Intranasal delivery of olanzapine by precision nasal device

文档序号:1026394 发布日期:2020-10-27 浏览:4次 中文

阅读说明:本技术 通过精密鼻装置的奥氮平的鼻内递送 (Intranasal delivery of olanzapine by precision nasal device ) 是由 J·D·赫克曼 K·H·萨特利 I·达舍夫斯基 A·R·达斯 于 2019-01-04 设计创作,主要内容包括:提供了用于躁动的急性治疗的方法,该躁动包括患有精神分裂症或双相障碍的患者中的躁动,所述方法包括向具有躁动的受试者施用有效剂量的包含奥氮平的干燥药物组合物,其中所述剂量通过鼻内递送装置施用,所述装置在鼻内施用后提供(a)至少30ng/mL的平均峰值血浆奥氮平浓度(C<Sub>max</Sub>)和(b)短于0.5小时的达到奥氮平的C<Sub>max</Sub>的平均时间(T<Sub>max</Sub>)。提供了适于奥氮平的鼻内递送的干燥药物组合物和装置。(Methods for acute treatment of agitation are providedComprising agitation in a patient suffering from schizophrenia or bipolar disorder, comprising administering to the subject with agitation an effective dose of a dry pharmaceutical composition comprising olanzapine, wherein the dose is administered by an intranasal delivery device that provides, following intranasal administration, (a) a mean peak plasma olanzapine concentration (Cmax) of at least 30ng/mL max ) And (b) C to olanzapine in less than 0.5 hour max Average time (T) of max ). Dry pharmaceutical compositions and devices suitable for intranasal delivery of olanzapine are provided.)

1. A method for acute treatment of agitation in a subject, comprising:

intranasally administering to a subject exhibiting agitation an effective dose of a dry pharmaceutical composition comprising olanzapine.

2. The method of claim 1, wherein the dry pharmaceutical composition is a powder.

3. The method of claim 2, wherein the powder comprises olanzapine in crystalline or amorphous form.

4. The method of claim 3, wherein the olanzapine is an amorphous solid obtained by spray drying.

5. The method of any one of claims 2-4, wherein the dry pharmaceutical composition comprises olanzapine in a partially crystalline and partially amorphous form.

6. The method of any one of claims 2-5, wherein the powder has a median diameter (D50) of the particle size distribution of olanzapine between 1 μm and 100 μm, between 1 μm and 50 μm, or between 1 μm and 15 μm.

7. The method of claim 6, wherein the olanzapine particle size distribution has a median diameter (D50) between 7.5 μm and 15 μm.

8. The method of any one of claims 1-70, wherein the dry pharmaceutical composition comprises no more than 70% olanzapine by weight.

9. The method of claim 8, wherein the dry pharmaceutical composition comprises no more than 60% olanzapine by weight.

10. The method of claim 9, wherein the dry pharmaceutical composition comprises 10-60% olanzapine by weight.

11. The method of claim 10, wherein the dry pharmaceutical composition comprises 25-55% olanzapine by weight.

12. The method of claim 11, wherein the dry pharmaceutical composition comprises 30-50% olanzapine by weight.

13. The method of claim 12, wherein the dry pharmaceutical composition comprises 40-50% olanzapine by weight.

14. The method of any one of claims 8-13, wherein the dry pharmaceutical composition further comprises a stabilizer, wherein the stabilizer is selected from the group consisting of: hydroxypropyl methylcellulose (HPMC), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus), vinyl pyrrolidine-vinyl acetate copolymer (Kollidon VA64), polyvinyl pyrrolidine K30(Kollidon K30), polyvinylpyrrolidone K90(Kollidon K90), hydroxypropyl cellulose (HPC), hydroxypropyl β -cyclodextrin (HPBCD), mannitol, and lactose monohydrate.

15. The method of claim 14, wherein the stabilizer is Hydroxypropylmethylcellulose (HPMC).

16. The method of any one of claims 8-15, wherein the dry pharmaceutical composition further comprises a penetration enhancer, wherein the penetration enhancer is selected from the group consisting of: n-tridecyl- β -D-maltoside, n-dodecyl- β -D-maltoside, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), propylene glycol, disodium EDTA, PEG400 monostearate, polysorbate 80, and polyethylene glycol (15) hydroxystearate.

17. The method of claim 16, wherein the penetration enhancer is 1, 2-distearoyl-sn-glycerol-3-phosphocholine (DSPC).

18. The method of any one of claims 8-17, wherein the dry pharmaceutical composition further comprises an antioxidant, wherein the antioxidant is selected from the group consisting of: alpha-tocopherol, ascorbic acid, ascorbyl palmitate, bronopol, Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), citric acid monohydrate, sodium ascorbate, ethylenediaminetetraacetic acid, fumaric acid, malic acid, methionine, propionic acid, sodium metabisulfite, sodium sulfite, sodium thiosulfate, thymol, and vitamin E polyethylene glycol succinate.

19. The method of any one of claims 1-18, wherein the dried pharmaceutical composition comprises less than 3%, less than 2%, less than 1.5%, less than 1%, or less than 0.5% by weight water.

20. The method of any one of claims 8-19, wherein the dry pharmaceutical composition consists essentially of:

50% by weight olanzapine;

42% by weight of HPMC; and

8% by weight of DSPC.

21. The method of any one of claims 1-20, wherein the dose is administered by an intranasal delivery device.

22. The method of claim 21, wherein the intranasal delivery device is a hand-held, manually-actuated, metered dose intranasal administration device.

23. The method of claim 21, wherein the intranasal delivery device is a hand-held, manually-actuated, propellant-driven, metered dose intranasal administration device.

24. The method of any one of claims 21-23, wherein the dry pharmaceutical composition is encapsulated within a capsule located within the device prior to device actuation.

25. The method of any one of claims 21-23, wherein the dry pharmaceutical composition is stored within a dosage container that is removably coupled to the device prior to device actuation.

26. The method of any one of claims 21-25, wherein the intranasal delivery device is capable of delivering the dry pharmaceutical composition to the upper nasal cavity.

27. The method of any one of claims 1-26, wherein the effective dose of the dry pharmaceutical composition comprises olanzapine in an amount effective to reduce agitation within 30 minutes.

28. The method of claim 27, wherein the effective dose of the dry pharmaceutical composition comprises 1-30mg olanzapine.

29. The method of claim 28, wherein the effective dose comprises 2-20mg olanzapine.

30. The method of claim 29, wherein the effective dose comprises 5-15mg olanzapine.

31. The method of claim 30, wherein the effective dose comprises 5mg olanzapine.

32. The method of claim 30, wherein the effective dose comprises 10mg olanzapine.

33. The method of claim 30, wherein the effective dose comprises 15mg olanzapine.

34. The method of any one of claims 27-33, wherein the effective dose is administered as a single undivided dose.

35. The method of any one of claims 27-33, wherein the effective dose is administered in multiple divided sub-dose forms.

36. The method of any one of claims 1-35, wherein the subject has schizophrenia.

37. The method of any one of claims 1-35, wherein the subject has bipolar disorder, optionally bipolar I disorder.

38. The method of any one of claims 1-35, wherein the patient has autism, dementia, PTSD, intoxication or drug-induced psychotic state.

39. The method of any one of claims 1-38, wherein the intranasal administration provides:

(a) a mean peak plasma olanzapine concentration (C) of at least 20ng/mLmax) And are and

(b) c to olanzapine in less than 1.5 hoursmaxAverage time (T) ofmax)。

40The method of claim 39, wherein said intranasal administration provides: c to olanzapine in less than 1.0 hourmaxAverage time (T) ofmax)。

41. The method of claim 40, wherein the intranasal administration provides: c to olanzapine in less than 0.75 hoursmaxAverage time (T) ofmax)。

42. The method of claim 41, wherein the intranasal administration provides: c to olanzapine in less than 0.50 hoursmaxAverage time (T) ofmax)。

43. The method of claim 40, wherein the intranasal administration provides: c to olanzapine in less than 0.25 hoursmaxAverage time (T) ofmax)。

44. The method of any one of claims 39-43, wherein the intranasal administration provides: a mean peak plasma olanzapine concentration (C) of at least 30ng/mLmax)。

45. The method of claim 44, wherein the intranasal administration provides: a mean peak plasma olanzapine concentration (C) of at least 40ng/mLmax)。

46. The method of claim 45, wherein the intranasal administration provides: a mean peak plasma olanzapine concentration (C) of at least 50ng/mLmax)。

47. The method of claim 46, wherein the intranasal administration provides: a mean peak plasma olanzapine concentration (C) of at least 60ng/mLmax)。

48. The method of claim 47, wherein the intranasal administration provides: a mean peak plasma olanzapine concentration (C) of at least 70ng/mLmax)。

49. The method of claim 48, wherein said intranasal administration provides: a mean peak plasma olanzapine concentration (C) of at least 80ng/mLmax)。

50. A dry pharmaceutical composition suitable for intranasal administration comprising:

olanzapine, and

at least one excipient.

51. The dry pharmaceutical composition of claim 50, wherein the composition is a powder.

52. The dry pharmaceutical composition of claim 51, wherein the composition comprises olanzapine in a crystalline or amorphous form.

53. The dry pharmaceutical composition of claim 51, wherein the composition comprises olanzapine in an amorphous form.

54. The dry pharmaceutical composition of claim 53, wherein the amorphous olanzapine is obtained by spray drying.

55. The dry pharmaceutical composition of any one of claims 52-54, wherein the composition comprises olanzapine in a partially crystalline and partially amorphous form.

56. The dry pharmaceutical composition of any one of claims 52-55, wherein the median diameter (D50) of the particle size distribution of olanzapine in the powder is between 1 μm and 100 μm, between 1 μm and 50 μm, or between 1 μm and 15 μm.

