WO2017164887A1 - Pulse dosing regimen and methods for treatment - Google Patents

Pulse dosing regimen and methods for treatment Download PDF

Info

Publication number
WO2017164887A1
WO2017164887A1 PCT/US2016/024232 US2016024232W WO2017164887A1 WO 2017164887 A1 WO2017164887 A1 WO 2017164887A1 US 2016024232 W US2016024232 W US 2016024232W WO 2017164887 A1 WO2017164887 A1 WO 2017164887A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutically acceptable
acceptable salt
dose
eriotinib
erlotinib
Prior art date
Application number
PCT/US2016/024232
Other languages
French (fr)
Other versions
WO2017164887A8 (en
Inventor
Stanley C. GILL
Kenneth K. Iwata
Gregory J. Riely
Jun Wu
Helena YU
Original Assignee
OSI Pharmaceuticals, LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by OSI Pharmaceuticals, LLC filed Critical OSI Pharmaceuticals, LLC
Priority to PCT/US2016/024232 priority Critical patent/WO2017164887A1/en
Publication of WO2017164887A1 publication Critical patent/WO2017164887A1/en
Publication of WO2017164887A8 publication Critical patent/WO2017164887A8/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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

  • the present disclosure describes a novel dosing regimen for erlotinib or a pharmaceutically acceptable salt thereof.
  • the dosing regimen demonstrates impressive control of central nervous system disease, which is better than that reported with standard dose erlotinib.
  • the present disclosure describes the use of the novel dosing regimen for treating patients in need thereof, including for controlling formation of metastatic brain, leptomeninges, or CNS lesions in a patient with non- small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation with or without pre-existing brain, leptomeninges, or CNS metastases.
  • NSCLC non- small cell lung cancer
  • EGFR epidermal growth factor receptor
  • TARCEVA® is a tyrosine kinase inhibitor (TKI).
  • TKI tyrosine kinase inhibitor
  • NSCLC metastatic non-small cell lung cancer
  • EGFR epidermal growth factor receptor
  • L858R exon 21 substitution mutations as detected by an FDA-approved test
  • maintenance treatment of patients with locally advanced or metastatic NSCLC whose disease has not progressed after four cycles of platinum based first-line chemotherapy
  • TARCEVA® (erlotinib), a tyrosine kinase inhibitor, is a quinazolinamine with the chemical name N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine.
  • TARCEVA® contains erlotinib as the hydrochloride salt that has the following structural formula:
  • Erlotinib hydrochloride has the molecular formula C22H23N3O4.HCI and a molecular weight of 429.90. The molecule has a pKa of 5.42 at 25°C. Erlotinib hydrochloride is very slightly soluble in water, slightly soluble in methanol and practically insoluble in acetonitrile,
  • Aqueous solubility of erlotinib hydrochloride is dependent on pH with increased solubility at a pH of less than 5 due to protonation of the secondary amine. Over the pH range of 1.4 to 9.6, maximal solubility of approximately 0.4 mg/mL occurs at a pH of approximately 2.
  • Epidermal growth factor receptor (EGFR) is expressed on the cell surface of both normal and cancer cells. In some tumor cells signaling through this receptor plays a role in tumor cell survival and proliferation irrespective of EGFR mutation status. Erlotinib reversibly inhibits the kinase activity of EGFR, preventing EGFR mutations.
  • T790M confers an increase in ATP binding affinity, thereby reducing the inhibitory efficacy of eriotinib. See, Yun et al., The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP, PNAS 105 (6): 2070-5, (2008).
  • EGFR-mutant lung cancers are highly responsive to EGFR tyrosine kinase inhibitors (TKIs) with superior progression free survival when compared to cytotoxic chemotherapy.
  • TKIs EGFR tyrosine kinase inhibitors
  • Acquired resistance occurs, however, with the central nervous system as a frequent site of relapse.
  • a parallel strategy to improve outcomes in patients with EGFR-mutant lung cancers is to adjust initial treatment to delay or prevent acquired resistance. While some have investigated EGFR TKIs in combination with other agents, modulating EGFR TKI dosing to prevent resistance in patients with EGFf?-mutant lung cancers has not been assessed. See, Johnson et al., Phase l/l I Study of HSP90 Inhibitor AUY922 and Eriotinib for EGFR-Mutant Lung Cancer With Acquired Resistance to 18
  • EGFR TKIs such as osimertinib, which inhibit EGFR T790M may be effective at the time of progression on eriotinib, afatinib, or gefitinib.
  • Eriotinib was initially developed to inhibit wild-type EGFR.
  • the 150 mg daily dose was the maximum tolerated dose established via a phase I study (Hildalgo et al.), which predated knowledge of EGFR mutation. The choice of a 150mg daily dose, therefore, did not take into consideration the development of drug resistance in patients whose tumors harbor EGFR mutations.
  • Mathematical modeling can predict the evolutionary dynamics that result in proliferation of resistant clones, and suggest potential alternative dosing schedules to delay resistance. See, Chmielecki et al., Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling, Science Translational Medicine, 2011 , 3(90):90ra59, and Foo et al., Evolution of resistance to targeted anti-cancer therapies during continuous and pulsed administration strategies, PLoS Computational Biology, 2009, 5(11):e1000557.
  • Yu et al. used these methods to evaluate different dosing schedules of eriotinib and selected twice weekly high dose of eriotinib plus daily low dose eriotinib as better able to delay progression in the setting of pre-existing resistant, EGFR T790M positive cells.
  • the mathematical prediction and hypotheses were confirmed in pre-clinical studies using 20 ⁇ erlotinib/1 ⁇ eriotinib and 100 nM afatinib/1 ⁇ eriotinib doses. See, Chmielecki et al., Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling, Science Translational Medicine, 2011 ,
  • Milton et al. conducted a study of weekly high dose eriotinib in unselected patients with advanced lung cancers. See, Milton et al., Weekly high-dose eriotinib in patients with non-small cell lung cancer (NSCLC): a phase l/l I study, Cancer, 2006; 107(5): 1034-41.
  • the maximum tolerated dose (MTD) was not reached at eriotinib 2000mg once weekly.
  • a separate Phase 1 study of twice-weekly pulse dose eriotinib identified the MTD to be eriotinib 1000mg twice-weekly, with a DLT of rash seen at higher dose levels.
  • Central nervous system (CNS) involvement is a major issue for patients with EGFf?-mutant lung cancers, with up to sixty percent of these patients developing brain or leptomeningeal metastases during their disease course. See, Heon et al., The impact of initial gefitinib or eriotinib versus chemotherapy on central nervous system progression in advanced non-small cell lung cancer with EGFR mutations,
  • Central nervous system-only progression may be a result of inadequate brain penetration, with cerebrospinal fluid (CSF) concentrations of eriotinib only 3 - 5 % of those in plasma.
  • CSF cerebrospinal fluid
  • One embodiment of the present disclosure includes a dosing regimen comprising: (i) orally administering one or more daily low dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more weekly high dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • a dosing regimen comprising: (i) orally administering one or more daily low dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more weekly high dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes a low dose of 50 mg of eriotinib or a
  • One aspect of the embodiment includes a high dose from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes a high dose of 1200 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the daily dose is once daily.
  • One aspect of the embodiment includes wherein the weekly dose is once or twice weekly.
  • One aspect of the embodiment includes wherein the regimen improves tolerability over a standard 100 mg to 150 mg daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the improved tolerability is manifested by a lower incidence in one or more of rash, diarrhea, nausea, fatigue, or mucositis.
  • One embodiment of the present disclosure includes a weekly dosing regimen comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • a weekly dosing regimen comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
  • the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
  • the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • One embodiment of the present disclosure includes a dosing regimen of eriotinib or a pharmaceutically acceptable salt thereof comprising: (i) administering to a patient in need thereof eriotinib or a pharmaceutically acceptable salt thereof in amounts that provide a steady state concentration (C trou g h ) of 0.05 - 10 ⁇ in the patient; and (ii) administering to a patient in need thereof eriotinib or a
  • a pulsed concentration (C max ) of 5 - 50 ⁇ in the patient includes wherein the steady state concentration (Ct rou g h ) is approximately 0.5 ⁇ .
  • the pulsed concentration (C max ) is approximately 20 ⁇ .
  • the pulsed concentration is maintained for 4 hours.
  • One embodiment of the present invention includes a method of treating or preventing central nervous system (CNS) metastases in a patient in need thereof comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • CNS metastases is leptomeningeal carcinomatosis.
  • the patient has extra-cranial metastatic non-small cell lung cancer.
  • the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
  • One aspect of the embodiment includes wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
  • the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the patient with or without pre-existing brain, leptomeninges, or CNS metastases has no progression.
  • One aspect of the embodiment includes wherein the patient's cerebrospinal fluid (CSF) is cleared of malignant cells.
  • CSF cerebrospinal fluid
  • One aspect of the embodiment includes wherein the patient achieves a clinical benefit.
  • One aspect of the embodiment includes wherein the clinical benefit is one or more of decrease in tumor size, suppression of tumor growth, delayed time to progression in CNS, unobservable increase in size of brain, leptomeninges, or CNS metastases or lesions, unobservable new brain, leptomeninges, or CNS metastases or lesion, delay in time from surgery to recurrence, increase in treatment options, delay or decrease in leptomeningeal seeding in the CSF, or an ability to delay whole brain radiation.
  • One embodiment of the present disclosure includes a method for controlling formation of metastatic brain, leptomeninges, or CNS lesions in a patient with non- small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation with or without pre-existing brain, leptomeninges, or CNS metastases comprising: (i) orally administering one or more pulse dose of erlotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of erlotinib or a pharmaceutically acceptable salt thereof.
  • NSCLC non- small cell lung cancer
  • EGFR epidermal growth factor receptor
  • One aspect of the embodiment includes wherein the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
  • One aspect of the embodiment includes wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
  • the pulse dose is from about 600 mg to about 1350 mg of erlotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is 1200 mg erlotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is from about 25 mg to about 50 mg of erlotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein the daily dose is 50 mg erlotinib or a pharmaceutically acceptable salt thereof.
  • the patient's cerebrospinal fluid is cleared of malignant cells.
  • the patient achieves an objective or complete response or disease control.
  • the control is durable.
  • One embodiment of the present disclosure includes a combination comprising: (a) erlotinib or a pharmaceutically acceptable salt thereof (i) orally administered in one or more pulse dose of erlotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administered in one or more daily dose of erlotinib or a pharmaceutically acceptable salt thereof; and (b) one or more therapeutic compounds administered in an effective amount thereof.
  • One aspect of the embodiment includes wherein the one or more therapeutic compounds are one or more oncology therapeutic compounds.
  • One aspect of the embodiment includes wherein the one or more therapeutic compounds are one or more inhibitors of epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • One aspect of the embodiment includes wherein the one or more therapeutic compounds are one or more inhibitors of EGFR is a mutant-selective inhibitor of EGFR.
  • the one or more therapeutic compounds are selected from one or more of rociletinib (CO-1686), osimertinib (AZD9291), HM61713, Bl 1482694, or other third or later generation EGFR TKI.
  • the pulse dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
  • One aspect of the embodiment includes wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
  • the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • each dose is selected from a solid or liquid form.
  • a solid form comprises one or more of eriotinib or the one or more therapeutic compounds complexed with a cyclodextrin and nano-encapsulated into a pharmaceutically acceptable carrier.
  • One embodiment of the present disclosure includes a unit dose of eriotinib or a pharmaceutically acceptable salt thereof, comprising a pharmaceutical composition comprising erotinib or a pharmaceutically acceptable salt thereof which achieves a peak concentration (C max ) of 5 - 50 ⁇ in a patient.
  • the unit dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the unit dose is about 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • the peak concentration (C max ) is approximately 20 ⁇ .
  • the peak concentration is maintained for 4 hours.
  • One aspect of the embodiment includes wherein the dose is selected from a solid or liquid form.
  • the solid form comprises eriotinib complexed with a cyclodextrin and nano-encapsulated into a pharmaceutically acceptable carrier.
  • One embodiment of the present disclosure includes a method of treating brain, leptomeninges, or CNS metastases in a patient diagnosed with EGFR-mutant lung cancer comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes wherein eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
  • One aspect of the embodiment includes wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
  • the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • One embodiment includes a method of treating a patient diagnosed with gliablastoma comprising: orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • the method includes wherein the pulse dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
  • the method includes wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
  • the method includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • GBM Glioblastoma multiforme
  • EGFR Epidermal growth factor receptor
  • mutation and re-arrangement are frequently observed in primary GBM.
  • Epidermal growth factor receptor (EGFR) and EGFRvlll analysis is of current interest in glioblastoma.
  • Fig. 1 illustrates a synthetic scheme for the manufacture of eriotinib.
  • Fig. 2 is a graphical representation of best response of target lesions
  • Fig. 3 is a graphical representation of progression-free survival probability from months since start of therapy.
  • Fig. 4 is an example of CNS response in a patient with untreated brain metastases on the regimen, namely pulse-continuous eriotinib or a pharmaceutically acceptable salt thereof.
  • Fig. 5 is a graphical representation of plasma concentration curves for eriotinib after multiple dosing. Blood samples were collected during cycle 1 week 1 day 1 at 0.5, 1 , 2, 4, and 6 hours after the first dose; before and four hours after eriotinib dosing on cycle 1 week 2 day 1 and cycle 1 week 4 day 1.
  • one embodiment of the present disclosure includes a dosing regimen comprising: (i) orally administering one or more daily low dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more weekly high dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • the terms "low dose” and "high dose” are used relative to the other as well as relative to a standard 100 mg to 150 mg daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • a low dose is lower than each of the standard dose and the high dose.
  • a high dose is higher than each of the standard dose and the low dose.
  • One aspect of the embodiment includes a low dose of 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes a high dose from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • One aspect of the embodiment includes a high dose of 1200 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • one embodiment of the present disclosure includes a weekly dosing regimen comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • pulse dose refers to a higher dose of eriotinib or a
  • pulse dose may be determined by determining a dose that provides higher CSF concentrations than a standard 100 mg to 150 mg daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
  • the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
  • unit dose refers to a dose of medicine in an individual packet for convenience, safety, or monitoring.
  • tolerability refers to the degree to which overt adverse effects of a drug can be tolerated by a patient. Tolerability of a particular drug can be discussed in a general sense, or it can be a quantifiable measurement as part of a clinical study. As one measure, tolerability may be determined by the rate of dropouts, or patients that forfeit participation in a clinical study due to one or more adverse effects.
  • the term "pharmaceutically acceptable” refers to carrier(s), diluent(s), excipient(s) or salt forms of the compounds of the present invention that are compatible with the other ingredients of the formulation and not deleterious to the recipient of the pharmaceutical composition.
  • the terms “effective amount”, “therapeutic amount”, and “effective dose” refer to an amount of the compound of the present invention sufficient to elicit a desired pharmacological or therapeutic effect. Moreover, the therapeutic amount of effective amount may thus result in an effective treatment of a disorder.
  • Treatment of a disorder may be manifested by delaying or preventing the onset or progression of the disorder, as well as the onset or progression of symptoms associated with the disorder. Treatment of a disorder may also be manifested by a decrease or elimination of symptoms, reversal of the progression of the disorder, as well as any other contribution to the well-being of the patient.
  • Erlotinib may exist in free form or, where appropriate, as a pharmaceutically acceptable derivative thereof.
  • a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a subject in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • the term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • a “pharmaceutically acceptable salt” means any non-toxic salt or salt of an ester of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an active metabolite or residue thereof.
  • active metabolite or residue thereof means that a metabolite or residue thereof is also an EGFR TKI.
  • Pharmaceutically acceptable salts include those derived from suitable inorganic and organic acids and bases.
  • suitable inorganic and organic acids and bases include those derived from suitable inorganic and organic acids and bases.
  • pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci. 4 alkyl) 4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counter-ions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
  • Polymorphism the occurrence of different crystal forms, is a property of some molecules and molecular complexes.
  • a single molecule like erlotinib, may give rise to a variety of crystalline forms having distinct crystal structures and physical properties like melting point, x-ray diffraction pattern, infrared absorption fingerprint, and solid state NMR spectrum.
  • One crystalline form may give rise to thermal behavior different from that of another crystalline form. Thermal behavior can be measured in the laboratory by such techniques as capillary melting point, thermogravimetric analysis (“TGA”), and differential scanning calorimetry (“DSC”), which have been used to distinguish polymorphic forms.
  • TGA thermogravimetric analysis
  • DSC differential scanning calorimetry
  • polymorphs are distinct solids sharing the same molecular formula yet having distinct advantageous physical properties compared to other crystalline forms of the same compound or complex.
  • the present invention includes pharmaceutical compositions comprising erlotinib and/or pharmaceutically acceptable salts thereof and one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • the manner in which the compound of the present invention is administered can vary.
  • the compound of the present invention is preferably administered orally.
  • Preferred pharmaceutical compositions for oral administration include tablets, capsules, caplets, syrups, solutions, and suspensions.
  • the pharmaceutical compositions of the present invention may be provided in modified release dosage forms such as time-release tablet and capsule formulations.
  • the compounds of the present invention may be used in the treatment of a variety of disorders and conditions and, as such, may be used in combination with a variety of other suitable therapeutic agents useful in the treatment or prophylaxis of those disorders or conditions.
  • compositions refer to erlotinib or a pharmaceutically acceptable salt thereof optionally admixed with one or more pharmaceutically acceptable carriers, diluents, or excipients.
  • Pharmaceutically acceptable compositions preferably exhibit a degree of stability to environmental conditions so as to make them suitable for manufacturing and commercialization purposes.
  • compositions may comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable carrier, adjuvant, or vehicle which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's Pharmaceutical Sciences Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except
  • composition its use is contemplated to be within the scope of this invention.
  • materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powder
  • excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
  • oils such as peanut oil, cottonseed oil; safflower oil; ses
  • compositions can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • a liquid dosage form may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid may be incorporated in an injectable product.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • intramuscular injection This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility.
  • the rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form.
  • delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle.
  • injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or (a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, (c) humectants such as glycerol, (d) disintegrating agents such as agar- agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, (e) solution retarding agents such as paraffin, (f) absorption accelerators such as quaternary ammonium compounds, (g) wetting agents such as, for example, cetyl
  • 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 sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. 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 sugar as well as high molecular weight polethylene glycols and the like.
  • Erlotinib or a pharmaceutically acceptable salt thereof may be formulated using nanotechnology. Nanoparticles are attractive for medical purposes based on their unique features, such as their surface to mass ratio being larger than that of other particles, their quantum properties, and their ability to adsorb and carry other compounds. Nanoparticles may have dimensions below 0.1 pm or 100 nm.
  • a pharmaceutical composition may comprise relatively large (size >100 nm) nanoparticles, as needed for loading a sufficient amount of drug onto the particles.
  • engineered particles may be used as carrier, but also the drug itself may be formulated at a nanoscale, and then function as its own carrier.
  • the composition of the engineered nanoparticles may vary.
  • Source materials may be of biological origin like phospholipids, lipids, lactic acid, dextran, chitosan, or have more chemical characteristics like various polymers, carbon, silica, and metals. Especially in the area of engineered nanoparticles of polymer origin there is a vast area of possibilities for the chemical composition. See, for example, Martins et al., Nanoparticle Drug Delivery Systems: Recent Patents and Applications in Nanomedicine, Recent Patents on Nanomedicine, 2013, 3(2), pp. 1 - 14. Many cytotoxic chemotherapeutic agents have poor aqueous solubility. These molecules are associated with poor physicochemical and biopharmaceutical properties, which makes the formulation difficult.
  • Cyclodextrins are chemically and physically stable macromolecules produced by enzymatic degradation of starch. They are water-soluble, biocompatible in nature with hydrophilic outer surface and lipophilic cavity. They have the shape of truncated cone or torus rather than perfect cylinder because of the chair conformation of glucopyranose unit. Cyclodextrins are classified as natural and derived
  • Natural cyclodextrins comprise three well-known industrially produced (major and minor) cyclic oligosaccharides. The most common natural cyclodextrins are ⁇ , ⁇ , and ⁇ consisting of 6, 7, and 8 glucopyranose units. They are crystalline, homogeneous, and nonhygroscopic substances. Amongst these, ⁇ -cyclodextrin is used for complexation due to perfect cavity size, efficient drug complexation and loading, availability, and relatively low cost.
  • Various hydrophilic, hydrophobic, and ionic derivatives have been developed and utilized to improve the physicochemical and biopharmaceutical properties of drug and inclusion capacity of natural cyclodextrins.
  • ⁇ - ⁇ -CD Hydroxypropyl ⁇ -cyclodextrin
  • RM ⁇ -CD randomly methylated- ⁇ - cyclodextrin
  • SBE- -CD sulfobutylether-p-cyclodextrin
  • Polymerized cyclodextrins are high molecular weight compounds, either water-soluble or insoluble. They offer the advantage of amorphous state and complexation without toxic effects.
  • Examples of polymerized cyclodextrins are soluble anionic ⁇ -cyclodextrin polymer, soluble ⁇ -cyclodextrin polymer, and epichlorohydrin ⁇ -cyclodextrin polymer.
  • Cyclodextrin-based nanocarriers may be prepared by utilizing the concept of dual approach, which involves combination of two different approaches in a single delivery system. This covers two aspects firstly; the anticancer drug is complexed with suitable cyclodextrin and secondly encapsulation of complexed drug into carrier.
  • Erlotinib or a pharmaceutically acceptable salt thereof may also be in microencapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. I n such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention.
  • the invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms are prepared by dissolving or dispensing the compound in the proper medium .
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the compounds of this invention may be made by a variety of methods, including well-established synthetic methods. Illustrative general synthetic methods are set out below and then specific compounds of the invention are prepared in the working Examples. The compounds can be prepared according to the methods described below using readily available starting materials and reagents. In these reactions, variants may be employed which are themselves known to those of ordinary skill in this art but are not described in detail here.
  • Pulse-continuous erlotinib results in impressive control of central nervous system disease [0061] Based on evolutionary mathematical modeling data, the present inventors tested a novel schedule of twice weekly pulse erlotinib in combination with daily erlotinib as initial EGFR TKI treatment in patients with EGFR-mutant lung cancers.
  • the trial was a prospective, open-label, single center phase 1 dose-escalation study of twice weekly pulse dose and daily erlotinib in patients with EGFR-mutant lung cancers.
  • the primary endpoint of the study was the identification of the maximum tolerated dose of the combination of twice weekly high dose and daily low dose erlotinib.
  • Secondary endpoints included the measurement of progression-free survival, overall survival, complete and partial response rate, determination of central nervous system progression and pharmacokinetic analysis.
  • the study was registered at clinicaltrials.gov (NCT01967095). The trial was conducted after approval from the institutional review board at Memorial Sloan Kettering. All patients provided written informed consent.
  • Cycle 1 week 1 (Days 1 -7) consisted of pulse dose erlotinib on days 1 and 2 without daily low dose erlotinib on days 3 to 7. For all subsequent weeks, patients received pulse dose erlotinib on days 1 and 2, and erlotinib 50 mg oral daily for 5 days on days 3 to 7 which was repeated weekly to complete 21 day cycles. Cycle 1 was 4 weeks to account for one week of lead in pulse dose erlotinib only. For each dose level, the dose of pulse erlotinib on days 1-2 was escalated (supplemental Table 1).
  • Dose level 4 1050mg 6 1 (grade 3 ALT/AST)
  • Dose level 6 1350mg 3 2 (grade 2 rash, grade 3
  • DLT dose-limiting toxicity
  • Toxicity assessments Patients were assessed five times during cycle 1 (28 days) and then every 21 days thereafter. Patient history, physical examination, complete blood count and serum chemistries were performed at each visit. Toxicity was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE), version 4. Dose-limiting toxicity was defined as any grade 4 hematologic toxicity lasting greater than 5 days. Grade 3
  • DLT thrombocytopenia with clinically significant bleeding
  • PK pharmacokinetic analysis
  • Plasma levels of erlotinib were determined using a validated liquid chromatography-tandem mass spectrometry assay.
  • LC-MS/MS was performed using a Phenomenex Kinetex 2.6 ⁇ core-shell Biphenyl column (50x3 mm) and Shimadzu HPLC.
  • MS detection was using an AB SCIEX API 4000 triple quadrupole mass spectrometer operating in positive ion electrospray ionization (ESI) mode.
  • Standard PK parameters were calculated using a noncompartmental method (WiNNonlin, Pharsight).
  • 34 patients were enrolled onto this study, including 24 patients on the dose escalation portion of the phase 1 study and 10 patients in an expansion cohort at the MTD. In total, sixteen patients were treated at the MTD. The clinical characteristics of all patients are listed in Table 1.
  • Bilirubin increased 4 (12%) 5 (15%) 2 (6%) 0 11 (32%)
  • Table 3 Molecular findings at diagnosis and at acquired resistance for patients who came off study for progression Baseline Acquired Resistance (off study for progression)
  • Pulse-continuous eriotinib was well tolerated. The most common treatment related adverse events were rash, diarrhea, nausea, fatigue and mucositis. When compared to prospective studies of standard daily dosed eriotinib and afatinib, BR.21 and Lux-Lung 3, respectively, toxicity was similar. See, Sequist et al. , Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung
  • Central nervous system metastases are a critical issue in the management of patients with EGFR-mutant lung cancers. Due to a prolonged disease course and improved overall survival, EGFR-mutant cancers may be enriched for CNS involvement with the cumulative incidence of CNS metastases approaching 60%.
  • Adjuvant therapy or care also called adjunct therapy or adjunctive therapy or care, is therapy that is given in addition to the primary, main, or initial therapy to maximize its effectiveness. As an adjuvant agent modifies the effect of another agent, so adjuvant therapy modifies other therapy.
  • NSCLC non-small-cell lung cancer

Abstract

A novel dosing regimen for erlotinib or a pharmaceutically acceptable salt thereof is described herein. The dosing regimen demonstrates impressive control of central nervous system disease, which is better than that reported with standard dose erlotinib. The use of the novel dosing regimen for treating patients in need thereof, including for controlling formation of metastatic brain, leptomeninges, or CNS lesions in a patient with non-small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation with or without pre-existing brain metastases, is also described herein.

