US20230157960A1 - Onapristone extended-release compositions and methods - Google Patents

Onapristone extended-release compositions and methods Download PDF

Info

Publication number
US20230157960A1
US20230157960A1 US18/151,846 US202318151846A US2023157960A1 US 20230157960 A1 US20230157960 A1 US 20230157960A1 US 202318151846 A US202318151846 A US 202318151846A US 2023157960 A1 US2023157960 A1 US 2023157960A1
Authority
US
United States
Prior art keywords
onapristone
cancer
formulation
release
dose
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.)
Abandoned
Application number
US18/151,846
Inventor
Alexander Zukiwski
Stefan Proniuk
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.)
Context Biopharma Inc
Original Assignee
Context Biopharma Inc
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 Context Biopharma Inc filed Critical Context Biopharma Inc
Priority to US18/151,846 priority Critical patent/US20230157960A1/en
Assigned to CONTEXT BIOPHARMA INC. reassignment CONTEXT BIOPHARMA INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ARNO THERAPEUTICS, INC.
Assigned to ARNO THERAPEUTICS, INC. reassignment ARNO THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZUKIWSKI, Alexander, PRONIUK, Stefan
Publication of US20230157960A1 publication Critical patent/US20230157960A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Onapristone is an anti-progestin drug and progesterone receptor antagonist which was originally developed for potential contraceptive use and the use in benign gynecological disorders such as the treatment of uterine leiomyomas.
  • ONA binds to the progesterone receptor (PR), preventing the PR from binding to DNA and thereby inhibiting or eliminating PR-induced DNA transcription.
  • PR progesterone receptor
  • Klijn et al. Progesterone antagonists and progesterone receptor modulation in the treatment of breast cancer, Steroids, v. 65, pp. 825-830 (2000); Jonat et al., The clinical efficacy of progesterone antagonists in breast cancer, Endocrine Therapy of Breast Cancer, pp. 117-124.
  • Onapristone is a type I progesterone receptor (PR) antagonist, which prevents PR-induced DNA transcription.
  • PR progesterone receptor
  • Onapristone anti-cancer activity is documented in multiple pre-clinical models and clinical studies in patients with hormone therapy-naive or tamoxifen-resistant breast cancer. Despite promising activity in breast cancer models, the development of onapristone as an oncology drug was terminated due to liver function test abnormalities. See, e.g., Robertson et al., Eur J Cancer. 35(4214-8 (Feb. 1999).
  • progesterone receptor has been described in breast [Mote 2000, Lange 2008], endometrial [Kim 2013, Mortel 1984], prostate [Lange 2007, Bonkhoff 2001], ovarian [Sieh 2013], and several other cancers [Yin 2010, Ishibashi 2005, Blankenstein 200].
  • Antiprogestins have been shown to have an inhibitory effect on the growth of different type of cancer cells, and antiprogestin treatment has been studied in breast [Jonat 2013], endometrial [Thigpen 1999], prostate [Taplin 2008] cancers and uterine sarcomas [Koivisto-Korander 2007].
  • progesterone is mediated by two distinct nuclear receptor proteins, PRA and PRB, two transcriptional isoforms of the single PR gene.
  • PRA and PRB two distinct nuclear receptor proteins
  • both PR isoforms are expressed and are required to mediate the physiological effects of progestin ligands [Mote 2002, Arnett-Mansfield 2004].
  • the two PR isoforms have both been detected in malignant tissues, such as breast, endometrial, ovarian and prostate cancers [Cotta 2015].
  • ONA is a type 1 antiprogestin which prevents PR monomers from dimerizing, inhibits ligand-induced phosphorylation, prevents association of the PR with its co-activators, and thus prevents PR-mediated. DNA transcription. ONA does not allow the PR complex to bind to DNA, does not or minimally modulates PR-mediated genes, and inhibits ligand-induced PR phosphorylation, in contrast to other antiprogestins [Beck 1996; Afhüppe 2009]. Preclinical activity has been shown in several models, including endometrial cancer [Mueller 2003] and the clinical anticancer activity of ONA has been previously documented in patients with hormone therapy-naive [Robertson 1999] or tamoxifen-resistant [Jonat 2002] breast cancer.
  • APR Transcriptionally activated PR
  • IHC immunohistochemistry
  • onapristone was provided to patients with cancer (e.g., breast, endometrial, others) in an immediate release formulation of 100 mg and provided QD (once per day).
  • Onapristone has also been given to patients in endocrinology studies, at immediate release doses of 1 and 10 mg doses resulting in a dose-dependent effect of onapristone on suppression of gonadotrophin (luteinizing hormone [LH] and follicle-stimulating hormone [FSH]) secretion. Cameron 2003.
  • these studies used immediate release formulations of onapristone of unknown purity.
  • these studies addressed the dose and formulation of onapristone suitable for potential contraceptive use rather than the dose and formulation suitable for treating a disease such as cancer.
  • extended-release pharmaceutical compositions comprising onapristone as the active ingredient in an amount from about 2 mg to about 100 mg.
  • the extended-release pharmaceutical compositions (also referred to herein as ER formulations) further comprise excipients suitable for the desired dosage form (e.g., tablet, capsule, etc.) and for delaying the release of the active ingredient.
  • onapristone ER formulations utilizing highly purified onapristone (e.g., at least about 98%).
  • the ratio of onapristone to inactive excipients in the ER formulations is about 0.05 to about 5%.
  • the AUC (area under the curve) of onapristone is at least about 1578 ng*h/ml over about an 8-12 hour period after administration of a 10 mg dose BID (i.e., twice per day) to a patient.
  • the Cmax (maximum plasma concentration) of onapristone is at least about 240 ng/ml over about an 8-12 hour period after administration of a 10 mg dose BID to a patient.
  • a steady state plasma concentration of onapristone is achieved at about 8 days following the initial dose of the extended release onapristone pharmaceutical composition.
  • the extended-release onapristone pharmaceutical composition comprises at least about 10 mg to about 50 mg of onapristone.
  • FIG. 1 shows the exemplary Cmax (maximum active ingredient concentration) levels per ONA dose level (10 mg, 20 mg, 30 mg, 40 mg, 50 mg extended-release BID (twice per day) and 100 mg QD (once per day));
  • FIG. 2 shows the exemplary AIX (area under the curve) per ONA dose level (10 mg, 20 mg, 30 mg, 40 mg, 50 mg extended-release formulations BID (twice per day) and 100 mg QD (once per day));
  • FIG. 3 shows the exemplary accumulation of ONA over time per ONA dose level (10 mg, 20 mg, 30 mg, 40 mg, 50 mg extended-release formulations BID (twice per day) and 100 mg QD (once per day)); and
  • FIGS. 4 A and 4 B show exemplary ONA plasma levels over time per dose levels for the extended-release formulations BID ( FIG. 4 A ) and the 100 mg formulation QD.
  • onapristone ER formulations comprise onapristone (ONA) ((8S,11R,13R,14S,17S)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one), an anti-progestin drug and progesterone receptor antagonist having the following structure:
  • ER formulations of onapristone arc provided.
  • extended release refers to a pharmaceutical compositions or drug formulation that is administered to a patient and has a mechanism to delay the release an active ingredient (i.e., drug).
  • ER pharmaceutical compositions include the active ingredient (e.g., onapristone) and excipients that delay release of the active ingredient (e.g., hydroxypropyl methylcellulose, ethyl cellulose, Eudragit (Evonik Industries) sustained release formulations (polymethacrylates), polyvinylpyrrolidone (PVP), carrageenan, etc.).
  • immediate release refers to pharmaceutical compositions or drug formulations that do not have a mechanism for delaying the release of the active ingredient following administration of the formulation to a patient.
  • exemplary extended release formulations are provided, for example, in Table 4 herein.
  • the terms “treat,” “prevent,” or similar terms, as used herein, do not necessarily mean 100% or complete treatment or prevention. Rather, these terms refer to various degrees of treatment or prevention of a particular disease (e.g., 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, or 1%) as recognized in the art as being beneficial.
  • treatment or “prevention” also refer to delaying onset of a disease for a period of time or delaying onset indefinitely.
  • treatment refers to administering a drug or treatment to a patient or prescribing a drug to a patient where the patient or a third party (e.g., caretaker, family member, or health care professional) administers the drug or treatment
  • a third party e.g., caretaker, family member, or health care professional
  • an extended-release pharmaceutical composition comprising onapristone wherein onapristone is present in an amount from about 2 mg to about 50 mg.
  • Onapristone can be provided, for example, in quantities of 2 mg, 2.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 37.5 mg, and 50 mg in any suitable extended release formulation (e.g., formulations of Table 4) multiple times per day (e.g., twice per day) or once per day.
  • ER formulations can include excipients that delay the dissolution of the tablet and the subsequent release of onapristone into the gastrointestinal track which then is absorbed into the bloodstream of a patient over time thereby reducing the C max concentration compared to an IR formulation.
  • a similar release profile can be achieved through the use of an osmotic tablet or a tablet film coated with a polymer that results in an extended release profile of the tablet.
  • onapristone ER formulations can be provided in any suitable dosage form (e.g., tablet, capsule, etc.) with a total weight of active ingredients plus excipients ranging from about 50 mg to 400 mg.
  • the tablet can be a matrix tablet, film coated tablet or osmotic pump.
  • onapristone ER formulations can be administered to a patient in need of treatment with onapristone once per day, twice per day (BID), or more to achieve the desired dose of onapristone.
