Stable Ready-to-use Carmustine Pharmaceutical Composition

Pramanick; Sougata ;   et al.

Patent Application Summary

U.S. patent application number 17/268022 was filed with the patent office on 2021-10-21 for stable ready-to-use carmustine pharmaceutical composition. This patent application is currently assigned to EMCURE PHARMACEUTICALS LTD.. The applicant listed for this patent is EMCURE PHARMACEUTICALS LTD.. Invention is credited to Aasiya Aslam Burhan, Deepak Pragjibhai Gondaliya, Mukund Keshav Gurjar, Hiren Pravinbhai Patel, Sougata Pramanick.

Application Number20210322304 17/268022
Document ID /
Family ID1000005737401
Filed Date2021-10-21

United States Patent Application 20210322304
Kind Code A1
Pramanick; Sougata ;   et al. October 21, 2021

STABLE READY-TO-USE CARMUSTINE PHARMACEUTICAL COMPOSITION

Abstract

The present invention relates to a ready-to-use solution of carmustine that does not require dissolution or dilution of the carmustine prior to addition to saline and dextrose parenteral solutions. In particular, the invention relates to a stable liquid pharmaceutical composition containing carmustine in the form of ready-to-use solution and method for preparing the same.


Inventors: Pramanick; Sougata; (Bhosari, IN) ; Burhan; Aasiya Aslam; (Bhosari, IN) ; Gurjar; Mukund Keshav; (Bhosari, IN) ; Patel; Hiren Pravinbhai; (Bhosari, IN) ; Gondaliya; Deepak Pragjibhai; (Bhosari, IN)
Applicant:
Name City State Country Type

EMCURE PHARMACEUTICALS LTD.

Pune

IN
Assignee: EMCURE PHARMACEUTICALS LTD.
Pune
IN

Family ID: 1000005737401
Appl. No.: 17/268022
Filed: September 3, 2019
PCT Filed: September 3, 2019
PCT NO: PCT/IB2019/057404
371 Date: February 11, 2021

Current U.S. Class: 1/1
Current CPC Class: A61K 9/0029 20130101; A61K 31/175 20130101; A61K 47/26 20130101
International Class: A61K 9/00 20060101 A61K009/00; A61K 31/175 20060101 A61K031/175; A61K 47/26 20060101 A61K047/26

Foreign Application Data

Date Code Application Number
Sep 5, 2018 IN 201821033221

Claims



1. A liquid, ready-to-use parenteral pharmaceutical composition of carmustine dissolved in polysorbate as the sole solvent, wherein the pharmaceutical composition has a concentration of about 2 mg/mL to about 500 mg/mL of carmustine and wherein the liquid ready-to-use parenteral pharmaceutical composition is water free.

2. The pharmaceutical composition according to claim 1, wherein the polysorbate is selected from polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 and mixtures thereof.

3. The pharmaceutical composition according to claim 1, wherein the polysorbate is super refined polysorbate 80 with a peroxide value below 10 meq 02/kg.

4. The pharmaceutical composition according to claim 1, wherein the pharmaceutical composition, after storage at 2-8.degree. C. for 3 months, contains at least 90% by weight of the initial amount of carmustine.

5. The pharmaceutical composition according to claim 1, wherein the pharmaceutical composition is admixed with 500 mL of 0.9% sodium chloride injection or 5% dextrose injection to form an administrable solution prior to administration to a patient.

6. The pharmaceutical composition according to claim 5, wherein the administrable solution has a concentration of about 0.2 mg/mL of carmustine.

7. A method for administering carmustine comprising: (a) adding the liquid, ready-to-use parenteral pharmaceutical composition of claim 1 to 500 mL of 0.9% sodium chloride solution for injection or 5% dextrose solution for injection to form an administrable solution, wherein the pharmaceutical composition consists of carmustine dissolved in polysorbate 80 as the sole solvent, wherein the pharmaceutical composition has concentration of about 100 mg/mL of carmustine and the pharmaceutical composition has been stored at 2 to 8.degree. C. in a sealed vial, and (b) parenterally administering the administrable solution to a patient in need thereof.

8. The pharmaceutical composition according to claim 3, wherein the polysorbate is super refined polysorbate 80 with a peroxide value about 0.5 meq 02/kg.

9. The pharmaceutical composition according to claim 3, wherein the polysorbate is super refined polysorbate 80 with a peroxide value about 0.2 meq 02/kg.

10. A liquid, ready-to-use parenteral pharmaceutical composition of carmustine dissolved in polysorbate as the sole solvent, wherein the pharmaceutical composition has a concentration of about 2 mg/mL to about 500 mg/mL of carmustine; the polysorbate is super refined polysorbate with a peroxide value below 10 meq 02/kg, the liquid ready-to-use parenteral pharmaceutical composition is water free, free of visible particles and is stored in a container that is free of polyvinyl chloride and free of di-2-ethylhexylphthalate.

11. The pharmaceutical composition according to claim 10, wherein the polysorbate is selected from polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 and mixtures thereof.

12. The pharmaceutical composition according to claim 10, wherein the polysorbate is super refined polysorbate with a peroxide value about 0.5 meq 02/kg.

13. The pharmaceutical composition according to claim 12, wherein the polysorbate is super refined polysorbate with a peroxide value about 0.2 meq 02/kg.

14. The pharmaceutical composition according to claim 10, wherein the pharmaceutical composition, after storage at 2-8.degree. C. for 3 months, contains at least 90% by weight of the initial amount of carmustine.

15. The pharmaceutical composition according to claim 10 wherein the composition is stored in a vial with a head space within the vial comprising not more than 6% oxygen.

16. The pharmaceutical composition according to claim 10, wherein the pharmaceutical composition is admixed with 500 mL of 0.9% sodium chloride injection or 5% dextrose injection to form an administrable solution prior to administration to a patient.

17. A method for administering carmustine comprising: (a) adding the liquid, ready-to-use parenteral pharmaceutical composition of claim 10 to 500 mL of 0.9% sodium chloride solution for injection or 5% dextrose solution for injection to form an administrable solution, wherein the pharmaceutical composition has concentration of about 100 mg/mL of carmustine and the pharmaceutical composition has been stored at 2 to 8.degree. C. in a sealed vial, and (b) parenterally administering the administrable solution to a patient in need thereof.

18. A liquid, ready-to-use parenteral pharmaceutical composition of carmustine dissolved in super refined polysorbate 80 with a peroxide value below 10 meq 02/kg as the sole solvent, wherein the pharmaceutical composition has a concentration of about 100 mg/mL, the liquid ready-to-use parenteral pharmaceutical composition is water free, free of visible particles and is stored in a container with a head space containing no more than 6% oxygen and wherein the container is free of polyvinyl chloride and free of di-2-ethylhexylphthalate.

19. The pharmaceutical composition as defined in claim 18 wherein the polysorbate 80 has a peroxide value about 0.2 meq 02/kg to about 0.5 meq 02/kg.
Description



[0001] The present application claims the benefit of Indian Patent Application No. 201821033221 Sep. 5, 2018, which is hereby incorporated by reference.

FIELD OF THE INVENTION

[0002] The present invention relates to a ready-to-use solution of carmustine that does not require dissolution or dilution of the carmustine prior to addition to saline and dextrose parenteral solutions. In particular, the invention relates to a stable liquid pharmaceutical composition containing carmustine in the form of ready-to-use solution and method for preparing the same.

BACKGROUND OF THE INVENTION

[0003] Carmustine (bischloroethyl nitrosurea also known as BCNU) is a nitrosurea drug for the treatment of brain cancers owing to its ability to cross the blood-brain barrier and excellent activity against brain tumours.

[0004] Carmustine chemically known as 1,3-bis(2-chloroethyl)-1-nitrosourea (shown below) alkylates DNA, RNA and interferes with its synthesis and functions. It also binds and modifies (carbamoylates) glutathione reductase, which consequently leads to cell death.

##STR00001##

[0005] Carmustine is highly soluble in alcohol and lipids. Carmustine, however, is poorly soluble in water and is unstable in many formulations. For instance, carmustine gets readily hydrolyzed in water at pH>6. The solubility and stability issues of carmustine have been discussed previously. See, for example, Levin et al., Selective Cancer Therapeutics, 1989, 5(1), 33-35.

[0006] Though the drug has poor oral bioavailability, following IV infusion, it is rapidly taken up by the tissues, and due to its high lipid solubility, it can cross the blood brain barrier. However, it is rapidly degraded, with no unmetabolized drug detectable after 15 minutes.

[0007] Carmustine is commercially available as a lyophilized 100 mg powder for injection under the trade name BiCNU.RTM. in single dose vials. See the March 2017 prescribing information for BiCNU.RTM., which is hereby incorporated by reference. Ethanol (dehydrated alcohol) (3 mL) is co-packaged with the drug product as a sterile diluent for reconstitution. To prepare the drug for administration, three preparation steps need to be performed. First, the lyophilized carmustine is reconstituted with the co-packed sterile dehydrated alcohol (3 mL) diluent. Second, the solution is further diluted with 27 mL of sterile water to form the reconstituted solution. Third, the reconstituted solution is further diluted with 5% Dextrose Injection, USP or Sodium Chloride Injection, USP (0.9% sodium chloride). This complicated preparation of carmustine solutions is time-consuming and can lead to errors in preparation and dosing.

[0008] The lyophilized formulation has several disadvantages including:

a) Extra handling, for example, due to the two step reconstitution; b) In some cases, complete dissolution of the lyophilized powder may require prolonged shaking because of solubilisation problems; c) Improper reconstitution of a lyophilized powder sometimes results in the formation of air-borne droplets ("blow-back"), which, in the case of a potent antitumor agent such as carmustine may be a health hazard to the personnel preparing the solution for injection; d) Risk of improper dosing of a patient due to dilution with the wrong quantity of diluents; and and e) The manufacturing of a lyophilized formulation is quite costly, since it not only requires capital investment for installation of a lyophilizer, but also its maintenance.

[0009] International patent application no. WO 2008/119260 discloses a pharmaceutical composition comprising a pharmaceutically effective amount of carmustine, Tween surfactant and a co-solvent selected from glycerol, polyethylene glycol, and a mixture thereof. The pharmaceutical composition may be in liquid form. Comparative example 2 of WO 2008/119260, which included carmustine, Tween 80, and 6 mL water for injection, was found to be less stable and failed to form a clear solution (turbidity) before freezing and freeze drying in 10 mL water for injection.

[0010] Indian patent application no. 1909/MUM/2015 discloses a ready-to-use injectable formulation which is free of surfactants.

[0011] There is an ongoing need for improved and simplified formulation of carmustine whose preparation and administration does not require either lyophilization or reconstitution. In particular, there is need for a stable, liquid, ready-to-use parenteral formulation of carmustine, which can be administered to a person in need thereof, without the hassles of reconstitution.

SUMMARY OF THE INVENTION

[0012] The present inventors while working on liquid, ready-to-use formulations of carmustine, surprisingly found that stable and clear solution of carmustine can be formulated with a suitable surfactant as the sole solvent. The present invention also provides a simple and cost-effective method of preparing a liquid, ready-to-use parenteral formulation of carmustine.

[0013] One embodiment of the invention is a liquid, ready-to-use parenteral pharmaceutical composition of carmustine.

[0014] Another embodiment is a liquid, ready-to-use parenteral pharmaceutical composition of carmustine dissolved in a suitable surfactant as the sole solvent.

[0015] The liquid, ready-to-use parenteral pharmaceutical composition of carmustine may have a concentration from about 2 mg/mL to about 500 mg/mL, preferably 100 mg/mL, 200 mg/mL or 300 mg/mL of carmustine.

[0016] The liquid, ready-to-use parenteral pharmaceutical composition of carmustine may be further admixed with 500 mL of 0.9% sodium chloride injection or 5% dextrose injection prior to actual clinical use.

[0017] One preferred embodiment is a liquid, ready-to-use parenteral pharmaceutical composition of 100 mg of carmustine dissolved in 1 mL of polysorbate (e.g., polysorbate 80) as the sole solvent. The pharmaceutical composition may be in a sealed vial. The polysorbate may be polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 or any combination of any of the foregoing. In a preferred embodiment, the polysorbate is polysorbate 80, such as super refined polysorbate 80 (and more preferably polysorbate 80 with a peroxide value below 10 meq O.sub.2/kg). In one embodiment, the peroxide value is about 0.5 meq O.sub.2/kg. In yet another embodiment the peroxide value is about 0.2 meq O.sub.2/kg.

[0018] Preferably, the pharmaceutical composition, after storage at 2-8.degree. C. for 3 months, contains at least 90% by weight of the initial amount of carmustine. In one embodiment, the pharmaceutical composition is admixed with 500 mL of 0.9% sodium chloride injection or 5% dextrose injection to form an administrable solution prior to administration to a patient.

[0019] Yet another embodiment is a process for the preparation of a liquid, ready-to-use parenteral formulation of carmustine comprising: [0020] a) dissolving carmustine in a sufficient quantity of a suitable surfactant, for example, to achieve a concentration of 100 mg/ml; [0021] b) aseptic filtration (e.g., with a sterile 0.22 micron filter) of solution obtained in step (a) to obtain a sterile product, and [0022] c) filling the solution obtained in step (b) into a suitable container/closure system. [0023] d) optionally, inert gas purging (nitrogen) can be carried out during any of the steps.

[0024] Another embodiment is a method of preparing an administrable solution of carmustine comprising diluting the ready-to-use parenteral formulation of carmustine with an aqueous 0.9% sodium chloride solution (preferably Sodium Chloride Injection, USP) or an aqueous 5% dextrose solution (preferably 5% Dextrose Injection, USP) to obtain the administrable solution.

[0025] In one embodiment, the ready-to-use parenteral formulation of carmustine is diluted with 500 mL of an aqueous 0.9% sodium chloride solution (preferably Sodium Chloride Injection, USP) or 500 mL of an aqueous 5% dextrose solution (preferably 5% Dextrose Injection, USP).

[0026] In one preferred embodiment, the administrable solution has a pH in the range of 4 to 7 and an osmolality in the range of 330-390 mOsmol/L.

[0027] Yet another embodiment is a method of administering carmustine comprising intravenously administering an administrable carmustine solution as described herein to a patient in need thereof. The administrable carmustine solution may be prepared as described herein.

[0028] Yet another embodiment is a method for administering carmustine comprising:

(a) adding a liquid, ready-to-use parenteral pharmaceutical composition of carmustine to 500 mL of 0.9% sodium chloride solution for injection or 5% dextrose solution for injection to form an administrable solution, wherein the pharmaceutical composition comprises, consists essentially of, or consists of 100 mg of carmustine dissolved in 1 mL of polysorbate 80 as the sole solvent, and (b) parenterally administering the administrable solution to a patient in need thereof.

[0029] In one embodiment, the ready-to-use pharmaceutical composition is stored at 2 to 8.degree. C. in a sealed vial (such as, for example, 3, 6, 9, or 12 months) prior to being added to 0.9% sodium chloride solution for injection or 5% dextrose solution for injection.

[0030] Yet another embodiment is a method of treating cancer in a patient in need thereof by intravenously administering an administrable carmustine solution as described herein to the patient. The administrable carmustine solution may be prepared as described herein. The patient may be suffering from brain tumors glioblastoma, brainstem glioma, medulloblastoma, astrocytoma, ependymoma, metastatic brain tumors, multiple myeloma, relapsed or refractory Hodgkin's lymphoma, or relapsed or refractory Non-Hodgkin's lymphomas.

DETAILED DESCRIPTION OF THE INVENTION

[0031] The present invention relates to a stable, liquid pharmaceutical composition containing carmustine in the form of a ready-to-use solution and method for preparing the same.

[0032] As used herein, a "ready-to-use" or "RTU" composition is a sterile, liquid, injectable composition that is stable and has not been reconstituted from a lyophilizate.

[0033] Pharmaceutical Composition

[0034] One embodiment is a liquid, ready-to-use parenteral composition of carmustine dissolved in a suitable surfactant as the sole solvent.

[0035] The concentration of carmustine in the liquid, ready-to-use parenteral composition may vary from about 2 mg/mL to about 500 mg/mL, preferably 100 mg/mL, 200 mg/mL or 300 mg/mL.

[0036] Suitable surfactants include, but are not limited to, sodium salts of fatty alcohol sulphates, partial fatty acid esters of polyhydroxyethylene sorbitan such as polyoxyethylene sorbitan monolaurate (e.g., polyoxyethylene (20) sorbitan monolaurate, which is referred to as polysorbate 20 or Tween.RTM. 20), polyoxyethylene sorbitan monopalmitate (e.g., polyoxyethylene (20) sorbitan monopalmitate, which is referred to as polysorbate 40 or Tween.RTM. 40), polyoxyethylene sorbitan monostearate (e.g., polyoxyethylene (20) sorbitan monosterate, which is referred to as polysorbate 60 or Tween.RTM. 60), and polyoxyethylene sorbitan monooleate (e.g., polyoxyethylene (20) sorbitan monooleate, which is referred to as polysorbate 80 or Tween.RTM. 80), polyoxyethylene sorbitan tristearate (e.g., polyoxyethylene (20) sorbitan tristearate, which is referred to as polysorbate 65 or Tween.RTM. 65), and polyoxyethylene sorbitan trioleate (e.g., polyoxyethylene (20) sorbitan trioleate, which is referred to as polysorbate 85 or Tween.RTM. 85), polyoxyethylene glycol esters, polyoxyethylene castor oil derivatives, Cremophor.RTM. EL (PEG-35 castor oil), Cremophor.RTM. RH40 (PEG-40 hydrogenated castor oil), polyoxyethylene 15 hydroxystearate, polyoxyethylene alkyl ethers (sold under the tradename Brij.RTM.); polyoxyethylene stearates (Myrj.RTM.), sorbitan derivatives, fatty acid esters of sorbitan, poloxamers (e.g., poloxamer 188, poloxamer 407, poloxamer 338, and poloxamer 184), poloxamine (e.g., poloxamine 304, poloxamine 904, and poloxamine 908), lecithin, an ethoxylated vegetable oil, vitamin E tocopherol propylene glycol succinate (vitamin E-TPGS), polyoxyethylene-polyoxypropylene block copolymers, incrocas 35, TPGS (D-.alpha.-tocopherol polyethylene glycol 1000 succinate), tyloxapol, sodium oleate, sodium deoxycholate and mixtures thereof.

[0037] In one preferred embodiment, the surfactant is selected from various grades of polysorbate, such as polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80 and mixtures thereof. More preferably, the surfactant is polysorbate (e.g., super refined polysorbate 80) with a peroxide value below 10 meq O.sub.2/kg. In one embodiment, the peroxide value is about 0.5 meq O.sub.2/kg. In yet another embodiment the peroxide value is about 0.2 meq O.sub.2/kg.

[0038] According to one preferred embodiment, 100 mg of carmustine is dissolved in a sufficient quantity of surfactant (up to 1 mL to make up the final volume). In one embodiment, the fill volumes of RTU liquid may be 1 mL or 3 mL.

[0039] The pharmaceutical composition may optionally include other optional pharmaceutical excipients such as antioxidants. Suitable antioxidants include, but are not limited to, acetone sodium bisulfite, argon, ascorbyl palmitate, ascorbic acid, sodium bisulfite, butylated hydroxy anisole (BHA), butylated hydroxy toluene (BHT), citric acid, cystein/cysteinate HCl, acetylcystein, dithionite sodium (Na hydrosulfite, Na sulfoxylate), gentisic acid, gentisic acid, ethanolamine, glutamate monosodium, glutathione, formaldehyde sulfoxylate sodium, metabisulfite sodium, metabisulfite potassium, methionine, monothioglycerol (thioglycerol), sulfite sodium, tocopherols alpha, alpha tocopherol hydrogen succinate, thioglycolate sodium, sodium formaldehyde sulfoxylate, thiourea, and any combination of any of the foregoing.

[0040] The concentration of antioxidant may range between 0.001 mg/mL to 5 mg/mL.

[0041] In one embodiment, the pharmaceutical composition, after storage at 2-8.degree. C. for 3, 6, 9, or 12 months, contains at least 90, 92, 94, 95, 96, 97, or 98% by weight of the initial amount of carmustine. In another embodiment, the pharmaceutical composition, after storage at 2-8.degree. C. for 3, 6, 9, or 12 months, contains at least 90% by weight of the initial amount of carmustine.

[0042] Process of Preparation for the RTU Composition

[0043] The liquid, ready-to-use parenteral composition of carmustine may be prepared by: [0044] a) dissolving carmustine in a sufficient quantity of a suitable surfactant (e.g., to achieve 100 mg/mL concentration), [0045] b) aseptic filtration (e.g., with a sterile 0.22 micron filter) of the carmustine solution obtained in step (a) to obtain a sterile product, and [0046] c) filling the solution obtained in step (b) into a suitable container/closure system

[0047] Optionally, purging inert gas (nitrogen) during any of the aforementioned steps.

[0048] The liquid, ready-to-use parenteral formulation of carmustine may be a clear, pale yellow and free from visible particles.

[0049] In one embodiment, the stable liquid, ready-to-use parenteral formulation of carmustine of present invention has a concentration of about 100 mg/mL of carmustine.

[0050] This liquid, ready-to-use parenteral formulation of carmustine can be filled in a suitable container/closure system, e.g., ampoules, vials, and prefilled syringe system. The ready-to-use solution may be stored in an amber type-I glass vial or polypropylene container (such as a polypropylene container which is polyvinyl chloride (PVC) free and di-2-ethylhexy phthalate (DEHP) free). These solutions are preferably not stored in a polyvinyl chloride container. In one embodiment, the head space of each vial contains no more than 6.0% by volume oxygen.

[0051] The liquid, ready-to-use parenteral formulation of carmustine may have a pH in the range of 4-7.

[0052] Prior to administration, the liquid, ready-to-use parenteral composition of carmustine may be further admixed with 0.9% sodium chloride injection (e.g., Sodium Chloride Injection, USP) or 5% dextrose injection (e.g., 5% Dextrose Injection, USP) to form an administrable solution. For instance, in one embodiment, the ready-to-use composition is further admixed with 500 mL of 0.9% sodium chloride injection or 5% dextrose injection.

[0053] The U.S. Pharmacopeia, USP 42-NF 37 (2019) is hereby incorporated by reference, including the entries for Sodium Chloride Injection, USP and 5% Dextrose Injection, USP.

[0054] The administrable solution may be a faint yellow colour with a pH in the range of 4 to 7 and osmolality in the range of 330-390 mOsmol/L.

[0055] The administrable solution may be stored in a glass or polypropylene container (such as a polypropylene container which is polyvinyl chloride (PVC) free and di-2-ethylhexy phthalate (DEHP) free). These solutions are preferably not stored in a polyvinyl chloride container.

[0056] The administrable carmustine solution can have a concentration of about 0.2 mg/mL of carmustine

[0057] As used herein, a "stable" composition means no aggregation observed when stored at conventional storage conditions like 2.degree. C. to 8.degree. C. (long term) for appropriate time and wherein the assay of carmustine is not less than 90% (based on 100% initial carmustine).

[0058] The carmustine content may be determined by methods known in the art, such as high performance liquid chromatography (HPLC method), and spectrophotometry (UV spectrophotometry). HPLC was used for performing the carmustine assay studies described herein.

[0059] Based on the results of table 2, it was concluded that the liquid, ready-to-use parenteral formulation of carmustine of the present invention, was stable for up to 3 months when stored at 2.degree. C.-8.degree. C.

[0060] Administration

[0061] The carmustine administrable solution may be administered by slow intravenous infusion over at least two hours. In one embodiment, the injected area is monitored during the administration. In another embodiment, the rate of administration of the intravenous infusion is no more than 1.66 mg/m.sup.2/min.

[0062] The carmustine administrable solution may be administered to a patient to treat brain tumors glioblastoma, brainstem glioma, medulloblastoma, astrocytoma, ependymoma, metastatic brain tumors, multiple myeloma, relapsed or refractory Hodgkin's lymphoma, or relapsed or refractory Non-Hodgkin's lymphomas.

[0063] In one embodiment, the carmustine administrable solution is administered to a patient as a single agent or in a combination therapy (such as with other chemotherapeutic agents) to treat (i) brain tumors glioblastoma, brainstem glioma, medulloblastoma, astrocytoma, ependymoma, or metastatic brain tumors, (ii) multiple myeloma in combination with prednisone, (iii) relapsed or refractory Hodgkin's lymphoma in combination with other approved drugs (such as chemotherapeutic agents), or (iv) relapsed or refractory Non-Hodgkin's lymphomas in combination with other approved drugs (such as chemotherapeutic agents).

[0064] The carmustine administrable solution may be administered as a single agent in previously untreated patients at a dose of 150 to 200 mg/m.sup.2 carmustine intravenously every 6 weeks. The carmustine administrable solution may be administered as a single dose or divided into daily injections such as 75 to 100 mg/m.sup.2 on two successive days. The dose may be lowered when the carmustine administrable solution is used with other myelosuppressive drugs or in patients in whom bone marrow reserve is depleted. The carmustine administrable solution may be administered for the duration according to the established regimen. In one embodiment, the patient is premedicated before each dose with antiemetics.

[0065] The dosing (after the initial dose) may be adjusted according to the hematologic response of the patient to the preceding dose. In one embodiment, the patient is dosed as follows:

TABLE-US-00001 Nadir After Prior Dose Percentage of Prior Leukocytes/mm.sup.3 Platelets/mm.sup.3 Dose to be Given >4000 >100,000 100% 3000-3999 75,000-99,999 100% 2000-2999 25,000-74,999 70% <2000 <25,000 50%

[0066] The hematologic toxicity can be delayed and cumulative. In one embodiment, the patient's blood counts are monitored weekly. In another embodiment, a repeat course of the carmustine administrable solution is not administered until circulating blood elements have returned to acceptable levels (platelets above 100 Gi/L, leukocytes above 4 Gi/L and absolute neutrophil count above 1 Gi/L). In yet another embodiment, the interval between courses is 6 weeks.

[0067] In yet another embodiment, renal function is evaluated prior to administration and/or periodically during treatment. In one embodiment, carmustine treatment is discontinued if the creatinine clearance is less than 10 mL/min. In another embodiment, carmustine is not administered to patients with compromised renal function. In yet another embodiment, transaminases and bilirubin are monitored periodically during treatment.

[0068] The following examples further illustrate the invention but should not be construed as in any way limiting its scope. In particular, the processing conditions are merely exemplary and can be varied by one of ordinary skill in the art.

[0069] All patents and other references cited herein are hereby incorporated by reference in their entireties

EXAMPLES

Example 1

TABLE-US-00002 [0070] TABLE 1 Composition of liquid, ready-to-use parenteral formulations of carmustine Composition Formulation 1 Carmustine 100 mg Polysorbate 80 NF q.s to 1 mL

[0071] (a) 100 mg of carmustine was dissolved in sufficient quantity (q.s. to 1 mL) of polysorbate 80 NF surfactant, under inert (nitrogen) gas purging. [0072] (b) The solution obtained in step (a) was aseptically filtered (sterile 0.22 micron filter) under inert (nitrogen) gas purging to obtain a sterile product. [0073] (c) The solution obtained in step (b) was filled into a sterile amber coloured type-I glass vial.

[0074] The stability of the formulation was tested after 3 months of storage at 2-8.degree. C. The results are provided in Table 2 below.

TABLE-US-00003 TABLE 2 Evaluation of liquid ready-to-use parenteral formulations of carmustine Stability data 3 months Test Initial (2.degree. C.-8.degree. C.) Description Clear pale yellow Clear pale yellow color solution color solution Assay 101.50% 97.21% Related substances Impurity A* 0.20% 1.80% Any unspecified impurity BLD** BLD** Total impurities 0.20% 1.80% *Impurity A refers to 1,3-bis(2-chloroethyl)urea **BLD: below limit of detection

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed