Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FORMULATIONS OF PTHRP ANALOGUES, TRANSDERMAL PATCHES THEREOF, AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2017/062922
Kind Code:
A1
Abstract:
Disclosed are PTHrP analogue formulations for transdermal delivery of a therapeutically effective amount of a PTHrP analogue, as well as transdermal patches prepared using these formulations, methods of preparing the disclosed formulations and patches, and methods of using these formulations and patches to treat osteoporosis, osteopenia, osteoarthritis, and/or bone fracture, improve bone mineral density (BMD), improve trabecular bone score (TBS), and treat, prevent, and/or reduce bone fractures.

Inventors:
HATTERSLEY GARY (US)
HARRIS ALAN (US)
SAEH JAMAL (US)
HAMED EHAB (US)
Application Number:
PCT/US2016/056196
Publication Date:
April 13, 2017
Filing Date:
October 08, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
RADIUS HEALTH INC (US)
International Classes:
A61M37/00; A61K38/29; C07K14/635
Foreign References:
US20140343499A12014-11-20
US20080039775A12008-02-14
US20140046293A12014-02-13
Other References:
AMERI ET AL.: "Demonstrated Solid-State Stability of Parathyroid Hormone PTH(134) Coated on a Novel Transdermal Microprojection Delivery System", PHARMACEUTICAL RESEARCH, vol. 26, no. Issue 11, November 2009 (2009-11-01), pages 2454 - 2463, XP019752630
See also references of EP 3359241A4
Attorney, Agent or Firm:
WANG, Yingli (US)
Download PDF:
Claims:
What is claimed is:

1. A preparation formulation suitable for coating a transdermal patch wherein said

preparation formulation comprises a therapeutically active substance, water and one or more excipients selected from the group consisting of Zn2+ salts, Mg2+ salts, Ca2+ salts, polyethylene glycols and hydroxypropyl beta-cyclodextrins.

2. The preparation formulation of claim 1 further comprising a bioactive peptide or

protein.

3. The preparation formulation of claims 1-2 comprising an antibody.

4. The preparation formulation of claims 1-2 comprising a bioactive peptide.

5. The preparation formulation of claim 4 wherein said bioactive peptide contains at least 10 amino acids.

6. The preparation formulation of claim 5 wherein said bioactive peptide is

abaloparatide as set forth in SEQ ID NO: 1.

7. The preparation formulation of any one of the preceding claims wherein said

excipients are selected from the group consisting of ZnCl2, Zn(OAc)2, Zn3(P04)2, ZnCitrate, ZnOxalate, MgO, MgCitrate, MgS04, MgOrotate, MgLactate, MgC03 CaSorbate, CaCitrate, CaAscorbate, Ca3(P04)2, CaCl2, CaC03, CaS04, and Ca(OAc)2.

8. The preparation formulation of claim 7 wherein said excipient is selected from ZnCl2 and Zn(OAc)2 and combinations thereof.

9. The preparation formulation of any of the preceding claims wherein the molar ratio of said excipient or excipients to said therapeutically active substance is 0.1 to 2.0.

10. The preparation formulation of any of the preceding claims wherein the molar ratio of said excipient or excipients to said therapeutically active substance is 0.2 to 1.5.

11. The preparation formulation of any of the preceding claims wherein the molar ratio of said excipient or excipients to said therapeutically active substance is 0.25 to 1.0.

12. A transdermal patch comprising a plurality of microprojections wherein at least one microprojection in the array is covered at least in part by a coating, said coating comprising a therapeutically active substance and one or more excipients selected from the group consisting of Zn2+ salts, Mg2+ salts, Ca2+ salts, polyethylene glycols and hydroxypropyl beta-cyclodextrins.

13. The transdermal patch of claim 12, wherein the microprojections are microneedles.

14. The transdermal patch of any one of claims 12-13 wherein said therapeutically active substance comprises a bioactive peptide or protein.

15. The transdermal patch according to any of claims 12-14 wherein said therapeutically active substance comprises an antibody.

16. The transdermal patch according to any of claims 12-14 comprising a bioactive

peptide.

17. The transdermal patch according to claim 17 wherein said bioactive peptide contains at least 10 amino acids.

18. The transdermal patch according to claim 17 wherein said bioactive bioactive peptide is abaloparatide as set forth in SEQ ID NO 1.

19. The transdermal patch according to claims 12-18 wherein and said excipients are selected from the group consisting of ZnCl2, Zn(OAc)2, Zn3(P04)2, ZnCitrate, ZnOxalate, MgO, MgCitrate, MgS04, MgOrotate, MgLactate, MgC03 CaSorbate, CaCitrate, CaAscorbate, Ca3(P04)2, CaCl2, CaC03, CaS04, and Ca(OAc)2.

20. The transdermal patch according to any one of claims 12-19 wherein said excipient is selected from ZnCl2 and Zn(OAc)2 and combinations thereof.

21. The transdermal patch according to any one of claims 12-20 wherein the molar ratio of said excipient or excipients to said therapeutically active substance is 0.1 to 2.0.

22. The transdermal patch according to any one of claims 12-21 wherein the molar ratio of said excipient or excipients to said therapeutically active substance is 0.2 to 1.5.

23. The transdermal patch according to any one of claims 12-22 wherein the molar ratio of said excipient or excipients to said therapeutically active substance is 0.25 to 1.0.

24. The transdermal patch according to any one of claims 18-23 wherein said

abaloparatide is present on said microprojection array in an amount between 90-110 μg, 140-160 μg, 185-220 μg, 225-275 μg or about 100 μg, about 150 μg, about 200 μg or about 250 μg.

25. A method of treating a condition selected from the group consisting of osteoporosis, osteopenia, osteoarthritis, and bone fracture in a subject comprising administering a transdermal patch according to any one of claims 18-24.

26. A method of preventing vertebral, non-vertebral, clinical and major osteoporotic fractures comprising administering a transdermal patch according to any one of claims 18-24.

27. A method of improving bone mineral density (BMD), improving trabecular bone score (TBS), and/or reducing bone fractures in a subject comprising administering to the subject a transdermal patch according to any one of claims 18-24.

28. The method according to any one of claims 25-27 wherein said patch comprises between 300-750 microprojections.

29. The method according to any one of claims 25-28 wherein said administration

comprises application of a force to the transdermal patch sufficient to drive one or more of the microprojections through the stratum corneum of the patient.

30. The method according to any one of claims 25-29 where the site of administration is the abdomen or the thigh.

Description:
FORMULATIONS OF PTHrP ANALOGUES, TRANSDERMAL PATCHES THEREOF, AND USES THEREOF

PRIORITY CLAIM

[0001] The present application claims priority to U.S. Provisional Application No.

62/239,773, filed October 9, 2015, U.S. Provisional Application No. 62/239,774, filed October 9, 2015, U.S. Provisional Application No. 62/239,801, filed October 9, 2015, U.S. Provisional Application No. 62/324,336, filed April 18, 2016, U.S. Provisional Application No. 62/353,249, filed June 22, 2016, and U.S. Provisional Application No. 62/396,196, filed September 18, 2016, all of which are incorporated herein by reference in their entirety, including drawings.

INTRODUCTION

[0002] Conventionally, osteoporosis is treated by administration of antiresorptive agents to suppress bone resorption. The most common of these treatments is oral or intravenous administration of bisphosphonates. However, an undesirable side effect of bisphosphonate administration is reduced bone formation (MacLean 2008). Anabolic agents provide an alternative to antiresorptives. The only anabolic agent currently available for treatment of osteoporosis is teriparatide (PTH (1 -34), Forteo®), a recombinant form of parathyroid hormone (PTH) that acts by a mechanism that involves stimulating new bone formation (along with resorption) and reconstituting internal bone microarchitecture (Recker 2009; Dempster 2012; Ma 201 1 ). The effects of teriparatide on bone mineral density (BMD) are superior to antiresorptive agents at the spine, but its effects at the hip are more modest, and often delayed until the second year of a two-year course of therapy (Leder 2014; Neer 2001).

[0003] Parathyroid hormone-related protein (PTHrP; UniProt Accession No. PI 2272) shares some homology with parathyroid hormone (PTH) at their N-terminal ends, and both proteins bind to the same G-protein coupled receptor, PTH receptor type-1 (PTH1R).

Despite a common receptor, PTH primarily acts as an endocrine regulator of calcium homeostasis whereas PTHrP plays a fundamental paracrine role in the mediation of endochondral bone development ( ronenberg 2006). The differential effects of these proteins may be related not only to differential tissue expression, but also to distinct receptor binding properties (Pioszak 2009; Okazaki 2008; Dean 2008). Over the past several years, PTHrP and its secretory forms (PTHrP(l-36), PTHrP(38-94), and osteostatin), as well as analogues thereof, have been investigated as potential treatments for osteoporosis.

Subcutaneous injection of PTHrP and its derivatives and analogues has been reported to be effective for treating osteoporosis and/or improving bone healing (Horwitz 2010; Horwitz 2006; Bostrom 2000; Augustine 2013).

[0004] Therefore, it is desirable to have an alternative delivery route that is both effective for treatment (e.g., a substantial bioequivalence of the subcutaneous delivery of PTHrP and/or derivatives and analogues thereof) and easy for administration to improve patients' satisfaction and compliance.

SUMMARY OF THE INVENTION

[0005] Provided herein in certain embodiments are preparation formulations for use in transdermal delivery of PTHrP analogues such as abaloparatide comprising a PTHrP analogue (e.g., abaloparatide) and one or more excipients selected from the group consisting of Zn 2+ salts (e.g., ZnCl 2 , Zn(OAc)2, Zn 3 (P04)2, ZnCitrate, ZnOxalate, etc., or combinations thereof), Mg 2+ salts (e.g., MgO, MgCitrate, MgS0 4) MgOrotate, MgLactate, MgC0 3 , MgCl 2 , Mg(OAc) 2 , etc., or combinations thereof) Ca 2+ salts (e.g., CaSorbate, CaCitrate,

CaAscorbate, Ca 3 (P0 4 )2, CaCl 2 , CaC0 3 , CaS0 4j Ca(0Ac) 2 , etc., or combinations thereof), PEG (polyethylene glycol), PVP (polyvinylpyrrolidone), cyclodextrin (CD, e.g., 2- hydroxypropyl-P-cyclodextrin (HPpCD)), salts of carboxylic acids including fatty acids, NaCl and histidine and various combinations thereof. In certain embodiments, the preparation formulation further comprises water for injection, saline or phosphate buffered saline (PBS). In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide ([Glu 22,25 , Leu 23,28 ' 31 , Aib 29 , Lys 6 ' 3 PTHrP(l-34)NH 2 ), which has the amino acid sequence set forth in SEQ ID NO: l . In certain embodiments, the PTHrP analogue is delivered by a transdermal patch comprising at least one microprojection (e.g., microneedle) prepared using the preparation formulation.

[0006] Provided herein in certain embodiments are patches for transdermal administration of a PTHrP analogue comprising one or more microprojections prepared using a preparation formulation as disclosed herein.

[0007] Provided herein in certain embodiments are methods of preparing a transdermal patch for administration of a PTHrP analogue comprising preparing at least a microprojection on a blank transdermal patch with a preparation formulation disclosed herein. In certain embodiments, the microprojections are microneedles.

[0008] Provided herein in certain embodiments are methods for treating osteoporosis, treating osteopenia, treating osteoarthritis, improving bone mineral density (BMD), improving trabecular bone score (TBS), and treating, preventing, and reducing bone fractures in a subject comprising transdermally administering a therapeutically effective amount of a PTHrP analogue via a transdermal patch comprising at least one microprojections prepared using PTHrP analog preparation formulation as disclosed herein. In some embodiments the osteoporosis being treated is postmenopausal osteoporosis. In some embodiments the osteoporosis being treated is glucocorticoid induced osteoporosis. In certain of these embodiments, the preparation formulation is administered via a transdermal patch as disclosed herein. The bone fractures being treated, prevented, or reduced and the bone with improved BMD or TBS may be vertebral or non- vertebral.

BRIEF DESCRIPTION OF DRAWINGS

[0009] Figures 1 A-1C: Pharmacokinetic profile of various formulations of abaloparatide administered by transdermal versus subcutaneous routes. Figure 1 A: One possible bioequivalence "window" of the abaloparatide-SC treatment, % scale on the vertical axis indicates the plasma abaloparatide concentration represented by % of its own maximum (Cmax), i.e. 100 = Cmax, hereinafter referred to as the "normalized plasma concentration." Figure IB: transdermal delivery in monkeys using a preparation formulation of abaloparatide comprising ZnCl 2 , the vertical axis indicates normalized peptide plasma concentration.

Figure 1C: transdermal delivery using a preparation formulation of abaloparatide comprising PEG.

[0010] Figure 2: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes (SC). The abaloparatide preparation formulation of the abaloparatide for transdermal delivery did not comprise ZnCl 2 or PEG, note the very quick Cmax of the transdermal delivery compare to SC and the increasing pulsatile nature of the delivery. Square: transdermal delivery (TD); and diamond: the abaloparatide-SC treatment. Administration in healthy postmenopausal women, % scale on vertical axis indicates the normalized plasma concentration of abaloparatide represented by % of C ma x for each group.

[0011] Figure 3: Pharmacokinetic profile of formulations of abaloparatide containing ZnCl 2 administered by transdermal (TD) versus subcutaneous (SC) routes, longitude of median plasma abaloparatide concentration v. time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnC^. Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined. Note the more prolonged release afforded by the ZnCl 2 addition.

[0012] Figure 4: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, median plasma abaloparatide concentration v. time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μ TD studies combined.

[0013] Figure 5: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, longitude of mean plasma abaloparatide concentration v, time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0014] Figure 6: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, mean plasma abaloparatide concentration v. time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0015] Figure 7: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, longitude of median of dose normalized plasma abaloparatide concentration v. time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 .

Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0016] Figure 8: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, median of dose normalized plasma abaloparatide concentration v. time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0017] Figure 9: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, longitude of mean of dose normalized plasma abaloparatide concentration v. time post administration. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0018] Figure 10: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal versus subcutaneous routes, mean of dose normalized plasma abaloparatide concentration v. time post administration in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0019] Figure 1 1 : Comparison of C max of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0020] Figure 12: Comparison of C max of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0021] Figure 13: Comparison of AUQ ast of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0022] Figure 14: Comparison of AUC| ast of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0023] Figure 15: Comparison of AUQ n f of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0024] Figure 16: Comparison of AUQnf of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0025] Figure 17: Comparison of C max /D (C max per dosage) of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCI 2 ) and represents all 150 μg TD combined.

[0026] Figure 18: Comparison of C max /D (C ma x per dosage) of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCI 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0027] Figure 19: Comparison of CL/F of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCI 2 ) and represents all 150 μg TD studies combined.

[0028] Figure 20: Comparison of CL/F of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCI 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 μg TD studies combined.

[0029] Figure 21 : Comparison of HL_Lambda_z of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCI 2 .

Historic TD used abaloparatide formulations in PBS buffer (no ZnCl ) and represents all 150 μg TD studies combined.

[0030] Figure 22: Comparison of HL_Lambda_z of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 .

Historic TD used abaloparatide formulations in PBS buffer (no ZnCl 2 ) and represents all 150 g TD studies combined.

[0031] Figure 23: Comparison of Tm ax of formulations of abaloparatide administered by transdermal versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 . Historic TD used abaloparatide formulations in PBS buffer (no ZnCI 2 ) and represents all 150 μg TD studies combined. [0032] Figure 24: Comparison of T max of formulations of abaloparatide administered by transdermal (abdomen) versus subcutaneous routes in healthy postmenopausal women. Transdermal administration was to the abdomen with a formulation including ZnCl 2 .

Historic TD used abaloparatide formulations in PBS buffer (no ZnCI 2 ) and represents all 150 μg TD studies combined.

[0033] Figure 25: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal route (abdomen) in a selected patient versus historical subcutaneous data in healthy postmenopausal women. TD used abaloparatide formulations PBS buffer with no Zn salt added.

[0034] Figure 26: Percent BMD changes from baseline at lumber spine of the subjects treated with abaloparatide via transdermal delivery or SC injection in postmenopausal women with osteoporosis. Transdermal delivery used abaloparatide formulations with PBS buffer (no Zn salt).

[0035] Figure 27: Percent BMD changes from baseline at total hip of the subjects treated with abaloparatide via transdermal delivery or SC injection in postmenopausal women with osteoporosis. Transdermal delivery used abaloparatide formulations with PBS buffer (no Zn salt).

[0036] Figure 28: Local tolerance data of the subjects treated with abaloparatide via transdermal delivery in postmenopausal women with osteoporosis. Transdermal delivery used abaloparatide formulations with PBS buffer (no Zn salt).

[0037] Figure 29: Pharmacokinetic profile of formulations of abaloparatide administered by transdermal delivery used abaloparatide formulations with PBS only (no Zn salt) in healthy postmenopausal women (diamond) versus subcutaneous (square) routes. Note the very rapid and pulsatile release in the transdermal delivery compared to the SC

administration.

[0038] Figures 30A-30B: PK/PD relationship of formulations of abaloparatide administered by transdermal and by subcutaneous routes all in postmenopausal women with osteoporosis. Figure 30A: C max v. BMD improvement (%) for formulations of abaloparatide administered by transdermal and by subcutaneous routes. Figure 30B: AUC v. BMD improvement (%) for formulations of abaloparatide administered by transdermal and by subcutaneous routes.

[0039] Figure 3 1 : Comparison of pK curves from a sc combination cohort (80 g), 1 st generation transdermal (200 μg abaloparatide + PBS buffer only) and 2 nd generation transdermal (200 μg abaloparatide + ZnCl 2 ). Values are the geometric means. [0040] Figure 32: Comparison of pK curves of selected individual patients being treated with the 2 nd generation transdermal (200 μg abaloparatide plus ZnCl 2 ) and compared to a reference set of sc treated patients. Values are the geometric means.

[0041] Figure 33 : Concentration-time graph after administration of patch formulated with PEG 3350 NF and 100 μg of abaloparatide to abdomen of healthy post-menopausal women (N=12) where plasma concentration time points are the arithmetic means.

[0042] Figure 34: Concentration-time graph after administration of patch formulated with PEG 3350 NF and 150 μg of abaloparatide to abdomen of healthy post-menopausal women (N=13) where plasma concentration time points are the arithmetic means.

[0043] Figure 35: Concentration-time graph after administration of patch formulated with PEG 3350 NF and 200 μg of abaloparatide to abdomen of healthy post-menopausal women (N=14) where plasma concentration time points are the arithmetic means.

[0044] Figure 36: Concentration-time graph after administration of patch formulated with PEG 3350 NF and ZnCl 2 and 100 μg of abaloparatide to abdomen of healthy postmenopausal women (N=8) where plasma concentration time points are the arithmetic means.

[0045] Figure 37: Concentration-time graph after administration of patch formulated with PEG 3350 NF and ZnCl 2 and 150 μg of abaloparatide to abdomen of healthy postmenopausal women (N=7) where plasma concentration time points are the arithmetic means.

[0046] Figure 38: Concentration-time graph after administration of patch formulated with PEG 3350 NF and ZnCl 2 and 200 μg of abaloparatide to abdomen of healthy postmenopausal women (N=8) where plasma concentration time points are the arithmetic means.

[0047] Figure 39: Transdermal administration utilized patches having different microneedle lengths with abaloparatide formulations (no Zn) in monkeys. Square:

subcutaneous delivery; triangle: transdermal delivery (short microneedles - 250 μι η );

diamond: transdermal delivery (regular microneedles - 500 μπι); and star: transdermal delivery (long microneedles - 700 μηη).

[0048] Figure 40: Comparison of Cmax (peak plasma concentration (pg/mL)) of various formulations of the PTHrP analogue of SEQ ID NO: l in monkeys.

[0049] Figure 41 : Comparison of AUC (area under the curve) of various formulations of the PTHrP analogue of SEQ ID NO: 1 administered by transdermal versus subcutaneous route.

[0050] Figure 42: Comparison of plasma concentration (pg/mL) of formulation of abaloparatide (ABL, SEQ ID NO: l) administered by subcutaneous route (SC) or transdermal administration (TD), wherein the transdermal administration utilized a transdermal patch coated using different transdermal formulations in monkeys.

[0051]

[0052] Table 1. Modeling of TD-A32 data for bioequivalence for the abaloparatide-SC treatment.

[0053] Table 2. PK Results of Abaloparatide 100 pg TD, Abaloparatide 150 pg TD, Abaloparatide 200 μg TD, Abaloparatide 80 pg SC, and Historical 150 pg TD.

[0054] Table 3. Comparisons of Abaloparatide 100 pg TD, Abaloparatide 150 pg TD, Abaloparatide 200 pg TD, Abaloparatide 80 pg SC, and Historical 150 pg TD to UnoPen 80 pg SC, respectively.

[0055] Table 4. Comparisons of Abaloparatide 100 pg TD, Abaloparatide 150 pg TD, Abaloparatide 200 pg TD, and Abaloparatide 80 pg SC to Historical 150 pg TD, respectively.

[0056] Table 5. Design for a Phase 2 study of transdermal delivery of abaloparatide using a transdermal patch prepared by a first general abaloparatide formulation (with PBS).

[0057] Table 6. C max , AUC, and BMD improvement of a Phase 2 study of transdermal delivery of abaloparatide (TD-50 meg, TD-100 meg, and TD-150 meg) using a transdermal patch prepared by a first general abaloparatide formulation (with PBS), and subcutaneous delivery of abaloparatide (SC-80 meg).

DETAILED DESCRIPTION

[0058] Abaloparatide is a synthetic PTHrP analogue having the sequence set forth in SEQ ID NO: 1. Abaloparatide has shown potent anabolic activity with decreased bone resorption, less calcium-mobilizing potential, and improved room temperature stability (Obaidi 2010). Studies performed in animals have demonstrated marked bone anabolic activity following administration of abaloparatide, with complete reversal of bone loss in ovariectomy-induced osteopenic rats and monkeys (Doyle 2013a; Doyle 2013b; Hattersley 2013). Abaloparatide has been developed as a promising anabolic agent for the treatment of osteopenia (e.g., glucocorticoid-induced osteopenia), osteoporosis (e.g. glucocorticoid-induced osteoporosis), and/or osteoarthritis.

[0059] Subcutaneous administration of 80 pg abaloparatide (hereinafter the "abaloparatide- SC treatment") has been shown to significantly reduce incidences of new vertebral, non- vertebral, major osteoporotic and clinical fractures versus a placebo. Subcutaneous abaloparatide administration has also been shown to improve bone mineral density (BMD) and/or trabecular bone score (TBS) of treated subjects at the lumbar spine, total hip, and femoral neck. In certain embodiments, the abaloparatide-SC treatment comprises subcutaneous administration of an aqueous formulation comprising abaloparatide (about 2 mg/mL) in an acetate buffer, with a pH of about 4.5 to about 5.6, or about 5.1. Optionally, the aqueous formulation further comprises phenol (about 5 mg/mL). In certain examples of these embodiments, the acetate buffer comprises tri-hydrate sodium acetate (about 5 mg/mL) with pH (e.g., about 4.5 to about 5.6, or about 5.1) adjusted with acetic acid.

[0060] Transdermal administration of abaloparatide is an attractive alternative to subcutaneous administration due to its less invasive nature. However, transdermal administration may have different PK profile compared to subcutaneous administration. It has been found that AUC of transdermal abaloparatide administration and subcutaneous abaloparatide administration (80 μg) had a linear relationship with the achieved BMD changes from the baseline after 6 months of treatments (Figure 30B). It is thus desired to develop transdermal abaloparatide administrations that are substantially bioequivalent to the subcutaneous abaloparatide administration to benefit from both the preferred osteoanabolic profile of subcutaneous abaloparatide administration and the convenience of transdermal administration.

[0061] As disclosed herein, it has been unexpectedly found that transdermal abaloparatide administration using a patch prepared with a preparation formulation comprising

abaloparatide and one or more excipients selected from the group consisting of ZnCl , PEG, and histidine produces a substantial bioequivalence to subcutaneous administration in a monkey model. Furthermore, preliminary clinical studies indicate that ZnCl 2 in a transdermal formulation blunts the pulsatile nature compared with a non-ZnCI 2 containing formulation and pushes the curve into one resembling the sc curve. This is a notable achievement in that achieving bioequivalence to a sc dose of abaloparatide would indicate exceptional fracture prevention effects as has been reported for 80 μ sc administration.

[0062] Based on these findings, provided herein are PTHrP analogue preparation formulations, transdermal patches prepared using these preparation formulations, transdermal patches comprising these preparation formulations, methods of making these patches, and methods of using the disclosed preparation formulations and patches to administer PTHrP analogues in a transdermal manner and to treat osteoporosis, osteopenia, and osteoarthritis, improve BMD, improve TBS, and treat, prevent, and reduce bone fractures in a subject. In certain embodiments of the preparation formulations, transdermal patches, and methods provided herein, the PTHrP analogue is abaloparatide consisting of the amino acid sequence set forth in SEQ ID NO: 1 , or an abaloparatide derivative comprising or consisting essentially of the amino acid sequence set forth in SEQ ID NO: 1. In certain embodiments of the preparation formulations, transdermal patches, and methods provided herein, the transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalcncc to a subcutaneous delivery of abaloparatide at the dosage of about 20 μg to about 200 μg, about 40 μg to about 120 g, about 60 μg to about 100 g, about 70 μg to about 90 μg, or about 80 μg. In certain embodiments of the preparation formulations, transdermal patches, and methods provided herein, the transdermal delivery of the PTHrP is a substantial

bioequivalence or bioequivalence of the abaloparatide-SC treatment.

[0063] As used herein, two treatments of an active agent are bioequivalent to one another if the 90% confidence interval of the ratio of area under the curve (AUC) and/or the peak serum concentration of the active agent (C max ) falls completely within the range 80-125%. See, e.g., Figure 1 A showing a bioequivalence window of the abaloparatide-SC treatment in Chinese Cynomolgus monkeys. Serum abaloparatide concentrations are presented as percentage of

[0064] As used herein, the term "substantially," "substantial" or "essentially" means nearly completely or completely. In particular, a normal bioequivalent range means a compound in a particular formulation for transdermal delivery within the 80%- 125% (for the mean in the 90% confidence interval (CI)) of AUC (o. t , o-inf) and C ma x of the reference compound in a reference formulation. In some embodiments the reference formulation is the SC delivery of 80 μg abaloparatide formulated as described herein. In certain embodiments, the transdermal delivery of a compound or more particularly abaloparatide falls within a substantially bioequivalent range wherein said range is 70%-136%, or 65%-141 %, or 60%-147%, or 50%- 158% (for the mean in the 90% confidence interval (CI)) of AUC ( 0 . t , o-inf) and Cm ax of the reference compound in a reference formulation.

[0065] As used herein, the term "about" or "approximately" means a range of ±0-10%, or ±0-5% or the numeral following the term.

[0066] As used herein, the term "transdermal delivery" refers to a delivery of an active agent through the stratum corneum to make contact with the intradermal space without significant pain upon penetration. Because the stratum corneum has no nerves, it may be pierced without stimulating nerves. The terms "transdermal" and "intradermal" are used interchangeably herein. The stratum corneum is composed primarily of several layers of dead skin cells and is not vascularized. Thus, the stratum corneum often poses a formidable barrier to the transdermal delivery of an active agent, especially for charged macromolecules such as peptides. Unlike active agents delivered by subcutaneous injection, which almost provides a complete entrance into the blood stream, many factors (and barriers) can affect the pharmacokinetics of drugs delivered by a transdermal route. For example, the site of application, the thickness, integrity, and hydration condition of the skin, the thickness and density of the adipose tissue under the skin of the application site, the size of the drug molecules, the pH condition and permeability of the membrane of the transdermal device, etc., all may affect the bioavailability of drugs delivered transdermally. In certain embodiments, transdermal delivery involves penetrating the skin through the stratum corneum into the dermis to a depth of up to about 700 μιη, or up to about 600 μ η ι, or up to about 500 μτη, or up to about 400 μπι, or up to about 300 μηι, or up to about 250 μηι, or up to about 150 μηι. In some embodiments, the average needle depth of penetration is approximately 800 μηι, or about 700 μηι, or about 600 μηι, or about 500 μηι, or about 400 μπι, or about 300 μπι, or about 250 μπι, or about 150 μιη.

/. Preparation formulation for transdermal delivery

[0067] Provided herein in certain embodiments are preparation formulations for transdermal delivery of a therapeutically active substance, e.g., a bioactive peptide, a bioactive peptide contains at least 10 amino acids, e.g., a PTHrP analogue (e.g., comprising, consisting essentially of, or consisting of abaloparatide). In certain embodiments, the transdermal delivery produces a substantial bioequivalence or bioequivalence to

subcutaneous delivery of the PTHrP analogue (e.g., at 80 μg). These formulations comprise a PTHrP analogue and one or more excipients selected from the group consisting of salts of Zn 2+ , salts of Mg 2+ , salts of Ca 2+ , salts of histidine, salts of carboxy lie acids (e.g., fatty acids), NaCl, PEG, PVP, cyclodextrin (CD, e.g., 2-hydroxypropyl-p-cyclodextrin (HPpCD)), and combinations thereof. In certain embodiments the salt of Zn 2+ is selected from the group consisting of Zn(OAc) 2 , ZnCl 2 , Zn 3 (P0 4 ) 2 , zinc citrate (ZnCitrate), zinc oxalate (ZnOxalate), and combinations thereof, the salt of Ca 2+ is selected from the group consisting of calcium sorbate (CaSorbate), calcium citrate (CaCitrate), calcium ascorbate (CaAscorbate),

Ca 3 (P0 4 ) 2l CaCl 2 , CaC0 3 , CaS0 4 , Ca(OAc) 2 and combinations thereof, the salt of Mg 2+ is selected from the group consisting of MgO, magnesium citrate (MgCitrate), MgS0 4j magnesium orotate (MgOrotate), magnesium lactate (MgLactate), MgC0 3 , MgCl 2 ,

Mg(OAc) 2 , and combinations thereof. In certain embodiments, two or more salts of Mg + , Zn 2+ and/or Ca 2+ as described herein are combined together for purposes of a transdermal formulation. In certain embodiments, the preparation formulation further comprises water for injection, brine or PBS. In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide. In certain embodiments, the transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to a subcutaneous delivery of abaloparatide at the dosage of about 20 μg to about 200 μg, about 40 g to about 120 μg, about 60 μg to about 100 μ¾ about 70 μg to about 90 μ^ or about 80 μg. In certain embodiments, the transdermal delivery of the PTHrP is a substantial bioequivalence or bioequivalence of the abaloparatide-SC treatment. In certain embodiments, the PTHrP analogue is delivered by a transdermal patch comprising at least one microprojection (e.g., microneedle) prepared using the preparation formulation.

[0068] In certain embodiments, the preparation formulation comprises PEG with a molecular weight of about 3,000 to about 3,700, about 2,000 to about 5,000, about 3,00 to about 3,500, or about 1 ,000 to about 6,000. A concentration by weight of PEG to the total amount of the preparation formulation is about 0.01 % to about 50%, about 5% to about 50%, about 5% to about 45%, about 5% to about 40%, about 5% to about 35%, about 5% to about 30%, about 5% to about 25%, about 5% to about 20%, about 5% to about 15%, about 10% to about 50%, about 10% to about 45%, about 10% to about 40%, about 10% to about 35%, about 10% to about 30%, about 10% to about 25%, about 10% to about 20%, about 10% to about 15%, about 15% to about 50%, about 1 % to about 45%, about 15% to about 40%, about 15% to about 35%, about 15% to about 30%, about 15% to about 25%, about 15% to about 20%, about 13% to about 17%, about 14% to about 16%, or about 14.9%.

[0069] In certain embodiments, the preparation formulation comprises water and a Zn salt (also referred to as Zn salt, salt of Zn, or salt of Zn 2+ ), in some embodiments said coating formulation comprises ZnC^, or Zn(OAc)2, or Zn 3 (P04)2, or ZnCitrate or ZnOxalate or combinations thereof. The concentration of Zn 2+ salt (e.g., ZnCh) in the preparation formulation, for example, by weight to the total amount of the preparation formulation is about 0.01 % to about 30%, 0. 1 % to about 30%, 0.3% to about 30%, about 0.5% to about 30%, about 0.8% to about 30%, about 1% to about 30%, about 1.5% to about 30%, about 2% to about 30%, about 5% to about 30%, 10% to about 30%, 15% to about 30%, about 20% to about 30%, about 25% to about 30%, about 0.01% to about 20%, 0. 1% to about 20%, 0.3% to about 20%, about 0.5% to about 20%, about 0.8% to about 20%, about 1 % to about 20%, about 1.5% to about 20%, about 2% to about 20%, about 5% to about 20%, 10% to about 20%, 15% to about 20%, about 0.01% to about 10%, 0. 1% to about 10%, 0.3% to about 10%, about 0.5% to about 10%, about 0.8% to about 10%, about 1% to about 10%, about 1.5% to about 10%, about 2% to about 10%, about 5% to about 10%, about 0.01% to about 5%, 0. 1 % to about 5%, 0.3% to about 5%, about 0.5% to about 5%, about 0.8% to about 5%, about 1% to about 5%, about 1.5% to about 5%, about 2% to about 5%, about 0.01 % to about 3%, 0. 1% to about 3%, 0.3% to about 3%, about 0.5% to about 3%, about 0.8% to about 3%, about 1% to about 3%, about 1.5% to about 3%, about 2% to about 3%, about 0.01% to about 30%, 0. 1 % to about 30%, 0.3% to about 30%, about 0.5% to about 30%, about 0.8% to about 2%, about 1 % to about 2%, about 1.5% to about 2%, about 0.01% to about 1 %, 0. 1% to about 1%, 0.3% to about 1%, about 0.5% to about 1%, about 0.8% to about 1%, or about 0.8%. In certain embodiments, the coating formulation for the described ranges comprises a Ca 2+ salt wherein said Ca 2+ salt can include CaSorbate, CaCitrate, CaAscorbate, Ca3(P0 4 )2, CaCl 2 , CaC0 3 , CaSC>4, Ca(OAc) 2 or combinations thereof. Ca 2+ salt is also referred to as Ca salt, salt of Ca, or salt of Ca 2+ . In some embodiments the coating solution comprises a Mg 2+ salt wherein said Mg + salt can include MgO, gCitrate, MgS04, MgOrotate, MgLactate, MgCC , MgCl 2 , Mg(OAc) 2 , or combinations thereof. Mg 2+ salt is also referred to as Mg salt, salt of Mg, or salt of Mg + .

[0070] In some embodiments of this invention, a formulated patch ready to package and use (the initial coating solution dried to remove water) is provided wherein said formulated patch, comprises a Zn 2+ salt. In certain embodiments the Zn + salt is Zn(OAc) 2 , in some embodiments the Zn 2+ salt is ZnCl 2 , in certain embodiments it is Zn 3 (P0 4 ) 2) in some embodiments it is ZnCitrate and in certain embodiments it is ZnOxalate or combinations thereof. In certain embodiments, the formulated patch is made by coating with the coating solution in one or multiple coating iterations and then drying said patch or allowing said patch to dry to a fairly constant weight and then characterizing said patch as weight by percent of the salt, the metal including its counterions. In certain embodiments, the coated patch, dried and ready to use comprises from 1.0 to 20% Zn salt by weight. In certain embodiments, said coated patch comprises from 1.5% to 15% Zn salt by weight. In some embodiments, the coated and dried patch comprises 1.5% to 10% Zn salt by weight, or 1.8% - 8.5%, or 1.9% to 5.9%, or about 1.9% to 8.5%, or about 2.0% to about 8%, or 5% to 8% by weight or is between 1.7% to 2.25%, or between 5 to 7%, or about 5.8%, or about 1.9%.

[0071] In some embodiments of this invention, a formulated patch ready to package and use (the initial coating solution dried to remove water) is provided wherein said formulated patch comprises a Ca 2+ salt. In some embodiments the Ca 2+ salt is CaSorbate, CaCitrate, CaAscorbate, Ca 3 (P0 4 )2, CaCl 2 , CaC0 , CaS0 4 , Ca(OAc) 2 or combinations thereof. In certain embodiments, the formulated patch is made by coating with the coating solution in one or multiple coating iterations and then drying said patch or allowing said patch to dry to a fairly constant weight and then characterizing said patch as weight by percent of the salt, the metal including its counterions. In certain embodiments, the coated patch, dried and ready to use comprises from 1.0 to 20% Ca salt by weight. In certain embodiments, said coated patch comprises from 1.5% to 15% Ca salt by weight. In some embodiments, the coated and dried patch comprises 1.5% to 10% Ca salt by weight, or 1.8% - 8.5%, or 1.9% to 5.9%, or about 1.9% to 8.5%, or about 2.0% to about 8%, or 5% to 8% by weight.

[0072] In some embodiments of this invention, a formulated patch ready to package and use (the initial coating solution dried to remove water) is provided wherein said formulated patch comprises a Mg 2+ salt. In some embodiments the Mg 2+ salt is MgO, MgCitrate, MgS0 4 , MgOrotate, MgLactate, MgC0 3 , MgCl 2 , Mg(OAc) 2 , or combinations thereof. In certain embodiments, the formulated patch is made by coating with the coating solution in one or multiple coating iterations and then drying said patch or allowing said patch to dry to a fairly constant weight and then characterizing said patch as weight by percent of the salt, the metal including its counterions. In certain embodiments, the coated patch, dried and ready to use comprises from 0.5 to 15% Mg salt by weight. In certain embodiments, said coated patch comprises from 1.0% to 10% Mg salt by weight. In some embodiments, the coated and dried patch comprises 1.5% to 10% Mg salt by weight, or 1.8% - 8.5%, or 1.9% to 5.9%, or about 1.9% to 8.5%, or about 2.0% to about 8%, or 5% to 8% by weight.

[0073] In certain embodiments, the formulated patch comprises two or more of the Zn 2+ , Ca 2+ and/or Mg 2+ salts described immediately above.

[0074] Example of coating solution

[0075] Example of formulation on patch ready to use (after drying)

[0076] Example of coating solution

[0081] Example of formulation on patch ready to use (after drying)

[0086] Example of formulation on patch ready to use (after drying)

[0091] Example of coating formulation for PEG, PVP, CD and histidine

[0092] Example of formulation on patch ready to use (after

[0093] Coating solution, doses and sites of administration for clinical and planned clinical studies

3 ZnCI 2 /PEG 100 Abdomen

150

200

4 Period 1 ZnCl 2 200 Thigh

4 Period 2 ZnCl 2 /PEG 200 Thigh

4 Period 3 ZnC 2 X 150 Abdomen

5 Period 1 ZnCl 2 200 Thigh

5 Period 2 Zn(OAc) 2 200 Thigh

5 Period 3 ZnCI 2 200 Thigh

[0094] In certain embodiments, the preparation formulation comprises histidine (e.g., monohydrochloride monohydrate). The concentration of histidine (by weight to the total amount of the preparation formulation) is about 1 % to about 15%, about 1 % to about 10%, about 1 % to about 5%, about 3% to about 15%, about 3% to about 10%, about 3% to about 5%, about 5% to about 1 %, about 5% to about 10%, about 3%, about 5%, or about 10%.

[0095] In certain embodiments, the preparation formulation comprises two or three excipients selected from the group consisting of PEG, ZnCl 2 , and histidine, wherein the concentration of each excipient is the same as disclosed herein.

[0096] In certain embodiments, the preparation formulation comprises two excipients selected from the group consisting of PEG, ZnC , and histidine, e.g., a combination of PEG and ZnCl 2 , a combination of histidine and PEG, and a combination of histidine and ZnCb.

[0097] In certain embodiments, the preparation formulation comprises a combination of PEG, ZnCl 2 , and histidine.

[0098] In certain embodiments, the preparation formulation comprises the PTHrP analogue at a concentration of about 5% to about 15%, about 12.5% to about 20%, about 15% to about 60%, about 40% to about 48%, about 43% to about 48%, about 40% to about 46%, about 40% to about 52%, about 46% to about 48%, about 46% to about 52%, about 50% to about 62%, about 52% to about 60%, or about 54% to about 58% by weight.

[0099] In certain embodiments, the preparation formulation has a viscosity at 25°C of greater than about 500 centipoise, greater than about 550 centipoise, greater than about 600 centipoise, greater than about 700 centipoise, greater than about 800 centipoise, greater than about 900 centipoise, greater than about 1,000 centipoise, greater than about 1,500 centipoise, greater than about 2,000 centipoise, greater than about 10,000 centipoise, about 5 . 00 to about 5,000 centipoise, about 500 to about 2,000 centipoise, or about 500 to about 1 ,000 centipoise, about 550 to about 5,000 centipoise, about 550 to about 2,000 centipoise, or about 550 to about 1 ,000 centipoise. [00100] In certain embodiments, the preparation formulations disclosed herein further comprise a bioactive peptide or protein. In certain embodiments, the preparation formulations disclosed herein comprise an antibody.

[00101] In certain embodiments, the preparation formulations disclosed herein further comprise excipients selected from the group consisting of ZnCl 2 , Zn(OAc) 2 , Zn 3 (P0 4 )2, ZnCitrate, ZnOxalate, MgO, MgCitrate, MgS0 4 , MgOrotate, MgLactate, MgC0 3 CaSorbate, CaCitrate, CaAscorbate, Ca 3 (P0 4 )2, CaCl 2 , CaC0 3 , CaS0 4 , and Ca(OAc) 2 . In certain embodiments, the preparation formulations disclosed herein comprise excipients selected from ZnCl 2 , Zn(OAc) 2 and combinations thereof. In certain embodiments, the preparation formulations disclosed herein have a molar ratio of the excipient or excipients to the therapeutically active substance selected from the ranges of about 0.1 to about 2.0, about 0.2 to about 1 .5, or about 0.25 to about 1.0.

II. Transdermal patches

[00102] Provided herein in certain embodiments are transdermal patches for administration of a PTHrP analogue comprising one or more microprojections prepared using a preparation formulation of the PTHrP analogue as disclosed herein, wherein transdermal delivery of the PTHrP analogue using the patch produces substantial bioequivalence or bioequivalence to subcutaneous delivery of the PTHrP. In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide. In certain embodiments, the transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to a subcutaneous delivery of abaloparatide at the dosage of about 20 g to about 250 pg, about 20 μg to about 200 μg, about 40 μg to about 120 μg, about 60 μg to about 100 about 70 μg to about 90 μ& about 80 μg, about 100 μg, about 150 μ& or about 200 μ& In certain embodiments, transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to the abaloparatide-SC treatment.

[00103] In certain embodiments, the transdermal patches provided herein are designed for passive diffusion of the PTHrP analogue as provided herein. In other embodiments, the transdermal patches are designed for active delivery of the PTHrP analogue using an external energy source.

[00104] In certain embodiments, the preparation formulation of the PTHrP analogue is used to prepare one or more microprojections on a transdermal patch, resulting in the transdermal patches comprising the PTHrP analogue. For example, at least part of the one or more microprojections on the transdermal patch comprises the PTHrP analogue. In certain embodiments, at least part of the one or more microprojections on the transdermal patch further comprises one or more excipients selected from the group consisting of PEG, ZnCl 2 and histidine.

[00105] In certain embodiments, the amount of each excipient per patch is about 1 μg to about 300 μ about 10 g to about 300 μ& about 100 μg to about 300 μ about 200 μg to about 300 about 1 μ to about 200 μg, about 10 to about 200 μg, about 100 μg to about 200 μg, about 150 μg to about 200 μg J about 1 μg to about 150 μ about 10 μg to about 150 μg, about 100 μg to about 150 μ¾ about 1 μg to about 100 μg, about 10 μg to about 100 & about 50 μg to about 100 μ& about 1 μg to about 50 μg, about 10 μg to about 50 μg, about 20 μg to about 50 μg, about 1 μg to about 20 μ about 10 μ to about 20 μ about 15 g to about 20 μg, about 1 μg, about 5 μg, about 10 μ about 20 μζ, about 30 μg, about 40 μ§, about 50 μg, about 60 μg, about 70 μ about 80 μ^ about 90 μg, about 100 μg, about 1 10 μg, about 120 μ about 130 μg, about 140 μg, about 150 μ about 160 μg, about 170 μ¾ about 180 μ¾ about 190 μ¾ about 200 μg ί about 210 μg, about 220 μ¾ about 230 μg, about 240 μg, about 250 μg, about 260 §, about 270 μg, about 280 μg, about 290 g, or about 300 μg.

[00106] In certain embodiments, the amount of the PTHrP analogue (e.g., abaloparatide) per patch is 1 μg to about 300 μg, about 10 μg to about 300 μg, about 100 μg to about 300 μ about 200 μg to about 300 μg, about 1 μg to about 200 μg, about 10 μg to about 200 μg, about 50 μg to about 200 μ about 80 μg to about 200 μg, about 100 μg to about 200 μg, about 150 μg to about 200 μg, about 1 μg to about 150 μg, about 10 μ ίο about 150 μ¾ about 50 μg to about 150 μg, about 80 μg to about 150 μg > about 100 μg to about 150 μ about 1 μg to about 100 μg, about 10 μg to about 100 μ^ about 50 μg to about 100 μ about 1 μg to about 50 μg, about 10 μg to about 50 μg, about 20 μg to about 50 μg, about 1 μg to about 20 μg, about 10 μg to about 20 μg, about 15 μg to about 20 μg, about 1 μ about 5 μ about 10 μ^ about 20 μg, about 30 μg, about 40 μ about 50 μ about 60 μ^ about 70 μ about 80 μ^ about 90 g, about 100 μ about 1 10 μg, about 120 μg, about 130 μ^ about 140 μ& about 150 μg, about 160 μg, about 170 about 180 ^ about 190 μg, about 200 μg, about 210 μg, about 220 μ about 230 μg, about 240 & about 250 μg, about 260 μg, about 270 μ¾ about 280 μg, about 290 μg, or about 300 μg.

[00107] In certain embodiments, the transdermal patches provided herein comprise a plurality of these microprojections. The term "microprojection" as used herein refers to a piercing element of any shape or size on a transdermal patch that is capable of piercing the stratum corneum of the skin. These small piercing elements can have various materials, shapes and dimensions. In certain embodiments, one or more of the microprojections on the disclosed transdermal patches are microneedles. The term "microneedle" as used herein refers to a microprojection comprising a base and a tip, wherein the tip has a smaller diameter, width, perimeter or circumference than the base. A transdermal patch comprising one or more microneedles may also be referred to as a "transdermal microneedle patch" or a "microneedle transdermal patch."

[00108] A microneedle in the transdermal patches provided herein may have any size, shape, or design commonly used in the art. In certain embodiments, the microneedles have their greatest diameter, width, perimeter, or circumference at the base. In certain embodiment, the microneedles have a tapered design, meaning that the microneedle from base to tip reflects a relatively constant narrowing over the length. In certain embodiments, the ratio of the diameter, width, perimeter, or circumference at the base of the microneedle to the diameter, width, perimeter, or circumference at the tip of the microneedle is greater than 2. In other embodiments, the ratio is greater than 4 or greater than 6. In certain embodiments, the microneedles have a generally circular perimeter about the axis that is broader at the base than the tip. In certain embodiments, the microneedles are pyramidal in shape, with an approximately rectangular base that tapers to an apex, wherein said apex is approximately rectangular. In certain embodiments, the microneedles are pyramidal in shape, with a square base that tapers to an apex wherein said apex is approximately square. In certain

embodiments, the microneedles are pyramidal in shape with a rectangular or square base and a shape that is not readily characterized as rectangular or square at the top.

[00109] The microprojection may have various length, e.g., about 30 μι η to about 1 ,500 μ η ι, about 50 μπι to about 1 ,500 μιη, about 100 μιη to about 1 ,500 μιη, about 250 μπι to about 1 ,500 μπι, about 500 μπι to about 1,500 μιη, about 600 μηι to about 1 ,500 μιη, about 750 μπι to about 1 ,500 μπι, about 800 μηι to about 1,500 μιη, about 1,000 μπι to about 1 ,500 μιυ, about 30 μιη to about 1 ,00 μιη, about 50 μπι to about 1 ,500 μπι, about 30 μπι to about 1,000 μηι, about 50 μιη to about 1 ,000 μπι, about 750 μπι to about 1 ,200 μηι, about 800 μηι to about 1,200 μπι, about 100 μηι to about 1 ,000 μπι, about 250 μηι to about 1 ,000 μπι, about 500 μπι to about 1 ,000 μπι, about 600 μπι to about 1,000 μιη, about 750 μπι to about 1 ,000 μπι, about 800 μηι to about 1,000 μπι, about 30 μπι to about 750 μπι, about 50 μιη to about 750 μηι, about 100 μηι to about 750 μηι, about 250 μπι to about 750μιη, about 500 μπι to about 750 μηι, about 600 μπι to about 750 μπι, about 600 μηι to about 800 μιτι, about 30 μ η ι to about 600 μπι, about 50 μηι to about 600 μπι, about 100 μπι to about 600 μι η , about 250 μπι to about 600 μπι, about 500 μιη to about 600 μηι, about 30 μπι to about 500 μιη, about 50 μηι to about 500 μιη, about 100 μηι to about 500 μηι, about 250 μηι to about 500 μπι, about 30 μιη to about 250 μιη, about 50 μηι to about 250 μιη, about 100 μιη to about 250 μηι, about 30 μηι, about 50 μιη, about 100 μπι, about 150 μπι, about 200 μηι, about 250 μ η ι, about 300 μηι, about 500 μηι, about 750 μηι, or about 1,500 μιη.

[00110] Microprojections on the transdermal patches provided herein can be made from any suitable material, including for example carbon, polymers, metals, or a combination thereof, to achieve a desirable flexural modulus. In some embodiments, the microprojection has a flexural modulus of greater than 1,000 MPa, greater than 2,000 MPa, greater than 3,000 MPa, or between 3,000 MPa and 15,000 MPa. As used herein, "ISO 178" refers to ISO test standards for determination of flexural properties of plastics.

[00111] In certain embodiments, the transdermal patches provided herein comprise a first backing layer on which the microprojections are arrayed. In these embodiments, the microprojections may be affixed to or integral with the first backing layer, In certain embodiments, the microprojections are made from the same material as the first backing layer. For example, the microprojections may be formed by etching or punching from the first backing layer. In certain embodiments, the microprojections are made by an injection molding process. In other embodiments, the microprojections may be made of a different material than the first backing layer. In certain of these embodiments, the microprojections are affixed to the first backing layer via an adhesive. In certain of these embodiments, the microprojections are detachable from the first backing layer and/or the second backing layer.

[00112] In certain embodiments, the transdermal patches provided herein further comprise a second backing layer on which the first backing layer is affixed. The second backing layer may be flexible or inflexible.

[00113] In certain embodiments, the transdermal patches provided herein comprise an adhesive material to facilitate the patch staying in place on a subject's skin before and/or during transdermal administration of the PTHrP analogue. In certain of these embodiments, the adhesive material is comprised on the first and/or second backing layer(s).

[00114] In certain embodiments of the transdermal patches provided herein, the vertical axis of the one or more microprojections extends at an angle of at least 45 degrees or at least 60 degrees from the first and/or second backing layer(s). In some embodiments, the microprojections are perpendicular to the first and/or second backing layer(s).

[00115] In certain embodiments of the transdermal patches provided herein, the patches have a microprojection density of about 20 to about 2,000 microprojections per cm 2 , about 50 to about 2,000 microprojections per cm 2 , about 100 to about 2,000 microprojections per cm 2 , about 250 to about 2,000 microprojections per cm 2 , about 500 to about 2,000 microprojections per cm 2 , about 750 to about 2,000 microprojections per cm 2 , about 1,000 to about 2,000 microprojections per cm 2 , about 1 ,500 to about 2,000 microprojections per cm 2 , about 300 to about 500 microprojections per cm . In certain embodiments, the patches comprise about 50 to about 4,000 microprojections, about 100 to about 4,000

microprojections, about 250 to about 4,000 microprojections, the patches comprise about 1 ,400 to about 4,000 microprojections, about 1,600 to about 4,000 microprojections, about 2,000 to about 4,000 microprojections, about 3,000 to about 4,000 microprojections, about 3,500 to about 4,000 microprojections, the patches comprise about 50 to about 3,500 microprojections, about 100 to about 3,500 microprojections, about 250 to about 3,500 microprojections, about 1,400 to about 3,500 microprojections, about 1,600 to about 3,500 microprojections, about 2,000 to about 3,500 microprojections, about 3,000 to about 3,500 microprojections, about 50 to about 3,000 microprojections, about 100 to about 3,000 microprojections, about 250 to about 3,000 microprojections, about 1 ,400 to about 3,000 microprojections, about 1,600 to about 3,000 microprojections, about 2,000 to about 3,000 microprojections, about 50 to about 600 microprojections, about 100 to about 500 microprojections, about 250 to about 400 microprojections, about 300 to about 375 microprojections, about 300 to about 750 microprojections, about 366 microprojections, about 316 microprojections, or about 320 microprojections.

[00116] In certain embodiments, the transdermal patch of a PTHrP analogue comprises at least one microprojection at least partially coated with a of the PTHrP analogue (hereinafter the "coated microprojection").

[00117] The term "coated" as used herein with regard to an individual microprojection means that the microprojection comprises a PTHrP analogue composition on at least part of its surface. In certain embodiments, the microprojection comprises a PTHrP analogue composition on about 1 % to about 100%, 1% to about 80%, about 1% to about 50%, about 2% to about 40%, about 5% to about 35%, 10% to about 30%, 15% to about 20%, or about 30% to about 50% of its total surface area. In certain embodiments, the microprojection comprises a PTHrP analogue composition on about 30% to about 50% of the top of the microprojection (as used herein, "top" means the end of the microprojection which would contact the skin).

[00118] The term "coated" as used herein with regard to a plurality of microprojections means that two or more of the microprojections in the plurality are coated as the term is used above with regard to an individual microprojection. In certain embodiments, more than 10%, more than 20%, more than 30%, more than 40%, more than 50%, more than 60%, more than 70%, more than 80%, or more than 90% of the microprojections in a plurality of microinjections are coated. In certain embodiments, about 1 % to about 100%, 1 % to about 80%, about 1% to about 50%, about 2% to about 40%, about 5% to about 35%, 10% to about 30%, 15% to about 20%, or about 30% to about 50% of the total microprojection surface area in the plurality of microinjections are coated. "Microprojection surface area" as used herein refers to the combined surface area of all microprojections on a single transdermal patch.

[00119] In certain embodiments, a transdermal patch comprising one or more coated microprojections may further comprise a PTHrP analogue composition on at least part of its surface of the first backing layer. For example, the transdermal patch comprises a PTHrP analogue composition on more than about 1% to about 100%, 1% to about 80%, about 1% to about 50%, about 2% to about 40%, about 5% to about 35%, 10% to about 30%, 15% to about 20%, or about 30% to about 50% of its total surface area of the first backing layer.

[00120] In certain embodiments, the transdermal patch of a PTHrP analogue comprises at least one microprojection comprising a plurality of layers arranged roughly parallel (e.g. at least about 80% parallel, at least about 90% parallel, or at least about 95% parallel) to the first backing layer, and at least one layer of the plurality of layers comprises the PTHrP analogue (hereinafter the "active agent layer") (hereinafter the "layered microprojection").

[00121] In certain embodiments, the first backing layer of the transdermal patch comprising at least one layered microprojection further comprises an active agent layer. In certain embodiments, the active agent layer forms a tip of the microprojection which can penetrate through the stratum corneum for the transdermal delivery. The tip of the microprojection may adopt any shape as disclosed supra regarding the shapes of microprojections (e.g., pyramid, square, rectangle, etc.).

[00122] In other embodiments, the microprojection provided herein comprises a reservoir that is in fluid communication with the skin when applied to the subject. The reservoir is loaded with the PTHrP analogue to be administered. The reservoir may be an inner space of the microprojection in fluid communication with the skin when applied, e.g.,

microprojections comprising a hollow portion. In certain embodiments, the hollow portion may have a side-opening.

[00123] In certain embodiments, the transdermal patches disclosed herein comprise a plurality of microprojections wherein at least one microprojection (e.g., microneedles)in the array is covered at least in part by a coating, said coating comprising a therapeutically active substance and one or more excipients selected from the group consisting of Zn 2+ salts, Mg 2+ salts, Ca 2+ salts, polyethylene glycols and hydroxypropyl beta-cyclodextrins. In certain embodiments, the therapeutically active substance comprises a bioactive peptide or protein. In certain embodiments, the therapeutically active substance comprises an antibody. In certain embodiments, the transdermal patches disclosed herein comprise a bioactive peptide or protein, e.g., bioactive peptide containing at least 10 amino acids, such as abaloparatide as · set forth in SEQ ID NO: 1. In certain embodiments, the one or more excipients the transdermal patches disclosed herein has are selected from the group consisting of ZnCl 2 , Zn(OAc) 2 , Zn 3 (P0 4 ) 2 , ZnCitrate, ZnOxalate, MgO, MgCitrate, MgS0 , MgOrotate,

MgLactate, MgC0 3 CaSorbate, CaCitrate, CaAscorbate, Ca 3 (P0 4 ) 2; CaCl 2 , CaC0 3 , CaS0 4 , and Ca(OAc) 2 . In certain embodiments, the one or more excipients the transdermal patches disclosed herein has are selected from the group consisting of ZnCl 2 and Zn(OAc) 2 and combinations thereof. In certain embodiments, the transdermal patches disclosed herein have a molar ratio of the excipient or excipients to the therapeutically active substance selected from the range of about 0.1 to about 2.0, about 0.2 to about 1.5, or about 0.25 to about 1.0. In certain embodiments, the transdermal patches disclosed herein have abaloparatide in an amount of between 90-1 10 μg, 140-160 g, 185-220 μ¾ 225-275 μg or about 100 μg, about 150 μg, about 200 μg or about 250 μg.

[00124] Transdermal patches may also be prepared as disclosed in US Application Nos. 14/361,787, 14/361 ,802, 13/452,412, 13/791 ,170, 13/791,360, which are incorporated herein by reference in their entirety, including drawings.

III. Method of preparing transdermal patches

[00125] Provided herein in certain embodiments are methods of preparing a transdermal patch for administration of a PTHrP analogue as disclosed herein, comprising preparing at least one microprojection on a transdermal patch with a preparation formulation disclosed herein. In certain embodiments the microprojections are microneedles. In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide. In certain embodiments, the transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to a subcutaneous delivery of abaloparatide at the dosage of about 20 g to about 200 μg, about 40 μg to about 120 μg, about 60 μg to about 100 μ about 70 μg to about 90 μg, or about 80 μ^ In certain embodiments,, and transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to the abaloparatide-SC treatment.

[00126] In certain embodiments, the preparation methods provided herein comprise contacting one or more microprojections on a blank (i.e., previously free of the PTHrP analogue) transdermal patch with the preparation formulations provided herein. In certain of these embodiments, the microprojections are coated with the preparation formulation by dipping a blank transdermal patch into the preparation formulation, then removing the patch and allowing it to dry.

[00127] In certain of these embodiments, the microprojections are layered microprojections and are prepared by casting or depositing the layers onto the first and/or second backing layer, then removing the patch and allowing it to dry.

[00128] In certain embodiments, accelerated drying conditions are applied to the transdermal patch, including for example circulating air flow, desiccants, vacuum, and/or heat.

[00129] In certain embodiments, transdermal delivery of the PTHrP analogue by applying the transdermal patches to a subject produces substantial bioequivalence or bioequivalence to subcutaneous delivery of the PTHrP analogue.

IV. Method of treatments

[00130] Provided herein in certain embodiments are methods of treating osteoporosis, osteopenia, and osteoarthritis, improving bone mineral density (BMD), improving trabecular bone score (TBS), and/or treating, preventing, and/or reducing bone fractures in a subject comprising transdermally administering a therapeutically effective amount of a PTHrP analogue comprised in a preparation formulation provided herein. In certain embodiments, transdermal administration is accomplished using a transdermal patch as provided herein, wherein the patch comprises at least one microprojection prepared using a preparation formulation provided herein. The bone fractures being treated, prevented, or reduced and/or the bone with improved BMD and/or TBS may be vertebral or non-vertebral, clinical and major osteoporotic fractures. In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide. In certain embodiments, the transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to a subcutaneous delivery of abaloparatide at the dosage of about 20 μg to about 200 μg, about 40 μg to about 120 μ& about 60 μg to about 100 μ about 70 g to about 90 μ& or about 80 μg. In certain embodiments, transdermal delivery of the PTHrP analogue produces substantial bioequivalence or bioequivalence to the abaloparatide-SC treatment.

[00131] The term "subject" as used herein refers to a mammalian subject. Examples of suitable subjects include, without limitation, subjects with one or more conditions selected from the group consisting of osteopenia, glucocorticoid-induced osteopenia, osteoporosis, glucocorticoid-induced osteoporosis, osteoarthritis, bone fractures, and high cortical porosity (e.g., subjects with diabetes, especially type II diabetes), female mammals, male mammals, dogs, cats, humans, men, women, women with osteoporosis, postmenopausal women, postmenopausal women with osteoporosis, mammals with high cortical porosity, and men and women with high cortical porosity.

[00132] As used herein, the term "cortical porosity" means the fraction of the cortical bone volume that is not occupied by the bone. The cortical porosity may be measured by Digital X-ray radiogrammetry (DXR) or other methods to provide an estimation of the local intensity minima ("holes") in the cortical bone regions using a recursive (climbing) algorithm starting from the outer region (Dhainaut 2013). A combined porosity measure is derived from the area percentage of holes found in the cortical part relative to the entire cortical area, by averaging over the involved bones and scaled to reflect a volumetric ratio rather than the projected area. A "high cortical porosity" means a porosity of about 10% higher, about 15% higher, about 20% higher, about 50% higher, about 100% higher, or about 150% higher than that of healthy subjects from the same age group as controls. For example, the subject may have a cortical porosity of about 0.01256, which the control group has a cortical porosity of about 0.01093 (Dhainaut 2013). Subjects with type II diabetes may have a cortical porosity up to twice that of controls (Oei 2013). Subject may have normal BMD or slightly lower BMD while have high cortical porosity.

[00133] The term "therapeutically effective amount" as used herein refers to an amount of a PTHrP formulation as provided herein that is sufficient to elicit the required or desired therapeutic and/or prophylactic response, as the particular treatment context may require. Examples of therapeutically effective amounts of a PTHrP analogue include, without limitation, 20 μ¾ 40 μ¾ 60 & 80 μg, 100 μ¾ 120 μg ί 140 μ¾ 160 μ & 180 μ & 200 μ β , 220 μg, 240 μg, 260 μg, 280 μg, or 300 μg. Other examples of therapeutically effective amounts of a PTHrP analogue may also include, without limitation, between 1 g kg and 50 μg kg, 5 μg/kg and 50 μg kg, 1 μg/kg and 40 μg kg ί 1 μg/kg and 30 μg/kg, 1 μg/kg and 20 μg kg, 1 μg/kg and 10 μg kg, 1 μg kg and 5 μg/kg, 5μg/kg and 40 μg/kg, 5 g/kg and 30 μg/kg, 5 μg/kg and 20 μg kg, 5 μg/kg and 10 μg kg, 10 μg/kg and 50 μξ/kg, 10 μg kg and 40 μg/kg, 10 μξ/kg and 30 μg/kg, 10 μg kg and 20 μg/kg, 10 μ§/¾ and 15 μg/kg, 20 μg/kg and 50 μg/kg, 20 μg/kg and 40 μg kg, or 20 μg/kg and 30 μg kg, of body weight of the subject.

[00134] Examples of bones which may exhibit improved BMD and/or TBS following the transdermal delivery of the PTHrP analogue include, without limitation, the lumbar spine, total hip, wrist, femur, cortical bone of the femur (femoral diaphysis), and/or femoral neck in the subject.

[00135] The transdermal delivery of the PTHrP analogue may be administered at any treatment interval necessary for therapeutic effectiveness. In certain embodiments, the transdermal delivery of the PTHrP analogue is administered on a daily basis. In other embodiments, the transdermal delivery of the PTHrP analogue may be administered every other day, every 3rd day, every 4th day, every 5th day, once a week, or once or twice a month. One of ordinary skill in the art will recognize that the treatment interval may vary over the course of treatment. For example, the transdermal delivery of the PTHrP analogue may be administered more frequently at the start of treatment, then less frequently over time as one or more therapeutic benchmarks are achieved. Alternatively, the transdermal delivery of the PTHrP analogue may be administered less frequently at the start of treatment, with the treatment interval decreasing over time.

[00136] In those embodiments of the methods provided herein wherein the transdermal delivery of the PTHrP analogue is administered using a transdermal patch provided herein. The transdermal patch may be placed in contact with the skin for any period of time necessary to achieve satisfactory analogue delivery. In certain embodiments, the transdermal patch may remain in contact with the skin for about 1 second to about 30 seconds, about 1 second to about 1 minute, about 15 second to about 30 seconds, about 15 second to about 1 minute, about 30 second to about 1 minute, about 1 minute to about 5 minutes, about 5 minutes to about 10 minutes, about 10 minutes to about 15 minutes, about 15 minutes to about 20 minutes, about 20 minutes to about 25 minutes, about 25 minutes to about 30 minutes, at least 5 minutes, at least 10 minutes, at least 15 minutes, at least 20 minutes, at least 25 minutes, at least 30 minutes, at least 35 minutes, at least 40 minutes, at least 45 minutes, at least 50 minutes, at least 55 minutes, at least 60 minutes, at least 75 minutes, at least 90 minutes, or at least 120 minutes. In certain embodiments, two or more transdermal patches may be placed in contact with the skin in a sequential manner to achieve the desired contact duration. In certain embodiments, more than one transdermal patch may be applied simultaneously.

[00137] In certain embodiments of the methods provided herein, the treatment is carried out for a set period determined in advance. In other embodiments, the treatment is carried out until one or more therapeutic benchmarks are reached. Examples of a suitable timeframe for treatment include, without limitation, 6 weeks, 12 weeks, 3 months, 24 weeks, 6 months, 48 weeks, 12 months, 18 months, and 24 months. In certain embodiments, the treatment is carried out via once a day administration of a transdermal patch for 18 months.

[00138] In certain embodiments, a subject administered a PTHrP analogue via transdermal delivery as provided herein achieves a C max which is about 80% to about 125% of the C max achieved by a subcutaneous administration of the same active agent. In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide, and the transdermal delivery of the PTHrP analogue achieves a C max which is about 80% to about 125% of the C max achieved by the abaloparatide-SC treatment.

[00139] In certain embodiments, a subject administered a PTHrP analogue via transdermal delivery as provided herein achieves an AUG which is about 80% to about 125% of the AUC achieved by a subcutaneous administration of a corresponding formulation. In certain embodiments, the PTHrP analogue comprises, consists of, or consists essentially of abaloparatide, and the transdermal delivery of the PTHrP analogue achieves a AUC which is about 80% to about 125% of the AUC achieved by the abaloparatide-SC treatment.

[00140] In certain embodiments, the PTHrP analogue formulation is administrated in combination with one or more additional osteoporosis therapies, including for example alendronate therapy. In these embodiments, the additional osteoporosis therapy may be administered before, during, or after the treatment with the PTHrP analogue formulation.

[00141] In certain embodiments of the methods disclosed herein, said administration comprises application of a force to the transdermal patch sufficient to drive one or more of the microprojections through the stratum corneum of the patient. In certain embodiments of the methods disclosed herein, the site of administration is the abdomen or the thigh.

[00142] The following examples are provided to better illustrate the claimed invention and are not to be interpreted as limiting the scope of the invention. To the extent that specific materials are mentioned, it is merely for purposes of illustration and is not intended to limit the invention. One skilled in the art may develop equivalent means or reactants without the exercise of inventive capacity and without departing from the scope of the invention. It will be understood that many variations can be made in the procedures herein described while still remaining within the bounds of the present invention. It is the intention of the inventors that such variations are included within the scope of the invention.

Examples

Example 1 : Pharmacokinetics of abaloparatide delivered via transdermal patch prepared using preparation formulations comprising PEG or ZnCb in non-human primates:

[00143] Microneedle transdermal patches coated with various formulations of abaloparatide were provided ready for use and stored refrigerated at 2-8°C. At least one hour prior to use, the transdermal patches in individual pouches were placed at room temperature.

[00144] Eight female non-naive Chinese Cynomolgus monkeys (2-4 kg at time of dosing) were included in the study. The same eight animals were used to test each' formulation, with a three day washout period between tests. Each animal received a fixed dose of abaloparatide without correcting for body weight.

[00145] The skin was prepared 24 hours prior to each transdermal patch application. A small area (5 x 5 cm) of the dorsal flank was prepared by close clipping of the hair with a small animal clipper. Care was taken during the clipping procedure to avoid abrasion of the skin. Both sides of the dorsal flank (thigh) were prepared for each administration to ensure a side without skin irritation was used for dose administration. The skin was wiped with an alcohol swab 15 minutes prior to patch application. Extra care was taken to ensure the collar of the path was firmly attached to the applicator prior to application and that the transdermal patch was firmly seated on the leg for administration.

[00146] Body weights of the animals were recorded prior to Day 1. 1.5 mL of whole blood from a peripheral vessel was collected pre-dose on Day 1 into a K 3 EDTA/aprotinin tube containing 15 μΙ_. (of 2.5 mg protein/mL/aprotinin solution) per ml of whole blood.

[00147] The transdermal patch was left in place for 15 minutes after placement. A line was drawn around the site of administration to enable post-dose observations. Each dose site was scored using the Draize scoring system pre-dose on Day I and at 1 hour and 24 hours post- dose. After patch removal, the transdermal patch was analyzed for residual content.

[00148] 1.5 mL of whole blood from a peripheral vessel was collected at 5, 10, 20, 30, 60, and 90 minutes after patch application into a K 3 EDTA/aprotinin tube containing 15 μ]_. (of 2.5 mg protein/mL/aprotinin solution) per ml of whole blood. Whole blood samples were collected within ± 5% of the scheduled collection time, with actual collection times recorded. Samples were kept on wet ice until processed. Animals were observed at each study blood collection time point. Any abnormalities were recorded by exception and reported immediately.

[00149] Whole blood samples were processed to plasma. Blood was centrifuged for 10 ± 2 minutes in a refrigerated centrifuge. Plasma samples were transferred to two approximately equal aliquots (aliquot 1 and aliquot 2). Samples were frozen at - 70°C ± 10 °C.

[00150] Abaloparatide concentrations were analyzed by LC-MS/MS. Abaloparatide serum concentrations were shown as percentage of the C max in Figures 1 A- IB and Figure 2.

[00151] The bioequivalence "window" for the abaloparatide-SC treatment was established by identifying the 80%- 125% serum concentration of abaloparatide versus time following the abaloparatide-SC treatment (Figure 1 A). The abaloparatide-SC treatment was carried out by single subcutaneous administration of an aqueous formulation of abaloparatide (2 mg/mL) in an acetate buffer (5 mg/mL tri-hydrate sodium acetate, pH 5.1 adjusted with acetic acid) further comprising phenol (5 mg mL) with a dose of 80 g abaloparatide.

[00152] Application of a transdermal patch (hereinafter, the "TD-A32") prepared by coating a microneedle array with an abaloparatide preparation formulation comprising 0.8% ZnCl 2 and 50-60% abaloparatide in water (Preparation Formulation Λ32, Figure IB) resulted in a pharmacokinetic profile that overlapped significantly with the bioequivalence window of Figure 1 A. The patch ("patch-A32")is loaded with 79 μg of abaloparatide.

[00153] Application of a transdermal patch prepared by coating a microneedle array with an abaloparatide preparation formulation comprising 14.9% PEG, and 50-60% abaloparatide in water (Preparation Formulation A31 , Figure 1C) resulted in a pharmacokinetic profile hat overlapped significantly with the bioequivalence window of Figure la. The patch is loaded with 125 μ¾ of abaloparatide.

[00154] Furthermore, modeling using fix increments were carried out to the pharmacokinetic profile of TD-A32 obtained as described supra. The abaloparatide-SC treatment data and TD-A32 data with dose of 79 μg were obtained from the experiments described supra (Table 1). The TD-A32 data with doses of 1 18.5 μ¾ 146.95 μ¾ 158 μg, and 177.75 μg (Table 1) were obtained by modeling of the experimental data of TD-A32 with dose of 79 μ with the following formulations:

Cmav of TD-A32 with dose of A ug = A

C j ax of TD- A32 with dose of 79 μg 79

AUC of TD-A32 with dose of A ug = A

AUC of TD- A32 with dose of 79 μ 79

[00155] The TD-A32 modeling data with C max 90% CI and AUC 90% CI both within the range of 80-125% were bioequivalent to the abaloparatide-SC treatment (e.g., Table 1 , TD- A32 with a dose of about 177.75 μg). Thus, Table shows that adjusting dose of abaloparatide of the transdermal administration may adjust the PK profile to achieve bioequivalence or substantial bioequivalence of the abaloparatide-SC treatment. Example 2: Pharmacokinetics of abaloparatide delivered via transdermal patch prepared using preparation formulations comprising PEG or ZnC in humans:

[00156] The pharmacokinetic profile of transdermal administration of abaloparatide and the abaloparatide-SC treatment was assessed in healthy postmenopausal women from 50 to 80 years of age, inclusive. Subjects received a single application of a transdermal patch (100 μg abaloparatide) prepared by coating with an abaloparatide formulation comprising 54% abaloparatide in l x PBS buffer (Figure 2, square), or SC-injection of 80 μg of abaloparatide in an aqueous formulation comprising acetate buffer (5 mg/mL tri-hydrate sodium acetate, pH 5.1 adjusted with acetic acid), 5 mg/mL phenol, and 2 mg/mL abaloparatide (Figure 2, diamond). Blood samples were collected at baseline and 5, 10, 15, 20, 30, 60, 90 and 120 minutes post dose. Abaloparatide concentrations were analyzed by LC-MS/MS method.

[00157] Transdermal delivery of abaloparatide using a transdermal patch prepared using an abaloparatide formulation without PEG or ZnCl 2 provided a much faster release of abaloparatide than the abaloparatide-SC treatment. Transdermal delivery using a transdermal patch prepared using an abaloparatide formulation with ZnCl 2 or PEG as an excipient as provided herein resulted in a PK profile that was much more similar to that of the abaloparatide-SC treatment.

Example 3: Pharmacokinetics of abaloparatide delivered via transdermal patch prepared using preparation formulations comprising ZnCb (Formulation A) in humans:

[00158] The pharmacokinetic profile of transdermal administration of abaloparatide and the abaloparatide-SC treatment was assessed in healthy postmenopausal women.

[00159] Subjects received a single application of a transdermal patch (500x550 patch configuration with microprojections with length of 500 micrometer) loaded with 100 μg, 150 μ or 200 μg abaloparatide, or a SC-injection of 80 g of abaloparatide.

[00160] Certain transdermal patches were prepared by coating with an abaloparatide formulation (Formulation A) comprising 0.7% ZnCl 2 , 39.2% abaloparatide, 60.1 % WFI (water for injection) (Abaloparatide 100 μg TD, Abaloparatide 150 μ§ TD, and

Abaloparatide 200 μg TD, respectively). Certain transdermal patches were loaded with 150 μg abaloparatide using a first general abaloparatide formulation comprising abaloparatide in PBS buffer (historical 150 μg TD).

[00161] Certain SC-injections of 80 μg of abaloparatide were administered by an injection pen of an aqueous formulation comprising acetate buffer (5 mg/mL tri-hydrate sodium acetate, pH 5.1 adjusted with acetic acid), 5 mg/mL phenol, and 2 mg/mL abaloparatide using an injection pen (UnoPen 80 μg SC). Certain SC-injections of 80 μg of abaloparatide were administered by injection of Formulation A (Abaloparatide 80 g SC).

[00162] Blood samples were collected at baseline and various time points up to 4 hours post dose. Abaloparatide concentrations were analyzed by LC-MS MS method. NCA (non- compartmental analyses) was performed using extravascular model. Relative actual times were used when possible, otherwise, relative nominal times were used. Nominal doses were used for the analysis. BQL were set to zero, and no subject or sample exclusions applied. Unless otherwise specified, in Figures 1 1 to 24, the box represent the 25th through 75th percentile of observations, the broken line represents the median of observations, the solid line represents the mean of observations, and the whiskers represents the extreme observations

[00163] PK results are summarized in Table 2, Figure 3 (longitude of median plasma abaloparatide concentration v. time post administration), Figure 4 (median plasma abaloparatide concentration v. time post administration), Figure 5 (longitude of mean plasma abaloparatide concentration v. time post administration), Figure 6 (mean plasma

abaloparatide concentration v. time post administration), Figure 7 (longitude of median of dose normalized plasma abaloparatide concentration v. time post administration), Figure 8 (median of dose normalized plasma abaloparatide concentration v. time post administration), Figure 9 (longitude of mean of dose normalized plasma abaloparatide concentration v. time post administration), and Figure 10 (mean of dose normalized plasma abaloparatide concentration v. time post administration), respectively.

[00164] PK results of treatment of Abaloparatide 100 g TD, Abaloparatide 150 μg TD, Abaloparatide 200 μg TD, Abaloparatide 80 μg SC, and historical 150 μg TD were compared to UnoPen 80 μg SC. (Table 3), and shown in Figures 1 1 , 13, 15, 17, 19, 21 , and 23 for C ma , AUCiast, AUCinf, Cmax/D (C max per dosage), CL/F, HL_Lambda_z, and T mM , respectively. Abaloparatide 100 μg TD, Abaloparatide 150 g TD, and Abaloparatide 200 μg TD treatments resulted in similar exposure, wherein Abaloparatide 200 μg TD showed the most promising results.

[00165] PK results of treatment of Abaloparatide 100 μg TD, Abaloparatide 150 μg TD, Abaloparatide 200 μg TD, Abaloparatide 80 μg SC, and UnoPen 80 μg SC were compared to historical 150 μg TD (Table 4). PK results of treatment of Abaloparatide 200 μg TD, and Abaloparatide 80 μg SC, compared to historical 150 μg TD were shown in Figures 12, 14, 16, 18, 20, 22, and 24 for C max , AUQ as( , AUC inf , C max /D (C max per dosage), CL/F, HL_Lambda_z, and T ma , respectively. Abaloparatide 100 μg TD, Abaloparatide 150 g TD, and Abaloparatide 200 μg TD treatments all significantly enhanced abaloparatide delivery (about twice of AUC), with lower C max (about 60% to about 70%), longer t a (about doubled), and later T max compared to the historical TD formulation. The variability was similar between the two routes of administration (SC and TD), although the range

(maximum-minimum) appeared lower for the TD administration (Figure 16). Comparison of the C max of the TD and SC delivery of Formulation A suggested a small incremental dose escalation may be needed to be more comparable (Figure 12). Figure 25 shows a PK profile of a subject treated with a transdermal patch prepared using Formulation A was within a comparable range of a UnoPen 80 μg SC treatment.

Example 4: Phase 2 study of transdermal patch prepared using first generation abaloparatide formulations in humans

[00166] Randomized, parallel-group, placebo-controlled, comparator-controlled, phase 2 study was carried out using transdermal patch prepared using a first generation abaloparatide formulation comprising abaloparatide and PBS. For 6 months, subjects received a daily TD application of a transdermal patch (with microprojections with length of 500 micrometer) loaded with 50 μ& 100 μ¾ or 150 μg abaloparatide (TD ABL 50 meg, TD ABL 100 meg, and TD ABL 150 meg, respectively), a daily SC-injection of 80 μg of abaloparatide (SC ABL 80 meg), or a placebo (TD Placebo) (Table 5).

[00167] Percent BMD changes from baseline of the subjects were determined at lumber spine (Figure 26), and total hip (Figure 27), respectively, N = 231 total. Local tolerance data were summarized in Figure 28 for % of subjects showing swelling or dermal response.

[00168] Summary of C ma x > AUC and percent BMD change from baseline of the subjects treated with the abaloparatide transdermal patch (TD-50 meg, TD-1O0 meg and TD-150 meg) and abaloparatide SC-injection (SC-80 meg) were further summarized in Table 6.

[00169] Analysis of the PK/PD relationship (C max vs BMD and AUC vs BMD) revealed a dose dependence and a linear relationship with AUC, suggesting that AUC rather than C max was a key driver of efficacy (Figures 30A-30B). AUC of the subjects treated with the abaloparatide transdermal patch (green diamonds) and the subjects treated with abaloparatide SC-injection (orange diamond) showed a linear relationship versus percent BMD changes from baseline of the subjects (Figure 30B), while C max of these subjects did not (Figure 30A). Such data suggested AUC was a key driver of the efficacy of abaloparatide treatments. [00170] As shown in Figure 28, the TD patches were well tolerated. POC for TD delivery of abaloparatide was shown, but with lower BMD gain achieved compared to the SC delivery.

[00171] PK profile of the TD patch prepared using the first generation abaloparatide formulation showed more pulsatile delivery than SC delivery with comparable C max and lower AUC (about 25-30% of SC) (Figure 29). Preliminary user experience surveys and US prescriber research suggested that both physicians and patients preferred the TD patch over SC injection by nearly 3: 1.

Example 5: Pharmacokinetics of abaloparatide delivered via transdermal patch prepared using preparation formulation comprising zinc (Formulation B) in humans

[00172] The pharmacokinetic profile of transdermal administration of abaloparatide and the abaloparatide-SC treatment was assessed in healthy postmenopausal women.

[00173] Subjects received a single application of a SC-injection of 80 μg of abaloparatide, or a single application to the thigh of a transdermal patch (500x550 patch configuration with microprojections with length of 500 micrometer) loaded with 200 μg abaloparatide prepared from preparation formulation B described below and compared to prior data generated from first generation preparation formulation containing 150 μg (historical td) abaloparatide in

PBS.

[00174] Certain transdermal patches were prepared by coating with an abaloparatide formulation (Formulation B) comprising abaloparatide and 2% ZnCl 2 in sterile water, e.g., WFI (water for injection) (Formula B TD, with 5-6% zinc chloride in the dried patch formulation). Certain transdermal patches were prepared using a first general abaloparatide formulation (First Generation TD, abaloparatide in PBS without ZnCl 2 ).

[00175] The SC-injections of 80 μ of abaloparatide were administered by an injection pen of an aqueous formulation comprising acetate buffer (5 mg/mL tri-hydrate sodium acetate, pH 5.1 adjusted with acetic acid), 5 mg/mL phenol, and 2 mg/mL abaloparatide using an injection pen (SC-injection).

[00176] Blood samples were collected at baseline and various time points up to 3 hours post dose. Abaloparatide concentrations were analyzed by LC-MS/MS method. The bars in Figure 31 represent the 25th through 75th percentile of observations.

[00177] PK results are summarized in Figures 31 and 32, showing the plasma abaloparatide concentration v. time post administration. [00178] Delivery of Formula B TD provided a PK profile much more comparable to that of SC than the first generation TD (Figures 31 and 32). Figure 31 shows the geometric mean PK profile of the subjects treated with SC-injection (SC, n = 60, average of sc dosing studies from multiple comparator transdermal v sc studies), with Formula B TD (TD Formulaion, n = 19) as geometric mean, and with First Generation TD (TD First Generation, n ~ 12).

Example 6 Pharmacokinetics of transdermal formulations containing PEG

[00179] Healthy postmenopausal volunteers were treated with an 80 μg sc injection as described previously or with a transdermal patch formulated to contain 100, 150 or 200 μg abaloparatide. The transdermal formulations were coated with PEG 3350 NF with a coating formulation consisting of approximately 40% abaloparatide, 15% PEG 3350 NF and 45% sterile water (all by weight %). The PEG patch upon drying consisted essentially of 74% abaloparatide and 26% PEG 33550 NF. The administration site was the abdomen and the pK parameters are shown in Figures 33-35.

Example 7 Pharmacokinetics of transdermal formulations containing PEG/ZnC

[00180] Healthy postmenopausal volunteers were treated with an 80 μg sc injection as described previously or with a transdermal patch formulated to contain 100, 150 or 200 μg abaloparatide. The transdermal formulations were coated with PEG 3350 NF/ZnC12 with a coating solution consisting of approximately 35% abaloparatide, 12.5% PEG3350NF, 0.7% ZnCl 2 and 52% water. The PEG/ZnCl 2 patch upon drying consisted essentially of 73% abaloparatide and 26% PEG 33550 NF, 1.5% ZnCl 2 . The administration site was the abdomen and the pK parameters are shown in Figures 36-38.

[00181] As stated above, the foregoing is merely intended to illustrate various embodiments of the present invention. The specific modifications discussed above are not to be construed as limitations on the scope of the invention. It will be apparent to one skilled in the art that various equivalents, changes, and modifications may be made without departing from the scope of the invention, and it is understood that such equivalent embodiments are to be included herein. All references in the present disclosure are hereby incorporated by reference herein in their entireties.

Example 8: Blood sample collection from female non-human primates following administration of the Abaloparatide via transdermal patch:

[00182] Microneedle transdermal patches coated with various formulations of the PTHrP analogue of SEQ ID NO: 1 were provided ready for use and stored refrigerated at 2-8°C. At least one hour prior to use, the transdermal patches in individual pouches were placed at room temperature. Eight female non-na ' ive Chinese Cynomolgus monkeys (2-4 kg at time of dosing) were included in the study. The same eight animals were used to test each formulation, with a three day washout period between tests.

[00183] Time points were calculated from the time of patch application at 5 minutes, 10 minutes, 20 minutes, 30 minutes, 1 hour, and 1.5 hours post-dose. Λ time point of pre-dose was taken at Day 1. 1.5 mL of whole blood was collected at each time point.

K EDTA/aprotinin was used as an anti-coagulant.

[00184] Each animal received a fixed dose of SEQ ID NO: 1 without correcting for body weight. The dose administration was performed as follows:

[00185] Day 1 : Test material was delivered by application of a transdermal patch. The skin was prepared 24 hours prior to each transdermal patch application as follows: a small area (5 x 5 cm) of the dorsal flank was prepared by close clipping of the hair with a small animal clipper. Care was taken during the clipping procedure to avoid abrasion of the skin. Both sides of the dorsal flank (thigh) were prepared for each administration to ensure a side without skin irritation was used for dose administration. Fifteen (15) minutes prior to patch application the skin was wiped with an alcohol swab. Extra care was taken to ensure the collar of the path was firmly attached to the applicator prior to application and that the transdermal patch was firmly seated on the leg for administration.

[00186] Days 4, 7, and 10: Test material was delivered by application of a transdermal patch. The skin was prepared 24 hours prior to each transdermal patch application as described above. Extra care was taken to ensure the collar of the path is firmly attached to the applicator prior to application and that the transdermal patch was firmly seated on the leg for administration.

[00187] Days 1, 4, 7, and 10: The transdermal patch was left in place for 15 minutes after placement. A line was drawn around the site of administration to enable post dose observations. After patch removal, the transdermal patch was analyzed for residual content.

[00188] Animals were observed at each study blood collection time point. Any

abnormalities were recorded by exception and reported immediately. Each dose site at pre- dose, 1 hour and 24 hours post-dose was scored using the Draize scoring system. Body weights of the animals were recorded prior to Day 1.

[00189] All blood samples were collected from a peripheral vessel. At Day 1, 1.5 mL of whole blood was collected pre-dose into a K 3 EDTA/aprotinin tube containing 15 (of 2.5 mg protein/mL/aprotinin solution) per ml of whole blood. At Days 1, 4, 7, and 10, 1.5 mL of whole blood was collected at each time point (5 minutes, 10 minutes, 20 minutes, 30 minutes, 60 minutes and 90 minutes after patch application) into a KsEDTA/aprotinin tube containing 15 pL (of 2.5 mg protein/mL/aprotinin solution) per ml of whole blood. Whole blood samples were collected within ± 5% of the scheduled collection time, with actual collection times recorded. Samples were kept on wet ice until processed.

[00190] The whole blood samples were processed to plasma. Blood was centrifugcd for 10 ± 2 minutes in a refrigerated centrifuge. Plasma samples were transferred to two

approximately equal aliquots (aliquot 1 and aliquot 2). Samples were frozen at -70°C ± 10°C.

A) : Pharmacokinetics of Abaloparatide formulations delivered via transdermal patch with different length microneedles:

[00191] As shown in Figure 39, abaloparatide (SEQ ID NO: 1 ) without any excipient was delivered by subcutaneous administration and by transdermal patches comprising different length of microneedles, short, regular, and long.

B ; Pharmacokinetics of various Abaloparatide formulations delivered via transdermal patch

[00192] Figures 40 and 41 show Cmax and AUC of delivery of Abaloparatide (SEQ ID NO: l) upon administration by transdermal patches coated with various coating formulations disclosed herein in comparison to those of Abaloparatide administered subcutaneously.

[00193] Figure 42 show the PK profile of subcutaneous (SC) delivery of SEQ ID NO: 1 (ABL), and transdermal delivery (TD) of SEQ ID NO: l (ABL) using patches prepared by various transdermal formulations for coating. Filled diamond: ABL administered SC;

unfilled triangle: an ABL formulation without excipient administered TD; filled circle: an ABL formulation comprising a PVP administered TD; filled square: an ABL formulation comprising a PLGA administered TD;.filled triangle: an ABL formulations comprising a PLGA administered TD; X: an ABL formulations comprising a ΗΡβΟϋ administered TD; star: an ABL formulations comprising a PLGA administered TD; unfilled circle: an ABL formulations comprising a PEG administered TD; +: an ABL formulations comprising a HP CD administered TD; unfilled square: an ABL formulations comprising ZnCl 2 administered TD. ABL plasma concentration at various time after each administration is summarized in the following table. Administra SC TD TD TD TD TD TD TD TD TD tion Route

Formulation AB AB ABL ABL ABL + ABL ABL ABL + ABL

L L + + + HPbC + + HPbC + only PVP PLG PLG D* PLG PEG D* ZnCl 2 C 17 A A A 3350

% 15% 4.90% 2.60% formulation

or

concentratio

n

Dose/patch, 145 120 96 88 1 13 253 188 84 1 19.0 meg 0

Time (min) Avarage AB plasma concentration (pg/mL)

0 0 0 0 0 0 0 4 0 0 0

5 1900 390 143 2605 2309 17283 7731 1669 5591 4448

3 2 0

10 880 242 160 1959 2385 18503 7651 1679 6496 10651

2 8 9

20 542 178 195 2250 2648 19764 7290 1383 6260 18404

9 3 0

30 222 698 893 1287 2675 20134 6390 1246 5391 20460

9

60 120 186 538 465 2539 15212 4634 8846 5082 17271

90 224 68 108 335 1927 13082 5042 7564 4913 14879

Cmax 3341 380 275 3237 3439 22261 8757 1764 7096 22090

0 8 0

Tmax 7 9 20 9 14 17 13 8 25 26

Tl/2 14 15 19 22 57 142 54 63 94 148

AUC 1316 743 743 9492 2042 14337 4742 9759 44458 14366

10 91 71 5 33 88 46 61 0 78

HPbCD is a HP CD [00194] References

References listed below are herein incorporated by reference.

1. Augustine et al., "Parathyroid hormone and parathyroid hormone-related protein analogs as therapies for osteoporosis," Curr Osteoporos Rep 1 1 :400-406 (2013).

2. Bostrom et al., "Parathyroid hormone-related protein analog RS-66271 is an effective therapy for impaired bone healing in rabbits on corticosteroid therapy," Bone 26:437-442 (2000).

3. Dean et al., "Altered selectivity of parathyroid hormone (PTH) and PTH-related protein (PTHrP) for distinct conformations of the PTH/PTHrP receptor," Mol Endocrinol 22: 156-166 (2008).

4. Dempster et al., "Skeletal histomorphometry in subjects on teriparatide or zoledronic acid therapy (SHOTZ) study: a randomized controlled trial," J Clin Endocrinol Metab 97:2799-2808 (2012).

5. Dhainaut et al., "Cortical hand bone porosity and its association with distal radius fracture in middle aged and elderly women," PLoS One 8:e68405 (2013).

6. Doyle et al., "BA058, a novel human PTHrP analog: reverses ovariectomy-induced bone loss and strength at the lumbar spine in aged cynomolgus monkeys," J Bone Miner Res 28 (Suppl 1) (2013a).

7. Doyle et al., "Long term effect of BA058, a hovel human PTHrP analog, restores bone mass in the aged osteopenic ovariectomized cynomolgus monkey," J Bone Miner Res 28 (Suppl 1) (2013a).

8. Hattersley G, Lesage E, Varela A, Mith SY. BA058, a Novel Human PTHrP Analog, Restores Bone Density and Increases Bone Strength At the Spine and Femur in Osteopenic Rats. Endocr Rev. 2013;34(abstract).

9. Horwitz et al., "Safety and tolerability of subcutaneous PTHrP(l-36) in healthy human volunteers: a dose escalation study," Osteoporos Int 17:225-230 (2006).

10. Horwitz et al., "Parathyroid hormone-related protein for the treatment of postmenopausal osteoporosis: defining the maximal tolerable dose," J Clin Endocrinol Metab 95: 1279-1287 (2010).

1 1. Kronenberg, "PTHrP and skeletal development," Ann N Y Acad Sci 1068: 1 -13 (2006). 12. Leder et al., "Two years of Denosumab and teriparatide administration in postmenopausal women with osteoporosis (The DATA Extension Study): a randomized controlled trial," J Clin Endocrinol Metab 99: 1694-1700 (2014).

13. a et al., "Comparative effects of teriparatide and strontium ranelate in the periosteum of iliac crest biopsies in postmenopausal women with osteoporosis," Bone 48:972-978 (201 1).

14. MacLean et al., "Systematic review: comparative effectiveness of treatments to prevent fractures in men and women with low bone density or osteoporosis," Ann Intern Med 148: 197-213 (2008).

15. Neer et al., "Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis," N Engl J Med 344: 1434-1441 (2001)

16. Obaidi et al., "Pharmacokinetics and Pharmacokinetics and pharmacodynamic of subcutaneously (SC) administered doses of BA058, a bone mass density restoring agent in healthy postmenopausal women," AAPS Abstract W5385 (2010)

17. Oei et al., "High bone mineral density and fracture risk in type 2 diabetes as skeletal complications of inadequate glucose control: the Rotterdam Study," Diabetes Care 36: 1619- 1628 (2013).

18. Okazaki et al., "Prolonged signaling at the parathyroid hormone receptor by peptide ligands targeted to a specific receptor conformation," Proc Natl Acad Sci USA 105: 16525- 16530 (2008)

19. Pioszak et al., "Structural basis for parathyroid hormone-related protein binding to the parathyroid hormone receptor and design of conformation-selective peptides," J Biol Chem 284:28382-28391 (2009).

20. Recker et al., "Comparative effects of teriparatide and strontium ranelate on bone biopsies and biochemical markers of bone turnover in postmenopausal women with usteoporosis," J Bone Mirier Res 24: 1358-1368 (2009).

Table 1. Modeling of TD-A32 data for bioequivalence for the abaloparatide-SC treatment (SC)

Table 2. PK Results of Abaloparatide 100 μ TD, Abaloparatide 150 μg TD, Abaloparatide 200 μg TD, Abaloparatide 80 μ SC, and Historical 150 μg TD— site of administration is abdomen

Table 3. Comparisons of Abaloparatide 100 μg TD, Abaloparatide 150 g TD, Abaloparatide 200 μ TD, Abaloparatide 80 μg SC, and Historical 150 μg TD to UnoPen 80 μg SC, respectively - Site of administration is abdomen

Table 4. Comparisons of Abaloparatide 100 μg TD, Abaloparatide 150 μ TD, Abaloparatide 200 μg TD, Abaloparatide 80 μ SC to Historical 150 μ TD, Respectively— Site of administration is abdomen

Table 5. Design for a Phase 2 study of transdermal delivery of abaloparatide using a transdermal patch prepared by a first generation (PBS) abaloparatide formulation

Table 6. C ma , AUC, and BMD improvement of a Phase 2 study of transdermal delivery of abaloparatide using a transdermal patch prepared by a first generation (PBS buffer) abaloparatide formulation