WO2022197293A1 - Procédés d'administration de tésétaxel à des patients présentant une déficience hépatique - Google Patents

Procédés d'administration de tésétaxel à des patients présentant une déficience hépatique Download PDF

Info

Publication number
WO2022197293A1
WO2022197293A1 PCT/US2021/022770 US2021022770W WO2022197293A1 WO 2022197293 A1 WO2022197293 A1 WO 2022197293A1 US 2021022770 W US2021022770 W US 2021022770W WO 2022197293 A1 WO2022197293 A1 WO 2022197293A1
Authority
WO
WIPO (PCT)
Prior art keywords
tesetaxel
dose
patient
administering
hepatic impairment
Prior art date
Application number
PCT/US2021/022770
Other languages
English (en)
Inventor
Thomas Wei
Adam Kamlet
Stew KROLL
Kevin Tang
Original Assignee
Odonate Therapeutics, 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 Odonate Therapeutics, Inc. filed Critical Odonate Therapeutics, Inc.
Priority to PCT/US2021/022770 priority Critical patent/WO2022197293A1/fr
Publication of WO2022197293A1 publication Critical patent/WO2022197293A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • Breast cancer is a heterogeneous disease comprised of several molecular subtypes, which are commonly grouped into clinical subtypes based on receptor status.
  • Receptors that are assessed in standard clinical practice include the estrogen receptor (ER) and the progesterone receptor (PR), which are collectively referred to as the hormone receptors (HR), and human epidermal growth factor receptor 2 (HER2).
  • ER estrogen receptor
  • PR progesterone receptor
  • HR hormone receptor
  • HER2 human epidermal growth factor receptor 2
  • Breast cancers generally are categorized by the presence or absence of these receptors. The most common form of breast cancer is HR-positive and HER2-negative, accounting for approximately 64% of newly diagnosed cases.
  • HER2- positive breast cancer and triple negative breast cancer (TNBC) which lacks all 3 receptors, are less common, accounting for approximately 13% and 11% of breast cancers, respectively.
  • Breast cancer typically is staged (Stage 0-IV) based on the size of the tumor, whether or not the tumor is invasive, whether or not the cancer is in the lymph nodes and whether or not the cancer has spread (metastasized) to other parts of the body beyond the breast, most often the bones, lungs, liver or brain.
  • the prognosis for women with locally advanced or metastatic breast cancer (LA/MBC) remains poor; the 5-year survival rate for metastatic disease is about 22%, making this an area of continued, high unmet medical need.
  • Tesetaxel is a novel, orally administered taxane.
  • Taxanes are an established class of anticancer agents that are broadly used in various cancers, including breast cancer.
  • the primary pharmacologic mechanism of tesetaxel is to stabilize cellular microtubule formation (inhibit tubulin depolymerization) in rapidly dividing cells, leading to arrest of unscheduled cell division at the Oh/M phase of the cell cycle and cell death.
  • Tesetaxel has several pharmacologic properties that make it unique among taxanes:
  • Tesetaxel is a capsule for oral administration with a low pill burden
  • Tesetaxel has a long ( ⁇ 8-day) terminal plasma half-life (ti/2) in humans, enabling the maintenance of adequate drug levels with relatively infrequent dosing;
  • Tesetaxel’ s formulation does not contain polyoxyethylated castor oil or polysorbate 80, solubilizing agents contained in other taxane formulations known to cause hypersensitivity reactions;
  • Tesetaxel has been shown to retain activity against taxane-resistant tumors in nonclinical studies.
  • tesetaxel includes the addition of two nitrogen- containing functional groups. Tesetaxel is chemically designed to: (1) not be substantially effluxed by the P-gly coprotein (P-gp) pump, with the intent of retaining activity against chemotherapy-resistant tumor cells; (2) have high oral bioavailability, which requires that a drug substantially survive first-pass metabolism in the liver; (3) have high solubility; and (4) have a long ti/2 in humans.
  • P-gp P-gly coprotein
  • the high oral bioavailability of tesetaxel has been attributed to various causes, including the fact that tesetaxel is a poor substrate for P-gp and CYP3A and thus is absorbed orally and survives first-pass liver metabolism.
  • patients with hepatic impairment may exhibit altered tesetaxel pharmacokinetics and/or altered sensitivity to the side effects of tesetaxel. It could not have been predicted whether tesetaxel could be safely administered to patients with hepatic impairment and, if so, whether any dosage modification would be required.
  • the present disclosure relates generally to the discovery that patients with hepatic impairment should receive a reduced dose of tesetaxel.
  • the present disclosure provides a method for safely administering tesetaxel to a patient diagnosed with hepatic impairment, comprising administering a first dose of tesetaxel to the patient on day 1 of a first 21 -day cycle, wherein the first dose of tesetaxel is 20- 80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the present disclosure provides a method for safely treating cancer in a patient diagnosed with hepatic impairment, comprising administering a first dose of tesetaxel to the patient on day 1 of a first 21 -day cycle, wherein the first dose of tesetaxel is 20-80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the hepatic impairment is moderate hepatic impairment.
  • the patient has been diagnosed with a cancer, such as breast cancer.
  • the present disclosure provides in various aspects methods for safely administering tesetaxel to patients diagnosed with hepatic impairment.
  • tesetaxel dosing methods discussed herein apply to any patient with hepatic impairment.
  • the population of affected patients thus includes, e.g., those who are diagnosed with hepatic impairment during a tesetaxel treatment cycle, as well as those who were diagnosed with hepatic impairment before beginning tesetaxel therapy.
  • the dose adjustments continue until the hepatic impairment in the patient is no longer effective to substantially alter the pharmacokinetics of tesetaxel.