Description

PULSE DOSING REGIMEN AND METHODS OF TREATMENT
FIELD OF THE INVENTION
[0001] The present disclosure describes a novel dosing regimen for erlotinib or a pharmaceutically acceptable salt thereof. The dosing regimen demonstrates impressive control of central nervous system disease, which is better than that reported with standard dose erlotinib. The present disclosure describes the use of the novel dosing regimen for treating patients in need thereof, including for controlling formation of metastatic brain, leptomeninges, or CNS lesions in a patient with non- small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation with or without pre-existing brain, leptomeninges, or CNS metastases.
BACKGROUND OF THE INVENTION
[0002] TARCEVA® (erlotinib HCI) is a tyrosine kinase inhibitor (TKI). As referenced in the FDA-approved label, TARCEVA® is indicated in the United States for first-line treatment of patients with metastatic non-small cell lung cancer (NSCLC) whose tumors have epidermal growth factor receptor (EGFR) exon 19 deletions or exon 21 (L858R) substitution mutations as detected by an FDA-approved test; maintenance treatment of patients with locally advanced or metastatic NSCLC whose disease has not progressed after four cycles of platinum based first-line chemotherapy; treatment of locally advanced or metastatic NSCLC after failure of at least one prior
chemotherapy regimen; and first-line treatment of patients with locally advanced, unresectable or metastatic pancreatic cancer, in combination with gemcitabine.
[0003] TARCEVA® (erlotinib), a tyrosine kinase inhibitor, is a quinazolinamine with the chemical name N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine. TARCEVA® contains erlotinib as the hydrochloride salt that has the following structural formula:
Figure imgf000003_0001
Reference is made to U.S. RE 41 ,065, herein incorporated by reference with regard to the description and synthesis of the compound.
[0004] Erlotinib hydrochloride has the molecular formula C22H23N3O4.HCI and a molecular weight of 429.90. The molecule has a pKa of 5.42 at 25°C. Erlotinib hydrochloride is very slightly soluble in water, slightly soluble in methanol and practically insoluble in acetonitrile,
acetone, ethyl acetate, and hexane. Aqueous solubility of erlotinib hydrochloride is dependent on pH with increased solubility at a pH of less than 5 due to protonation of the secondary amine. Over the pH range of 1.4 to 9.6, maximal solubility of approximately 0.4 mg/mL occurs at a pH of approximately 2.
[0005] Epidermal growth factor receptor (EGFR) is expressed on the cell surface of both normal and cancer cells. In some tumor cells signaling through this receptor plays a role in tumor cell survival and proliferation irrespective of EGFR mutation status. Erlotinib reversibly inhibits the kinase activity of EGFR, preventing
autophosphorylation of tyrosine residues associated with the receptor and thereby inhibiting further downstream signaling. Erlotinib binding affinity for EGFR exon 19 deletion or exon 21 (L858R) mutations is higher than its affinity for the wild type receptor. Erlotinib inhibition of other tyrosine kinase receptors has not been fully characterized.
[0006] As with other ATP competitive small molecule tyrosine kinase inhibitors, patients may develop resistance. Over 50% of resistance is caused by a mutation in the ATP binding pocket of the EGFR kinase domain involving substitution of a small polar threonine residue with a large nonpolar methionine residue, T790M. See, Balak et al., Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors, Clin. Cancer Res 12 (1): 6494-501 , (2006). While proponents of the 'gatekeeper' mutation hypothesis suggest this mutation prevents the binding of eriotinib through steric hindrance, research suggests that T790M confers an increase in ATP binding affinity, thereby reducing the inhibitory efficacy of eriotinib. See, Yun et al., The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP, PNAS 105 (6): 2070-5, (2008).
[0007] EGFR-mutant lung cancers are highly responsive to EGFR tyrosine kinase inhibitors (TKIs) with superior progression free survival when compared to cytotoxic chemotherapy. See, for example, Lee et al., Impact of Specific Epidermal Growth Factor Receptor (EGFR) Mutations and Clinical Characteristics on Outcomes after Treatment with EGFR Tyrosine Kinase Inhibitors Versus Chemotherapy in EGFR- Mutant Lung Cancer: A Meta- Analysis, Journal of clinical oncology.
2015;33(17): 1958-65; Mok et al. , Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma, The New England Journal of Medicine, 2009;361 (10):947-57; Rosell et al. , Eriotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial, The Lancet
Oncology, 2012; 13(3):239-46; and Sequist et al. , Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations, Journal of Clinical Oncology, 2013; 31 (27):3327-34. The majority of patients will respond to eriotinib, gefitinib, and afatinib, but in less than a year develop resistance to further therapy with these agents. See, Sequist et al. , Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations, Journal of Clinical Oncology. 2013;
31 (27):3327-34; Maemondo et al., Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR, The New England Journal of Medicine,
2010;362(25):2380-8; Mok et al., Gefitinib or Carboplatin-Paclitaxel in Pulmonary Adenocarcinoma. N. Engl. J. Med. , 2009; 361 (10):947-57; and Janne et al.,
Randomized Phase II Trial of Eriotinib Alone or With Carboplatin and Paclitaxel in Patients Who Were Never or Light Former Smokers With Advanced Lung
Adenocarcinoma: CALGB 30406 Trial, Journal of Clinical Oncology, 2012; 39(17): 2063-9. As noted hereinabove, the most common mechanism of resistance is acquisition of an EGFR T790M mutation, identified in 60% of patients with acquired resistance to EGFR TKIs. See also, Kobayashi et al. , EGFR mutation and resistance of non-small-cell lung cancer to gefitinib, The New England Journal of Medicine. 2005; 352 (8): 786-92, and Yu et al. , Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers, Clinical Cancer Research, 2013, 19(8):2240-7. Acquired resistance occurs, however, with the central nervous system as a frequent site of relapse. A parallel strategy to improve outcomes in patients with EGFR-mutant lung cancers is to adjust initial treatment to delay or prevent acquired resistance. While some have investigated EGFR TKIs in combination with other agents, modulating EGFR TKI dosing to prevent resistance in patients with EGFf?-mutant lung cancers has not been assessed. See, Johnson et al., Phase l/l I Study of HSP90 Inhibitor AUY922 and Eriotinib for EGFR-Mutant Lung Cancer With Acquired Resistance to 18
Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors, Journal of Clinical Oncology, 2015, 33(15):1666-73, Riely et al., Prospective assessment of discontinuation and reinitiation of eriotinib or gefitinib in patients with acquired resistance to eriotinib orgefitinib followed by the addition of everolimus, Clinical Cancer Research, 2007, 13(17):5150-5, Reguart et al., Phase l/l I trial of vorinostat (SAHA) and eriotinib for non-small cell lung cancer (NSCLC) patients with epidermal growth factor receptor (EGFR) mutations after eriotinib progression, Lung Cancer, 2014, 84(2):161-7, Johnson et al., Phase II trial of dasatinib for patients with acquired resistance to treatment with the epidermal growth factor receptor tyrosine kinase inhibitors eriotinib orgefitinib, Journal of Thoracic Oncology. 2011 , 6(6):1128-31 , and Goldberg et al., A phase I study of eriotinib and hydroxychloroquine in advanced non- small-cell lung cancer, Journal of Thoracic Oncology, 2012, 7(10): 1602-8. Third generation EGFR TKIs, such as osimertinib, which inhibit EGFR T790M may be effective at the time of progression on eriotinib, afatinib, or gefitinib.
[0008] Eriotinib was initially developed to inhibit wild-type EGFR. The 150 mg daily dose was the maximum tolerated dose established via a phase I study (Hildalgo et al.), which predated knowledge of EGFR mutation. The choice of a 150mg daily dose, therefore, did not take into consideration the development of drug resistance in patients whose tumors harbor EGFR mutations. Hidalgo et al., Phase I and pharmacologic study of OSI-774, an epidermal growth factor receptor tyrosine kinase inhibitor in patients with advanced solid malignancies, Journal of Clinical Oncology, 2001 , 19(13): 3267-79. Mathematical modeling can predict the evolutionary dynamics that result in proliferation of resistant clones, and suggest potential alternative dosing schedules to delay resistance. See, Chmielecki et al., Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling, Science Translational Medicine, 2011 , 3(90):90ra59, and Foo et al., Evolution of resistance to targeted anti-cancer therapies during continuous and pulsed administration strategies, PLoS Computational Biology, 2009, 5(11):e1000557. Yu et al., used these methods to evaluate different dosing schedules of eriotinib and selected twice weekly high dose of eriotinib plus daily low dose eriotinib as better able to delay progression in the setting of pre-existing resistant, EGFR T790M positive cells. The mathematical prediction and hypotheses were confirmed in pre-clinical studies using 20 μΜ erlotinib/1 μΜ eriotinib and 100 nM afatinib/1 μΜ eriotinib doses. See, Chmielecki et al., Optimization of dosing for EGFR-mutant non-small cell lung cancer with evolutionary cancer modeling, Science Translational Medicine, 2011 ,
3(90):90ra59.
[0009] Milton et al., conducted a study of weekly high dose eriotinib in unselected patients with advanced lung cancers. See, Milton et al., Weekly high-dose eriotinib in patients with non-small cell lung cancer (NSCLC): a phase l/l I study, Cancer, 2006; 107(5): 1034-41. The maximum tolerated dose (MTD) was not reached at eriotinib 2000mg once weekly. A separate Phase 1 study of twice-weekly pulse dose eriotinib identified the MTD to be eriotinib 1000mg twice-weekly, with a DLT of rash seen at higher dose levels. See, Chia et al., A Phase 1 dose escalation pharmacokinetic (PK) and pharmacodynamic (PD) study of weekly and twice weekly erltoinib in advanced stage malignancies, Journal of Clinical Oncology, 2007:25, SuppI; Abstract 3594.
[0010] Due to a wide inter-subject variability in bioavailability, lower daily doses of eriotinib may be effective with significantly less toxicity, although not proven in a randomized trial setting. Yamada et al., A prospective, multicentre phase II trial of low-dose eriotinib in non-small cell lung cancer patients with EGFR mutations pretreated with chemotherapy: Thoracic Oncology Research Group 0911, Eur J Cancer, 2015 Sep; 51 (14): 1904-10; Eriotinib at a dose of 25 mg daily for non-small cell lung cancers with EGFR mutations, Journal of Thoracic Oncology,
2010;5(7):1048-53; and Satoh et al., Low-dose gefitinib treatment for patients with advanced non-small cell lung cancer harboring sensitive epidermal growth factor receptor mutations. Journal of Thoracic Oncology, 201 1 ; 6(8): 1413-7.
[0011] The available data suggest that EGFR TKI high pulse dosing is tolerable and low daily dosing is effective but these have not previously been administered together in patients.
[0012] Central nervous system (CNS) involvement is a major issue for patients with EGFf?-mutant lung cancers, with up to sixty percent of these patients developing brain or leptomeningeal metastases during their disease course. See, Heon et al., The impact of initial gefitinib or eriotinib versus chemotherapy on central nervous system progression in advanced non-small cell lung cancer with EGFR mutations,
Clinical Cancer Research, 2012; 18(16):4406-14; and Omuro et al., High incidence of disease recurrence in the brain and leptomeninges in patients with non-small cell lung carcinoma after response to gefitinib, Cancer, 2005, 103(11):2344-8. Although benefit is commonly seen with EGFR TKIs when CNS disease is already present, these medications inconsistently lead to durable CNS control and do not prevent the emergence of CNS metastases. Up to 33% of patients with EGFR-mutant lung cancers have CNS progression during initial EGFR TKI therapy and in a significant subset of patients, the CNS progression occurs in the setting of continued systemic control. See, Heon et al., The impact of initial gefitinib or eriotinib versus
chemotherapy on central nervous system progression in advanced non-small cell lung cancerwith EGFR mutations, Clinical Cancer Research, 2012, 18(16):4406-14; Omuro et al., High incidence of disease recurrence in the brain and leptomeninges in patients with nonsmall cell lung carcinoma after response to gefitinib Cancer, 2005; 103(11):2344-8; and Lee et al., Frequent central nervous system failure after clinical benefit with epidermal growth factor receptor tyrosine kinase inhibitors in Korean patients with nonsmall-cell lung cancer, Cancer, 2010, 116(5): 1336-43. Central nervous system-only progression may be a result of inadequate brain penetration, with cerebrospinal fluid (CSF) concentrations of eriotinib only 3 - 5 % of those in plasma. See, Togashi et al., Cerebrospinal fluid concentration of eriotinib and its active metabolite OSI-420 in patients with central nervous system metastases of non-small cell lung cancer, Journal of Thoracic Oncology, 2010, 5(7):950-5.
SUMMARY OF THE INVENTION
[0013] Consequently, the CNS becomes a common sanctuary site of disease progression due to inadequate drug delivery, not acquired drug resistance. The present inventors demonstrate that pulse dose eriotinib or a pharmaceutically acceptable salt thereof produces higher CSF concentrations and appears effective in the treatment of CNS metastases.
[0014] With eriotinib 1500mg once weekly, a peak plasma concentration of 1 1 ,300 nM was reached with a concurrent CSF concentration of 120 nM which is above the IC50 of eriotinib. See, Clarke et al., High dose weekly eriotinib achieves therapeutic concentrations in CSF and is effective in leptomeningeal metastases from epidermal growth factor receptor mutant lung cancer, Journal of Neuro-Oncology, 2010, 99(2):283-6. In an earlier series, patients with EGFft-mutant lung cancers with CNS involvement were treated with a median dose of eriotinib 1500mg once weekly. Six of nine had a partial response in the CNS. See, Grammes et al., "Pulsatile" high-dose weekly eriotinib for CNS metastases from EGFR mutant non-small cell lung cancer, Neuro-Oncology, 2011 , 13(12):1364-9.
[0015] One embodiment of the present disclosure includes a dosing regimen comprising: (i) orally administering one or more daily low dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more weekly high dose of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes a low dose of 50 mg of eriotinib or a
pharmaceutically acceptable salt thereof. One aspect of the embodiment includes a high dose from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes a high dose of 1200 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is once daily. One aspect of the embodiment includes wherein the weekly dose is once or twice weekly. One aspect of the embodiment includes wherein the regimen improves tolerability over a standard 100 mg to 150 mg daily dose of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the improved tolerability is manifested by a lower incidence in one or more of rash, diarrhea, nausea, fatigue, or mucositis.
[0016] One embodiment of the present disclosure includes a weekly dosing regimen comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule. One aspect of the embodiment includes wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule. One aspect of the embodiment includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
[0017] One embodiment of the present disclosure includes a dosing regimen of eriotinib or a pharmaceutically acceptable salt thereof comprising: (i) administering to a patient in need thereof eriotinib or a pharmaceutically acceptable salt thereof in amounts that provide a steady state concentration (Ctrough) of 0.05 - 10 μΜ in the patient; and (ii) administering to a patient in need thereof eriotinib or a
pharmaceutically acceptable salt thereof in amounts that provide a pulsed concentration (Cmax) of 5 - 50 μΜ in the patient. One aspect of the embodiment includes wherein the steady state concentration (Ctrough) is approximately 0.5 μΜ. One aspect of the embodiment includes wherein the pulsed concentration (Cmax) is approximately 20 μΜ. One aspect of the embodiment includes wherein the pulsed concentration is maintained for 4 hours.
[0018] One embodiment of the present invention includes a method of treating or preventing central nervous system (CNS) metastases in a patient in need thereof comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the CNS metastases is leptomeningeal carcinomatosis. One aspect of the embodiment includes wherein the patient has extra-cranial metastatic non-small cell lung cancer. One aspect of the embodiment includes wherein the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule. One aspect of the embodiment includes wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule. One aspect of the embodiment includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the patient with or without pre-existing brain, leptomeninges, or CNS metastases has no progression. One aspect of the embodiment includes wherein the patient's cerebrospinal fluid (CSF) is cleared of malignant cells. One aspect of the embodiment includes wherein the patient achieves a clinical benefit. One aspect of the embodiment includes wherein the clinical benefit is one or more of decrease in tumor size, suppression of tumor growth, delayed time to progression in CNS, unobservable increase in size of brain, leptomeninges, or CNS metastases or lesions, unobservable new brain, leptomeninges, or CNS metastases or lesion, delay in time from surgery to recurrence, increase in treatment options, delay or decrease in leptomeningeal seeding in the CSF, or an ability to delay whole brain radiation.
[0019] One embodiment of the present disclosure includes a method for controlling formation of metastatic brain, leptomeninges, or CNS lesions in a patient with non- small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation with or without pre-existing brain, leptomeninges, or CNS metastases comprising: (i) orally administering one or more pulse dose of erlotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of erlotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule. One aspect of the embodiment includes wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule. One aspect of the embodiment includes wherein the pulse dose is from about 600 mg to about 1350 mg of erlotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is 1200 mg erlotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is from about 25 mg to about 50 mg of erlotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is 50 mg erlotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the patient experiences no tumor formation in the central nervous system. One aspect of the embodiment includes wherein the patient's cerebrospinal fluid is cleared of malignant cells. One aspect of the embodiment includes wherein the patient achieves an objective or complete response or disease control. One aspect of the embodiment includes wherein the control is durable.
[0020] One embodiment of the present disclosure includes a combination comprising: (a) erlotinib or a pharmaceutically acceptable salt thereof (i) orally administered in one or more pulse dose of erlotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administered in one or more daily dose of erlotinib or a pharmaceutically acceptable salt thereof; and (b) one or more therapeutic compounds administered in an effective amount thereof. One aspect of the embodiment includes wherein the one or more therapeutic compounds are one or more oncology therapeutic compounds. One aspect of the embodiment includes wherein the one or more therapeutic compounds are one or more inhibitors of epidermal growth factor receptor (EGFR). One aspect of the embodiment includes wherein the one or more therapeutic compounds are one or more inhibitors of EGFR is a mutant-selective inhibitor of EGFR. One aspect of the embodiment includes wherein the one or more therapeutic compounds are selected from one or more of rociletinib (CO-1686), osimertinib (AZD9291), HM61713, Bl 1482694, or other third or later generation EGFR TKI. One aspect of the embodiment includes wherein the pulse dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule. One aspect of the embodiment includes wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule. One aspect of the embodiment includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the
embodiment includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein each dose is selected from a solid or liquid form. One aspect of the embodiment includes wherein a solid form comprises one or more of eriotinib or the one or more therapeutic compounds complexed with a cyclodextrin and nano-encapsulated into a pharmaceutically acceptable carrier.
[0021] One embodiment of the present disclosure includes a unit dose of eriotinib or a pharmaceutically acceptable salt thereof, comprising a pharmaceutical composition comprising erotinib or a pharmaceutically acceptable salt thereof which achieves a peak concentration (Cmax) of 5 - 50 μΜ in a patient. One aspect of the embodiment includes wherein the unit dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the unit dose is about 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the peak concentration (Cmax) is approximately 20 μΜ. One aspect of the embodiment includes wherein the peak concentration is maintained for 4 hours. One aspect of the embodiment includes wherein the dose is selected from a solid or liquid form. One aspect of the embodiment wherein the solid form comprises eriotinib complexed with a cyclodextrin and nano-encapsulated into a pharmaceutically acceptable carrier.
[0022] One embodiment of the present disclosure includes a method of treating brain, leptomeninges, or CNS metastases in a patient diagnosed with EGFR-mutant lung cancer comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule. One aspect of the embodiment includes wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule. One aspect of the embodiment includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
[0023] One embodiment includes a method of treating a patient diagnosed with gliablastoma comprising: orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the pulse dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule. In one aspect, the method includes wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule. In one aspect, the method includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. In one aspect, the method includes wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
[0024] Glioblastoma multiforme (GBM) is the most common brain tumour and has the worst prognosis. Epidermal growth factor receptor (EGFR) gene amplification, mutation and re-arrangement (all of which enhance tumor growth, survival, progression, and resistance to therapy) are frequently observed in primary GBM. The most common EGFR variant in GBM, the EGFRvll l, is characterized by a deletion of 267 amino acids in the extracellular domain, leading to a receptor which is unable to bind ligand yet is constitutively active. Epidermal growth factor receptor (EGFR) and EGFRvlll analysis is of current interest in glioblastoma. See, e.g., Gan et al., The Epidermal Growth Factor Receptor Variant III (EGFRvlll): Where the Wild Things are Altered, FEBS J, 2013 Nov., 280(21): 5350 - 70; Gan et al., The EGFRvlll Variant in Glioblastoma Multiforme, J. Clin. Neurosci., 2009 Jun., 16(6): 748- 54; Padfield et al., Current Therapeutics Advances Targeting EGFR and EGFRvlll in Glioblastoma, Front. Oncol., v. 5, published online 2015 Jan 29; and Iwata et al., Inhibition of mutant EGFRvlll transformed cells by tyrosine kinase inhibitor OSI-774 (Tarceva) [Abstract No. 79]. Proc. ASCO2002; 21.
[0025] The scope of the present invention includes all combinations of aspects and embodiments.
BRIEF DESCRIPTION OF THE FIGURES
[0026] Fig. 1 illustrates a synthetic scheme for the manufacture of eriotinib.
[0027] Fig. 2 is a graphical representation of best response of target lesions
(RECIST 1.1) in all patients with a radiographic assessment of response.
[0028] Fig. 3 is a graphical representation of progression-free survival probability from months since start of therapy.
[0029] Fig. 4 is an example of CNS response in a patient with untreated brain metastases on the regimen, namely pulse-continuous eriotinib or a pharmaceutically acceptable salt thereof.
[0030] Fig. 5 is a graphical representation of plasma concentration curves for eriotinib after multiple dosing. Blood samples were collected during cycle 1 week 1 day 1 at 0.5, 1 , 2, 4, and 6 hours after the first dose; before and four hours after eriotinib dosing on cycle 1 week 2 day 1 and cycle 1 week 4 day 1.
DETAILED DESCRIPTION
[0031] The following definitions are meant to clarify, but not limit, the terms defined. If a particular term used herein is not specifically defined, such term should not be considered indefinite. Rather, terms are used within their accepted meanings.
[0032] As referenced herein, one embodiment of the present disclosure includes a dosing regimen comprising: (i) orally administering one or more daily low dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more weekly high dose of eriotinib or a pharmaceutically acceptable salt thereof. As used herein, the terms "low dose" and "high dose" are used relative to the other as well as relative to a standard 100 mg to 150 mg daily dose of eriotinib or a pharmaceutically acceptable salt thereof. A low dose is lower than each of the standard dose and the high dose. Likewise, a high dose is higher than each of the standard dose and the low dose. One aspect of the embodiment includes a low dose of 50 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes a high dose from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes a high dose of 1200 mg of eriotinib or a pharmaceutically acceptable salt thereof.
[0033] As referenced herein, one embodiment of the present disclosure includes a weekly dosing regimen comprising: (i) orally administering one or more pulse dose of eriotinib or a pharmaceutically acceptable salt thereof; and (ii) orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof. As used herein, the term "pulse dose" refers to a higher dose of eriotinib or a
pharmaceutically acceptable salt thereof as compared to the daily dose. The term "pulse dose," as used herein may be determined by determining a dose that provides higher CSF concentrations than a standard 100 mg to 150 mg daily dose of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof. One aspect of the embodiment includes wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
[0034] As used herein, the term "unit dose" refers to a dose of medicine in an individual packet for convenience, safety, or monitoring.
[0035] As used herein, the term "tolerability" refers to the degree to which overt adverse effects of a drug can be tolerated by a patient. Tolerability of a particular drug can be discussed in a general sense, or it can be a quantifiable measurement as part of a clinical study. As one measure, tolerability may be determined by the rate of dropouts, or patients that forfeit participation in a clinical study due to one or more adverse effects.
[0036] As used herein, the term "pharmaceutically acceptable" refers to carrier(s), diluent(s), excipient(s) or salt forms of the compounds of the present invention that are compatible with the other ingredients of the formulation and not deleterious to the recipient of the pharmaceutical composition.
[0037] As used herein, the terms "effective amount", "therapeutic amount", and "effective dose" refer to an amount of the compound of the present invention sufficient to elicit a desired pharmacological or therapeutic effect. Moreover, the therapeutic amount of effective amount may thus result in an effective treatment of a disorder.
[0038] Treatment of a disorder may be manifested by delaying or preventing the onset or progression of the disorder, as well as the onset or progression of symptoms associated with the disorder. Treatment of a disorder may also be manifested by a decrease or elimination of symptoms, reversal of the progression of the disorder, as well as any other contribution to the well-being of the patient.
[0039] Erlotinib may exist in free form or, where appropriate, as a pharmaceutically acceptable derivative thereof. According to the invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a subject in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
[0040] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. A "pharmaceutically acceptable salt" means any non-toxic salt or salt of an ester of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an active metabolite or residue thereof. As used herein, the term "active metabolite or residue thereof" means that a metabolite or residue thereof is also an EGFR TKI.
[0041] Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge, et al., describe pharmaceutically acceptable salts in detail in J.
Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
Pharmaceutically acceptable salts include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci.4alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counter-ions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate and aryl sulfonate.
[0042] Polymorphism, the occurrence of different crystal forms, is a property of some molecules and molecular complexes. A single molecule, like erlotinib, may give rise to a variety of crystalline forms having distinct crystal structures and physical properties like melting point, x-ray diffraction pattern, infrared absorption fingerprint, and solid state NMR spectrum. One crystalline form may give rise to thermal behavior different from that of another crystalline form. Thermal behavior can be measured in the laboratory by such techniques as capillary melting point, thermogravimetric analysis ("TGA"), and differential scanning calorimetry ("DSC"), which have been used to distinguish polymorphic forms. The difference in the physical properties of different crystalline forms results from the orientation and intermolecular interactions of adjacent molecules or complexes in the bulk solid. Accordingly, polymorphs are distinct solids sharing the same molecular formula yet having distinct advantageous physical properties compared to other crystalline forms of the same compound or complex.
[0043] All forms of erlotinib or a pharmaceutically acceptable salt thereof are intended to be within the scope of the present invention.
[0044] Although it is possible to administer the compound of the present invention in the form of a bulk active chemical, it is preferred to administer the compound in the form of a pharmaceutical composition or formulation. Thus, one aspect the present invention includes pharmaceutical compositions comprising erlotinib and/or pharmaceutically acceptable salts thereof and one or more pharmaceutically acceptable carriers, diluents, or excipients.
[0045] The manner in which the compound of the present invention is administered can vary. The compound of the present invention is preferably administered orally. Preferred pharmaceutical compositions for oral administration include tablets, capsules, caplets, syrups, solutions, and suspensions. The pharmaceutical compositions of the present invention may be provided in modified release dosage forms such as time-release tablet and capsule formulations.
[0046] The compounds of the present invention may be used in the treatment of a variety of disorders and conditions and, as such, may be used in combination with a variety of other suitable therapeutic agents useful in the treatment or prophylaxis of those disorders or conditions.
[0047] As used herein, the terms "pharmaceutically acceptable composition" or "pharmaceutical composition" refer to erlotinib or a pharmaceutically acceptable salt thereof optionally admixed with one or more pharmaceutically acceptable carriers, diluents, or excipients. Pharmaceutically acceptable compositions preferably exhibit a degree of stability to environmental conditions so as to make them suitable for manufacturing and commercialization purposes.
[0048] Pharmaceutically acceptable compositions may comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. As an example, Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with erlotinib or a pharmaceutically acceptable salt thereof, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable
composition, its use is contemplated to be within the scope of this invention. Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc;
excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
[0049] Pharmaceutically acceptable compositions can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like.
[0050] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compound(s), a liquid dosage form may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[0051] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid may be incorporated in an injectable product. The injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[0052] In order to prolong the effect of a compound of the invention, it is often desirable to slow the absorption of the compound from subcutaneous or
intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[0053] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body
temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[0054] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or (a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, (c) humectants such as glycerol, (d) disintegrating agents such as agar- agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, (e) solution retarding agents such as paraffin, (f) absorption accelerators such as quaternary ammonium compounds, (g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, (h) absorbents such as kaolin and bentonite clay, and (i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[0055] 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 sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. 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 sugar as well as high molecular weight polethylene glycols and the like.
[0056] Erlotinib or a pharmaceutically acceptable salt thereof may be formulated using nanotechnology. Nanoparticles are attractive for medical purposes based on their unique features, such as their surface to mass ratio being larger than that of other particles, their quantum properties, and their ability to adsorb and carry other compounds. Nanoparticles may have dimensions below 0.1 pm or 100 nm.
Alternatively, a pharmaceutical composition may comprise relatively large (size >100 nm) nanoparticles, as needed for loading a sufficient amount of drug onto the particles. In addition, for drug delivery, not only engineered particles may be used as carrier, but also the drug itself may be formulated at a nanoscale, and then function as its own carrier. The composition of the engineered nanoparticles may vary.
Source materials may be of biological origin like phospholipids, lipids, lactic acid, dextran, chitosan, or have more chemical characteristics like various polymers, carbon, silica, and metals. Especially in the area of engineered nanoparticles of polymer origin there is a vast area of possibilities for the chemical composition. See, for example, Martins et al., Nanoparticle Drug Delivery Systems: Recent Patents and Applications in Nanomedicine, Recent Patents on Nanomedicine, 2013, 3(2), pp. 1 - 14. Many cytotoxic chemotherapeutic agents have poor aqueous solubility. These molecules are associated with poor physicochemical and biopharmaceutical properties, which makes the formulation difficult. One approach in this regard is the use of combination of cyclodextrin and nanotechnology in delivery system. See, for example, Gidwani et al., A Comprehensive Review on Cyclodextrin-Based Carriers for Delivery of Chemotherapeutic Cytotoxic Anticancer Drugs, BioMed Research International, Volume 2015, Article ID 198268, 15 pages. Cyclodextrins are chemically and physically stable macromolecules produced by enzymatic degradation of starch. They are water-soluble, biocompatible in nature with hydrophilic outer surface and lipophilic cavity. They have the shape of truncated cone or torus rather than perfect cylinder because of the chair conformation of glucopyranose unit. Cyclodextrins are classified as natural and derived
cyclodextrins. Natural cyclodextrins comprise three well-known industrially produced (major and minor) cyclic oligosaccharides. The most common natural cyclodextrins are α, β, and γ consisting of 6, 7, and 8 glucopyranose units. They are crystalline, homogeneous, and nonhygroscopic substances. Amongst these, β-cyclodextrin is used for complexation due to perfect cavity size, efficient drug complexation and loading, availability, and relatively low cost. Various hydrophilic, hydrophobic, and ionic derivatives have been developed and utilized to improve the physicochemical and biopharmaceutical properties of drug and inclusion capacity of natural cyclodextrins. Hydroxypropyl^-cyclodextrin (ΗΡ-β-CD), randomly methylated-β- cyclodextrin (RM^-CD), and sulfobutylether-p-cyclodextrin (SBE- -CD) are mostly preferred for complexation. Polymerized cyclodextrins are high molecular weight compounds, either water-soluble or insoluble. They offer the advantage of amorphous state and complexation without toxic effects. Examples of polymerized cyclodextrins are soluble anionic β-cyclodextrin polymer, soluble γ-cyclodextrin polymer, and epichlorohydrin β-cyclodextrin polymer. Cyclodextrin-based nanocarriers may be prepared by utilizing the concept of dual approach, which involves combination of two different approaches in a single delivery system. This covers two aspects firstly; the anticancer drug is complexed with suitable cyclodextrin and secondly encapsulation of complexed drug into carrier.
[0057] Erlotinib or a pharmaceutically acceptable salt thereof may also be in microencapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. I n such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g. , tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[0058] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms are prepared by dissolving or dispensing the compound in the proper medium . Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[0059] The compounds of this invention may be made by a variety of methods, including well-established synthetic methods. Illustrative general synthetic methods are set out below and then specific compounds of the invention are prepared in the working Examples. The compounds can be prepared according to the methods described below using readily available starting materials and reagents. In these reactions, variants may be employed which are themselves known to those of ordinary skill in this art but are not described in detail here.
[0060] Reference is made to U.S. RE 41 ,065 and incorporated herein by reference with regard to the description and synthesis of TARCEVA® (erlotinib hydrochloride).
EXAMPLES
Specific Example 1 : Pulse-continuous erlotinib results in impressive control of central nervous system disease [0061] Based on evolutionary mathematical modeling data, the present inventors tested a novel schedule of twice weekly pulse erlotinib in combination with daily erlotinib as initial EGFR TKI treatment in patients with EGFR-mutant lung cancers.
[0062] The trial was a prospective, open-label, single center phase 1 dose-escalation study of twice weekly pulse dose and daily erlotinib in patients with EGFR-mutant lung cancers. The primary endpoint of the study was the identification of the maximum tolerated dose of the combination of twice weekly high dose and daily low dose erlotinib. Secondary endpoints included the measurement of progression-free survival, overall survival, complete and partial response rate, determination of central nervous system progression and pharmacokinetic analysis. The study was registered at clinicaltrials.gov (NCT01967095). The trial was conducted after approval from the institutional review board at Memorial Sloan Kettering. All patients provided written informed consent.
[0063] Patients had stage IV or recurrent EGFR-mutant lung adenocarcinomas and no prior treatment with an EGFR TKI . Prior cytotoxic chemotherapy was allowed. Patients were required to have measurable disease per RECIST (Response
Evaluation Criteria in Solid Tumours), version 1.1). Patients must have had adequate organ function and a Karnofsky performance status of > 70%, with reference to Karnofsky et al. , The Use of the Nitrogen Mustards in the Palliative Treatment of Carcinoma - with Particular Reference to Bronchogenic Carcinoma, Cancer, 1948; 1 (4):634-56. Patients with clinically stable brain metastases, either treated or untreated, were eligible.
Study Design
[0064] The study used a standard 3+3 dose escalation design. Three patients were enrolled at each dose level and assessed for 1 full cycle before a dose escalation decision was made. No intra-patient dose escalation was allowed. Cycle 1 , week 1 (Days 1 -7) consisted of pulse dose erlotinib on days 1 and 2 without daily low dose erlotinib on days 3 to 7. For all subsequent weeks, patients received pulse dose erlotinib on days 1 and 2, and erlotinib 50 mg oral daily for 5 days on days 3 to 7 which was repeated weekly to complete 21 day cycles. Cycle 1 was 4 weeks to account for one week of lead in pulse dose erlotinib only. For each dose level, the dose of pulse erlotinib on days 1-2 was escalated (supplemental Table 1).
[0065] Supplemental Table 1 : Dose levels and the number of patients treated and any DLTs observed in the first cycle: Number treated DLT
Dose level 1 : 600mg 3 None
Dulse
Dose level 2: 750mg 3 None
Dulse
Dose level 3: 900mg 3 None
Dulse
Dose level 4: 1050mg 6 1 (grade 3 ALT/AST)
Dulse
Dose level 5: 1200mg 6 (+10 in None
Dulse exDansion1)
Dose level 6: 1350mg 3 2 (grade 2 rash, grade 3
Dulse mucositis')
[0066] Patients who did not experience a dose-limiting toxicity (DLT) continued treatment at the assigned dose until progression of disease, unacceptable toxicity, or withdrawal of informed consent. Dose reductions of the pulse dose were allowed for toxicity, in 150 mg increments.
[0067] Toxicity assessments: Patients were assessed five times during cycle 1 (28 days) and then every 21 days thereafter. Patient history, physical examination, complete blood count and serum chemistries were performed at each visit. Toxicity was graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE), version 4. Dose-limiting toxicity was defined as any grade 4 hematologic toxicity lasting greater than 5 days. Grade 3
thrombocytopenia with clinically significant bleeding was considered a DLT. All grade 3 or greater non-hematologic toxicities were considered DLTs with diarrhea, nausea and vomiting needing to be grade 3 for 72 hours despite maximal supportive care to qualify as a DLT. Once the maximum tolerated dose (MTD) was determined, an additional ten patients were enrolled at the MTD.
[0068] Response assessments: Response to therapy was assessed by interval imaging every 6 weeks with a CT scan with response evaluated per RECIST 1.1. After six cycles on treatment, patients could reduce the radiographic assessments to every 4 cycles (12 weeks).
[0069] Pharmacokinetic analysis: Whole-blood samples for plasma were collected at prescheduled time points for pharmacokinetic (PK) analysis. Plasma levels of erlotinib were determined using a validated liquid chromatography-tandem mass spectrometry assay. LC-MS/MS was performed using a Phenomenex Kinetex 2.6 μίτι core-shell Biphenyl column (50x3 mm) and Shimadzu HPLC. MS detection was using an AB SCIEX API 4000 triple quadrupole mass spectrometer operating in positive ion electrospray ionization (ESI) mode. Standard PK parameters were calculated using a noncompartmental method (WiNNonlin, Pharsight).
[0070] Statistical Analysis: Descriptive statistics were used to summarize the clinical characteristics of the patients. Progression-free survival was estimated using the Kaplan-Meier method, and defined as the time from start of study therapy until progression or death. Patients who did not experience the event of interest were censored at the date they came off study or date of last assessment if still receiving study therapy. Response rates were calculated using binomial proportions and exact 95% CIs. All statistical analyses were performed using R 3.2.2 (R Development Core Team) including the "survival" and "Hmisc" packages.
Results
[0071] 34 patients were enrolled onto this study, including 24 patients on the dose escalation portion of the phase 1 study and 10 patients in an expansion cohort at the MTD. In total, sixteen patients were treated at the MTD. The clinical characteristics of all patients are listed in Table 1.
[0072] Table 1 : Patient characteristics
Figure imgf000025_0001
SRS 1
Craniotomy
Untreated 1
6
No 22
[0073] Eleven patients (32%) had brain metastases at diagnosis, 5 of whom received treatment (surgery or radiation) for their CNS disease prior to study enrollment.
[0074] Determination of the maximum tolerated dose: There were no DLTs seen at dose levels at the 600, 750 and 900mg pulse dose levels. At the erlotinib 1050mg pulse dose, there was one DLT of grade 3 transaminitis (Supplemental Table 1 , above).
[0075] At 1350mg pulse dose level, there were two DLTs: grade 3 rash and grade 3 mucositis. We then enrolled an additional three patients on to the 1200mg erlotinib pulse dose level with no DLTs seen. The 1200mg pulse on days 1 and 2 and 50mg erlotinib on days 3 to 7 was determined to be the MTD.
[0076] Adverse events: All 34 patients were evaluable for toxicity (Table 2). No grade four or five toxicities were reported with no deaths on study. Most drug-related toxicities were grade 1 and 2. The most common (>25%) drug-related adverse events were rash, diarrhea, nausea, fatigue, mucositis, pruritus, vomiting, increased bilirubin and dry skin (Table 2). Of the sixteen patients treated at the MTD, only three of sixteen required a dose reduction of the pulse dose erlotinib; the reasons for dose reduction were grade 3 rash, intolerable grade 2 rash and intolerable grade 2 diarrhea. All three patients eventually were lowered to erlotinib 900mg as the pulse dose. Six patients were removed from the study for toxicity: hyperbilirubinemia (1 ), possible pneumonitis (1), diarrhea (2), transaminitis (1) and mucositis (1).
[0077] Table 2: Study drug-related adverse events seen in >10% of patients
Figure imgf000026_0001
Adverse Event Grade 1 Grade 2 Grade 3 Grade 4 Total (any grade)
Bilirubin increased 4 (12%) 5 (15%) 2 (6%) 0 11 (32%)
Dry skin 10 (29%) 0 0 0 10 (29%)
ALT elevated 6 (18%) 1 (3%) 1 (3%) 0 8 (24%)
Alopecia 7 (21 %) 0 0 0 7 (21 %)
AST increased 5 (15%) 1 (3%) 0 0 6 (18%)
Paronychia 4 (12%) 2 (6%) 0 0 6 (18%)
Anorexia 3 (9%) 2 (6%) 0 0 5 (15%)
Anemia 2 (6%) 1 (3%) 1 (3%) 0 4 (12%)
[0078] Efficacy: All 34 patients were evaluable for response. Four patients came off study prior to the first follow-up radiographic assessment (3 due to toxicity, 1 due to noncompliance) and were counted as non-responders in our intent to treat analysis. Twenty-four patients had confirmed partial responses and one patient had a complete response (Figure 2). The overall response rate was 74% (95% CI: 60- 84%). All patients had a decrease in the sum of their target lesions. The maximum radiographic tumor change of target lesions for each patient with a follow up radiographic assessment is depicted in Figure 2. The median progression-free survival is 9.9 months (95% CI 5.8-15.4 months) (Figure 3). The overall survival estimate is not yet mature. Six subjects have died: four due to lung cancer progression, one due to Parkinson's disease and one due to a second primary gastric cancer.
[0079] Central nervous system activity: A brain MRI was required at enrollment. Eleven patients (32%) had brain metastases at study entry. No patient had leptomeningeal disease. Prior to the trial, 3 patients had whole brain radiation, 1 had stereotactic radiosurgery and 1 had a surgical resection. All 6 patients with untreated brain metastases had evidence of tumor regression in the CNS (Figure 4). No patients had clinical CNS progression on study treatment. Of the 19 patients who came off study for progression of disease, 16 had CNS imaging within three months of coming off study and none had radiographic evidence of new or progressing brain metastases.
[0080] Table 3: Molecular findings at diagnosis and at acquired resistance for patients who came off study for progression Baseline Acquired Resistance (off study for progression)
Pt EGFR Concurrent mutations EGFR T790M Concurrent mutations
1 Ex19 del Not done Ex19 del Positive Not done
2 Ex19 del Not done Ex19 del Positive AURKA amp, GNAS amp, MEF2B gain,
CCNE1 gain, AKT2 gain, MCL1 gain
3 Ex19 del Not done Ex19 del Negative CDK2NB del, CDKN2A del, BAP1 G109V,
ETV1 R163Q, NOTCH3 G1689A
4 Ex19 del Not done Ex19 del Positive Not done
5 Ex19 del Not done Ex19 del Positive CCND2 amp, VEGFA amp, TERT gain, MDC1 gain, CDKN2B del, CDKN2A del, PAX5 del, AKT3 R166*, CSF1 R A74T, INSR D1259A
6 L858R Not done L858R Negative Not done
7 L858R Not done L858R Failed Failed
8 L858R Not done L858R Positive TP53 M246V, APC S1 100fs, BRIP1 I983fs,
FBXW7 C573Y, RUNX1 ex5 del
9 Ex19 del Not done Ex19 del Positive Not done
10 Ex19 del Not done Ex19 del Positive Not done
11 Ex19 del TP53 E298X, EGFR amp, MYC Ex19 del Positive TP53 E298X, EGFR amp, ERBB2 amp,
amp, RECQL4 amp, RB1 del, RAD 21 amp, MYC amp, SDHA amp, TERT PDGFR R804Q, SETD2 N16343fs amp, IL7R amp, RICTOR amp, RECQL4 amp
, RB1 del, PDGFR R804Q, SETD2
12 Ex19 del TP53 Q104X, EGFR amp, MYC Ex19 del Positive Failed
amp, FLT1 R1 146T, MCL1 G3S,
PTPRD S569L
13 Ex19 del Not done Ex19 del Positive Failed
14 L858R EGFR V689F, FGFR3 R399C, L858R Negative EGFR V689F, FGFR3 R399C, TP53 P36fs, AR
TP53 P36fs, AR G578X, ATRX G578X, ATRX S784F, BARD1 N73S, EPHA3 S784F, BARD1 N73S, EPHA3 F152S, RET E164K
F152S, SMARCA4 P1277L
15 Ex19 del Not done Ex19 del Positive EGFR L747P, TP53 Y163N, PTEN F271 S
16 Ex19 del GNA1 1 R210L, TP53 C275W, Ex19 del Positive GNA1 1 R210L, TP53 C275W, NKX2-1 amp,
PMS2 amp, RAC1 amp, EGFR FOXA1 amp, CENPA E50fs, PLCG2 P999T, amp, NKX2-1 amp, FOXA1 amp, RB1 ex10 SV, STAG2 D26E, TET1 Q683P PTEN del, GNAS K25N, RB1 ex10
SV, STAG2 D26E, TET1
17 Ex19 del Not done Ex19 del Positive PIK3CA E542K, TP53 V73fs, CCNE1 amp,
EGFR amp, RICTOR gain, IL7R I121fs, MAP2K4 R304*, MLL3 D3461 N
18 Ex19 del Not done Ex19 del Positive PIK3CA E542K, CTNNB1 S33F, LATS1
K652R, TGFBR2 K399fs
19 Ex19 del PIK3CA E545K, TP53 ex5 del, Ex19 del Negative PIK3CA E545K, TP53 ex5 del, MET gain,
EGFR gain, EPHB1 E144K, EGFR gain, SETD2 T2316A
Amp= amplification, del= deletion [0081] Mechanisms of acquired resistance: Of the nineteen patients that progressed on study therapy, six continued daily eriotinib at standard doses for > 2 months after discontinuation of study therapy. Nineteen patients had a tumor biopsy at the time of progression. One patient's biopsy sample was inadequate for molecular analysis. Fourteen of eighteen (78%, 95% CI 54%-91%) patients had EGFR T790M identified in their rebiopsy specimen. Of the eighteen patients, eleven patients had next generation sequencing of their acquired resistance biopsy sample, and four had a paired pre-treatment tumor sample as well. Multiple concurrent mutations were found in both baseline and acquired resistance tumor samples in addition to the sensitizing EGFR mutation which was identified in all samples. Acquired molecular alterations not present in baseline samples included HER2 and MET amplification which have been previously reported (33, 34) as well as amplification in RAD21 , SDHA, TERT, IL7R and RICTOR, and mutations in RET E164K, CENPA E50fs, and PLCG2 P999T.
[0082] Pharmacokinetic analysis: Samples were collected from all 34 patients in the phase 1 study at the pre-specified time points. Median plasma concentration-time profiles are shown in Figure 5. There was significant intra-patient variability in erlotinib plasma concentrations. No significant increase in peak plasma concentration was observed with increasing pulse doses of erlotinib from 600 mg to 1 ,350 mg, at any time point examined during Cycle 1. This was also confirmed by analysis of the major erlotinib drug metabolites (OSI-420 and M1 1), which showed no significant dose-dependence with increasing pulse doses of erlotinib. The peak plasma concentration occurred after the second day of weekly pulse dose at Week 2, with median plasma concentration at 4 hr post-administration reaching 5,393 ng/mL
(range 2,600 - 9355 ng/mL), approximately 13.7 micromolar, more than 5 times the peak plasma concentration seen with standard 150mg dosing (35). The median plasma trough concentration before the pulse dosing at the start of week 4 was 435 ng/mL (range: 29 - 1655 ng/mL), a concentration of 1.1 micromolar. This level at week 4 is consistent with 24 hr plasma concentrations observed following standard 150 mg dosing (35).
[0083] Patient disposition: six patients remain enrolled in the study. Nineteen patients discontinued study therapy because of progressive disease, and six others for adverse events discussed above. Two patients stopped study therapy due to non- compliance and one due to the development of an unrelated gastric
adenocarcinoma.
[0084] We believe this is the first time evolutionary cancer modeling was used to optimize dosing of a targeted therapy. This dosing schedule derived from
mathematical modeling did not delay the development of resistance or prevent the emergence of EGFR T790M despite validation in vitro and in vivo pre-clinical models. A likely explanation is that the peak plasma concentrations in patients may not have been high enough to correspond to the peak concentrations utilized in the pre-clinical models. The pulse and low dose concentrations used in the pre-clinical models were 20 micromolar and 1 micromolar, respectively. The median peak and trough concentration obtained at the MTD in our study was 13.7 micromolar and 1.1 micromolar, respectively. We were limited in exploring higher pulse dose levels due by toxicity resulting in DLTs. I n addition, in the relatively few samples obtained in this phase 1 study (n=3-6 for dose levels 1 -4 and 6), there were no significant increases in plasma peak concentration with increasing pulse doses from 600mg to 1 ,350mg, presumably due to limitations in drug absorption or due to drug clearance or metabolism. Even if toxicity were not an issue at higher dose levels, it is uncertain whether shorter dosing intervals or higher doses would result in the peak plasma concentrations required to delay EGFR T790M. The peak concentrations of eriotinib required to delay EGFR T790M do not appear achievable clinically. There are other potential limitations to our clinical model that may have contributed to our negative findings. The original modeling utilized both pulse afatinib and eriotinib, with the combination of pulse afatinib and daily eriotinib as superior at delaying resistance. Differences between eriotinib and afatinib may translate to disparate activity against EGFR T790M , and our results may have been different if we had utilized pulse afatinib. I n addition, there are limitations inherent to any model system that considers multiple variables but cannot fully account for the entire complexity of all host and tumor factors.
[0085] Pulse-continuous eriotinib was well tolerated. The most common treatment related adverse events were rash, diarrhea, nausea, fatigue and mucositis. When compared to prospective studies of standard daily dosed eriotinib and afatinib, BR.21 and Lux-Lung 3, respectively, toxicity was similar. See, Sequist et al. , Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung
Adenocarcinoma With EGFR Mutations, Journal of Clinical Oncology, 2013,
31 (27):3327-34, and Shepherd et al. , Eriotinib in previously treated non-small-cell lung cancer, The New England Journal of Medicine, 2005, 353(2) : 123-32.
[0086] Surprisingly, the study showed no significant increase in toxicity with pulse continuous eriotinib, despite a five times increase in the median peak plasma concentration of eriotinib compared to standard dosing. See, Hamilton et al. , Effects of smoking on the pharmacokinetics of eriotinib, Clinical Cancer Research, 2006, 12(7 Pt 1 ):2166-71 . Pulse dosing may result in decreased toxicity compared to the same amount of drug divided among daily doses, as toxicity may be related to drug trough or steady state concentrations, rather than peak concentrations. [0087] This observation is corroborated by our previous study of once weekly pulse dose eriotinib at 2000mg once weekly which also showed a similar toxicity profile to standard daily dose eriotinib. See, Milton et al., A phase l/ll study of weekly high- dose eriotinib in previously treated patients with nonsmall cell lung cancer, Cancer, 2006, 107(5): 1034-41 . The present study demonstrates that pulse-continuous eriotinib is a feasible dosing schedule that is tolerated by most people.
[0088] The efficacy of pulse continuous eriotinib was similar to standard dose eriotinib for lung cancer. The overall response rate was 74% (95% CI 60-84%) and the median progression free survival was 9.9 months (95% CI 5.8-15.4 months). The dose and schedule of eriotinib assessed in this clinical trial did not improve response rates, prolong progression free survival or delay systemic resistance. The 78% (95% CI 54%-91 %) frequency of EGFR T790M at the time of acquired resistance was comparable what is seen with standard dosing. Further study is needed to investigate whether drug dosing alters the mechanisms of resistance that emerge.
Central nervous system metastases are a critical issue in the management of patients with EGFR-mutant lung cancers. Due to a prolonged disease course and improved overall survival, EGFR-mutant cancers may be enriched for CNS involvement with the cumulative incidence of CNS metastases approaching 60%. See, for example, Heon et al., The impact of initial gefitinib or eriotinib versus chemotherapy on central nervous system progression in advanced non-small cell lung cancer with EGFR mutations, Clinical Cancer Research, 2012, 18(16):4406-14; Omuro et al., High incidence of disease recurrence in the brain and leptomeninges in patients with nonsmall cell lung carcinoma after response to gefitinib, Cancer, 2005, 103(1 1 ):2344-8; Lee et al., Frequent central nervous system failure after clinical benefit with epidermal growth factor receptor tyrosine kinase inhibitors in Korean patients with nonsmall-cell lung cancer, Cancer, 2010, 116(5): 1336-43; Heon et al., Development of central nervous system metastases in patients with advanced non- small cell lung cancer and somatic EGFR mutations treated with gefitinib or eriotinib, Clinical Cancer Research, 2010, 16(23): 5873-82; Rangachari et al. , Brain
metastases in patients with EGFR-mutated or ALK-rearranged non-small-cell lung cancers, Lung Cancer, 2015, 88(1): 108-1 1 ; and Kris et al., Using Multiplexed Assays of Oncogenic Drivers in Lung Cancers to Select Targeted Drugs, J. Am. Med. Assoc. , 2014; 31 1 (19): 1998-2006.
[0089] Despite new treatments, such as osimertinib, that have demonstrated impressive systemic efficacy, CNS failures on treatment are common. In addition, patients with CNS involvement demonstrate lower response rate and shorter progression free survival with the 3rd generation EGFR TKIs compared to patients without CNS metastases. See, Camidge et al., Activity of Rociletinib in EGFR Mutant NSCLC Patients With a History of CNS Involvement World Conference on Lung Cancer, 2015, Oral presentation at the World Conference on Lung Cancer, Denver, CO.
[0090] This is believed to be the first demonstration of pre-treatment incidence of CNS involvement in study subjects and also the rate of CNS progression on an EGFR TKI therapy. None of the prior large prospective studies of EGFR tyrosine kinase inhibitors include this information. See, for example, Rosell et al., Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial, The Lancet Oncology, 2012, 13(3):239-46; Sequist et al., Phase III Study of Afatinib or Cisplatin Plus Pemetrexed in Patients With Metastatic Lung Adenocarcinoma With EGFR Mutations, Journal of Clinical Oncology, 2013, 31 (27):3327-34; Janne et al., Randomized Phase II Trial of Eriotinib Alone or With Carboplatin and Paclitaxel in Patients Who Were Never or Light Former Smokers With Advanced Lung Adenocarcinoma: CALGB 30406 Trial, Journal of Clinical Oncology, 2012, 39(17): 2063-9; Zhou et al., Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation- positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open- label, randomised, phase 3 study, The Lancet Oncology, 2011 , 12(8):735-42; and Mitsudomi et al., Gefitinib versus cisplatin plus docetaxel in patients with non-small- cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial, The Lancet Oncology, 2010, 11 (2): 121-8.
[0091] As site of failure has prognostic as well as treatment implications, the present inventors suggest the rate of CNS failure be explicitly reported in all future prospective studies in lung cancers. Within EGFR-mutant lung cancers,
understanding the CNS efficacy of drugs is critically important and will help in the selection between and among drugs that may have similar systemic effect.
[0092] The pulse continuous erlotinib dosing resulted in response in pre-existing brain metastases and prevention of new brain metastases while on treatment. Using retrospective data, CNS progression with or without concurrent systemic progression can occur in up to 33% of patients treated with standard dosing of EGFR TKI, with isolated CNS progression in up to 19% of cases. Heon et al., The impact of initial gefitinib or erlotinib versus chemotherapy on central nervous system progression in advanced non-small cell lung cancer with EGFR mutations, Clinical Cancer
Research, 2012, 18(16):4406-14; Omuro et al., High incidence of disease recurrence in the brain and leptomeninges in patients with nonsmall cell lung carcinoma after response to gefitinib, Cancer, 2005, 103(11):2344-8; and Lee et al., Frequent central nervous system failure after clinical benefit with epidermal growth factor receptor tyrosine kinase inhibitors in Korean patients with nonsmall-cell lung cancer, Cancer, 2010, 116(5):1336-43.
[0093] The present data suggests that the CNS efficacy of this dosing schedule may be superior. The pulse continuous dosing allows for increased CNS penetration but continued daily dosing maintains systemic control of disease.
[0094] In summary, this is the first prospective study to assess an alternative dosing of an approved tyrosine kinase inhibitor based on evolutionary modeling. The regimen was well tolerated with a similar toxicity profile to standard dose erlotinib. Pulse continuous erlotinib did not lengthen progression-free survival or prevent the emergence of EGFR T790M as was hypothesized, perhaps due to the inability to achieve high enough plasma concentrations with the pulse doses used in this study. Surprisingly, however, none of the patients on study had evidence of new or progressing CNS disease on this regimen. Specific Example 2 - Adjuvant therapy
[0095] Adjuvant therapy or care, also called adjunct therapy or adjunctive therapy or care, is therapy that is given in addition to the primary, main, or initial therapy to maximize its effectiveness. As an adjuvant agent modifies the effect of another agent, so adjuvant therapy modifies other therapy.
[0096] For example, resectable non-small-cell lung cancer (NSCLC) accounts for 20% to 25% of lung cancer cases diagnosed annually; however, only 60% of patients survive 5 years after surgery. The first trial to demonstrate a significant survival benefit with adjuvant cisplatin-based chemotherapy was reported a decade ago. Subsequently, additional randomized trials confirmed the role of adjuvant chemotherapy in patients with pathologic stage II and III NSCLC, and subset analyses suggested a benefit in patients with large IB tumors. A meta-analysis provided further support for adjuvant chemotherapy. Kelly et al., Adjuvant Erlotinib Versus Placebo in Patients With Stage IB-IIIA Non-Small-Cell Lung Cancer (RADIANT): A Randomized, Double-Blind, Phase III Trial, Journal of Clinical Oncology, Published Ahead of Print on August 31 , 2015 as
10.1200/JCO.2015.61.8918, sets forth a study design, patient population, assignment and masking protocol, study assessment, outcomes, and statistical analysis that could be used for a study of adjuvant efficacy. As such, that reference is incorporated herein in its entirety for such purpose. As set forth in Kelly et al., id, the signal of activity observed with eriotinib in patients with GFf?-expressing tumors combined with its oral availability and mild nonhematologic toxicity profile led to its evaluation in earlier stages of lung cancer. The Randomized Double-Blind Trial in Adjuvant NSCLC With Tarceva (RADIANT) study evaluated whether eriotinib would increase disease-free survival (DFS) in patients with completely resected stage IB to IIIA NSCLC whose tumors express EGFR.
[0097] As a further reference for conducting an adjuvant trial, Pennell et al.,
SELECT: A Multicenter Phase II Trial of Adjuvant Eriotinib in Resected Early-stage EGFR Mutation-positive NSCLC, presented at the 2014 ASCO meeting, provides study design, statistical considerations, patient characteristics, disease
characteristics, treatment durations, dose monitoring, and outcomes and is hereby incorporated by reference in its entirety for such purpose.
[0098] The specific pharmacological responses observed may vary according to and depending on the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of
administration employed, and such expected variations or differences in the results are contemplated in accordance with practice of the present invention.
[0099] Although specific embodiments of the present invention are herein illustrated and described in detail, the invention is not limited thereto. The above detailed descriptions are provided as exemplary of the present invention and should not be construed as constituting any limitation of the invention. Modifications will be obvious to those skilled in the art, and all modifications that do not depart from the spirit of the invention are intended to be included with the scope of the appended claims.

Claims

CLAIMS What is claimed is:
1. A dosing regimen comprising:
a. orally administering one or more daily low dose of erlotinib or a
pharmaceutically acceptable salt thereof; and
b. orally administering one or more weekly high dose of erlotinib or a pharmaceutically acceptable salt thereof
2. The dosing regimen of claim 1 , wherein the low dose is of 50 mg of erlotinib or a pharmaceutically acceptable salt thereof.
3. The dosing regimen of any one of claims 1 or 2, wherein the high dose is from about 600 mg to about 1350 mg of erlotinib or a pharmaceutically acceptable salt thereof.
4. The dosing regimen of any one of claims 1 - 4, wherein the high dose is 1200 mg of erlotinib or a pharmaceutically acceptable salt thereof.
5. The dosing regimen of any one of claims 1 - 5, wherein the daily dose is once daily.
6. The dosing regimen of any one of claims 1 - 6, wherein the weekly dose is once or twice weekly.
7. The dosing regimen of any one of claims 1 - 7, wherein the regimen improves tolerability over a standard 100 mg to 150 mg daily dose of erlotinib or a pharmaceutically acceptable salt thereof.
8. The dosing regimen of claim 7, wherein the improved tolerability is manifested by a lower incidence in one or more of rash, diarrhea, nausea, fatigue, or mucositis.
9. A weekly dosing regimen comprising:
a. orally administering one or more pulse dose of erlotinib or a pharmaceutically acceptable salt thereof; and
b. orally administering one or more daily dose of erlotinib or a pharmaceutically acceptable salt thereof.
10. The dosing regimen of claim 9, wherein the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
11. The dosing regimen of any one of claims 9 or 10, wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
12. The dosing regimen of claims 9 - 11 , wherein the pulse dose is from about
600 mg to about 1350 mg of erlotinib or a pharmaceutically acceptable salt thereof.
13. The dosing regimen of claims 9 - 12, wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
14. The dosing regimen of claims 9 - 13, wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
15. The dosing regimen of claims 9 - 14, wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
16. A dosing regimen of eriotinib or a pharmaceutically acceptable salt thereof comprising:
a. administering to a patient in need thereof eriotinib or a
pharmaceutically acceptable salt thereof in amounts that provide a steady state concentration (Ctr0ugh) of 0.05 - 10 μΜ in the patient; b. administering to a patient in need thereof eriotinib or a
pharmaceutically acceptable salt thereof in amounts that provide a pulsed concentration (Cmax) of 5 - 50 μΜ in the patient.
17. The dosing regimen of claim 16, wherein the steady state concentration
(Ctrougn) is approximately 0.5 μΜ.
18. The dosing regimen of claim 16, wherein the pulsed concentration (Cmax) is approximately 20 μΜ.
19. The dosing regimen of any one of claims 16 or 18, wherein the pulsed
concentration is maintained for 4 hours.
20. A method of treating or preventing central nervous system (CNS) metastases in a patient in need thereof comprising:
a. orally administering one or more pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof; and
b. orally administering one or more daily dose of eriotinib or a
pharmaceutically acceptable salt thereof.
21. The method of claim 20, wherein the CNS metastases is leptomeningeal carcinomatosis.
22. The method of any one of claims 20 or 21 , wherein the patient has extracranial metastatic non-small cell lung cancer.
23. The method of any one of claims 20 - 22, wherein the pulse dose is
administered once daily on each of days 1 and 2 of a weekly dosing schedule.
24. The method of any one of claims 20 - 23, wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
25. The method of any one of claims 20 - 24, wherein the pulse dose is from
about 600 mg to about 1350 mg of erlotinib or a pharmaceutically acceptable salt thereof.
26. The method of any one of claims 20 - 25, wherein the pulse dose is 1200 mg erlotinib or a pharmaceutically acceptable salt thereof.
27. The method of any one of claims 20 - 26, wherein the daily dose is from
about 25 mg to about 50 mg of erlotinib or a pharmaceutically acceptable salt thereof.
28. The method of any one of claims 20 - 27, wherein the daily dose is 50 mg erlotinib or a pharmaceutically acceptable salt thereof.
29. The method of any one of claims 20 - 28, wherein the patient with or without pre-existing brain metastases has no progression.
30. The method of any one of claims 20 - 29, wherein the patient's cerebrospinal fluid (CSF) is cleared of malignant cells.
31. The method of any one of claims 20 - 30, wherein the patient achieves a clinical benefit.
32. The method of claim 31 , wherein the clinical benefit is one or more of
decrease in tumor size, suppression of tumor growth, delayed time to progression in CNS, unobservable increase in size of brain, leptomeninges, or CNS metastases or lesions, unobservable new brain, leptomeninges, or CNS metastases or lesion, delay in time from surgery to recurrence, increase in treatment options, delay or decrease in leptomeningeal seeding in the CSF, or an ability to delay whole brain radiation.
33. A method for controlling formation of metastatic brain, leptomeninges, or CNS lesions in a patient with non-small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation with or without pre-existing brain metastases comprising:
a. orally administering one or more pulse dose of erlotinib or a
pharmaceutically acceptable salt thereof; and
b. orally administering one or more daily dose of erlotinib or a
pharmaceutically acceptable salt thereof.
34. The method of claim 33, wherein the pulse dose is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
35. The method of any one of claims 33 or 34, wherein the daily dose is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
36. The method of any one of claims 33 - 35, wherein the pulse dose is from about 600 mg to about 1350 mg of erlotinib or a pharmaceutically acceptable salt thereof.
37. The method of any one of claims 33 - 36, wherein the pulse dose is 1200 mg erlotinib or a pharmaceutically acceptable salt thereof.
38. The method of any one of claims 33 - 37, wherein the daily dose is from
about 25 mg to about 50 mg of erlotinib or a pharmaceutically acceptable salt thereof.
39. The method of any one of claims 33 - 38, wherein the daily dose is 50 mg erlotinib or a pharmaceutically acceptable salt thereof.
40. The method of any one of claims 33 - 39, wherein the patient experiences no tumor formation in the central nervous system.
41. The method of any one of claims 33 - 40, wherein the patient's cerebrospinal fluid is cleared of malignant cells.
42. The method of any one of claims 33 - 41 , wherein the patient achieves an objective or complete response or disease control.
43. The method of claim 42, wherein the control is durable.
44. A combination comprising:
a. erlotinib or a pharmaceutically acceptable salt thereof
i. orally administered in one or more pulse dose of erlotinib or a pharmaceutically acceptable salt thereof; and ii. orally administered in one or more daily dose of erlotinib or a pharmaceutically acceptable salt thereof; and b. one or more dose of one or more therapeutic compounds
administered in an effective amount thereof.
45. The combination of claim 44, wherein the one or more therapeutic
compounds are one or more oncology therapeutic compounds.
46. The combination of any one of claims 44 or 45, wherein the one or more therapeutic compounds are one or more inhibitors of epidermal growth factor receptor (EGFR).
47. The combination of any one of claims 44 - 46, wherein the one or more
therapeutic compounds are one or more inhibitors of EGFR is a mutant- selective inhibitor of EGFR.
48. The combination of claim 44, wherein the one or more therapeutic
compounds are selected from one or more of rociletinib (CO-1686), osimertinib (AZD9291), HM61713, Bl 1482694, or other third or later generation EGFR TKI.
49. The combination of any one of claims 44 - 48, wherein the pulse dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
50. The combination of any one of claims 44 - 49, wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
51. The combination of any one of claims 44 - 50, wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
52. The combination of claims 44 - 51 , wherein the pulse dose is 1200 mg
eriotinib or a pharmaceutically acceptable salt thereof.
53. The combination of claims 44 - 52, wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
54. The combination of claims 44 - 53, wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
55. The combination of claims 44 - 54, wherein each dose is selected from a solid or liquid form.
56. The combination of claim 55, wherein the solid form comprises one or more of eriotinib or the one or more therapeutic compounds complexed with a cyclodextrin and nano-encapsulated into a pharmaceutically acceptable carrier.
57. A method of treating brain, leptomeninges, or CNS metastases in a patient diagnosed with EGFR-mutant lung cancer comprising:
a. orally administering one or more pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof; and
b. orally administering one or more daily dose of eriotinib or a
pharmaceutically acceptable salt thereof.
58. The method of claim 57, wherein the pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
59. The method of any one of claims 57 or 58, wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
60. The method of any one of claims 57 - 59, wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
61. The method of any one of claims 57 - 60, wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
62. The method of any one of claims 57 - 61 , wherein the daily dose is from
about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
63. The method of any one of claims 57 - 62, wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
64. A unit dose of eriotinib or a pharmaceutically acceptable salt thereof,
comprising a pharmaceutical composition comprising erotinib or a pharmaceutically acceptable salt thereof which achieves a peak concentration (Cmax) of 5 - 50 μΜ in a patient.
65. The unit dose of claim 64, wherein the unit dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
66. The unit dose of any one of claims 64 or 65, wherein the unit dose is about 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
67. The unit dose of any one of claims 64 - 66, wherein the peak concentration (Cmax) is approximately 20 μΜ.
68. The unit dose of any one of claims 64 - 67, wherein the peak concentration is maintained for 4 hours.
69. The unit dose of claims 64 - 68, wherein the dose is selected from a solid or liquid form.
70. The unit dose of claim 69, wherein the solid form comprises eriotinib
complexed with a cyclodextrin and nano-encapsulated into a
pharmaceutically acceptable carrier.
71. A method of treating a patient diagnosed with non-small cell lung cancer (NSCLC) that harbors epidermal growth factor receptor (EGFR) mutation comprising:
a. orally administering one or more pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof; and b. orally administering one or more daily dose of eriotinib or a pharmaceutically acceptable salt thereof.
72. The method of claim 71 , wherein the pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
73. The method of any one of claims 71 or 72, wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
74. The method of any one of claims 71 - 73, wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
75. The method of any one of claims 71 - 74, wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
76. The method of any one of claims 71 - 75, wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
77. The method of any one of claims 71 - 76, wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
78. A method of treating a patient diagnosed with gliablastoma comprising: a. orally administering one or more pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof; and
b. orally administering one or more daily dose of eriotinib or a
pharmaceutically acceptable salt thereof.
79. The method of claim 78, wherein the pulse dose of eriotinib or a
pharmaceutically acceptable salt thereof is administered once daily on each of days 1 and 2 of a weekly dosing schedule.
80. The method of any one of claims 78 or 79, wherein the daily dose of eriotinib or a pharmaceutically acceptable salt thereof is administered once daily on each of days 3 to 7 of a weekly dosing schedule.
81. The method of any one of claims 78 - 80, wherein the pulse dose is from about 600 mg to about 1350 mg of eriotinib or a pharmaceutically acceptable salt thereof.
82. The method of any one of claims 78 - 81 , wherein the pulse dose is 1200 mg eriotinib or a pharmaceutically acceptable salt thereof.
83. The method of any one of claims 78 - 82, wherein the daily dose is from about 25 mg to about 50 mg of eriotinib or a pharmaceutically acceptable salt thereof.
84. The method of any one of claims 78 - 83, wherein the daily dose is 50 mg eriotinib or a pharmaceutically acceptable salt thereof.
PCT/US2016/024232 2016-03-25 2016-03-25 Pulse dosing regimen and methods for treatment WO2017164887A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2016/024232 WO2017164887A1 (en) 2016-03-25 2016-03-25 Pulse dosing regimen and methods for treatment

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2016/024232 WO2017164887A1 (en) 2016-03-25 2016-03-25 Pulse dosing regimen and methods for treatment

Publications (2)

Publication Number Publication Date
WO2017164887A1 true WO2017164887A1 (en) 2017-09-28
WO2017164887A8 WO2017164887A8 (en) 2017-11-09

Family

ID=59900662

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/024232 WO2017164887A1 (en) 2016-03-25 2016-03-25 Pulse dosing regimen and methods for treatment

Country Status (1)

Country Link
WO (1) WO2017164887A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022251539A3 (en) * 2021-05-26 2023-01-05 C4 Therapeutics, Inc. Egfr degraders to treat cancer metastasis to the brain or cns

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008127659A2 (en) * 2007-04-13 2008-10-23 University Of Texas Southwestern Medical Center Combination therapy for cancer
US20140066465A1 (en) * 2012-09-04 2014-03-06 The Cleveland Clinic Foundation CANCER TREATMENT USING TYROSINE KINASE AND NF-kB INHIBITORS
WO2014140989A2 (en) * 2013-03-14 2014-09-18 Pfizer Inc. Combination of an egfr t790m inhibitor and an egfr inhibitor for the treatment of non-small cell lung cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008127659A2 (en) * 2007-04-13 2008-10-23 University Of Texas Southwestern Medical Center Combination therapy for cancer
US20140066465A1 (en) * 2012-09-04 2014-03-06 The Cleveland Clinic Foundation CANCER TREATMENT USING TYROSINE KINASE AND NF-kB INHIBITORS
WO2014140989A2 (en) * 2013-03-14 2014-09-18 Pfizer Inc. Combination of an egfr t790m inhibitor and an egfr inhibitor for the treatment of non-small cell lung cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
PASTORINO ET AL.: "Abstract 4652: Cerebrospinal fluid pharmacokinetics and Pharmacodynamics following high-dose erlotinib treatment in brain cancer patients", CANCER RES., vol. 74, 1 October 2014 (2014-10-01), XP055423209 *
YU ET AL.: "A phase I study of twice weekly pulse dose and daily low dose erlotinib as initial treatment for patients (pts) with EGFR-mutant lung cancers", JOURNAL OF CLINICAL ONCOLOGY, vol. 33, no. 15, 29 May 2015 (2015-05-29), XP055423203 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022251539A3 (en) * 2021-05-26 2023-01-05 C4 Therapeutics, Inc. Egfr degraders to treat cancer metastasis to the brain or cns

Also Published As

Publication number Publication date
WO2017164887A8 (en) 2017-11-09

Similar Documents

Publication Publication Date Title
Erlichman et al. Phase I study of EKB-569, an irreversible inhibitor of the epidermal growth factor receptor, in patients with advanced solid tumors
US20230103366A1 (en) Combination therapies and methods of use thereof for treating cancer
TWI798199B (en) Cancer treatment
US20120308562A1 (en) Methods of treating mesothelioma with a pi3k inhibitor compound
CN112057452A (en) Quinoline derivatives for treating non-small cell lung cancer
WO2011092338A1 (en) Combined treatment of pancreatic cancer with gemcitabine and masitinib
WO2021183318A2 (en) Methods and compositions relating to improved combination therapies
JP2019112416A (en) Methods for the treatment of cancer
US11833149B2 (en) Pulse dosing regimen and methods of treatment
JP2023022190A (en) cancer treatment
WO2013086002A1 (en) Compositions, process of preparation of said compositions and method of treating cancer
BR112021010110A2 (en) Cancer Treatment Methods Resistant to cdk4/6 Inhibitors
JP2021523190A (en) A composition comprising a bisfluoroalkyl-1,4-benzodiazepinone compound for treating adenoid cystic carcinoma
JP6973456B2 (en) Side effect reducing agent during cancer chemotherapy
TW202045173A (en) Cancer treatment
US20220117963A1 (en) Elacestrant in combination with abemaciclib in women with breast cancer
WO2017164887A1 (en) Pulse dosing regimen and methods for treatment
JP2022516252A (en) Pharmaceutical combination for treating cancer
US20230233567A1 (en) Belvarafenib for use in cancer treatment
US20130150386A1 (en) Methods for the treatment of lung cancer
JP2023509191A (en) Combination therapy to treat cancer
WO2024023766A1 (en) P13k inhibitor combination therapy
WO2023158715A1 (en) Oxphos inhibitors for use in treating cancer
CN112533600A (en) Quinoline derivatives for the treatment of small cell lung cancer

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 16895690

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 16895690

Country of ref document: EP

Kind code of ref document: A1