  • onapristone ER formulations wherein the purity of the onapristone is at least about 98%. Without being bound by theory, it is believed that using a highly purified form of onapristone in part decreases the liver function test abnormalities resulting in clinical benefits for cancer patients at all doses.
  • the ratio of onapristone to inactive excipients in the onapristone ER formulation is about 0.05 (e.g., Table 4) to about 5%.
  • ER formulations wherein the AUC of onapristone following the administration of 10 mg of the onapristone ER formulation to a patient BID is at least about 1578 ng*h/ml over about 8-12 hours. in one aspect, the time period can vary by about plus or minus two hours.
  • Another aspect provides onapristone ER formulations where the Cmax of onapristone following the administration of 10 mg of the onapristone ER formulation to a patient BID is at least about 240 ng/ml over about 8-12 hours. In one aspect, the time period can vary by about plus or minus two hours.
  • Another aspect provides onapristone ER formulations where a steady state plasma concentration of onapristone is achieved at about 8 days following the administration of the onapristone ER formulations to a patient twice a day (BID).
  • FIG. 1 For brevity, the figures in this specification may be administered using two onapristone ER formulation twice per day (BID) to a cancer patient, where the onapristone ER formulation comprises of at least about 10 mg to about 50 mg of onapristone.
  • the ER formulation is administered once per day.
  • the onapristone in the onapristone ER formulation is at least about 98% pure.
  • onapristone administered to a patient it at least about 98% pure.
  • onapristone in the onapristone ER formulations can be provided, for example, in quantities of 2 mg, 2.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 37.5 mg, and 50 mg.
  • the onapristone ER formulations can be administered twice per day (BID) to a human subject in need of treatment, where the onapristone ER formulation comprises of at least about 10 mg to about 50 mg of onapristone. In one aspect, the ER formulation is administered once per day. In another aspect, the disorder is selected from the group consisting of breast cancer, endometrial cancer, prostate cancer, ovarian, uterine endometrioid cancers, and other types of cancer which express the PR.
  • the onapristone ER formulation is administered to a human subject having a disorder capable of treatment with onapristone wherein the the AUC of onapristone following the administration of 10 mg of the onapristone ER formulation to a patient BID is at least about 1578 ng*h/ml over about 8-12 hours.
  • the time period can vary by about plus or minus two hours.
  • the onapristone ER formulation is administered to a human subject having a disorder capable of treatment with onapristone by administering an onapristone ER formulation to the subject twice per day (BID) where the Cmax of onapristone in the human subject is at least about 240 ng/ml over about 8-12 hours.
  • BID twice per day
  • the onapristone ER formulation is administered once per day.
  • the time period can vary by about plus or minus two hours.
  • an onapristone ER formulation is administered to a human subject having a disorder capable of treatment with onapristone twice per day (BID) where a steady state plasma concentration is achieved at about 8 days.
  • BID onapristone twice per day
  • PK results for onapristone are available for 52 patients from the a first study (ARN-AR18-CT-101) (Table 1). Variability for onapristone PK is moderate and greater for the IR versus the ER formulation.
  • Onapristone Cmax and AUC values for the ER form arc proportional to administered dose ( FIGS. 1 and 2 ). Based on observed mean AUC values, oral bioavailability fir the ER versus the IR formulation is approximately 50% ( FIG. 24 ).
  • a later Tmax value for the ER form results in somewhat lower dose-corrected Cmax values for the ER form compared to the IR form. Steady state is attained before day 8 with a mean t1 ⁇ 2 of 7.5 hrs.
  • Table 1 compares descriptive statistics for the primary onapristone pharmacokinetic exposure parameters following single oral doses from 10 to 50 mg of extended -release onapristone compared to that from 100 mg immediate-release onapristone (Study ARN-AR18-CT-101). Exposure following ER onapristone appears later than that for IR onapristone, consistent with extended release formulations. However, the extended-release aspects are not reflected in the overall duration of exposure. Although study size is small, onapristone exposure generally increases in proportion to ER onapristone dose. Exposure at 50 mg ER onapristone is approximately 20-50% that of 100 mg IR onapristone depending on the formulation. Variability in these parameters is similar for both formulations and across ER onapristone dose levels.
  • PK results arc available in 19 patients from a second study (ARN-AR18-CT-102) and show linear dose relationships for C max and AUC (Table 2) following single oral doses from 10 to 50 mg of extended -release onapristone.
  • the ER formulation appears to be performing according to the dose release specifications with a t 1/2 of approximately 8 hours and a T max of approximately 3-4 hours. Steady state is also achieved. within 8 days in this study. Day 29 and 57 data indicate no evidence of accumulation over time, once steady state is reached.
  • Onapristone exposure generally increases less than proportionally with the ER onapristone dose formulation. Variability in these parameters is similar across ER onapristone dose levels.
  • FIGS. 1 and 2 show the results of an exemplary comparison of the relative systemic onapristone exposure following single oral doses from 10 to 50 mg of extended release onapristone compared to that from 100 mg immediate-release onapristone (Study ARN-AR18-CT-101).
  • Onapristone exposure assessed by Cmax ( FIG. 1 ) and AUC ( FIG. 2 ), increases linearly across the ER onapristone dose range and is lower than that for IR onapristone at all ER dose levels.
  • the ER onapristone formulations provided clinical benefit to patients despite tower onapristone exposure.
  • FIG. 3 shows the results of an exemplary comparison of the degree of onapristone accumulation following twice-daily oral doses from 10 to 50 mg of extended -release onapristone compared. to that from daily oral 100 mg immediate-release onapristone (Study ARN-ARI8-CT-101). Accumulation for the ER onapristone formulation given twice daily is measurably greater than that for IR onapristone given daily.
  • FIGS. 4 A and 4 B show exemplary plasma onapristone concentration-time profiles fix individual subjects following single oral doses of 50 mg extended -release onapristone compared to that from 100 mg immediate-release onapristone (Study ARN-AR18-CT-101).
  • the profiles for ER onapristone generally reach maximum concentrations more slowly than those for IR onapristone, supporting the extended release of drug from the ER formulation. Concentrations at all dose levels of ER onapristone are generally lower than those for 100 mg IR onapristone.
  • the ER onapristone formulations provided clinical benefit to patients despite lower onapristone exposure.
  • Table 4 provide exemplary onapristone extended release formulations.
  • the tablets can be provided to a patient alone or in any desired combination to achieve the desired dose.
  • Onapristone extended-release formulations can be prepared by the following exemplary method:
  • Step 1 De-lump onapristone drug substance by milling or by passing through a wire screen followed by further passing the resulting de-lumped onapristone through a wire screen of appropriate mesh size (e.g., 425 or 710 microns).
  • appropriate mesh size e.g., 425 or 710 microns.
  • Step 2 Screen the colloidal silicon dioxide and approximately half of the pregelatinized starch separately through a screen of appropriate mesh size (e.g., 425 or 710 microns) into a stainless steel blending container. The previously-screened onapristone drug substance from Step 1 is added to this blend.
  • appropriate mesh size e.g. 425 or 710 microns
  • Step 3 The mixture is blended anal screened through a screen of appropriate mesh size (e.g., 425 or 710 microns).
  • Step 4 The remaining pregelatinized starch is screened through a screen of appropriate mesh size (e.g., 425 or 710 microns) into the stainless steel blending container (from Step 2). The previously screened mixture from Step 3 is added to the container.
  • appropriate mesh size e.g. 425 or 710 microns
  • Step 5 The mixture is blended to achieve a homogenous mix.
  • Step 6 Approximately half of the microcrystalline cellulose, half of the lactose monohydrate and half of the hydroxypropyl methylcellulose are separately screened into a larger stainless steel blending container through a screen of appropriate mesh size (e.g., 425 or 710 microns). The blend from Step 5 is added to this container, and the remaining microcrystalline cellulose, lactose monohydrate and hydroxypropyl methylcellulose are screened into the container through a screen of appropriate mesh size (e,g., 425 or 710 microns).
  • a screen of appropriate mesh size e.g., 425 or 710 microns
  • Step 7 The mixture is blended further to achieve a homogeneous mix.
  • Step 8 The mixture from Step 7 is co-screened with magnesium stearate through a screen of appropriate mesh size (e.g., 425 or 710 microns) into the container from Step 4.
  • appropriate mesh size e.g., 425 or 710 microns
  • the PR determination for inclusion purposes was performed on archived tissue blocks in the pathology department of each participating center. Central PR/APR evaluation was planned, but retrospective relative to inclusion and treatment.
  • R2D recommended phase 2 dose
  • Highly purified ONA tablets can be by standard pharmaceutical chemistry purification methods by those skilled in the art. ER formulation with release kinetics from 10-12 hours depending on tablet dose. The original study design included a. 20-patient expansion component. An 8-week dose-limiting toxicity (DLT) observation period was utilized to characterize thoroughly the safety profile, as previous ONA studies demonstrated a spike in the LFTs at approximately 6 weeks of treatment.
  • DLT dose-limiting toxicity