57. The dry pharmaceutical composition of claim 56, wherein the olanzapine particle size distribution has a median diameter (D50) between 7.5 μm and 15 μm.

58. The dry pharmaceutical composition of any one of claims 50-57, wherein the dry pharmaceutical composition comprises no more than 70% olanzapine by weight.

59. The dry pharmaceutical composition of claim 58, wherein the dry pharmaceutical composition comprises no more than 60% olanzapine by weight.

60. The dry pharmaceutical composition of claim 59, wherein the dry pharmaceutical composition comprises 10-60% olanzapine by weight.

61. The dry pharmaceutical composition of claim 60, wherein the dry pharmaceutical composition comprises 25-55% olanzapine by weight.

62. The dry pharmaceutical composition of claim 61, wherein the dry pharmaceutical composition comprises 30-50% olanzapine by weight.

63. The dry pharmaceutical composition of claim 62, wherein the dry pharmaceutical composition comprises 30-40% olanzapine by weight.

64. The dry pharmaceutical composition of claim 62, wherein the dry pharmaceutical composition comprises 40-50% olanzapine by weight.

65. The dry pharmaceutical composition of any one of claims 50-64, further comprising a stabilizer, wherein the stabilizer is selected from the group consisting of: hydroxypropyl methylcellulose (HPMC), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus), vinylpyrrolidone-vinyl acetate copolymer (Kollidon VA64), polyvinylpyrrolidone K30(Kollidon K30), polyvinylpyrrolidone K90(Kollidon K90), hydroxypropyl cellulose (HPC), hydroxypropyl β -cyclodextrin (HPBCD), mannitol, and lactose monohydrate.

66. The dry pharmaceutical composition of claim 65, wherein the stabilizer is Hydroxypropylmethylcellulose (HPMC).

67. The dry pharmaceutical composition of any one of claims 50-66, further comprising a penetration enhancer, wherein the penetration enhancer is selected from the group consisting of: n-tridecyl- β -D-maltoside, n-dodecyl- β -D-maltoside, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), propylene glycol, disodium EDTA, PEG400 monostearate, polysorbate 80, and polyethylene glycol (15) hydroxystearate.

68. The dry pharmaceutical composition of claim 67, wherein the penetration enhancer is 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC).

69. The dry pharmaceutical composition of any one of claims 50-68, further comprising an antioxidant, wherein the antioxidant is selected from the group consisting of: alpha-tocopherol, ascorbic acid, ascorbyl palmitate, bronopol, Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), citric acid monohydrate, sodium ascorbate, ethylenediaminetetraacetic acid, fumaric acid, malic acid, methionine, propionic acid, sodium metabisulfite, sodium sulfite, sodium thiosulfate, thymol, and vitamin E polyethylene glycol succinate.

70. The dry pharmaceutical composition of any one of claims 50-69, wherein the dry pharmaceutical composition comprises less than 3%, less than 2%, less than 1.5%, less than 1%, or less than 0.5% by weight water.

71. The dry pharmaceutical composition of any one of claims 50-70, wherein the dry pharmaceutical composition consists essentially of:

50% by weight olanzapine;

42% by weight of HPMC; and

8% by weight of DSPC.

72. A unit dosage form comprising a dry pharmaceutical composition according to any one of claims 50-71.

73. The unit dosage form of claim 72, wherein the unit dosage form contains 1-30mg olanzapine.

74. The unit dosage form of claim 73, wherein the unit dosage form contains 2-20mg olanzapine.

75. The unit dosage form of claim 74, wherein the unit dosage form contains 5-15mg olanzapine.

76. The unit dosage form of claim 75, wherein the unit dosage form contains 5mg olanzapine.

77. The unit dosage form of claim 75, wherein the unit dosage form contains 10mg of olanzapine.

78. The unit dosage form of claim 75, wherein the unit dosage form contains 15mg of olanzapine.

79. The unit dosage form of any one of claims 72-78, wherein the unit dosage form is a capsule encapsulating the dry pharmaceutical composition.

80. The unit dosage form of any one of claims 72-78, wherein the unit dosage form is a dosage container storing the dry pharmaceutical composition, wherein the dosage container is configured to be removably coupled to an intranasal delivery device.

Background

In 1.3 million U.S. emergency room visits per year, it is estimated that 170 million patients are involved in agitation, including patients whose agitation is a manifestation of schizophrenia or bipolar disorder.

The current standard of care for treating acute and exacerbating manic events in schizophrenia or bipolar I mania is by administering 5mg, 7.5mg or 10mg olanzapine (an atypical antipsychotic) by intramuscular Injection (IM). Although olanzapine IM is characterized by a rapid onset (reaching mean maximum plasma concentrations within 15 to 45 minutes), this route of administration is characterized by several injection-related acute side effects including injection site pain, excessive sedation, extrapyramidal syndrome and akathisia (Atkins et al, BMC Psychiatry 14,7 (2014); Battaglia et al, am.j.emerg.med.21: 192-charge 198 (2003); Kishi et al, j.psychiatr.res.68: 198-charge 209 (2015)). Moreover, the invasive intramuscular injection procedure may cause emotional trauma to the patient, whether collaborative or uncooperative, and may result in physical attack on hospital personnel attempting to inject. In addition, IM injections are contraindicated in cooperative patients (Nordstrom et al, west.j.emerg.med.13(1):3-10 (2012)).

Oral administration of olanzapine in standard tablets or orally disintegrating tablets has been approved for acute treatment of mania or mixed episodes associated with bipolar 1 disorder and does not suffer from many of the disadvantages of intramuscular injections in this patient population; however, there is a significant lag before effective blood levels are reached and agitation is reduced.

Pulmonary delivery of the typical antipsychotic drug loxapine by oral inhalation has been approved in 2017 for acute treatment of agitation associated with schizophrenia or bipolar 1 disorder in adults. However, the product label contains a black box warning: administration may cause bronchospasm, which may lead to respiratory distress and cessation (adasvre FDA product label, 8 months 2017), and the product is only available under risk assessment and mitigation strategies (REMS).

An effective non-invasive treatment of acute agitation can divert treatment from the emergency room to the "community" early in the onset of the agitation, with obvious benefits, including reduced emergency department visits and health economic burdens. Thus, there is a need for acute treatment of agitation, including agitation associated with schizophrenia and manic depression, which is rapid in onset and does not require parenteral injection.

Disclosure of Invention

We have developed a dry powder formulation of olanzapine suitable for intranasal delivery by a hand-held, manually actuated, propellant driven, metered dose intranasal applicator. After a single dose PK study in cynomolgus monkeys and rodents, we performed a phase I trial in healthy human subjects. In this phase I study, intranasal delivery of olanzapine formulations yielded similar or slightly higher plasma exposure (AUC) and maximum C compared to olanzapine administered IM at the same dosemax. In addition, median T after intranasal delivery of the formulationmax-three test doses in the range of 0.16-0.17 hours-significantly shorter than the median T measured for intramuscular and oral administrationmaxShows that the olanzapine can be quickly and effectively absorbed across nasal epithelial cells。

Three standardized behavioral tests were used to measure pharmacodynamic effects. Behavioral testing has shown that intranasal administration of olanzapine produces similar or better sedation than IM or oral administration of olanzapine. Consistent with pharmacokinetic data, behavioral effects of olanzapine were observed significantly earlier in the group of subjects treated with intranasal olanzapine (INP105) than in the group of subjects treated with oral olanzapine (Zyprexa Zydis). These results indicate that intranasal delivery of olanzapine may be an effective method for acute treatment of agitation.

Thus, in a first aspect, a method for acute treatment of agitation is presented. The method comprises intranasally administering to a subject exhibiting agitation an effective dose of a dry pharmaceutical composition comprising olanzapine.

In typical embodiments, the dry pharmaceutical composition is a powder. In some embodiments, the powder comprises olanzapine in crystalline or amorphous form. In some embodiments, olanzapine is an amorphous solid obtained by spray drying. In some embodiments, the dry pharmaceutical composition comprises olanzapine in a partially crystalline and partially amorphous form.

In some embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the powder is between 1 μm and 100 μm, between 1 μm and 50 μm, or between 1 μm and 15 μm, as measured by a laser diffraction particle size analyzer, such as a Malvern panalytical mastersizer 3000. In some embodiments, the olanzapine particle size distribution has a median diameter (D50) between 7.5 μm and 15 μm.

In some embodiments, the dose is administered by an intranasal delivery device. In some embodiments, the intranasal delivery device is a hand-held, manually-actuated, metered dose intranasal administration device. In some embodiments, the intranasal delivery device is a hand-held, manually-actuated, propellant-driven, metered dose intranasal administration device.

In some embodiments, the dry pharmaceutical composition is encapsulated within a capsule located within the device prior to actuation of the device. In some embodiments, the dry pharmaceutical composition is stored within a dosage container that is removably coupled to the device prior to actuation of the device.

In some embodiments, the intranasal delivery device is capable of delivering the dry pharmaceutical composition to the upper nasal cavity.

In some embodiments, the dry pharmaceutical composition comprises no more than 70% by weight, or no more than 60% by weight olanzapine. In some embodiments, the dry pharmaceutical composition comprises 10-60% olanzapine by weight, 20-60% olanzapine by weight, 25-55% olanzapine by weight, 30-50% olanzapine by weight, or 40-50% olanzapine by weight.

In some embodiments, the dry pharmaceutical composition further comprises a stabilizing agent, wherein the stabilizing agent is selected from the group consisting of: hydroxypropyl methylcellulose (HPMC), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus), vinyl pyrrolidine-vinyl acetate copolymer (Kollidon VA64), polyvinyl pyrrolidine K30(Kollidon K30), polyvinylpyrrolidone K90(Kollidon K90), hydroxypropyl cellulose (HPC), hydroxypropyl β -cyclodextrin (HPBCD), mannitol, and lactose monohydrate. In some embodiments, the stabilizer is Hydroxypropylmethylcellulose (HPMC).

In some embodiments, the dry pharmaceutical composition further comprises a penetration enhancer, wherein the penetration enhancer is selected from the group consisting of: n-tridecyl- β -D-maltoside, n-dodecyl- β -D-maltoside, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), propylene glycol, disodium EDTA, PEG400 monostearate, polysorbate 80, and polyethylene glycol (15) hydroxystearate. In some embodiments, the penetration enhancer is 1, 2-distearoyl-sn-glycerol-3-phosphocholine (DSPC).

In some embodiments, the dry pharmaceutical composition further comprises an antioxidant, wherein the antioxidant is selected from the group consisting of: alpha-tocopherol, ascorbic acid, ascorbyl palmitate, bronopol Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), citric acid monohydrate, sodium ascorbate, ethylenediaminetetraacetic acid, fumaric acid, malic acid, methionine, propionic acid, sodium metabisulfite, sodium sulfite, sodium thiosulfate, thymol, and vitamin E polyethylene glycol succinate.

In some embodiments, the dry pharmaceutical composition comprises less than 3%, less than 2%, less than 1.5%, less than 1%, or less than 0.5% by weight water.

In some embodiments, the dry pharmaceutical composition consists essentially of: 50% by weight olanzapine; 42% by weight of HPMC; and 8 wt% DSPC.

In some embodiments, the effective dose is a dose of olanzapine effective to reduce agitation within 60 minutes. In some embodiments, an effective dose of the dry pharmaceutical composition comprises 1-30mg olanzapine; 2-20mg olanzapine; 5-15mg olanzapine; 5mg olanzapine; 10mg olanzapine; or 15mg olanzapine.

In some embodiments, the effective dose is administered as a single undivided dose. In some embodiments, the effective dose is administered in multiple aliquots of sub-dose forms.

In some embodiments, the subject has schizophrenia. In some embodiments, the subject has bipolar disorder, optionally bipolar I disorder. In some embodiments, the subject has autism, dementia, PTSD, intoxication or drug-induced psychotic state.

In some embodiments, intranasal administration provides: (a) a mean peak plasma olanzapine concentration (C) of at least 20ng/mLmax) And (b) C to olanzapine in less than 1.5 hoursmaxAverage time (T) ofmax)。

In some embodiments, intranasal administration provides: c to olanzapine in less than 1.0 hourmaxAverage time (T) ofmax) (ii) a C to olanzapine in less than 0.75 hoursmaxAverage time (T) ofmax) (ii) a C to olanzapine in less than 0.50 hoursmaxAverage time (T) ofmax) Or less than 0.25 hours to reach olanzapine CmaxAverage time (T) ofmax)。

In some embodiments, intranasal administration provides: average of at least 40ng/mLPeak plasma olanzapine concentration (C)max) (ii) a A mean peak plasma olanzapine concentration (C) of at least 50ng/mLmax) (ii) a A mean peak plasma olanzapine concentration (C) of at least 60ng/mLmax) (ii) a A mean peak plasma olanzapine concentration (C) of at least 70ng/mLmax) (ii) a Or a mean peak plasma olanzapine concentration (C) of at least 80ng/mLmax)。

In another aspect, the present invention provides a dry pharmaceutical composition suitable for intranasal administration comprising: olanzapine and at least one excipient.

In some embodiments, the composition is a powder. In some embodiments, the composition comprises olanzapine in a crystalline or amorphous form. In some embodiments, the composition comprises olanzapine in an amorphous form. In some embodiments, the amorphous olanzapine is obtained by spray drying. In some embodiments, the composition comprises olanzapine in a partially crystalline and partially amorphous form.

In some embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the powder is between 1 μm and 100 μm, between 1 μm and 50 μm, or between 1 μm and 15 μm. In some embodiments, the olanzapine particle size distribution has a median diameter (D50) between 7.5 μm and 15 μm.

In some embodiments, the dry pharmaceutical composition comprises no more than 70% olanzapine by weight, or no more than 60% olanzapine by weight. In some embodiments, the dry pharmaceutical composition comprises 10-60% olanzapine by weight, 20-60% olanzapine by weight, 25-55% olanzapine by weight, 30-50% olanzapine by weight, 30-40% olanzapine by weight, or 40-50% olanzapine by weight.

In some embodiments, the dry pharmaceutical composition further comprises a stabilizing agent, wherein the stabilizing agent is selected from the group consisting of: hydroxypropyl methylcellulose (HPMC), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus), vinylpyrrolidone-vinyl acetate copolymer (Kollidon VA64), polyvinylpyrrolidone K30(Kollidon K30), polyvinylpyrrolidone K90(Kollidon K90), hydroxypropyl cellulose (HPC), hydroxypropyl β -cyclodextrin (HPBCD), mannitol, and lactose monohydrate. In some embodiments, the stabilizer is Hydroxypropylmethylcellulose (HPMC).

In some embodiments, the dry pharmaceutical composition further comprises a penetration enhancer, wherein the penetration enhancer is selected from the group consisting of: n-tridecyl- β -D-maltoside, n-dodecyl- β -D-maltoside, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), propylene glycol, disodium EDTA, PEG400 monostearate, polysorbate 80, and polyethylene glycol (15) hydroxystearate. In some embodiments, the penetration enhancer is 1, 2-distearoyl-sn-glycerol-3-phosphocholine (DSPC).

In some embodiments, the dry pharmaceutical composition further comprises an antioxidant, wherein the antioxidant is selected from the group consisting of: alpha-tocopherol, ascorbic acid, ascorbyl palmitate, bronopol Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), citric acid monohydrate, sodium ascorbate, ethylenediaminetetraacetic acid, fumaric acid, malic acid, methionine, propionic acid, sodium metabisulfite, sodium sulfite, sodium thiosulfate, thymol, and vitamin E polyethylene glycol succinate.

In some embodiments, the dry pharmaceutical composition comprises less than 3%, less than 2%, less than 1.5%, less than 1%, or less than 0.5% by weight water.

In some embodiments, the dry pharmaceutical composition consists essentially of: 50% by weight olanzapine; 42% by weight of HPMC; and 8 wt% DSPC.

In yet another aspect, the present invention provides a unit dosage form containing a dry pharmaceutical composition provided herein.

In some embodiments, the unit dosage form contains 1-30mg olanzapine; 2-20mg olanzapine; 5-15mg olanzapine; 5mg olanzapine; 10mg olanzapine; or 15mg olanzapine.

In some embodiments, the unit dosage form is a capsule encapsulating the dry pharmaceutical composition. In some embodiments, the unit dosage form is a dosage container storing a dry pharmaceutical composition, wherein the dosage container is configured to be removably coupled to an intranasal delivery device.

Other features and advantages of the present disclosure will become apparent from the following detailed description, including the drawings. However, it should be understood that the detailed description and specific examples are provided for illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.

Drawings

Figure 1 shows intramuscular administration (IM) followed by or using precision nasal deliveryDevice mean plasma olanzapine levels in non-human primates (NHPs) after intranasal administration of three different dry powder formulations of olanzapine as a function of time.

Fig. 2 shows an image of an NHP-POD device for administering olanzapine to NHPs as described in examples 1 and 2.

Figure 3 shows the change in plasma olanzapine concentration over time (time shown is 0-2 hours) after administration of a nasal powder formulation of olanzapine (F-OLZ #1-6) delivered to a non-human primate (NHP) by an NHP-POD device.

Fig. 4 shows the change in plasma olanzapine concentration over time (time shown is 0-24 hours) after administration of a nasal powder formulation of olanzapine (F-OLZ #1-6) delivered to NHPs by an NHP-POD device.

Fig. 5A is an intranasal drug delivery device according to one or more embodiments.

Fig. 5B illustrates a partial cross-sectional view of the intranasal delivery device with a removable tip attached thereto and an isolated perspective view of the removable tip in its detached state according to one or more embodiments.

Fig. 5C is a perspective view of a tip and capsule according to one or more embodiments.

Fig. 5D is a cross-sectional view of a tip and a capsule coupled to a device according to one or more embodiments.

Fig. 5E is an exploded view of a tip and capsule according to one or more embodiments.

Fig. 5F is a perspective view of a tip with a capsule attached thereto according to one or more embodiments.

Fig. 5G is a cross-sectional view of a tip with a capsule attached according to one or more embodiments.

Fig. 5H is a cross-sectional view of a tip according to one or more embodiments.

Fig. 5I is a cross-sectional view of a tip according to one or more embodiments.

Fig. 5J is a cross-sectional view of an inlet interface with a capsule-attached tip according to one or more embodiments.

Fig. 5K-5N are perspective views of a tip of a device according to one or more embodiments.

Fig. 5O is a perspective view of a tip according to one or more embodiments.

Fig. 5P is a perspective view of a tip according to one or more embodiments.

Fig. 5Q is a perspective view of a tip coupled to a device according to one or more embodiments.

Fig. 5R is a cross-sectional view of a tip coupled to a device according to one or more embodiments.

Fig. 5S is an enlarged view of a capsule-attached inlet interface according to one or more embodiments.

Fig. 5T is a perspective view of a second embodiment of a tip according to one or more embodiments.

Fig. 5U is a perspective view of the tip of fig. 5T with a capsule attached according to one or more embodiments.

Fig. 5V is a perspective view of a piercing member according to one or more embodiments.

Fig. 5W is a perspective view of a piercing member according to one or more embodiments.

Fig. 5X illustrates a flow path of a second embodiment of a piercing member according to one or more embodiments.

Fig. 6 illustrates an example of a non-human primate precision nasal delivery device according to one or more embodiments.

Fig. 7A illustrates another example of a non-human primate precision nasal delivery device according to one or more embodiments.

Fig. 7B illustrates a side view and a cross-sectional view of an actuator body of the inter-nasal device of fig. 7A, according to one or more embodiments.

Fig. 7C illustrates a side view of an extension tube of the intranasal device of fig. 7A according to one or more embodiments.

Fig. 7D illustrates an enlarged view of two embodiments of a connection interface at the end of the extension tube of fig. 7C according to one or more embodiments.

Fig. 7E illustrates a side view and a cross-sectional view of the tip of the inter-nasal device of fig. 7A, according to one or more embodiments.

Figures 8A-C show intranasal administration of 5mg, 10mg, or 15mg olanzapine (INP105) in a human subject; intramuscular administration of 5mg or 10mg olanzapine (Zyprexa IM); or the mean plasma concentration-time curve measured after oral administration of 10mg olanzapine (Zyprexa Zydis). Data were obtained from the phase 1 clinical trial study described in example 3, and for clarity, figure 8A plots results without error bars, figure 8B includes error bars for shorter PK time points (0-1 hour), and figure 8C plots results without error bars for longer PK time points (0-8 hours).

Figure 9 shows intranasal administration of 5mg, 10mg, or 15mg olanzapine (INP105) in a human subject; intramuscular administration of 5mg or 10mg olanzapine (Zyprexa IM); or to three categories after oral administration of 10mg olanzapine (Zyprexa Zydis): change from baseline in maximum VAS scores measured by alertness/dull, confusion/waking and energetic/not conscious. Data were obtained from the study described in example 3 and plotted with error bars.

Figure 10 shows intranasal administration of 5mg, 10mg, or 15mg olanzapine (INP105) in a human subject; intramuscular administration of 5mg or 10mg olanzapine (Zyprexa IM); or the change from baseline in the maximum ACES score measured after oral administration of 10mg olanzapine (Zyprexa Zydis). Data were obtained from the study described in example 3 and plotted with error bars.

Figures 11A-B show intranasal administration of 5mg, 10mg, or 15mg olanzapine (INP105) in a human subject; intramuscular administration of 5mg or 10mg olanzapine (Zyprexa IM); or the average ACES score-time curve measured after oral administration of 10mg olanzapine (Zyprexa Zydis). Data were obtained from the study described in example 3, with FIG. 11A plotting results for longer PK time points (0-8 hours) and FIG. 11B plotting results for shorter PK time points (0-1 hour).

Figure 12 shows intranasal administration of 5mg, 10mg, or 15mg olanzapine (INP105) in a human subject; intramuscular administration of 5mg or 10mg olanzapine (Zyprexa IM); or the change from baseline in the maximum DSST score measured after oral administration of 10mg olanzapine (Zyprexa Zydis). Data were obtained from the study described in example 3 and plotted with error bars.

Figures 13A-B show intranasal administration of 5mg, 10mg, or 15mg olanzapine (INP105) in a human subject; intramuscular administration of 5mg or 10mg olanzapine (Zyprexa IM); or the average DSST score-time curve measured after oral administration of 10mg olanzapine (Zyprexa Zydis). Data were obtained from the study described in example 3, with FIG. 13A plotting results for longer PK time points (0-4 hours) and FIG. 13B plotting results for shorter PK time points (0-1 hour).

Figures 14A-F show average DSST score-time curves and average plasma concentration-time curves measured after intramuscular administration of 5mg olanzapine (figure 14A), intramuscular administration of 10mg olanzapine (figure 14B), oral administration of 10mg olanzapine (figure 14C), intranasal administration of 5mg olanzapine (figure 14D), intranasal administration of 10mg olanzapine (figure 14E), or intranasal administration of 15mg olanzapine (figure 14F) in a human subject. Data were obtained from the study described in example 3, with results plotted for longer PK time points (0-12 hours).

Figures 15A-F show the mean DSST score-time curves and mean plasma concentration-time curves measured after intramuscular administration of 5mg olanzapine (figure 15A), intramuscular administration of 10mg olanzapine (figure 15B), oral administration of 10mg olanzapine (figure 15C), intranasal administration of 5mg olanzapine (figure 15D), intranasal administration of 10mg olanzapine (figure 15E), or intranasal administration of 15mg olanzapine (figure 15F) in a human subject. Data were obtained from the study described in example 3, with results plotted for shorter PK time points (0-1 hour).

Figures 16A-F show the average ACES score-time curve and average plasma concentration-time curve measured after intramuscular administration of 5mg olanzapine (figure 16A), intramuscular administration of 10mg olanzapine (figure 16B), oral administration of 10mg olanzapine (figure 16C), intranasal administration of 5mg olanzapine (figure 16D), intranasal administration of 10mg olanzapine (figure 16E), or intranasal administration of 15mg olanzapine (figure 16F) in a human subject. Data were obtained from the study described in example 3, with results plotted for longer PK time points (0-12 hours).

Figures 17A-F show average ACES score-time curves and average plasma concentration-time curves measured after intramuscular administration of 5mg olanzapine (figure 17A), intramuscular administration of 10mg olanzapine (figure 17B), oral administration of 10mg olanzapine (figure 17C), intranasal administration of 5mg olanzapine (figure 17D), intranasal administration of 10mg olanzapine (figure 17E), or intranasal administration of 15mg olanzapine (figure 17F) in a human subject. Data were obtained from the study described in example 3, with results plotted for shorter PK time points (0-1 hour).

Detailed Description

Definition of

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

A pharmaceutical composition is "dry" if its residual moisture content does not exceed 5%.

Other understanding conventions

The range is as follows: throughout this disclosure, various aspects of the present invention are presented in a range format. Ranges are inclusive of the recited endpoints. It is to be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have explicitly disclosed all the possible sub-ranges as well as individual numerical values within that range. For example, a description of a range such as 1 to 6 should be read as having explicitly disclosed sub-ranges such as 1 to 3, 1 to 4, 1 to 5, 2 to 4, 2 to 6, 3 to 6, and the like, as well as individual numbers within that range, such as 1,2, 2.7, 3, 4, 5, 5.3, and 6. This applies regardless of the breadth of the range.

The term "or" as used herein is to be understood as being inclusive, unless specified otherwise or apparent from the context.

The terms "a", "an" and "the" as used herein are to be construed as singular or plural unless specifically stated or apparent from the context. That is, the articles "a" and "an" are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. For example, "an element" refers to one element or more than one element.

In this disclosure, "comprise," "include," "contain," "have," "include," and variants thereof have the meaning attributed to them in U.S. patent law, permitting the presence of additional components beyond those expressly recited.

Unless specifically stated or otherwise apparent from the context, the term "about" as used herein is to be understood as being within the normal tolerance of the art, e.g., within 2 standard deviations of the mean, and is intended to encompass variations of ± 20% or ± 10%, more preferably ± 5%, even more preferably ± 1%, and still more preferably ± 0.1% from the stated value.

Summary of Experimental observations

We performed two single dose PK studies in cynomolgus monkeys to examine the pharmacokinetics following administration of a multi-powder olanzapine formulation delivered by the intranasal route using a non-human primate precision nasal delivery ("nhpPOD" or "NHP-POD") device. The formulations examined included unmodified crystalline powder, formulations containing HPMC and 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), and formulations containing HPMC and Pluronic F68. A placebo control also delivered intranasally by the nhpPOD device was microcrystalline cellulose.

PK results indicate that intranasal delivery of olanzapine formulations containing HPMC and DSPC using an nhpPOD device resulted in similar plasma exposure (AUC) and T as intramuscular administration of olanzapinemax. The formulated (HPMC/DSPC) powder produced 1.7 times higher AUC and 2.8 times shorter T compared to unformulated olanzapine (Cipla API)max

To further optimize Olanzapine (OLZ) formulation, approximately thirty different formulations were designed and manufactured for upper nasal delivery via the POD device. Formulations were tested, characterized and optimized for POD device compatibility. Stabilizers, penetration enhancers, particle size and manufacturing processes were also screened as part of the formulation development process.

In total twenty formulations were evaluated in a single dose PK study in rats (data not shown) and non-human primates (NHPs). The results show that administration of formulations F-OLZ #2, F-OLZ #5 and F OLZ #6 to NHPs via an NHP-POD device resulted in rapid absorption with a median T achieved in a short timemax(15, 15 and 23 minutes, respectively) and in less than 7 minutes more than 40ng/mL, which is approximately the plasma concentration achieved after 3X 10mg intramuscular injection in stable non-agitated patients (as reported in Zyprexa NDA 21253). Delivery of formulations F-OLZ #1, F OLZ #3 and F-OLZ #4 to NHPs via the NHP-POD device resulted in slower plasma absorption than the other 3 formulations, but still resulted in 30-60 min TmaxThis is compared to the time to reach peak plasma concentration (T) for oral Olanzapine (OLZ) tablets or orally disintegrating tabletsmax5-8 hours) is significantly faster.

The pharmacodynamic effects of each nasal olanzapine formulation administered to NHPs were collected throughout each study. For TmaxThe shorter lead formulation, with visible sedation observed in NHP by 7 min blood draw, but not over-sedation, and this effect lasted 24 hours. This reported sedative effect was observed in all groups receiving nasal olanzapine, but with a delayed onset of action and less pronounced in the group with slower time to peak plasma concentrations and lower peak exposure.

The pharmacokinetic and pharmacodynamic effects of the intranasally administered formulation F-OLZ #2 (olanzapine formulation with HPMC and DSPC (INP105)) were further tested in a phase 1 clinical trial in healthy human subjects. In this study, intranasal delivery of olanzapine formulations yielded similar or slightly higher plasma exposure (AUC) and maximum C compared to olanzapine administered IM at the same dosemax. In addition, median T after intranasal delivery of the formulationmaxSignificantly shorter than the median T measured for olanzapine administered IM or orallymaxTo show thatOlanzapine is rapidly and efficiently absorbed across nasal epithelial cells.

Three standardized behavioral tests were used to measure pharmacodynamic effects — Visual Analog Scale (VAS); restlessness/calm rating scale (ACES); and digital sign replacement testing (DSST). These tests all show that intranasal olanzapine administration produces similar or better sedation than IM or oral administration of olanzapine. Furthermore, the behavioral effects of olanzapine were observed significantly earlier in the group of subjects treated with intranasal olanzapine (INP105) or IM olanzapine (Zyprexa IM) compared to the group of subjects treated with oral olanzapine (Zyprexa Zydis). This is consistent with pharmacokinetic results where it was found that intranasal delivery of olanzapine had a significantly shorter median T compared to IM or oral deliverymax. These results indicate that intranasal delivery of olanzapine may be an effective method for acute treatment of agitation.

Method for treating agitation

Accordingly, in a first aspect, a method is provided for acute treatment of agitation. The method comprises intranasally administering to a subject exhibiting agitation an effective dose of a dry pharmaceutical composition comprising olanzapine.

Dry powder composition

In typical embodiments, the dry pharmaceutical composition is a powder.

In typical embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the powder is from 1 μm to 500 μm as measured by a laser diffraction particle size analyzer such as a Malvern Panalytical Mastersizer 3000. In some embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the powder is from 1 μm to 250 μm, from 1 μm to 100 μm, from 1 μm to 75 μm, from 1 μm to 50 μm, from 1 μm to 25 μm, from 1 μm to 20 μm, from 1 μm to 15 μm, or from 2 μm to 15 μm. In certain embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the composition is from 2 μm to 5 μm or from 7.5 μm to 15 μm.

In some embodiments, the powder comprises olanzapine in crystalline form. In some embodiments, the powder comprises olanzapine in an amorphous form. In some embodiments, the dry pharmaceutical composition comprises olanzapine in both crystalline and amorphous forms. In some embodiments, the dry pharmaceutical composition comprises olanzapine in a partially crystalline and partially amorphous form. In a particular embodiment olanzapine is an amorphous solid obtained by spray drying.

In various embodiments, the dry powder composition comprises no more than 70% olanzapine by weight. In some embodiments, the dry pharmaceutical composition comprises no more than 60% olanzapine by weight. In some embodiments, the composition comprises 10-70% olanzapine by weight, 20-70% olanzapine by weight, 10-60% olanzapine by weight, 20-60% olanzapine by weight, 25-55% olanzapine by weight, 30-50% olanzapine by weight, 30-40% olanzapine by weight, or 40-50% olanzapine by weight.

In some embodiments, the dry powder composition further comprises a stabilizer selected from the group consisting of: hydroxypropyl methylcellulose (HPMC), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus), vinyl pyrrolidine-vinyl acetate copolymer (Kollidon VA64), polyvinyl pyrrolidine K30(Kollidon K30), polyvinylpyrrolidone K90(Kollidon K90), hydroxypropyl cellulose (HPC), hydroxypropyl β -cyclodextrin (HPBCD), mannitol, and lactose monohydrate. In some embodiments, the stabilizer is Hydroxypropylmethylcellulose (HPMC).

In some embodiments, the dry powder composition further comprises a penetration enhancer selected from the group consisting of: n-tridecyl- β -D-maltoside, n-dodecyl- β -D-maltoside, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), propylene glycol, disodium EDTA, PEG400 monostearate, polysorbate 80, and polyethylene glycol (15) hydroxystearate. In some embodiments, the penetration enhancer is 1, 2-distearoyl-sn-glycerol-3-phosphocholine (DSPC).

In some embodiments, the dry powder composition comprises both HPMC and DSPC.

In various embodiments, the dry powder composition further comprises a nonionic surfactant. In certain embodiments, the nonionic surfactant is an alkyl maltoside. In a particular embodiment, the alkyl maltoside is n-dodecyl β -D-maltoside. In some embodiments, the nonionic surfactant is present in the dry powder composition from 0.1 to 10 weight percent, more typically from 1 to 5 weight percent. In a particular embodiment, the nonionic surfactant is present at 1% by weight.

In some embodiments, the nonionic surfactant is Pluronic PF 68. In some embodiments, the nonionic surfactant is present in the dry powder composition from 20 to 40 weight percent, more typically from 25 to 35 weight percent. In a particular embodiment, the nonionic surfactant is present at 31 wt%.

In some embodiments, the dry powder composition further comprises an antioxidant selected from the group consisting of: alpha-tocopherol, ascorbic acid, ascorbyl palmitate, bronopol Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), citric acid monohydrate, sodium ascorbate, ethylenediaminetetraacetic acid, fumaric acid, malic acid, methionine, propionic acid, sodium metabisulfite, sodium sulfite, sodium thiosulfate, thymol, and vitamin E polyethylene glycol succinate.

In some embodiments, the dry powder composition further comprises an acid. In certain embodiments, the acid is citric acid. In some embodiments, the acid is present in the dry powder composition at 10 to 20 wt.%, more typically 15 to 20 wt.%. In a particular embodiment, the citric acid is present at 18% by weight.

In various embodiments, the dry powder composition further comprises a salt of a monovalent inorganic cation. Typically, the salt is NaCl. In some embodiments, the composition comprises 1-5% by weight NaCl or 2-4% by weight NaCl.

In some embodiments, the dry powder composition comprises less than 3 wt.%, less than 2.5 wt.%, less than 2 wt.%, less than 1.5 wt.%, less than 1 wt.%, less than 0.9 wt.%, less than 0.8 wt.%, less than 0.7 wt.%, less than 0.6 wt.%, or less than 0.5 wt.% water.

In a currently preferred embodiment, the dry powder composition comprises 50% olanzapine by weight, 42% HPMC by weight and 8% DSPC. In some embodiments, the dry powder composition is a spray-dried composition comprising amorphous olanzapine. In some embodiments, olanzapine is spray dried in the presence of HPMC and/or DSPC. In other embodiments, HPMC and/or DSPC is added after spray drying of olanzapine.

Device for measuring the position of a moving object

In the methods described herein, the dose is administered by an intranasal delivery device that delivers the powder to the nasal cavity.

In some embodiments, the intranasal delivery device is a hand-held, manually-actuated, metered dose intranasal administration device. In certain embodiments, the device is a manually actuated, propellant driven, metered dose intranasal administration device. In particular embodiments, the dry pharmaceutical composition is encapsulated within a capsule present within the device prior to actuation of the device. In some embodiments, the dry pharmaceutical composition is stored within a dose container that is removably coupled to the device prior to actuation of the device. For example, the dosage container may be inserted into a portion of the device or may be coupled to the device such that the dosage container is in fluid communication with the device.

In various embodiments, the intranasal delivery device includes a housing body, a propellant canister contained within the housing body, a compound chamber containing a drug compound or designed to receive a drug compound, a channel in fluid communication with the propellant canister and the compound chamber, and an outlet orifice at a distal end of the channel. In this configuration, the propellant released from the canister travels through the channel, contacts the drug compound in the compound chamber, and pushes the drug compound out of the outlet orifice for delivery into the upper nasal cavity.

In typical embodiments, the intranasal delivery device is capable of delivering the dry pharmaceutical composition to the upper nasal cavity.

Nasal drug delivery device

In various embodiments, the intranasal applicator device is a non-human primate precision nasal delivery ("nhpPOD") device described in fig. 7A-E, also described in U.S. patent No. 9,550,036, which is incorporated herein by reference in its entirety. In one embodiment, the intranasal device is one of the embodiments of fig. 1,2 and 9 of U.S. patent No. 9,550,036. In these embodiments, the pharmaceutical compound is loaded directly into the compound chamber.

An example nhpPOD apparatus is shown in figure 6.

Referring to fig. 6, a Metered Dose Inhaler (MDI) canister 602 dispensing 25 μ Ι of hydrofluoroalkane is attached to a plastic actuator 604. The actuator was in gas communication with a polytetrafluoroethylene filter cartridge (frit)1704 with a pore size of 50 μm. The filter cartridge 606 communicates with a dose retention cylinder 610, which is placed inside the body 612 of the POD to generate the aerosolized flow. Upon actuation, the HFA propellant 802 is converted to a gas by passing through the filter material 606, which then mixes with the dose 610; the mixture of the dose and propellant then exits from a 23 gauge stainless steel tube nozzle 614 covered with a fluorinated ethylene-propylene liner that is placed over the outside of the metal tip to protect the nasal epithelium from damage by the nozzle 614 during use. In one embodiment, the dose 610 is loaded directly into the body 612 without the need for a holding cylinder.

Medical unit dose container

In various embodiments, the intranasal administration device is a medical unit dose container as described in US 2016/0101245a1, the disclosure of which is incorporated herein by reference in its entirety.

Intranasal device with access port

In various embodiments, the intranasal applicator is a medical unit dose container as described in U.S. application No. 16/198,312 filed on 21.11.2018, the disclosure of which is incorporated by reference herein in its entirety and repeated below for completeness.

As shown in fig. 5A and 5B, the intranasal device 500 is designed to deliver consistent quality of the compound into the nasal cavity. For example, but not limited to, the compound may be an intranasal formulation in the form of a powder. The device 500 targets specific regions of the nasal cavity with a narrow targeted delivery plume. Specifically, device 500 provides the compound to the upper third of the nasal cavity. In one embodiment, device 500 is used to administer a compound into the upper nasal cavity of a human. The upper nasal cavity includes the olfactory region and the middle and upper turbinate regions. In another embodiment, device 500 is used to administer a compound into the upper nasal cavity of a non-human primate. Device 500 is also designed to simplify loading of the compound into device 500 by a clinician and use thereof. The device 500 may be reused to administer several doses of the compound.

Fig. 5B illustrates a partial cross-sectional view of a device 500 for intranasal delivery of a compound with a coupled tip, and separately illustrates a perspective view of the tip when disengaged. In the embodiment of fig. 5B, the device 500 includes an actuator body 502, a propellant canister 504, and a tip 506. The tip 506 includes an outer wall 508 and an inner wall 510, an outlet channel 512, an inlet interface 514, one or more grooves 528 (shown in fig. 5C), an outlet orifice 516, and a nozzle 518. Fig. 5B illustrates a compound container 520 coupled to the inlet interface 514. The compound contained in the compound container 520 may be a liquid or a powder. In the embodiment of fig. 5B, the compound is a powder.

As shown in fig. 5B, the device 500 includes a propellant canister 504 located within an actuator body 502. The propellant canister 504 contains a propellant. In one embodiment, the propellant may be pressurized. The propellant is a fluid, e.g., a liquid or a gas. In one aspect, the propellant is a liquid. In another aspect, the propellant is a gas. The propellant includes a pharmaceutically suitable propellant. Some examples of pharmaceutically suitable propellants include Hydrofluoroalkanes (HFAs), including but not limited to HFA, HFA 227, HFA134a, HFA-FP, HFA-BP, and similar HFAs. In one aspect, the propellant is a liquid HFA. In another aspect, the propellant is gaseous HFA. Additional examples of suitable propellants include nitrogen or chlorofluorocarbons (CFCs). Additionally, the propellant may be pressurized air (e.g., ambient air). The canister 504 may be a Metered Dose Inhaler (MDI) device that includes a pressurized canister and a metering valve 522 (including a valve stem) to meter propellant upon actuation. In one embodiment, a pump fitting (not shown) secures the metering valve 522 to the canister 504 and holds the two components in place during use of the device 500. A range of embodiments of the pump fitting include a fixed interface that retains the pump fitting within the actuator body 502, provides vertical displacement, and prevents rotation during installation of the canister 504.

The propellant canister 504 may have the ability to dispense propellant for a certain number of doses. In one embodiment, the device 500 may be shipped without the canister 504 and the canister 504 may be loaded into the actuator body 502 by a user. In some embodiments, the propellant canister may be replaced with a new propellant canister so that the device 500 may be reused. In one aspect, when the MDI device is actuated, a discrete amount of pressurized HFA fluid will be released. The MDI may contain between about 30 and about 300 actuations, inclusive, of the HFA propellant. The amount of fluid propellant released upon actuation may be between about 20 microliters (μ l) to about 200 μ l of liquid propellant, inclusive.

The actuator body 502 includes a propellant channel 524 that is in fluid communication with the propellant canister 504. The propellant channel 524 is in fluid communication with an inlet interface 514 configured to be coupled to the compound container 520 such that propellant released from the propellant canister 504 can be introduced into the compound container 520 via one or more grooves 528 on the inlet interface 514. In the embodiment of fig. 5B, the propellant channel 524 includes a port 526 at a distal end to receive the tip 506. In this configuration, the tip 506 may be coupled to and decoupled from the actuator body 502 by inserting the tip 506 into the port 526. In other embodiments, the port 526 may be inserted into the tip 506. In some embodiments, the port 526 and/or the tip 506 may include a sealed interface that creates a hermetic seal between the propellant channel 524 and the tip 506 such that propellant released from the canister 504 does not escape the propellant channel 524 but is directed toward the inlet interface 514.

The tip 506 may be coupled to the actuator body 502 and decoupled from the actuator body 502, which enables a user to load and unload the compound container 520 to and from the inlet interface 514. Tip 506 includes an outer wall 508 and an inner wall 510, wherein the inner wall forms an outlet passage 512 extending between the proximal and distal ends of tip 506. An inlet interface 514 is positioned around the distal end of the outer wall 508, the inlet interface 514 coupling a compound container 520. In the embodiment of fig. 5B, the inlet interface 514 is a collar that is insertable into the compound container 520. In other embodiments, the inlet interface 514 can be a ring, band, port, or band that interfaces with the compound container 520. The inlet interface 514 includes one or more grooves 528 (shown in fig. 5C) to direct propellant released from the canister 504 into the compound container 520 coupled to the inlet interface 514. The released propellant then contacts the compound within the compound container 520, agitating and entraining the compound and pushing the compound through the outlet channel 512 and out of the outlet orifice 516 located at the distal end of the outlet channel 512. In the embodiment of fig. 5B, the tip 506 includes a nozzle at the distal end of the outlet channel 512 to direct the released propellant and compound out of the outlet orifice in a narrow plume.

Fig. 5C is a perspective view of a tip 506 and a compound container according to one or more embodiments. In the embodiment of fig. 5C, the compound container 520 is a capsule. The capsule may be made of two parts that fit together. When separated, a portion of the capsule (e.g., a half-capsule, as shown in fig. 5E-5G) may be coupled to the tip 506. In use, the compound container 520 may contain a compound within a capsule. In one example, the compound is a powder. As shown in fig. 5E, the half-capsule includes an outlet opening 532 of the compound container 520. The outlet opening 532 may be coupled to the inlet interface 514 as shown in fig. 5F-5G. In the embodiment of fig. 5F-5G, the inlet interface 514 is inserted into the outlet opening 532, and the compound container 520 may be secured to the inlet interface 514 via an interference fit. In an alternative embodiment, the outlet opening 532 may be inserted into the inlet interface 514. As shown in fig. 5G-5H, the tip 506 has an outer wall 508 and an inner wall 510, with an outlet passage 512 formed by a bore or lumen through the inner wall 510. The outlet opening 532 fits around the inlet port 514 such that the compound container 520 and the outlet channel 512 are in fluid communication.

As shown in fig. 5F, 5G, and 5J, the inlet interface 514 is, for example, a ring, band, port, collar, or band that interfaces with the compound container 520. As shown in fig. 5C, 5E, 5F, 5K, 5L, 5M, 5N, 5O, and 5P, one or more grooves 528 are located on the inlet interface 514 and create a flow path for propellant released from the propellant canister 504 to travel into the compound container 520. Examples of grooves 528 include, but are not limited to, channels, slots, radial ports, or passages. The groove 528 provides a path through the inlet interface 514 through which propellant flows into the compound container 520. In one example, there are a plurality of grooves 528. The grooves 528 may be equally spaced around the inlet interface 514. The grooves 528 may be of equal size to one another or may be of different sizes. The grooves 528 extend along the length of the inlet interface 514 such that when the compound container 520 is coupled to the inlet interface 514, a first portion of each groove 528 is exposed within the propellant channel 524 and a second portion of each groove 528 is positioned within the compound container 520. As shown in fig. 5C, inlet interface 514 includes a ledge 530 that is designed to abut compound container 520 when coupled to inlet interface 514 and groove 528 extends beyond ledge 530 such that groove 528 is not completely covered by compound container 520.

In use, propellant released from the canister 504 flows through the propellant channels 524 and into the compound container 520 via the grooves 528, as shown by the direction of the arrows in fig. 5D. The outlet passage 512 is aligned with the outlet opening 532 of the compound container 520. Propellant flows into the compound container 520 in the groove 528 of the inlet interface 514 to agitate the powder, and powder and propellant exit the compound container 520 via the outlet opening 532 corresponding to the outlet channel 512. The propellant and powder mixture is carried through the outlet channel 512 through the nozzle 518 and exits the device 500 at the outlet orifice 516. In one example, the tip 506 may have one or more outlet orifices. The plume exiting the outlet aperture 516 has a narrow spray plume.

In one example of use of the device 500, at the time of use, the user divides the pre-filled capsule in half. In one example, the capsule is pre-filled with a powdered compound. The half-capsule is coupled to the tip 506 via an inlet interface 514. As shown in fig. 5P and 5Q, the tip 506 is then coupled to the actuator body 502. Propellant gas, for example from a refrigerant or compressed gas source, is directed through propellant channels 524 and toward the filled powder capsules. The groove 528 around the inlet interface 514 of the tip 506 introduces a high velocity jet of propellant gas that agitates the dry powder into a suspension within the propellant gas (data not shown, but confirmed by high speed close-up video). The grooves 528, which introduce gas tangentially to the hemispherical bottom of the compound container 520, create jets that will enhance powder agitation and entrainment. Once the powder is suspended, it is evacuated through the outlet opening 532 into the outlet channel 512 and out of the outlet orifice 516 of the device 500.

Generally, any shrinkage joint will cause powder blockage when accelerating the powder formulation through the restrictive orifice. Since the powder administered by the device 500 is suspended in the propellant gas prior to evacuation, it can be further throttled and directed without clogging the device. Thus, a much greater mass of powder can be delivered through a much smaller exit orifice without the device 500 being too long. The time from propellant actuation to the end of compound delivery is less than 1 second.

A groove 528 in the proximal end of the tip 506 will facilitate the flow of gas into the compound container 520. In one example, HFA gas is directed at the surface of the powder dose present in the compound container 520 (e.g., orthogonally or near orthogonally), which results in rapid agitation and entrainment of the powder. The hemispherical shape of the compound container 520 will facilitate redirection of the gas to the outlet channel 512 of the tip 506, as shown in fig. 5D. The arrows of fig. 5B and 5D illustrate the direction of propellant flow after the device 500 is actuated.

The actuator body 502 is attached and sealed to a propellant canister 504 and a tip 506, creating a pressurized flow path for the propellant gas. In certain aspects, the actuator body 502 is a reusable component. In certain aspects, the canister 504 is a reusable component.

In one example, compound container 520 is a standard size 3 drug capsule, but one skilled in the art would know how to use other sized drug capsules and modify device 500 to fit it. Additionally, in another example, the compound container 520 may not be a capsule, but another container capable of holding a compound, such as, but not limited to, an ampoule. In one example, the ampoule may be made of plastic, and in one example, it may be a blow-filled ampoule. To load the device 500, the user or clinician detaches the capsule containing the pre-filled formulation, discards the cap, and mounts the capsule over the tip 506. It is also possible for a clinician to fill an empty compound container 520 at the time of use and then mount the compound container 520 to the tip 506. In certain examples, the capsule is a disposable component.