  • the present disclosure provides a method for safely administering tesetaxel to a patient diagnosed with hepatic impairment, comprising administering a first dose of tesetaxel to the patient on day 1 of a first 21 -day cycle, wherein the first dose of tesetaxel is 20- 80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the present disclosure provides a method for safely treating cancer in a patient diagnosed with hepatic impairment, comprising administering a first dose of tesetaxel to the patient on day 1 of a first 21 -day cycle, wherein the first dose of tesetaxel is 20-80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the first dose of tesetaxel is 60-80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment, such as 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the first dose of tesetaxel comprises 6-24 mg/m 2 tesetaxel, such as 6, 9, 12, 15, 18, or 21 mg/m 2 tesetaxel.
  • the first dose of tesetaxel comprises 18 or 21 mg/m 2 tesetaxel.
  • the method further comprises administering a second dose of tesetaxel to the patient on day 1 of a second 21 -day cycle, wherein the second dose of tesetaxel is 20-80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the second dose of tesetaxel is 60-80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment, such as 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the second dose comprises 13.5, 12, 10.5, or 9 mg/m 2 tesetaxel.
  • the second dose of tesetaxel comprises 6-24 mg/m 2 tesetaxel, such as 6, 9, 12, 15, 18, or 21 mg/m 2 tesetaxel.
  • the first dose of tesetaxel comprises 18 or 21 mg/m 2 tesetaxel.
  • the first dose of tesetaxel referred to herein may not be the absolute first dose of tesetaxel administered to the patient.
  • the patient may have previously received tesetaxel at a higher dose, or may have previously been receiving tesetaxel.
  • the present disclosure provides a method for safely administering tesetaxel to a patient diagnosed with hepatic impairment, comprising administering a first dose of tesetaxel to the patient on day 1 of a first 21 -day cycle, wherein the first dose is an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment, diagnosing the patient with hepatic impairment, and administering a second dose of tesetaxel to the patient on day 1 of a second 21 -day cycle, wherein the second dose of tesetaxel is 20-80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • This treatment protocol is useful, for example, when a patient is diagnosed with hepatic impairment after administration of the first dose of tesetaxel but before the second dose of tesetaxel.
  • the second dose of tesetaxel is 60-80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment, such as 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80% of the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment.
  • the second dose comprises 13.5, 12, 10.5, or 9 mg/m 2 tesetaxel.
  • the second dose of tesetaxel comprises 6-24 mg/m 2 tesetaxel, such as 6, 9, 12, 15, 18, or 21 mg/m 2 tesetaxel.
  • the first dose of tesetaxel comprises 18 or 21 mg/m 2 tesetaxel.
  • first 21 -day cycle and/or a second 21 -day cycle does not necessarily reflect the absolute first 21 -day cycle and the absolute second 21 -day cycle, respectively, but may instead refer to any two consecutive 21 -day cycles.
  • a first 21 -day cycle may correspond to a patient’s third absolute 21 -day cycle
  • a second 21 -day cycle may correspond to the patient’s fourth absolute 21 -day cycle.
  • a first 21 -day cycle may correspond to a patient’s third absolute 21 -day cycle of tesetaxel administration but which is the first 21 -day cycle that is characterized by a reduced dose of tesetaxel (e.g., the first 21 -day cycle that is characterized by a dose of tesetaxel that is 20-80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment).
  • a second 21 -day cycle may correspond to a patient’s fifth absolute 21 -day cycle of tesetaxel administration but the first 21 -day cycle that is characterized by a reduced dose of tesetaxel (e.g., the first 21-day cycle that is characterized by a dose of tesetaxel that is 20-80% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment).
  • the first dose of tesetaxel may comprise less than 27 mg/m 2 of tesetaxel, such as 1-24 mg/m 2 tesetaxel, such as 6, 9, 12, 15, 18, 21, or 24 mg/m 2 tesetaxel.
  • the first dose may comprise 27 mg/m 2 of tesetaxel.
  • the second dose of tesetaxel may comprise less than 27 mg/m 2 of tesetaxel, such as 6-24 mg/m 2 tesetaxel, such as 6, 9, 12, 15, 15, 18, 21, or 24 mg/m 2 tesetaxel.
  • the first dose and the second dose may be equal or different. In certain embodiments, the first dose and the second dose are equal.
  • the method further comprises administering an antiemetic agent, such as dexamethasone, ondansetron, dolasetron, palonosetron, aprepitant, or rolapitant, or a combination of any of the foregoing.
  • an antiemetic agent such as dexamethasone, ondansetron, dolasetron, palonosetron, aprepitant, or rolapitant, or a combination of any of the foregoing.
  • the hepatic impairment is moderate hepatic impairment.
  • normal hepatic function and moderate hepatic impairment are defined consistent with the definitions of the National Cancer Institute (NCI) Organ Dysfunction Working Group (ODWG) criteria.
  • Normal hepatic function is characterized by total bilirubin (TBIL) ⁇ upper limit of normal (ULN) and aspartate aminotransferase (AST) ⁇ ULN.
  • Moderate hepatic impairment is characterized by 1.5 x ULN ⁇ TBIL ⁇ 3 x ULN and any AST.
  • the patient has been diagnosed with a cancer.
  • the cancer is breast cancer.
  • the cancer is locally advanced breast cancer.
  • the cancer is metastatic breast cancer.
  • the breast cancer is hormone receptor (HR)-positive.
  • the patient has previously received endocrine therapy.
  • the breast cancer is estrogen receptor (ER)-positive.
  • the breast cancer is ER-negative.
  • the breast cancer is progesterone receptor (PR)-positive.
  • the breast cancer is PR-negative.
  • the breast cancer is HER2-negative.
  • the breast cancer is HER2-positive.
  • the breast cancer is HR-positive and HER2-negative.
  • the breast cancer is HR negative and HER2-negative.
  • the method comprises repeating the 21-day cycle at least once (e.g., a first 21-day cycle and a second 21 -day cycle). In some embodiments, the method comprises repeating the 21- day cycle until the cancer progresses or until unacceptable toxicity is observed.
  • the tesetaxel may also be conjointly administered with other suitable therapeutic agents.
  • tesetaxel and capecitabine may be effectively used in conjoint therapy, as described in International Patent Application PCT/US18/35653, published as WO 2018/223029, which is hereby incorporated by reference herein in its entirety.
  • the combination can provide greater efficacy than capecitabine alone.
  • the methods disclosed herein may result in longer progression-free survival (PFS), longer survival, a greater treatment response, a longer duration of response and/or better disease control.
  • PFS progression-free survival
  • the combination is more efficacious as administration of capecitabine alone (e.g., at a dose of 2,500 mg/m 2 or 2,000 mg/m 2 daily for 14 consecutive days of a 21 -day cycle), and in some embodiments, the combination is more efficacious and has a more tolerable safety profile. More tolerable treatment regimens, such as those disclosed herein, are more likely to be continued by patients, and thus may be more likely to be effective.
  • the method comprises administering a therapeutically effective amount of tesetaxel and a therapeutically effective amount of capecitabine conjointly.
  • the therapeutically effective amount of tesetaxel is the dose of tesetaxel described herein (e.g., 40-60% of an indicated tesetaxel dose for a patient not diagnosed with hepatic impairment).
  • the therapeutically effective amount of tesetaxel is 27 mg/m 2 .
  • the method further comprises administering capecitabine daily starting on day 1 of the 21 -day cycle for 14 consecutive 24-hour periods.
  • any suitable dose of capecitabine may be used.
  • the daily dosage may be divided into a number of smaller, divided doses, such as 2, 3, 4, 5, 6 or more divided doses.
  • the daily dosage of capecitabine is divided into two divided doses.
  • a daily dosage regimen may begin with a partial dose on the first day and end with a partial dose on the last day, such that the daily dosage is delivered in a number of 24-hour periods, which may or may not correspond to calendar days.
  • dosing of capecitabine is alternately discussed herein in terms of the total daily dosage (i.e., the total amount administered in a day or in a 24-hour period) or in terms of divided doses (i.e., the individual doses administered over the course of a day or a 24- hour period that combine to meet the total daily dosage).
  • capecitabine is administered at twice-daily intervals (i.e., 2 times per 24-hour period) for a period of time, such as for 14 consecutive 24-hour periods.
  • a first dose of capecitabine is administered on day 1
  • subsequent doses are administered at twice-daily intervals with a final dose administered on day 15.
  • capecitabine is administered twice daily for 14 consecutive calendar days (i.e., 2 doses of capecitabine are administered on each of days 1-14).
  • reference to a number of “daily dosages” of capecitabine herein refers to administering capecitabine for that number of 24-hour periods and encompasses administering capecitabine for that number of calendar days.
  • administering a therapeutically effective amount of capecitabine comprises administering capecitabine twice daily on days 1-14 of the 21 -day cycle. In some embodiments, the method comprises administering a therapeutically effective amount of capecitabine in 28 doses at twice-daily intervals beginning on day 1 of the 21 -day cycle. In some embodiments, the method comprises administering a first dose of capecitabine on day 1 of the 21 -day cycle and administering a final 28 th dose on day 15 of the 21 -day cycle.
  • administering a therapeutically effective amount of capecitabine comprises administering a first dose of capecitabine after noon (e.g., in the evening) of day 1 of the 21 -day cycle and administering a final 28 th dose before noon ( e.g ., in the morning) on day 15 of the 21- day cycle.
  • administering a therapeutically effective amount of capecitabine comprises administering 14 daily dosages of 300-2,000 mg/m 2 (such as 1,000-1,800 mg/m 2 ) of capecitabine beginning on day 1 of the 21 -day cycle. In some embodiments, administering a therapeutically effective amount of capecitabine comprises administering 14 daily dosages of 1,650 mg/m 2 of capecitabine beginning on day 1 of the 21 -day cycle.
  • administering a therapeutically effective amount of capecitabine comprises administering 825 mg/m 2 of capecitabine at twice-daily intervals for 14 consecutive 24-hour periods beginning on day 1 of the 21 -day cycle. In some such embodiments, administering a therapeutically effective amount of capecitabine comprises administering 825 mg/m 2 of capecitabine twice daily on days 1-14 of the 21-day cycle. In other such embodiments, administering capecitabine comprises administering a first dose of 825 mg/m 2 of capecitabine on day 1, administering subsequent doses of 825 mg/m 2 of capecitabine at twice-daily intervals, and administering a final dose of 825 mg/m 2 of capecitabine on day 15.
  • administering a therapeutically effective amount of capecitabine comprises administering 14 daily dosages of 1,750 mg/m 2 of capecitabine beginning on day 1 of the 21 -day cycle. In some embodiments, administering a therapeutically effective amount of capecitabine comprises administering 875 mg/m 2 of capecitabine at twice-daily intervals for 14 consecutive 24-hour periods beginning on day 1 of the 21 -day cycle. In some such embodiments, administering a therapeutically effective amount of capecitabine comprises administering 875 mg/m 2 twice daily on days 1-14 of the 21 -day cycle.
  • administering a therapeutically effective amount of capecitabine comprises administering a first dose of 875 mg/m 2 on day 1, administering subsequent doses of 875 mg/m 2 of capecitabine at twice-daily intervals, and administering a final dose of 875 mg/m 2 of capecitabine on day 15.
  • administering a therapeutically effective amount of capecitabine comprises administering 28 doses of 150-1,000 mg/m 2 of capecitabine at twice-daily intervals. In some embodiments, administering a therapeutically effective amount of capecitabine comprises administering 150-1,000 mg/m 2 of capecitabine at twice-daily intervals for 14 consecutive 24- hour periods. In some such embodiments, administering a therapeutically effective amount of capecitabine comprises administering 150-1,000 mg/m 2 twice daily on days 1-14 of the 21-day cycle.
  • administering a therapeutically effective amount of capecitabine comprises administering a first dose of 150-1,000 mg/m 2 of capecitabine on day 1, and administering subsequent doses of 150-1,000 mg/m 2 of capecitabine at twice-daily intervals and concluding by administering a final dose of 150-1,000 mg/m 2 of capecitabine on day 15.
  • administering a therapeutically effective amount of capecitabine comprises administering 28 doses of 150-1,000 mg/m 2 of capecitabine at twice-daily intervals beginning with the first dose on day 1 of the 21 -day cycle and ending with the 28 th dose on day 15 of the 21 -day cycle. In some embodiments, administering a therapeutically effective amount of capecitabine comprises administering 28 doses of 825 mg/m 2 of capecitabine at twice-daily intervals.
  • administering a therapeutically effective amount of capecitabine comprises administering 28 doses of 825 mg/m 2 of capecitabine at twice-daily intervals beginning with the first dose on day 1 of the 21 -day cycle and ending with the 28 th dose on day 15 of the 21 -day cycle. In some embodiments, administering a therapeutically effective amount of capecitabine comprises administering 28 doses of 875 mg/m 2 of capecitabine at twice-daily intervals.
  • administering a therapeutically effective amount of capecitabine comprises administering 28 doses of 875 mg/m 2 of capecitabine at twice-daily intervals beginning with the first dose on day 1 of the 21 -day cycle and ending with the 28 th dose on day 15 of the 21 -day cycle.
  • the patient has previously been treated with a taxane. In some embodiments, the patient has previously been treated with a taxane in the neoadjuvant or adjuvant setting. In some embodiments, the taxane is paclitaxel, docetaxel or albumin-bound (nab) paclitaxel. In some embodiments, the patient has not previously been treated with a taxane.
  • the primary cancer is breast cancer, such as MBC or LA/MBC.
  • the breast cancer is locally advanced breast cancer.
  • the breast cancer is metastatic breast cancer.
  • Metastatic breast cancer may include, but is not limited to, breast cancer that has spread to the central nervous system (CNS). Treatment of CNS cancers, including CNS cancers that are metastases of breast cancer, is described in PCT/US2019/049642, filed September 5, 2019 and published as WO 2020/081165, which is hereby incorporated by reference herein in its entirety. Tesetaxel is brain-penetrant; that is, it crosses the blood-brain barrier.
  • tesetaxel unlike docetaxel and paclitaxel, may be conveniently utilized in the treatment of tumors of the CNS, such as brain tumors.
  • the structures of tesetaxel, docetaxel and paclitaxel are shown below:
  • the breast cancer is HR-positive, such as ER-positive or PR- positive. In some embodiments, the patient has previously received endocrine therapy. In some embodiments, the breast cancer is HER2-negative. In some embodiments, the breast cancer is HR-positive and HER2-negative. In some embodiments, the breast cancer is HR-negative (i.e., ER-negative and PR-negative) and HER2-negative. In some embodiments, the breast cancer is HER2-positive.
  • a therapeutic that “prevents” a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample, or delays the onset or reduces the severity of one or more symptoms of the disorder or condition relative to the untreated control sample.
  • prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population ( e.g ., by a statistically and/or clinically significant amount).
  • treating includes prophylactic and/or therapeutic treatments.
  • prophylactic or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal), then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition); whereas, if it is administered after manifestation of the unwanted condition, the treatment is therapeutic (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • the unwanted condition e.g., disease or other unwanted state of the host animal
  • terapéuticaally effective amount means the concentration of a compound that is sufficient to elicit the desired therapeutic effect.
  • the phrases “conjoint administration,” “administered conjointly,” “receiving conjoint treatment,” and grammatical variations refer to any form of administration of two or more different therapeutic compounds such that the second compound is administered while the previously administered therapeutic compound is still therapeutically effective in the body (e.g ., the two compounds are simultaneously therapeutically effective in the patient, which may include synergistic effects of the two compounds).
  • the different therapeutic compounds can be administered either in the same formulation or in a separate formulation, either concomitantly (i.e., at substantially the same time) or sequentially (i.e., with one compound administered first and the other compound administered at a later time).
  • the different therapeutic compounds can be administered within one hour, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours, 7 days, 14 days or 15 days of one another, or wherein the different therapeutic compounds are administered within the same treatment cycle as one another.
  • an individual who receives such treatment can benefit from a combined effect of different therapeutic compounds.
  • prodrug is intended to encompass compounds which, under physiologic conditions, are converted into the therapeutically active agents of the present invention.
  • a common method for making a prodrug is to include one or more selected moieties that are hydrolyzed under physiologic conditions to reveal the desired molecule.
  • the prodrug is converted by an enzymatic activity of the host animal.
  • esters or carbonates e.g., esters or carbonates of alcohols or carboxylic acids
  • some or all of the compounds of the invention in a formulation represented above can be replaced with the corresponding suitable prodrug (e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate or carboxylic acid present in the parent compound is presented as an ester).
  • Tesetaxel is a taxane having the following structure:
  • Tesetaxel and its preparation are described in U.S. Patent No. 6,677,456, which is incorporated by reference in its entirety.
  • Various crystal forms of tesetaxel are described in U.S. Patent No. 7,410,980, which is hereby incorporated by reference in its entirety.
  • An “indicated tesetaxel dose for a patient not diagnosed with hepatic impairment” is the recited tesetaxel dose on a prescribing information label.
  • the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment is 6-27 mg/m 2 , preferably 18- 27 mg/m 2 tesetaxel.
  • the indicated tesetaxel dose for a patient not diagnosed with hepatic impairment is 18, 21, 24, or 27 mg/m 2 tesetaxel, preferably 27 mg/m 2 tesetaxel.
  • compositions and methods of the present invention may be utilized to treat an individual in need thereof.
  • the individual is a human.
  • the composition or the compound is preferably administered as a pharmaceutical composition comprising, for example, a compound of the invention and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, oils such as olive oil or injectable organic esters.
  • the aqueous solution is pyrogen-free, or substantially pyrogen-free.
  • the excipients can be chosen, for example, to effect delayed release of an agent or to selectively target one or more cells, tissues or organs.
  • the pharmaceutical composition can be in dosage unit form such as a tablet, capsule (including sprinkle capsule and gelatin capsule), granule, lyophile for reconstitution, powder, solution, syrup, suppository, injection or the like.
  • the composition can also be present in a transdermal delivery system (e.g ., a skin patch).
  • the composition can also be present in a solution suitable for topical administration, such as an eye drop.
  • a pharmaceutically acceptable carrier can contain physiologically acceptable agents that act, for example, to stabilize, to increase solubility or to increase the absorption of a compound such as a compound of the invention.
  • physiologically acceptable agents include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients.
  • the choice of a pharmaceutically acceptable carrier, including a physiologically acceptable agent depends, for example, on the route of administration of the composition.
  • the preparation or pharmaceutical composition can be a self-emulsifying drug delivery system or a self- microemulsifying drug delivery system.
  • the pharmaceutical composition also can be a liposome or other polymer matrix, which can have incorporated therein, for example, a compound of the invention.
  • Liposomes for example, which comprise phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications, commensurate with a reasonable benefit-risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials that can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide;
  • a pharmaceutical composition can be administered to a subject by any of a number of routes of administration including, for example, orally (e.g ., as drenches in aqueous or non-aqueous solutions or suspensions, tablets, capsules [including sprinkle capsules and gelatin capsules], boluses, powders, granules or pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (e.g., as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (e.g., as a patch applied to the skin); and topically (e.g., as a cream, ointment or spray applied to the skin, or as an eye drop).
  • routes of administration including, for example, orally (e.g
  • the compound may also be formulated for inhalation.
  • a compound may be simply dissolved or suspended in sterile water. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973, 5,763,493, 5,731,000, 5,541,231, 5,427,798, 5,358,970 and 4,172,896, as well as in patents cited therein.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated and the particular mode of administration.
  • the amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound that produces a therapeutic effect. Generally, out of 100 percent, this amount will range from about 1 percent to about 99 percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.
  • Methods of preparing these formulations or compositions include the step of bringing into association an active compound, such as a compound of the invention, with the carrier and, optionally, one or more accessory ingredients.
  • an active compound such as a compound of the invention
  • the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • Formulations of the invention suitable for oral administration may be in the form of capsules (including sprinkle capsules and gelatin capsules), cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), lyophile, powders, granules or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in-water or water- in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient.
  • Compositions or compounds may also be administered as a bolus, electuary or paste.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as
  • absorbents such as kaolin and bentonite clay
  • lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof
  • complexing agents such as, modified and unmodified cyclodextrins
  • coloring agents such as, modified and unmodified cyclodextrins
  • capsules including sprinkle capsules and gelatin capsules
  • the pharmaceutical compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high-molecular-weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared using a binder (e.g ., gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g., sodium starch glycolate or cross-linked sodium carboxymethyl cellulose) or surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine, a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions that can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art, such
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.
  • Formulations of the pharmaceutical compositions for rectal, vaginal or urethral administration may be presented as a suppository, which may be prepared by mixing one or more active compounds with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • Formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash, oral spray or oral ointment.
  • compositions can be formulated for delivery via a catheter, stent, wire or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum or intestine.
  • Formulations that are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.
  • Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers or propellants that may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Powders and sprays can contain, in addition to an active compound, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances.
  • Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body.
  • dosage forms can be made by dissolving or dispersing the active compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
  • Ophthalmic formulations eye ointments, powders, solutions and the like are also contemplated as being within the scope of this invention.
  • Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056, 2005/0059744, 2005/0031697 and 2005/004074 and U.S. Patent No. 6,583,124, the contents of which are incorporated herein by reference.
  • liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatible with such fluids.
  • a preferred route of administration is local administration (e.g ., topical administration, such as eye drops, or administration via an implant).
  • parenteral administration and “administered parenterally” as used herein mean modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.
  • compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like) and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents that delay absorption such as aluminum monostearate and gelatin.
  • the absorption of the drug in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue.
  • active compounds can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5%, more preferably, 0.5 to 90%, of active ingredient in combination with a pharmaceutically acceptable carrier.
  • Methods of introduction may also be provided by rechargeable or biodegradable devices.
  • Various slow-release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinaceous biopharmaceuticals.
  • a variety of biocompatible polymers including hydrogels), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a compound at a particular target site.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors, including the activity of the particular compound or combination of compounds employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound(s) being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound(s) employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a suitable daily dosage of an active compound used in the compositions and methods of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • the effective daily dosage of the active compound may be administered as 1 , 2, 3, 4, 5, 6 or more sub-doses (or divided doses) administered separately at appropriate intervals throughout a day, optionally, in unit dosage forms.
  • an active compound may be administered one or two times daily on the days on which it is administered.
  • the methods of the invention may be used alone or the compounds administered may be used conjointly with another type of therapeutic agent.
  • contemplated salts of the invention include, but are not limited to, alkyl, dialkyl, trialkyl or tetra- alkyl ammonium salts.
  • contemplated salts of the invention include, but are not limited to, L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N-methylglucamine, hydrabamine, lH-imidazole, lithium, L-lysine, magnesium, 4-(2- hydroxyethyl (morpholine, piperazine, potassium, 1 -(2-hydroxy ethyl)pyrrolidine, sodium, triethanolamine, tromethamine and zinc salts.
  • contemplated salts of the invention include, but are not limited to, Na, Ca, K, Mg, Zn or other metal salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, 1 -hydroxyl- naphthoic acid, 2,2-dichloroacetic acid, 2-hydroxyethanesulfonic acid, 2-oxoglutaric acid, 4- acetamidobenzoic acid, 4-aminosalicylic acid, acetic acid, adipic acid, L-ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, (+)-camphoric acid, (+)-camphor-10-sulfonic acid, capric acid (decanoic acid), caproic acid (hexanoic acid), caprylic acid (octanoic acid), carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, e
  • the pharmaceutically acceptable acid-addition salts can also exist as various solvates, such as with water, methanol, ethanol, dimethylformamide, and the like. Mixtures of such solvates can also be prepared.
  • the source of such solvate can be from the solvent of crystallization, inherent in the solvent of preparation or crystallization, or adventitious to such solvent.
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: (1) water-soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal- chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water-soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT),
  • Patients are divided into those with normal hepatic function (Cohort 1) and those with moderate hepatic impairment (Cohort 2). Patients undergo screening 7 days prior to the first Study treatment.
  • TBIL and AST are measured in two blood samples taken at least 72 hours apart with one sample obtained within 72 hours prior to Cycle 1, Day 1.
  • the second blood sample may be drawn and analyzed a t alocal lab and does not need to be drawn at the study site lab.
  • Hepatic function classification according to NCI-ODWG criteria must remain the same for the two samples for the patient to be eligible for enrollment.
  • Patients in Cohort 1 are treated with tesetaxel (27 mg/m 2 ) orally once on the morning of Day 1 of a 21 -day cycle.
  • Patients in Cohort 2 are treated with tesetaxel (21 mg/m 2 ) orally once on the morning of Day 1 of a 21 -day cycle.
  • Patients are treated for one 21 -day cycle and may continue onto an optional treatment extension, wherein tesetaxel capsules are administered orally on Day 1 of each 21 -day cycle at the same dose used in Cycle 1 or a reduced dose based on tolerability. Treatment continues until disease progresses or unacceptable toxicity is observed in the patient.
  • the primary objective of the study is to assess the effect of moderate hepatic impairment on the pharmacokinetics of tesetaxel and tesetaxel metabolites.
  • the secondary objective of the study is to assess safety and tolerability of tesetaxel in patients with moderate hepatic impairment.
  • the primary endpoint of the study is the ratio of the geometric mean Cmax for unbound tesetaxel for Cohort 2 compared to Cohort 1 and the ratio of the geometric mean AUCo-t of unbound tesetaxel for Cohort 2 compared to Cohort 1.
  • Secondary endpoints include the ratio of the geometric mean Cmax of tesetaxel for Cohort 2 compared to Cohort 1 and the ratio of the geometric mean AUCo-t of tesetaxel for Cohort 2 compared to Cohort 1, the ratio of the geometric mean Cmax of unbound tesetaxel metabolites (t-butyl-OH tesetaxel, 5-pyridine-OH tesetaxel, and N-desmethyl tesetaxel) for Cohort 2 compared to Cohort 1 and the ratio of the geometric mean AUCo-t of unbound tesetaxel metabolites (t-butyl-OH tesetaxel, 5-pyridine-OH tesetaxel, and N-desmethyl tesetaxel) for Cohort 2 compared to Cohort 1, the ratio of the geometric mean Cmax of tesetaxel metabolites (t-butyl-OH tesetaxel, 5-pyridine-OH tesetaxel, and N-desmethyl tese

Abstract

La présente invention concerne des procédés d'administration de tésétaxel à un patient atteint d'un cancer, tel que le cancer du sein, comprenant l'administration de tésétaxel à un patient atteint d'une déficience hépatique.
PCT/US2021/022770 2021-03-17 2021-03-17 Procédés d'administration de tésétaxel à des patients présentant une déficience hépatique WO2022197293A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2021/022770 WO2022197293A1 (fr) 2021-03-17 2021-03-17 Procédés d'administration de tésétaxel à des patients présentant une déficience hépatique

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2021/022770 WO2022197293A1 (fr) 2021-03-17 2021-03-17 Procédés d'administration de tésétaxel à des patients présentant une déficience hépatique

Publications (1)

Publication Number Publication Date
WO2022197293A1 true WO2022197293A1 (fr) 2022-09-22

Family

ID=83320653

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/022770 WO2022197293A1 (fr) 2021-03-17 2021-03-17 Procédés d'administration de tésétaxel à des patients présentant une déficience hépatique

Country Status (1)

Country Link
WO (1) WO2022197293A1 (fr)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021034335A1 (fr) * 2019-08-16 2021-02-25 Odonate Therapeutics, Inc. Procédés d'administration de tésétaxel avec des glucocorticoïdes qui sont des inducteurs de cyp3a4

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021034335A1 (fr) * 2019-08-16 2021-02-25 Odonate Therapeutics, Inc. Procédés d'administration de tésétaxel avec des glucocorticoïdes qui sont des inducteurs de cyp3a4

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Tesetaxel | C46H60FN3O13 - PubChem", INTERNET ARCHIVE WAYBACK MACHINE, 16 August 2019 (2019-08-16), pages 1 - 16, XP055975447, Retrieved from the Internet <URL:https://web.archive.org/web/20190816162638/https://pubchem.ncbi.nlm.nih.gov/compound/Tesetaxel> [retrieved on 20221027] *
BAAS PAUL; SZCZESNA A; ALBERT I; MILANOWSKI J; JUHÁSZ E; SZTANCSIK Z; VON PAWEL J; OYAMA R; BURGERS S: "Phase I/II study of a 3 weekly oral taxane (DJ-927) in patients with recurrent, advanced non-small cell lung cancer.", JOURNAL OF THORACIC ONCOLOGY, LIPPINCOTT WILLIAMS & WILKINS,, US, vol. 3, no. 7, 30 June 2008 (2008-06-30), US , pages 745 - 750, XP009539094, ISSN: 1556-1380, DOI: 10.1097/JTO.0b013e31817c73ff *
HOSSAIN RAJIB, JANNAT RUMMATUL ANNESHA, CHADNI SHADIA AKTER, DAS CHANDON, HAFIZ SM, HASSAN, ISLAM MOHAMMAD TOREQUL: "DOSAGE ADJUSTMENT IN HEPATIC PATIENTS: A LITERATURE-BASED REVIEW", PHARMACOLOGYONLINE, 30 August 2019 (2019-08-30), pages 113 - 137, XP055975443, Retrieved from the Internet <URL:https://pharmacologyonline.silae.it/files/newsletter/2019/vol2/PhOL_2019_2_NL011_Hossain.pdf> [retrieved on 20221027] *
ROGER K. VERBEECK: "Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction", EUROPEAN JOURNAL OF CLINICAL PHARMACOLOGY, SPRINGER, BERLIN, DE, vol. 64, no. 12, 2 September 2008 (2008-09-02), Berlin, DE , pages 1147 - 1161, XP019653453, ISSN: 1432-1041, DOI: 10.1007/s00228-008-0553-z *
SYN NICHOLAS LI‐XUN, WANG LINGZHI, WONG ANDREA LI‐ANN, SOE MU‐YAR, CHUAH BENJAMIN, CHAN DANIEL, TAN SING‐HUANG, SOO ROSS ANDREW, L: "Dose modifications in Asian cancer patients with hepatic dysfunction receiving weekly docetaxel: A prospective pharmacokinetic and safety study", CANCER SCIENCE, JAPANESE CANCER ASSOCIATION, TOKYO, JP, vol. 107, no. 2, 8 February 2016 (2016-02-08), JP , pages 173 - 180, XP055975440, ISSN: 1347-9032, DOI: 10.1111/cas.12856 *

Similar Documents

Publication Publication Date Title
CN110420211A (zh) 治疗炎症、自身免疫性疾病和疼痛的方法
US20070072938A1 (en) Dronabinol treatment of delayed chemotherapy-induced nausea and vomiting
US20220072011A1 (en) Treatments of accumulated fat with deoxycholic acid and salts thereof
CN115990181A (zh) 4-苯胺基-3-氰基喹啉和卡培他滨的抗肿瘤组合
JP2000516198A (ja) セロトニンの再取り込み阻害剤の胃腸に対する作用を克服するための5ht4レセプター拮抗剤の使用
AU2014248928B2 (en) Onapristone polymorphic forms and methods of use
WO2021034335A1 (fr) Procédés d&#39;administration de tésétaxel avec des glucocorticoïdes qui sont des inducteurs de cyp3a4
CN104994877A (zh) 用于癌症治疗的反式-克罗米芬
AU2018275122A1 (en) Dosing schedule for tesetaxel and capecitabine
EP2106792A1 (fr) Utilisation d&#39;une combinaison d&#39;udénafil et d&#39;alfuzosine ou oxybutynine pour le traitement de la vessie hyperactive
US11179365B2 (en) Pharmaceutical combination comprising LSZ102 and ribociclib
WO2022197293A1 (fr) Procédés d&#39;administration de tésétaxel à des patients présentant une déficience hépatique
WO2022197290A1 (fr) Procédés d&#39;administration de tésétaxel avec des inhibiteurs de cyp3a4
JPH10512270A (ja) 肥満の治療又は予防のための医薬製剤の製造のための3,4−ジフェニル・クロマンの使用
WO2020081165A1 (fr) Méthodes de traitement de tumeurs du système nerveux central au moyen de tesetaxel
WO2011044105A1 (fr) Traitement avec des agonistes cholinergiques
CZ211897A3 (en) Use of 3,4-diphenylchromans for preparing pharmaceutical preparations used for dilation of vessels
JP2000514441A (ja) 月経前症候群の1又は複数の症状を抑制するための医薬組成物の製造のための3,4―ジフェニルクロマンの使用
WO2009056256A1 (fr) Utilisation d&#39;acétate de mégestrol à solubilité améliorée pour traiter la cachexie cancéreuse
TW202114717A (zh) 含有對硼苯基丙胺酸之注射液劑
TW202333675A (zh) 用於治療癌症之組合療法之用途
WO2020205608A1 (fr) Utilisations d&#39;antagonistes du récepteur des androgènes et d&#39;inhibiteurs de la voie jnk et compositions pharmaceutiques associées
WO2015001359A1 (fr) Combinaison de médicaments et son utilisation dans la thérapie de l&#39;obésité

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21931888

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21931888

Country of ref document: EP

Kind code of ref document: A1