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)

Abstract

Onapristone extended-release formulations and methods of administering onapristone extended-release formulations are provided. Onapristone extended-release formulations provide sufficient therapeutic activity as compared to immediate-release formulations with reduced potential for adverse side effects.

Description

    PRIORITY CLAIM
  • This application is a continuation of U.S. patent application Ser. No. 17/035,544 tiled Sep. 28, 2020, which is a continuation of U.S. patent application Ser. No. 14/942.809 filed Nov. 16, 2015, which claims the benefit of U.S. Provisional Patent Application No. 62/080,868 filed Nov. 17, 2014, each of which is hereby incorporated by reference in its entirety.
  • BACKGROUND
  • Onapristone (ONA) is an anti-progestin drug and progesterone receptor antagonist which was originally developed for potential contraceptive use and the use in benign gynecological disorders such as the treatment of uterine leiomyomas. However, onapristone has demonstrated substantial activity in advanced breast cancer. It is thought that ONA binds to the progesterone receptor (PR), preventing the PR from binding to DNA and thereby inhibiting or eliminating PR-induced DNA transcription. See, e.g., Klijn et al., Progesterone antagonists and progesterone receptor modulation in the treatment of breast cancer, Steroids, v. 65, pp. 825-830 (2000); Jonat et al., The clinical efficacy of progesterone antagonists in breast cancer, Endocrine Therapy of Breast Cancer, pp. 117-124.
  • Onapristone is a type I progesterone receptor (PR) antagonist, which prevents PR-induced DNA transcription. Presence of transcriptionally activated PR (APR) in tissue samples from a cancer patient, measured using, for example, an immunohistochemistry companion diagnostic procedure, indicates susceptibility to treatment with onapristone anticancer activity. Onapristone anti-cancer activity is documented in multiple pre-clinical models and clinical studies in patients with hormone therapy-naive or tamoxifen-resistant breast cancer. Despite promising activity in breast cancer models, the development of onapristone as an oncology drug was terminated due to liver function test abnormalities. See, e.g., Robertson et al., Eur J Cancer. 35(4214-8 (Feb. 1999).
  • Expression of the progesterone receptor (PR) has been described in breast [Mote 2000, Lange 2008], endometrial [Kim 2013, Mortel 1984], prostate [Lange 2007, Bonkhoff 2001], ovarian [Sieh 2013], and several other cancers [Yin 2010, Ishibashi 2005, Blankenstein 200]. Antiprogestins have been shown to have an inhibitory effect on the growth of different type of cancer cells, and antiprogestin treatment has been studied in breast [Jonat 2013], endometrial [Thigpen 1999], prostate [Taplin 2008] cancers and uterine sarcomas [Koivisto-Korander 2007].
  • The effects of progesterone are mediated by two distinct nuclear receptor proteins, PRA and PRB, two transcriptional isoforms of the single PR gene. In luminal epithelial cells of the normal breast and in normal endometrium, both PR isoforms are expressed and are required to mediate the physiological effects of progestin ligands [Mote 2002, Arnett-Mansfield 2004]. The two PR isoforms have both been detected in malignant tissues, such as breast, endometrial, ovarian and prostate cancers [Cotta 2015].
  • ONA is a type 1 antiprogestin which prevents PR monomers from dimerizing, inhibits ligand-induced phosphorylation, prevents association of the PR with its co-activators, and thus prevents PR-mediated. DNA transcription. ONA does not allow the PR complex to bind to DNA, does not or minimally modulates PR-mediated genes, and inhibits ligand-induced PR phosphorylation, in contrast to other antiprogestins [Beck 1996; Afhüppe 2009]. Preclinical activity has been shown in several models, including endometrial cancer [Mueller 2003] and the clinical anticancer activity of ONA has been previously documented in patients with hormone therapy-naive [Robertson 1999] or tamoxifen-resistant [Jonat 2002] breast cancer.
  • Transcriptionally activated PR (APR) can be detected by observational evaluation of the subnuclear distribution pattern using immunohistochemistry (IHC). Using this method, APR can be used as a potentially predictive biomarker in endometrioid cancer of the uterus. See U.S. Pat. No. 9,046,534. APR detection is being developed as a companion diagnostic to identify patients more likely to respond to ONA [Bonneterre 2015].
  • Early clinical studies employing the original immediate release (IR) formulation of ONA have shown that ONA is well-tolerated with the exception of abnormalities in liver function tests (LFTs) [Cameron 1996, Cameron 2003, Croxatto 1994, Jonat 2002, Robertson 1999]. Studies with the original IR formulation were discontinued due to these LFT abnormalities. Id.
  • Previously, onapristone was provided to patients with cancer (e.g., breast, endometrial, others) in an immediate release formulation of 100 mg and provided QD (once per day). Onapristone has also been given to patients in endocrinology studies, at immediate release doses of 1 and 10 mg doses resulting in a dose-dependent effect of onapristone on suppression of gonadotrophin (luteinizing hormone [LH] and follicle-stimulating hormone [FSH]) secretion. Cameron 2003. However, these studies used immediate release formulations of onapristone of unknown purity. Importantly, these studies addressed the dose and formulation of onapristone suitable for potential contraceptive use rather than the dose and formulation suitable for treating a disease such as cancer.
  • What is needed is an improved formulation of onapristone which allow for a continuous suppression of the PR and methods of administering the same resulting in sufficient bioavailability to provide clinical benefit to cancer patients at doses which result in less toxicity than the previous clinical experience with onapristone.
  • SUMMARY
  • Aspects described herein provide extended-release pharmaceutical compositions comprising onapristone as the active ingredient in an amount from about 2 mg to about 100 mg. The extended-release pharmaceutical compositions (also referred to herein as ER formulations) further comprise excipients suitable for the desired dosage form (e.g., tablet, capsule, etc.) and for delaying the release of the active ingredient.
  • Further aspects provide onapristone ER formulations utilizing highly purified onapristone (e.g., at least about 98%). In another aspect, the ratio of onapristone to inactive excipients in the ER formulations is about 0.05 to about 5%.
  • In a further aspect, the AUC (area under the curve) of onapristone is at least about 1578 ng*h/ml over about an 8-12 hour period after administration of a 10 mg dose BID (i.e., twice per day) to a patient.
  • In another aspect, the Cmax (maximum plasma concentration) of onapristone is at least about 240 ng/ml over about an 8-12 hour period after administration of a 10 mg dose BID to a patient. In yet another aspect, a steady state plasma concentration of onapristone is achieved at about 8 days following the initial dose of the extended release onapristone pharmaceutical composition. in another aspect, the extended-release onapristone pharmaceutical composition comprises at least about 10 mg to about 50 mg of onapristone.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the exemplary Cmax (maximum active ingredient concentration) levels per ONA dose level (10 mg, 20 mg, 30 mg, 40 mg, 50 mg extended-release BID (twice per day) and 100 mg QD (once per day));
  • FIG. 2 shows the exemplary AIX (area under the curve) per ONA dose level (10 mg, 20 mg, 30 mg, 40 mg, 50 mg extended-release formulations BID (twice per day) and 100 mg QD (once per day));
  • FIG. 3 shows the exemplary accumulation of ONA over time per ONA dose level (10 mg, 20 mg, 30 mg, 40 mg, 50 mg extended-release formulations BID (twice per day) and 100 mg QD (once per day)); and
  • FIGS. 4A and 4B show exemplary ONA plasma levels over time per dose levels for the extended-release formulations BID (FIG. 4A) and the 100 mg formulation QD.
  • DETAILED DESCRIPTION
  • Before describing several exemplary aspects described herein, it is to be understood that the invention is not limited to the details of construction or process steps set forth in the following description. The aspects described herein are capable of being practiced or being carried out in various ways.
  • In another aspect onapristone ER formulations comprise onapristone (ONA) ((8S,11R,13R,14S,17S)-11-[4-(dimethylamino)phenyl]-17-hydroxy-17-(3-hydroxypropyl)-13-methyl-1,2,6,7,8,11,12,14,15,16-decahydrocyclopenta[a]phenanthren-3-one), an anti-progestin drug and progesterone receptor antagonist having the following structure:
  • Figure US20230157960A1-20230525-C00001
  • In one aspect, ER formulations of onapristone arc provided. The term “extended release” refers to a pharmaceutical compositions or drug formulation that is administered to a patient and has a mechanism to delay the release an active ingredient (i.e., drug). For example, ER pharmaceutical compositions include the active ingredient (e.g., onapristone) and excipients that delay release of the active ingredient (e.g., hydroxypropyl methylcellulose, ethyl cellulose, Eudragit
    Figure US20230157960A1-20230525-P00001
    (Evonik Industries) sustained release formulations (polymethacrylates), polyvinylpyrrolidone (PVP), carrageenan, etc.). The term “immediate release” (IR) refers to pharmaceutical compositions or drug formulations that do not have a mechanism for delaying the release of the active ingredient following administration of the formulation to a patient. Exemplary extended release formulations are provided, for example, in Table 4 herein. The terms “treat,” “prevent,” or similar terms, as used herein, do not necessarily mean 100% or complete treatment or prevention. Rather, these terms refer to various degrees of treatment or prevention of a particular disease (e.g., 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 10%, 5%, or 1%) as recognized in the art as being beneficial. The terms “treatment” or “prevention” also refer to delaying onset of a disease for a period of time or delaying onset indefinitely. The term “treatment” or “treating” refers to administering a drug or treatment to a patient or prescribing a drug to a patient where the patient or a third party (e.g., caretaker, family member, or health care professional) administers the drug or treatment
  • One aspect provides an extended-release pharmaceutical composition comprising onapristone wherein onapristone is present in an amount from about 2 mg to about 50 mg. Onapristone can be provided, for example, in quantities of 2 mg, 2.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 37.5 mg, and 50 mg in any suitable extended release formulation (e.g., formulations of Table 4) multiple times per day (e.g., twice per day) or once per day. ER formulations can include excipients that delay the dissolution of the tablet and the subsequent release of onapristone into the gastrointestinal track which then is absorbed into the bloodstream of a patient over time thereby reducing the Cmax concentration compared to an IR formulation. A similar release profile can be achieved through the use of an osmotic tablet or a tablet film coated with a polymer that results in an extended release profile of the tablet.
  • In another aspect, onapristone ER formulations can be provided in any suitable dosage form (e.g., tablet, capsule, etc.) with a total weight of active ingredients plus excipients ranging from about 50 mg to 400 mg. In another aspect, the tablet can be a matrix tablet, film coated tablet or osmotic pump. In yet another aspect, onapristone ER formulations can be administered to a patient in need of treatment with onapristone once per day, twice per day (BID), or more to achieve the desired dose of onapristone.
  • Further aspects provide onapristone ER formulations wherein the purity of the onapristone is at least about 98%. Without being bound by theory, it is believed that using a highly purified form of onapristone in part decreases the liver function test abnormalities resulting in clinical benefits for cancer patients at all doses.
