N-or C-terminus of the first Fc domain, and a second RNase1 domain operably linked, with or without a linker, to the N-or C-terminus of the second Fc domain, such that the first RNase1 and the second RNase1 domains are at the same end (N-or C-terminus) of the homodimer. In some embodiments, the first and second RNase1 domains of the homodimer are the same. In some embodiments, the homodimer comprises one RNase1 domain operably linked, with or without a linker, to the N-or C-terminus of one Fc domain and one DNase domain operably linked, with or without a linker, to the N-or C-terminus of a second Fc domain. In some embodiments, the RNase1 and DNase domains are located at the same end (N-or C-terminus) of the Fc domain. In some embodiments, the RNase1 and DNase domains are located at opposite ends of the Fc domain.

As used herein, the term "dimer" refers to a macromolecular complex formed by two macromolecules (e.g., polypeptides). "homodimer" refers to a dimer formed from two identical macromolecules (e.g., polypeptides). "heterodimer" refers to a dimer formed from two different macromolecules (e.g., polypeptides).

As used herein, the term "variant" refers to a polypeptide derived from a wild-type nuclease (e.g., RNase) or Fc domain and differs from wild-type by one or more alterations, i.e., substitutions, insertions, and/or deletions at one or more positions. Substitution refers to the replacement of an amino acid occupying a position with a different amino acid. Deletion refers to the removal of an amino acid occupying a position. Insertion refers to the addition of 1 or more (e.g., 1-3 amino acids) immediately adjacent to the amino acid occupying the position. A variant polypeptide must have less than 100% sequence identity or similarity to the wild-type polypeptide. In some embodiments, a variant polypeptide will have an amino acid sequence that has from about 75% to less than 100% amino acid sequence identity or similarity to the amino acid sequence of a wild-type polypeptide, or from about 85% to less than 100%, or from about 90% to less than 100% (e.g., 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%) or from about 95% to less than 100%, e.g., over the length of the variant polypeptide.

In certain aspects, the RNase-Fc fusion protein employs one or more "linker domains," such as polypeptide linkers. As used herein, the term "linker domain" refers to one or more amino acids that link two or more peptide domains in a linear polypeptide sequence. As used herein, the term "polypeptide linker" refers to a peptide or polypeptide sequence (e.g., a synthetic peptide or polypeptide sequence) that links two or more polypeptide domains in a linear amino acid sequence of a protein. For example, polypeptide linkers can be used to operably link a nuclease domain (e.g., RNase) to an Fc domain. In some embodiments, such polypeptide linkers provide flexibility to the polypeptide molecule. In some embodiments, a polypeptide linker is used to link (e.g., genetically fuse) the RNase domain to the Fc domain. The RNase-Fc fusion protein may comprise more than one linker domain or peptide linker. Various peptide linkers are known in the art.

As used herein, the term "gly-ser polypeptide linker" refers to a peptide consisting of glycine and serine residues. An exemplary Gly/ser polypeptide linker comprises the amino acid sequence (Gly)4Ser) n. In some embodiments, n is 1 or more, such as 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 9 or more, or 10 or more (e.g., (Gly)4Ser) 10). Another exemplary Gly/Ser polypeptide linker comprises the amino acid sequence Ser (Gly)4Ser) n. In some embodiments, n is 1 or more, e.g., 2 or more, 3 or more, 4 or more, 5 or more, 6 or more, 7 or more, 8 or more, 9 or more, or 10 or more (e.g., Ser (Gly)4Ser)10)。

As used herein, the terms "couple," "conjugate," "link," "fusion" or "fusion" are used interchangeably. These terms refer to the joining together of two or more elements (elements) or components or domains by any means including chemical conjugation or recombinant means. Methods of chemical conjugation (e.g., using heterobifunctional crosslinkers) are known in the art.

A polypeptide or amino acid sequence "derived from" a given polypeptide or protein refers to the source of the polypeptide. Preferably, the polypeptide or amino acid sequence derived from a particular sequence has substantially the same amino acid sequence as the sequence or a portion thereof, wherein the portion consists of at least 10-20 amino acids, preferably at least 20-30 amino acids, more preferably at least 30-50 amino acids, or the sequence from which it originates can be otherwise identified by one of ordinary skill in the art. A polypeptide derived from another polypeptide may have one or more mutations relative to the starting polypeptide, for example one or more amino acid residues have been replaced by another amino acid residue or have an insertion or deletion of one or more amino acid residues.

In one embodiment, there is one amino acid difference between the starting polypeptide sequence and the sequence from which it is derived. Identity or similarity with respect to this sequence is defined herein as the percentage of amino acid residues in the candidate sequence that are identical (i.e., the same residues) as the starting amino acid residue, after aligning the sequences and introducing gaps (gaps), if necessary, to achieve the maximum percentage of sequence identity.

In one embodiment, the polypeptide of the present disclosure consists of, consists essentially of, or comprises an amino acid sequence as set forth in the sequence listing or sequence listing disclosed herein and functionally active variants thereof. In one embodiment, the polypeptide comprises an amino acid sequence that is at least 80%, such as at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to the sequence listing disclosed herein or an amino acid sequence set forth in the sequence listing. In some embodiments, the polypeptide comprises a contiguous amino acid sequence that is at least 80%, e.g., at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to the contiguous amino acid sequence set forth in the sequence listing or sequence listings disclosed herein. In some embodiments, the polypeptide comprises contiguous amino acids having at least 10, e.g., at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 200, at least 300, at least 400, or at least 500 (or any integer within these numbers) of the amino acid sequences set forth in the sequence listings or sequence listings disclosed herein.

In some embodiments, the RNase-Fc fusion proteins of the present disclosure are encoded by a nucleotide sequence. The nucleotide sequences of the present disclosure can be used in a variety of applications, including: cloning, gene therapy, protein expression and purification, mutation introduction, DNA vaccination of a host in need, antibody production for e.g. passive immunization, PCR, primer and probe generation, siRNA design and generation (see e.g. dharmacosidesign website), etc. In some embodiments, the nucleotide sequence of the present disclosure comprises, consists of, or consists essentially of a nucleotide sequence encoding an amino acid sequence of an RNase-Fc fusion protein selected from the group consisting of the sequence Listing (Table) or the sequence Listing (Listing). In some embodiments, the nucleotide sequence comprises a nucleotide sequence that is at least 80%, such as at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a nucleotide sequence encoding an amino acid sequence of the sequence listing or sequence listings disclosed herein. In some embodiments, the nucleotide sequence comprises a contiguous nucleotide sequence that is at least 80%, such as at least 81%, at least 82%, at least 83%, at least 84%, at least 85%, at least 86%, at least 87%, at least 88%, at least 89%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a contiguous nucleotide sequence encoding an amino acid sequence listed in the sequence listing or sequence listing disclosed herein. In some embodiments, the nucleotide sequence comprises contiguous nucleotides having at least 10, such as at least 15, e.g., at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 200, at least 300, at least 400, or at least 500 (or any integer within these numbers) nucleotide sequences encoding an amino acid sequence listed in the sequence listing or sequence listing disclosed herein.

It is also understood by those of ordinary skill in the art that RNase-Fc fusion proteins can be altered such that they differ in sequence from the naturally occurring or native sequence from which their components (e.g., nuclease domain, linker domain, and Fc domain) are derived, while retaining the desired activity of the native sequence. For example, nucleotide or amino acid substitutions or changes in "non-essential" amino acid residues that result in conservative substitutions may be made. An isolated nucleic acid molecule encoding a non-natural variant may be produced by introducing one or more nucleotide substitutions, additions or deletions in the nucleotide sequence of the RNase-Fc fusion protein, thereby introducing one or more amino acid substitutions, additions or deletions into the encoded protein. Mutations can be introduced by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis.

The RNase-Fc fusion protein may comprise conservative amino acid substitutions at one or more amino acid residues, for example at essential or non-essential amino acid residues. A "conservative amino acid substitution" is one in which an amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art, including basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), β -branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine tryptophan, histidine). Thus, a non-essential amino acid residue in an RNase-Fc fusion protein is preferably replaced with another amino acid residue from the same side chain family. In another embodiment, the amino acid string may be replaced with a structurally similar string that differs in the order and/or composition of the side chain family members. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of the coding sequence, such as by saturation mutagenesis, and the resulting mutants can be incorporated into an RNase-Fc fusion protein and screened for their ability to bind to the desired target.

As used herein, the term "modulator of the innate immune system" refers to the expression and secretion of any gene, protein, nucleic acid, or microrna, including cytokines and chemokines, associated with the expression or regulation of the innate immune response; dendritic cell activation; and the complement cascade (complement cascade). In some embodiments, the modulator of the innate immune system comprises an inflammation-associated molecule. In some embodiments, the modulator of the innate immune system comprises an inflammation-associated gene. In some embodiments, the modulator of the innate immune system comprises an inflammation-associated protein.

In some embodiments, the modulator of the innate immune system comprises a gene or protein associated with modulation of signal transduction, interferon family members, complement, antigen processing, signal transduction, ubiquitination, chemotaxis, cell adhesion, and polymerase activity.

The term "innate immune system" refers to a non-specific defense mechanism against antigens. The innate immune response is driven not by specificity for a particular antigen, but by the presence of the antigen. The functions of the innate immune system include acting as a physical and chemical barrier to infectious agents, recognition and removal of foreign substances by leukocytes, dendritic cell activation, cytokine and chemokine secretion, complement cascade activation, and adaptive immune system activation.

As used herein, the term "inflammation-associated molecule" refers to a molecule that plays a role in inflammation or inflammatory response. In some embodiments, the inflammation-associated molecule is a pro-inflammatory molecule. In some embodiments, the inflammation-associated molecule is an anti-inflammatory molecule. In some embodiments, the inflammation-associated molecule is an inflammation-associated gene. In some embodiments, the inflammation-associated molecule is an inflammation-associated protein. In some embodiments, the inflammation-associated molecule is an inflammation-associated cytokine. In some embodiments, the inflammation-associated molecule is an inflammatory mediator.

As used herein, the term "inflammation-associated gene" refers to a gene that plays a role in inflammation or inflammatory response. In some embodiments, the inflammation-associated gene is a pro-inflammatory gene. In some embodiments, the inflammation-associated gene is an anti-inflammatory gene. In some embodiments, the inflammation-associated gene encodes an inflammation-associated protein. In some embodiments, the inflammation-associated gene encodes a cytokine.

As used herein, the term "inflammation-associated protein" refers to a protein that plays a role in inflammation or inflammatory response. In some embodiments, the inflammation-associated protein is a pro-inflammatory protein. In some embodiments, the inflammation-associated protein is an anti-inflammatory protein. In some embodiments, the inflammation-associated protein is a cytokine.

As used herein, the term "pro-inflammatory molecule" refers to a molecule that enhances or stimulates an inflammatory response. In some embodiments, the proinflammatory molecule is a "proinflammatory gene". In some embodiments, the proinflammatory molecule is a "proinflammatory protein". In some embodiments, the proinflammatory gene encodes a proinflammatory protein. In some embodiments, the proinflammatory molecule is an "inflammatory cytokine.

In some embodiments, the term "stimulating an inflammatory response" refers to stimulating the production of inflammatory cytokines.

As used herein, the term "inflammatory cytokine" refers to a signaling molecule (cytokine) that is secreted from immune cells (e.g., helper T cells and macrophages) and plays a role in inflammatory responses.

As used herein, the term "gene expression profile" refers to a technique for identifying genes expressed in a sample and/or determining their degree of expression at a particular time. The term "inflammation-associated gene expression profile" refers to a gene expression profile that identifies the expression of inflammation-associated genes at a particular time and/or determines their degree of expression.

The term "ameliorating" refers to any therapeutically beneficial result in the treatment of a disease state, e.g., an autoimmune disease state (e.g., SLE, sjogren's syndrome), including prevention, lessening of severity or progression, alleviation or cure.

As used herein, the terms "primary Sjogren's syndrome", "Sjogren's disease", and "Sjogren's" may be used interchangeably.

The term "in situ" refers to a process that occurs in living cells that grow separately from a living organism, e.g., growth in tissue culture.

The term "in vivo" refers to a process that occurs in a living organism.

The term "mammal" or "subject" or "patient" as used herein includes humans and non-humans, including, but not limited to, humans, non-human primates, canines, felines, murines, bovines, equines, and porcines.

The term "identity," in the context of two or more nucleic acid or polypeptide sequences, refers to two or more sequences or subsequences that have a specified percentage of nucleotides or amino acid residues that are the same when compared and aligned for maximum correspondence, such as using the sequence comparison algorithms described below (e.g., BLASTP and BLASTN or other algorithms available to the skilled artisan) or by visual inspection.

For sequence comparison, typically one sequence serves as a reference sequence to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. The sequence comparison algorithm then calculates the percent sequence identity of the test sequence relative to the reference sequence based on the specified program parameters.

Optimal alignment of sequences for comparison can be performed, for example, by the local homology algorithm of Smith & Waterman (Adv Appl Math 1981; 2:482), by the homology alignment algorithm of Needleman & Wunsch (J Mol Biol 1970; 48:443), by the similarity search method of Pearson & Lipman (PNAS 1988; 85:2444), by the computerisation of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group,575Science Dr., Madison, Wis.) or by visual inspection (see generally Ausubel et al, infra).

One example of an algorithm suitable for determining percent sequence identity and sequence similarity is the BLAST algorithm, described in Altschul et al, J Mol Biol 1990; 215:403-10 are described. Software for performing BLAST analysis is publicly available through the National Center for Biotechnology Information (National Center for Biotechnology Information) website.

The term "sufficient amount" refers to an amount sufficient to produce a desired effect.

The term "therapeutically effective amount" is an amount effective to ameliorate the symptoms of a disease. A therapeutically effective amount may be a "prophylactically effective amount" since prophylaxis may be considered treatment.

The term "about" will be understood by one of ordinary skill and will vary to some extent depending on the context in which it is used. If the use of this term is not clear to the ordinarily skilled artisan in view of the context in which it is used, "about" will mean less than plus or minus 10% of the specified value.

It must be noted that, as used in the specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise.

RNase-containing nuclease fusion protein

RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, of the present disclosure include at least one enzymatically active RNase domain or fragment or variant thereof (e.g., human RNase1, or a fragment or variant thereof) operably linked to a PK moiety that provides a scaffold and/or increases the RNase domain half-life in vivo as compared to a nuclease domain without such PK moiety. In some aspects, the RNase-containing nuclease fusion protein is an RNase-Fc fusion protein comprising at least one enzymatically active RNase domain or fragment or variant thereof (e.g., human RNase1, or a fragment or variant thereof) operably linked to an Fc domain (e.g., human IgGl Fc domain, or a variant or fragment thereof) that alters the serum half-life of the nuclease molecule fused thereto as compared to a nuclease molecule not fused to an Fc domain or a variant or fragment thereof.

In some embodiments, RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, of the present disclosure are operably coupled to an Fc domain, or variant or fragment thereof, via a linker domain. In some embodiments, the linker domain is a linker peptide. In some embodiments, the linker domain is a linker nucleotide.

In some embodiments, RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, of the present disclosure include leader sequences, such as leader peptides. In some embodiments, the leader molecule is a leader peptide located N-terminal to the nuclease domain. In some embodiments, the RNase-Fc fusion protein comprises a leader peptide at the N-terminus of the molecule, wherein the leader peptide is subsequently cleaved from the RNase-Fc fusion protein. Methods for generating nucleic acid sequences encoding leader peptides fused to recombinant proteins are well known in the art. In some embodiments, the RNase-Fc fusion proteins are expressed with or without a leader fused to their N-terminus. One skilled in the art can predict and/or infer the protein sequence of the RNase-Fc fusion proteins of the present disclosure after cleavage of the fusion leader peptide.

In some embodiments, the leader peptide is a VK3 leader peptide (VK3LP), wherein the leader peptide is fused to the N-terminus of the RNase-Fc fusion protein. Such leader sequences can increase the level of synthesis and secretion of the RNase-Fc fusion protein in mammalian cells. In some embodiments, the leader sequence is cleaved to produce an RNase-Fc fusion protein. In some embodiments, the RNase-Fc fusion protein of the present disclosure is expressed without a leader peptide fused to its N-terminus, and the resulting RNase-Fc fusion protein has an N-terminal methionine.

In some embodiments, the RNase-Fc fusion protein of the present disclosure includes a VK3 leader peptide, e.g., SEQ ID NO:49(RSLV-132), fused to the N-terminus of the RNase-Fc fusion protein. In some embodiments, the RNase-Fc fusion protein of the present disclosure does not comprise a leader sequence, e.g., SEQ ID NO:50 (RSLV-132).

In some embodiments, the RNase-Fc fusion protein comprises an RNase domain, or variant or fragment thereof, operably coupled to the N-or C-terminus of an Fc domain. In some embodiments, the RNase-Fc fusion protein comprises both an RNase domain and a DNase domain.

In some embodiments, the RNase-Fc fusion protein comprises two nuclease domains (e.g., two RNase domains) operably linked to each other in tandem and further operably coupled to the N-or C-terminus of the same or different Fc domains or variants or fragments thereof.

The sequence listing provides sequences of exemplary RNase-Fc fusion proteins of various configurations.

In some embodiments, the RNase-Fc fusion protein is a polynuclease protein (e.g., two RNA nucleases, or one RNase and one DNase) fused to the same or different Fc domain or variant or fragment thereof that specifically binds to an extracellular immune complex.

In one embodiment, the nuclease domain is operably coupled (e.g., chemically coupled or genetically fused (e.g., directly or through a polypeptide linker)) to the N-terminus of the Fc domain or variant or fragment thereof. In another embodiment, the nuclease domain is operably coupled (e.g., chemically coupled or genetically fused (e.g., directly or through a polypeptide linker)) to the C-terminus of the Fc domain or variant or fragment thereof. In other embodiments, the nuclease domain is operably coupled (e.g., chemically coupled or genetically fused (e.g., directly or through a polypeptide linker)) through an amino acid side chain of an Fc domain or variant or fragment thereof.

In certain embodiments, the RNase-Fc fusion proteins of the present disclosure comprise two or more nuclease domains and at least one Fc domain, or variants or fragments thereof. For example, a nuclease domain can be operably coupled to the N-terminus and C-terminus of the same or different Fc domains, variants or fragments thereof through an optional linker between the nuclease domain and one or more Fc domains, variants or fragments thereof. In some embodiments, the nuclease domains are the same, e.g., RNase and RNase. In other embodiments, the nuclease domains are different, e.g., two different RNA nucleases or an RNase and a DNase.

In some embodiments, two or more nuclease domains are operably linked in tandem to each other (e.g., via a polypeptide linker), and the tandem array of nuclease domains are operably coupled (e.g., chemically conjugated or genetically fused (e.g., directly or via a polypeptide linker)) to the C-terminus or N-terminus of the same or different Fc domains or variants or fragments thereof. In other embodiments, the tandem array of nuclease domains is operably coupled to the N-terminus and the C-terminus of the same Fc domain or variant or fragment thereof. In some embodiments, the nuclease domain is operably linked in series, with or without a linker, to the N-or C-terminus of the same or a different Fc domain (e.g., N-RNase-C, N-RNase-DNase-C or N-DNase-RNase-C). In some embodiments, the tandem RNase-Fc fusion protein forms a homodimer or a heterodimer.

In other embodiments, one or more nuclease domains are inserted between two Fc domains or variants or fragments thereof. For example, one or more nuclease domains may form all or part of a polypeptide linker of an RNase-Fc fusion protein of the present disclosure.

In some embodiments, the RNase-Fc fusion protein comprises at least two nuclease domains (e.g., RNase and RNase or RNase and DNase), at least one linker domain, and at least one Fc domain, or variants or fragments thereof.

In some embodiments, the RNase-Fc fusion proteins of the present disclosure comprise an Fc domain, or a variant or fragment thereof, as described herein, thereby increasing the serum half-life and bioavailability of the RNase-Fc fusion protein. In some embodiments, the RNase-Fc fusion protein comprises one or more polypeptides, such as a polypeptide comprising the amino acid sequence set forth in any one of SEQ ID NOS: 44-58.

One skilled in the art will appreciate that other configurations of nuclease domains and Fc domains are possible, including optional linkers between the nuclease domains and/or between the nuclease domains and Fc domains. It is also understood that the domain orientation can be altered so long as the nuclease domain is active in the particular configuration tested.

In certain embodiments, the RNase-Fc fusion proteins of the present disclosure have at least one nuclease domain specific for a target molecule that mediates a biological effect. In another embodiment, binding of an RNase-Fc fusion protein of the present disclosure to a target molecule (e.g., RNA or DNA) results in the reduction or elimination of the target molecule, e.g., from a cell, tissue, or circulation.

In other embodiments, the RNase-Fc fusion proteins of the present disclosure can be assembled together or with other polypeptides to form binding proteins having two or more polypeptides ("multimers"), wherein at least one polypeptide of the multimers is an RNase-Fc fusion protein of the present disclosure. Exemplary multimeric forms include dimeric, trimeric, tetrameric, and hexamer altered binding proteins, and the like. In one embodiment, the polypeptides of the multimer are identical (i.e., homomeric altered binding proteins, e.g., homodimers, homotetramers). In another embodiment, the polypeptides of the multimer are different (e.g., heteromeric). In one embodiment, the RNase-Fc fusion proteins of the present disclosure assemble together to form a dimer. In one embodiment, the dimer is a homodimer. In one embodiment, the dimer is a heterodimer.

In some embodiments, the RNase-Fc fusion protein has a serum half-life that is increased at least about 1.5 fold, such as at least 3 fold, at least 5 fold, at least 10 fold, at least about 20 fold, at least about 50 fold, at least about 100 fold, at least about 200 fold, at least about 300 fold, at least about 400 fold, at least about 500 fold, at least about 600 fold, at least about 700 fold, at least about 800 fold, at least about 900 fold, at least about 1000 fold, or 1000 fold or more, relative to a corresponding nuclease molecule that is not fused to an Fc domain or variant or fragment thereof. In other embodiments, the RNase-Fc fusion protein has a serum half-life that is reduced by at least about 1.5 fold, such as at least 3 fold, at least 5 fold, at least 10 fold, at least about 20 fold, at least about 50 fold, at least about 100 fold, at least about 200 fold, at least about 300 fold, at least about 400 fold, at least about 500 fold, or 500 fold or less relative to a corresponding nuclease molecule that is not fused to an Fc domain or variant or fragment thereof. The serum half-life of the RNase-Fc fusion proteins of the present disclosure can be determined using art-recognized routine methods.

In some embodiments, the RNase activity in the RNase-Fc fusion protein is not less than about 10-fold, e.g., 9-fold, 8-fold, 7-fold, 6-fold, 5-fold, 4-fold, 3-fold, or 2-fold greater than the activity of a control RNase molecule. In some embodiments, the activity of the RNase in the RNase-Fc fusion protein is about equal to the activity of a control RNase molecule.

In some embodiments, the RNase-Fc fusion protein may be active on extracellular immune complexes (e.g., in soluble form or deposited insoluble complexes) containing DNA and/or RNA.

In some embodiments, the activity of the RNase-Fc fusion protein is detectable in vitro and/or in vivo.

In another aspect, a multifunctional RNase molecule is provided that is attached to another enzyme or antibody with binding specificity, such as an scFv targeting RNA or DNA or a second nuclease domain with the same or different specificity as the first domain.

In some embodiments, the linker domain comprises a (gly4ser)3, 4, or 5 variant that alters the length of the linker by a 5 amino acid progression. In another embodiment, the linker domain is about 18 amino acids in length and includes an N-linked glycosylation site that is susceptible to in vivo protease cleavage. In some embodiments, the N-linked glycosylation site can protect the RNase-Fc fusion protein from cleavage in the linker domain. In some embodiments, the N-linked glycosylation site can help separate the folding of the independent functional domains separated by the linker domain.

In some embodiments, the linker domain is an NLG linker (VDGASSPVNVSSPSVQDI) (SEQ ID NO: 37).

In some embodiments, the RNase-Fc fusion protein comprises substantially all or at least an enzymatically active fragment of a DNase. In some embodiments, the DNase is a type I secreted DNase, preferably a human DNase, such as mature human pancreas DNase1(UniProtKB entry P24855, SEQ ID NO: 6). In some embodiments, the naturally occurring variant allele A114F (SEQ ID NO:8) that exhibits reduced sensitivity to actin is contained in the RNase 1 of the RNase-Fc fusion protein (see Pan et al, JBC 1998; 273: 18374-81; Zhen et al, BBRC 1997; 231: 499-504; Rodriguez et al, Genomics 1997; 42: 507-13). In other embodiments, a naturally occurring variant allele G105R (SEQ ID NO:9) that exhibits high DNase activity relative to wild-type DNase1 is included in the DNase1 of an RNase-Fc fusion protein (see Yasuda et al, Int J Biochem Cell Biol 2010; 42: 1216-25). In some embodiments, the mutation is introduced into the RNase-FC fusion protein to produce a more stable derivative of human DNase 1. In some embodiments, the DNase is human wild-type DNase1 or human mutant DNase 1A 114F which removes all potential N-linked glycosylation sites, i.e., asparagine residues at positions 18 and 106 of the DNase1 domain as set forth in SEQ ID NO:6 (i.e., human DNase 1N 18S/N106S/A114F, SEQ ID NO:11), corresponding to asparagine residues 40 and 128, respectively, of full-length pancreatic DNase1 having a native leader sequence (SEQ ID NO: 5).