The tip 506 receives the compound container 520 during loading and then couples to the actuator body 502 prior to use. When the propellant canister 504 is actuated, expanding propellant gas is introduced into the compound container 520 via the groove 528 around the inlet interface 514 of the tip 506. The resulting jet of propellant gas agitates and entrains the powder formulation within the compound container 520 which then exits through the outlet passage 512 and the outlet orifice 516 of the tip 506. In one example, the tip 506 is a disposable component. FIG. 5K illustrates an example measurement of the tip 506 in inches. In the embodiment of fig. 5N, the inlet interface 514 can include a radius along the bottom edge 222 to aid in placement of the compound container 520 onto the tip 506. The radius of curvature may range between about 0.005 inches and 0.025 inches, inclusive.

Fig. 5T and 5U illustrate perspective views of a second embodiment 534 of the tip. Similar to the tip 506, the tip 534 can be coupled to and decoupled from the actuator body 502, which enables a user to load and unload the compound container 536 from the tip 534 to use the device 500 for delivery to the user's upper nasal cavity. As shown in fig. 5T and 5U, the compound container 536 is a capsule. In one example, the compound container 536 can contain a powder. In the embodiment of fig. 5T and 5U, the tip 534 includes an inlet interface 538 for coupling with a compound container 536, wherein the inlet interface 538 has a piercing member 540. The piercing member 540 is designed to pierce the compound container 536 to create an opening in the compound container 536. The piercing member 540 may include a sharp point, a sharp corner, a knife-like edge, or other suitable geometry for piercing the compound container 536. In one embodiment, the inlet interface 538 comprises more than one piercing member 540, wherein each piercing member 540 is designed to pierce a compound container 536. The piercing members 540 may be positioned around the inlet interface 538 in a symmetrical or random pattern. In one embodiment, in use, a user can remove the spike 534 from the actuator body 502, load the compound container 536 into the port 526 of the propellant channel 524, and then insert the spike 534 back into the port 526. As the tip 534 is coupled to the actuator body 502, the piercing member 540 pierces the capsule. In this configuration, as shown in fig. 5U, the pierced capsule fits around piercing member 540. In an alternative embodiment, piercing member 542 can include a plurality of piercing points 544 that respectively pierce compound container 536. The plurality of puncture points 544 can be spaced around puncture member 542, or each

Fig. 5V and 5W illustrate perspective views of a piercing member 542 that can be used with the tip 534 according to one or more embodiments. As shown in fig. 5V, piercing member 542 can be a collar, ring, band, port, or band coupled to the pierced compound container 536. The piercing member 542 includes one or more piercing grooves 546, similar to the grooves 528, which form a flow path between the propellant channel 524 and the compound container 536. Propellant from the propellant canister 504 enters via one or more piercing grooves 546 of the piercing member 542, flows along the piercing grooves 546 and into the pierced compound container 536. As shown in fig. 5V and 5W, piercing member 542 includes a plurality of piercing openings 548. In the embodiment of fig. 5V, 5W, 5X, the puncture opening 548 is in fluid communication with the outlet channel 512. Propellant from the propellant canister 504 flows into the piercing recess 546, mixes with the powder in the pierced compound container 536, and flows into the piercing opening 544 to the outlet channel 512. The arrows in fig. 5X illustrate the flow path of the propellant. The outlet channels 512 provide a path for propellant and powder to the nozzle 518 and outlet orifice 516. The mixture of propellant and powder exits the device 500 via the outlet orifice 516. The plume exiting the device 500 is a narrow spray plume. In this embodiment, piercing member 542 may be integrally molded as a single piece or may be comprised of two or more pieces. In one example, the piercing member 542 may be a separate molded piece that acts in association with the inlet interface 538 (where the capsule is attached). In some embodiments, the inlet interface may include more than one piercing member 542.

As an alternative to a capsule being manually separated prior to placement on the tip 534, the tip 534 may include an integral piercing member 542 and piercing recess 546, as shown in fig. 5V and 5W. To create repeatable puncturing of the compound container 536, the piercing member 542 forms a single point, piercing point 544. In one example, puncture point 544 includes puncture openings 546 spaced radially about puncture point 544. The piercing opening 546 is in fluid communication with the outlet passage 512 to evacuate the powder from the compound container 536.

By allowing the propellant flow path to be created by the inline piercing motion, as shown in fig. 5X, loading of the compound container 536 onto the nib 534 is simplified for the user because the compound container 536 does not require manual manipulation and separation. In one example, piercing member 542 is integrally formed with tip 534. In one example, the filled compound container 536 can be filled and installed into the actuator body 502 or the tip 534 during manufacture of the device 500. In use, a user may apply linear motion to drive the piercing member 542 into the pre-filled compound container 536, thereby creating a complete gas flow path for metering prior to propellant actuation.

The present invention is further described in the following examples, which are not intended to limit the scope of the present invention.

Powder capsule

In one embodiment, a device was constructed and tested. The residual powder in the compound container after actuation was tested. When 2 or more but less than 6 grooves are used on the inlet interface, the device has comparable powder delivery performance as measured by the residue after actuation. In this example, the groove was combined with 63mg HFA propellant and the.040 "outlet orifice of the nozzle. Four grooves (every 90 degrees) were found to provide uniform gas delivery.

Dose mass

A dose mass reproducibility test was performed. The standard deviation of dose delivery indicates that the device is capable of delivering consistent dose quality. The average residual dose amount remaining in the device was < 5%, indicating that very little dose was lost in the device.

Figure BDA0002663715770000241

Intranasal devices with multiple cartridges

Fig. 7A illustrates another example of a non-human primate precision nasal delivery device 700, and fig. 7B illustrates a side view and a cross-sectional view of an actuator body 710 of the inter-nasal device 700 of fig. 7A. The device 700 may deliver the compound as a liquid, a powder, or some combination thereof. The device 700 includes a propellant canister 705, an actuator body 710, an extension tube 715 and a tip 720. Similar to the device 1, the propellant canister 705 is in fluid communication with the actuator body 710 such that propellant released from the propellant canister 705 travels through the actuator body 710, through the extension tube 715, through the nib 720, and out the outlet opening 725 of the nib 720. The compound may be loaded into the tip 720 such that as the propellant travels through the tip 720, the propellant contacts the compound and propels the compound to the outlet opening 725, where the propellant and compound exit in a plume.

Figure 7C illustrates a side view of the extension tube 715 of the inter-nasal device 700 of figure 7A. The extension tube 715 is a tube that includes an internal passage that creates fluid communication between the actuator body 710 and the tip 720. In the embodiment of fig. 7A-7D, a first end 730 of the extension tube 715 is coupled to the actuator body 710, while a second end 735 of the extension tube 715 is coupled to the tip 720, each of which is coupled via a respective connection interface 740a, 740b (collectively "740"). The connection hub 740 includes a luer lock with a male or female end on each side of the luer lock. In the embodiment of fig. 7A-7D, each connection interface 740 includes a luer lock having two male ends. Thus, the male ends of the connection interface 740a are inserted into the actuator body 710 and the first end 730, respectively, while the male ends of the connection interface 740b are inserted into the tip 720 and the second end 735, respectively. As illustrated in fig. 7C, the second end 735 may include a plurality of filter cartridges 745 positioned within the interior passage of the luer lock. The filter element 745 may be configured to convert the liquid propellant to a gas as the propellant passes through the filter element 745. Alternatively, the extension tube 715 in fig. 7B may be configured to convert the liquid propellant into a gas. The filter element 745 may be comprised of a porous material. The number of filter elements 745 may vary in different embodiments. As the number of filter elements increases, the intensity of the plume may decrease, for example, in terms of its impact force, velocity, plume width, other similar criteria, or some combination thereof. Similarly, the length of the extension tube 715 may be adjusted so that the propellant travels through a longer or shorter distance. Calibrating the intensity of the plume may enable the device 700 to accurately deliver the compound to the nasal cavity. Fig. 7D illustrates an enlarged view of the connection interface 740b at the second end 735 of the extension tube 715 of fig. 7C — the first example embodiment 750 includes a single filter element 745 and the second example embodiment 755 includes three filter elements 745 stacked in series. The number of filter elements 745 may be selected based on the type of compound. For example, a single cartridge 745 may be used for powder compounds while three cartridges 745 may be used for liquid compounds, or vice versa.

Figure 7E illustrates a side view and a cross-sectional view of the tip 720 of the inter-nasal device of figure 7A. The tip 720 is designed to be inserted into a nostril. The tip 720 includes an internal passage 760 and an outlet opening 725 for delivering the compound to the nasal cavity. In the embodiment of fig. 7E, the tip 720 includes a filter element 745 disposed within the interior passage 760. The filter element 745 may be configured to convert the liquid propellant to a gas as the propellant passes through the filter element 745. The filter element 745 may be comprised of a porous material. In the embodiment of fig. 7E, the tip 720 further includes a nozzle 765 at the distal end of the tip 720 proximate the outlet opening 725. The nozzles 765 can enhance deposition of the compound within the nasal cavity, such as to the upper olfactory region of the user. In some embodiments, the nozzle 765 can comprise a single orifice, while in alternative embodiments, the nozzle 765 can comprise multiple orifices (e.g., between 2 and 11 orifices). In some embodiments, the tip 720 may not include a nozzle. Different embodiments of the tip may be used based on the different types of compounds to be delivered to the nasal cavity of the user. For example, a tip for delivering a powder compound may not include a nozzle, while a tip for delivering a liquid compound may include a nozzle, or vice versa. In addition, the number of orifices in the nozzle may similarly vary based on the type of compound. A compound may be loaded into the tip 720 such that the compound is contained within the internal passage 760. In the embodiment of fig. 7E, compound is loaded into the tip 720 through the opening 770 at the proximal end of the tip 720 prior to disposing the filter element 745 within the internal passage 760. The cartridge 745 is then inserted to contain the compound inside the tip 720. In an alternative embodiment, such as one in which the tip 720 does not include a nozzle 765, the compound can be loaded into the tip through the outlet opening 725. In the configuration of fig. 7E, propellant travels from the propellant canister 705 through the actuator body 710 and extension tube 715, through the nib 720 and contacts the filter element 745, and then contacts the compound within the internal passage 760, pushing the compound through the outlet opening 725 where the propellant and compound exit in a plume that is delivered within the user's nasal cavity.

Effective dose

In the methods described herein, an effective dose is a dose of a dry powder composition comprising olanzapine in an amount effective to reduce agitation. In some embodiments, the effective dose is a dose comprising olanzapine in an amount effective to reduce agitation within 60 minutes, within 50 minutes, within 40 minutes, within 30 minutes, within 20 minutes, or within 10 minutes.

In some embodiments, an effective dose of the dried pharmaceutical composition comprises 1-30mg, 2-20mg, 5-15mg, 5mg, 6mg, 7mg, 8mg, 9mg, 10mg, 11mg, 12mg, 13mg, 14mg, 19,15mg, 16mg, 17mg, 18mg, 19mg, or 20mg olanzapine.

In some embodiments, the effective dose is administered as a single undivided dose. In some embodiments, the effective dose is administered in multiple divided sub-doses.

Patient's health

In the methods described herein, intranasal administration of olanzapine is used for the acute treatment of agitated patients. In some embodiments, the patient is a restless emergency department patient.

In some embodiments, the patient has schizophrenia, bipolar disorder, dementia, or autism. In some embodiments, the patient has bipolar I disorder. In some embodiments, the patient has acute agitation not associated with schizophrenia, bipolar disorder, or autism. In certain embodiments, the patient has refractory panic disorder, post-traumatic stress disorder, agitation associated with dementia, agitation associated with drug-induced psychotic states, intoxication, or agitation/aggression plus mental disability.

PK

In various embodiments of the methods described herein, intranasal administration provides (a) a mean peak plasma olanzapine concentration (C) of at least 20ng/mLmax) And (b) C to olanzapine in less than 1.5 hoursmaxAverage time (T) ofmax)。

In some embodiments, the intranasal administration provides a mean peak plasma olanzapine concentration (Cmax) of at least 25ng/mL, at least 30ng/mL, at least 40ng/mL, at least 50ng/mL, at least 60ng/mL, at least 70ng/mL, or at least 80ng/mLmax)。

In some embodiments, the intranasal administration provides a C to olanzapine of less than 1.0 hour, less than 0.75 hour, less than 0.50 hour, or less than 0.25 hourmaxAverage time (T) ofmax)。

In a currently preferred embodiment, the intranasal administration provides a mean peak plasma olanzapine concentration of at least 40ng/mL and a Cmax of less than 30 minutes, or more preferably less than 20 minutesmaxAverage time (T) ofmax)。

Dry pharmaceutical composition

In another aspect, a dry pharmaceutical composition suitable for intranasal administration is provided. The composition comprises olanzapine and at least one excipient.

In typical embodiments, the dry pharmaceutical composition is a powder.

In some embodiments, the composition comprises olanzapine in crystalline form. In some embodiments, the composition comprises olanzapine in an amorphous form. In some embodiments, the composition comprises olanzapine in a partially crystalline and partially amorphous form. In a particular embodiment olanzapine is an amorphous solid obtained by spray drying. In some embodiments, the composition comprises olanzapine in a crystalline form and an amorphous form.

In typical embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the powder is from 1 μm to 500 μm as measured by a laser diffraction particle size analyzer such as a Malvern Panalytical Mastersizer 3000. In some embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the powder is from 1 μm to 250 μm, from 1 μm to 100 μm, from 1 μm to 75 μm, from 1 μm to 50 μm, from 1 μm to 25 μm, from 1 μm to 20 μm, from 1 μm to 15 μm, or from 2 μm to 15 μm. In certain embodiments, the median diameter (D50) of the particle size distribution of olanzapine in the composition is from 2 μm to 5 μm or from 7.5 μm to 15 μm.

In various embodiments, the dry pharmaceutical composition comprises no more than 70% olanzapine by weight. In some embodiments, the composition comprises no more than 60% olanzapine by weight. In some embodiments, the composition comprises 10-70% olanzapine by weight, 20-70% olanzapine by weight, 10-60% olanzapine by weight, 20-60% olanzapine by weight, 25-55% olanzapine by weight, 30-50% olanzapine by weight, 30-40% olanzapine by weight, or 40-50% olanzapine by weight.

In some embodiments, the pharmaceutical composition further comprises a stabilizer selected from the group consisting of: hydroxypropyl methylcellulose (HPMC), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer (Soluplus), vinylpyrrolidone-vinyl acetate copolymer (Kollidon VA64), polyvinylpyrrolidone K30(Kollidon K30), polyvinylpyrrolidone K90(Kollidon K90), hydroxypropyl cellulose (HPC), hydroxypropyl β -cyclodextrin (HPBCD), mannitol, and lactose monohydrate. In some embodiments, the stabilizer is Hydroxypropylmethylcellulose (HPMC).

In some embodiments, the dry pharmaceutical composition further comprises a penetration enhancer selected from the group consisting of: n-tridecyl- β -D-maltoside, n-dodecyl- β -D-maltoside, 1, 2-distearoyl-sn-glycero-3-phosphocholine (DSPC), 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1, 2-dioleoyl-sn-glycero-3-phosphocholine (DOPC), propylene glycol, disodium EDTA, PEG400 monostearate, polysorbate 80, and polyethylene glycol (15) hydroxystearate. In some embodiments, the penetration enhancer is 1, 2-distearoyl-sn-glycerol-3-phosphocholine (DSPC).

In some embodiments, the dry pharmaceutical composition comprises both HPMC and DSPC.

In various embodiments, the dry pharmaceutical composition further comprises a nonionic surfactant. In certain embodiments, the nonionic surfactant is an alkyl maltoside. In a particular embodiment, the alkyl maltoside is n-dodecyl β -D-maltoside. In some embodiments, the nonionic surfactant is present in the dry powder composition from 0.1 to 10 weight percent, more typically from 1 to 5 weight percent. In a particular embodiment, the nonionic surfactant is present at 1% by weight. In some embodiments, the nonionic surfactant is Pluronic PF 68. In some embodiments, the nonionic surfactant is present in the dry powder composition from 20 to 40 weight percent, more typically from 25 to 35 weight percent. In a particular embodiment, the nonionic surfactant is present at 31 wt%.

In some embodiments, the pharmaceutical composition further comprises an antioxidant selected from the group consisting of: alpha-tocopherol, ascorbic acid, ascorbyl palmitate, bronopol Butylated Hydroxyanisole (BHA), Butylated Hydroxytoluene (BHT), citric acid monohydrate, sodium ascorbate, ethylenediaminetetraacetic acid, fumaric acid, malic acid, methionine, propionic acid, sodium metabisulfite, sodium sulfite, sodium thiosulfate, thymol, and vitamin E polyethylene glycol succinate.

In some embodiments, the dry pharmaceutical composition further comprises an acid. In certain embodiments, the acid is citric acid. In some embodiments, the acid is present in the dry powder composition at 10 to 20 wt.%, more typically 15 to 20 wt.%. In a particular embodiment, the citric acid is present at 18% by weight.

In various embodiments, the dry pharmaceutical composition further comprises a salt of a monovalent inorganic cation. Typically, the salt is NaCl. In some embodiments, the composition comprises 1-5% by weight NaCl or 2-4% by weight NaCl.

In some embodiments, the dry pharmaceutical composition further comprises less than 3%, less than 2.5%, less than 2%, less than 1.5%, less than 1%, less than 0.9%, less than 0.8%, less than 0.7%, less than 0.6%, or less than 0.5% by weight water.

In a currently preferred embodiment, the dry pharmaceutical composition comprises 50% olanzapine by weight, 42% HPMC by weight and 8% DSPC. In some embodiments, the dry pharmaceutical composition is a spray-dried composition comprising amorphous olanzapine. In some embodiments, olanzapine is spray dried in the presence of HPMC and/or DSPC. In other embodiments, HPMC and/or DSPC is added after spray drying of olanzapine.

Unit dosage form

In another aspect, a unit dosage form is provided. The unit dosage form contains a dry pharmaceutical composition as described above in section 5.5.

In typical embodiments, the unit dosage form contains 1-30mg olanzapine. In some embodiments, the unit dosage form contains 2 to 20mg olanzapine. In some embodiments, the unit dosage form contains 5-15mg olanzapine. In some embodiments, the unit dosage form contains 5mg olanzapine. In some embodiments, the unit dosage form contains 10mg olanzapine. In some embodiments, the unit dosage form contains 15mg olanzapine.

In some embodiments, the unit dosage form is a capsule encapsulating the dry pharmaceutical composition. In some embodiments, the capsule is a hard capsule. In some embodiments, the hard capsule is an HPMC hard capsule.

In some embodiments, the unit dosage form is a dosage container storing a dry pharmaceutical composition, wherein the dosage container is configured to be removably coupled to an intranasal delivery device. In particular embodiments, the dose container is a tip configured to be removably coupled to an intranasal delivery device.

Experimental examples

The invention is further described by reference to the following experimental examples. These embodiments are provided for illustrative purposes only and are not intended to be limiting.

79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:包含至少一种PI在5.8与8.5之间的基础胰岛素和带有羧酸根电荷及疏水基的共聚氨基酸的PH 7为7的可注射溶液

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!