  • In another aspect, the ratio of onapristone to inactive excipients in the onapristone ER formulation is about 0.05 (e.g., Table 4) to about 5%.
  • Further aspects provide ER formulations wherein the AUC of onapristone following the administration of 10 mg of the onapristone ER formulation to a patient BID is at least about 1578 ng*h/ml over about 8-12 hours. in one aspect, the time period can vary by about plus or minus two hours.
  • Another aspect provides onapristone ER formulations where the Cmax of onapristone following the administration of 10 mg of the onapristone ER formulation to a patient BID is at least about 240 ng/ml over about 8-12 hours. In one aspect, the time period can vary by about plus or minus two hours.
  • Another aspect provides onapristone ER formulations where a steady state plasma concentration of onapristone is achieved at about 8 days following the administration of the onapristone ER formulations to a patient twice a day (BID).
  • Further aspects provide methods of administering onapristone to a patient comprising administering an onapristone ER formulation twice per day (BID) to a cancer patient, where the onapristone ER formulation comprises of at least about 10 mg to about 50 mg of onapristone. In one aspect, the ER formulation is administered once per day. In another aspect, the onapristone in the onapristone ER formulation is at least about 98% pure.
  • In one aspect, the onapristone administered to a patient it at least about 98% pure. In yet another aspect, onapristone in the onapristone ER formulations can be provided, for example, in quantities of 2 mg, 2.5 mg, 5 mg, 10 mg, 20 mg, 25 mg, 37.5 mg, and 50 mg.
  • In yet another aspect, the onapristone ER formulations can be administered twice per day (BID) to a human subject in need of treatment, where the onapristone ER formulation comprises of at least about 10 mg to about 50 mg of onapristone. In one aspect, the ER formulation is administered once per day. In another aspect, the disorder is selected from the group consisting of breast cancer, endometrial cancer, prostate cancer, ovarian, uterine endometrioid cancers, and other types of cancer which express the PR.
  • In another aspect, the onapristone ER formulation is administered to a human subject having a disorder capable of treatment with onapristone wherein the the AUC of onapristone following the administration of 10 mg of the onapristone ER formulation to a patient BID is at least about 1578 ng*h/ml over about 8-12 hours. In another aspect, the time period can vary by about plus or minus two hours.
  • In another aspect, the onapristone ER formulation is administered to a human subject having a disorder capable of treatment with onapristone by administering an onapristone ER formulation to the subject twice per day (BID) where the Cmax of onapristone in the human subject is at least about 240 ng/ml over about 8-12 hours. In another aspect, the onapristone ER formulation is administered once per day. In yet another aspect, the time period can vary by about plus or minus two hours.
  • In another aspect, an onapristone ER formulation is administered to a human subject having a disorder capable of treatment with onapristone twice per day (BID) where a steady state plasma concentration is achieved at about 8 days.
  • PK results for onapristone are available for 52 patients from the a first study (ARN-AR18-CT-101) (Table 1). Variability for onapristone PK is moderate and greater for the IR versus the ER formulation. Onapristone Cmax and AUC values for the ER form arc proportional to administered dose (FIGS. 1 and 2 ). Based on observed mean AUC values, oral bioavailability fir the ER versus the IR formulation is approximately 50% (FIG. 24 ). A later Tmax value for the ER form results in somewhat lower dose-corrected Cmax values for the ER form compared to the IR form. Steady state is attained before day 8 with a mean t½ of 7.5 hrs.
  • Table 1 compares descriptive statistics for the primary onapristone pharmacokinetic exposure parameters following single oral doses from 10 to 50 mg of extended -release onapristone compared to that from 100 mg immediate-release onapristone (Study ARN-AR18-CT-101). Exposure following ER onapristone appears later than that for IR onapristone, consistent with extended release formulations. However, the extended-release aspects are not reflected in the overall duration of exposure. Although study size is small, onapristone exposure generally increases in proportion to ER onapristone dose. Exposure at 50 mg ER onapristone is approximately 20-50% that of 100 mg IR onapristone depending on the formulation. Variability in these parameters is similar for both formulations and across ER onapristone dose levels.
  • TABLE 1
    Summary of PK results for 52 Patients
    From Study ARN-AR18-CT-101
    Form
    ER ER ER ER ER IR
    Dose (mg)
    10 20 30 40 50 100
    n
    12 12 6 10 6 6
    AUCtau (ng*h/mL)
    Mean 1578 4228 4856 6833 8966 40800
    CV % 75 94 19 65 53 51
    Cmax (ng/mL)
    Mean 240 586 767 870 1459 4296
    CV % 67 77 15 67 48 54
    tmax (hrs)
    Mean 3.4 3.8 3.8 5.2 2.5 1.3
    CV % 47 50 51 68 55 61
    t1/2 (hrs)
    Mean 8.9 7.9 3.9 23.9 11.1 23.6
    CV % 120 39 31 183 140 165
  • PK results arc available in 19 patients from a second study (ARN-AR18-CT-102) and show linear dose relationships for Cmax and AUC (Table 2) following single oral doses from 10 to 50 mg of extended -release onapristone. Confirming the ARN-AR.18-CT-101 study, the ER formulation appears to be performing according to the dose release specifications with a t1/2 of approximately 8 hours and a Tmax of approximately 3-4 hours. Steady state is also achieved. within 8 days in this study. Day 29 and 57 data indicate no evidence of accumulation over time, once steady state is reached. Onapristone exposure generally increases less than proportionally with the ER onapristone dose formulation. Variability in these parameters is similar across ER onapristone dose levels.
  • TABLE 2
    Summary of PK results for 19 Patients From Study ARN-AR18-CT-102.
    Onapristone ER twice-daily dose
    Parameter
    10 mg 20 mg 30 mg 40 mg 50 mg
    mean (CV %) n = 5 n = 5 n = 3 n = 3 n = 3
    Tmax, h   4.0 (43)   3.6 (46)   4.0 (50)   3.0 (88)   3.3 (35)
    Cmax, ng/mL  260 (51)  362 (41)  325 (62)  680 (14)  538 (44)
    AUCi, 7013 (53) 9745 (44) 14380 (18) 17300 (27) 23541 (39)
    ng/mL * h
    CL, L/h  1.85 (48)  2.04 (31)   2.13 (18)   2.18 (10)   2.4 (46)
    t1/2, h  5.46 (63)  5.61 (30)   9.46 (44)   5.45 (55)   15.9 (53)
  • FIGS. 1 and 2 show the results of an exemplary comparison of the relative systemic onapristone exposure following single oral doses from 10 to 50 mg of extended release onapristone compared to that from 100 mg immediate-release onapristone (Study ARN-AR18-CT-101). Onapristone exposure, assessed by Cmax (FIG. 1 ) and AUC (FIG. 2 ), increases linearly across the ER onapristone dose range and is lower than that for IR onapristone at all ER dose levels. Surprisingly, as disclosed herein, the ER onapristone formulations provided clinical benefit to patients despite tower onapristone exposure.
  • FIG. 3 shows the results of an exemplary comparison of the degree of onapristone accumulation following twice-daily oral doses from 10 to 50 mg of extended -release onapristone compared. to that from daily oral 100 mg immediate-release onapristone (Study ARN-ARI8-CT-101). Accumulation for the ER onapristone formulation given twice daily is measurably greater than that for IR onapristone given daily.
  • FIGS. 4A and 4B show exemplary plasma onapristone concentration-time profiles fix individual subjects following single oral doses of 50 mg extended -release onapristone compared to that from 100 mg immediate-release onapristone (Study ARN-AR18-CT-101). The profiles for ER onapristone generally reach maximum concentrations more slowly than those for IR onapristone, supporting the extended release of drug from the ER formulation. Concentrations at all dose levels of ER onapristone are generally lower than those for 100 mg IR onapristone. Surprisingly, as disclosed herein, the ER onapristone formulations provided clinical benefit to patients despite lower onapristone exposure.
  • TABLE 3
    Efficacy In Study ARN-AR18-CT-101
    %
    Tumor change Duration
    type Dose Response STL weeks
    Serous
    10 PR −52 40
    OC
    Serous
    50 SD  −7 34
    OC
    Granulosa
    40 SD −24 24
    OC
    Granulosa
    30 SD  +5 32
    OC
    EC
    30 SD −13  30+
    EC 20 SD  +5 32
    BC 50 SD  −7  32+
    BC 20 SD NA 28
    BC 40 SD −10 24
  • Clinical benefit (PR (partial response or SD (stable disease) for ≥24 weeks) was observed in ovarian, breast and uterine endometrioid cancers using the onapristone ER formulation. One patient with serous ovarian cancer experienced a PR (32 week duration) and 8 patients had SD for at least 24 weeks (Table 3). The median progression free survival (PFS) was 57.5 days (range 21-281).
  • In study ARN-AR18-CT-101, in 52 female patients with PR-positive solid tumors, 9/46 patients (20%) receiving the onapristone ER formulation at doses from 10-50 mg BID demonstrated clinical benefit, vs. 0/6 (0%) patients receiving the 100mg once-daily onapristone IR formulation. Clinical benefit responses, defined as RECIST 1.1 partial response or stable disease for at least 24 weeks, were seen only in patients receiving ER. Of interest, 7/9 of the patients with clinical benefit (78%) received doses below the established 100 mg IR dose and the patient with a partial response was treated at the lowest ER dose level, 10 mg BID.
  • With respect to ARN-AR18-CT-102, 2 of 21 patients with prostate cancer had SD after week 12. Median duration of treatment was 8 weeks.
  • EXAMPLE
  • The following non-limiting examples illustrate aspects described herein. Not every element described herein is required. Indeed, a person of skill in the art will find numerous additional uses of and variations to the methods described herein, which the inventors intend to be limited only by the claims, All references cited herein are incorporated by reference in their entirety.
  • Example 1
  • ER Formulations
  • TABLE 4
    Onapristone Extended-Release Formulations
    Amount per tablet (mg)
    Component 2.5 mg 5 mg 10 mg 20 mg Function
    Onapristone 2.50 5.00 10.00 20.00 Active
    Lactose monohydrate 10.25 20.50 41.00 82.00 Filler
    Microcrystalline 10.25 20.50 41.00 82.00 Filler
    cellulose
    Pregelatinized starch 10.00 20.00 40.00 80.00 Disintegrant
    Hydroxypropyl 16.50 33.00 66.00 132.00 Binder/
    methylcellulose modified
    release agent
    Colloidal silicon 0.25 0.50 1.00 2.00 Glidant
    dioxide
    Magnesium stearate 0.25 0.50 1.00 2.00 Lubricant
    Tablet weight (mg) 50.00 100.00 200.00 400.00
  • Table 4 provide exemplary onapristone extended release formulations. In one aspect, the tablets can be provided to a patient alone or in any desired combination to achieve the desired dose.
  • Example 2 Preparing Exemplary Onapristone ER Formulations
  • Onapristone extended-release formulations can be prepared by the following exemplary method:
  • Step 1: De-lump onapristone drug substance by milling or by passing through a wire screen followed by further passing the resulting de-lumped onapristone through a wire screen of appropriate mesh size (e.g., 425 or 710 microns).
  • Step 2: Screen the colloidal silicon dioxide and approximately half of the pregelatinized starch separately through a screen of appropriate mesh size (e.g., 425 or 710 microns) into a stainless steel blending container. The previously-screened onapristone drug substance from Step 1 is added to this blend.
  • Step 3: The mixture is blended anal screened through a screen of appropriate mesh size (e.g., 425 or 710 microns).
  • Step 4: The remaining pregelatinized starch is screened through a screen of appropriate mesh size (e.g., 425 or 710 microns) into the stainless steel blending container (from Step 2). The previously screened mixture from Step 3 is added to the container.
  • Step 5: The mixture is blended to achieve a homogenous mix.
  • Step 6: Approximately half of the microcrystalline cellulose, half of the lactose monohydrate and half of the hydroxypropyl methylcellulose are separately screened into a larger stainless steel blending container through a screen of appropriate mesh size (e.g., 425 or 710 microns). The blend from Step 5 is added to this container, and the remaining microcrystalline cellulose, lactose monohydrate and hydroxypropyl methylcellulose are screened into the container through a screen of appropriate mesh size (e,g., 425 or 710 microns).
  • Step 7: The mixture is blended further to achieve a homogeneous mix.
  • Step 8: The mixture from Step 7 is co-screened with magnesium stearate through a screen of appropriate mesh size (e.g., 425 or 710 microns) into the container from Step 4.
  • Example 3
  • Patients and Methods
  • Eligibility
  • Inclusion criteria included:
  • (1) post-menopausal female patients ≥18 years of age that have been previously treated recurrent or metastatic progesterone receptor-expressing cancer (e.g., endometrial, ovarian, breast cancer or uterine sarcoma) with evaluable disease per Response Evaluation Criteria In Solid Tumors, version 1.1 (RECIST 1.1);
  • (2) patients having available tissue blocks or biopsy specimens to determine progesterone receptor (PR) and activated progesterone receptor (APR) status; and
  • (3) patients having Eastern Cooperative Oncology Group (ECOG) performance status 0-1, and signed informed consent.
  • The PR determination for inclusion purposes was performed on archived tissue blocks in the pathology department of each participating center. Central PR/APR evaluation was planned, but retrospective relative to inclusion and treatment.
  • Key exclusion criteria included significantly impaired liver or kidney function, creatinine clearance lower than 60 ml/min, total bilirubin>upper limit of normal (ULN), alkaline phosphatase>ULN (or >2.5×ULN with liver or >5×ULN with bone metastases), ALT/AST>ULN (or >2.5×ULN with liver metastases), QTcF>480 msec, chronic inflammatory liver condition, severe concomitant disease, uncontrolled brain metastases, inadequate washout from previous therapy, inability to swallow or absorb tablets, use of inhibitors, inducers or substrates of CYP3A4, or use of progestin-based hormone replacement therapy.
  • Example 4
  • Study Design and Treatment
  • The study was an open-label, multicenter, randomized, parallel-group, two part phase 1-2 study with phase I part of the trial discussed herein. To determine the recommended phase 2 dose (RP2D), patients enrolled in this phase 1 study were randomized in parallel fashion to six (6) cohorts: five (5) cohorts of ER ONA tablets (10 mg BID, 20 mg BID, 30 mg BID, 40 mg BID, 50 mg BID) and one (1) cohort using the IR tablet formulation (100 mg QD). The trial was conducted in five (5) centers in France (registered on ClinicalTrials.gov as NCT02052128).
  • The study was approved by the Ile de France III Comite pour la Protection des Personnes (a French national ethics committee), the ANSM (French regulatory authority) and individual site scientific review boards, and written informed consent was obtained from each study patient.
  • Highly purified ONA tablets can be by standard pharmaceutical chemistry purification methods by those skilled in the art. ER formulation with release kinetics from 10-12 hours depending on tablet dose. The original study design included a. 20-patient expansion component. An 8-week dose-limiting toxicity (DLT) observation period was utilized to characterize thoroughly the safety profile, as previous ONA studies demonstrated a spike in the LFTs at approximately 6 weeks of treatment.
  • Patients were treated until documented. progressive disease (PD) or intolerance to medication. We consider the design of this study to be in agreement with the recently-proposed guidance for phase 1 protocols for dose escalation [Iasonos 2015].
  • Example 5
  • Pharmacokinetics Methods
  • Blood samples were collected at 0, 1, 2, 3, 4, 6, 8, 12 (before next BID dose), and 24 (before next dose—for 100 mg IR only) hours post-ONA, as well as hour 0 on days 8, 29 and 57 (just before drug intake). Plasma concentrations of ONA, mono-demethylated onapristone (M1) and other metabolites in plasma and urine were analyzed with a validated. ultra-performance liquid chromatography with tandem mass spectrometry detection (UPLC-MS/MS) assay. Pharmacokinetic modeling was performed using Monolix software in order to estimate PK parameters Cmax, Tmax, AUC0-last, AUC0-8, t1/2, Vd, CL, and Vc. Although the above description refers to particular aspects, it is to be understood that these aspects are merely illustrative. It will be apparent to those skilled in the art that various modifications and variations can be made to the polymorphic forms and methods described herein. Thus, it is intended that the present description include modifications and variations that are within the scope of the appended claims and their equivalents.
  • REFERENCES
    • 1. Afhüppe W, Sommer A, Muller J et al. Global gene expression profiling of progesterone receptor modulators in T47D cells provides a new classification system. J Steroid Biochem Mol Biol 2009; 113:101-115.
    • 2. Arnett-Mansfield R L, DeFazio A, Mote P A et al. Sub-nuclear Distribution of Progesterone Receptors A and B in Normal and Malignant Endometrium. The Journal of Clinical Endocrinology & Metabolism 2004; 89: 1429-1442.
    • 3. Beck C A, Zhang Y, Weigel N et al. Two Types of Anti-progestins Have Distinct Effects on Site-specific Phosphorylation of Human Progesterone Receptor. The Journal of Biological Chemistry 1996; 271:1209-1217.
    • 4. Benagiano G, Bastianelli C, Farris M. Selective progesterone receptor modulators 3: use in oncology, endocrinology and psychiatry. Expert Opin. Pharmacother 2008; 9:2487-2496.
    • 5. Blankenstein M A, Verheijen F M, Jacobs J M et al. Occurrence, regulation, and significance of progesterone receptors in human meningioma. Steroids 2000; 65: 795-800
    • 6. Bonkhoff H, Fixemer T, Hunsicker I, and Remberger K. Progesterone Receptor Expression in Human Prostate Cancer: Correlation With Tumor Progression. Prostate 2001; 48:285-291.
    • 7. Bonneterre J, Hutt E, Bosq J et al. Development of a technique to detect the activated form of the progesterone receptor and correlation with clinical and histopathological characteristics of endometrioid adenocarcinoma of the uterine corpus. Gynecologic Oncology 2015; doi:10.1016/j.ygyno.2015.06.037
    • 8. Cameron S, Critchley H O D, Buckley C H et al. The effects of post-ovulatory administration of onapristone on the development of a secretory endometrium. Human Reproduction 1996; 11 (1):40-49.
    • 9. Cameron S T, Glasier A F, Narvekar N et al. Effects of onapristone on postmenopausal endometrium. Steroids 2003; 68:1053-1059.
    • 10. Cottu P, A Italiano, A Varga et al. Onapristone (ON_) in progesterone receptor (PR)-expressing tumors: Efficacy and biomarker results of a dose-escalation phase 1 study. J Clin Oncol 2015; 33 (suppl; abstr 5593).
    • 11. Croxatto H, Salvatierra A A, Fuentealba B et al. Effect of the antiprogestin onapristone on follicular growth in women. Human Reproduction 1994; 9: 1442-1447.
    • 12. Goyeneche A A and Telleria C M. Antiprogestins in gynecological diseases. Reproduction 2015 149: R15-R33.
    • 13. Graham D, Bosq J, Caillaud J M et al. Determination of the activated form of the progesterone receptor (PR) in endometrial cancer (EC). J Clin Oncol 2013; 31(suppl; abstr 5602).
    • 14. Hopp T A, Weiss H L, Hilsenbeck S G, et al. Breast Cancer Patients with Progesterone Receptor PR-A-Rich Tumors Have Poorer Disease-Free Survival Rates. Clin Cancer Res 2004 10; 2751
    • 15. Hutt E, Bosq J, Powell M A, Leblanc E, Fujiwara K, Herzog T J, Coleman R L, Graham D, Clarke C, Gilles E M, Zukiwski A A, Monk B J. Clinical and pathological correlation of the activated form of the progesterone receptor (APR) in Endometrial Cancer (EC). ECC 2013, #1.002
    • 16. Iasonos A, Gönen M, Bosl G J. Scientific Review of Phase I Protocols With Novel Dose-Escalation Designs: How Much Information Is Needed? Journal of Clinical Oncology 2015:JCO. 2014.59. 8466.
    • 17. Ishibashi H, Suzuki T, Suzuki S, et al. Progesterone receptor in non-small cell lung cancer—a potent prognostic factor and possible target for endocrine therapy. Cancer Res 2005; 65(14):6450-8.
    • 18. Jonat W, Giurescu M, Robertson J F R. The clinical efficacy of progesterone antagonists in breast cancer, Endocrine Ther Breast Cancer 2002 (8):117-124.
    • 19. Jonat W, Bachelot T, Ruhstaller P et al. Randomized phase 2 study of lonaprisan as second line therapy for progesterone receptor positive breast cancer. Ann Oncol 2013; 24: 2543-2548.
    • 20. Kim J J, Kurita T, and Bulun S E. Progesterone Action in Endometrial Cancer, Endometriosis, Uterine Fibroids, and. Breast Cancer. Endocrine Rev 2013; 34: 13
    • 21. Klijn J G M, Setyono-Han B, Foekens J A. Progesterone antagonists and progesterone receptor modulators in the treatment of breast cancer, Steroids 2000; 65: 825-830.
    • 22. Koivisto-Korander R, Leminen A and Heikinheimo O. Mifepristone as treatment of recurrent progesterone receptor-positive uterine leiomyosarcoma. Obstetrics and Gynecology 2007; 109: 512-514.
    • 23. Lanari C, Wargon V, Rojas P and Molinoio A A. Antiprogestins in breast treatment: are we ready? Endocrine-Related Cancer 2012; 19: R35-R50.
    • 24. Lange C A, Gioeli D, Hammes S R, and P C Marker. Integration of Rapid. Signaling Events with Steroid Hormone Receptor Action in Breast and Prostate Cancer, Anna Rev Physiol 2007; 69:171-99,
    • 25. Lange C A, Sartorius C A, Abdel-Hafiz H, et al. Progesterone Receptor Action: Translating Studies in Breast Cancer Models to Clinical Insights, Innov Endocrinol Cancer 2008; 7: 94-110.
    • 26. Mortel R, Zaino R, and Satyaswaroop P G. Heterogeneity and Progesterone- Receptor Distribution in Endometrial Adenocarcinoma. Cancer 1984; 53:113-116,
    • 27. Mote P and Clarke C. Relative expression of progesterone receptors A and Bin premalignant and invasive breast lesions. Breast Cancer Research 2000; 2 (Suppl 1): P2.01 doi:10.1186/bcr103.
    • 28. Mote P A, Bartow S, Tran N, Clarke C L. Loss of co-ordinate expression of progesterone receptors A and B is an early event in breast carcinogenesis. Breast Cancer Res Treat 2002; 72(2):163-72.
    • 29. Mote P A, Graham J D, Clarke C L. Progesterone receptor isoforms in normal and malignant breast. Ernst Schering Found Symp Proc. 2007; (1):77-107.
    • 30. Mueller M D, Vigne J L, Pritts E A et al. Progestins activate vascular endothelial growth factor gene transcription in endometrial adenocarcinoma cells, Fertil Steril 2003; 79: 386- 392.
    • 31. Rezai K, Cottu P H, Huguet S et al. Population pharmacokinetic (PPK) modeling of onapristone in patients (pts) with progesterone receptor (PR)-expressing cancers. AACR Annual Meeting 2015. Abstract 4523.
    • 32. Robertson J F R, Willsher P C, Winterbottom L et al. Onapristone, a Progesterone Receptor Antagonist, as First-line Therapy in Primary Breast Cancer. Eur J Cancer 1999; 35: 214-218.
    • 33. Sieh W, Köbel M, Longacre T A, et al. Hormone-receptor expression and ovarian cancer survival: an Ovarian Tumor Tissue Analysis consortium study. Lancet Oncol 2013; http://dx.doi.org/10.1016/S1470-2045(13)70253-5.
    • 34. Taplin M E, Manola J, Oh W et al. A phase II study of mifepristone (RU-486) in castration-resistant prostate cancer, with a correlative assessment of androgen-related hormones. J Compil BJU Int 2008; 101: 1084-1089.
    • 35. Thigpen J T, Brady M, Alvarez R et al. Oral Medroxyprogesterone Acetate in the Treatment of Advanced or Recurrent Endometrial Carcinoma: A Dose-Response Study by the Gynecologic Oncology Group. J Clin Oncol 1999; 17: 1736-1744.
    • 36. Yin P, Lin Z, Reicrstad S, et al. Transcription Factor KLF11 Integrates Progesterone Receptor Signaling and Proliferation in Uterine Leiomyoma. Cells. Cancer Res 2010; 70(4); 1722-30.