In some embodiments, the DNase is human DNase1 comprising one or more basic (i.e. positively charged) amino acid substitutions to increase DNase function and chromatin cleavage. In some embodiments, basic amino acids are introduced into human DNase1 at the DNA binding interface to enhance binding to negatively charged phosphates on DNA substrates (see US 7407785; US 6391607). This hyperactive DNase1 may be referred to as "chromatin cutter".

In some embodiments, 1, 2, 3, 4, 5, or 6 basic amino acid substitutions are introduced into DNase 1. For example, one or more of the following residues are mutated to enhance DNA binding: gln9, Glu13, Thr14, His44, Asn74, Asn110, Thr 205. In some embodiments, one or more of the foregoing amino acids is replaced with a basic amino acid, such as arginine, lysine, and/or histidine. For example, a human DNase may comprise one or more of the following substitutions: Q9R, E13R, T14K, H44K, N74K, N110R, T205K. In some embodiments, human DNase1 further comprises an a114F substitution, which reduces sensitivity to actin (see US 6348343). In one embodiment, human DNase1 includes the following substitutions: E13R, N74K, a114F, and T205K.

In some embodiments, human DNase1 further includes mutations that remove potential glycosylation sites, for example, asparagine residues at positions 18 and 106 of the DNase1 domain as shown in SEQ ID No. 6, which correspond to asparagine residues at positions 40 and 128, respectively, of full-length pancreatic DNase1 having the native leader sequence. In one embodiment, human DNase1 includes the following substitutions: E13R/N74K/A114F/T205K/N18S/N106S.

In some embodiments, the DNase is a DNase 1-like (DNaseL) enzyme, 1-3(UniProtKB entry Q13609; SEQ ID NO: 15). In some embodiments, the DNase is three-primer repair exonuclease 1(TREX 1; UniProtKB entry Q9NSU 2; SEQ ID NO: 16). In some embodiments, the DNase is DNase 2. In some embodiments, DNase2 is DNase2 α (i.e., DNase 2; UnitRotKB entry O00115SEQ ID NO:18) or DNase2 β (i.e., DNase 2-like acid DNase; UnitRotKB entry Q8WZ 79; SEQ ID NO: 19). In some embodiments, the N-linked glycosylation site of DNase 1L3, TREX1, DNase2 α or DNase2 β is mutated to remove potential N-linked glycosylation sites. In some embodiments, DNase-linker-Fc domains are prepared that contain linker domains of 20 or 25 amino acids.

In some embodiments, the activity of the DNase in the RNase-Fc fusion protein is not less than about 10 fold, such as 9 fold, 8 fold, 7 fold, 6 fold, 5 fold, 4 fold, 3 fold or 2 fold, of the activity of a control DNase molecule. In some embodiments, the activity of the DNase in the RNase-Fc fusion protein is about equal to the activity of the control DNase molecule.

In some embodiments, the RNase-Fc fusion proteins of the present disclosure include human RNase 1. In some embodiments, the RNase-Fc fusion protein comprises a wild-type human RNase1 domain. In some embodiments, the RNase-Fc fusion protein comprises human pancreatic RNase1 of the RNase A family (UniProtKB entry P07998; SEQ ID NO: 1). In some embodiments, the RNase-Fc fusion protein comprises the mature form of human pancreatic RNase1 as set forth in SEQ ID NO: 2. In some embodiments, the RNase-Fc domain comprises a human RNase1 domain having one or more mutations. In some embodiments, human RNase1 is mutated to remove all potential N-linked glycosylation sites, namely asparagine residues at positions 34, 76 and 88 of the RNase1 domain shown in SEQ ID NO:2 (human RNase 1N 34S/N76S/N88S, SEQ ID NO:4), corresponding to asparagine residues at positions 62, 104 and 116, respectively, of full-length pancreatic RNase1 with the native leader sequence (SEQ ID NO: 1). In some embodiments, an RNase 1-linker-Fc containing a linker domain of 20 or 25 amino acids is prepared.

In some embodiments, the RNase-Fc fusion protein comprises mammalian RNase 1. In some embodiments, the RNase-Fc fusion protein comprises primate RNase 1. In some embodiments, the RNase-Fc fusion protein comprises rodent RNase 1. In some embodiments, the RNase-Fc fusion protein comprises mouse RNase 1. In some embodiments, the RNase-Fc fusion protein comprises rat RNase 1. In some embodiments, the RNase-Fc fusion protein comprises monkey RNase 1. In some embodiments, the RNase-Fc fusion protein comprises goat RNase 1. In some embodiments, the RNase-Fc fusion protein comprises rabbit RNase 1. In some embodiments, the RNase-Fc fusion protein comprises equine RNase 1. In some embodiments, the RNase-Fc fusion protein comprises canine RNase 1. In some embodiments, the RNase1 domain is a mutated RNase1 domain.

In some embodiments, the RNase-Fc fusion protein comprises an RNase molecule linked to an Fc domain that specifically binds to an extracellular immune complex. In some embodiments, the Fc domain does not bind effectively to an Fc γ receptor. In one aspect, the RNase-Fc fusion protein does not bind Clq efficiently. In other aspects, the RNase-Fc fusion protein comprises an in-frame Fc domain of IgG 1. In other aspects, the RNase-Fc fusion protein further comprises mutations in the hinge, CH2, and/or CH3 domains. In other aspects, the mutations are P238S, P331S, or N297S, and may include mutations in one or more of the three hinge cysteines. In some such aspects, residues 220, 226, and 229 located in one or more of the three hinge cysteines are mutated, such as by substitution of one or more cysteine residues with serine, e.g., C220S, C226S, and/or C229S, according to EU index numbering. In some embodiments, one of the three hinge region cysteines is replaced with a serine, e.g., C220S is also referred to herein as an "SCC hinge". In some embodiments, all three hinge region cysteines are replaced with serine, C220S, C226S, and C229S, also referred to herein as "SSS hinges". In other aspects, the RNase-Fc fusion protein comprises SCC hinges, but its human IgG1 FcCH2 and CH3 domains are wild-type and bind efficiently to Fc receptors, facilitating uptake of the RNase-Fc fusion protein into the endocytosis chamber to which they bind. In other aspects, the RNase-Fc fusion protein has activity against single-stranded and/or double-stranded RNA substrates.

In some aspects, the RNase-Fc fusion protein comprises a mutated Fc domain. In some aspects, the RNase-Fc fusion protein comprises a mutated IgGl Fc domain. In some aspects, the mutant Fc domain comprises one or more mutations in the hinge, CH2, and/or CH3 domain. In some aspects, the mutant Fc domain comprises the P238S mutation. In some aspects, the mutant Fc domain comprises the P331S mutation. In some aspects, the mutant Fc domain comprises the P238S mutation and the P331S mutation. In some aspects, the mutant Fc domain comprises P238S and/or P331S, and may include a mutation in one or more of the three hinge cysteines. In some aspects, the mutant Fc domain comprises one or more mutations in P238S and/or P331S, and/or three hinge cysteines. In some aspects, the mutant Fc domain comprises P238S and/or P331S, and/or three hinge cysteines mutated to SSS or one hinge cysteine mutated to SCC. In some aspects, the mutant Fc domain comprises mutations in P238S and P331S and three hinge cysteines. In some aspects, the mutant Fc domain comprises P238S and P331S and SCC or SSS. In some aspects, the mutant Fc domain comprises P238S and P331S, and SCC. In some aspects, the mutant Fc domain comprises P238S SSS. In some aspects, the mutant Fc domain comprises P331S and SCC or SSS. In some aspects, the mutant Fc domain comprises a mutation of one or more of the three hinge cysteines. In some aspects, the mutant Fc domain comprises mutations in three hinge cysteines. In some aspects, the mutant Fc domain comprises three hinge cysteine mutations to SSS. In some aspects, the mutant Fc domain comprises a SCC mutation of one of the three hinge cysteines. In some aspects, the mutant Fc domain comprises SCC or SSS. In some aspects, the mutated Fc domain is as set forth in any one of SEQ ID NOS: 21-28. In some aspects, the RNase-Fc fusion protein is set forth in any one of SEQ ID NOs 44-58. In some aspects, the RNase-Fc fusion protein comprises a wild-type human RNasel domain linked to a mutant human IgGlFc domain comprising SCC, P238S, and P331S, or a mutant human IgGlFc domain comprising SSS, P238S, and P331S. In some embodiments, the RNase-Fc fusion protein is shown in SEQ ID NO 45-46. In some embodiments, the RNase-Fc fusion protein is shown in SEQ ID NO 50.

In some aspects, the RNase-Fc fusion protein comprises a wild-type human RNasel domain linked by a (Gly4Ser)4 linker domain to a mutant human IgGlFc domain comprising SCC, P238S, and P331S, or a mutant human IgGlFc domain comprising SSS, P238S, and P331S. In some aspects, the RNase-Fc fusion protein is shown in SEQ ID NO 47-48.

In some embodiments, the RNase-Fc fusion protein comprises a human DNase 1G 105R a114F domain linked by a (Gly4Ser)4 linker domain to a mutant human IgG1 Fc domain comprising SCC, P238S, and P331S linked by an NLG linker domain to a wild-type human RNase1 domain. In some embodiments, the RNase-Fc fusion protein comprises a human DNase 1G 105R a114F domain linked by a (Gly4Ser)4 linker domain to a mutant human IgG1 Fc domain comprising SSS, P238S, and P331S linked by an NLG linker domain to a wild-type human RNase1 domain. In some aspects, the RNase-Fc fusion protein is shown in SEQ ID NO 51-52.

In some aspects, the RNase-Fc fusion protein comprises a wild-type human RNase1 domain linked by a (Gly4Ser)4 linker domain to a mutant human IgG1 Fc domain comprising SCC, P238S, and P331S linked by an NLG linker domain to a human DNase 1G 105R a114F domain. In some embodiments, the RNase-Fc fusion protein comprises a wild-type human RNase1 domain linked by a (Gly4Ser)4 linker domain to a mutant human IgG1 Fc domain comprising SSS, P238S, and P331S linked by an NLG linker domain to a human DNase 1G 105R a114F domain. In some aspects, the RNase-Fc fusion protein is shown in SEQ ID NO 53-54.

In some aspects, the RNase-Fc fusion protein comprises a wild-type human RNase1 domain linked to a mutant human IgG1 Fc domain comprising SCC, P238S, and P331S linked to a human DNase 1G 105R a114F domain by an NLG linker domain. In some aspects, the RNase-Fc fusion protein comprises a wild-type human RNase1 domain linked to a mutant human IgG1 Fc domain comprising SSS, P238S, and P331S linked to a human DNase 1G 105R a114F domain by an NLG linker domain. In some aspects, the RNase-Fc fusion protein is shown in SEQ ID NO: 55-58.

In some aspects, the activity of the RNase-Fc fusion protein is detectable in vitro and/or in vivo.

In some embodiments, the RNase-Fc fusion protein comprises an RNase domain and an Fc domain, wherein the RNase1 domain is located on the COOH side of the Fc. At itIn other embodiments, the RNase-Fc fusion protein comprises an RNase domain and an Fc domain, wherein the RNase1 domain is located at the NH of the Fc2And (3) side. In some embodiments, the RNase-Fc fusion protein comprises: RNase-Fc; Fc-RNase; Fc-linker-RNase; RNase-linker-Fc, RNase-Fc-DNase; DNase-Fc-RNase; RNase-linker-Fc-linker-DNase; DNase-linker-Fc-linker-RNase; RNase-Fc-linker-DNase; DNase-Fc-linker-RNase; RNase-linker-Fc-DNase; DNase-linker-Fc-RNase.

In some embodiments, the fusion linkage between the enzyme domain and the other domain of the RNase-Fc fusion protein is optimized.

In some embodiments, the target of RNase enzymatic activity of the RNase-Fc fusion protein is predominantly extracellular, consisting of, for example, RNA contained in an immune complex with anti-RNP autoantibodies and RNA expressed on the surface of cells undergoing apoptosis. In some embodiments, the RNase-Fc fusion protein is active in the acidic environment of endocytic vesicles. In some embodiments, the RNase-Fc fusion protein includes a wild-type (wt) Fc domain to, for example, allow the molecule to bind FcR and enter the endocytic compartment through the entry route used by the immune complex. In some embodiments, an RNase-Fc fusion protein comprising an Fc domain or variant or fragment thereof is adapted to be active in both extracellular and endocytic environments (in which TLR7 may be expressed). In some aspects, this allows RNase-Fc fusion proteins including wild-type Fc domains or variants or fragments thereof to stop TLR7 signaling through previously phagocytosed immune complexes or by activating RNA of TLR7 following viral infection. In some embodiments, the wild-type RNase of the RNase-Fc fusion protein is not resistant to inhibition by RNase cytoplasmic inhibitors. In some embodiments, the wild-type RNase of the RNase-Fc fusion protein is not active in the cytoplasm of the cell.

In some embodiments, the RNase-Fc fusion protein comprises RNase. In some embodiments, the RNase-Fc fusion protein comprises DNase and RNase. In some embodiments, these RNase-Fc fusion proteins improve the treatment of xerosis as they digest or degrade immune complexes containing RNA, DNA, or a combination of RNA and DNA, and are extracellularly active. In some embodiments, the RNase-Fc fusion proteins of the present disclosure reduce fatigue in xerosis patients.

In some embodiments, the present disclosure provides nucleic acids encoding one or more RNase-Fc fusion proteins for use in gene therapy methods for treating or preventing disorders, diseases, and conditions. Gene therapy methods involve introducing RNase-Fc fusion protein nucleic acid (DNA, RNA and antisense DNA or RNA) sequences into an animal in need thereof to achieve expression of one or more polypeptides of the disclosure. The method may comprise introducing one or more polynucleotides encoding the RNase-Fc fusion proteins of the disclosure operably coupled to a promoter and any other genetic elements necessary for expression of the RNase-Fc fusion protein in the target tissue.

In gene therapy applications, the RNase-Fc fusion protein gene is introduced into cells to achieve in vivo synthesis of a therapeutically effective gene product. "Gene therapy" includes conventional gene therapy and administration of gene therapy agents to achieve a sustained effect by a single treatment, which involves the single or repeated administration of therapeutically effective DNA or mRNA. Oligonucleotides can be modified to enhance their uptake, for example, by substituting their negatively charged phosphodiester groups with uncharged groups.

Fc domains

In some embodiments, a polypeptide comprising one or more nuclease domains or variants or fragments thereof is operably coupled, with or without a linker domain, to an Fc domain that serves as a scaffold and means to increase the serum half-life of the polypeptide. In some embodiments, one or more nuclease domains and/or Fc domains are aglycosylated, deglycosylated, or under-glycosylated. In some embodiments, the Fc domain is a mutant or variant Fc domain, or a fragment of an Fc domain.

Suitable Fc domains are well known in the art and include, but are not limited to, Fc and Fc variants such as those disclosed in WO2011/053982, WO02/060955, WO02/096948, WO05/047327, WO05/018572, and US 2007/0111281 (the foregoing are incorporated herein by reference). It is within the ability of the skilled person to introduce an Fc domain into an RNase-Fc fusion protein disclosed herein (with or without altered glycosylation) using conventional methods (e.g. cloning, conjugation).

In some embodiments, the Fc domain is a wild-type human IgGl Fc, e.g., as shown in SEQ ID NO: 20. In some embodiments, the Fc domain is a human IgG1 Fc domain with one or more mutations.

In some embodiments, the Fc domain is wild-type human IgG4 Fc, as shown in SEQ ID NOS: 30-31. In some embodiments, the Fc domain is a human IgG4 Fc domain with one or more mutations.

In some embodiments, the Fc domain is altered or modified, for example by mutations that result in amino acid additions, deletions, or substitutions. As used herein, the term "Fc domain variant" refers to an Fc domain having at least one amino acid modification (such as one amino acid substitution) as compared to the wild-type Fc from which the Fc domain is derived. For example, when the Fc domain is derived from a human IgGl antibody, the variant comprises at least one amino acid mutation (e.g., substitution) as compared to the wild-type amino acid at a corresponding position in the human IgGl Fc region. The amino acid substitution of the Fc variant may be at a position within the Fc domain that corresponds to the position numbering given for that residue in the Fc region of the antibody (numbering according to the EU index).

In one embodiment, the Fc variant comprises one or more amino acid substitutions at one or more amino acid positions located in the hinge region or portion thereof. In another embodiment, the Fc variant comprises one or more amino acid substitutions at one or more amino acid positions located in the CH2 domain or portion thereof. In another embodiment, the Fc variant comprises one or more amino acid substitutions at one or more amino acid positions located in the CH3 domain or portion thereof. In another embodiment, the Fc variant comprises one or more amino acid substitutions at one or more amino acid positions located in the CH4 domain or portion thereof.

In some embodiments, the Fc domain comprises one or more of the following amino acid substitutions: T350V, L351Y, F405A and Y407V. In some embodiments, the Fc domain comprises one or more of the following amino acid substitutions: T350V, T366L, K392L and T394W.

In some embodiments, a human IgGl Fc domain has a mutation at N83 (i.e., N297 by Kabat numbering) resulting in an aglycosylated Fc domain (e.g., Fc N83S; SEQ ID NO: 21). In some embodiments, the human IgG1 Fc domain includes mutations in one or more of the three hinge region cysteines (residues 220, 226, and 229, numbered according to the EU index). In some embodiments, one or more of the three hinge cysteines in the Fc domain can be mutated to SCC (SEQ ID NO:24) or SSS (SEQ ID NO:25), wherein "S" represents an amino acid in which a cysteine is replaced with a serine amino acid (wherein CCC refers to the three cysteines present in the wild-type hinge domain). Thus, "SCC" indicates that only the first cysteine of the three hinge region cysteines (residues 220, 226, and 229, numbered according to the EU index) is replaced by serine, and "SSS" indicates that all three cysteines in the hinge region are replaced by serine (residues 220, 226, and 229, numbered according to the EU index).

In some aspects, the Fc domain is a human IgGl Fc domain having one or more mutations.

In some aspects, the mutant Fc domain comprises one or more mutations in the hinge, CH2, and/or CH3 domain.

In some aspects, the Fc domain is a human IgG4 Fc domain with one or more mutations. In some embodiments, the mutations in the IgG4 Fc domain comprise one or more mutations selected from the group of mutations consisting of: F296Y, E356K, R409K and H345R. In some embodiments, the mutations in the IgG4 Fc domain comprise one or more mutations selected from the group of mutations consisting of: F296Y, R409K and K439E. In some embodiments, the RNase-Fc fusion proteins disclosed herein comprise a first polypeptide comprising a mutant IgG4 Fc domain, wherein the Fc domain comprises mutations F296Y, E356K, R409K, and H345R, and a second polypeptide comprising a mutant IgG4 Fc domain, wherein the CH3 domain comprises mutations F296Y, R409K, and K439E. In some embodiments, the mutated IgG4 Fc domain comprises one or more mutations in the hinge, CH2, and/or CH3 domains.

Substitution of CH2

In some aspects, the mutant Fc domain comprises the P238S mutation. In some aspects, the mutant Fc domain comprises the P331S mutation. In some aspects, the mutant Fc domain comprises the P238S mutation and the P331S mutation. In some aspects, the mutant Fc domain comprises P238S and/or P331S, and may include mutations in one or more of the three hinge cysteines (residues 220, 226, and 229), according to EU index numbering. In some aspects, the mutant Fc domain comprises one or more mutations in P238S and/or P331S, and/or three hinge cysteines (residues 220, 226, and 229), according to EU index numbering. In some aspects, the mutant Fc domain comprises P238S and/or P331S, and/or the hinge cysteine is mutated to SCC or the three hinge cysteines are mutated to SSS. In some aspects, the mutant Fc domain comprises mutations in P238S and P331S and at least one of the three hinge cysteines. In some aspects, the mutant Fc domain comprises P238S and P331S, and SCC. In some aspects, the mutant Fc domain comprises P238S and P331S and SSS. In some aspects, the mutant Fc domain comprises P238S and SCC or SSS. In some aspects, the mutant Fc domain comprises P331S and SCC or SSS. (all numbering according to the EU index).

In some aspects, a mutant Fc domain includes a mutation at an N-linked glycosylation site, e.g., asparagine in place of another amino acid, e.g., N297S. In some aspects, a mutant Fc domain includes a mutation at an N-linked glycosylation site (e.g., N297), e.g., a mutation of asparagine for another amino acid (e.g., serine), e.g., N297S and one or more of the three hinge cysteines. In some aspects, a mutant Fc domain includes a mutation at an N-linked glycosylation site (e.g., N297), e.g., asparagine substituted for another amino acid (e.g., serine), e.g., N297S and one of the three hinge cysteines mutated to SCC or all three cysteines mutated to SSS. In some aspects, a mutant Fc domain includes a mutation at an N-linked glycosylation site (e.g., N297), e.g., asparagine in place of another amino acid (e.g., serine), e.g., N297 and one or more mutations at the CH2 domain, which reduces fcyr binding and/or complement activation, e.g., mutations at P238 or P331 or both, e.g., P238S or P331S, or both P238S and P331S. In some aspects, such mutant Fc domains may also include mutations in the hinge region, such as SCC or SSS. (all numbering according to the EU index.) in some aspects, the mutant Fc domains are as shown in the sequence listing or sequence listing herein.

Substitution of CH3

In some embodiments, the heterodimer is formed by a mutation in the CH3 domain of the Fc domain on the heterodimeric RNase-Fc fusion proteins disclosed herein. The heavy chain was first engineered for heterodimerization using the "knob-to-hole" strategy (Rigway B, et al, Protein Eng.,9(1996) pp.617-621, incorporated herein by reference). The term "knob" refers to a technique for pairing two polypeptides in vitro or in vivo by introducing a protuberance (knob) into one polypeptide and a cavity (knob) into the other polypeptide at their interface of interaction. See, e.g., WO 96/027011, WO98/050431, US 5,731,168, US2007/0178552, WO2009089004, US 20090182127. In particular, combinations of mutations in the CH3 domain can be used to form heterodimers, such as S354C, T366W in the "knob" heavy chain and Y349C, T366S, L368A, Y407V in the "hole" heavy chain. In another example, T366Y in the "pestle" heavy chain and Y407T in the "hole" heavy chain. In some embodiments, the heterodimeric RNase-Fc fusion proteins disclosed herein comprise a first CH3 domain with knob mutation T366W and a second CH3 domain with hole mutations T366S, L368A, and Y407V. (numbering according to the EU index.) in some embodiments, an RNase-Fc fusion protein disclosed herein comprises a first CH3 domain having a knob mutation T366Y and a second CH3 domain having a hole mutation Y407T. In some embodiments, the CH3 mutations are those described in US2012/0149876a1, US2017/0158779, US9574010, and US9562109, each of which is incorporated herein by reference; and Von Kreudenstein, t.s.et al.mabs,5(2013), pp.646-654, incorporated herein by reference) and including the following mutations: T350V, L351Y, F405A and Y407V (first CH3 domain); and T350V, T366L, K392L, T394W (second CH3 domain). In some embodiments, the heterodimeric RNase-Fc fusion proteins disclosed herein comprise a first CH3 domain having T350V, L351Y, F405A, and Y407V mutations and a second CH3 domain having T350V, T366L, K392L, T394W mutations. (numbering according to the EU index.)

In some embodiments, the heterodimer is formed by a mutation in the CH3 domain of the Fc domain on an RNase-Fc fusion protein disclosed herein. In particular, combinations of mutations in the CH3 domain can be used to form heterodimers with high heterodimer stability and purity; see, for example, Von Kreudenstein et al, mAbs 5:5, 646-654; months 9 to 10 in 2013, and US2012/0149876a1, US2017/0158779, US9574010, and US9562109, each of which is incorporated herein by reference in its entirety. In some embodiments, the mutations in the Fc domain comprise one or more mutations selected from the group consisting of: T350V, L351Y, F405A and Y407V. In some embodiments, the mutations in the Fc domain comprise one or more mutations selected from the group consisting of: T350V, T366L, K392L and T394W. In some embodiments, the RNase-Fc fusion proteins disclosed herein comprise a CH3 domain with mutations T350V, L351Y, F405A, and Y407V. In some embodiments, the RNase-Fc fusion proteins disclosed herein comprise a CH3 domain with mutations T350V, T366L, K392L, and T394W. In some embodiments, the RNase-Fc fusion proteins disclosed herein comprise a first polypeptide comprising a mutant Fc domain, wherein the CH3 domain comprises mutations T350V, L351Y, F405A, and Y407V, and a second polypeptide comprising a mutant Fc domain, wherein the CH3 domain comprises mutations T350V, T366L, K394 392L, and T394W.

Other mutations in the CH3 domain of the Fc domain are considered to preferentially form heterodimers. See, for example, Von Kreudenstein et al, mAbs 5:5, 646-654; month 9 to month 10 2013, incorporated herein by reference). In some embodiments, the mutation in the Fc domain of the first polypeptide comprises one or more mutations selected from the group consisting of: the mutations in the Fc domain of T350V, L351Y, F405A and Y407V, and the second polypeptide include one or more mutations selected from the following group of mutations: T350V, T366L, K392M and T394W. In some embodiments, the mutation in the Fc domain of the first polypeptide comprises one or more mutations selected from the group consisting of: L351Y, F405A and Y407V, the mutation in the Fc domain of the second polypeptide comprising one or more mutations selected from the following group of mutations: T366L, K392M and T394W.

In some embodiments, the CH3 mutations are those described by Moore, g.l.et al (mABs,3(2011), pp.546-557) and include the following mutations: S364H and F405A (first CH3 domain); and Y349T and T394F (second CH3 domain). In some embodiments, the heterodimeric RNase-Fc fusion proteins disclosed herein comprise a first CH3 domain having the S364H and F405A mutations and a second CH3 domain having the Y349T and T394F mutations. (numbering according to the EU index.)

In some embodiments, the CH3 mutations are those described by Gunasekaran, k.et al. (j.biol.chem.,285(2010), pp.19637-19646) and include the following mutations: K409D and K392D (first CH3 domain); and D399K and E365K (second CH3 domain). In some embodiments, the heterodimeric RNase-Fc fusion proteins disclosed herein comprise a first CH3 domain having K409D and K392D mutations and a second CH3 domain having D399K and E365K mutations. (numbering according to the EU index.)

The RNase-Fc fusion proteins of the present disclosure may employ art-recognized Fc variants known to confer alterations in effector function and/or FcR binding. For example, changes (e.g., substitutions) in one or more amino acid positions are disclosed in International PCT publications WO88/07089A1, WO96/14339A1, WO98/05787A1, WO98/23289A1, WO99/51642A1, WO99/58572A1, WO00/09560A2, WO00/32767A1, WO00/42072A2, WO02/44215A2, WO02/060919A2, WO03/074569A2, WO 04/04A 04, WO04/029207A 04, WO 04/04A 04/04A 04, WO 04A 04/04A 04, WO 04/04A 3636363672A 04A 363672A 04A; U.S. patent publication nos. US2007/0231329, US2007/0231329, US2007/0237765, US2007/0237766, US2007/0237767, US2007/0243188, US20070248603, US20070286859, US 20080057056; or U.S. Pat. nos. 5,648,260; 5,739,277; 5,834,250; 5,869,046; 6,096,871, respectively; 6,121,022; 6,194,551; 6,242,195, respectively; 6,277,375; 6,528,624, respectively; 6,538,124, respectively; 6,737,056; 6,821,505, respectively; 6,998,253, respectively; 7,083,784, respectively; and 7,317,091, each of which is incorporated herein by reference. In one embodiment, specific changes may be made at one or more of the disclosed amino acid positions (e.g., specific substitutions of one or more amino acids as disclosed in the art). In another embodiment, different alterations (e.g., different substitutions of one or more amino acid positions disclosed in the art) can be made at one or more of the disclosed amino acid positions.

Other amino acid mutations in the Fc domain are contemplated to reduce binding to Fc γ receptors and Fc γ receptor subtypes. Amino acid residue numbering is assigned to the Fc domain according to the Kabat definition. See, for example, Sequences of Proteins of Immunological Interest (Table of contexts, Introduction and Constant Region Sequences), 5th edition, Bethesda, MD: NIH vol.1:647-723 (1991); kabat et al, "Introduction" Sequences of Proteins of Immunological Interest, US Dept of Health and Human Services, NIH,5th edition, Bethesda, MD vol.1: xiii-xcvi (1991); chothia & Lesk, J.mol.biol.196:901-917 (1987); chothia et al, Nature 342:878-883(1989), each of which is incorporated herein by reference for all purposes. "

For example, mutations at positions 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 279, 280, 283, 285, 298, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 312, 315, 322, 324, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 356, 360, 373, 376, 378, 379, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 382, 438, or 439 of the Fc region may be combined with alterations as described in U.S. patent No. 6,737,056 issued 5, 18, 2004, and the entire contents of which are incorporated herein by reference. This patent reports that changing Pro331 to Ser in IgG3 results in a six-fold decrease in affinity compared to unmutated IgG3, indicating that Pro331 is involved in Fc γ RI binding. In addition, amino acid modifications at positions 234, 235, 236 and 237, 297, 318, 320 and 322 are disclosed in U.S. Pat. No. 5,624,821, published 4/29 1997, which is incorporated herein by reference in its entirety, as potentially altering receptor binding affinity. (numbering according to the EU index.)

Other mutations contemplated for use include, for example, those described in U.S. patent application publication No. 2006/0235208, which is disclosed at 19/10.2006, the entire contents of which are incorporated herein by reference. This disclosure describes Fc variants with reduced binding to Fc γ receptors, reduced antibody-dependent cell-mediated cytotoxicity, or reduced complement-dependent cytotoxicity comprising at least one amino acid modification in the Fc region, including 232G, 234H, 235D, 235G, 235H, 236I, 236N, 236P, 236R, 237K, 237L, 237N, 237P, 238K, 239R, 265G, 267R, 269R, 270H, 297S, 299A, 299I, 299V, 325A, 325L, 327R, 328R, 329K, 330I, 330L, 330N, 330P, 330R, and 331L (numbering according to the EU index), and double mutants 236R/237K, 236R/325L, 236R/328R, 237K/325L, 237K/328R, 325L/328R, 235G/236R, 267R/269R, 234G/235G, 236R/237K/325L, 236R/325L/328R, 235G/236R/237K, and 237K/325L/328R. Other mutations described in this disclosure for contemplated use include 227G, 234D, 234E, 234G, 234I, 234Y, 235D, 235I, 235S, 236S, 239D, 246H, 255Y, 258H, 260H, 2641, 267D, 267E, 268D, 268E, 272H, 272I, 272R, 281D, 282G, 283H, 284E, 293R, 295E, 304T, 324G, 324I, 327D, 327A, 328D, 328E, 328F, 328I, 328M, 328N, 328Q, 328T, 328V, 328Y, 330I, 330L, 330Y, 332D, 332E, 335D, insertion G between positions 235 and 236, insertion a between positions 235 and 236, insertion S between positions 235 and 236, insertion T between positions 235 and 236, insertion N between positions 235 and 236, insertion D between positions 235 and 236, insertion V between positions 235 and 236, insertion between positions 236 and 236V, insertion between positions 235 and 236L, insertion of position V, insertion of position between positions 235 and 236, G between positions 235 and 236, a between positions 235 and 236, S between positions 235 and 236, T between positions 235 and 236, N between positions 235 and 236, D between positions 235 and 236, V between positions 235 and 236, L between positions 235 and 236, G between positions 297 and 298, a between positions 297 and 298, D between positions 297 and 298, G between positions 326 and 327, a between positions 326 and 327, T between positions 326 and 327, D between positions 326 and 327, and E (numbering according to the EU index) between positions 326 and 327. Furthermore, the mutations described in U.S. patent application publication No. 2006/0235208 include 227G/332E, 234D/332E, 234E/332E, 234Y/332E, 234I/332E, 234G/332E, 235I/332E, 235S/332E, 235D/332E, 235E/332E, 236S/332E, 236A/332E, 236S/332D, 236A/332D, 239D/268E, 246H/332E, 255Y/332E, 258H/332E, 260H/332E, 264I/332E, 267E/332E, 267D/332E, 268D/332D, 268E/332E, 268D/332E, 268E/330Y, 268D/330Y, 272R/332E, 272H/332E, 268E/332E, 268E/332E, 272R/330Y, 272R/332E, 272H/332E, and, 283H/332E, 284E/332E, 293R/332E, 295E/332E, 304T/332E, 324I/332E, 324G/332E, 324I/332D, 324G/332D, 327D/332E, 328A/332E, 328T/332E, 328V/332E, 328I/332E, 328F/332E, 328Y/332E, 328M/332E, 328D/332E, 328E/332E, 328N/332E, 328Q/332E, 328A/332D, 328T/332D, 328V/332D, 328I/332D, 328F/332D, 328Y/332D, 328M/332D, 328D/332D, 328E/332D, 328N/332D, Q/332D, 330L/332E, 328E/332D, 330Y/332E, 330I/332E, 332D/330Y, 335D/332E, 239D/332E/330Y, 239D/332E/330L, 239D/332E/330I, 239D/332E/268E, 239D/332E/268D, 239D/332E/327D, 239D/332E/284E, 239D/268E/330Y, 239D/332E/327D/332E/268E/A, 239D/332E/330Y/327A, 332E/330Y/268E/327A, 239D/332E/268E/330E/327A, inserts G > 297-298/332E/327, Insert A >297-298/332E, insert S >297-298/332E, insert D >297-298/332E, insert G >326-327/332E, insert A >326-327/332E, insert T >326-327/332E, insert D >326-327/332E, insert E >326-327/332E, insert G >235-236/332E, insert A >235-236/332E, insert S >235-236/332E, insert T >235-236/332E, insert N >235-236/332E, insert D >235-236/332E, insert V >235-236/332E, insert L >235-236/332E, insert G >235-236/332D, insert A >235-236/332D, Insertions S >235-236/332D, insertions T >235-236/332D, insertions N >235-236/332D, insertions D >235-236/332D, insertions V >235-236/332D, and insertions L >235-236/332D (numbering according to the EU index) are contemplated for use. Mutant L234A/L235A is described, for example, in U.S. patent application publication No. 2003/0108548, which is published at 12/6/2003 and is incorporated herein by reference in its entirety. In embodiments, the modifications described are included individually or in combination. (numbering according to the EU index.)

PK moiety

In some embodiments, the RNase is operably coupled to a PK moiety, which acts as a scaffold and means to increase the serum half-life of the RNase.

Suitable PK moieties are well known in the art and include, but are not limited to, albumin, transferrin, Fc and variants thereof, and polyethylene glycol (PEG) and derivatives thereof. Suitable PK moieties include, but are not limited to, HSA or variants or fragments thereof, such as those disclosed in US5,876,969, WO2011/124718 and WO 2011/0514789; fc and Fc variants, such as those disclosed in WO2011/053982, WO02/060955, WO02/096948, WO05/047327, WO05/018572, and US 2007/0111281; transferrin or a variant or fragment thereof, as disclosed in US7,176,278 and US8,158,579; and PEG or derivatives such as those disclosed in Zalipsky et al ("Use of Functionalized Poly (Ethylene Glycols) for Modification of Polypeptides" in Polyethylene Glycol Chemistry: Biotechnical and biological Applications, J.M.Harris, plus Press, New York (1992)), and Zalipsky et al advanced Drug Reviews 1995:16:157 and U.S. Pat. Nos. 4,640,835, 4,496,689, 4,301,144, 4,670,417, 4,791,192, 4,179,337, and 5,932,462 (the foregoing are incorporated herein by reference). It is within the ability of the skilled person to operably couple a PK moiety (e.g. cloning, conjugation) to an RNase of the invention using conventional methods.

In some embodiments, the PK moiety is natural aglycosylated HSA.

In some embodiments, the PK moiety is wild-type Fc (SEQ ID NO: 20).

In certain embodiments, the Fc domain is altered or modified, for example by amino acid mutation (e.g., addition, deletion, or substitution). As used herein, the term "Fc domain variant" refers to an Fc domain having at least one amino acid modification (e.g., amino acid substitution) as compared to the wild-type Fc from which the Fc domain is derived. For example, wherein the Fc domain is derived from a human IgGl antibody, the variant comprises at least one amino acid mutation (e.g., substitution) as compared to a wild-type amino acid at a corresponding position in a human IgGl Fc region. For example, wherein the Fc domain is derived from a human IgG4 antibody, the variant comprises at least one amino acid mutation (e.g., substitution) as compared to the wild-type amino acid at a corresponding position in the Fc region of human IgG 4.

In some embodiments, the PK moiety is any Fc variant described herein.

In some embodiments, the PK moiety is wild type HST. In other embodiments, the PK moiety is an HST with mutations at N413 and/or N611 and/or S12 (S12 is a potential O-linked glycosylation site), resulting in an HST with altered glycosylation (i.e., HST N413S, HST N611S, HST N413S/N611S, and HST S12A/N413S/N611S).

Joint domain

In some embodiments, the RNase-Fc fusion protein includes a linker domain. In some embodiments, the RNase-Fc fusion protein comprises a plurality of linker domains. In some embodiments, the linker domain is a polypeptide linker. In certain aspects, it is desirable to fuse an Fc or variant or fragment thereof with one or more nuclease domains using a polypeptide linker to form an RNase-Fc fusion protein.

In one embodiment, the polypeptide linker is synthetic. As used herein, the term "synthetic" with respect to a polypeptide linker includes a peptide (or polypeptide) comprising an amino acid sequence (which may or may not naturally occur) that is linked in a linear amino acid sequence to a sequence (which may or may not naturally occur) to which it is not naturally linked in nature (e.g., an Fc sequence). For example, a polypeptide linker may comprise a non-naturally occurring polypeptide that is a modified form of a naturally occurring polypeptide (e.g., comprising a mutation, such as an addition, substitution, or deletion) or comprises a first amino acid sequence (which may or may not be naturally occurring). Polypeptide linkers of the invention may be used, for example, to ensure juxtaposition of Fc or variants or fragments thereof to ensure proper folding and formation of functional Fc or variants or fragments thereof. Preferably, a polypeptide linker compatible with the present invention will be relatively non-immunogenic and will not inhibit any non-covalent binding between the monomeric subunits of the binding protein.

In certain embodiments, the RNase-Fc fusion protein employs an NLG linker as set forth in SEQ ID NO: 37.

In certain embodiments, the RNase-Fc fusion proteins of the present disclosure employ polypeptide linkers to join any two or more in-frame (in frame) domains into a single polypeptide chain. In one embodiment, the two or more domains may be independently selected from any of the Fc domains, or variants or fragments thereof, or nuclease domains discussed herein. In some embodiments, the RNase domain of the RNase-Fc fusion protein is operably coupled to the Fc domain by a linker domain. In some embodiments, polypeptide linkers can be used to fuse the same Fc fragments, thereby forming a homodimeric Fc region. In other embodiments, polypeptide linkers can be used to fuse different Fc fragments, thereby forming a heterodimeric Fc region. In other embodiments, the polypeptide linker of the invention can be used to genetically fuse the C-terminus of a first Fc fragment to the N-terminus of a second Fc fragment to form a complete Fc domain.

In one embodiment, the polypeptide linker comprises a portion of an Fc domain, or a variant or fragment thereof. For example, in one embodiment, the polypeptide linker may comprise different portions of an Fc fragment (e.g., a C or N domain), or an Fc domain or variant thereof.

In another embodiment, the polypeptide linker comprises or consists of a gly-ser linker. As used herein, the term "gly-ser linker" refers to a peptide consisting of glycine and serine residues. Exemplary Gly/ser linkers comprise the formula (Gly)4Ser) n, wherein n is a positive integer (e.g. 1, 2, 3, 4 or 5). A preferred Gly/ser linker is (Gly)4Ser) 4. Another preferred Gly/ser linker is (Gly)4Ser) 3. Another preferred Gly/ser linker is (Gly)4Ser) 5. In certain embodiments, the gly-ser linker can be inserted between two other sequences of the polypeptide linker (e.g., any of the polypeptide linker sequences described herein). In other embodiments, the gly-ser linker is attached at one or both ends of another sequence of a polypeptide linker (e.g., any of the polypeptide linker sequences described herein). In other embodiments, two or more gly-ser linkers are incorporated in tandem (in series) into the polypeptide linker.

In other embodiments, the polypeptide linker of the invention comprises a biologically relevant peptide sequence or a sequence portion thereof. For example, biologically relevant peptide sequences may include, but are not limited to, sequences derived from anti-rejection (rejection) or anti-inflammatory peptides. The anti-rejection or anti-inflammatory peptide may be selected from the group consisting of cytokine inhibitory peptides, cell adhesion inhibitory peptides, thrombin inhibitory peptides and platelet inhibitory peptides. In a preferred embodiment, the polypeptide linker comprises a peptide sequence selected from the group consisting of an IL-1 inhibitory or antagonistic peptide sequence, an Erythropoietin (EPO) -mimetic peptide sequence, a Thrombopoietin (TPO) -mimetic peptide sequence, a G-CSF mimetic peptide sequence, a TNF antagonist peptide sequence, an integrin binding peptide sequence, a selectin antagonist peptide sequence, an antipathogenic peptide sequence, a Vasoactive Intestinal Peptide (VIP) mimetic peptide sequence, a calmodulin antagonist peptide sequence, a mast cell antagonist, an SH3 antagonist peptide sequence, a urokinase receptor (UKR) antagonist peptide sequence, a somatostatin or cortistatin mimetic peptide sequence, and a macrophage and/or T cell inhibitory peptide sequence. Exemplary peptide sequences are disclosed in U.S. patent No. 6,660,843, which is incorporated herein by reference, any of which can be used as a polypeptide linker.

Other linkers suitable for RNase-Fc fusion proteins are known in the art, e.g., serine-rich linkers disclosed in US5,525,491, Arai et al, Protein Eng 2001; helix-forming peptide linkers disclosed in 529-32 (e.g., a (eaaak) nA (n-2-5)), and Chen et al, Mol Pharm 2011; 8:457-65, a dipeptide linker LE, a thrombin sensitive dithiocyclic peptide linker, and an alpha-helix forming linker LEA (EAAAK)4ALEA(EAAAK)4ALE(SEQ ID NO:39)。

Other exemplary linkers include a GS linker (i.e., (GS) n), a GGSG (SEQ ID NO:40) linker (i.e., (GGSG) n), a GSAT linker (SEQ ID NO:41), an SEG linker, and a GGS linker (i.e., (GGSGGS) n), where n is a positive integer (e.g., 1, 2, 3, 4, or 5). Other linkers suitable for RNase-Fc fusion proteins can be found using publicly available databases, such as linker databases (ibi. vu. nl/programs/linkerdbwww). Linker databases are databases of inter-domain linkers in multifunctional enzymes, which serve as potential linkers in novel fusion proteins (see, e.g., George et al, Protein Engineering 2002; 15: 871-9).

It will be appreciated that variant forms of these exemplary polypeptide linkers may be produced by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence encoding the polypeptide linker, thereby introducing one or more amino acid substitutions, additions or deletions into the polypeptide linker. Mutations can be introduced by standard techniques such as site-directed mutagenesis and PCR-mediated mutagenesis.

The polypeptide linkers of the present disclosure are at least one amino acid in length and can be of varying lengths. In one embodiment, the polypeptide linker of the invention is about 1 to about 50 amino acids in length. The term "about" as used in this context means +/-two amino acid residues. Since the linker length must be a positive integer, a length of about 1 to about 50 amino acids refers to a length of 1 to 48-52 amino acids. In another embodiment, a polypeptide linker of the present disclosure is about 10-20 amino acids in length. In another embodiment, a polypeptide linker of the present disclosure is about 15 to about 50 amino acids in length.

In another embodiment, a polypeptide linker of the present disclosure is about 20 to about 45 amino acids in length. In another embodiment, a polypeptide linker of the present disclosure is about 15 to about 25 amino acids in length. In another embodiment, the polypeptide linker of the disclosure is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, or 61 or more amino acids in length.

Polypeptide linkers can be introduced into the polypeptide sequence using techniques known in the art. Modification can be confirmed by DNA sequence analysis. Plasmid DNA may be used to transform a host cell for stable production of the produced polypeptide.

Fusion proteins with altered glycosylation comprising an RNase nuclease

Glycosylation (e.g., O-linked or N-linked glycosylation) can affect the serum half-life of RNase-containing nuclease fusion proteins of the present disclosure, for example, by minimizing clearance of mannose and asialoglycoprotein receptors and other lectin-like receptors from the circulation. Thus, in some embodiments, RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc proteins, are prepared in an aglycosylated, deglycosylated, or under-glycosylated form. Preferably, the N-linked glycosylation is altered and the RNase-Fc fusion protein is aglycosylated.

In some embodiments, all asparagine residues in the RNase-Fc fusion protein that correspond to the Asn-X-Ser/Thr (X can be any other naturally occurring amino acid other than Pro) consensus are mutated to residues that do not act as N-linked glycosylation receptors (e.g., serine, glutamine) to eliminate glycosylation of the RNase-Fc fusion protein when synthesized in cells that glycosylate proteins

In some embodiments, the RNase-Fc fusion protein lacking N-linked glycosylation sites is produced in mammalian cells. In one embodiment, the mammalian cell is a CHO cell. Thus, in one embodiment, the aglycosylated RNase-Fc fusion protein is produced in CHO cells.

In other embodiments, the reduction or absence of N-glycosylation is achieved by producing an RNase-Fc fusion protein in, for example, a host (e.g., a bacterium such as E.coli), a mammalian cell engineered to lack one or more enzymes important for glycosylation, or a mammalian cell treated with an agent that prevents glycosylation, such as tunicamycin (an inhibitor of Dol-PP-GlcNAc formation).

In some embodiments, the RNase-Fc fusion protein is produced in lower eukaryotes engineered to produce glycoproteins with complex N-glycans, rather than high mannose type sugars (see, e.g., US 2007/0105127).

In some embodiments, glycosylated RNase-Fc fusion proteins (e.g., those produced in mammalian cells such as CHO cells) are chemically or enzymatically treated to remove one or more carbohydrate residues (e.g., one or more mannose, fucose, and/or N-acetylglucosamine residues) or to modify or mask one or more carbohydrate residues. Such modifications or masking may reduce the binding of the RNase-Fc fusion protein to mannose receptors, and/or asialoglycoprotein receptors, and/or other lectin-like receptors. Chemical deglycosylation can be achieved by treating the RNase-Fc fusion protein with trifluoromethanesulfonic acid (TFMS), as described, for example, in Sojar et al, JBC 1989; 264:2552-9and Sojar et al, Methods Enzymol 1987; 138:341-50 or by treatment with hydrogen fluoride, as disclosed in Sojar et al (1987, supra). Enzymatic removal of N-linked carbohydrates from RNase-Fc fusion proteins can be achieved by treating the RNase-Fc fusion protein with protein N-glycosidases (PNGase) A or F, as disclosed in Thotakura et al (Methods Enzymol 1987; 138: 350-9). Other art-recognized commercially available deglycosylases suitable for use include endo- α -N-acetyl-galactosaminidase, endoglycosidase F1, endoglycosidase F2, endoglycosidase F3, and endoglycosidase H. In some embodiments, one or more of these enzymes can be used to deglycosylate the RNase-Fc fusion proteins of the present disclosure. An alternative method of deglycosylation is disclosed in, for example, US 8,198,063.

In some embodiments, the RNase-Fc fusion protein is partially deglycosylated. Partial deglycosylation can be achieved by treating the RNase-Fc fusion protein with an endoglycosidase (e.g., endoglycosidase H) that cleaves N-linked high mannose carbohydrates, but does not cleave complex types of carbohydrates, leaving a GlcNAc residue attached to asparagine. RNase-Fc fusion protein treated with endoglycosidase H will lack high mannose carbohydrates, resulting in reduced interaction with hepatic mannose receptors. Although this receptor recognizes terminal GlcNAc, the probability of effective interaction with a single GlcNAc on the protein surface is not as good as an intact high mannose structure.

In other embodiments, the glycosylation of the RNase-Fc fusion protein is modified, e.g., by oxidation, reduction, dehydration, displacement, esterification, alkylation, sialylation, carbon-carbon bond cleavage, etc., to reduce the clearance of the RNase-Fc fusion protein from the blood. In some embodiments, the RNase-Fc fusion protein is treated with periodate and sodium borohydride to modify the carbohydrate structure. Periodate treatment oxidizes vicinal diols, cleaves carbon-carbon bonds and replaces hydroxyl groups with aldehyde groups; borohydride reduces aldehydes to hydroxyl groups. Many sugar residues include vicinal diols and are therefore cleaved by this treatment. Sequential treatment of the lysosomal enzyme β -glucuronidase with these drugs can demonstrate an extended serum half-life using periodate and sodium borohydride (see, e.g., Houba et al (1996) bioconjugate Chem 1996:7: 606-11; Stahl et al PNAS 1976; 73: 4045-9; Achord et al Pediat. Res 1977; 11: 816-22; Achord et al cell 1978; 15: 15-78). One method of treatment with periodate and sodium borohydride is described in Hickman et al, BBRC 1974; 57: 55-61. One method of treatment with periodate and cyanoborohydride increases serum half-life and tissue distribution of ricin, which is described in Thorpe et al eur J Biochem 1985; 147: 197-.

In one embodiment, the carbohydrate structure of the RNase-Fc fusion protein may be masked by the addition of one or more additional interfering mannose or asialoglycoprotein receptor or other lectin-like receptor recognition moieties.

In some embodiments, one or more potential glycosylation sites are removed by mutating the nucleic acid encoding the RNase-Fc fusion protein, thereby reducing glycosylation (lack of glycosylation) of the RNase-Fc fusion protein when synthesized in a cell (e.g., a mammalian cell such as a CHO cell) that glycosylates the protein. In some embodiments, it may be desirable to selectively glycosylate an under-glycosylated RNase-Fc fusion protein by mutating potential N-linked glycosylation sites therein if, for example, the under-glycosylated RNase-Fc fusion protein exhibits increased activity or contributes to increased serum half-life. In other embodiments, if, for example, such a modification improves the serum half-life of the RNase-Fc fusion protein, it may be desirable to have insufficient glycosylation of portions of the RNase-Fc fusion protein such that certain domains lack N-glycosylation. Alternatively, other amino acids in the vicinity of the glycosylation receptor can be modified to disrupt the recognition motif of the glycosylase without having to change the amino acid that is normally glycosylated.

In some embodiments, the glycosylation of the RNase-Fc fusion protein can be altered by introducing glycosylation sites. For example, the amino acid sequence of an RNase-Fc fusion protein can be modified to introduce a consensus sequence for N-linked glycosylation of Asn-X-Ser/Thr (X is any amino acid except proline). Additional N-linked glycosylation sites can be added anywhere in the amino acid sequence of the entire RNase-Fc fusion protein. Preferably, the glycosylation site is introduced into the amino acid sequence at a position that does not substantially reduce the RNase-Fc fusion protein activity.

The addition of O-linked glycosylation sites has been reported to alter the serum half-life of proteins such as growth hormone, follicle stimulating hormone, IGFBP-6, factor IX and many other proteins (e.g., as disclosed in Okada et al, Endocr Rev 2011; 32: 2-342; Weenen et al, J Clin Endocrinol Metab 2004; 89: 5204-12; Marinaro et al, European Journal of Endocrinology 2000; 142: 512-6; US 2011/0154516). Thus, in some embodiments, the O-linked glycosylation (on serine/threonine residues) of the RNase-Fc fusion protein is altered. Methods for altering O-linked glycosylation are routine in the art and can be accomplished, for example, by β -elimination (see, e.g., Huang et al, Rapid Communications in Mass Spectrometry 2002; 16: 1199-; by using a commercially available kit (e.g., GlycoProfile) TMBeta-Elimination Kit, Sigma); or by using a series of exoglycosidases (such as but not limited to beta)1-4 galactosidase and β -N-acetylglucosaminidase) until only Gal β 1-3GalNAc and/or GlcNAc β 1-3GalNAc remain, and then treated with, for example, endo- α -N-acetylgalactosaminidase (i.e., O-glycosidase). Such enzymes are commercially available from, for example, New England Biolabs. In other embodiments, the RNase-Fc fusion protein is altered to introduce O-linked glycosylation in the RNase-Fc fusion protein, as described, for example, in Okada et al (supra), Weenen et al (supra), US 2008/0274958; and US 2011/0171218. In some embodiments, one or more O-linked glycosylation consensus sites are introduced into an RNase-Fc fusion protein, such as CXXGGT/S-C (SEQ ID NO:59) (van den Steen et al, In Critical Reviews In Biochemistry and Molecular Biology, Michael Cox, ed., 1998; 33:151-208), NST-E/D-A (SEQ ID NO:60), NITQS (SEQ ID NO:61), QSTQSTQS (SEQ ID NO:62), D/E-FT-R/K-V (SEQ ID NO:63), C-E/D-SN (SEQ ID NO:64), and GGSC-K/R (SEQ ID NO: 65). Additional O-linked glycosylation sites can be added anywhere in the amino acid sequence of the entire RNase-Fc fusion protein. Preferably, the glycosylation site is introduced into the amino acid sequence at a position that does not substantially reduce the RNase-Fc fusion protein activity. Alternatively, as described, for example, in WO 87/05330 and Aplin et al, CRC Crit Rev Biochem 1981; 259-306), O-linked sugar moieties were introduced by chemical modification of amino acids in the RNase-Fc fusion protein.

In some embodiments, both N-linked and O-linked glycosylation sites are introduced into the RNase-Fc fusion protein, preferably at positions in the amino acid sequence that do not substantially reduce the RNase-Fc fusion protein activity.

It is well within the ability of the skilled person to introduce, reduce or eliminate glycosylation (e.g.N-linked or O-linked glycosylation) in an RNase-Fc fusion protein and to determine whether modification of such glycosylation state increases or decreases the activity or serum half-life of the RNase-Fc fusion protein using routine methods in the art.

In some embodiments, the RNase-Fc fusion protein may comprise an altered glycoform (e.g., a glycan that is hypofucosylated or does not contain fucose).

In some embodiments, the serum half-life of an RNase-Fc fusion protein having altered glycosylation is increased at least about 1.5 fold, such as at least 3 fold, at least 5 fold, at least 10 fold, at least about 20 fold, at least about 50 fold, at least about 100 fold, at least about 200 fold, at least about 300 fold, at least about 400 fold, at least about 500 fold, at least about 600 fold, at least about 700 fold, at least about 800 fold, at least about 900 fold, at least about 1000 fold, or 1000 fold or more, relative to a corresponding glycosylated RNase-Fc fusion protein (e.g., an RNase-Fc fusion protein in which the potential N-linked glycosylation site is not mutated). The serum half-life of the RNase-Fc fusion protein with altered glycosylation state can be determined using conventional methods recognized in the art.

In some embodiments, an RNase-Fc fusion protein having altered glycosylation (e.g., an RNase-Fc fusion protein that is not glycosylated, deglycosylated, or under-glycosylated) retains at least 50% of the activity of the corresponding glycosylated RNase-Fc fusion protein (e.g., an RNase-Fc fusion protein in which potential N-linked glycosylation sites are not mutated), such as at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 99.5%, or 100%

In some embodiments, altering the glycosylation state of the RNase-Fc fusion protein can increase activity by directly increasing activity or by increasing bioavailability (e.g., serum half-life). Thus, in some embodiments, the activity of an RNase-Fc fusion protein having altered glycosylation is increased at least 1.3 fold, such as at least 1.5 fold, at least 2 fold, at least 2.5 fold, at least 3 fold, at least 3.5 fold, at least 4 fold, at least 4.5 fold, at least 5 fold, at least 5.5 fold, at least 6 fold, at least 6.5 fold, at least 7 fold, at least 7.5 fold, at least 8 fold, at least 8.5 fold, at least 9 fold, at least 9.5 fold, or 10 fold or more, relative to a corresponding glycosylated RNase-Fc fusion protein (e.g., an RNase-Fc fusion protein in which the potential N-linked glycosylation site is not mutated).

The glycosylation state of the RNase-Fc fusion protein can be easily determined by the skilled person using art-recognized methods. In a preferred embodiment, the glycosylation state is determined using mass spectrometry. In other embodiments, the interaction with concanavalin a (con a) can be evaluated to determine whether the RNase-Fc fusion protein is under-glycosylated. An under-glycosylated RNase-Fc fusion protein is expected to have reduced binding to Con A-Sepharose compared to the corresponding glycosylated RNase-Fc fusion protein. SDS-PAGE analysis can also be used to compare the mobility of under-glycosylated proteins and the corresponding glycosylated proteins. Proteins that are not glycosylated enough are expected to have greater mobility in SDS-PAGE compared to glycosylated proteins. Other suitable art-recognized methods for analyzing the glycosylation state of proteins are described, for example, in Roth et al, International Journal of Carbohydrate Chemistry 2012; 1-10.

Pharmacokinetics (e.g., serum half-life) of RNase-Fc fusion proteins with different glycosylation states can be determined using conventional methods, e.g., by introducing the RNase-Fc fusion protein into a mouse, e.g., intravenously, taking blood samples at predetermined time points, and determining and comparing the level and/or activity of the RNase-Fc fusion protein in the samples.

Method for preparing RNase-Fc fusion protein

RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, of the present disclosure are prepared in transformed or transfected host cells using recombinant DNA techniques. To this end, recombinant DNA molecules encoding RNase-Fc fusion proteins were prepared. Methods for preparing such DNA molecules are well known in the art. For example, the sequence encoding the RNase-Fc fusion protein can be cleaved from the DNA using a suitable restriction enzyme. Alternatively, chemical synthesis techniques (e.g., the phosphoramidate method) can be used to synthesize the DNA molecule. Further, a combination of these techniques may be used.

The present invention also includes vectors capable of expressing the RNase-Fc fusion protein in a suitable host. The vector comprises a DNA molecule encoding an RNase-Fc fusion protein operably coupled with appropriate expression control sequences. Methods of effecting such operative ligation either before or after insertion of the DNA molecule into the vector are well known. Expression control sequences include promoters, activators, enhancers, operators, ribosomal nuclease domains, initiation signals, termination signals, cap signals, polyadenylation signals, and other signals involved in transcriptional or translational control.

The resulting vector with the DNA molecule thereon is used to transform or transfect a suitable host. In some embodiments, the RNase-Fc fusion proteins of the present disclosure can be prepared by co-transfecting or co-transforming two or more expression vectors comprising DNA encoding the RNase-Fc fusion proteins into a suitable host. Such transformation or transfection may be performed using methods well known in the art.

Any of a wide variety of available and well known host cells can be used in the practice of the present invention. The choice of a particular host depends on many factors recognized in the art. These include, for example, compatibility with the chosen expression vector, toxicity of the RNase-Fc fusion protein encoded by the DNA molecule, transformation or transfection efficiency, ease of recovery of the RNase-Fc fusion protein, expression characteristics, biosafety and cost. It is necessary to understand the balance between these factors, i.e., not all hosts may be equally efficient for expression of a particular DNA sequence. In these general guidelines, useful microbial hosts include cultured bacterial (e.g., E.coli), yeast (e.g., Saccharomyces), and other fungal, insect, plant, mammalian (including human) cells or other hosts known in the art. In some embodiments, the RNase-Fc fusion protein is produced in CHO cells.

Next, the transformed or transfected host is cultured and purified. The host cell may be cultured under conventional fermentation or culture conditions in order to express the desired compound. Such fermentation and culture conditions are well known in the art. Finally, the RNase-Fc fusion protein was purified from the culture by methods well known in the art.

The compounds may also be prepared by synthetic methods. For example, solid phase synthesis techniques may be used. Suitable techniques are well known in the art and include those described in Merrifield (1973), chem.polypetides, pp.335-61(Katsoyannis and Panayotis eds.); merrifield (1963), J.Am.chem.Soc.85: 2149; davis et al, Biochem Intl 1985; 10: 394-414; stewart and Young (1969), Solid Phase Peptide Synthesis; U.S. Pat. No.3,941,763; finn et al (1976), The Proteins (3rd ed.)2: 105-253; and Erickson et al (1976), The Proteins (3rd ed.)2: 257-. In some embodiments, the compounds containing derivatized peptides or containing non-peptide groups may be synthesized by well-known organic chemistry techniques.

Other methods of molecular expression/synthesis are generally known to those of ordinary skill in the art.

Pharmaceutical composition

In certain embodiments, RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins, are administered alone. In certain embodiments, the RNase-Fc fusion protein is administered prior to the administration of the at least one additional therapeutic agent. In certain embodiments, the RNase-Fc fusion protein is administered concurrently with the administration of the at least one additional therapeutic agent. In certain embodiments, the RNase-Fc fusion protein is administered after the administration of the at least one additional therapeutic agent. In other embodiments, the RNase-Fc fusion protein is administered prior to the administration of the at least one additional therapeutic agent. One skilled in the art will appreciate that, in some embodiments, the RNase-Fc fusion protein is combined with other agents/compounds. In some embodiments, the RNase-Fc fusion protein and the additional agent are administered simultaneously. In some embodiments, the RNase-Fc fusion protein and the other agent are not administered simultaneously, and the RNase-Fc fusion protein is administered before or after administration of the agent. In some embodiments, the subject receives the RNase-Fc fusion protein and the additional agent simultaneously during the same prophylactic, disease-onset, and/or therapeutic period.

The pharmaceutical compositions of the present disclosure may be administered in a combination therapy, i.e., in combination with other agents. In certain embodiments, the combination therapy comprises an RNase-Fc fusion protein in combination with at least one additional agent. Agents include, but are not limited to, chemical compositions, antibodies, antigen-binding regions, and combinations and conjugates thereof, prepared synthetically in vitro. In certain embodiments, the agent may act as an agonist, antagonist, allosteric modulator, or toxin.

In certain embodiments, the present disclosure provides pharmaceutical compositions comprising an RNase-Fc fusion protein and a pharmaceutically acceptable diluent, carrier, solubilizer, emulsifier, preservative, and/or adjuvant.

In certain embodiments, the present disclosure provides pharmaceutical compositions comprising an RNase-Fc fusion protein and a therapeutically effective amount of at least one additional therapeutic agent, together with pharmaceutically acceptable diluents, carriers, solubilizers, emulsifiers, preservatives and/or adjuvants.

In certain embodiments, acceptable formulation materials are preferably non-toxic to recipients at the dosages and concentrations employed. In some embodiments, the formulation material is for subcutaneous and/or intravenous administration. In certain embodiments, the pharmaceutical composition may comprise formulation materials for altering, maintaining or maintaining, for example, the pH, osmotic pressure, viscosity, clarity, color, isotonicity, odor, sterility, stability, dissolution or release rate, adsorption or permeation of the composition. In certain embodiments, suitable formulation materials include, but are not limited to, amino acids (such as glycine, glutamine, asparagine, arginine, or lysine); an antibacterial agent; antioxidants (such as ascorbic acid, sodium sulfite or sodium bisulfite); buffering agents (such as borate, bicarbonate, Tris-HCl, citrate, phosphate or other organic acids); bulking agents (such as mannitol or glycine); chelating agents (such as ethylenediaminetetraacetic acid (EDTA)); complexing agents (such as caffeine, polyvinylpyrrolidone, beta-cyclodextrin or hydroxypropyl-beta-cyclodextrin); a filler; a monosaccharide; a disaccharide; and other carbohydrates (such as glucose, mannose, or dextrins); proteins (e.g., gelatin); coloring, flavoring and diluting agents; an emulsifier; hydrophilic polymers (such as polyvinylpyrrolidone); a low molecular weight polypeptide; salt-forming counterions (e.g., sodium); preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorhexidine, sorbic acid or hydrogen peroxide); solvents (such as glycerol, propylene glycol or polyethylene glycol); sugar alcohols (such as mannitol or sorbitol); a suspending agent; surfactants or wetting agents (e.g., pluronic, PEG, sorbitan esters, polysorbates such as polysorbate 20, polysorbate 80, tritium, tromethamine, lecithin, cholesterol, tyloxapol); stability enhancers (such as sucrose or sorbitol); tonicity enhancing agents (e.g., alkali metal halides, preferably sodium or potassium chloride, mannitol sorbitol); delivery vehicles (delivery vehicles); a diluent; excipients and/or pharmaceutical adjuvants. (Remington's Pharmaceutical Sciences,18th Edition, A.R. Gennaro, ed., Mack Publishing Company (1995.) in some embodiments, the formulation comprises PBS, 20mM NaOAC, pH 5.2,50mM NaCl, and/or 10mM NAOAC, pH 5.2, 9% sucrose.

In certain embodiments, the RNase-Fc fusion protein and/or therapeutic molecule is linked to a half-life extending vehicle known in the art. Such vehicles include, but are not limited to, polyethylene glycol, glycogen (e.g., glycosylation of RNase-Fc fusion proteins), and dextran. Such vehicles are described, for example, in U.S. application serial No. 09/428,082 (now U.S. patent No. 6,660,843) and published PCT application No. WO 99/25044.

In certain embodiments, the optimal pharmaceutical composition will be determined by one of skill in the art based on, for example, the intended route of administration, delivery form, and desired dosage. See, for example, Remington's Pharmaceutical Sciences, supra. In certain embodiments, such compositions can affect the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the fusion proteins of the present disclosure.

In certain embodiments, the primary vehicle or carrier in the pharmaceutical composition may be aqueous or non-aqueous in nature. For example, in certain embodiments, a suitable vehicle or carrier may be water for injection, a physiological saline solution, or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. In some embodiments, the saline comprises isotonic phosphate buffered saline. In certain embodiments, the pharmaceutical composition comprises a Tris buffer at about pH 7.0-8.5 or an acetate buffer at about pH 4.0-5.5, which may further comprise sorbitol or a suitable substitute thereof. In certain embodiments, a composition comprising an RNase-Fc fusion protein (with or without at least one additional therapeutic agent) for storage may be prepared by mixing the selected composition with the desired purity, optionally in the form of a lyophilized cake or aqueous solution (Remington's Pharmaceutical Sciences, supra). Furthermore, in certain embodiments, the composition comprising the RNase-Fc fusion protein (with or without at least one additional therapeutic agent) may be prepared as a lyophilizate using a suitable excipient (e.g., sucrose).

In certain embodiments, the pharmaceutical composition may be selected for parenteral delivery. In certain embodiments, the composition may be selected for inhalation or delivery through the digestive tract, such as oral administration. The preparation of such pharmaceutically acceptable compositions is within the ability of those skilled in the art.

In certain embodiments, the formulation components are present at concentrations acceptable to the site of administration. In certain embodiments, buffers are used to maintain the composition at physiological pH or in a slightly low pH range, typically in a pH range of about 5 to about 8.

In certain embodiments, when parenteral administration is contemplated, the therapeutic composition may be in the form of a pyrogen-free, parenterally acceptable aqueous solution comprising the desired RNase-Fc fusion protein in a pharmaceutically acceptable vehicle, with or without additional therapeutic agents. In certain embodiments, the parenterally injected vehicle is sterile distilled water, wherein the RNase-Fc fusion protein is prepared as a sterile isotonic solution for proper storage, with or without at least one additional therapeutic agent. In certain embodiments, preparation may involve formulating the desired molecule with an agent that provides controlled or sustained release of the product, which may then be delivered by depot injection. In certain embodiments, hyaluronic acid may also be used and may have the effect of promoting circulation duration. In certain embodiments, implantable drug delivery devices can be used to introduce desired molecules.

In certain embodiments, the pharmaceutical composition may be prepared for inhalation. In certain embodiments, the RNase-Fc fusion protein, with or without at least one additional therapeutic agent, can be prepared as a dry powder for inhalation. In certain embodiments, an inhalation solution comprising an RNase-Fc fusion protein, with or without at least one additional therapeutic agent, may be prepared with a propellant for aerosol delivery. In certain embodiments, the solution may be atomized. Pulmonary administration is further described in PCT application No. PCT/US94/001875, which describes pulmonary delivery of chemically modified proteins.

In certain embodiments, it is contemplated that the formulation may be administered orally. In certain embodiments, the RNase-Fc fusion protein administered in this manner may be prepared with or without carriers typically used in mixed solid dosage forms (e.g., tablets and capsules), with or without at least one additional therapeutic agent. In certain embodiments, the capsule may be designed to release the active portion of the formulation at a point where bioavailability is maximized and pre-systemic degradation is minimized in the gastrointestinal tract. In certain embodiments, at least one additional agent may be included to promote uptake of the RNase-Fc fusion protein and/or any additional therapeutic agent. In certain embodiments, diluents, flavoring agents, low melting waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binding agents may also be used.

In certain embodiments, the pharmaceutical composition may include an effective amount of the RNase-Fc fusion protein, with or without at least one additional therapeutic agent, in admixture with non-toxic excipients suitable for the manufacture of tablets. In certain embodiments, the solution may be prepared in unit dosage form by dissolving the tablet in sterile water or another suitable vehicle. In certain embodiments, suitable excipients include, but are not limited to, inert diluents such as calcium carbonate, sodium carbonate or bicarbonate, lactose, or calcium phosphate; or a binder, such as starch, gelatin or acacia; or a lubricant such as magnesium stearate, stearic acid or talc.

Additional pharmaceutical compositions will be apparent to those skilled in the art, including formulations of the RNase-Fc fusion protein with or without at least one additional therapeutic agent in a sustained or controlled delivery formulation. In certain embodiments, techniques for preparing a variety of other sustained or controlled delivery means, such as liposome carriers, bioerodible microparticles or porous beads, and depot injections, are also known to those of skill in the art. See, for example, PCT application No. PCT/US93/00829, which describes the controlled release of porous polymeric microparticles for the delivery of pharmaceutical compositions. In certain embodiments, the sustained release formulation may include a semipermeable polymer matrix in the form of a shaped article (e.g., a film or microcapsule). Sustained release matrices may include polyesters, hydrogels, polylactic acid (U.S. Pat. No. 3,773,919 and EP058,481), copolymers of L-glutamic acid and gamma-ethyl-L-glutamic acid (Sidman et al, Biopolymers,22:547-556(1983)), poly (2-hydroxyethyl methacrylate) (Langer et al, J Biomed Mater Res,15:167-277(1981) and Langer, Chem Tech,12:98-105(1982)), ethylene vinyl acetate (Langer et al, supra) or poly-D (-) -3-hydroxybutyric acid (EP 133,988). In certain embodiments, the sustained release composition may further comprise liposomes, which may be prepared by any of several methods known in the art. See, e.g., Eppstein et al, PNAS,82: 3688-; EP 036,676; EP 088,046 and EP 143,949.

Pharmaceutical compositions for in vivo administration are typically sterile. In certain embodiments, this may be achieved by filtration through sterile filtration membranes. In certain embodiments, if the composition is lyophilized, the sterilization process may be used before or after lyophilization and reconstitution. In certain embodiments, compositions for parenteral administration may be stored in lyophilized form or in solution form. In certain embodiments, the parenteral composition is typically placed into a container having a sterile access port, such as an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.

In certain embodiments, once the pharmaceutical composition is prepared, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. In certain embodiments, such formulations may be stored in a ready-to-use form or in a form that is reconstituted prior to administration (e.g., lyophilized).

In certain embodiments, kits for producing a single dose administration unit are provided. In certain embodiments, the kit may comprise a first container having a dried protein and a second container having an aqueous formulation. In certain embodiments, kits comprising single-chamber and multi-chamber pre-filled syringes (e.g., liquid syringes and hemolysis syringes) are included. In certain embodiments, the effective amount of a pharmaceutical composition comprising an RNase-Fc fusion protein for therapeutic use, with or without at least one additional therapeutic agent, will depend, for example, on the therapeutic context and objectives. One skilled in the art will appreciate that, according to certain embodiments, the appropriate dosage level for treatment will thus depend, in part, on the indication of the molecule delivered, the RNase-Fc fusion protein, with or without at least one additional therapeutic agent, the route of administration, and the size (body weight, body surface or organ size) and/or condition (age and general health) of the patient. In certain embodiments, the clinician may titrate the dosage and modify the route of administration to obtain the optimal therapeutic effect. In certain embodiments, typical dosages may range from about 0.5mg/kg up to about 50mg/kg or more, depending on the factors described above. In certain embodiments, the dose range may be about 5-10mg/kg, about 2-8mg/kg, about 3-6mg/kg, about 3mg/kg, about 5mg/kg, or about 10 mg/kg.

In certain embodiments, the frequency of administration will take into account pharmacokinetic parameters of the RNase-Fc fusion protein and/or any additional therapeutic agents in the formulation used. In certain embodiments, the clinician will administer the composition until a dosage is reached that achieves the desired effect. In certain embodiments, the composition may thus be administered as a single dose, or over time as two or more doses (which may or may not contain the same amount of the desired molecule) or as a continuous infusion via an implanted device or catheter. Further refinement of appropriate dosages is routinely made by those of ordinary skill in the art and is within the scope of the tasks they routinely perform. In certain embodiments, the appropriate dose may be determined by using appropriate dose response data.

In certain embodiments, the route of administration of the pharmaceutical composition is consistent with known methods, e.g., oral, by injection by intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, subcutaneous, intraocular, intraarterial, intraportal, or intralesional routes; by slow release systems or by implanted devices. In certain embodiments, the composition may be administered continuously by bolus injection or by infusion or by an implanted device.

In certain embodiments, the composition may be administered topically through an implant membrane, sponge, or another suitable material onto which the desired molecule has been absorbed or encapsulated. In certain embodiments, if an implantation device is used, the device may be implanted into any suitable tissue or organ, and the desired molecule may be delivered by diffusion, timed release bolus, or continuous administration.

In certain embodiments, it may be desirable to use a pharmaceutical composition comprising an RNase-Fc fusion protein in an ex vivo manner, with or without at least one additional therapeutic agent. In such cases, cells, tissues and/or organs that have been removed from the patient are exposed to a pharmaceutical composition comprising an RNase-Fc fusion protein, with or without at least one additional therapeutic agent, and then the cells, tissues and/or organs are transplanted back into the patient.

In certain embodiments, the RNase-Fc fusion protein and/or any additional therapeutic agent may be delivered by implantation of certain cells that have been genetically engineered, using methods as described herein, to express and secrete the polypeptide. In certain embodiments, such cells may be animal or human cells, and may be autologous, allogeneic or xenogeneic. In certain embodiments, the cell may be immortalized. In certain embodiments, to reduce the chance of an immune response, cells may be encapsulated to avoid infiltration into surrounding tissues. In certain embodiments, the encapsulating material is generally a biocompatible, semi-permeable polymeric shell or membrane that allows for the release of protein products but prevents the cells from being damaged by the patient's immune system or other harmful factors from surrounding tissues.

In vitro assay

Various in vitro assays known in the art can be used to assess the efficacy of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins.

For example, cultured human PBMCs from normal subjects, lupus patient PBMCs, or xerosis patient PBMCs are isolated, cultured and treated with various stimuli (e.g., TLR ligands, co-stimulatory antibodies, immune complexes, and normal or autoimmune serum), in the presence or absence of RNase-Fc fusion proteins. Cytokines produced by stimulated cells can be measured using commercially available reagents such as antibody pair kits for various cytokines (e.g., IL-6, IL-8, IL-10, IL-4, IFN-. gamma., and TNF-. alpha.) from Biolegend (San Diego, Calif.). Culture supernatants were collected at various time points (e.g., 24, 48 hours or later) appropriate for the assay to determine the effect of the RNase-Fc fusion protein on cytokine production. IFN- α production is measured using anti-human IFN- α antibodies, such as those available from PBL interferon source (Piscataway, NJ), and standard curve reagents. Similar assays were performed using human lymphocyte subpopulations (isolated monocytes, B cells, pDC, T cells, etc.); purification is carried out using a commercially available magnetic bead-based isolation kit, e.g., available from Miltenyi Biotech (Auburn, CA).

Multicolor flow cytometry can be used to assess the effect of RNase-Fc fusion proteins on immune cell activation by measuring expression of lymphocyte activating receptors (e.g., CD5, CD23, CD69, CD80, CD86, and CD25 in PBMC), or to isolate cell subsets at different time points after stimulation using routine methods recognized in the art.

The efficacy of RNase-Fc fusion proteins can also be tested by incubating SLE or xerosis (Sjogren's) patient sera with normal human pDC to activate IFN output, as described, for example, in Ahlin et al, Lupus 2012:21: 586-95; mathsson et al, Clin Expt Immunol 2007; 147: 513-20; and Chiang et al, J Immunol 2011; 186:1279 and 1288. Without being bound by theory, circulating immune complexes containing nucleic acids in the serum of SLE or xerosis patients promote entry of nucleic acid antigens into pDC endosomes through Fc receptor mediated endocytosis, followed by nucleic acid binding and activation of endosomal TLRs 7, 8 and 9. To assess the effect of RNase-Fc fusion proteins, serum or plasma from SLE or xerosis patients was pretreated with RNase-Fc fusion proteins and then added to pDC cell cultures isolated from healthy volunteers. The level of IFN- α produced at various time points was then determined. By degrading the immune complex containing the nucleic acid, an effective RNase-Fc fusion protein is expected to reduce the amount of IFN-. alpha.produced.

The effectiveness of the RNase-Fc fusion protein was demonstrated by comparing the assay results of cells treated with the RNase-Fc fusion protein disclosed herein with the assay results of cells treated with a control formulation. After treatment, the levels of the various markers described above (e.g., cytokines, cell surface receptors, proliferation) are generally improved in the treated group for an effective RNase-Fc fusion protein relative to the levels of the marker present prior to treatment, or relative to levels measured in a control group.

Method of treatment

The RNase-containing nuclease fusion proteins of the present disclosure, including the RNase-Fc fusion proteins of the present disclosure, are particularly effective in treating autoimmune diseases or abnormal immune responses. In this regard, it is understood that RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins of the present disclosure, are useful for controlling, inhibiting, modulating, treating, or eliminating unwanted immune responses to external and self-antigens. In some embodiments, the RNase-containing nuclease fusion proteins of the present disclosure, including the RNase-Fc fusion proteins of the present disclosure, are used to treat or reduce fatigue in patients with autoimmune diseases. In some embodiments, the fatigue is sjogren's syndrome-associated fatigue.

In some aspects, the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins, are provided in an effective or sufficient amount to a human patient in need thereof by administering an RNase-containing nuclease fusion protein of the present disclosure (e.g., an RNase-Fc fusion protein), and can be used to treat autoimmune diseases in the human patient, thereby treating the disease. The present disclosure encompasses any route of administration (e.g., intravenous, intramuscular, subcutaneous) suitable for achieving the desired effect. Treatment of a disease condition can result in a reduction in symptoms associated with the condition, which can be long-term or short-term, or even a transient beneficial effect. In some embodiments, treatment of xerosis results in reduced fatigue associated with the disease or condition.

Biochemical assay

In some embodiments, RNase-containing nuclease fusion proteins, including RNase-Fc fusion proteins, of the present disclosure are administered to a human patient in need thereof to treat sjogren's syndrome. In some aspects, the effectiveness of an RNase-Fc fusion protein is demonstrated by comparing IFN- α levels, IFN- α response gene levels, autoantibody titers, renal function, and levels of pathological and/or circulating immune complexes in human patients treated with an RNase-Fc fusion protein disclosed herein as compared to placebo.

For example, a human subject in need of treatment (e.g., a patient meeting the american-european consensus's criteria for the classification of xerosis) is selected or identified. A subject may need, for example, to alleviate the cause or symptoms of sjogren's syndrome, e.g., pSS. In some embodiments, the patient has sjogren's syndrome and is in need of fatigue reduction. The identification of the subject may be performed in a clinical setting or elsewhere, for example by the subject himself using a self-test kit in the subject's home.

At baseline (day 1), a suitable first dose of an RNase-containing nuclease fusion protein of the present disclosure, including an RNase-Fc fusion protein, is administered to a patient in need thereof. RNase-Fc fusion proteins were prepared as described herein. The condition of the patient is assessed at baseline (day 1) and some time after the first dose, e.g., day 8, day 15, day 29, day 43, day 57, day 71, day 85, day 99 or at the end of the study, e.g., by measuring IFN- α levels, IFN- α response gene levels, autoantibody titers, renal function and pathology and/or circulating immune complex levels. Other relevant criteria may also be measured. The number and intensity of administration is adjusted to the needs of the subject. Following treatment, the subject's IFN- α levels, IFN- α response gene levels, autoantibody titers, renal function, and pathology and/or circulating immune complex levels are reduced and/or improved relative to levels present prior to treatment, or relative to levels measured in similarly diseased but untreated/control subjects.

Fatigue determination

Various Patient Report Outcome (PRO) instruments have been used and validated in measuring fatigue in chronically ill subjects. Such PRO are known in the art and can be used to assess the efficacy of RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins. EULAR Sicca Syndrome Patient Reporting Index (ESSPRI)

The European Association of antirheumatics (EULAR) sicca (SS) patient report index (ESSPRI) aims at assessing the symptoms of patients with primary sicca (Seror et al, Ann. Rheum. Dis.2011; 70: 968-. ESSPRI was developed as a global score to measure all the important and disabling symptoms of primary sjogren's syndrome: dryness, pain in the extremities and fatigue. ESSPRI has proven to be sufficient to measure each symptom without losing content effectiveness, and the score is easy to calculate.

ESSPRI is a patient-administered questionnaire used to assess the symptoms of patients with primary sjogren's syndrome. The questionnaire includes three scales, one for each symptom: (1) dryness, (2) limb pain, and (3) fatigue. Each component of the ESSPRI is measured by a numerical scale from 0 to 10, and the global ESSPRI score is the average of three scales: (dryness + pain in limbs + fatigue)/3. A reduction in the espri score of at least 1 point is clinically significant.

In some embodiments, the effectiveness of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof, is demonstrated by assessing the improvement in fatigue in a patient after treatment with the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof. After treatment, fatigue of the patient as measured by ESSPRI is generally reduced compared to the level of fatigue in the patient prior to treatment, and/or compared to the patient treated with the control formulation.

FACIT-fatigue

The chronic disease treatment Fatigue function assessment scale (FACIT-Fatigue) is used to assess the degree of Fatigue in an individual's daily activities over the past week.

The FACIT-fatigue questionnaire and scoring and interpretation materials are available from FACIT. The FACIT-Fatigue questionnaire provides a series of general and targeted measurements. The FACIT fatigue scale has many advantages, including high internal efficacy, high retest reliability, reliability and sensitivity to changes in patients with various chronic health conditions, ease of use, and use in various environments (K.F. Tennant, Try This: best Practices in Nurseng Car to holder adapters, Issue 30,2012; Chandran et al, Ann. Rheum. Dis.2007; 66: 936-.

FACIT-Fatigue is a questionnaire containing 13 items, originally developed to measure Fatigue in cancer patients and now in xerosis patients. The patient was asked to answer 13 questions, with a score ranging from 0 to 4(0 ═ none at all, 1 ═ some, 2 ═ some, 3 ═ many, 4 ═ very). The fatigue scale has 13 items, with 52 being the highest score. A higher score in the fatigue scale corresponds to a lower fatigue level and indicates a better quality of life.

To calculate the FACIT fatigue score, the answer scores of the negative phrase questions are reversed and then the 13 item answers are added. The scores of the 11 items with answers are reversed (item score 4-answer if the answer is not lost) and the answers of the two items (items 7-8) are unchanged. All items are added so that a higher score corresponds to less fatigue. In the case of skipping individual questions, the scores are scaled using the average of the other answers in the scale.

FACIT-fatigue 13 [ totalscore (inverted item) + total score (item 7-8) ]/number of items answered

In some embodiments, the effectiveness of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof, is demonstrated by assessing the improvement in fatigue in a patient after treatment with the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof. After treatment, fatigue in patients measured by the FACIT fatigue scale is generally reduced compared to the level of fatigue in patients before treatment, and/or compared to patients treated with control formulations.

General overview of fatigue

Fatigue profiles (ProF) were developed in order to establish an assessment tool that can effectively characterize fatigue associated with primary sjogren's syndrome. Thus, the words used by primary sjogren's syndrome patients to express their complaints about fatigue, discomfort and pain were used in the ProF questionnaire. ProF has proven to be a reliable and effective tool for measuring the severity of fatigue and general discomfort in patients with primary sjogren syndrome.

ProF is a 16-item self-filling questionnaire divided into two fields, one being physical fatigue and one being mental fatigue. The body fatigue field comprises 12 items, which are divided into four aspects: (a) requiring rest (4 projects), (b) poor launch (3 projects), (c) low endurance (3 projects), and (d) muscle weakness (2 projects). The field of mental fatigue includes 4 items, which are divided into two aspects: (a) inattention (two items) and (b) poor memory (two items). Based on the worst feeling the patient had over the past two weeks, the patient scored each item in the range of 0-7(0 ═ no problems at all "and 7 ═ as bad as can be imagined). The score for each facet may be obtained by adding the item scores within each facet and dividing the sum by the number of items in each facet. The score for each domain (e.g., somatic, mental) can be obtained by adding the facet scores within each domain and dividing the sum by the number of facets within each domain. Higher scores indicate higher fatigue. (Bowman et al, Rheumatology, 2004; 43: 758-.

In some embodiments, the effectiveness of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof, is demonstrated by assessing the improvement in fatigue in a patient after treatment with the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof. After treatment, fatigue in patients measured by PROF is generally reduced compared to the level of fatigue in patients before treatment, and/or compared to patients treated with control formulations.

Assessment of reduction of Sjogren syndrome-associated fatigue

In some embodiments, the effectiveness of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof, is demonstrated by assessing the improvement in fatigue in a patient after treatment with the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof. In some embodiments, a patient treated with an RNase-Fc fusion protein will exhibit reduced (demonstrate) fatigue when compared to the level of fatigue in the patient prior to treatment and/or when compared to a patient treated with a control formulation. In some embodiments, the fatigue is sjogren's syndrome-associated fatigue.

In some embodiments, the condition of the patient is assessed by measuring the patient's fatigue through one or more patient reported indices (e.g., espri, PROF, FACIT) as compared to the patient's fatigue prior to treatment or to the fatigue of similarly afflicted untreated or control patients. In some embodiments, the effectiveness of the RNase-Fc fusion protein is demonstrated by assessing the EULAR SS patient reporting index (espri), profile or fatigue (PROF), and/or chronic disease treatment Function Assessment (FACIT) fatigue scale for patients treated with the RNase-Fc fusion proteins disclosed herein when compared to patients treated with control formulations. In some embodiments, a patient treated with an RNase-Fc fusion protein will exhibit an improvement in the (demonstrate) espri index, PROF, and/or FACIT fatigue scale as compared to the espri index, PROF, and/or FACIT fatigue scale of the patient prior to treatment, or as compared to a patient treated with a control formulation.

For example, a human subject in need of treatment is selected or identified (e.g., a patient meeting american society for rheumatism SLE criteria, or a patient meeting american-european consensus xerosis classification criteria). The subject may be in need of, e.g., alleviation of the cause or symptoms (e.g., fatigue) of SLE or sjogren's syndrome. The identification of the subject may be performed in a clinical setting or elsewhere, for example by the subject himself using a self-test kit in the subject's home.

At baseline (day 1), a suitable first dose of an RNase-containing nuclease fusion protein of the present disclosure, including an RNase-Fc fusion protein, is administered to a patient in need thereof. RNase-Fc fusion proteins were prepared as described herein. The patient's condition is assessed by the ESSPRI index, PROF and/or FACIT fatigue scale at baseline (day 1) and after a period of time following the first dose, e.g., day 8, day 15, day 29, day 43, day 57, day 71, day 85, day 99 or at the end of the study. Other relevant criteria may also be measured. The number and intensity of administration is adjusted to the needs of the subject. After treatment, improvements in one or more of the following outcomes may be noted: (1) an improvement in the ESSPRI index relative to pre-treatment or relative to an analogous diseased but untreated/control subject, (2) an improvement in the PROF relative to pre-treatment or relative to an analogous diseased but untreated/control subject, (3) an improvement in the FACIT fatigue scale relative to pre-treatment or relative to an analogous diseased but untreated/control subject may be noted. In some embodiments, the improvement in the ESSPRI index is a clinically meaningful improvement. A clinically significant improvement in the ESSPRI index is a reduction in the ESSPRI score of at least 1 point.

Neuropsychological analysis of fatigue determination

Various neuropsychological assays known in the art can be used to assess the efficacy of RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins.

Digit symbol replacement testing

Digital Symbol Substitution Testing (DSST) provides an effective and sensitive test to measure cognitive dysfunction affected by many areas. DSST is sensitive to the presence of cognitive dysfunction and changes in cognitive function in a range of clinical populations, including patients with sjogren's syndrome. Such neuropsychological tests are widely used, highly validated and extremely sensitive tests that read executive function-related inputs.

DSST is a time-limited paper-pen cognitive test given on a piece of paper. The test requires the patient to match a symbol to a number according to a key on top of the paper. The patient copies the symbol into a space below a row of numbers and counts the number of correct symbols within an allowed time (e.g., 90 or 120 seconds). The test provides data on the accuracy and rate at which tasks are performed. Patient performance on DSSTs correlates with real-world functional outcomes, such as the ability to perform daily tasks, and recovery from dysfunction in a range of psychiatric disorders. The DSST test may be used to assess the attention and/or concentration of a patient.

DSST is a multifactorial test that can measure a series of cognitive operations and provides a practical and effective method to monitor cognitive function over time. In order to perform well on DSSTs, patients must have complete motor speed, attention, and visual perception capabilities, including the ability to scan and write or draw (i.e., basic mental dexterity). DSST offers high sensitivity for detecting cognitive impairment and has many advantages, including compactness, reliability, sensitivity to changes, and minimal impact of language, culture, and education on test performance. (Jaeger, J., Journal of Clinical Psycopharmacology,38 (5)), 513-.

In addition, DSST is used in clinical development to define pharmacokinetic/pharmacodynamic (PK/PD) relationships. This test also serves as a PD biomarker in CNS studies, allowing discrimination between two doses of Selective Serotonin Reuptake Inhibitors (SSRIs).

In some embodiments, the effectiveness of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof, is demonstrated by assessing the improvement in fatigue in a patient after treatment with the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof. After treatment, the cognitive function of the patient as measured by the DSST test is generally improved compared to the level of cognitive function in the patient prior to treatment, and/or compared to the patient treated with the control formulation.

Assessing improvement in cognitive function associated with sjogren's syndrome

In some embodiments, the effectiveness of the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof, is demonstrated by assessing the improvement in fatigue in a patient after treatment with the RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins or pharmaceutical compositions thereof. In some embodiments, a patient treated with an RNase-Fc fusion protein exhibits an improvement in cognitive function when compared to the level of cognitive function in the patient prior to treatment and/or when compared to a patient treated with a control formulation. In some embodiments, the patient has sjogren's syndrome.

In some embodiments, the condition of the patient is assessed by measuring cognitive function of the patient by one or more neuropsychological assays (e.g., DSST) as compared to cognitive function of the patient prior to treatment or as compared to cognitive function of a similarly afflicted untreated or control patient. In some embodiments, the effectiveness of the RNase-Fc fusion protein is demonstrated by evaluating the results of a DSST test for patients treated with the RNase-Fc fusion proteins disclosed herein when compared to patients treated with a control formulation. In some embodiments, a patient treated with the RNase-Fc fusion protein will exhibit an improvement in the (demonstrate) DSST test as compared to a DSST test in a patient prior to treatment, or as compared to a patient treated with a control formulation.

For example, a human subject in need of treatment is selected or identified (e.g., a patient meeting american society for rheumatism SLE criteria, or a patient meeting american-european consensus xerosis classification criteria). The subject may be in need of, e.g., alleviation of the cause or symptoms (e.g., fatigue) of SLE or sjogren's syndrome. The identification of the subject may be performed in a clinical setting or elsewhere, for example by the subject himself using a self-test kit in the subject's home.

At baseline (day 1), a suitable first dose of an RNase-containing nuclease fusion protein of the present disclosure, including an RNase-Fc fusion protein, is administered to a patient in need thereof. RNase-Fc fusion proteins were prepared as described herein. The condition of the patient is assessed, e.g., by a DSST test, at baseline (day 1) and after a period of time following the first dose, e.g., day 8, day 15, day 29, day 43, day 57, day 71, day 85, day 99, or at the end of the study. Other relevant criteria may also be measured. The number and intensity of administration is adjusted to the needs of the subject. Post-treatment, the DSST test score improved relative to the pre-treatment DSST test score or relative to similar diseased but untreated/control subjects.

Dosing regimens

RNase-containing nuclease fusion proteins of the present disclosure, including RNase-Fc fusion proteins of the present disclosure and/or pharmaceutical compositions of the present disclosure, are prepared into pharmaceutically acceptable dosage forms for human subjects by conventional methods known to those skilled in the art. In some embodiments, the actual dosage level of the active ingredient (i.e., the RNase-Fc fusion) in the pharmaceutical compositions of the present disclosure is varied (are varied) to obtain an amount of the active ingredient effective to achieve a desired therapeutic response for a particular human patient, composition, and/or mode of administration without unacceptable toxicity to the patient.

The selected dosage level will depend upon a variety of factors including the activity of the particular RNase-Fc fusion protein, the route of administration, the time of administration, the rate of excretion or metabolism of the particular RNase-Fc fusion protein administered, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular RNase-Fc fusion protein, the age, sex, body weight, condition, general health and past medical history of the patient undergoing treatment, and factors well known in the medical arts.

In general, a suitable dose of an RNase-Fc fusion protein or composition of the present disclosure is the amount of the active ingredient that is the lowest dose effective to produce a therapeutic effect in a human subject. Such effective dosages will generally depend on the factors described above. Generally, intravenous, oral, and subcutaneous doses of the RNase-Fc fusion proteins or compositions of the present disclosure for use in human patients range from about 0.5mg to about 50mg per kilogram body weight per week when used for the indicated effects. In some embodiments, the RNase-Fc fusion protein or pharmaceutical composition of the present disclosure is administered to a human patient in need thereof by injection (e.g., by intravenous injection, e.g., by infusion) at a dose of about 0.5mg to about 50mg per kilogram body weight per week.

In some embodiments, the RNase-Fc fusion protein or composition of the present disclosure is administered to a human patient at a dose of typically about 1-20mg/kg weekly, 2-10mg/kg weekly, 5-15mg/kg weekly, 5-10mg/kg weekly, or 2-5mg/kg weekly. In some embodiments, a dose of greater than 10mg/kg, or greater than 15mg/kg or greater than 20mg/kg per week may be necessary. In some embodiments, a dose of less than 20mg/kg, or less than 15mg/kg, or less than 10mg/kg per week may be necessary. In some embodiments, parenteral administration, such as, for example, intravenous injection, is about 5-10mg/kg per week to a human patient. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dosage of about 0.5mg/kg, 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, 8mg/kg, 9mg/kg10mg/kg, 11mg/kg, 12mg/kg, 14mg/kg, 15mg/kg, 16mg/kg, 17mg/kg, 18mg/kg, 19mg/kg20mg/kg, 21mg/kg, 22mg/kg, 23mg/kg, 24mg/kg 25mg/kg weekly, biweekly, monthly or semi-monthly (e.g., every 2 months, every 3 months). In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 1mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 2mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 3mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 4mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 5mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 6mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 7mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 8mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 9mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 10mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 12mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the RNase-Fc fusion protein is administered to the human patient at a dose of 15mg/kg weekly, biweekly, monthly, or semi-monthly. In some embodiments, the aforementioned dose is prepared for intravenous injection.

If desired, weekly, biweekly, monthly or semi-monthly effective doses of an RNase-Fc fusion protein or composition of the disclosure, optionally in unit dosage form, are administered to a human patient as two, three, four, five, six or more sub-doses, administered separately at appropriate intervals throughout the day (e.g., as intravenous injections or infusions). In some embodiments, the administration is once daily administration. In some embodiments, administration is one or more administrations every 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 days, or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 weeks, or every 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 months, as needed, to obtain a therapeutic effect (e.g., sufficient to digest circulating RNA complexed with autoantibodies and/or RNA containing immune complexes). In some embodiments, the administration is once weekly administration (e.g., intravenous injection or infusion). In some embodiments, the administration is once or multiple administrations every two weeks. In some embodiments, the administration is once every two weeks. In some embodiments, the administration is one or more administrations per month. In some embodiments, the administration is a monthly administration. In some embodiments, the administration is one or more administrations per half month (e.g., every 2 months, every 3 months). In some embodiments, the administration is once every 2 months or every 3 months. In some embodiments, the aforementioned dose is prepared for intravenous injection.

In some embodiments, administration is once weekly administration (e.g., intravenous injection or infusion) for two weeks, then once every two weeks to achieve or maintain a therapeutic effect. In some embodiments, administration is once weekly for three weeks, then every two weeks to achieve or maintain a therapeutic effect. In some embodiments, administration is once weekly for four weeks, then every two weeks to achieve or maintain a therapeutic effect. In some embodiments, administration is once weekly for two weeks, then monthly to achieve or maintain a therapeutic effect. In some embodiments, the administration is once weekly for three weeks, then monthly to achieve or maintain a therapeutic effect. In some embodiments, administration is once weekly for four weeks, followed by once monthly administration to achieve or maintain a therapeutic effect. In some embodiments, the aforementioned dose is prepared for intravenous injection. As used herein, an initial weekly administration followed by a bi-weekly, monthly, or semi-monthly administration is referred to as a "loading dose".

In some embodiments, the RNase-Fc fusion protein or composition of the present disclosure is administered weekly (e.g., by intravenous injection or infusion) at a dose of about 1 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 2 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 3 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 4 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 5 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 6 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 7 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 8 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 9 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 10 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 12 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly at a dose of about 15 mg/kg. In some embodiments, the aforementioned dose is prepared for intravenous injection. As used herein, weekly (weekly) is understood to have the art-recognized meaning of weekly (every week).

In some embodiments, the RNase-Fc fusion protein or composition of the present disclosure is administered (e.g., by intravenous injection or infusion) at a dose of about 1mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 2mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 3mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 4mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 5mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 6mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 7mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 8mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 9mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 12mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 15mg/kg every two weeks. In some embodiments, the aforementioned dose is prepared for intravenous injection. As used herein, every two weeks (biweekly) is understood to have the art-recognized meaning of every two weeks (every two weeks).

In some embodiments, the RNase-Fc fusion protein or composition of the present disclosure is administered (e.g., by intravenous injection or infusion) once every three weeks at a dose of about 1 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 2mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 3mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 4mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 5mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 6mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 7mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 8mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 9mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 12mg/kg once every three weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 15mg/kg once every three weeks. In some embodiments, the aforementioned dose is prepared for intravenous injection. As used herein, every three weeks (every third week) is understood to have the art-recognized meaning of once every three weeks (once every three weeks).

In some embodiments, the RNase-Fc fusion protein or composition of the present disclosure is administered monthly (e.g., by intravenous injection or infusion) at a dose of about 1 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 2 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 3 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 4 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 5 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 6 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 7 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 8 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 9 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 10 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 12 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered monthly at a dose of about 15 mg/kg. In some embodiments, the aforementioned dose is prepared for intravenous injection. As used herein, monthly is understood to have the art-recognized meaning of monthly.

In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered (e.g., by intravenous injection or infusion) at a dose of about 2mg/kg weekly for two weeks, and then at a dose of about 2mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 2mg/kg weekly for three weeks, and then at a dose of about 2mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 2mg/kg weekly for four weeks, and then at a dose of about 2mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 2mg/kg weekly for two weeks, and then monthly at a dose of about 2 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 2mg/kg weekly for three weeks, and then monthly at a dose of about 2 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly for four weeks at a dose of about 2mg/kg, and then monthly at a dose of about 2 mg/kg. In some embodiments, the aforementioned dose is prepared for intravenous injection.

In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered (e.g., by intravenous injection or infusion) at a dose of about 5mg/kg weekly for two weeks, and then at a dose of about 5mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 5mg/kg weekly for three weeks, and then at a dose of about 5mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 5mg/kg weekly for four weeks, and then at a dose of about 5mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 5mg/kg weekly for two weeks, and then monthly at a dose of about 5 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 5mg/kg weekly for three weeks, and then monthly at a dose of about 5 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered weekly for four weeks at a dose of about 5mg/kg, and then monthly at a dose of about 5 mg/kg. In some embodiments, the aforementioned dose is prepared for intravenous injection.

In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered (e.g., by intravenous injection or infusion) at a dose of about 10mg/kg weekly for two weeks, and then at a dose of about 10mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg weekly for three weeks, and then at a dose of about 10mg/kg every two weeks. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg weekly for four weeks, and then every two weeks at a dose of about 10 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg weekly for two weeks, and then monthly at a dose of about 10 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg weekly for three weeks, and then monthly at a dose of about 10 mg/kg. In some embodiments, an RNase-Fc fusion protein or composition of the present disclosure is administered at a dose of about 10mg/kg weekly for four weeks, and then monthly at a dose of about 10 mg/kg. In some embodiments, the aforementioned dose is prepared for intravenous injection.

As understood in the art, weekly, biweekly, triweekly, or monthly administration may be one or more administrations or divided doses as described above.

In one embodiment, the effective amount of the RNase-Fc fusion protein is about 2mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 3mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 4mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 5mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 6mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 7mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 8mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 9mg/kg per week per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 10mg/kg per week per human subject. In some embodiments, the aforementioned dose is prepared for intravenous injection.

In one embodiment, the effective amount of the RNase-Fc fusion protein is about 2mg/kg every two weeks per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 3mg/kg every two weeks per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 4mg/kg biweekly per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 5mg/kg every two weeks per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 6mg/kg biweekly per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 7mg/kg every two weeks per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 8mg/kg every two weeks per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 9mg/kg biweekly per human subject. In one embodiment, the effective amount of the RNase-Fc fusion protein is about 10mg/kg every two weeks per human subject. In some embodiments, the aforementioned dose is prepared for intravenous injection.

Methods and uses of inflammation-associated molecules

Provided herein are methods and uses for the diagnosis and treatment of inflammation-related molecules (e.g., inflammation-related genes, inflammation-related proteins, pro-inflammatory molecules) described herein. Further provided herein are methods of identifying a subject having xerosis who is likely to respond to treatment with an RNA nuclease agent described herein by detecting the presence of or determining the amount or expression level of one or more inflammation-associated molecules (e.g., amount or expression level) in a sample obtained from the subject, wherein the presence or amount or expression level of the one or more inflammation-associated molecules indicates that the subject is likely to respond to treatment with the RNA nuclease agent.

Inflammation-associated molecules

In some aspects, the present disclosure provides methods for detecting the presence of or determining the amount or level of expression of an inflammation-associated molecule (e.g., an inflammation-associated gene) in a sample from a subject (e.g., a sample from a subject with sjogren's syndrome). In some aspects, the disclosure provides methods of identifying a subject with xerosis as a candidate for treatment with a ribonuclease agent (e.g., RSLV-132) by determining an inflammation-related gene expression profile in a sample obtained from the subject, and comparing the inflammation-related gene expression profile determined in the sample obtained from the subject to an inflammation-related gene expression profile in a sample obtained from a suitable control subject, wherein the inflammation-related gene expression profile indicates that the subject is a candidate for treatment with an RNA nuclease agent (e.g., RSLV-132).

As used herein, the term "inflammation-associated molecule" refers to a molecule that plays a role in inflammation or inflammatory response. In some embodiments, the inflammation-associated molecule is a pro-inflammatory molecule. In some embodiments, the inflammation-associated molecule is an anti-inflammatory molecule. In some embodiments, the inflammation-associated molecule is an inflammatory mediator. In some embodiments, the inflammation-associated molecule is an inflammation-associated protein. In some embodiments, the inflammation-associated molecule is an inflammation-associated cytokine. In some embodiments, the inflammation-associated molecule is an inflammation-associated gene.

In some embodiments, the inflammation-associated gene is IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, STAT5B, CXCL10(IP-10), CD163, RIPK2, and/or CCR 2.

In some embodiments, the inflammation-associated gene is IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, and/or STAT 5B.

In some embodiments, the inflammation-associated gene is CXCL10(IP-10), CD163, RIPK2, and/or CCR 2.

In some embodiments, the inflammation-associated gene is IL-5.

In some embodiments, the inflammation-associated gene is a TNF receptor.

In some embodiments, the inflammation-associated gene is an IL-6 receptor.

In some embodiments, the inflammation-associated gene is an IL-1 accessory protein.

In some embodiments, the inflammation-associated gene is CXCL 1.

In some embodiments, the inflammation-associated gene is IL-17 receptor A.

In some embodiments, the inflammation-associated gene is LTBR 4.

In some embodiments, the inflammation-associated gene is STAT 5B.

In some embodiments, the inflammation-associated gene is CXCL10 (IP-10).

In some embodiments, the inflammation-associated gene is CD 163.

In some embodiments, the inflammation-associated gene is RIPK 2.

In some embodiments, the inflammation-associated gene is CCR 2.

In some embodiments, the inflammation-associated gene is IL5, which is a gene encoding the protein interleukin 5 (IL-5).

In some embodiments, the inflammation-associated gene is TNFRSF1A, which is a gene encoding the protein "TNF receptor superfamily member 1A".

In some embodiments, the inflammation-associated gene is IL6R, which is a gene encoding the protein interleukin 6 receptor (IL-6 receptor).

In some embodiments, the inflammation-associated gene is IL1RAP, which is a gene encoding the protein "interleukin 1 receptor accessory protein".

In some embodiments, the inflammation-associated gene is CXCL1, which is a gene encoding the protein "C-X-C motif chemokine ligand 1(CXCL 1)".

In some embodiments, the inflammation-associated gene is IL17RA, which is a gene encoding the protein "interleukin 17 receptor a".

In some embodiments, the inflammation-associated gene is LTB4R, which is a gene encoding the protein "leukotriene B4 receptor".

In some embodiments, the inflammation-associated gene is STAT5B, a gene that encodes the protein "signal transducer and activator of transcription 5B (transcription factor STAT 5B)".

In some embodiments, the inflammation-associated gene is CXCL10, which is a gene encoding the protein "C-X-C motif chemokine ligand 10 (IP-10)".

In some embodiments, the inflammation-associated gene is CD163, which is a gene encoding the protein "CD 163".

In some embodiments, the inflammation-associated gene is RIPK2, which is a gene encoding the protein receptor-interacting serine/threonine kinase 2.

In some embodiments, the inflammation-associated gene is CCR2, which is a gene encoding the protein "C-C motif chemokine receptor 2(CCR 2)".

In some embodiments, the inflammation-associated gene is IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, STAT5B, CXCL10, CD163, RIPK2, and/or CCR 2.

In some embodiments, the inflammation-associated gene is IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and/or STAT 5B.

In some embodiments, the inflammation-associated gene is CXCL10, CD163, RIPK2, and/or CCR 2.

In some embodiments, the inflammation-associated gene is APOL, HGF, TBC1D, SETD, CCR, CD163, CD, CYBB, PLA2G, IDO, RIPK, ACER, CXCL, AIMP, BIRC, SNX, PTPN, VAMP, APPL, CSF, GBA, GPS, AKT, MAPKAPK, PGLYRP, NUPR, TNFRSF1, MAPK, ORM, CCN, F11, NFAM, IL17, MMP, ADAM, NDST, FOS, NLRP, PIK3, IL1R, STAT5, TREM, SIRPA, IL6, SLC11A, LTB4, BCL, MMP, FPR, xa tb2, NOD, IL1, IL, CXCL, TPST, ZC3H12, TYROBP, and/or CDK.

In some embodiments, the inflammation-associated gene is APOL 3. In some embodiments, the inflammation-associated gene is HGF. In some embodiments, the inflammation-associated gene is TBC1D 23. In some embodiments, the inflammation-associated gene is SETD 6. In some embodiments, the inflammation-associated gene is CCR 2. In some embodiments, the inflammation-associated gene is CD 47. In some embodiments, the inflammation-associated gene is CD 163. In some embodiments, the inflammation-associated gene is CD 36. In some embodiments, the inflammation-associated gene is CYBB. In some embodiments, the inflammation-associated gene is PLA2G 7. In some embodiments, the inflammation-associated gene is IDO 1. In some embodiments, the inflammation-associated gene is RIPK 2. In some embodiments, the inflammation-associated gene is ACER 3. In some embodiments, the inflammation-associated gene is CXCL 10. In some embodiments, the inflammation-associated gene is AIMP 1. In some embodiments, the inflammation-associated gene is BIRC 3. In some embodiments, the inflammation-associated gene is SNX 4. In some embodiments, the inflammation-associated gene is PTPN 2. In some embodiments, the inflammation-associated gene is VAMP 7. In some embodiments, the inflammation-associated gene is APPL 1. In some embodiments, the inflammation-associated gene is CSF 1. In some embodiments, the inflammation-associated gene is GBA. In some embodiments, the inflammation-associated gene is GPS 2. In some embodiments, the inflammation-associated gene is AKT 1. In some embodiments, the inflammation-associated gene is MAPKAPK 2. In some embodiments, the inflammation-associated gene is PGLYRP 1. In some embodiments, the inflammation-associated gene is NUPR 1. In some embodiments, the inflammation-associated gene is TNFRSF 1A. In some embodiments, the inflammation-associated gene is MAPK 13. In some embodiments, the inflammation-associated gene is ORM 2. In some embodiments, the inflammation-associated gene is CCN 3. In some embodiments, the inflammation-associated gene is F11R. In some embodiments, the inflammation-associated gene is NFAM 1. In some embodiments, the inflammation-associated gene is IL17 RA. In some embodiments, the inflammation-associated gene is MMP 25. In some embodiments, the inflammation-associated gene is ADAM 8. In some embodiments, the inflammation-associated gene is NDST 1. In some embodiments, the inflammation-associated gene is FOS. In some embodiments, the inflammation-associated gene is NLRP 12. In some embodiments, the inflammation-associated gene is PIK3 CD. In some embodiments, the inflammation-associated gene is IL1 RAP. In some embodiments, the inflammation-associated gene is IL1R 2. In some embodiments, the inflammation-associated gene is STAT 5B. In some embodiments, the inflammation-associated gene is TREM 1. In some embodiments, the inflammation-associated gene is SIRPA. In some embodiments, the inflammation-associated gene is IL 6R. In some embodiments, the inflammation-associated gene is SLC11a 1. In some embodiments, the inflammation-associated gene is LTB 4R. In some embodiments, the inflammation-associated gene is BCL 6. In some embodiments, the inflammation-associated gene is MMP 9. In some embodiments, the inflammation-associated gene is FPR 1. In some embodiments, the inflammation-associated gene is FPR 2. In some embodiments, the inflammation-associated gene is TBXA 2R. In some embodiments, the inflammation-associated gene is NOD 2. In some embodiments, the inflammation-associated gene is IL1 RN. In some embodiments, the inflammation-associated gene is IL 5. In some embodiments, the inflammation-associated gene is CXCL 1. In some embodiments, the inflammation-associated gene is TPST 1. In some embodiments, the inflammation-associated gene is ZC3H 12A. In some embodiments, the inflammation-associated gene is TYROBP. In some embodiments, the inflammation-associated gene is CDK 19.

In some embodiments, the inflammation-associated gene is STAT1, STAT2, ZNF606, TRIM37, ACKR3, and/or MAP3K 8.

In some embodiments, the inflammation-associated genes are STAT1 and STAT 2.

In some embodiments, the inflammation-associated gene is STAT 1.

In some embodiments, the inflammation-associated gene is STAT 2.

In some embodiments, the inflammation-associated genes are ZNF606 and TRIM 37.

In some embodiments, the inflammation-associated gene is ZNF 606.

In some embodiments, the inflammation-associated gene is TRIM 37.

In some embodiments, the inflammation-associated genes are ACKR3 and MAP3K 8.

In some embodiments, the inflammation-associated gene is ACKR 3.

In some embodiments, the inflammation-associated gene is MAP3K 8.

In some embodiments, the inflammation-associated gene is "STAT 1," a gene that encodes the protein "signal transducer and activator of transcription 1," a key mediator of cytokine signaling pathways.

In some embodiments, the inflammation-associated gene is "STAT 2," a gene that encodes the protein "signal transducer and activator of transcription 2," a key mediator of cytokine signaling pathways.

In some embodiments, the inflammation-associated gene is "ZNF 606," which is a gene encoding the protein "zinc finger protein 606," which plays a role in the host's response to viral infection.

In some embodiments, the inflammation-associated gene is "TRIM 37," which is a gene encoding the protein "protein 37 containing a tripartite motif," which functions in the MHC class I-mediated antigen presentation pathway.

In some embodiments, the inflammation-associated gene is "MAP 3K8," which is a gene encoding the protein "mitogen-activated protein kinase 8," which is an inducer of nfkb, TNF, IL-2, and TLR4 signaling.

In some embodiments, "ACKR 3" refers to a gene encoding the protein "atypical chemokine receptor 3" that serves as a receptor for CXCL11 and CXCL 12.

In some embodiments, the inflammation-related gene is CRELD1, PARVG, ACAP1, RXRB, COX19, CERS4, B4GALT7, ZNF329, ZFAND2B, NELFB, EMD, UBTF, PYCR2, RNF216, SEC24C, NUMA1, CARD11, EMG1, ZNF576, TRAF2, MAP2K7, CDK4, KHDC4, GIPC1, ILF3, GBP4, FCER1G, STAT1, STAT2, DTX3L, EPSTI1, PARP9, TRIM 9, 140, TRIM 9, PSMB9, MAP3K 9, ACOT9, XRCC 9, KLF 9, pf 9, PRUNE 9, LACTB 36241, ccm 9, nchl 9, trincl 36169, MR 9, or kr 9.

In some embodiments, the inflammation-related gene is PLCB1, EFHC2, RING1, REV1, HIBADH, C2ORF68, PPP2R2A, hadna, eni 2, ZNF671, ERP29, TOB1, NUDT16L1, ZNF329, ZFAND2B, YIPF2, SNUPN, ZNF606, ELAC1, ECI1, HAX1, PFDN6, COQ8 6, GOLGA8 6, TOMM 6, PIK3C2 6, loxgd 6, FAM122 6, IGHD, SYS 6, OR2a 6, IL4 6, GRB 6, RAB 6, MOB3 6, KLHL 6, USF 6, fisbbp 72, CD 6, plak 6, melk 6, OR 3 b 6, kopl 6, and/OR abc 365.

In some embodiments, the inflammation-related gene is KHDC4, PMS2, GIMAP1-GIMAP5, SLC25a25, EML2, ZNF790, VSIG1, AXIN2, DHRS3, TESPA1, RGPD5, SPOUT1, TRAF3IP3, RPL13A, NUDT16L A, ACKR A, TFPT, SPAG A, TOB A, ZFAND 2A, ZNF329, UBTF, HIC A, TRMT61A, ZNF 324A, PRKCE, PLEKHA A, BCL7A, ZNF608, tisess, FCHSD A, SMG A, ATXN A, CNNM A, SIPA1L A, CDKL A, ktu A, ktsu A, gtsn A, tetsn A, bthsd A, spx A, spnf A, spx A, MR A, spx A, spnf A, spx A, spf A, spx A, MR A, spx A, spf A, MR A, spf A, spx A, spf A, or spnf A, spx A, MR A, spf A, spnf A, or spnf A.

In some embodiments, the inflammation-related gene is CRELD1, PARVG, ACAP1, RXRB, COX1, CERS 1, B4GALT 1, ZNF329, ZFNAND 21, NELFB, EMD, UBTF, PYCR 1, RNF216, SEC24 1, NUMA1, CARD1, EMG1, ZNF576, TRAF 1, MAP2K 1, CDK 1, KHDC 1, GIPC1, ZNF 1, GBP 1, FCER 11, STAT 36DTX 3 1, EPSTI1, PARP 1, TRIM1, SP140, TRIM1, PSMB 1, MAP3K 1, ACOT 1, XRCC 1, KLF 1, PF1, PRUNE 1, LABENCP 1, LACTB 1, CTC 1, ZFAND2B, ZNF329, UBTF, HIC2, TRMT61A, ZNF324B, PRKCE, PLEKHA2, BCL7A, ZNF608, TIMELESS, FCHSD2, SMG7, ATXN1, CNNM2, SIPA1L2, CDKL 56 5, TSKU, GGA3, TESK2, BTN2a2, UBXN7, CHP2, MAP3K8, POU5F2, NF1, XRCC2, NME9, KLHL33, MR1, and/or USF 3.

In some embodiments, the inflammation-associated gene is CRELD 1. In some embodiments, the inflammation-associated gene is PARVG. In some embodiments, the inflammation-associated gene is ACAP 1. In some embodiments, the inflammation-associated gene is RXRB. In some embodiments, the inflammation-associated gene is COX 19. In some embodiments, the inflammation-associated gene is CERS 4. In some embodiments, the inflammation-associated gene is B4GALT 7. In some embodiments, the inflammation-associated gene is ZNF 329. In some embodiments, the inflammation-associated gene is ZFAND 2B. In some embodiments, the inflammation-associated gene is NELFB. In some embodiments, the inflammation-associated gene is EMD. In some embodiments, the inflammation-associated gene is UBTF. In some embodiments, the inflammation-associated gene is PYCR 2. In some embodiments, the inflammation-associated gene is RNF 216. In some embodiments, the inflammation-associated gene is SEC 24C. In some embodiments, the inflammation-associated gene is NUMA 1. In some embodiments, the inflammation-associated gene is CARD 11. In some embodiments, the inflammation-associated gene is EMG 1. In some embodiments, the inflammation-associated gene is ZNF 576. In some embodiments, the inflammation-associated gene is TRAF 2. In some embodiments, the inflammation-associated gene is MAP2K 7. In some embodiments, the inflammation-associated gene is CDK 4. In some embodiments, the inflammation-associated gene is KHDC 4. In some embodiments, the inflammation-associated gene is GIPC 1. In some embodiments, the inflammation-associated gene is ILF 3. In some embodiments, the inflammation-associated gene is GBP 4. In some embodiments, the inflammation-associated gene is FCER 1G. In some embodiments, the inflammation-associated gene is STAT 1. In some embodiments, the inflammation-associated gene is STAT 2. In some embodiments, the inflammation-associated gene is DTX 3L. In some embodiments, the inflammation-associated gene is EPSTI 1. In some embodiments, the inflammation-associated gene is PARP 9. In some embodiments, the inflammation-associated gene is TRIM 22. In some embodiments, the inflammation-associated gene is SP 140. In some embodiments, the inflammation-associated gene is TRIM 5. In some embodiments, the inflammation-associated gene is PSMB 9. In some embodiments, the inflammation-associated gene is MAP3K 8. In some embodiments, the inflammation-associated gene is ACOT 9. In some embodiments, the inflammation-associated gene is XRCC 2. In some embodiments, the inflammation-associated gene is KLF 5. In some embodiments, the inflammation-associated gene is NBPF 10. In some embodiments, the inflammation-associated gene is PRUNE 2. In some embodiments, the inflammation-associated gene is LACTB. In some embodiments, the inflammation-associated gene is FAM 241A. In some embodiments, the inflammation-associated gene is CCDC 169. In some embodiments, the inflammation-associated gene is KLHL 33. In some embodiments, the inflammation-associated gene is KDM 1B. In some embodiments, the inflammation-associated gene is FANCL. In some embodiments, the inflammation-associated gene is MR 1. In some embodiments, the inflammation-associated gene is TRIM 13. In some embodiments, the inflammation-associated gene is PLCB 1. In some embodiments, the inflammation-associated gene is EFHC 2. In some embodiments, the inflammation-associated gene is RING 1. In some embodiments, the inflammation-associated gene is REV 1. In some embodiments, the inflammation-associated gene is HIBADH. In some embodiments, the inflammation-associated gene is C2ORF 68. In some embodiments, the inflammation-associated gene is PPP2R 2A. In some embodiments, the inflammation-associated gene is hadoa. In some embodiments, the inflammation-associated gene is ENY 2. In some embodiments, the inflammation-associated gene is ZNF 671. In some embodiments, the inflammation-associated gene is ERP 29. In some embodiments, the inflammation-associated gene is TOB 1. In some embodiments, the inflammation-associated gene is NUDT16L 1. In some embodiments, the inflammation-associated gene is ZNF 329. In some embodiments, the inflammation-associated gene is ZFAND 2B. In some embodiments, the inflammation-associated gene is YIPF 2. In some embodiments, the inflammation-associated gene is SNUPN. In some embodiments, the inflammation-associated gene is ZNF 606. In some embodiments, the inflammation-associated gene is ELAC 1. In some embodiments, the inflammation-associated gene is ECI 1. In some embodiments, the inflammation-associated gene is HAX 1. In some embodiments, the inflammation-associated gene is PFDN 6. In some embodiments, the inflammation-associated gene is COQ 8B. In some embodiments, the inflammation-associated gene is GOLGA 8N. In some embodiments, the inflammation-associated gene is TOMM 7. In some embodiments, the inflammation-associated gene is PIK3C 2B. In some embodiments, the inflammation-associated gene is LOXHD 1. In some embodiments, the inflammation-associated gene is FAM 122C. In some embodiments, the inflammation-associated gene is IGHD. In some embodiments, the inflammation-associated gene is SYS 1. In some embodiments, the inflammation-associated gene is OR2a 42. In some embodiments, the inflammation-associated gene is OR2a 1. In some embodiments, the inflammation-associated gene is IL 4R. In some embodiments, the inflammation-associated gene is GRB 10. In some embodiments, the inflammation-associated gene is RAB 20. In some embodiments, the inflammation-associated gene is MOB 3C. In some embodiments, the inflammation-associated gene is KLHL 33. In some embodiments, the inflammation-associated gene is USF 3. In some embodiments, the inflammation-associated gene is PFFIBP 1. In some embodiments, the inflammation-associated gene is CD 40. In some embodiments, the inflammation-associated gene is PLEKHA 2. In some embodiments, the inflammation-associated gene is ABL 2. In some embodiments, the inflammation-associated gene is PI 3. In some embodiments, the inflammation-associated gene is TIMELESS. In some embodiments, the inflammation-associated gene is CLHC 1. In some embodiments, the inflammation-associated gene is KMT 5A. In some embodiments, the inflammation-associated gene is BCL 7A. In some embodiments, the inflammation-associated gene is HACE 1. In some embodiments, the inflammation-associated gene is TRIM 37. In some embodiments, the inflammation-associated gene is C5ORF 22. In some embodiments, the inflammation-associated gene is KHDC 4. In some embodiments, the inflammation-associated gene is PMS 2. In some embodiments, the inflammation-associated gene is GIMAP1-GIMAP 5. In some embodiments, the inflammation-associated gene is SLC25a 25. In some embodiments, the inflammation-associated gene is EML 2. In some embodiments, the inflammation-associated gene is ZNF 790. In some embodiments, the inflammation-associated gene is VSIG 1. In some embodiments, the inflammation-associated gene is AXIN 2. In some embodiments, the inflammation-associated gene is DHRS 3. In some embodiments, the inflammation-associated gene is ESPA 1. In some embodiments, the inflammation-associated gene is RGPD 5. In some embodiments, the inflammation-associated gene is SPOUT 1. In some embodiments, the inflammation-associated gene is TRAF3IP 3. In some embodiments, the inflammation-associated gene is RPL 13A. In some embodiments, the inflammation-associated gene is NUDT16L 1. In some embodiments, the inflammation-associated gene is ACKR 3. In some embodiments, the inflammation-associated gene is TFPT. In some embodiments, the inflammation-associated gene is SPAG 7. In some embodiments, the inflammation-associated gene is TOB 1. In some embodiments, the inflammation-associated gene is ZFAND 2B. In some embodiments, the inflammation-associated gene is ZNF 329. In some embodiments, the inflammation-associated gene is UBTF. In some embodiments, the inflammation-associated gene is HIC 2. In some embodiments, the inflammation-associated gene is TRMT 61A. In some embodiments, the inflammation-associated gene is ZNF 324B. In some embodiments, the inflammation-associated gene is PRKCE. In some embodiments, the inflammation-associated gene is PLEKHA 2. In some embodiments, the inflammation-associated gene is BCL 7A. In some embodiments, the inflammation-associated gene is ZNF 608. In some embodiments, the inflammation-associated gene is TIMELESS. In some embodiments, the inflammation-associated gene is FCHSD 2. In some embodiments, the inflammation-associated gene is SMG 7. In some embodiments, the inflammation-associated gene is ATXN 1. In some embodiments, the inflammation-associated gene is CNNM 2. In some embodiments, the inflammation-associated gene is SIPA1L 2. In some embodiments, the inflammation-associated gene is CDKL 5. In some embodiments, the inflammation-associated gene is TSKU. In some embodiments, the inflammation-associated gene is GGA 3. In some embodiments, the inflammation-associated gene is TESK 2. In some embodiments, the inflammation-associated gene is BTN2a 2. In some embodiments, the inflammation-associated gene is UBXN 7. In some embodiments, the inflammation-associated gene is CHP 2. In some embodiments, the inflammation-associated gene is MAP3K 8. In some embodiments, the inflammation-associated gene is POU5F 2. In some embodiments, the inflammation-associated gene is NF 1. In some embodiments, the inflammation-associated gene is XRCC 2. In some embodiments, the inflammation-associated gene is NME 9. In some embodiments, the inflammation-associated gene is KLHL 33. In some embodiments, the inflammation-associated gene is MR 1. In some embodiments, the inflammation-associated gene is USF 3.

Accordingly, in some aspects, the present disclosure provides methods for treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent (e.g., RSLV-132), wherein the treatment results in a reduction of one or more inflammation-associated genes. In some aspects, the inflammation-associated gene is IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and/or STAT 5B. In some aspects, the inflammation-associated gene is IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and/or STAT 5B.

Accordingly, in some aspects, the present disclosure provides methods for treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent (e.g., RSLV-132), wherein the treatment results in reduction of one or more inflammation-associated genes and improvement of fatigue. In some aspects, the inflammation-associated gene is IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL-1, IL-17 receptor A, LTBR4, and/or STAT 5B. In some aspects, the inflammation-associated gene is IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and/or STAT 5B.

In some aspects, the disclosure provides methods for treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent (e.g., RSLV-132), wherein the treatment results in an increase in one or more inflammation-associated genes. In some aspects, the inflammation-associated gene is CXCL10(IP-10), CD163, RIPK2, and/or CCR 2.

In some aspects, the disclosure provides methods for treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent (e.g., RSLV-132), wherein the treatment results in an increase in one or more inflammation-associated genes and an improvement in fatigue. In some aspects, the inflammation-associated gene is CXCL10(IP-10), CD163, RIPK2, and/or CCR 2.

In some aspects, the disclosure provides methods for treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent (e.g., RSLV-132), wherein the treatment results in an increase in one or more cytokines. In some aspects, the cytokine is CXCL10 (IP-10).

In some aspects, the disclosure provides methods for treating xerosis in a patient in need thereof, the method comprising administering to the patient an effective amount of an RNA nuclease agent (e.g., RSLV-132), wherein the treatment results in an increase in one or more cytokines and an improvement in fatigue. In some aspects, the cytokine is CXCL10 (IP-10).

In some aspects, the present disclosure provides methods of identifying a subject with xerosis as a candidate for treatment with an RNA nuclease agent by determining an inflammation-associated gene expression profile in a sample obtained from the subject; and comparing the inflammation-associated gene expression profile of the subject with a sample obtained from a suitable control subject, wherein the inflammation-associated gene expression profile indicates that the subject is a candidate for treatment with an RNA nuclease agent. In some aspects, the inflammation-associated genes include MAP3K8, ACKR3, STAT1, STAT2, TRIM37, and/or ZNF 606.

Thus, in some aspects, the disclosure provides methods of identifying a subject with sjogren's syndrome who is likely to respond to treatment with an RNA nuclease agent described herein (e.g., RSLV-132) by detecting the presence of an inflammation-associated molecule (e.g., an inflammation-associated gene) in a sample obtained from the subject, wherein the presence of the inflammation-associated molecule (e.g., the inflammation-associated gene) indicates that the subject is likely to respond to treatment with the agent. In some aspects, the amount or expression level of an inflammation-related molecule (e.g., an inflammation-related gene) in a sample is determined and compared to a reference amount or reference expression level of the inflammation-related molecule (e.g., an inflammation-related gene). In some aspects, when the amount or expression level of an inflammation-associated molecule (e.g., an inflammation-associated gene) in a sample is increased relative to a reference amount or reference expression level of the inflammation-associated molecule (e.g., an inflammation-associated gene), then the patient may be responsive to treatment with an RNA nuclease agent disclosed herein. In some aspects, when the amount or expression level of an inflammation-related molecule (e.g., an inflammation-related gene) in a sample is reduced relative to a reference amount or reference expression level of the inflammation-related molecule (e.g., an inflammation-related gene), then the patient may be responsive to treatment with an RNA nuclease agent disclosed herein.

In some aspects, the disclosure provides a method of identifying a patient who is likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132) described herein, wherein a sample from the patient is contacted with a nucleic acid probe that hybridizes to a complementary target sequence in DNA or RNA of an inflammation-associated molecule (e.g., an inflammation-associated gene) such that a hybridization complex is formed between the nucleic acid probe and the DNA or RNA of the inflammation-associated molecule (e.g., an inflammation-associated gene). In order to detect hybridization of the probe to a target DNA or RNA sequence of an inflammation-related molecule (e.g., an inflammation-related gene), the probe is labeled with a molecular marker; for example, a radioactive label, a fluorescent label, an enzymatic label or digoxigenin. In some aspects, the presence of the probe-target complex indicates that the patient is likely to respond to the treatment. In some aspects, the amount of probe-target complex in the sample is compared to a control. In some aspects, when the amount of probe-target complex in the sample is increased relative to the control, the patient may respond to treatment with an RNA nuclease agent (e.g., RSLV-132). In some aspects, a patient may respond to treatment with an RNA nuclease agent (e.g., RSLV-132) when the amount of probe-target complex in the sample is reduced relative to a control.

In some aspects, the disclosure provides a method of identifying a patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132) described herein, wherein a sample from the patient is contacted with an antibody that specifically binds an inflammation-associated molecule (e.g., an inflammation-associated protein) or an antigen-binding fragment thereof, thereby forming a complex with the inflammation-associated molecule, detecting the presence of the antibody-inflammation-associated molecule complex, wherein the presence of the complex indicates that the patient is likely to respond to treatment. In some aspects, the amount of antibody-inflammation-associated molecule complex in the sample is compared to a control. In some aspects, a patient may respond to treatment with an RNA nuclease agent (e.g., RSLV-132) when the amount of antibody-inflammation-associated molecule complex in the sample is increased relative to a control. In some aspects, a patient may respond to treatment with an RNA nuclease agent (e.g., RSLV-132) when the amount of antibody-inflammation-associated molecule complex in the sample is reduced relative to a control.

Diagnostic method

The present disclosure provides methods related to detecting and/or quantifying one or more inflammation-related molecules (e.g., inflammation-related genes) described herein (e.g., STAT1, STAT2, ZNF606, TRIM37, ACKR3, and/or MAP3K8) in one or more samples, wherein the detection and/or quantification of the one or more inflammation-related molecules, alone or in combination, would indicate the likelihood that an RNA nuclease agent (e.g., RSLV-132) would provide a therapeutic effect or benefit to a patient suffering from xerosis.

Provided herein are methods of identifying a subject with xerosis as a candidate for treatment with an RNA nuclease agent by determining an inflammation-associated gene expression profile in a sample obtained from the subject; and comparing the inflammation-associated gene expression profile of the subject with the inflammation-associated gene expression profile in the sample obtained from the suitable control subject, wherein the inflammation-associated gene expression profile indicates that the subject is a candidate for treatment with an RNA nuclease agent (e.g., RSLV-132). In some aspects, the inflammation-associated genes in the gene expression profile include MAP3K8, ACKR3, STAT1, STAT2, TRIM37, and/or ZNF 606.

Further provided herein are methods of identifying a subject with xerosis as a candidate for treatment with an RNA nuclease agent by determining an inflammation-associated gene expression profile in a sample obtained from the subject; and comparing the inflammation-associated gene expression profile of the subject with xerosis to the inflammation-associated gene expression profile in the sample obtained from a suitable control subject, wherein the inflammation-associated gene expression profile indicates that the subject is a candidate for treatment with an RNA nuclease agent (e.g., RSLV-132) when the amount of the one or more inflammation-associated genes in the sample from the subject with xerosis is equal to or greater than the amount of the one or more inflammation-associated genes in the sample obtained from a suitable control subject. In some embodiments, the inflammation-associated genes in the gene expression profile include ZNF606 and/or ACKR 3.

Further provided herein are methods of identifying a subject with xerosis as a candidate for treatment with an RNA nuclease agent by determining an inflammation-associated gene expression profile in a sample obtained from the subject; and comparing the inflammation-associated gene expression profile of the subject with xerosis to the inflammation-associated gene expression profile in the sample obtained from a suitable control subject, wherein the inflammation-associated gene expression profile indicates that the subject is a candidate for treatment with an RNA nuclease agent (e.g., RSLV-132) when the amount of the one or more inflammation-associated genes in the sample from the subject with xerosis is less than the amount of the one or more inflammation-associated genes in the sample obtained from a suitable control subject. In some embodiments, the inflammation-associated genes in the gene expression profile include STAT1, STAT2, TRIM37, and/or MAP3K 8.

Further provided herein are methods of identifying a subject with xerosis as a candidate for treatment with an RNA nuclease agent by determining an inflammation-related gene expression profile in a sample obtained from the subject, wherein the inflammation-related genes in the profile include MAP3K8, ACKR3, STAT1, STAT2, TRIM37, and/or ZNF 606; and comparing the inflammation-associated gene expression profile of the subject with a subject with xerosis to the inflammation-associated gene expression profile in a sample obtained from a suitable control subject and identifying the subject as a candidate for treatment with an RNA nuclease agent, wherein a) the expression level of ZNF606 in the sample is increased relative to the control; b) increased expression levels of ACKR3 in the sample relative to a control; c) a decrease in the expression level of STAT1 in the sample relative to a control; d) a decrease in the expression level of STAT2 in the sample relative to a control; e) a decrease in the expression level of TRIM37 in the sample relative to a control; f) (ii) a decrease in the expression level of MAP3K8 in the sample relative to a control; or g) any combination of (a), (b), (c), (d), (e) and (f).

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining an amount of one or more inflammation-related molecules (e.g., inflammation-related genes) (e.g., STAT1, STAT2, ZNF606, TRIM37, ACKR3, and/or MAP3K8) in a sample obtained from the patient relative to a reference amount of the inflammation-related molecules (e.g., inflammation-related genes), wherein the amount of the inflammation-related molecules (e.g., inflammation-related genes) in the sample relative to the reference amount of the inflammation-related molecules (e.g., inflammation-related genes) indicates a likelihood that the patient will respond to the treatment.

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the amount of one or more inflammation-associated genes in a sample obtained from the patient; and comparing the amount of the one or more inflammation-associated genes in the sample to a reference amount of the one or more inflammation-associated genes, wherein the patient is likely to be responsive to the treatment when the amount of the one or more inflammation-associated genes in the sample is equal to or greater than the reference amount of the one or more inflammation-associated genes. In some embodiments, the inflammation-associated gene is ZNF606 and/or ACKR 3.

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the amount of one or more inflammation-associated genes in a sample obtained from the patient; and comparing the amount of the one or more inflammation-associated genes in the sample to a reference amount of the one or more inflammation-associated genes, wherein the patient is likely to be responsive to the treatment when the amount of the one or more inflammation-associated genes in the sample is less than the reference amount of the one or more inflammation-associated genes. In some embodiments, the inflammation-associated gene is STAT1, STAT2, TRIM37, and/or MAP3K 8.

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level of a panel of inflammation-associated genes in a patient sample, wherein the panel comprises STAT1, STAT2, ZNF606, TRIM37, ACKR3, and/or MAP3K 8; comparing the expression level of the group in the sample to the expression level of the group in a control, wherein the amount of the inflammation-associated gene in the sample relative to the amount of the inflammation-associated gene in the control is indicative of the likelihood that the patient will respond to the treatment.

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level of a panel of inflammation-associated genes in a patient sample, wherein the panel comprises STAT1, STAT2, ZNF606, TRIM37, ACKR3, and/or MAP3K 8; comparing the expression level of the group in the sample to the expression level of the group in a control and identifying a patient likely to respond to treatment with an RNA nuclease agent, wherein a) the expression level of ZNF606 in the sample is increased relative to the control; b) increased expression levels of ACKR3 in the sample relative to a control; c) a decrease in the expression level of STAT1 in the sample relative to a control; d) a decrease in the expression level of STAT2 in the sample relative to a control; e) a decrease in the expression level of TRIM37 in the sample relative to a control; f) (ii) a decrease in the expression level of MAP3K8 in the sample relative to a control; or g) any combination of (a), (b), (c), (d), (e), and (f).

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: contacting the sample with a nucleic acid probe that hybridizes to a complementary target sequence in the DNA or RNA of an inflammation-related molecule (e.g., an inflammation-related gene), thereby forming a hybridization complex between the nucleic acid probe and the DNA or RNA of the inflammation-related molecule (e.g., an inflammation-related gene); determining the amount of complex in the sample relative to the amount of complex in a reference sample, wherein the amount of complex in the sample relative to the amount of complex in the reference sample indicates the likelihood that the patient is susceptible to treatment with an RNA nuclease agent (e.g., RSLV-132).

Further provided herein are methods of identifying a xerosis patient likely to respond to treatment with an RNA nuclease agent (e.g., RSLV-132), the method comprising: contacting the sample with at least one diagnostic antibody or antigen-binding fragment thereof that specifically binds to an inflammation-associated molecule (e.g., an inflammation-associated gene), thereby forming a diagnostic antibody-inflammation-associated molecule complex; determining the amount of complex in the sample relative to the amount of complex in a reference sample, wherein the amount of complex in the sample relative to the amount of complex in the reference sample indicates the likelihood that the patient is susceptible to treatment with an RNA nuclease agent (e.g., RSLV-132).

Further provided herein are methods for monitoring the response of a xerosis patient treated with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level and/or activity of one or more inflammation-associated molecules (e.g., inflammation-associated genes) in a first sample of the patient obtained prior to treatment with the RNA nuclease agent; determining the expression level and/or activity of one or more inflammation-associated molecules (e.g., inflammation-associated genes) in a second sample from the patient obtained after treatment with the RNA nuclease agent; comparing the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the second sample to the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the first sample, wherein a change in the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the second sample relative to the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the first sample is indicative of a response of the patient being treated with the RNA nuclease agent. In some aspects, the one or more inflammation-associated molecules are inflammation-associated genes. In some aspects, the inflammation-associated gene is IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, STAT5B, CXCL10(IP-10), CD163, RIPK2, and/or CCR 2. In some aspects, the inflammation-associated gene is IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, STAT5B, CXCL10, CD163, RIPK2, and/or CCR 2.

Further provided herein are methods for monitoring the response of a xerosis patient treated with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level and/or activity of one or more inflammation-associated molecules (e.g., inflammation-associated genes) in a first sample of the patient obtained prior to treatment with the RNA nuclease agent; determining the expression level and/or activity of one or more inflammation-associated molecules (e.g., inflammation-associated genes) in a second sample from the patient obtained after treatment with the RNA nuclease agent; comparing the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the second sample to the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the first sample, wherein a decrease in the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the second sample relative to the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the first sample is indicative of a response of the patient being treated with the RNA nuclease agent. In some aspects, the one or more inflammation-associated molecules are inflammation-associated genes. In some aspects, the inflammation-associated gene is IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, and/or STAT 5B. In some aspects, the inflammation-associated gene is IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, and/or STAT 5B.

Further provided herein are methods for monitoring the response of a xerosis patient treated with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level and/or activity of one or more inflammation-associated molecules (e.g., inflammation-associated genes) in a first sample of the patient obtained prior to treatment with the RNA nuclease agent; determining the expression level and/or activity of one or more inflammation-associated molecules (e.g., inflammation-associated genes) in a second sample from the patient obtained after treatment with the RNA nuclease agent; comparing the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the second sample to the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the first sample, wherein an increase in the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the second sample relative to the level of expression and/or activity of the one or more inflammation-associated molecules (e.g., inflammation-associated genes) in the first sample is indicative of a response of the patient being treated with the RNA nuclease agent. In some aspects, the one or more inflammation-associated molecules are inflammation-associated genes. In some aspects, the inflammation-associated gene is CXCL10(IP-10), CD163, RIPK2, and/or CCR 2.

Further provided herein are methods for monitoring the response of a xerosis patient treated with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level and/or activity of one or more cytokines in a first sample of the patient obtained prior to treatment with the RNA nuclease agent; determining the expression level and/or activity of one or more cytokines in a second sample from the patient obtained after treatment with the RNA nuclease agent; comparing the expression level and/or activity of the one or more cytokines in the second sample to the expression level and/or activity of the one or more cytokines in the first sample, wherein an increase in the expression level and/or activity of the one or more cytokines in the second sample relative to the expression level and/or activity of the one or more cytokines in the first sample is indicative of a response of the patient being treated with the RNA nuclease agent. In some aspects, the cytokine is CXCL10 (IP-10).

Further provided herein are methods for monitoring the response of a xerosis patient treated with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level and/or activity of one or more cytokines in a first sample of the patient obtained prior to treatment with the RNA nuclease agent; determining the expression level and/or activity of one or more cytokines in a second sample from the patient obtained after treatment with the RNA nuclease agent; comparing the expression level and/or activity of the one or more cytokines in the second sample to the expression level and/or activity of the one or more cytokines in the first sample, wherein a decrease in the expression level and/or activity of the one or more cytokines in the second sample relative to the expression level and/or activity of the one or more cytokines in the first sample is indicative of a response of the patient being treated with the RNA nuclease agent.

Further provided herein are methods for monitoring the response of a xerosis patient treated with an RNA nuclease agent (e.g., RSLV-132), the method comprising: determining the expression level and/or activity of one or more cytokines in a first sample of the patient obtained prior to treatment with the RNA nuclease agent; determining the expression level and/or activity of one or more cytokines in a second sample from the patient obtained after treatment with the RNA nuclease agent; comparing the expression level and/or activity of the one or more cytokines in the second sample to the expression level and/or activity of the one or more cytokines in the first sample, wherein a change in the expression level and/or activity of the one or more cytokines in the second sample relative to the expression level and/or activity of the one or more cytokines in the first sample is indicative of a response of the patient being treated with the RNA nuclease agent. In some aspects, the cytokine is CXCL10 (IP-10).

Determination of inflammation-associated molecules

The presence or amount or level of expression of one or more of the inflammation-associated molecules described herein (e.g., an inflammation-associated gene, an inflammation-associated protein, a proinflammatory molecule such as a proinflammatory gene or a proinflammatory protein) in a sample can be detected or determined by a variety of methods and techniques that detect those that are known in the art and understood by the skilled artisan, including but not limited to Immunohistochemistry (IHC), Immunofluorescence (IF), western blot analysis, immunoprecipitation, molecular binding assays, enzyme-linked immunosorbent assay (ELISA), enzyme-linked immunosorbent assay (ELIFA), flow cytometry, MassARRAY, proteomics, blood-based quantitative assays (e.g., serum ELISA), biochemical enzyme activity assays, in situ hybridization, Fluorescence In Situ Hybridization (FISH), Southern analysis, Northern analysis, whole genome sequencing, Polymerase Chain Reaction (PCR) including quantitative real-time PCR (qRT-PCR) and other amplification-type detection methods (e.g., branched DNA, PCR, mass spectrometry, and the like, SISBA, TMA, etc.), RNA-Seq, microarray analysis, gene expression profiling and/or Serial Analysis of Gene Expression (SAGE), and any of a variety of analyses that can be performed by protein, gene, and/or tissue array analysis. Typical Protocols for assessing the status of genes and gene products are found, for example, In Ausubel et al, eds.,1995, Current Protocols In Molecular Biology, Units 2(Northern Blotting),4(Southern Blotting),15 (immunology) and 18(PCR Analysis). Multiplex immunoassays such as those available from rule-based medical or mesoscale discovery ("MSD") may also be used. Diagnostic antibodies that bind the inflammation-associated molecules of the present disclosure are available from a variety of commercial sources, such as BD Biosciences, ebiosciences, BioLegend, Abcam, and the like.

Nucleic acid inflammation related molecular technology

In some embodiments, the expression level of an inflammation-related molecule (e.g., an inflammation-related gene) described herein can be a nucleic acid expression level. In some embodiments, the nucleic acid expression level is determined using qPCR, rtPCR, RNA-Seq, multiplex qPCR or RT-qPCR, microarray analysis, gene expression profiling, SAGE, MassARRAY techniques, or in situ hybridization (e.g., FISH). In some embodiments, the expression level of an inflammation-related molecule (e.g., an inflammation-related gene) is determined in a cell from a xerosis patient. In some embodiments, the expression level of an inflammation-related molecule (e.g., an inflammation-related gene) described herein is determined in blood cells from a xerosis patient.

Methods for assessing mRNA in cells are known in the art and include, for example, hybridization assays using complementary DNA probes (e.g., in situ hybridization using labeled riboprobes specific for one or more genes, Northern blotting, and related techniques) and various nucleic acid amplification assays (e.g., RT-PCR using complementary primers specific for one or more genes, as well as other amplification type detection methods, such as branched DNA, SISBA, TMA, and the like). In addition, such methods can include one or more steps that allow for the determination of the level of a target mRNA in a biological sample (e.g., by simultaneously examining the level of a comparative control mRNA sequence of a "housekeeping" gene, such as an actin family member).

Nucleic acid sequences of inflammation-associated molecules (e.g., inflammation-associated genes, pro-inflammatory genes) of the present disclosure are known in the art. In some embodiments, the inflammation-related molecules (e.g., inflammation-related genes) of the present disclosure include IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, STAT5B, CXCL10(IP-10), CD163, RIPK2, and/or CCR 2. In some embodiments, an inflammation-related molecule of the present disclosure (e.g., an inflammation-related gene) comprises IL5, TNFRSF1A, IL6R, IL1RAP, CXCL1, IL17RA, LTB4R, STAT5B, CXCL10, CD163, RIPK2, and/or CCR 2. In some embodiments, the inflammation-related molecules (e.g., inflammation-related genes) of the present disclosure include STAT1, STAT2, ZNF606, TRIM37, ACKR3, and/or MAP3K 8.

In some embodiments, the sequence of the amplified cDNA of interest may be determined. The methods include protocols for examining or detecting mRNA (e.g., mRNA of interest) in a tissue or cell sample by microarray technology. Using nucleic acid microarrays, test and control mRNA samples from test and control tissue samples are reverse transcribed and labeled to generate cDNA probes. The probes are then hybridized to an array of nucleic acids immobilized on a solid support. The array is configured such that the order and location of each member of the array is known. For example, selection of genes whose expression correlates with increased or decreased clinical benefit of a treatment comprising an RNA nuclease agent (e.g., RSLV-132) can be arrayed on a solid support. Hybridization of a labeled probe to a particular array member indicates that the sample derived from that probe expresses that gene.

Any of the diagnostic methods described herein are included as part of any method involving methods for identifying patients who may benefit from treatment described herein (e.g., selecting treatment or intervention), or involving the development of treatment (e.g., recruiting patients in a clinical trial) that is advantageous over those methods that do not include a diagnostic method, as a population of patients whose members are expected to require and/or do not require, benefit, or respond to treatment can be determined.

Thus, in some embodiments, an inflammation-related molecule (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) suitable for use in the present disclosure is a diagnostic inflammation-related molecule (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)). In some embodiments, the inflammation-associated molecule (e.g., an inflammation-associated gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) is a monitoring inflammation-associated molecule (e.g., an inflammation-associated gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)). In some embodiments, the inflammation-associated molecule (e.g., an inflammation-associated gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) is a predictive inflammation-associated molecule (e.g., an inflammation-associated gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)).

An inflammation-related molecule (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) can be a substance or biological event whose detection is indicative of a particular physiological state (e.g., a disease state). For example, the presence of an inflammation-related gene in the serum of a patient may be indicative of a disease (e.g., sjogren's syndrome). Inflammation-related molecules (e.g., inflammation-related genes or pro-inflammatory molecules (e.g., pro-inflammatory genes)) measured in a patient prior to treatment can be used to determine patients suitable for inclusion in a clinical trial. Changes in inflammation-associated molecules (e.g., inflammation-associated genes or pro-inflammatory molecules (e.g., pro-inflammatory genes)) after treatment can predict or identify safety issues associated with drug candidates, or reveal pharmacological activities that are expected to predict the ultimate benefit of treatment. Inflammation-related molecules (e.g., inflammation-related genes or pro-inflammatory molecules (e.g., pro-inflammatory genes)) can reduce uncertainty in drug development and evaluation by providing quantifiable predictions about drug performance, and they can aid in dose selection. A composite inflammation-related molecule (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) includes several individual inflammation-related molecules (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) in a prescribed algorithm that achieves a single interpretation reading (single interpretation) when a single inflammation-related molecule (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) fails to provide all of the relevant information needed for evaluation.

The surrogate endpoint is an inflammation-related molecule (e.g., an inflammation-related gene or a pro-inflammatory molecule (e.g., a pro-inflammatory gene)) that is intended to replace a clinical endpoint and is expected to predict clinical benefit based on epidemiological, therapeutic, pathophysiological, or other scientific evidence.

Protein inflammation related molecular technology

In some embodiments, the amount of an inflammation-associated molecule (e.g., an inflammation-associated protein) is measured by determining the protein expression level of the inflammation-associated molecule (e.g., an inflammation-associated protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)). There are many techniques known in the art and described herein for measuring or determining the level of protein expression that can be used in the methods provided by the present disclosure. For example, in some embodiments, the protein expression level of an inflammation-associated molecule (e.g., an inflammation-associated protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)) is selected from flow cytometry (e.g., Fluorescence Activated Cell Sorting (FACS))TM) Western blot, enzyme linked immunosorbent assay (ELISA), immunoprecipitation, Immunohistochemistry (IHC), immunofluorescence, radioimmunoassay, dot blot, immunodetection methods, HPLC, surface plasmon resonance, spectroscopy, mass spectrometry, and HPLC.

In some embodiments, a sample is contacted with an antibody that specifically binds an inflammation-related molecule described herein (e.g., an inflammation-related protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)) under conditions that allow binding to the inflammation-related molecule (e.g., an inflammation-related protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)), and the presence of a complex formed by the antibody and the inflammation-related molecule (e.g., an inflammation-related protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)) is detected. In some embodiments, the sample is contacted with a combination of antibodies that specifically bind to a combination of inflammation-associated molecules described herein (e.g., an inflammation-associated protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)). In some embodiments, the protein expression level of an inflammation-associated molecule (e.g., an inflammation-associated protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)) is determined in cells from a patient with xerosis. In some embodiments, the protein expression level of an inflammation-associated molecule (e.g., an inflammation-associated protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)) is determined in blood cells from a xerosis patient.

In some embodiments, the amount of an inflammation-related molecule (e.g., an inflammation-related protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)) in a sample is determined using a diagnostic antibody that binds to the inflammation-related molecule (e.g., the inflammation-related protein or the pro-inflammatory molecule (e.g., a pro-inflammatory protein)). In some embodiments, the anti-inflammatory-related molecular diagnostic antibody specifically binds to an inflammation-related molecule (e.g., an inflammation-related protein or a pro-inflammatory molecule (e.g., a pro-inflammatory protein)). In some embodiments, the diagnostic antibody is a non-human antibody. In some embodiments, the diagnostic antibody is a rat, mouse, or rabbit antibody. In some embodiments, the diagnostic antibody is a monoclonal antibody. In some embodiments, the diagnostic antibody is directly labeled. In other embodiments, the diagnostic antibody is indirectly labeled.

Reagent kit

The present disclosure provides kits comprising the RNase-containing nuclease fusion proteins of the present disclosure, including an RNase-Fc fusion protein and instructions for use. The kit may comprise, in a suitable container, the RNase-Fc fusion protein disclosed herein, one or more controls, as well as various buffers, reagents, enzymes, and other standard components well known in the art. In some embodiments, the kit comprises an injectable solution comprising an RNase-Fc fusion protein and one or more pharmaceutically acceptable carriers and/or diluents. In some embodiments, the injectable solution is prepared for intravenous administration. In some embodiments, the kit comprises instructions for use.

The container may comprise at least one vial, well, tube, flask, bottle, syringe, or other container means into which the RNase-Fc fusion protein may be placed and, in some cases, suitably aliquoted. Where additional components are provided, the kit may comprise additional containers in which the components may be placed. The kit may also include means for containing the RNase-Fc fusion protein and any other closed reagent containers for commercial sale. Such containers may include injection or blow molded plastic containers in which the desired vials are retained. The container and/or kit may include a label with instructions and/or warnings for use.

Examples

The following are examples of specific embodiments for carrying out the invention. The examples are provided for illustrative purposes only and are not intended to limit the scope of the present invention in any way. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental error and deviation should, of course, be allowed for.

The practice of the present invention will employ, unless otherwise indicated, conventional methods of protein chemistry, biochemistry, recombinant DNA technology and pharmacology within the skill of the art. These techniques are explained fully in the literature. See, e.g., T.E.Creighton, Proteins: Structures and Molecular Properties (W.H.Freeman and Company, 1993); l. leininger, Biochemistry (Worth Publishers, inc., current addition); sambrook, et al, Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); methods In Enzymology (s.Colowick and n.kaplan eds., Academic Press, Inc.); remington's Pharmaceutical Sciences,18th Edition (Easton, Pennsylvania: Mack Publishing Company, 1990); carey and Sundberg Advanced Organic Chemistry 3rd Ed.(Plenum Press)Vols A and B(l992)。

Example 1

Production of RNase-Fc fusion protein-encoding expression vector

Various embodiments of RNase-containing nuclease fusion proteins of the present disclosure are presented in the sequence listing (table 1). An exemplary RNase-Fc fusion protein, RSLV-132, was constructed, the structure of which is shown in FIG. 1. Specifically, polynucleotides encoding RNase-Fc fusion proteins were synthesized directly, starting from the amino acid sequence of the RNase-Fc fusion protein, using codon optimization of Genescript (Genescript, Piscatawy, n.j.) to achieve optimal expression in mammalian cells. The optimization process involves, for example, avoiding regions of very high (> 80%) or very low (< 30%) GC content as much as possible, and avoiding cis-acting sequence motifs such as internal TATA boxes, chi sites and ribosome entry sites, AT-rich or GC-rich sequence extensions, RNA instability motifs, repeats and RNA secondary structures, as well as cryptic splice donor and acceptor sites in higher eukaryotes. The DNA encoding the RNase-Fc fusion protein was cloned into pcDNA3.1+ mammalian expression vector. RSLV-132 is an RNase-Fc fusion protein having the following configuration (FIG. 1), producing RSLV-132.

RSLV-132 is a homodimer comprising two polypeptides, each having the amino acid sequence shown in SEQ ID NO: 50. Each polypeptide of the homodimer has an RNase-Fc configuration in which the wild-type human RNase 1 domain (SEQ ID NO:2) is operably coupled, without a linker, to the N-terminus of the human IgGl Fc domain comprising the SCC hinge and the CH2 mutations P238S and P331S (SEQ ID NO: 22).

Example 2

Stable mammalian cell lines transiently expressing and expressing RNase-Fc fusion proteins

For transient expression, FreeStyle is usedTMMAX reagent expression vectors containing RNase-Fc fusion protein inserts from example 1 were transiently transfected into Chinese Hamster Ovary (CHO) cells, e.g., CHO-S cells (e.g., FreeStyle) using transfection protocols recommended by the manufacturerTMCHO-S cells, Invitrogen). CHO-S cells were stored in FreeStyle containing 2mM L-glutamine and penicillin-streptomycinTMCHO expression medium.

Stable CHO-S cell lines expressing RNase-Fc fusion proteins were generated using conventional methods known in the art. For example, CHO-S cells can be used containing RNAsNucleic acid sequences of e-Fc fusion proteins and nucleic acid sequences encoding a marker (e.g., GFP, a surface marker selectable by magnetic beads) selected for, e.g., flow cytometry or magnetic bead isolation (e.g., MACSelect)TMA system). Alternatively, any transfection method known in the art may be used (e.g., electroporation (Lonza) or FreeStyle as mentioned above)TMMAX reagent) CHO-S cells are transfected with a vector comprising the nucleic acid sequence of the RNase-Fc fusion protein and a selectable marker and then selected using, for example, flow cytometry. The selectable marker may be incorporated into the same vector as the vector encoding the RNase-Fc fusion protein or a separate vector.

The RNase-Fc fusion protein is purified from the culture supernatant by capturing the molecules using a column packed with protein a agarose beads, followed by washing in a column wash buffer (e.g., 90mM Tris, 150mM NaCl, 0.05% sodium azide) and releasing the molecules from the column using a suitable elution buffer (e.g., 0.1M citrate buffer, pH 3.0). The eluted material was further concentrated by buffer exchange using a Centricon concentrator with continuous rotation in PBS, then filtered through a 0.2 μm filter set. The concentration of the RNase-Fc fusion protein is determined using standard spectrophotometry (e.g., Bradford, BCA, Lowry, Biuret assay).

Example 3

Study design and patient characteristics

A multicenter, double-blind placebo-controlled study was conducted to evaluate the effect of 8 intravenous infusions of RSLV-132 on 28 patients with primary sicca syndrome (pSS). According to the american consensus group (AEGC) 2002, study participants aged between 18 and 85 years and were diagnosed with primary sjogren's syndrome. In particular, the study was conducted in a subset of xerosis patients who had elevated anti-Ro 52/60 autoantibody levels and elevated interferon-stimulated gene expression levels in blood cells (e.g., positive interferon character at screening). Subjects were asked to maintain a stable combination of drugs (comitant mediations) for 30 days prior to baseline visit. Hydroxychloroquine is disabled within 30 days of baseline; belimumab, abelep or TNF inhibitors were contraindicated within 90 days of baseline; or cyclophosphamide or rituximab is contraindicated within 180 days of baseline. It is further required that the patient has not previously had a head and neck radiotherapy, lymphoma, graft versus host disease or IgG4 related disease. Potential subjects were screened to assess their eligibility to enter the study within 60 days prior to study entry (i.e., prior to baseline visit). Thirty subjects were screened and randomized into the study. Both subjects gave consent back prior to receiving study treatment. 28 subjects participated in this randomized, double-blind, placebo-controlled phase 2 study (clinicaltirials. gov: NCT 03247686). Baseline assessments were performed on study day 1. Following baseline assessment, patients received a first infusion of RSLV-132 or placebo.

Each subject was randomized into 3:1 (active drug: placebo) groups and received 8 infusions of 10mg/kg RSLV-132 or placebo at baseline, once a week for two weeks (three doses), and then once every two weeks for the next 10 weeks of the study (i.e., intravenous infusions on days 1 (baseline), 8, 15, 29, 43, 57, 71 and 85). Patient reports measured by EESPRI, FACIT and fatigue Profile (PROF) were used to assess the active versus control groups by comparing the baseline and day 99 of the study. Patient reported results were measured on days 1, 29, 57, 85, and 99 (or end of treatment) prior to receiving the daily dose. Efficacy endpoints were measured on day 99 and safety follow-up was performed on days 141, 176 and 211.

RSLV-132 was present at a concentration of 9.5mg/mL in a disposable vial containing 5.3mL of a sterile preservative-free solution, including buffer for dilution by intravenous infusion. A 0.9% sodium chloride solution was used as a placebo infusion.

The study was conducted in accordance with the principles of the Helsinki declaration and the International conference on harmonization (ICH) drug clinical trial administration Specification (GCP) guidelines. Approval by the ethics committee and the institutional review committee was obtained and written informed consent was provided by all patients.

Example 4

Assessment of patient characteristics at baseline

To assess differences between treatment groups (i.e., placebo-treated patients compared to RSLV-132-treated patients) and establish baseline levels for analysis, demographic and disease characteristics were obtained prior to treatment. Baseline patient profile analysis included analysis of complement C3 and complement C4 levels, IgG levels (mg/dL), ESR levels, ESSDAI score, ESSPRI score, FACIT score, and fatigue profile (ProF).

Complement C3 and complement C4(C3/C4) measurements were used to assess activation of the immune system. The measurement of C3/C4 in blood was used as a reading of immunological activity. Blood tests measure specific complement proteins (C3 or C4) and are reported in milligrams per deciliter. Low levels of C3/C4 in the blood may be indicative of disease or autoimmunity.

Immunoglobulin g (igg) comprises about 80% of serum immunoglobulins. Measuring IgG levels in blood samples may be indicative of a disease state. The amount of IgG in blood is typically reported in milligrams per deciliter.

Erythrocyte Sedimentation Rate (ESR) was used as a screen for in vivo inflammation. Typically, erythrocytes settle more rapidly in certain disease states due to an increase in plasma fibrinogen, immunoglobulins, and other acute phase response proteins. Changes in the shape or number of red blood cells may also affect the ESR. Anticoagulated whole blood is placed in a narrow vertical tube and red blood cells are precipitated from the plasma by gravity. Their rate of sedimentation is measured in millimeters (mm/hr) of clear plasma that appears at the top of the column after one hour.

The European Association of antirheumatics (EULAR) Sicca Syndrome (SS) disease Activity index (ESSDAI) was developed as a homogeneous assessment of systemic activity (Seror et al, Ann Rheum Dis.2010; 69(6): 1103-1109). ESSDAI includes 12 fields (i.e. organ system: skin, respiration, kidney, joint, muscle, peripheral nervous system, central nervous system, hematology, gland, constitutional lymphadenopathy, biology). Each domain is divided into 3-4 activity levels. The definition of each activity level is provided by a detailed description of what should be considered in the field. The possible score ranges between 0-123, with about 80% of patients scoring ≦ 13.

Baseline demographics, disease characteristics, and biochemical data were similar between treatment groups (table 2). In particular, the study population had mild to moderate disease as determined by the ESSDAI score and high disease activity as determined by the ESSPRI score. The study subjects also reported extreme fatigue. The ESSDAI and ESSPRI scores were slightly higher in the placebo group than in the RSLV-132 group. Complement 3, complement 4, ESR and IgG measurements were comparable to healthy values and were similar between the two groups (table 2).

Table 2 study demographics and mean clinical characteristics at baseline

Example 5

Clinically significant improvement in ESSPRI score and ESSPRI fatigue in patients receiving RSLV-132 treatment

pSS is an autoimmune disease characterized by lymphatic infiltration of the salivary and lacrimal glands with subsequent inflammation, gland damage and loss of function leading to dry eyes and dry mouth. The clinical features of pSS can be divided into two groups: (1) can be disabling and affect benign symptoms such as dryness, pain and fatigue in most patients; and (2) potentially severe systemic manifestations affecting 20-40% of patients (Seror et al. Ann Rheum Dis 2011; 70: 968-.

The EULAR SS patient reporting index (ESSPRI) is intended to assess symptoms in patients with primary sjogren's syndrome and has been validated and accepted by the FDA. ESSPRI evaluates patients for dryness, pain, and fatigue, and evaluates each symptom on a numerical scale of 0-10. A reduction in the espri score of at least 1 point is clinically significant.

The ESSPRI score of patients receiving RSLV-132 treatment dropped more than 1 point over the course of the study (FIG. 2). Specifically, the ESSPRI score for the patient receiving RSLV-132 dropped from about 6 points at baseline to about 4.5 points at day 99 (FIG. 2), with a change from baseline of-1.20 for the patient (FIG. 3 and Table 3). This improvement in the ESSPRI score is of clinical significance. In contrast, as shown in figure 3 and table 3, the change from baseline in patients receiving placebo treatment was-0.54. Furthermore, ESSPRI fatigue in patients receiving RSLV-132 treatment improved by approximately-1.4 from baseline to day 99 of the study compared to 0 in the placebo group (FIG. 4). When three components of the ESSPRI score were evaluated separately, there was a reduction in the incidence of dryness-related fatigue in patients treated with RSLV-132 compared to placebo-treated control group patients (FIGS. 5A-5C). Subject level data showed that 25% of subjects in the placebo group and 55% or RSLV-132 treated subjects had minimal clinically significant improvement (MCII) in ESSPRI (reduction ≧ 1 point) (table 3). These data provide evidence that RSLV-132 treatment improved symptoms in pSS patients and had clinically significant fatigue reduction.

TABLE 3 measurement of clinical efficacy at day 99

Example 6

Clinically significant improvement in FACIT fatigue score in patients receiving RSLV-132 treatment

The chronic disease test Functional Assessment (FACIT) fatigue scale is used to measure the degree of fatigue in chronic disease and is widely used in Sjogren's syndrome patients. The FACIT fatigue questionnaire includes 13 questions about fatigue, which are measured on the 4-point Likert scale (Likert scale). The total score ranged from 0 to 52, with higher scores representing less fatigue (Chandran et al, Ann Rheum Dis 2007; 66:936- & 939).

As shown in FIG. 6, there was a clinically significant improvement in FACIT fatigue score in patients receiving RSLV-132 treatment. In particular, the FACIT score of RSLV-132 treated patients increased by about 6 points between baseline and day 57 of the study. In contrast, on study day 57, the FACIT score of patients receiving placebo treatment increased by approximately 1 point. On study day 99, the FACIT score for patients receiving RSLV-132 treatment increased by approximately 6 points from baseline (mean 5.9 points increase). In contrast, on study day 99, the FACIT score of patients receiving placebo treatment increased by approximately 1 point from baseline (mean increase of 1.13). The subject level data showed that 25% of placebo subjects and 45% of RSLV-132 treated subjects had the least clinically significant improvement (MCII) (increase ≧ 6 points) in the FACIT score (Table 3). These data provide evidence that treatment with RSLV-132 improved fatigue in pSS patients.

Example 7

Fatigue Profile (PROF) improvement in patients receiving RSLV-132 treatment

Fatigue profile (ProF) is used to measure fatigue associated with chronic disease and has been used to assess fatigue in patients with sjogren's syndrome. Fatigue profile consists of 16 items, divided into two areas: (1) physical fatigue and (2) mental fatigue. Fatigue profile scores ranged from 0 to 7, with higher scores representing more fatigue. The score may be displayed as a profile or as a calculated total score (Strombeck et al, Scand J Rheumatotol 2005; 34: 455-.

The RSLV-132 treated patients experienced an improvement in fatigue profile during the study. Specifically, the fatigue profile score for RSLV-132 treated patients decreased by more than 1 point from baseline to day 99 of the study (average decrease of 1.04 point) (fig. 7). In contrast, the fatigue profile of patients receiving placebo treatment was not reduced, with a mean reduction of 0.02 points (fig. 7).

Notably, during the course of the study, patients receiving RSLV-132 treatment experienced a clinically significant improvement in the mental program of fatigue profile, as mental scores dropped by about 1.5 points from baseline to day 99 of the study (mean drop in mental fatigue program of 1.53 points) (fig. 8). In contrast, patients receiving placebo treatment had a mean decrease in fatigue profile score of 0.06 points (fig. 8). As shown in Table 3, mental fatigue response (reduction ≧ 1 point) was observed in 25% of placebo patients and 55% of RSLV-132-treated patients on day 99. These data provide evidence that the active group (patients receiving RSLV-132 treatment) experienced a clinically significant improvement in the mental component of fatigue profile (score ≧ 1 reduction).

Patients receiving RSLV-132 treatment also improved in the somatic program of fatigue profile, as the somatic score decreased by about 0.8 points from baseline during the study (fig. 9). As shown in Table 3, physical fatigue response (reduction ≧ 1 point) was observed in 25% of placebo patients and 50% of RSLV-132 treated patients on day 99. Neither the mental nor physical items of fatigue profile were reduced in patients receiving placebo treatment. These data provide evidence that treatment with RSLV-132 improved fatigue in pSS patients.

Example 8

Statistically significant improvement in DSST in patients receiving RSLV-132 treatment

The Digital Symbol Substitution Test (DSST) is used to measure cognitive functions (e.g., attention and concentration) in patients with sjogren's syndrome. DSST is a highly validated, sensitive instrument widely used in clinical studies involving CNS drugs as a readout of executive function. DSST is a time-limited paper-pen cognitive test. The test requires the patient to match a symbol to a number according to a key on top of the paper. The patient copies the symbol into a space below a row of numbers and calculates the number of correct symbols within the allowable time.

A portion of 12 patients were tested for numeric symbol replacement (DSST). The results of the DSST neuropsychological tests support the above-mentioned fatigue results. Patients received DSST at baseline and at follow-up (day 99). The total number of symbols matching the number completed in 90 seconds and the time (in seconds) to complete the test are measured. An increase in the number of matches completed within a specified time indicates an improvement. A reduction in the time to complete the test also indicates an improvement. Notably, there was a statistically significant improvement in the time to complete the test between baseline and follow-up in patients receiving RSLV-132 treatment compared to change-2.80 in patients receiving placebo treatment, with a change of 16.40 (fig. 10A). As shown in FIG. 10B, RSLV-132 patients completed the task 16.40 seconds faster than baseline (lost 16.4 seconds), while placebo patients completed the task 2.80 seconds slower than the original time at baseline (increased 2.80 seconds). Patients receiving RSLV-132 treatment also showed an improvement in the number of matches completed within 90 seconds between baseline and follow-up (fig. 10A). Improvements in the DSST test support the discovery of reduced fatigue in patients with sjogren's syndrome, as reduced fatigue corresponds to improved cognitive ability.

Example 9

Responders to RSLV-132 express key inflammatory genes

Gene expression analysis was performed to determine biochemical evidence of reduced inflammation in Sjogren syndrome patients treated with RSLV-132 and was performed as follows.

At Q2Solutions | EA Genomics in Morrisville, NC RNA sequencing of whole blood samples. On days 1 and 99 before study treatmentWhole blood was collected in a collection tube. RNA was extracted and quantified spectrophotometrically using NanoDrop 8000 from Thermo-Fisher, and integrity was assessed on a Bioanalyzer 2100 using RNA 6000Nano Assay. rRNA was consumed using the Illumina TruSeq Stranded Total RNA protocol and RiboZero Magnetic Gold to generate 50 base pair, chain and paired-end sequencing libraries. The library was sequenced on an Illumina HiSeq to a target depth of 5000 ten thousand reads (reads). Before mapping (mapping), an adapter trimming (homopolymer filtering), homopolymer filtering and low quality reading filtering are performed. The processed reads were then mapped to the hg19 genome using STAR v 2.4. Gene and transcript quantification was performed using RSEM 1.2.14.

The results of the gene expression analysis provided biochemical evidence of reduced inflammation in patients who responded clinically to RSLV-132 with RSLV-132 treatment (FIG. 11). These patients showed a widespread reduction in key inflammatory pathways, which was not observed in RSLV-132 treated patients who did not reach clinical response. Clinical response was defined as patients who experienced minimal clinically significant improvement (MCII) in two of the three tools; ESSPRI (reduction by 1 or more), FACIT (increase by 6 or more) or fatigue profile (reduction by 1 or more). Using these criteria, clinical responses were seen in RSLV-132 group 9/20 (45%) and placebo group 2/8 (25%).

Changes in gene expression at day 99 were compared to day 1 for RSLV-132 subjects who reached or did not reach clinical response. Whole blood was collected from 7 patients in the non-responsive group and 7 patients in the responsive group, and gene expression analysis was performed using RNAseq according to the above experimental protocol. FIG. 11 the genes shown in the heatmap (heat map) are involved in regulating proactivelyThe key inflammatory genes of the innate immune system are highly correlated with the outcome of the FACIT tool (R)2>0.6). RSLV-132 treated patients who achieved clinical response showed a broad decrease in inflammation-associated gene expression and were not observed in subjects who did not achieve clinical response (fig. 11). It was observed that expression of key inflammatory genes (e.g., IL-5, TNF receptor, IL-6 receptor, IL-1 accessory protein, CXCL1, IL-17 receptor A, LTBR4, and STAT5B) was reduced in patients treated with RSLV-132 who experienced a clinical response, but not in patients who did not develop a clinical response. Increases in other genes such as CXCL10(IP-10), CD163, RIPK2 and CCR2 were also observed in those patients who achieved clinical responses. Two subjects in the placebo group experienced clinical responses but did not have similar gene expression profiles to those of the RSLV-132 treatment responder (data not shown).

These data provide evidence that subjects treated with RSLV-132 that achieved a clinical response show reduced expression of inflammation-associated genes.

Example 10

Responders to RSLV-132 showed different gene expression profiles

Examination of gene expression profiles to determine gene expression "fingerprints" can be used to identify patients who are likely to respond to treatment with RSLV-132. A different gene expression pattern was observed between the baseline gene expression profiles (prior to study drug administration) of RSLV-132 treated subjects who developed a clinical response (MCII) on the subsequent day 99 compared to the baseline gene expression profile of RSLV-132 treated subjects who did not develop a clinical response.

RNAseq was performed on blood samples from patients at baseline (RSLV-132 pre-administration) as described in example 9. Baseline gene expression profiles were analyzed for non-responders (patients treated with RSLV-132 showed no clinical response) and responders (patients treated with RSLV-132 showed clinical response). Examining the gene expression profiles of a subset of responders versus non-responders RSLV-132 revealed interesting profiles among responders. As shown in figures 12A-12C and table 4, different gene expression profiles were observed on day 1 prior to study drug administration in patients with a positive clinical response on day 99 of the subsequent study.

When baseline geneSpecific profiles were revealed in RSLV-132 responders when expression was correlated with FACIT (fig. 12A), ProF (fig. 12B), or espfri (fig. 12C). The reduction of STAT1 and STAT2 expression was associated with the FACIT test (fig. 12A), the increase of ZNF606 expression and the reduction of TRIM37 expression were associated with the ProF test (fig. 12B), and the increase of ACKR3 expression and the reduction of MAPK3K8 expression were associated with the espfri test (fig. 12C). As shown in Table 4, MAP3K8 and ACKR3 are highly correlated with ESSPRI (R)2>0.9), STAT1 and STAT2 are highly related to FACIT (R)2>0.76), and TRIM37 and ZNF606 are highly correlated with ProF (R)2>0.71). These data provide evidence that specific RNA molecules circulating in certain patients promote chronic activation of inflammatory pathways in these patients.

Table 4: genes showing a strong correlation with MCII in a given tool (ESSPRI, FACIT or ProF)

Example 11

Safety and tolerability of RSLV-132 treatment

To assess the overall safety and tolerability of RSLV-132 treatment, adverse events were measured throughout the study. Adverse events were monitored within 211 days after final treatment. The incidence of treatment-emergent adverse events, severe adverse events and drug-related adverse events was comparable between the RSLV-132 treated group and the placebo group (table 5). No mortality occurred during the study. Fatigue is the most common Adverse Event (AE) in the study. Most adverse events from fatigue were reported early in the study. No serious infection or infusion reactions were observed in any of the treatment groups during the study. No patients discontinued the drug study due to adverse events AE. One patient in the RSLV-132 group presented with a severe adverse event and was hospitalized for mumps 88 days after the last dose of study drug. This adverse event appeared to be unrelated to RSLV-132 treatment.

TABLE 5 handling of Emergency adverse events (TEAE) (safety analysis set)

Hospitalization for parotitis 88 days after the last dose of study medication; independent of study drug

Sequence listing (Table 1)

94页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:经由存在于体腔中的导管来多用途地递送治疗性非紫外线电磁辐射的方法和装置

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类