US20160297842A1 - Process for the Preparation of Dicycloplatin - Google Patents

Process for the Preparation of Dicycloplatin Download PDF

Info

Publication number
US20160297842A1
US20160297842A1 US15/097,071 US201615097071A US2016297842A1 US 20160297842 A1 US20160297842 A1 US 20160297842A1 US 201615097071 A US201615097071 A US 201615097071A US 2016297842 A1 US2016297842 A1 US 2016297842A1
Authority
US
United States
Prior art keywords
carboplatin
dicycloplatin
combining step
dicarboxylic acid
solvent
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
US15/097,071
Other versions
US9447130B1 (en
Inventor
Xiaozhong Liu
Jirui Xi
Ye Zhao
Liang Zhang
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.)
Medoc Pharmaceutical Co Ltd
Syn Nat Products Enterprise LLC
Original Assignee
Medoc Pharmaceutical Co Ltd
Syn Nat Products Enterprise LLC
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
Application filed by Medoc Pharmaceutical Co Ltd, Syn Nat Products Enterprise LLC filed Critical Medoc Pharmaceutical Co Ltd
Assigned to Syn-Nat Products Enterprise LLC, Medoc Pharmaceutical Co., Ltd. reassignment Syn-Nat Products Enterprise LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LIU, XIAOZHONG, XI, JIRUI, ZHANG, LIANG, ZHAO, YE
Application granted granted Critical
Publication of US9447130B1 publication Critical patent/US9447130B1/en
Publication of US20160297842A1 publication Critical patent/US20160297842A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F15/00Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic System
    • C07F15/0006Compounds containing elements of Groups 8, 9, 10 or 18 of the Periodic System compounds of the platinum group
    • C07F15/0086Platinum compounds
    • C07F15/0093Platinum compounds without a metal-carbon linkage
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/13Crystalline forms, e.g. polymorphs

Definitions

  • This invention relates to a new process for preparing an antitumor derivative of a double dicarboxylic acid diaminoplatin complex, named as dicycloplatin.
  • Cisplatin has been used widely in clinical medicine as an antitumor drug since an antitumor effect was discovered for cis-dichlorodiaminoplatin. Rosenberg et al. Nature, 1965, 205: 698; Nature, 1972, 222: 385. Although a drug of this type exhibits therapeutic effects in cancers such as genitourinary cancer, nasopharyngeal cancer, cephalocircular cancer and lung cancer, it is toxic and leads to severe side effects. Some undesirable effects, such as nephrotoxicity, neurotoxicity, ototoxicity, nausea, and vomiting, are all constraints to its dosage and long term use.
  • Carboplatin one of the second-generation antitumor drugs of platin analogues, has an antitumor spectrum similar to that of cisplatin and is susceptible to cross drug-resistance.
  • the therapeutic effect of carboplatin is slightly inferior to that of cisplatin.
  • the toxicity and associated side effects of carboplatin is significantly less than that of cisplatin, myelosuppression still exists, and cisplatin is not stable as an aqueous solution.
  • active studies in search of highly potent antitumor drugs from platin analogues with low toxicity and broad-spectrum effects have continued.
  • Dicycloplatin is a super molecule composed of carboplatin (CBP) and 1,1-cyclobutane dicarboxylate (CBDCA) joined together by hydrogen bonds.
  • CBP carboplatin
  • CBDCA 1,1-cyclobutane dicarboxylate
  • Preclinical studies have shown that DCP overcomes the problem of CBP instability in aqueous solution while maintaining CBP's anticancer effects.
  • Clinical evaluation in a Phase I dose-escalation study in patients with tumors showed that DCP was tolerated at doses ranging from 100 to 550 mg/m 2 and showed potential efficacy in Chinese cancer patients.
  • DCP showed favorable bioavailability and stability in vivo, with the recommended Phase II dosage for DCP-containing chemotherapy being 450 mg/m 2 .
  • DCP is currently being investigated as a monotherapy in several cancer types, such as prostatic carcinoma, and in combination with paclitaxel in a Phase II non-lung cancer study.
  • the chemical structure of DCP is shown as formula I:
  • DCP Dicycloplatin
  • the present invention provides a surprisingly discovered new process for preparing dicycloplatin with the benefits of easy operation, mild reactive conditions, short reaction times and high reproducibility.
  • the present invention provides a process for preparing dicycloplatin, comprising: suspending per unit carboplatin in a corresponding proportion of 1,1-cyclobutane dicarboxylic acid and solvent(s); followed by crystallizing or precipitating the dicycloplatin through stirring, evaporating the solvent(s), and/or cooling.
  • An aspect of the invention is a process for preparing dicycloplatin of formula (I),
  • the process further comprises cooling the precipitating solids before the separating step.
  • the process further comprises agitating the suspension after the combining step and before the separating step.
  • the solids consist essentially of dicycloplatin.
  • the solids consist of dicycloplatin.
  • the solvent used in the process is water.
  • the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:15 to about 1:50.
  • the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:1.15 to 1:2.
  • the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from 1:3 to about 1:50.
  • the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from 1:5 to 1:10.
  • the proportion of the weight of carboplatin in grams and the volume of the solvent in mL in the combining step ranges from about 1:2 to about 1:50.
  • the proportion of the weight of the carboplatin in grams and the volume of the solvent in mL in the combining step ranges from about 1:3 to about 1:5.
  • the dicycloplatin has an X-ray powder diffraction pattern substantially as depicted in FIG. 2 .
  • reaction temperature of the combining step ranges from 0° C. to 80° C.
  • reaction temperature of the combining step ranges from 10 to 25° C.
  • the solvent is an organic solvent selected from the group consisting of: tetrahydrofuran (THF), dichloromethane (DCM), isopropanol, ethanol, ethyl acetate (EtOAc), dimethylformamide (DMF), 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, methyl isobutyl ketone (MIBK), methyl tert-butyl ether (MTBE), 1,2-dichloro ethane and a mixture thereof.
  • THF tetrahydrofuran
  • DCM dichloromethane
  • EtOAc ethyl acetate
  • DMF dimethylformamide
  • 1,4-dixoane 1,4-dixoane
  • acetone toluene
  • isobutyl acetate isopropyl acetate
  • chloroform 2-methoxyethanol
  • the solvent is a mixture of water and an organic solvent selected from the group consisting of: THF, DCM, isopropanol, ethanol, EtOAc, DMF, 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, MIBK, MTBE, 1,2-dichloro ethane and a mixture thereof.
  • an organic solvent selected from the group consisting of: THF, DCM, isopropanol, ethanol, EtOAc, DMF, 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, MIBK, MTBE, 1,2-dichloro ethane and a mixture thereof.
  • reaction temperature of the combining step ranges from 0° C. to 80° C.
  • reaction temperature of the combining step ranges from 20 to 35° C.
  • the final product does not contain carboplatin in an amount detectable with X-ray Powder Diffraction (XRPD).
  • XRPD X-ray Powder Diffraction
  • the final product does not contain carboplatin in an amount detectable with Differential Scanning Calorimetry (DSC).
  • DSC Differential Scanning Calorimetry
  • the final product does not contain carboplatin in an amount detectable with either XRPD or DSC.
  • FIG. 1 shows a representative 1 H-NMR spectrum of the product, where the product contains dicycloplatin and is dissolved in DMSO-d 6 .
  • FIG. 2 shows a representative X-ray powder diffraction (XRPD) pattern of the product, where the product contains dicycloplatin (bottom pattern) and is compared to a standard XRPD pattern of dicycloplatin (top pattern).
  • XRPD X-ray powder diffraction
  • FIG. 3 shows a representative XRPD pattern of the product, where the product contains dicycloplatin (bottom pattern) and is compared to an XRPD pattern of carboplatin (top pattern).
  • FIG. 4 shows a representative differential scanning calorimetric (DSC) thermogram of the product, where the product contains dicycloplatin (bottom pattern) and is compared to a DSC thermogram of 1,1-cyclobutane dicarboxylate (top pattern).
  • DSC differential scanning calorimetric
  • FIG. 5 shows a representative thermal gravimetric analysis (TGA) thermogram of the product, where the product contains dicycloplatin (bottom pattern).
  • FIG. 6 shows representative XRPD patterns of the product and the patterns are compared to standard dicycloplatin and carboplatin patterns.
  • FIG. 7 shows a representative XRPD pattern of the product, where the product contains dicycloplatin and is compared to a standard XRPD pattern of dicycloplatin and carboplatin.
  • the present invention provides a new process for preparing dicycloplatin this is easy to operate, involves mild reactive conditions and has a short reaction time.
  • the new process is reproducible and readily scaled up for industrial application.
  • only water is used as a solvent (i.e., no organic solvent).
  • only one organic solvent or a mixture of organic solvents is used.
  • a mixture of organic solvent and water is used.
  • the present invention provides a process for producing dicycloplatin, comprising: suspending carboplatin in corresponding proportion of 1,1-cyclobutane dicarboxylic acid and a solvent to form a suspension; stirring the suspension to form precipitating solids; and drying the solids, wherein the solids comprise dicycloplatin of formula I and does not contain carboplatin.
  • the process further comprises cooling the precipitating solids. In some embodiments, the process further comprises filtering the precipitating solids. In some embodiments, the process further comprises re-crystallizing the solids.
  • the solvent is water.
  • the molar ratio of carboplatin and 1,1-cyclobutane dicarboxylic acid is lower than 1:1. In some embodiments, the molar ratio of carboplatin and 1,1-cyclobutane dicarboxylic acid is lower than 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5, 1:5, 1:5.5, 1:6, 1:6.5, 1:7, 1:7.5, 1:8, 1:8.5, 1:9, 1:9.5, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, or 1:50.
  • the molar ratio of carboplatin and 1,1-cyclobutane dicarboxylic acid ranges from about 1:1.15 to about 1:2, from about 1:1.15 to about 1:3, from about 1:1.15 to about 1:4, from about 1:1.15 to about 1:5, from about 1:1.15 to about 1:6, from about 1:1.15 to about 1:7, from about 1:1.15 to about 1:8, from about 1:1.15 to about 1:9, from about 1:1.15 to about 1:10, from about 1:1.15 to about 1:15, from about 1:1.15 to about 1:20, from about 1:1.15 to about 1:25, from about 1:1.15 to about 1:30, from about 1:1.15 to about 1:40, from about 1:1.15 to about 1:50.
  • the proportion of carboplatin (weight, g) and solvent (volume, mL) ranges from about 1:2 to about 1:50. In some specific embodiments, the proportion of carboplatin (weight, g) and solvent (volume, mL) ranges from about 1:3 to about 1:5.
  • the final dicycloplatin product does not contain carboplatin in an amount of more than 5%, 3%, 1%, 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, or 0.001% of the final product by weight.
  • the final dicycloplatin product is at least 95%, 96%, 97%, 98%, 99% or 99.9% pure.
  • the reactions are typically carried out under mild temperature. However, the reactions can also be processed under high temperatures or low temperatures. The reactions are typically carried out under normal pressure. However, the reactions can also be processed under increased pressure or decreased pressure.
  • organic solvents may be used as the solvent to produce the desired products.
  • exemplary organic solvents include, but are not limited to, THF, DCM, isopropanol, ethanol, EtOAc, DMF, 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, MIBK, MTBE, 1,2-dichloro ethane and mixtures of two or more solvents, or mixtures of water and organic solvents.
  • the organic solvent is DCM.
  • Analytical Instrument Panalytical Empyrean.
  • the X-ray powder diffraction was conducted by mounting a sample of the crystalline material on a Si single crystal low-background holder and spreading out the sample into a thin layer with the aid of a microscope slide.
  • the 2-theta (2- ⁇ ) position was calibrated against a Panalytical 640 Si powder standard.
  • the collimated X-ray source was passed through a programmed divergence slit set at 10 mm and the reflected radiation directed through a 5.5 mm anti-scatter slit.
  • the sample was exposed for 16.3 seconds per 0.013° 2-theta increment (continuous scan mode) over the range of 3 degrees to 40 degrees 2-theta in theta-theta mode.
  • the running time was 3 minutes and 57 seconds.
  • the instrument was equipped with a RTMS detector (X'Celerator). Control and data capture was by means of a Dell Optiplex 780 XP operating with data collector software.
  • the relative intensities of peaks can be affected by, for example, grains above 30 microns in size and that non-unitary aspect ratios may affect analysis of samples. It is also known that the position of reflections can be affected by the precise height at which the sample sits in the diffractometer and the zero calibration of the diffractometer.
  • the surface planarity of the sample may also have a limited effect.
  • the diffraction pattern data presented are not intended to be limited to the absolute values.
  • DSC thermoanalytical method to measure the difference in the amount of heat required to increase the temperature of a sample and reference was measured as a function of temperature.
  • the general process of DSC is known and the specific instruments and conditions employed in the described Examples were as follows:
  • Heating rate 10° C. per minute
  • Purge gas nitrogen.
  • TGA was used to measure changes in physical and chemical properties of samples as a function of increasing temperature (with a constant heating rate), or as a function of time (with constant temperature and/or constant mass loss).
  • the general process of TGA is known and the specific instruments and conditions in the described Examples were as follows:
  • Heating rate 10° C. per minute
  • Purge gas nitrogen.
  • the product was analyzed by XRPD, DSC, TGA and 1 H NMR.
  • the product was analyzed by XRPD, DSC, TGA and 1 H NMR.
  • the product was analyzed by XRPD, DSC, TGA and 1 H NMR.
  • the product was analyzed by XRPD, DSC, TGA and 1 H NMR.
  • the product was analyzed by XRPD, DSC, TGA and 1 H NMR.
  • the product was analyzed by XRPD, DSC, TGA and 1 H NMR.
  • FIG. 1 shows a representative 1 H NMR spectrum of a product obtained in Example 1.
  • the 1 H NMR spectrum indicates the product contained dicycloplatin and that the molar ratio of the two components (1,1-cyclobutane dicarboxylic acid and carboplatin) in dicycloplatin is 1:1.
  • FIG. 2 shows the representative XRPD pattern of a product obtained in Examples 1 and 2 compared to a standard XRPD pattern of dicycloplatin (based on Yang et al., Science China Chemistry, 40(5): 485-491 (2010)), indicating that the sample was dicycloplatin.
  • FIG. 3 shows a representative XRPD pattern of a product obtained in Example 5 compared to a standard XRPD pattern of carboplatin, indicating that there was no traceable carboplatin in the sample obtained in the above Examples.
  • FIG. 4 shows a representative DSC thermogram of dicycloplatin obtained in Example 5, showing that there was no evidence of 1,1-cyclobutane dicarboxylic acid (i.e., no signal of the melting point of 1,1-cyclobutane dicarboxylic at 159.7° C.) present in the products obtained in the above Examples.
  • thermograms from Examples 1-6 arc also highly similar.
  • FIG. 5 shows a representative TGA thermogram of a product obtained in Example 5.
  • the dicycloplatin has a TGA thermogram comprising about 0.5% weight loss up to 150° C.
  • the product was analyzed by XRPD, DSC and TGA.
  • the product was analyzed by XRPD, DSC and TGA.
  • the product was analyzed by XRPD, DSC and TGA.
  • Examples 7-9 represent experiments conducted according to the reported process in U.S. Pat. No. 6,699,901.
  • carboplatin was dissolved in water and cyclobutane dicarboxylic acid was added to the solution while stirring.
  • the molar ratio of the carboplatin and the cyclobutane dicarboxylic acid was 1:1.
  • the mixture was completely dissolved and the solution was evaporated to produce a dried product.
  • the XRPD pattern results for the product are shown in FIG. 6 .
  • the XRPD results arc shown in FIG. 6 .
  • the final product contained not only dicycloplatin, but also a significant amount of carboplatin, demonstrated by the XRPD patterns in FIG. 6 , where the product XRPD patterns illustrate similar peaks compared to the standard carboplatin XRPD.
  • carboplatin proved difficult to remove, rendering the entire product unusable.
  • the high level of carboplatin present in the final product may be due to several reasons. Using a 1:1 molar ratio may be one reason that carboplatin are still present.
  • FIG. 7 shows a representative XRPD pattern for the product obtained according to the process of Example 10. The results indicate that the product contained dicycloplatin, with less than traceable amounts of carboplatin.
  • Table 10 shows the initial product after precipitation. 1,1-cyclobutane carboxylic acid can be removed easily but carboplatin cannot be removed. Therefore, to obtain pure dicycloplatin, co-precipitation of carboplatin should be prevented.

Abstract

The present invention relates to a process to prepare dicycloplatin under mild reactive conditions and at fast reaction rates. The process is reproducible and easy to scale up for industrial application.

Description

    FIELD OF THE INVENTION
  • This invention relates to a new process for preparing an antitumor derivative of a double dicarboxylic acid diaminoplatin complex, named as dicycloplatin.
  • BACKGROUND OF THE INVENTION
  • Cisplatin has been used widely in clinical medicine as an antitumor drug since an antitumor effect was discovered for cis-dichlorodiaminoplatin. Rosenberg et al. Nature, 1965, 205: 698; Nature, 1972, 222: 385. Although a drug of this type exhibits therapeutic effects in cancers such as genitourinary cancer, nasopharyngeal cancer, cephalocircular cancer and lung cancer, it is toxic and leads to severe side effects. Some undesirable effects, such as nephrotoxicity, neurotoxicity, ototoxicity, nausea, and vomiting, are all constraints to its dosage and long term use. Carboplatin, one of the second-generation antitumor drugs of platin analogues, has an antitumor spectrum similar to that of cisplatin and is susceptible to cross drug-resistance. The therapeutic effect of carboplatin is slightly inferior to that of cisplatin. Although the toxicity and associated side effects of carboplatin is significantly less than that of cisplatin, myelosuppression still exists, and cisplatin is not stable as an aqueous solution. As a result, active studies in search of highly potent antitumor drugs from platin analogues with low toxicity and broad-spectrum effects have continued.
  • Dicycloplatin (DCP) is a super molecule composed of carboplatin (CBP) and 1,1-cyclobutane dicarboxylate (CBDCA) joined together by hydrogen bonds. The solubility and stability of platinum complexes have a direct bearing on their activity, toxicity and pharmacokinetics. Preclinical studies have shown that DCP overcomes the problem of CBP instability in aqueous solution while maintaining CBP's anticancer effects. Clinical evaluation in a Phase I dose-escalation study in patients with tumors showed that DCP was tolerated at doses ranging from 100 to 550 mg/m2 and showed potential efficacy in Chinese cancer patients. DCP showed favorable bioavailability and stability in vivo, with the recommended Phase II dosage for DCP-containing chemotherapy being 450 mg/m2. DCP is currently being investigated as a monotherapy in several cancer types, such as prostatic carcinoma, and in combination with paclitaxel in a Phase II non-lung cancer study. The chemical structure of DCP is shown as formula I:
  • Figure US20160297842A1-20161013-C00001
  • Dicycloplatin (DCP) was first reported in U.S. Pat. No. 6,699,901, which disclosed the chemical structure and process for preparation of DCP.
  • The crystal structure of DCP has been determined and reported in “Structural studies of dicycloplatin-an antitumor supramolecule.” Yang X. et al., Science China Chemistry, 40(5): 485-491 (2010), which shows dicycloplatin as a co-crystal composed of carboplatin and 1,1-cyclobutane dicarboxylate.
  • However, the processes disclosed in U.S. Pat. No. 6,699,901, which were tested by the inventors of the present invention, cannot produce satisfying results and are difficult to scale up for industrial-scale production. The preparation of dicycloplatin according to the method of U.S. Pat. No. 6,699,901 produced a mixture of carboplatin and dicycloplatin, resulting in high toxicity and unacceptable products.
  • Therefore, it is desirable to provide robust and easy-to-scale-up crystallization processes to efficiently obtain pure dicycloplatin in high yield.
  • SUMMARY OF THE INVENTION
  • The present invention provides a surprisingly discovered new process for preparing dicycloplatin with the benefits of easy operation, mild reactive conditions, short reaction times and high reproducibility.
  • Accordingly, the present invention provides a process for preparing dicycloplatin, comprising: suspending per unit carboplatin in a corresponding proportion of 1,1-cyclobutane dicarboxylic acid and solvent(s); followed by crystallizing or precipitating the dicycloplatin through stirring, evaporating the solvent(s), and/or cooling.
  • An aspect of the invention is a process for preparing dicycloplatin of formula (I),
  • Figure US20160297842A1-20161013-C00002
  • comprising: combining carboplatin with a corresponding proportion of 1,1-cyclobutane dicarboxylic acid and a solvent to form a suspension; separating formed precipitated solids from the suspension; and drying the solids, wherein the solids comprise the dicycloplatin of formula (I) without any amount of carboplatin detectable by XRPD.
  • In an embodiment, the process further comprises cooling the precipitating solids before the separating step.
  • In an embodiment, the process further comprises agitating the suspension after the combining step and before the separating step.
  • In an embodiment, the solids consist essentially of dicycloplatin.
  • In an embodiment, the solids consist of dicycloplatin.
  • In an embodiment, the solvent used in the process is water.
  • In an embodiment, the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:15 to about 1:50.
  • In an embodiment, the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:1.15 to 1:2.
  • In an embodiment, the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from 1:3 to about 1:50.
  • In an embodiment, the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from 1:5 to 1:10.
  • In an embodiment, the proportion of the weight of carboplatin in grams and the volume of the solvent in mL in the combining step ranges from about 1:2 to about 1:50.
  • In an embodiment, the proportion of the weight of the carboplatin in grams and the volume of the solvent in mL in the combining step ranges from about 1:3 to about 1:5.
  • In an embodiment, the dicycloplatin has an X-ray powder diffraction pattern substantially as depicted in FIG. 2.
  • In an embodiment, the reaction temperature of the combining step ranges from 0° C. to 80° C.
  • In an embodiment, the reaction temperature of the combining step ranges from 10 to 25° C.
  • In an embodiment, the solvent is an organic solvent selected from the group consisting of: tetrahydrofuran (THF), dichloromethane (DCM), isopropanol, ethanol, ethyl acetate (EtOAc), dimethylformamide (DMF), 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, methyl isobutyl ketone (MIBK), methyl tert-butyl ether (MTBE), 1,2-dichloro ethane and a mixture thereof.
  • In an embodiment, the solvent is a mixture of water and an organic solvent selected from the group consisting of: THF, DCM, isopropanol, ethanol, EtOAc, DMF, 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, MIBK, MTBE, 1,2-dichloro ethane and a mixture thereof.
  • In an embodiment, the reaction temperature of the combining step ranges from 0° C. to 80° C.
  • In an embodiment, the reaction temperature of the combining step ranges from 20 to 35° C.
  • In some embodiments, the final product does not contain carboplatin in an amount detectable with X-ray Powder Diffraction (XRPD).
  • In some embodiments, the final product does not contain carboplatin in an amount detectable with Differential Scanning Calorimetry (DSC).
  • In some embodiments, the final product does not contain carboplatin in an amount detectable with either XRPD or DSC.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The following figures find a basis in Examples 7-9 as described herein and are employed as merely illustrative of particular embodiments of the present invention, and with no intention of limiting the scope of the invention.
  • FIG. 1 shows a representative 1H-NMR spectrum of the product, where the product contains dicycloplatin and is dissolved in DMSO-d6.
  • FIG. 2 shows a representative X-ray powder diffraction (XRPD) pattern of the product, where the product contains dicycloplatin (bottom pattern) and is compared to a standard XRPD pattern of dicycloplatin (top pattern).
  • FIG. 3 shows a representative XRPD pattern of the product, where the product contains dicycloplatin (bottom pattern) and is compared to an XRPD pattern of carboplatin (top pattern).
  • FIG. 4 shows a representative differential scanning calorimetric (DSC) thermogram of the product, where the product contains dicycloplatin (bottom pattern) and is compared to a DSC thermogram of 1,1-cyclobutane dicarboxylate (top pattern).
  • FIG. 5 shows a representative thermal gravimetric analysis (TGA) thermogram of the product, where the product contains dicycloplatin (bottom pattern).
  • FIG. 6 shows representative XRPD patterns of the product and the patterns are compared to standard dicycloplatin and carboplatin patterns.
  • FIG. 7 shows a representative XRPD pattern of the product, where the product contains dicycloplatin and is compared to a standard XRPD pattern of dicycloplatin and carboplatin.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The following description of certain embodiment(s) is merely exemplary in nature and is in no way intended to limit the invention, its application, or uses. As used throughout, ranges are used as shorthand for describing each and every value that is within the range. Any value within the range can be selected as the terminus of the range. In addition, all references cited herein are hereby incorporated by reference in their entireties. In the event of a conflict in a definition in the present disclosure and that of a cited reference, the present disclosure controls.
  • The present invention provides a new process for preparing dicycloplatin this is easy to operate, involves mild reactive conditions and has a short reaction time. The new process is reproducible and readily scaled up for industrial application. In some embodiments, only water is used as a solvent (i.e., no organic solvent). In some embodiments, only one organic solvent or a mixture of organic solvents is used. In some embodiments, a mixture of organic solvent and water is used.
  • In an exemplary embodiment, the present invention provides a process for producing dicycloplatin, comprising: suspending carboplatin in corresponding proportion of 1,1-cyclobutane dicarboxylic acid and a solvent to form a suspension; stirring the suspension to form precipitating solids; and drying the solids, wherein the solids comprise dicycloplatin of formula I and does not contain carboplatin.
  • In some embodiments, the process further comprises cooling the precipitating solids. In some embodiments, the process further comprises filtering the precipitating solids. In some embodiments, the process further comprises re-crystallizing the solids.
  • In some embodiments, the solvent is water.
  • In some embodiments, the molar ratio of carboplatin and 1,1-cyclobutane dicarboxylic acid is lower than 1:1. In some embodiments, the molar ratio of carboplatin and 1,1-cyclobutane dicarboxylic acid is lower than 1:1.5, 1:2, 1:2.5, 1:3, 1:3.5, 1:4, 1:4.5, 1:5, 1:5.5, 1:6, 1:6.5, 1:7, 1:7.5, 1:8, 1:8.5, 1:9, 1:9.5, 1:10, 1:11, 1:12, 1:13, 1:14, 1:15, 1:20, 1:25, 1:30, 1:35, 1:40, 1:45, or 1:50.
  • In some specific embodiments, the molar ratio of carboplatin and 1,1-cyclobutane dicarboxylic acid ranges from about 1:1.15 to about 1:2, from about 1:1.15 to about 1:3, from about 1:1.15 to about 1:4, from about 1:1.15 to about 1:5, from about 1:1.15 to about 1:6, from about 1:1.15 to about 1:7, from about 1:1.15 to about 1:8, from about 1:1.15 to about 1:9, from about 1:1.15 to about 1:10, from about 1:1.15 to about 1:15, from about 1:1.15 to about 1:20, from about 1:1.15 to about 1:25, from about 1:1.15 to about 1:30, from about 1:1.15 to about 1:40, from about 1:1.15 to about 1:50.
  • In some embodiments, the proportion of carboplatin (weight, g) and solvent (volume, mL) ranges from about 1:2 to about 1:50. In some specific embodiments, the proportion of carboplatin (weight, g) and solvent (volume, mL) ranges from about 1:3 to about 1:5.
  • In some embodiments, the final dicycloplatin product does not contain carboplatin in an amount of more than 5%, 3%, 1%, 0.5%, 0.1%, 0.05%, 0.01%, 0.005%, or 0.001% of the final product by weight.
  • In some embodiments, the final dicycloplatin product is at least 95%, 96%, 97%, 98%, 99% or 99.9% pure.
  • The reactions are typically carried out under mild temperature. However, the reactions can also be processed under high temperatures or low temperatures. The reactions are typically carried out under normal pressure. However, the reactions can also be processed under increased pressure or decreased pressure.
  • In some embodiments, organic solvents may be used as the solvent to produce the desired products. Exemplary organic solvents include, but are not limited to, THF, DCM, isopropanol, ethanol, EtOAc, DMF, 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, MIBK, MTBE, 1,2-dichloro ethane and mixtures of two or more solvents, or mixtures of water and organic solvents. In some specific embodiments, the organic solvent is DCM.
  • The following non-limiting examples further illustrate certain aspects of the present invention.
  • EXAMPLES X-Ray Powder Diffraction (XRPD)
  • Analytical Instrument: Panalytical Empyrean. The X-ray powder diffraction was conducted by mounting a sample of the crystalline material on a Si single crystal low-background holder and spreading out the sample into a thin layer with the aid of a microscope slide. The 2-theta (2-θ) position was calibrated against a Panalytical 640 Si powder standard. The sample was irradiated with X-rays generated by a copper long-fine focus tube operated at 45 kV and 40 mA with a wavelength of Kα1=1.540589 angstroms and Kα2=1.544426 angstroms (Kα2/Kα1 intensity ratio is 0.50). The collimated X-ray source was passed through a programmed divergence slit set at 10 mm and the reflected radiation directed through a 5.5 mm anti-scatter slit. The sample was exposed for 16.3 seconds per 0.013° 2-theta increment (continuous scan mode) over the range of 3 degrees to 40 degrees 2-theta in theta-theta mode. The running time was 3 minutes and 57 seconds. The instrument was equipped with a RTMS detector (X'Celerator). Control and data capture was by means of a Dell Optiplex 780 XP operating with data collector software.
  • It is known in the art that the relative intensities of peaks can be affected by, for example, grains above 30 microns in size and that non-unitary aspect ratios may affect analysis of samples. It is also known that the position of reflections can be affected by the precise height at which the sample sits in the diffractometer and the zero calibration of the diffractometer.
  • The surface planarity of the sample may also have a limited effect. As a result, the diffraction pattern data presented are not intended to be limited to the absolute values.
  • Differential Scanning Calorimetry (DSC)
  • DSC was used as a thermoanalytical method to measure the difference in the amount of heat required to increase the temperature of a sample and reference was measured as a function of temperature. The general process of DSC is known and the specific instruments and conditions employed in the described Examples were as follows:
  • Analytical Instrument: TA Instruments Q2000 DSC;
  • Heating rate: 10° C. per minute; and
  • Purge gas: nitrogen.
  • Thermal Gravimetric Analysis (TGA)
  • TGA was used to measure changes in physical and chemical properties of samples as a function of increasing temperature (with a constant heating rate), or as a function of time (with constant temperature and/or constant mass loss). The general process of TGA is known and the specific instruments and conditions in the described Examples were as follows:
  • Analytical Instrument: TA Instruments Q5000 TGA;
  • Heating rate: 10° C. per minute; and
  • Purge gas: nitrogen.
  • 1H Nuclear Magnetic Resonance (1H NMR)
  • The general process of 1H NMR spectroscopy is known and the specific instruments and conditions employed in the described Examples were as follows: solution NMR was collected on a Bruker 400M NMR Spectrometer using DMSO-d6.
  • Example 1 Preparation Procedures for Dicycloplatin
    • 1) Charged 5.0 g of carboplatin (13.47 mmol) and 2.232 g of 1,1-cyclobutane dicarboxylic acid (15.50 mmol) into a 20-mL glass vial.
    • 2) Added 12.5 mL of deionized water into the same vial and stirred the mixture at RT (room temperature) to form uniform suspension while keeping the vial away from light.
    • 3) Cooled the mixture to 5° C. at a rate of 0.33° C./min and aged at 5° C. for 20 hours to form a precipitation cake.
    • 4) Vacuum filtered and washed the cake with 5 mL of deionized water, and then vacuum dried the wet cake.
    • 5) Collected the solids from the dried cake and measured the amount of dicycloplatin.
    • 6) 6.49 g dicycloplatin was obtained with an overall yield of 93.2%.
  • The product was analyzed by XRPD, DSC, TGA and 1H NMR.
  • 1H NMR results: (400 MHz, DMSO-d6) δ 12.63 (s, 2H), 4.09 (s, 6H), 2.67 (t, J=7.9 Hz, 4H), 2.37 (t, J=8.0 Hz, 4H), 1.95-1.76 (m, 2H), 1.73-1.55 (m, 2H).
  • The XRPD data are listed in Table 1.
  • TABLE 1
    2-theta d-spacing intensity %
    7.5 11.8 100.0
    15.0 5.9 46.7
    16.4 5.4 6.0
    16.8 5.3 12.7
    18.5 4.8 9.8
    20.8 4.3 8.6
    21.1 4.2 5.1
    21.5 4.1 7.7
    22.0 4.0 10.7
    22.8 3.9 15.6
    24.0 3.7 16.6
    26.5 3.4 5.4
    29.7 3.0 5.5
    30.5 2.9 6.5
    33.2 2.7 4.6
  • Example 2 Preparation Procedures for Dicycloplatin
    • 1) Charged 1.0 g of carboplatin (2.69 mmol) and 776.4 mg of 1,1-cyclobutane dicarboxylic acid (5.39 mmol) into a 20-mL glass vial.
    • 2) Added 5 mL of deionized water into the same vial and stirred the suspension at RT for 6 hours while keeping the vial away from light to form a precipitation cake.
    • 3) Vacuum filtered and washed the cake with 2.8 mL of deionized water rapidly, and then vacuum dried the wet cake.
    • 4) Collected the solids from the dried cake and measured the amount of dicycloplatin.
  • The product was analyzed by XRPD, DSC, TGA and 1H NMR.
  • 1H NMR results: (400 MHz, DMSO-d6) δ 12.65 (s, 2H), 4.09 (s, 6H), 2.67 (t, J=7.9 Hz, 4H), 2.37 (t, J=8.0 Hz, 4H), 1.92-1.77 (m, 2H), 1.72-1.57 (m, 2H).
  • The XRPD data are listed in Table 2.
  • TABLE 2
    2-theta d-spacing intensity %
    7.6 11.7 100.0
    15.1 5.9 66.3
    16.5 5.4 11.8
    16.8 5.3 23.6
    18.5 4.8 17.1
    20.8 4.3 15.3
    21.5 4.1 12.1
    22.1 4.0 17.8
    22.8 3.9 33.8
    24.0 3.7 34.2
    26.5 3.4 12.3
    29.8 3.0 11.6
    30.6 2.9 18.6
    31.5 2.8 11.7
    33.3 2.7 11.8
  • Example 3 Preparation Procedures for Dicycloplatin
    • 1) Charged 5.0 g of carboplatin (13.47 mmol) and 2.523 g of 1,1-cyclobutane dicarboxylic acid (17.52 mmol) into a 100-mL glass vial.
    • 2) Added 25 mL of deionized water into the same vial and stirred the suspension at RT for 5 hours while keeping the vial away from light to form a precipitation cake.
    • 3) Vacuum filtered and washed the cake with 5 mL of deionized water rapidly, and then vacuum dried the wet cake.
    • 4) Collected the solids from the dried cake and measured the amount of dicycloplatin.
    • 5) 5.88 g of dicycloplatin was obtained with an overall yield of 84.2%.
  • The product was analyzed by XRPD, DSC, TGA and 1H NMR.
  • 1H NMR results: (400 MHz, DMSO-d6) δ 12.63 (s, 2H), 4.09 (s, 6H), 2.67 (t, J=7.9 Hz, 4H), 2.37 (t, J=8.0 Hz, 4H), 1.90-1.80 (m, 2H), 1.70-1.59 (m, 2H).
  • The XRPD data are listed in Table 3.
  • TABLE 3
    2-theta d-spacing intensity %
    7.5 11.8 100.0
    15.0 5.9 90.9
    16.4 5.4 13.8
    16.8 5.3 24.2
    18.5 4.8 22.9
    21.1 4.2 10.8
    22.0 4.0 11.4
    22.8 3.9 30.9
    24.0 3.7 37.4
    26.5 3.4 6.5
    29.7 3.0 8.8
    30.5 2.9 16.9
    31.4 2.8 8.4
    31.6 2.8 8.0
    33.2 2.7 10.6
  • Example 4 Preparation Procedures for Dicycloplatin
    • 1) Weighed 5.0 g of carboplatin (13.47 mmol) and 2.523 g of 1,1-cyclobutane dicarboxylic acid (17.52 mmol) into a 100-mL glass vial.
    • 2) Added 25 mL of deionized water into the same vial and stirred the mixture at RT to form a uniform suspension while keeping the vial away from light.
    • 3) Cooled the mixture to 5° C. at a rate of 0.33° C./min and aged at 5° C. for 19 hours to form a precipitation cake.
    • 4) Vacuum filtered and dried the wet cake.
    • 5) Collected the solids from the dried cake and measured the amount of dicycloplatin.
    • 6) 6.11 g of dicycloplatin was obtained with an overall yield of 87.1%.
  • The product was analyzed by XRPD, DSC, TGA and 1H NMR.
  • 1H NMR results: (400 MHz, DMSO-d6) δ 12.65 (s, 2H), 4.09 (s, 6H), 2.67 (t, J=7.9 Hz, 4H), 2.37 (t, J=8.0 Hz, 4H), 1.94-1.77 (m, 2H), 1.74-1.51 (m, 2H).
  • The XRPD data are listed in Table 4.
  • TABLE 4
    2-theta d-spacing intensity %
    7.6 11.7 100.0
    15.1 5.9 20.6
    16.4 5.4 2.9
    16.8 5.3 8.9
    18.5 4.8 4.7
    20.3 4.4 2.8
    20.8 4.3 4.0
    21.5 4.1 2.5
    22.1 4.0 4.5
    22.8 3.9 11.4
    24.0 3.7 9.6
    26.5 3.4 3.0
    29.8 3.0 2.4
    30.6 2.9 5.1
    31.5 2.8 2.7
  • Example 5 Preparation Procedures for Dicycloplatin
    • 1) Added 60.08 g of 1,1-cyclobutane dicarboxylic acid (417.22 mmol) and 593 mL of deionized water into a 1-L shaded crystallizer.
    • 2) Charged 118.6 g of carboplatin (319.46 mmol) into the same crystallizer and stirred the mixture for 6 hours at RT.
    • 3) Cooled the mixture to 5° C. at a rate of 0.33° C./min and aged at 5° C. for 19 hours to form a precipitation cake.
    • 4) Vacuum filtered and washed the cake with 120 mL of deionized water, and then vacuum dried the wet cake.
    • 5) Collected the solids from the dried cake and measured the amount of dicycloplatin.
    • 6) 148.52 g of dicycloplatin was obtained with overall yield of 90.2%.
  • The product was analyzed by XRPD, DSC, TGA and 1H NMR.
  • 1H NMR results: (400 MHz, DMSO-d6) δ 12.63 (s, 2H), 4.09 (s, 6H), 2.67 (t, J=7.9 Hz, 4H), 2.37 (t, J=8.0 Hz, 4H), 1.95-1.77 (m, 2H), 1.73-1.55 (m, 2H).
  • The XRPD data are listed in Table 5.
  • TABLE 5
    2-theta d-spacing intensity %
    7.5 11.8 100.0
    15.0 5.9 23.5
    16.4 5.4 3.1
    16.8 5.3 8.3
    18.5 4.8 4.7
    20.3 4.4 2.8
    21.1 4.2 2.4
    22.0 4.0 1.5
    22.8 3.9 17.4
    24.0 3.7 9.5
    26.5 3.4 1.0
    27.7 3.2 2.4
    30.6 2.9 7.3
    31.4 2.8 3.3
    39.2 2.3 2.5
  • Example 6 Preparation Procedures for Dicycloplatin
    • 1) Added 2.239 g of 1,1-cyclobutane dicarboxylic acid (15.55 mmol) and 15 mL of deionized water into a 20-mL glass vial.
    • 2) Charged 5.0 g of carboplatin (13.47 mmol) into the same vial and stirred the mixture at RT.
    • 3) Cooled the mixture to 5° C. at a rate of 0.33° C./min and aged at 5° C. for 19 hours to form a precipitation cake.
    • 4) Vacuum filtered and washed the cake with 5 mL of deionized water, and then vacuum dried the wet cake.
    • 5) Collected the solids and measured the amount of cycloplatin.
    • 6) 6.40 g of dicycloplatin was obtained with overall yield of 92.2%.
  • The product was analyzed by XRPD, DSC, TGA and 1H NMR.
  • 1H NMR results: (400 MHz, DMSO-d6) δ 12.63 (s, 2H), 4.09 (s, 6H), 2.67 (t, J=7.9 Hz, 4H), 2.37 (t, J=8.0 Hz, 4H), 1.93-1.75 (m, 2H), 1.74-1.51 (m, 2H).
  • The XRPD data are listed in Table 6.
  • TABLE 6
    2-theta d-spacing intensity %
    7.6 11.7 100.0
    15.2 5.8 30.9
    16.5 5.4 3.7
    16.9 5.2 9.8
    18.6 4.8 5.9
    20.4 4.3 3.6
    20.9 4.3 5.5
    22.1 4.0 5.6
    22.9 3.9 21.0
    24.1 3.7 12.4
    26.6 3.3 3.0
    27.8 3.2 3.4
    30.7 2.9 8.7
    31.6 2.8 4.5
    39.3 2.3 3.6

    Representative Results from Examples 1-6
  • The 1H NMR spectra from Examples 1-6 are highly similar. FIG. 1 shows a representative 1H NMR spectrum of a product obtained in Example 1. The 1H NMR spectrum indicates the product contained dicycloplatin and that the molar ratio of the two components (1,1-cyclobutane dicarboxylic acid and carboplatin) in dicycloplatin is 1:1.
  • The X-ray powder diffraction (XRPD) patterns from Examples 1-6 are also highly similar. FIG. 2 shows the representative XRPD pattern of a product obtained in Examples 1 and 2 compared to a standard XRPD pattern of dicycloplatin (based on Yang et al., Science China Chemistry, 40(5): 485-491 (2010)), indicating that the sample was dicycloplatin.
  • FIG. 3 shows a representative XRPD pattern of a product obtained in Example 5 compared to a standard XRPD pattern of carboplatin, indicating that there was no traceable carboplatin in the sample obtained in the above Examples.
  • The differential scanning calorimetry (DSC) thermograms from the Examples 1-6 are also highly similar. FIG. 4 shows a representative DSC thermogram of dicycloplatin obtained in Example 5, showing that there was no evidence of 1,1-cyclobutane dicarboxylic acid (i.e., no signal of the melting point of 1,1-cyclobutane dicarboxylic at 159.7° C.) present in the products obtained in the above Examples.
  • The representative thermal gravimetric analysis (TGA) thermograms from Examples 1-6 arc also highly similar. FIG. 5 shows a representative TGA thermogram of a product obtained in Example 5. The dicycloplatin has a TGA thermogram comprising about 0.5% weight loss up to 150° C.
  • Example 7 Preparation Procedures for Dicycloplatin
    • 1) Dissolved 50.0 mg of carboplatin (135 μmol) in 2.8 mL of water in a 3-mL glass vial at RT.
    • 2) Added 19.4 mg of 1,1-cyclobutane dicarboxylic acid (135 μmol) into the same vial.
    • 3) Stirred the suspension at RT to obtain a clear solution.
    • 4) Transferred the clear solution to a 25-mL round-bottom flask and rotary evaporated at a temperature between 50° C. and 55° C. to obtain white crystal solid.
    • 5) Air-dried the solid to obtain a product.
  • The product was analyzed by XRPD, DSC and TGA.
  • The XRPD data are listed in Table 7.
  • TABLE 7
    2-theta d-spacing intensity %
    7.6 11.6 100.0
    15.1 5.9 82.9
    16.5 5.4 23.4
    16.9 5.3 13.9
    18.6 4.8 44.9
    21.2 4.2 15.0
    22.9 3.9 16.5
    24.1 3.7 33.2
    24.1 3.7 15.8
    24.8 3.6 7.8
    30.6 2.9 10.3
    31.7 2.8 11.9
    33.3 2.7 11.7
    33.4 2.7 6.2
    37.6 2.4 9.2
  • Example 8 Preparation Procedures for Dicycloplatin
    • 1) Dissolved 15.1 mg of carboplatin (41 μmol) and 5.9 mg of 1,1-cyclobutane dicarboxylic acid (41 μmol) in 1.0 mL of water in a 3-mL glass vial.
    • 2) After filtering by 0.45 μm filter membrane, the filtrate was transferred to a 4-mL glass. Covered the vial with aluminum-foil paper to keep solution away from light and evaporated the solution freely at RT.
  • The product was analyzed by XRPD, DSC and TGA.
  • The XRPD data are listed in Table 8.
  • TABLE 8
    2-theta d-spacing intensity %
    7.5 11.8 100.0
    15.1 5.9 41.3
    16.8 5.3 18.0
    18.5 4.8 11.4
    20.8 4.3 11.1
    22.0 4.0 11.2
    22.8 3.9 34.8
    24.0 3.7 28.9
    26.5 3.4 10.5
    27.6 3.2 8.4
    30.6 2.9 19.4
    31.5 2.8 8.0
  • Example 9 Preparation Procedures for Dicycloplatin
    • 1) Charged 49.7 mg of carboplatin (134 μmol) in 2.8 mL of water in a 3-mL of vial and stirred the suspension to obtain a clear solution at RT.
    • 2) Added 19.4 mg of 1,1-cyclobutane dicarboxylic acid (135 μmol) into the same vial.
    • 3) Stirred the solution in a vial wrapped with aluminum-foil at RT.
    • 4) Solid was observed after slurry for about 15 min and isolated by centrifuging.
  • The product was analyzed by XRPD, DSC and TGA.
  • The XRPD data are listed in Table 9.
  • TABLE 9
    2-theta d-spacing intensity %
    7.6 11.6 23.4
    14.7 6.0 6.1
    15.0 5.9 14.2
    15.2 5.8 100.0
    15.4 5.8 4.6
    16.5 5.4 9.0
    22.9 3.9 5.9
    24.1 3.7 9.2
    24.2 3.7 4.4
    29.8 3.0 11.3
    29.9 3.0 6.0
    31.7 2.8 13.7
    31.8 2.8 7.3
    33.4 2.7 6.7
    33.5 2.7 4.0
  • Test According to a Reported Process.
  • Examples 7-9 represent experiments conducted according to the reported process in U.S. Pat. No. 6,699,901. In brief, carboplatin was dissolved in water and cyclobutane dicarboxylic acid was added to the solution while stirring. The molar ratio of the carboplatin and the cyclobutane dicarboxylic acid was 1:1. The mixture was completely dissolved and the solution was evaporated to produce a dried product. The XRPD pattern results for the product are shown in FIG. 6.
  • The XRPD results arc shown in FIG. 6. On scales of or higher than approximately 250 mg, the final product contained not only dicycloplatin, but also a significant amount of carboplatin, demonstrated by the XRPD patterns in FIG. 6, where the product XRPD patterns illustrate similar peaks compared to the standard carboplatin XRPD. Moreover, carboplatin proved difficult to remove, rendering the entire product unusable.
  • The high level of carboplatin present in the final product may be due to several reasons. Using a 1:1 molar ratio may be one reason that carboplatin are still present.
  • Since evaporation was required in the reported process, dicycloplatin was degraded in the product because it is sensitive to high temperature.
  • Moreover, in additional polymorph studies of dicycloplatin, it was recognized that the use of shorter aliphatic alcohols (as reported in U.S. Pat. No. 6,699,901) such as methanol, ethanol, isopropanol and butanol led to a significant amount of carboplatin being left in the product because of disproportionation. In addition, the water solubility of dicycloplatin is limited (40 mg/mL at room temperature), and it was observed that a slurry of dicycloplatin in hot water caused partial hydrolysis). Thus, recrystallization in water was not efficient and was difficult to scale up.
  • As indicated by the results of the current Examples, the reported process was not robust and was difficult to scale up.
  • Example 10 Preparation of Dicycloplatin Using Dichloromethane (DCM) as a Solvent
    • 1) Charged 500 mg of carboplatin (1.347 mmol) in 20 mL of DCM in a 50-mL of flask and stirred the suspension to obtain a clear solution at RT.
    • 2) Added 582 mg of 1,1-cyclobutane dicarboxylic acid (4.038 mmol) into the same flask.
    • 3) Stirred the solution in vial wrapped with aluminum-foil at 20-35° C.
    • 4) Stirred over 12 hours and then filtered, washed with DSM.
  • The product obtained by this example was analyzed by XRPD, DSC and TGA. FIG. 7 shows a representative XRPD pattern for the product obtained according to the process of Example 10. The results indicate that the product contained dicycloplatin, with less than traceable amounts of carboplatin.
  • Example 11 Preparation of Dicycloplatin Using Different Solvents
  • Different solvents, including organic solvents, were used for the preparation of dicycloplatin. A summary of the products is provided in Table 10.
  • TABLE 10
    mol ratio (A/B)
    solvent (v/v) 10:1 2:1 1:1 1:2 1:5 1:10
    EtOH/H2O (19:1) A A A A + C (A + B + C)
    ACN/H2O (9:1) A A A A (A + B + C)
    Acetone/H2O (19:1) A A A A (B + C)*
    1,4-dioxane/H2O A A A A (B + C)*
    (9:1)
    H2O A A (A + B + C) (B + C)* (B + C)* (B + C)*
    DCM A A A (A + B + C) (B + C)* (B + C)*
    Isopropanol A A A A + C (A + B + C)
    Isopropanol/H2O (A + B + C)
    (3:1)
    Toluene (B + C)*
    THF (A + B + C)
    Acetone (B + C)*
    EtOAc A A A A + B (B + C)*
    (1) A: carboplatin; B: 1,1-cyclobutane carboxylic acid; C: dicycloplatin
    (2) *indicates that substantially pure dicycloplatin was obtained in a significant quantity after further processing such as filtering, washing and drying.
  • Table 10 shows the initial product after precipitation. 1,1-cyclobutane carboxylic acid can be removed easily but carboplatin cannot be removed. Therefore, to obtain pure dicycloplatin, co-precipitation of carboplatin should be prevented.

Claims (20)

1. A process for preparing dicycloplatin of formula (I),
Figure US20160297842A1-20161013-C00003
comprising:
combining carboplatin with a corresponding proportion of 1,1-cyclobutane dicarboxylic acid and a solvent to form a suspension;
separating formed precipitated solids from the suspension; and
drying the solids,
wherein the solids comprise the dicycloplatin of formula (I) without a detectable amount of carboplatin present, and
wherein the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:1.15 to 1:50.
2. The process of claim 1, wherein the solids comprise dicycloplatin of formula (I) without any amount of carboplatin detectable by XRPD.
3. The process of claim 1, further comprising cooling the precipitating solids before the separating step.
4. The process of claim 1, further comprising agitating the suspension after the combining step and before the separating step.
5. The process of claim 1, wherein the solids consist essentially of dicycloplatin.
6. The process of claim 1, wherein the solids consist of dicycloplatin.
7. The process of claim 1, wherein the solvent is water.
8. The process of claim 1, wherein the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:15 to about 1:50.
9. The process of claim 1, wherein the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from about 1:1.15 to 1:2.
10. The process of claim 1, wherein the proportion of the weight of carboplatin in grams and the volume of the solvent in mL in the combining step ranges from about 1:2 to about 1:50.
11. The process of claim 1, wherein the proportion of the weight of the carboplatin in grams and the volume of the solvent in mL in the combining step ranges from about 1:3 to about 1:5.
12. The process of claim 1, wherein the dicycloplatin has an X-ray powder diffraction pattern substantially as depicted in FIG. 2.
13. The process of claim 1, wherein the reaction temperature of the combining step ranges from 0° C. to 80° C.
14. The process of claim 1, wherein the reaction temperature of the combining step ranges from 10 to 25° C.
15. The process of claim 1, wherein the solvent is an organic solvent selected from the group consisting of: tetrahydrofuran (THF), dichloromethane (DCM), isopropanol, ethanol, ethyl acetate (EtOAc), dimethylformamide (DMF), 1,4-dixoane, acetone, toluene, isobutyl acetate, isopropyl acetate, chloroform, 2-methoxyethanol, methyl isobutyl ketone (MIBK), methyl tert-butyl ether (MTBE), 1,2-dichloro ethane, and a mixture thereof.
16. The process of claim 1, wherein the solvent is a mixture of water and an organic solvent selected from the group consisting of: THF, DCM, isopropanol, ethanol, EtOAc, DMF, 1,4-dixoane, acetone, and a mixture thereof.
17. The process of claim 12, wherein the reaction temperature of the combining step ranges from 0° C. to 80° C.
18. The process of claim 12, wherein the reaction temperature of the combining step ranges from 20 to 35° C.
19. The process of claim 12, wherein the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from 1:3 to about 1:50.
20. The process of claim 12, wherein the molar ratio of the carboplatin and the 1,1-cyclobutane dicarboxylic acid in the combining step ranges from 1:5 to 1:10.
US15/097,071 2015-04-10 2016-04-12 Process for the preparation of dicycloplatin Active US9447130B1 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2015/025334 WO2016164040A1 (en) 2015-04-10 2015-04-10 Process for the preparation of dicycloplatin

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/025334 Continuation WO2016164040A1 (en) 2015-04-10 2015-04-10 Process for the preparation of dicycloplatin

Publications (2)

Publication Number Publication Date
US9447130B1 US9447130B1 (en) 2016-09-20
US20160297842A1 true US20160297842A1 (en) 2016-10-13

Family

ID=56895556

Family Applications (1)

Application Number Title Priority Date Filing Date
US15/097,071 Active US9447130B1 (en) 2015-04-10 2016-04-12 Process for the preparation of dicycloplatin

Country Status (14)

Country Link
US (1) US9447130B1 (en)
EP (1) EP3280408B1 (en)
JP (1) JP6925318B2 (en)
KR (1) KR102432526B1 (en)
CN (1) CN106132408B (en)
AU (1) AU2015390714B2 (en)
CA (1) CA2981158C (en)
DK (1) DK3280408T3 (en)
ES (1) ES2765648T3 (en)
IL (1) IL254894B (en)
MX (1) MX2017012985A (en)
RU (1) RU2676269C1 (en)
WO (1) WO2016164040A1 (en)
ZA (1) ZA201706795B (en)

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108697093A (en) 2015-06-19 2018-10-23 新纳特产品公司 Composition and application thereof containing carboplatin
CA2988992A1 (en) 2015-06-19 2016-12-22 Syn-Nat Products Enterprise LLC Pharmaceutical composition of carboplatin based co-crystals and use thereof
CN116854745A (en) * 2015-06-25 2023-10-10 新纳特产品公司 Pharmaceutical co-crystal composition and use thereof
CN106995467B (en) * 2017-05-03 2019-06-21 江苏大红鹰恒顺药业有限公司 A kind of method of microwave catalysis synthesis bicycloplatin
CN109053808B (en) * 2017-06-21 2020-12-29 宋勤华 Industrial preparation method of high-purity dicycloplatin needle crystal
CA3082295A1 (en) * 2017-11-21 2019-05-31 Medoncare Pharmaceutical Co., Ltd Combination products containing dicycloplatin, preparation and use thereof
CN108780053A (en) * 2018-02-09 2018-11-09 北京索普兴大医药研究有限公司 Bicycloplatin drug test method
WO2019161526A1 (en) * 2018-02-22 2019-08-29 昆明贵研药业有限公司 One-pot method for preparing twin dicarboxylic acid diamine complex platinum (ii) derivatives
WO2018171371A2 (en) * 2018-02-22 2018-09-27 昆明贵研药业有限公司 Preparation method for bis-dicarboxylic acid diammine platinum (ii) derivative
CN112262123B (en) * 2018-09-18 2023-12-15 昆明贵研药业有限公司 Method for purifying bis-dicarboxylic acid diammine platinum (II) derivative

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2067099C1 (en) * 1991-03-23 1996-09-27 Санкионг Индастриз Лтд. Platinum (ii) complex compounds, method of their synthesis, intermediate compounds and methods of their synthesis
US5455270A (en) * 1993-08-11 1995-10-03 Bristol-Myers Squibb Co. Stabilized solutions of platinum(II) antitumor agents
CN1121380C (en) * 2000-03-03 2003-09-17 北京兴大豪斯科技有限公司 Anti-tumor bis-dicarboxylic diamino platinum derivatives and its medicinal composition
WO2003045962A1 (en) * 2001-11-30 2003-06-05 Jingzun Wang Supermolecular carboplatinum derivatives, their preparation and pharmaceutical composition containing them as active ingredient and applications of the compositions
US20090082438A1 (en) * 2005-03-18 2009-03-26 Kazunori Kataoka Coordination Compound Composed of Diaminocyclohexane Platinum (II) and Block Copolymer and Anti-Cancer Agent Comprising the Same
WO2006103691A1 (en) * 2005-03-28 2006-10-05 Dabur Pharma Ltd. Stable pharmaceutical compositions of platinum (ii) antitumour agents
ZA200806748B (en) * 2006-01-30 2010-01-27 Platco Technologies Proprietar Preparation of platinum (II) complexes
AU2008264866A1 (en) * 2007-06-18 2008-12-24 Platco Technologies (Proprietary) Limited Platinum (IV) complexes

Also Published As

Publication number Publication date
KR102432526B1 (en) 2022-08-12
DK3280408T3 (en) 2020-02-03
BR112017021590A2 (en) 2018-07-03
US9447130B1 (en) 2016-09-20
IL254894B (en) 2021-02-28
CA2981158A1 (en) 2016-10-13
ZA201706795B (en) 2019-02-27
WO2016164040A1 (en) 2016-10-13
AU2015390714A1 (en) 2017-10-26
RU2676269C1 (en) 2018-12-27
JP2018510914A (en) 2018-04-19
JP6925318B2 (en) 2021-08-25
EP3280408A1 (en) 2018-02-14
MX2017012985A (en) 2018-05-22
CN106132408B (en) 2018-02-13
IL254894A0 (en) 2017-12-31
AU2015390714B2 (en) 2021-02-04
KR20170134737A (en) 2017-12-06
EP3280408A4 (en) 2018-10-17
ES2765648T3 (en) 2020-06-10
CA2981158C (en) 2022-05-24
CN106132408A (en) 2016-11-16
EP3280408B1 (en) 2019-10-23

Similar Documents

Publication Publication Date Title
US9447130B1 (en) Process for the preparation of dicycloplatin
JP2018533617A (en) Process for preparing amorphous ibrutinib and novel crystalline forms
Liu et al. sp2 CH⋯ Cl hydrogen bond in the conformational polymorphism of 4-chloro-phenylanthranilic acid
RU2648990C1 (en) Lobaplatin crystals, methods of production and applications in pharmaceuticals
JP7351061B2 (en) Salts and crystal forms of fused ring pyrimidine compounds and their production and use
CN114746412A (en) Novel crystal form of KD-025 and preparation method thereof
EP3322709B1 (en) Crystalline forms of (3r)-3-cyclopentyl-3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]propanenitrile salts and preparation thereof
Hu et al. Solution growth and thermal treatment of crystals lead to two new forms of 2-((2, 6-dimethylphenyl) amino) benzoic acid
CA2776361A1 (en) Crystalline forms of substituted pyrazolopyrimidines
Fernandes et al. Studies on polymorph conversion in a new cyclodextrin inclusion compound
CN105440082B (en) A kind of lobaplatin crystal, preparation method and medicinal application
Samie et al. Orientation-dependent conformational polymorphs in two similar pyridine/pyrazine phenolic esters
WO2017009316A1 (en) Crystalline forms of n-[(3-amino-3-oxetanyl)methyl]-2-(2,3-dihydro-1,1 -dioxido-1,4-benzothiazepin-4(5 h)-yl)-6-methyl-4-quinazolinamine for the treatment of respiratory syncytial virus (rsv) infections
CA3130247C (en) Fgfr inhibitor compound in solid form and preparation method therefor
KR101653521B1 (en) 1,2-cyclohexanediaminplatinum(ii)-bis-(4-methylbenzenesulfonate) and the hydrates thereof
CN105440083B (en) A kind of lobaplatin crystal, preparation method and medicinal application
AU2017329753A1 (en) Crystalline polymorphic form of 3-hydroxy-4,5-bis-benzyloxy-6-benzyloxymethyl-2-phenyl-2-oxo-2lambda5-(1,2)oxaphosphinane
CN110776450B (en) Siponimod crystal form and preparation method thereof
BR112017021590B1 (en) DICYCLOPLATIN PREPARATION PROCESS
US20170298087A1 (en) L-proline complex of sodium-glucose cotransporter 2 inhibitor, monohydrate and crystal form thereof
EP2771312B1 (en) Agomelatine-urea complex and crystalline forms thereof
WO2017032349A1 (en) Salts of 5-fluoro-3-phenyl-2-[(1s)-1-(9h-purin-6-ylamino)propyl]quinazolin-4-one and preparation thereof
US20140275102A1 (en) Solid State Forms of 6-[4-[3-((R)-2-Methylpyrrolidine-1-yl)-propoxy]phenyl] 2H-pyridazine-3-one Hydrochloride
CA3183361A1 (en) Succinate salts of octahydrothienoquinoline compound and crystals thereof
CN115246830A (en) Solid form of acid and preparation method thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: MEDOC PHARMACEUTICAL CO., LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIU, XIAOZHONG;XI, JIRUI;ZHAO, YE;AND OTHERS;REEL/FRAME:038904/0922

Effective date: 20150522

Owner name: SYN-NAT PRODUCTS ENTERPRISE LLC, NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:LIU, XIAOZHONG;XI, JIRUI;ZHAO, YE;AND OTHERS;REEL/FRAME:038904/0922

Effective date: 20150522

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4