Claims (17)

What is claimed is:
1. A method of administering onapristone to a patient having cancer comprising administering a pharmaceutical composition comprising onapristone to the subject, wherein the pharmaceutical composition comprises onapristone, lactose monohydrate, microcrystalline cellulose, pregelatinized starch, hydroxypropyl methylcellulose, silicon dioxide, and magnesium stearate.
2. The method of claim 1, wherein the composition comprises about 10 mg to about 50 mg of onapristone.
3. The method of claim 1, wherein the composition comprises about 10 mg, about 20 mg, or about 25 mg of onapristone.
4. The method of claim 1, wherein the pharmaceutical composition is administered twice per day.
5. The method of claim 1, wherein the onapristone is at least about 98% pure.
6. The method of claim 1, wherein the cancer expresses the progesterone receptor.
7. The method of claim 1, wherein the cancer is selected from the group consisting of breast, prostate, ovarian, and uterine cancer.
8. The method of claim 1, wherein the cancer is ovarian cancer.
9. The method of claim 1, wherein the cancer is breast cancer.
10. The method of claim 1, wherein the cancer is uterine cancer.
11. The method of claim 1, wherein the cancer is prostate cancer.
12. The method of claim 1, wherein the composition consists essentially of onapristone, lactose monohydrate, microcrystalline cellulose, pregelatinized starch, hydroxypropyl methylcellulose, silicon dioxide, and magnesium stearate.
13. The method of claim 1, wherein the composition consists of onapristone, lactose monohydrate, microcrystalline cellulose, pregelatinized starch, hydroxypropyl methylcellulose, silicon dioxide, and magnesium stearate.
14. A method of treating ovarian, breast or uterine cancer in a subject, the method comprising administering to the subject a pharmaceutical composition comprising onapristone, lactose monohydrate, microcrystalline cellulose, pregelatinized starch, hydroxypropyl methylcellulose, silicon dioxide, and magnesium stearate.
15. The method of claim 14, wherein the cancer is ovarian cancer.
16. The method of claim 14, wherein the cancer is uterine cancer.
17. The method of claim 14, wherein the cancer is breast cancer.
US18/151,846 2014-11-17 2023-01-09 Onapristone extended-release compositions and methods Abandoned US20230157960A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/151,846 US20230157960A1 (en) 2014-11-17 2023-01-09 Onapristone extended-release compositions and methods

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201462080868P 2014-11-17 2014-11-17
US14/942,809 US10786461B2 (en) 2014-11-17 2015-11-16 Onapristone extended-release compositions and methods
US17/035,544 US11672762B2 (en) 2014-11-17 2020-09-28 Onapristone extended-release compositions and methods
US18/151,846 US20230157960A1 (en) 2014-11-17 2023-01-09 Onapristone extended-release compositions and methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US17/035,544 Continuation US11672762B2 (en) 2014-11-17 2020-09-28 Onapristone extended-release compositions and methods

Publications (1)

Publication Number Publication Date
US20230157960A1 true US20230157960A1 (en) 2023-05-25

Family

ID=56014429

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/942,809 Active US10786461B2 (en) 2014-11-17 2015-11-16 Onapristone extended-release compositions and methods
US17/035,544 Active US11672762B2 (en) 2014-11-17 2020-09-28 Onapristone extended-release compositions and methods
US18/151,846 Abandoned US20230157960A1 (en) 2014-11-17 2023-01-09 Onapristone extended-release compositions and methods

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US14/942,809 Active US10786461B2 (en) 2014-11-17 2015-11-16 Onapristone extended-release compositions and methods
US17/035,544 Active US11672762B2 (en) 2014-11-17 2020-09-28 Onapristone extended-release compositions and methods

Country Status (11)

Country Link
US (3) US10786461B2 (en)
EP (1) EP3247362A4 (en)
JP (1) JP6655075B2 (en)
KR (1) KR20170084086A (en)
CN (2) CN113559075A (en)
AU (2) AU2015350241B2 (en)
CA (1) CA2966753A1 (en)
HK (1) HK1246653A1 (en)
MX (1) MX2017005163A (en)
RU (1) RU2017112748A (en)
WO (1) WO2016081383A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2017005163A (en) 2014-11-17 2018-01-18 Arno Therapeutics Inc Onapristone extended-release compositions and methods.
BR112018005999A2 (en) 2015-09-25 2019-01-08 Context Biopharma Inc methods for the production of onapristone intermediates
CN108883067B (en) 2015-12-15 2021-03-09 康特斯生物制药公司 Amorphous onapristone composition and preparation method thereof
US20180148471A1 (en) 2016-11-30 2018-05-31 Arno Therapeutics, Inc. Methods for onapristone synthesis dehydration and deprotection

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050085470A1 (en) * 2003-10-09 2005-04-21 Puwen Zhang 6-Amino-1,4-dihydro-benzo[d][1,3] oxazin-2-ones and analogs useful as progesterone receptor modulators
CA2925175A1 (en) * 2007-10-12 2009-04-16 Novartis Ag Compositions comprising sphingosine 1 phosphate (s1p) receptor modulators
US20140271819A1 (en) * 2013-03-12 2014-09-18 Stefan Proniuk Onapristone polymorphic forms and methods of use

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES533260A0 (en) 1983-06-15 1985-02-01 Schering Ag PROCEDURE FOR THE PREPARATION OF 13A-ALQUILGONANOS
US4780461A (en) 1983-06-15 1988-10-25 Schering Aktiengesellschaft 13α-alkyl-gonanes, their production, and pharmaceutical preparations containing same
DE3321826A1 (en) 1983-06-15 1984-12-20 Schering AG, 1000 Berlin und 4709 Bergkamen 13 alpha -Alkylgonanes, the preparation thereof and pharmaceutical products containing these
US4742000A (en) 1986-05-02 1988-05-03 University Of Chicago Antibody to human progesterone receptor and diagnostic materials and methods
DE3630030A1 (en) 1986-09-01 1988-03-03 Schering Ag 13 (ALPHA) -ALKYLGONAN- (DELTA) (UP ARROW) 9 (UP ARROW) (UP ARROW) ((UP ARROW) (UP ARROW) 1 (UP ARROW) 1 (UP ARROW)) (UP ARROW) -5, 10-EPOXIDE
US4774236A (en) 1986-09-17 1988-09-27 Research Triangle Institute 17α-(substituted-methyl)-17β-hydroxy/esterified hydroxy steroids and pharmaceutical compositions containing them
IE60780B1 (en) 1987-01-23 1994-08-10 Akzo Nv New 11-aryl steroid derivatives
DE3822770A1 (en) 1988-07-01 1990-01-04 Schering Ag 13-ALKYL-11SS-PHENYLGONANE
US5283190A (en) 1989-07-31 1994-02-01 Traish Adbulmaged M Specific monoclonal antibodies against a defined epitope of progesterone receptor and methods for their use
DE4008584A1 (en) 1990-03-15 1991-09-26 Schering Ag METHOD FOR PRODUCING INTERMEDIATE PRODUCTS FOR THE ANTIGESTAGE SYNTHESIS (ONAPRISTONE SYNTHESIS)
US5141961A (en) 1991-06-27 1992-08-25 Richrdson-Vicks Inc. Process for solubilizing difficulty soluble pharmaceutical actives
MX9301121A (en) 1992-03-02 1993-09-01 Schering Ag METHOD AND EQUIPMENT FOR ORAL CONTRACEPTION AND REGULATION OF MENSTRUATION WITH ESTROGEN / PROGESTIN / ANIPROGESTIN.
CN1053450C (en) 1992-11-19 2000-06-14 北京第三制药厂 Total synthesis method of 17-substituted 11 beta-substituted aromatic group-4, 9-estradiene compound
DE4332283A1 (en) 1993-09-20 1995-04-13 Jenapharm Gmbh Novel 11-benzaldoximestradiene derivatives, processes for their preparation and medicaments containing these compounds
JPH10507461A (en) 1994-10-24 1998-07-21 シエーリング アクチエンゲゼルシヤフト Competitive progesterone antagonists to regulate female fertility as needed
US5759577A (en) 1995-01-17 1998-06-02 American Home Products Corporation Controlled release of steroids from sugar coatings
US6900193B1 (en) 1996-05-01 2005-05-31 The United States Of America As Represented By The Department Of Health And Human Services Structural modification of 19-norprogesterone I: 17-α-substituted-11-β-substituted-4-aryl and 21-substituted 19-norpregnadienedione as new antiprogestational agents
IL122740A (en) 1997-01-15 2003-09-17 Akzo Nobel Nv 16-hydroxy-11-(substituted phenyl)-estra-9,4-diene derivatives, their preparation and pharmaceutical compositions containing them
US5962444A (en) 1998-05-29 1999-10-05 Research Triangle Institute 17β-nitro-11β-arylsteroids and their derivatives having agonist or antagonist hormonal properties
US6537584B1 (en) 1999-11-12 2003-03-25 Macromed, Inc. Polymer blends that swell in an acidic environment and deswell in a basic environment
ATE441433T1 (en) 2000-05-19 2009-09-15 Genentech Inc GENETIC DETECTION METHOD TO IMPROVE THE PROBABILITY OF AN EFFECTIVE RESPONSE TO CANCER THERAPY WITH A HERITAGE ANTAGONIST
US6750015B2 (en) 2000-06-28 2004-06-15 Kathryn B. Horwitz Progesterone receptor-regulated gene expression and methods related thereto
UY26966A1 (en) 2000-10-18 2002-06-20 Schering Ag USE OF ANTIPROGESTINES FOR THE INDUCTION OF APOPTOSIS IN A CELL
US7381976B2 (en) 2001-03-13 2008-06-03 Triton Thalassic Technologies, Inc. Monochromatic fluid treatment systems
AUPR546801A0 (en) 2001-06-05 2001-06-28 Commonwealth Scientific And Industrial Research Organisation Recombinant antibodies
US7105642B2 (en) 2001-08-03 2006-09-12 Cell Signalling Technology, Inc. Monoclonal antibodies specific for phosphorylated estrogen receptor alpha (Ser118) and uses thereof
US20040121304A1 (en) 2001-12-21 2004-06-24 Ulrike Fuhrmann Method for screening for progesterone receptor isoform-specific ligands and for tissue-selective progesterone receptor ligands
US20060111577A1 (en) 2003-02-28 2006-05-25 Kim Hyun K Method for preparing 17 alpha-acetoxy-11beta-(4-n,n-dimethylaminophenyl)-19-norpregna-4,9-diene-3,20-dione, intermediates thereof, and methods for the preparation of such intermediates
US20040265371A1 (en) 2003-06-25 2004-12-30 Looney Dwayne Lee Hemostatic devices and methods of making same
JP5017109B2 (en) 2004-07-09 2012-09-05 ザ・ポピュレイション・カウンシル,インコーポレイテッド Sustained release composition containing a progesterone receptor modulator
KR20070061893A (en) 2004-09-22 2007-06-14 트리패스 이미징, 인코포레이티드 Methods and compositions for evaluating breast cancer prognosis
CA2604218A1 (en) 2005-04-20 2006-10-26 Pfizer Limited Pyrazole derivatives as progesterone receptor antagonists
WO2007078599A2 (en) 2005-12-16 2007-07-12 The Board Of Trustees Of The Leland Stanford Junior University Functional arrays for high throughput characterization of gene expression regulatory elements
US20070167971A1 (en) 2006-01-17 2007-07-19 Raymond Huey Devices and methods for promoting the formation of blood clots in esophageal varices
US20070166372A1 (en) 2006-01-19 2007-07-19 Mai De Ltd. Preparation of solid coprecipitates of amorphous valsartan
PT1989217E (en) 2006-02-17 2012-08-30 Janssen Pharmaceutica Nv 11-phosphorous steroid derivatives useful as progesterone receptor modulators
DE102006054535A1 (en) 2006-11-15 2008-05-21 Bayer Schering Pharma Aktiengesellschaft Progesterone receptor antagonist
WO2008128783A2 (en) 2007-04-24 2008-10-30 Dsm Ip Assets B.V. Photochemical process for the preparation of a previtamin d
CA2596204C (en) 2007-08-07 2019-02-26 Historx, Inc. Method and system for determining an optimal dilution of a reagent
WO2009025759A1 (en) 2007-08-17 2009-02-26 Progenics Pharmaceuticals (Nevada), Inc. Tight junction proteins associated with infection and entry of hepatitis c virus (hcv), methods and uses thereof
EP2075246A1 (en) 2007-12-27 2009-07-01 M. J. Institute of Research A process for preparation of amorphous form of atorvastatin hemi-calcium salt
EP2249838A4 (en) 2008-02-05 2012-05-02 Harbor Biosciences Inc Pharmaceutical solid state forms
TWI539953B (en) 2008-04-28 2016-07-01 瑞波若斯治療學公司 Compositions and methods for treating breast cancer
TW201002736A (en) 2008-04-28 2010-01-16 Repros Therapeutics Inc Compositions and methods for treating progesterone-dependent conditions
JP5174952B2 (en) 2009-03-27 2013-04-03 株式会社小松製作所 Fuel saving control device for work vehicle and fuel saving control method for work vehicle
WO2010126590A1 (en) 2009-04-27 2010-11-04 Cold Spring Harbor Laboratory Ptp1b inhibitors
WO2010141485A1 (en) 2009-06-01 2010-12-09 Samuel Waxman Cancer Research Foundation Compositions and methods to induce differentiation and growth inhibition in breast cancer
US9062074B2 (en) 2009-07-15 2015-06-23 Cti Biopharma Corp. (9E)-15-(2-pyrrolidin-1-yl-ethoxy)-7,12,25-trioxa-19,21,24-triaza-tetracyclo[18.3.1.1(2.5).1(14,18)]hexacosa-1(24),2,4,9,14,16,18(26),20,22-nonaene citrate salt
US20110293511A1 (en) 2009-09-29 2011-12-01 Terrance Grant Johns Specific binding proteins and uses thereof
US8261855B2 (en) * 2009-11-11 2012-09-11 Flanders Electric, Ltd. Methods and systems for drilling boreholes
US9295649B2 (en) 2009-12-15 2016-03-29 Bind Therapeutics, Inc. Therapeutic polymeric nanoparticle compositions with high glass transition temperature or high molecular weight copolymers
CN102906103A (en) * 2010-03-22 2013-01-30 利普生物药剂公司 Compositions and methods for non-toxic delivery of antiprogestins
JP6336755B2 (en) 2010-11-12 2018-06-06 ザ ジェネラル ホスピタル コーポレイション Non-coding RNA related to polycomb
WO2012083017A2 (en) 2010-12-16 2012-06-21 Celgene Corporation Controlled release oral dosage forms of poorly soluble drugs and uses thereof
CA2822462A1 (en) 2010-12-20 2012-06-28 The General Hospital Corporation Polycomb-associated non-coding rnas
JP2014510080A (en) 2011-03-09 2014-04-24 セントローズ, エルエルシー Extracellular targeted drug complex
AR085651A1 (en) 2011-03-11 2013-10-16 Celgene Corp METHODS TO TREAT CANCER USING 3- (5-AMINO-2-METIL-4-OXO-4H-QUINAZOLIN-3-IL) -PIPERIDIN-2,6-DIONA
ES2656988T3 (en) 2011-04-15 2018-03-01 J-Pharma Co., Ltd. Biomarker for breast cancer
EP3421496A1 (en) 2011-05-13 2019-01-02 Ganymed Pharmaceuticals GmbH Antibodies for treatment of cancer expressing claudin 6
US20130029953A1 (en) * 2011-07-28 2013-01-31 Klaus Nickisch Progesterone antagonists
JP6092880B2 (en) 2011-10-04 2017-03-08 インヴァイヴィス ファーマスーティカルズ インコーポレーテッドInvivis Pharmaceuticals Inc. Methods and systems for identifying and treating anti-progestin sensitive tumors
MX2014006820A (en) 2011-12-08 2015-05-11 Fundacion Sales Methods and compositions for treating antiprogestin-resistant cancers.
US20130338016A1 (en) 2012-04-17 2013-12-19 Vala Sciences, Inc. Method For Integrated Pathology Diagnosis And Digital Biomarker Pattern Analysis
MX2014013044A (en) 2012-04-27 2015-06-23 Univ Minnesota Breast cancer prognosis, prediction of progesterone receptor subtype and|prediction of response to antiprogestin treatment based on gene expression.
JP2015529808A (en) 2012-07-24 2015-10-08 シーダーズ−サイナイ メディカル センター A novel method for detecting resistance to chemotherapy in patients with lung cancer
EP2931292B1 (en) 2012-12-13 2018-06-13 Warsaw Orthopedic, Inc. Compositions and methods comprising polyethylene glycol and magnesium for treatment of neuronal injury
US20140363425A1 (en) 2013-03-13 2014-12-11 J. Dinny Graham Systems and methods for identifying cancers having activated progesterone receptors
US9096641B2 (en) 2013-06-05 2015-08-04 Evestra, Inc. Imidazolyl progesterone antagonists
CN103483449A (en) 2013-08-20 2014-01-01 东北农业大学 Two ScFv (Single Chain Variable Fragment ) antibodies, encoding genes and application thereof for preparing preparation for treating or preventing infectious bursal disease of chicken
KR20170023771A (en) 2014-04-08 2017-03-06 아르노 테라퓨틱스 인코포레이티드 Systems and methods for identifying progesterone receptor subtypes
MX2017005163A (en) 2014-11-17 2018-01-18 Arno Therapeutics Inc Onapristone extended-release compositions and methods.
AU2016235298B2 (en) 2015-03-23 2021-03-11 Evestra, Inc. Novel cytotoxic agents that preferentially target leukemia inhibitory factor (LIF) for the treatment of malignancies and as new contraceptive agents
BR112018005999A2 (en) 2015-09-25 2019-01-08 Context Biopharma Inc methods for the production of onapristone intermediates
CN108883067B (en) 2015-12-15 2021-03-09 康特斯生物制药公司 Amorphous onapristone composition and preparation method thereof
JP2019513706A (en) 2016-03-21 2019-05-30 コンテキスト・バイオファーマ・インコーポレイテッド Onapristone metabolite composition and method
JP7082065B2 (en) 2016-05-02 2022-06-07 テトラジェネティクス, インコーポレイテッド Anti-Kv1.3 antibody and its production method and usage method
WO2018067198A1 (en) 2016-10-03 2018-04-12 The Regents Of The University Of California Inhibitory antibodies and methods of use thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050085470A1 (en) * 2003-10-09 2005-04-21 Puwen Zhang 6-Amino-1,4-dihydro-benzo[d][1,3] oxazin-2-ones and analogs useful as progesterone receptor modulators
CA2925175A1 (en) * 2007-10-12 2009-04-16 Novartis Ag Compositions comprising sphingosine 1 phosphate (s1p) receptor modulators
US20140271819A1 (en) * 2013-03-12 2014-09-18 Stefan Proniuk Onapristone polymorphic forms and methods of use

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Cottu et al. (Gynecological cancer, 2015 ASCO Annual Meeting I. "Onapristone (ONA) in progesterone receptor (PR)-expressing tumors: Efficacy and biomarker results of a dose-escalation phase 1 study"). (Year: 2015) *

Also Published As

Publication number Publication date
RU2017112748A (en) 2018-12-19
AU2015350241A1 (en) 2017-05-04
US20210220279A1 (en) 2021-07-22
AU2015350241B2 (en) 2021-05-20
CN113559075A (en) 2021-10-29
MX2017005163A (en) 2018-01-18
CA2966753A1 (en) 2016-05-26
JP6655075B2 (en) 2020-02-26
US11672762B2 (en) 2023-06-13
CN106999501A (en) 2017-08-01
HK1246653A1 (en) 2018-09-14
US10786461B2 (en) 2020-09-29
RU2017112748A3 (en) 2019-06-26
AU2021218093A1 (en) 2021-09-09
JP2017537883A (en) 2017-12-21
EP3247362A1 (en) 2017-11-29
WO2016081383A1 (en) 2016-05-26
US20160166583A1 (en) 2016-06-16
KR20170084086A (en) 2017-07-19
CN106999501B (en) 2021-08-13
EP3247362A4 (en) 2018-10-10

Similar Documents

Publication Publication Date Title
US11672762B2 (en) Onapristone extended-release compositions and methods
US20220000857A1 (en) Method of Treating Renal Cell Carcinoma Using N-(4-(6,7-dimethoxyquinolin-4-yloxy)phenyl)-N?-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide, (2S)-hydroxybutanedioate
BR112012033391B1 (en) CONTRACEPTIVE KIT, AND PHARMACEUTICAL COMPOSITION UNDERSTANDING DROSPYRENONE
CN103648487B (en) Drug delivery system
MX2007012081A (en) Tablet containing steroid hormones.
US20230270744A1 (en) Methods of Administering Elagolix
JP2007517899A (en) Treatment of osteoporosis associated with aromatase inhibitor therapy
EP4021420A1 (en) System and method of multi-drug delivery
US20230414618A1 (en) Methods of Administering Elagolix
CN114340625A (en) Method of treating cancer with oral dosage forms of estrogen receptor-alpha inhibitors
WO2022040896A1 (en) System and method of multi-drug delivery
WO2024005634A1 (en) Anti-angiogenic therapy as treatment for benign uterine disorders

Legal Events

Date Code Title Description
AS Assignment

Owner name: ARNO THERAPEUTICS, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZUKIWSKI, ALEXANDER;PRONIUK, STEFAN;SIGNING DATES FROM 20160612 TO 20160706;REEL/FRAME:062327/0183

Owner name: CONTEXT BIOPHARMA INC., UNITED STATES

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ARNO THERAPEUTICS, INC.;REEL/FRAME:062327/0322

Effective date: 20171215

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION