AU2013202598A1 - Sustained release small molecule drug formulation - Google Patents

Sustained release small molecule drug formulation Download PDF

Info

Publication number
AU2013202598A1
AU2013202598A1 AU2013202598A AU2013202598A AU2013202598A1 AU 2013202598 A1 AU2013202598 A1 AU 2013202598A1 AU 2013202598 A AU2013202598 A AU 2013202598A AU 2013202598 A AU2013202598 A AU 2013202598A AU 2013202598 A1 AU2013202598 A1 AU 2013202598A1
Authority
AU
Australia
Prior art keywords
formulation
small molecule
molecule drug
ratio
drug
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2013202598A
Other versions
AU2013202598B2 (en
Inventor
Guohua Chen
Gunjan H. Junnarkar
Andrew S Luk
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Durect Corp
Original Assignee
Durect Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2006299657A external-priority patent/AU2006299657B2/en
Application filed by Durect Corp filed Critical Durect Corp
Priority to AU2013202598A priority Critical patent/AU2013202598B2/en
Publication of AU2013202598A1 publication Critical patent/AU2013202598A1/en
Application granted granted Critical
Publication of AU2013202598B2 publication Critical patent/AU2013202598B2/en
Ceased legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

An injectable depot formulation includes a biocompatible polymer, an organic solvent combined with the biocompatible polymer to form a viscous gel, and a small molecule drug incorporated in the viscous gel such that the formulation exhibits an in vivo release profile having Cmax to Cmin ratio less than 200 and lag time less than 0.2. 100 +- Formulation 22 -E- Formulation 25 + Formulation 32 C I'- Formulation 33 Formulation 53 10 Formualtion 59 -l0 . C-, 0. 5 10 15 20 25 30 35 40 45 Time (days) 1 100. -- Formulation 23 E3,'- Formulation 12 0) Formulation 32 c 0 5 10 15 20 25 30 35 40 45 Time (days) -v--Formulation 55 _ -.- Formulation 64 10+- Formulation 70 0) -j0. 0 s 1 15 20 25 30 35 40 45 Time (days)

Description

1 AUSTRALIA Patents Act 1990 ALZA CORPORATION COMPLETE SPECIFICATION STANDARD PATENT Invention Title: Sustained release small molecule drug formulation The following statement is a full description of this invention including the best method of performing it known to us:- SUSTAINED RELEASE SMALL MOLECULE DRUG FORMULATION BACKGROUND OF THE INVENTION [0001] The invention relates generally to delivery of small molecule drugs. [0002] The term "small molecule drug," as used herein, refers to beneficial agents having low molecular weight. The beneficial agents are usually synthesized by organic chemistry, but may also be isolated from natural sources such as plants, fungi, and microbes. The common routes for delivering small molecule drugs are oral, injection, pulmonary, and transdermal. [0003] Many psychotherapeutic drugs are small molecule drugs and are usually provided as oral pills or bolus injections that can be administered one or more times daily. However, oral pills and bolus injections may not be optimal routes for administering small molecule psychotherapeutic drugs because of the peaks and troughs observed in plasma concentration after dosing. Adverse effects and loss of therapeutic effect have been associated with plasma concentration peaks and troughs, respectively. [0004] From the foregoing, psychotherapy as well as other forms of therapy presently relying on small molecule drugs administered in the form of oral pills and bolus injections may benefit from a sustained release dosage form designed to minimize variations in plasma concentration following dosing. Administration of psychotherapeutic agents as sustained release formulations will also increase patient compliance. BRIEF SUMMARY OF THE INVENTION [0005] In one aspect, the invention relates to an injectable depot formulation which comprises a biocompatible polymer, an organic solvent combined with the biocompatible polymer to form a viscous gel, and a small molecule drug incorporated in the viscous gel such that the formulation exhibits an in vivo release profile having C, to Cmin ratio less than 200 and lag time less than 0.2. 1fA [0006] In another aspect, the invention relates to a method of administering a small molecule drug to a subject in a controlled manner which comprises implanting in the subject an effective amount of an injectable depot formulation comprising a biocompatible polymer, an organic solvent combined with the biocompatible polymer to form a viscous gel, and a small molecule drug incorporated in the viscous gel such that the formulation exhibits an in vivo release profile having Cma, to Cmin ratio less than 200 and lag time less than 0.2. [0007] Other features and advantages of the invention will be apparent from the following description. BRIEF DESCRIPTION OF THE DRAWINGS [0008] FIG. 1 shows influence of drug salt form on in vivo release profile of formulations according to embodiments of the invention. [0009] FIG. 2 shows influence of solvent type on in vivo release profile of formulations according to embodiments of the invention. [0010] FIG. 3 shows influence of polymer type on in vivo release profile of formulations according to embodiments of the invention. [0011] FIG. 4 shows formulations having near zero-order release profiles according to embodiments of the invention. DETAILED DESCRIPTION OF THE INVENTION [0012] The invention will now be described in detail with reference to a few preferred embodiments, as illustrated in accompanying drawings. In the following description, numerous specific details are set forth in order to provide a thorough understanding of the invention. However, it will be apparent to one skilled in the art that the invention may be practiced without some or all of these specific details. In other instances, well-known features and/or process steps have not been described in detail in order to not unnecessarily obscure the invention. The features and advantages of the invention may be better understood with reference to the drawings and discussions that follow. 2 [0013] The invention is based in part on the discovery that incorporation of a sparingly soluble small molecule drug in a depot gel vehicle produces a small molecule drug formulation that has near zero-order release in vivo. The release profile shows minimal lag time and burst. For a depot formulation, this release profile is surprising because the prevailing thought in the art is that a low burst, near zero-order release is virtually impossible to attain unless special steps are taken, such as coatings for drugs and microencapsulation. Several small drug formulations have been identified in this invention with in vivo release profiles having a Cmax to Cmin ratio less than 200 and lag time, Tiag, less than 0.2. [0014] The variable "Cm" is the minimum drug concentration in plasma or serum. The variable "Cma" is the maximum drug concentration in plasma or serum. The variable "Tag" is the ratio of Tvaniey to Troa, where Tvaiey is less than Tt 0
,
8 s. The variable "Tvaney" is the time to reach Cvaniey. The variable "Cv 1 1 y" is the first trough of drug concentration in plasma or serum during release. The variable "Tt 0 ,t" is the total release duration. [0015] Small molecule drug formulations according to embodiments of the invention can be prepared as depot injections. The environment of use is a fluid environment and may include a subcutaneous, intramuscular, intramyocardial, adventitial, intratumoral, or intracerebral portion, a wound site, or tight joint spaces or body cavity of a human or animal. Multiple or repeated injections may be administered to the subject, for example, when the therapeutic effect of the drug has subsided or the period of time for the drug to have a therapeutic effect has lapsed or when the subject requires further administration of the drug for any reason. The formulation serves as an implanted sustained release drug delivery system after injection into the subject. Such controlled release can be over a period of one week, more than one week, one month, or more than one month. Preferably, the controlled release is over at least a period of one week, more preferably over a period of at least one month. [0016] A small molecule drug formulation according to an embodiment of the invention includes a depot gel vehicle. The depot gel vehicle includes a biocompatible polymer, i.e., a polymer that would not cause irritation or necrosis in the environment of use. Biocompatible polymers that may be useful in the invention may be bioerodible, i.e., gradually decompose, dissolve, hydrolyze and/or erode in situ. Examples of bioerodible 3 polymers include, but are not limited to, polylactides, polyglycolides, polycaprolactones, polyanhydrides, polyamines, polyurethanes, polyesteramides, polyorthoesters, polydioxanones, polyacetals, polyketals, polycarbonates, polyorthocarbonates, polyphosphazenes, succinates, poly(malic acid), poly(amino acids), polyvinylpyrrolidone, polyethylene glycol, polyhydroxycellulose, polysaccharides, chitin, chitosan, and copolymers, terpolymers and mixtures thereof The polymer is typically present in the depot gel vehicle in an amount ranging from about 5 to 80% by weight, preferably from about 20 to 70%, often from about 40 to 60% by weight. [0017] In one embodiment, the polymer is a polylactide. A polylactide polymer is a polymer based on lactic acid or a copolymer based on lactic acid and glycolic acid. The polylactide polymer can include small amounts of other comonomers that do not substantially affect the advantageous results that can be achieved in accordance with the invention. The term "lactic acid" includes the isomers L-lactic acid, D-lactic acid, DL-lactic acid, and lactide. The term "glycolic acid" includes glycolide. The polymer may have a lactic-acid to glycolic-acid monomer ratio of from about 100:0 to 15:85, preferably from about 60:40 to 75:25, often about 50:50. The polylactide polymer has a number average molecular weight ranging from about 1,000 to about 120,000, preferably from about 5,000 to about 30,000, as determined by gel permeation chromatography. Suitable polylactide polymers are available commercially. [0018] The depot gel vehicle further includes a biocompatible solvent which when combined with the polymer forms a viscous gel, typically exhibiting viscosity in a range from 500 poise to 200,000 poise, preferably from about 1,000 poise to 50,000 poise. The solvent used in the depot gel vehicle is typically an organic solvent and may be a single solvent or a mixture of solvents. To limit water intake by the depot gel vehicle in the environment of use, the solvent, or at least one of the components of the solvent in the case of a multi-component solvent, preferably has limited miscibility with water, e.g., less than 7% by weight, preferably less than 5% by weight, more preferably less than 3% by weight miscibility with water. Examples of suitable solvents include, but are not limited to, benzyl benzoate (BB), benzyl alcohol (BA), ethyl benzoate (EB), triacetin, and N-methyl-2-pyrrolidone (NMP). The solvent is typically present in the depot gel vehicle in an amount ranging from about 20 to 4 95% by weight, preferably in an amount ranging from about 30 to 80% by weight, often in an amount ranging from about 40 to 60 % by weight. [0019] A formulation according to an embodiment of the invention includes a small molecule drug dispersed or dissolved in a depot gel vehicle as described above. The term "dispersed or dissolved" is intended to encompass all means of establishing the presence of the small molecule drug in the viscous gel and includes dissolution, dispersion, suspension, and the like. Small molecule drugs used in formulations of the invention are sparingly soluble in water. In a preferred embodiment, small molecule drugs used in formulations of the invention have less than I mg/ml solubility in water. In one embodiment, small molecule drugs used in formulations of the invention have a molecular weight in a range from 200 to 2,000 Daltons. Small molecule drugs used in formulations of the invention may have a narrow or wide therapeutic window. However, the invention generally delivers salubrious results in terms of C,, and toxicity control for small molecule drugs having a narrow therapeutic window. The small molecule drug is typically present in the formulation in an amount ranging from about 1 to 50% by weight, more preferably in an amount ranging from about 5 to 40% by weight, often in an amount ranging from about 10 to 30% by weight. [0020] In one embodiment, a small molecule drug formulation includes a small molecule psychotherapeutic drug, such as a small molecule antipsychotic, dopamine receptor agonist, dopamine receptor antagonist, serotonin receptor agonist, serotonin receptor antagonist, and serotonin uptake inhibitor drug. Table 1 below shows physiochemical properties of some small molecule psychotherapeutic drugs. R209130-base has the molecular formula Cj 9
H
20 FNO. R209130-mandelic acid salt (R209130) has the molecular formula
C
19
H
20
FNO.C
8
H
8 0 3 . R209130-tartaric acid salt (R167154) has the molecular formula
C
19
H
20
FNO.C
4
H
6 0 6 . R209130 and its analogs possess putative atypical antipsychotic properties and have demonstrated antianxiety, antidepressive, and socializing effects in animal models. These characteristics may be attributed to R209130 dual antagonism of central dopamine D2 receptors, serotonin 5-HT2A and 5-HT 2 c receptors, and the inhibition norepinephrine uptake. Risperidone-base has the molecular formula C 23
H
27
FN
4 0 2 . Risperidone-pamoate has the molecular formula C 2 3
H
2 7
FN
4 0 2
.C
23
H
16
O
6 . Risperidone is a combined serotonin (5-HT 2 ) and dopamine (D2) receptor antagonist. 5 TABLE 1 Property R209130 R167154 R209130 Risperidone Risperidone base base pamoate pKa 9.2 9.2 9.2 8.2/3.1 8.2/3.1 Solubility in 0.32 (pH 41.84 (pH 0.008 (pH 0.09 (pH 8.8) 0.2 (pH 7.2)
H
2 0 (mg/ml) 4.9) 3.4) 9.5) Solubility at 0.35 6.1 (pH 6.5) 2 1 0.2 (pH 7.2) pH 7 (mg/ml) Solubility in 58.6 at 10.3 at 40*C > 200,000 32,000 50 BB (ptg/ml) 40"C Solubility in 7.3 at 40 0 C 41.3 at 4 0 "C > 200,000 407 2.97 BA (mg/ml) Intrinsic 0.054 3.7 0.7 0.0025 N/A dissolution rate (mg/cn 2 .min) LogP 3.9 4.0 N/A 3.04 N/A (CsOH/pH 7 buffer) Molecular 449.5 447.5 297.4 410.5 798.5 weight [0021] A study was conducted to determine the PK profile of a small molecule drug delivered from a depot gel vehicle according to the invention and the influence of salt form of the drug, solvent type, polymer type, polymer molecular weight, polymer/solvent ratio, drug loading, and particle size on the PK profile. [0022] The following examples are presented for illustration purposes and are not intended to limit the invention as otherwise described herein. EXAMPLE 1 [0023] A depot gel vehicle was prepared as follows: A HDPE container was tared on a Mettler PJ3000 top loader balance. Poly D,L-lactide-co-glycolide (PLGA), (L/G ratio of 50/50), available as RESOMER@ RG 502 (PLGA-502), was weighed into the container. The container containing PLGA-502 was tared, and the corresponding solvent was added to the PLGA-502. Amounts expressed as percentages for various combinations of PLGA-502 and 6 solvent are set forth below in Table 2. A hybrid mixer was used to mix the PLGA-502 and solvent mixture, resulting in a clear gel-like solution of the polymer in the solvent. TABLE 2 Formulation PLGA-502 (wt%, g) Benzyl Benzoate Benzyl Alcohol (wt%, g) (wt%, g) A 50.067 50.044 B 50.023 24.988 24.988 C 50.365 45.093 5.1780 D 50.139 37.553 12.560 E 50.350 45.193 [0024] Additional depot gel vehicles were prepared with solvents, selected from benzyl benzoate (BB), benzyl alcohol (BA), ethyl benzoate (EB), ethyl hydroxide (EtOH), triacetin, and N-methyl-2-pyrrolidone (NMP), and mixtures thereof, and polymers, selected from Poly D,L-lactide, available as RESOMER® L 104, RESOMER® R 104, RESOMER® 202, RESOMER@ 203, RESOMER® 206, RESOMER® 207, RESOMER® 208; PLGA, L/G ratio of 50/50, available as RESOMER@ RG 502H; PLGA, L/G ratio of 50/50, available as RESOMER@ RG 503; PLGA, L/G ratio of 50/50, available as RESOMER@ RG 755; Poly L-lactide, molecular weight of 2000, available as RESOMER® L 206, RESOMER® L 207, RESOMER@ L 209, RESOMER® L 214; Poly L-lactide-co-D,L lactide, L/G ratio of 90/10, available as RESOMER@ LR 209; PLGA, L/G ratio of 75/25, available as RESOMER@ RG 752, RESOMER® RG 756, PLGA, L/G ratio of 85/15, available as RESOMER® RG 858; Poly L-lactide-co-trimethylene carbonate, L/G ratio of 70/30, available as RESOMER® LT 706, and Poly dioxanone, available as RESOMER® X210 (Boebringer Ingelheim Chemicals, Inc. Petersburg, VA); DL-lactide/glycolide (DL), L/G ratio of 100/0, available as MEDISORB@ Polymer 100 DL High, MEDISORB® Polymer 100 DL Low; DL-lactide/glycolide (DL), L/G ratio of 85/15, available as MEDISORB@ Polymer 8515 DL High, MEDISORB@ Polymer 8515 DL Low; DL lactide/glycolide (DL), L/G ratio of 75/25, available as MEDISORB® Polymer 7525 DL High, MEDISORB@ Polymer 7525 DL Low; DL-lactide/glycolide (DL), L/G ratio of 65/35, available as MEDISORB® Polymer 6535 DL High, MEDISORB@ Polymer 6535 DL Low; 7 DL-lactide/glycolide (DL), L/G ratio of 54/46, available as MEDISORB@ Polymer 5050 DL High, MEDISORB@ Polymer 5050 DL Low, MEDISORB® 5050 Polymer DL 2A(3), MEDISORB@ 5050 Polymer DL 3A(3), MEDISORB@ 5050 Polymer DL 4A(3) (Medisorb Technologies International L.P., Cincinnati, OH); and PLGA (L/G ratio of 50/50), PLGA (L/G ratio of 65/35), PLGA (L/G ratio of 75/25), PLGA (L/G ratio of 85/15), Poly D,L lactide, Poly L-lactide, Poly glycolide, Poly E-caprolactone, Poly D,L-lactide-co-caprolactone (L/C ratio of 25/75), and Poly D,L-lactide-co-caprolactone (L/C ratio of 75/25), available from Birmingham Polymers, Inc., Birmingham, AL. Polycaprolactone-glycolic acid-lactic acid copolymer (PCL-GA-LA) was also used either mixed with polyvinylpyrrolidone (PVP) or by itself. Typical molecular weights of these polymers are in the range of 6,000 to 20,000. EXAMPLE 2 [0025] Drug particles were prepared as follows: R209130, R167154, risperidone base, or risperidone pamoate drug was passed through sieves of different sizes to obtain drug particles having a certain range of particle size distribution. Particles in the range of 20 to 63 Rm, 63 to 125 jim, 75 to 125 jim, or less than 38 Rm were obtained. Micronized particles received were also used as drug particles. EXAMPLE 3 [0026] Depot formulations were prepared as follows: sieved drug particles prepared as described in Example 2 were added into the depot gel vehicles prepared as described in Example 1 in an amount of 0 to 50% by weight and blended manually until the drug particles were wetted completely. Then, the mixture of drug particles and depot gel was thoroughly blended by conventional mixing using a Caframo mechanical stirrer with an attached square tip metal spatula. Final homogeneous gel formulations were transferred to 3, 10, or 30 cc disposable syringes for storage or dispensing. 8 EXAMPLE 4 [0027] A representative number of implantable gels were prepared in accordance with the foregoing procedures and tested in vivo in rats to determine release of the drug as determined by blood serum or plasma concentration of drug as a function of time. [0028] In general, in vivo studies in rats were performed following an open protocol to determine plasma levels of the drug (e.g., R209130, R167154, risperidone base, risperidone pamoate) upon systemic administration of the drug via the implant systems of the invention. Depot gel formulations containing the drug, prepared as described in the Examples above, were loaded into 0.5 cc disposable syringes. Disposable needles (18 gauge) were attached to the syringes and heated to 37*C using a circulator bath. The depot gel formulations were injected into rats. Blood was drawn at specified time intervals and analyzed for drug content. All plasma samples were stored at 4*C prior to analysis. EXAMPLE 5 [0029] This example investigates influence of drug salt form on in vivo release of small molecule drugs from depot gel vehicles. [0030] Particles of R209130 and R167154, in appropriate size range, were incorporated in depot gel vehicles, as per procedure in Example 3. Resulting formulations are illustrated in Table 2 below. Final homogeneous depot formulations were transferred to 3, 10, or 30 cc disposable syringes for storage or dispensing. In vivo release of the drugs were analyzed, as per procedure in Example 4. In vivo release profiles of the formulations are shown in FIG. 1. Cmax to Cmin ratio and Tiag of the formulations are shown in Table 2. R167154 and R209130 are different salt forms of the same drug. Formulation 7 (R209130) has Cmax to Cmin ratio of 19.2 and Tiag of 0.61, while formulation 3 (R167154) has Cax to Cmnin ratio of 25.7 and Trag of 0.33. This example shows that in vivo release is influenced by salt form of the formulation. Even though Tias for formulation 7 (R209130) is higher than Tiag for formulation 3 CR167154), formulation 7 appears to have better release rate profile and duration of release in comparison to formulation 3. 9 TABLE 2 No. PLGA BA BB (wt%) Triacetin Drug Cmax/Cmin Tiag (wt%) (wt/o) (wt%) (wt%) 3 ",aj,!A 45 22.5 22.5 0 10 25.7 0.33 7 1,al,,B 45 22.5 22.5 0 10 19.2 0.61 1 = R209130, 2 = R167154, 3 = risperidone base, 4 risperidone pamoate; a = 50150 PLGA-502 (MW = 16,000), b = 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d = 40/55/5 PCL-GA-LA (MW = -13,500), e = 75/25 PLGA (MW = 14,300), f = 80/20 PCL-GA-LA/PVP, g = RG502:RGS02H (1:1); a = P/S ratio of 50/50, f= P/S ratio of 40/60, x= P/S ratio of 45/55, 8 = P/S ratio of 60/40,6 = P/S ratio of 55/45; A = 63-125 pm, B = 20-63 pm, C = 75-125 pm, D = <38 pm, E = micronized, F = as is, G = not applicable; NV = no valley EXAMPLE 6 [0031] This example investigates influence of solvent type on in vivo release of small molecule drugs from depot gel vehicles. [0032] Depot gel vehicles were prepared with PLGA-502 and a solvent selected from BA, BB, EB, EtOH, NMP, and triacetin, and combinations thereof, as per procedure in Example 1. The depot gel vehicles were loaded with drug substance, in appropriate range, as per procedure in Example 3. Resulting formulations are illustrated in Table 3 below. Final homogeneous depot formulations were transferred to 3, 10 or 30 cc disposable syringes for storage or dispensing. In vivo release profiles of the formulations in Table 3 are shown in FIG. 2. Cmax to Cmin ratio and Tiag of the formulations are shown in Table 3. TABLE 3 Target content in formulation (% w/w) No. PLGA BA BB BtOH NMP Triacetin EB Drug Cnax/ Tiag Cmin 2T1'' 45 0 45 0 0 0 0 10 59.86 NV 3aaA 45 22.5 22.5 0 0 0 0 10 25.68 0.33 10 ,a, Iu, 40 40 0 0 0 0 0 20 4.35 0.61 14a,uiAc 40 20 20 0 0 0 0 20 3.15 0.50 63"'72' ' 43.3 0 0 0 0 43.3 0 13,4 1364.43 0.14 10 Target content in formulation (% w/w) No. PLGA BA BB EtOH NMP Triacetin EB Drug Cmax/ Tiag Cmin 73 ".'** 43.3 0 0 0 0 0 43.3 13.4 5.20 N/A 1 = R209130, 2 R167154, 3 =risperidone base, 4 =risperidone pamoate, a= 50/50 PLGA-502 (MW= 16,000), b 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d = 40/55/5 PCL-GA-LA (MW =-13,500), e 75/25 PLGA (MW = 14,300), f = 80/20 PCL-GA-LA/PVP, g = RG502:RG502H (1:1); a= P/S ratio of 50/50, 0 = P/S ratio of 40/60, x= P/S ratio of 45/55, 6 = P/S ratio of 60/40, e = P/S ratio of 55/45; A = 63-125 pm, B = 20 63 pm, C = 75-125 pm, D = <38 pm, E = micronized, F = as is, G = not applicable; NV = no valley [0033] In Table 3 above, formulation 63 (risperidone base/PLGA/triacetin depot) has a Cmax to Cmin ratio of 1364.64. On the other hand, formulation 73 (risperidone base/PLGA/EB depot) has a Cmax to Cmin ratio of 5.20, which is significantly lower than the Cmax to Cmin ratio for formulation 63. Formulation 2 (R167154/PLGA/BB depot) has a Cmax to Cmin ratio of 59.68. On the other hand, formulation 3 (R167154/PLGA/BA/BB) has a Cmax to Cmini ratio of 25.68, which is less than half the Cmax to Cmiin ratio for formulation 2. This indicates that solvent type can influence in vivo release profile of the formulation. EXAMPLE 7 [0034] This example investigates influence of polymer type on in vivo release of small molecule drugs from depot gel vehicles. [0035] Depot gel vehicles were prepared with different polymers and loaded with R209130, in appropriate size range, as per procedure in Example 3. Resulting formulations are illustrated in Table 4 below, Final homogeneous depot formulations were transferred to 3, 10 or 30 cc disposable syringes for storage or dispensing. Table 4 shows Cmax to Cmin ratio and Tlag for in vivo release profiles of the formulations. FIG. 3 shows in vivo release profiles of formulations in Table 4. TABLE 4 Target content in formulation (% w/w) No. Polymer BA BB Drug Cmax/Cmin Tiag 21,av,a,C 35 35 0 30 9.86 0.17 23 1,a,Iva,C 35 0 35 30 6.83 0.17 11 Target content in formulation (% w/w) No. Polymer BA BB Drug Cmax/Cmin Tiag 2 4 1Ia,IV,x,E 35 0 35 30 44.0 NV 2 5 1,c,1v,a,c 35 0 35 30 29.49 0.45 32 L'd'i'"' 35 0 35 30 10.65 0.12 3 3 "tjgC 35 0 35 30 6.35 0.14 3 4 1a,,vc 35 35 0 30 8.75 0.23 35 ['I','4U 35 0 35 30 44.21 NV 4 8 1 ,cQ~b 35 0 35 30 163.12 NV 53 *'N'u 35 0 35 30 31.16 0.25 5 9 ''dIV'' 35 0 35 30 6.26 0.07 1 = R209130, 2 R 167154, 3 = risperidone base, 4 = risperidone pamoate, a = 50/50 PLGA-502 (MW 16,000), b = 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d = 40/55/5 PCL-GA-LA (MW = -13,500), e =75/25 PLGA (MW = 14,300), f = 80/20 PCL-GA-LA/PVP, g = RG502:RG502H (1:1); a = P/S ratio of 50/50, $= P/S ratio of 40/60, x= P/S ratio of 45/55,6 = P/S ratio of 60/40, e = P/S ratio of 55/45; A = 63-125 pm, B = 20-63 pm, C = 75-125 pm, D = <38 pm, E = micronized, P = as is, G = not applicable; NV = no valley EXAMPLE 8 [0036] This example investigates influence of molecular weight of polymers on in vivo release of small molecule drugs from depot gel vehicles. [0037] Depot gel vehicles were prepared with polymers with different molecular weights and loaded with drug substance, in appropriate size range, as per procedure in Example 3. Resulting formulations are illustrated in Table 5 below. Final homogeneous depot formulations were transferred to 3, 10 or 30 cc disposable syringes for storage or dispensing. Table 5 shows Cmax to Cmin ratio and Tla for in vivo release profiles of the formulations. TABLE 5 Target content in Formulation (% w/w) No. PLGA BA BB Triacetin Drug Cax/Cmin Tiag 10 ,a,mIIIaC 40 40 0 0 20 4.35 0.61 1 1 ba,m,a,D 40 40 0 0 20 12.06 0.61 12',a,IV,a,C 35 35 0 0 30 4.78 0.14 12 Target content in Formulation (% w/w) No. PLGA BA BB Triacetin Drug Cmax/Cmn Tiag 13 a,1vD 35 35 0 0 30 5.29 0.36 21 40 40 0 0 20 48.55 No valley 251,r,1v,a,c 35 0 35 0 30 29.49 0.45 2 6 1,cNV,,D 35 0 35 0 30 41.67 No valley 4 8 1,c,v,(x,E 35 0 35 0 30 163.12 No valley 4 9 1,c,Iv,&,E 42 0 28 0 30 66.31 0.39 63 3,aVII,a,C 43.3 0 0 43.3 13.4 1364.43 0.14 64 4,cvl,a,c 36.9 0 36.9 0 26.1 11.66 No valley 6 9 4,aV111,a,E 36.9 0 36.9 0 26.1 14.12 0.90 704,cmc 36.9 0 36.9 0 26.1 22.11 no valley 723,a,vra,G 43.3 0 43.3 0 13.4 24.48 N/A 1 = R209130, 2= R167154, 3 = risperidone base, 4 = risperidone pamoate, a = 50/50 PLGA-502 (MW = 16,000), b= 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d= 40/55/5 PCL-GA-LA (MW = -13,500), e = 75125 PLGA (MW = 14,300), f= 80/20 PCL-GA-LA/PVP, g RG502:RG502H (1:1); a = P/S ratio of 50/50, f = P/S ratio of 40/60, x= P/S ratio of 45/55, 6 = P/S ratio of 60/40, E = P/S ratio of 55145; A = 63-125 jim, B = 20-63 pm, C = 75-125 pm, D = <38 pm, E = micronized, F = as is, G = not applicable; NV = no valley EXAMPLE 9 [0038] This example investigates influence of polymer/solvent ratios on in vivo, release of small molecule drags from depot gel vehicles. [0039] Depot gel vehicles were prepared with different polymer/solvent ratios and loaded with drug substance, in appropriate size range, as per procedure in Example 3. Resulting formulations are illustrated in Table 6 below. Final homogeneous depot formulations were transferred to 3, 10 or 30 cc disposable syringes for storage or dispensing. Table 6 shows Cmx to Cmin ratio and Tiag for in vivo release profiles of the formulations. TABLE 6 Target content in Formulation (% w/w) 13 No. PLGA BB EtOH Drug Cmax/Cmin Tias 22"O"v'a'c 35 0 0 30 9.86 0.17 231,"v'm'c 35 35 0 30 6.83 0.17 2 4 1,aIva,E 35 35 0 30 44.00 NV 251cv,a,c 35 35 0 30 29.49 0.45 2 6 .c,VAD 35 35 0 30 41.67 NV 2 7 1,cVsc 28 42 0 30 54.16 NV 28 ac,IvpD 28 42 0 30 120.74 NV 2 9 1,ajV,x,C 31.5 34.65 3.85 30 1.93 NV 301a,wV, 31.5 34.65 3.85 30 7.07 0.29 4 8 1,jV~apE 35 35 0 30 163.12 NV 4 9 1,c,IV,6,E 42 28 0 30 66.31 0.39 521,eIv'OE 28 42 0 30 47.86 NV 5 3 1eAWCE 35 35 0 30 31.16 0.25 5 6 1,b,V,,F 38.5 31.5 0 30 17.10 NV 6 5 4,cVIII,aE 36.9 36.9 0 26.1 50.87 NV 6 6 4,c,VUIc,,G 40.6 33.2 0 26.1 38.39 NV 6 7 4,enVII,s,G 33.2 40.6 0 26.1 43.55 NV 1 = R209130, 2= R167154, 3 risperidone base, 4 = risperidone pamoate, a = 50/50 PLGA-502 (MW = 16,000), b = 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d = 40/55/5 PCL-GA-LA (MW = -13,500), e = 75/25 PLGA (MW = 14,300), f = 80/20 PCL-GA-LA/PVP, g= RG502:RG502H (1:1); a= P/S ratio of 50/50, # = P/S ratio of 40/60, x= P/S ratio of 45/55, 8 = P/S ratio of 60/40, e = P/S ratio of 55/45; A = 63-125 in, B = 20-63 im, C = 75-125 gm, D =<38 mi, E = micronized, F = as is, G = not applicable; NV = no valley EXAMPLE 10 [0040] This example investigates influence of drug loading on in vivo release of small molecule drugs from depot gel vehicles [0041] Depot gel vehicles were prepared with varying percentages of drug, in appropriate size range, as per procedure in Example 3. Resulting formulations are illustrated in Table 7 below. Final homogeneous depot formulations were transferred to 3, 10 or 30 cc disposable syringes for storage or dispensing. Table 7 shows Cmax to Cmrn ratio and Tas for in vivo release profiles of the formulations. 14 TABLE 7 Target content in Formulation (% w/w) Formulation No. PLGA BA BB Drug Cmax/Cmin Tiag 4 ,a,1,a,B 45 45 0 10 4.37 0.50 5 ]a1,1, 40 20 20 20 10.66 0.61 7 1,a,II,a,B 45 22.5 22.5 10 19.17 0.61 1 0 ,aI1aC 40 40 0 20 4.35 0.61 1 1 1,a,III,a,D 40 40 0 20 12.06 0.61 12 1,a,IV,cC 35 35 0 30 4.78 0.14 13 1,aV,,D 35 35 0 30 5.29 0.36 14 1,a,m,a,c 40 20 20 20 3.15 0.50 151admIaD 40 20 20 20 9.60 0.61 161,a,1vac 35 17.5 17.5 30 7.16 0.61 1 7 1,a,V,aD 35 17.5 17.5 30 17.35 0.36 181,av,ac 30 30 0 40 3.54 0.39 1 = R209130, 2 =R167154, 3 = risperidone base, 4 =risperidone panoate, a= 50/50 PLGA-502 (MW = 16,000), b = 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d = 40/5515 PCL-GA-LA (MW = -13,500), e =75/25 PLGA (MW = 14,300), f = 80/20 PCL-GA-LA/PVP, g = RG502:RG502H (1:1); a = P/S ratio of 50/50, 3= P/S ratio of 40/60, X= P/S ratio of 45/55, 5 = P/S ratio of 60/40, e = P/S ratio of 55/45; A = 63-125 pn, B = 20-63 pin, C = 75-125 in, D = <38 pm, E = micronized, F = as is, G = not applicable; NV = no valley EXAMPLE 11 [0042] This example investigates influence of drug particle size on in vivo release of small molecule drags from depot gel vehicles. [0043] Depot gel vehicles were prepared and loaded with drug particles in appropriate size range, as per procedure in Example 3. Resulting formulations are illustrated in Table 8 below. Final homogeneous depot formulations were transferred to 3, 10 or 30 cc disposable syringes for storage or dispensing. Table 8 shows Cmax to Cmin ratio and Tia, for in vivo release profiles of the formulations. TABLE 8 Target content in Formulation (% w/w) 15 Formulation No. PLGA BA BB Drug Cmax/Cmin Tiag 71,aj,,,B 45 22.5 22.5 10 19.17 0.61 101,a,rr,aC 40 40 0 20 4.35 0.61 11 40 40 0 20 12.06 0.61 231,a,c 35 0 35 30 6.83 0.17 2 4 1,a,V,a,3 35 0 35 30 44.00 NV 2 5 1,olv,a,c 35 0 35 30 29.49 0.45 261 35 0 35 30 41.67 NV 6 4 4,,VmI,a,c 36.9 0 36.9 26.1 11.66 NV 654,Cv,3 36.9 0 36.9 26.1 50.87 NV 664,c,vm,,G 40.6 0 33.2 26.1 38.39 NV 7 2 3,a,VI[a,G 43.3 0 43.3 13.4 24.48 N/A 1= R209130, 2 R167154, 3 = risperidone base, 4 =risperidone pamoate, a= 50/50 PLGA-502 (MW = 16,000), b = 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d= 40/55/5 PCL-GA.LA (MW = -13,500), e = 75/25 PLGA (MW = 14,300), f= 80/20 PCL-GA-LA/PVP, g = RG502:RG502H (1:1); a = P/S ratio of 50/50, # = P/S ratio of 40/60, X= P/S ratio of 45/55, 0 = P/S ratio of 60/40, e = P/S ratio of 55/45; A = 63-125 pm, B = 20-63 pm, C = 75-125 pm, D = <38 prm, E = micronized, F = as is, G = not applicable; NV no valley EXAMPLE 12 [0044] A formulation is described as near zero-order if the ratio of Cmax to Cmin is less than 200, preferably less than 50, more preferably less than 30. Tiag in release of formulation is preferably less than 0.2. Formulations that do not show Cvaney do not exhibit lag. Table 9 shows a number of formulations that exhibited the characteristic near zero-order release. FIG. 4 shows in vivo release profiles of selected formulations in Table 9. TABLE 9 Target content in Formulation (% w/w) Formulation Polymer BA BB EtOH Drug Cmax/Cmin Tiag No. Particles 121,a'iv',e 35 35 0 0 30 4.78 0.14 221,vC 35 35 0 0 30 9.86 0.17 35 0 35 0 30 6.83 0.17 291,v,xC 31.5 0 34.65 3.85 30 1.93 NV 16 Target content in Formulation (% w/w) Formulation Polymer BA BB EtOH Drug Cmax/Cmin Tag No. Particles 32 35 35 0 0 30 10.65 0.12 33 1Tv4U 35 0 35 0 30 6.35 0.14 354'I4' 35 0 35 0 30 44.21 NV 55 .*',4' 35 0 35 0 30 6.33 0.11 56 38.5 0 31.5 0 30 17.10 NV 60"4' 25 0 25 0 50 12.90 0.07 61I'l4' 35 0 35 0 30 26.53 0.11 64 4,c,vImizc 36.9 0 36.9 0 26.1 11.66 NV 704'4C4' 36.9 0 36.9 0 26.1 22.11 NV I = R209130, 2 = R167154, 3 = risperidone base, 4 = risperidone pamoate; a = 50/50 PLGA-502 (MW = 16,000), b = 50/50 PLGA-502H (MW = 11,000), c = 50/50 PLGA (MW = 6400), d = 40/55/5 PCL-GA-LA (MW = -13,500), e 75/25 PLGA (MW = 14,300), f = 80/20 PCL-GA-LA/PVP, g = RG502:RG502H (1:1); a = P/S ratio of 50/50, j= P/S ratio of 40/60, x= P/S ratio of 45/55, 8 = P/S ratio of 60/40, = P/S ratio of 55/45; A = 63-125 si, B = 20-63 gm, C= 75-125 a, D = <38 im, E = micronized, F = as is, G= not applicable; NV = no valley [0045] While the invention has been described with respect to a limited number of embodiments, those skilled in the art, having benefit of this disclosure, will appreciate that other embodiments can be devised which do not depart from the scope of the invention as disclosed herein. 17

Claims (19)

  1. 2. The formulation of claim 1, wherein the in vivo release profile has Cmx to Cmin ratio less than 30.
  2. 3. The formulation of claim 1, wherein the small molecule drug is sparingly soluble in water.
  3. 4. The formulation of claim 1, wherein a solubility of the small molecule in water is less than 1 mg/ml.
  4. 5. The formulation of claim 1, wherein the polymer is a polylactide.
  5. 6. The formulation of claim 5, wherein the polymer has a monomer ratio of lactic acid to glycolic acid in the range from 100:0 to 15:85.
  6. 7. The formulation of claim 1, wherein the polymer is selected from the group consisting of polylactides, polyglycolides, polycaprolactones, polyanhydrides, polyamines, polyurethanes, polyesteramides, polyorthoesters, polydioxanones, polyacetals, polyketals, polycarbonates, polyorthocarbonates, polyphosphazenes, succinates, poly(malic acid), poly(amino acids), polyvinylpyrrolidone, polyethylene glycol, polyhydroxycellulose, polysaccharides, chitin, chitosan, and copolymers, terpolymers and mixtures thereof. 18
  7. 8. The formulation of claim 1, wherein the solvent is selected from the group consisting of benzyl alcohol, benzyl benzoate, ethyl benzoate, ethyl hydroxide, triacetin, N methyl-2-pyrrolidone, and mixtures thereof.
  8. 9. The formulation of claim 1, wherein the small molecule drug comprises an antipsychotic agent.
  9. 10. The formulation of claim 1, wherein the small molecule drug is selected from the group consisting of dopamine receptor agonist, dopamine receptor antagonist, serotonin receptor agonist, serotonin receptor antagonist, serotonin uptake inhibitor, and combinations thereof.
  10. 11. The formulation of claim 1, wherein the small molecule drug is selected from the group consisting of base and salt forms of R209130 and risperidone.
  11. 12. The formulation of claim 1, wherein the small molecule drug has a molecular weight in a range from 200 to 2,000 Daltons.
  12. 13. The formulation of claim 1, wherein the solvent has a miscibility in water less than 7% by weight.
  13. 14. The formulation of claim 1, wherein the small molecule drug .is in the form of particles having an average particle size in a range from 0.1 to 125 pm.
  14. 15. The formulation of claim 1, which releases the small molecule drug at or near target rate over at least one week.
  15. 16. The formulation of claim 1, which releases the small molecule drug at or near target rate for up to one month. 19
  16. 17. A method of administering a small molecule drug to a subject in a controlled manner, comprising: implanting in the subject an effective amount of an injectable depot formulation comprising a biocompatible polymer, an organic solvent combined with the biocompatible polymer to form a viscous gel, and a small molecule drug incorporated in the viscous gel such that the formulation exhibits an in vivo release profile having Cma to Cmin ratio less than 200 and lag time less than 0.2.
  17. 18. The method of claim 17, wherein the small molecule drug is delivered systemically in a controlled manner over a release duration.
  18. 19. The method of claim 17, wherein the release duration is at least one week.
  19. 20. The method of claim 17, wherein the small molecule drug comprises an antipsychotic agent. 20
AU2013202598A 2005-09-30 2013-04-05 Sustained release small molecule drug formulation Ceased AU2013202598B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013202598A AU2013202598B2 (en) 2005-09-30 2013-04-05 Sustained release small molecule drug formulation

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/722,845 2005-09-30
AU2006299657A AU2006299657B2 (en) 2005-09-30 2006-09-28 Sustained release small molecule drug formulation
AU2013202598A AU2013202598B2 (en) 2005-09-30 2013-04-05 Sustained release small molecule drug formulation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2006299657A Division AU2006299657B2 (en) 2005-09-30 2006-09-28 Sustained release small molecule drug formulation

Publications (2)

Publication Number Publication Date
AU2013202598A1 true AU2013202598A1 (en) 2013-05-02
AU2013202598B2 AU2013202598B2 (en) 2016-06-09

Family

ID=48408505

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013202598A Ceased AU2013202598B2 (en) 2005-09-30 2013-04-05 Sustained release small molecule drug formulation

Country Status (1)

Country Link
AU (1) AU2013202598B2 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7833543B2 (en) * 1995-06-07 2010-11-16 Durect Corporation High viscosity liquid controlled delivery system and medical or surgical device
US6143314A (en) * 1998-10-28 2000-11-07 Atrix Laboratories, Inc. Controlled release liquid delivery compositions with low initial drug burst
US20050281879A1 (en) * 2003-11-14 2005-12-22 Guohua Chen Excipients in drug delivery vehicles

Also Published As

Publication number Publication date
AU2013202598B2 (en) 2016-06-09

Similar Documents

Publication Publication Date Title
US11110093B2 (en) Sustained release small molecule drug formulation
RU2355385C2 (en) Compositions of prolongedaction with controlled release
JP5078217B2 (en) Injectable depot compositions and their use
KR20050083605A (en) Injectable multimodal polymer depot compositions and uses thereof
KR20060125749A (en) Excipients in drug delivery vehicles
KR20060125748A (en) Excipients in drug delivery vehicles
JP2006503004A (en) Injectable depot composition and use thereof
US20080206303A1 (en) Prolonged Release Formulations Comprising Anastrozole
US20180250354A1 (en) COMPOSITIONS AND METHODS FOR LONG TERM RELEASE OF GONADOTROPIN-RELEASING HORMONE (GnRH) ANTAGONISTS
US20200352935A1 (en) Sustained release buprenorphine microspheres (srbm) and methods of use thereof
AU2013202598A1 (en) Sustained release small molecule drug formulation
EP2054037A2 (en) Subcutaneous implants releasing an active principle over an extended period of time
US20140335135A1 (en) Pharmaceutical composition

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired