WO2020068979A1 - Low-intensity treatment of hematological disorders - Google Patents

Low-intensity treatment of hematological disorders Download PDF

Info

Publication number
WO2020068979A1
WO2020068979A1 PCT/US2019/052952 US2019052952W WO2020068979A1 WO 2020068979 A1 WO2020068979 A1 WO 2020068979A1 US 2019052952 W US2019052952 W US 2019052952W WO 2020068979 A1 WO2020068979 A1 WO 2020068979A1
Authority
WO
WIPO (PCT)
Prior art keywords
cpx
days
composition
cytarabine
treatment
Prior art date
Application number
PCT/US2019/052952
Other languages
French (fr)
Inventor
Ronald CHEUNG
Stefan FADERL
Qi Wang
Original Assignee
Celator Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Celator Pharmaceuticals, Inc. filed Critical Celator Pharmaceuticals, Inc.
Priority to EP19864378.5A priority Critical patent/EP3856198A4/en
Priority to MX2021003527A priority patent/MX2021003527A/en
Priority to JP2021540394A priority patent/JP2022502498A/en
Priority to AU2019350759A priority patent/AU2019350759A1/en
Priority to KR1020217011132A priority patent/KR20210065962A/en
Priority to CN201980063289.2A priority patent/CN113164502A/en
Priority to BR112021005539-8A priority patent/BR112021005539A2/en
Priority to CA3114002A priority patent/CA3114002A1/en
Publication of WO2020068979A1 publication Critical patent/WO2020068979A1/en
Priority to IL281729A priority patent/IL281729A/en
Priority to US17/338,561 priority patent/US20210393665A1/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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/63Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide
    • A61K31/635Compounds containing para-N-benzenesulfonyl-N-groups, e.g. sulfanilamide, p-nitrobenzenesulfonyl hydrazide having a heterocyclic ring, e.g. sulfadiazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the invention relates to compositions and methods for treating patients who are ineligible for treatment with conventional chemotherapy. More particularly, the invention concerns the treatment of hematological conditions that are susceptible to treatment with combinations of cytidine analogues and anthracyclines.
  • Combination chemotherapies comprising cytidine analogues and anthracycline agents have been well studied for treatment against various cancers or hematologic proliferative disorders.
  • Drug cocktails of the cytidine analogue, cytarabine, and an anthracycline such as daunorubicin demonstrate some efficacy in patients with hematologic malignancies. See, e.g. , Tallum et al., Blood 106:2243 (2005).
  • Cytarabine cytosine arabinoside, Ara-C or 1 -b-D-arabinofuranosylcytosinc
  • cytarabine-5’ -triphosphate ara-CTP
  • the mechanism of action is not completely understood, but it appears that ara-CTP acts primarily through inhibition of DNA polymerase. Incorporation into DNA and RNA may also contribute to cytarabine cytotoxicity. Cytarabine is cytotoxic to a wide variety of proliferating mammalian cells in culture.
  • Daunorubicin hydrochloride is the hydrochloride salt of an anthracycline cytotoxic antibiotic produced by a strain of Streptomyces coeruleorubidus .
  • Daunorubicin has antimitotic and cytotoxic activity through a number of proposed mechanisms of action.
  • Daunorubicin forms complexes with DNA by intercalation between base pairs. It inhibits topoisomerase II activity by stabilizing the DNA-topoisomerase II complex, preventing the religation portion of the ligation-religation reaction that topoisomerase II catalyzes. Single strand and double strand DNA breaks result.
  • Daunorubicin may also inhibit polymerase activity, affect regulation of gene expression, and produce free radical damage to DNA. Daunorubicin possesses an antitumor effect against a wide spectrum of animal tumors, either grafted or spontaneous.
  • cytarabine combined with an anthracy cline has been the standard first-line therapy for acute myelogenous leukemia (AML), against which other regimens are compared.
  • AML acute myelogenous leukemia
  • the standard of care for AML has been a combination of cytarabine and daunorubicin administered in the classic“7 + 3” regimen with cytarabine administered for 7 consecutive days and daunorubicin for the first 3 of those 7 consecutive days.
  • Vyxeos® also known as“CPX-351” and used interchangeably here
  • CPX-351 a fixed-dose liposomal combination of cytarabine and daunorubicin at a 5: 1 ratio which delivers the administered ratio to the patient over time was approved by the FDA for the treatment of adults with two types of high-risk acute myeloid leukemia (AML): newly diagnosed therapy-related AML (t-AML) or AML with myelodysplasia-related changes (AML-MRC).
  • AML high-risk acute myeloid leukemia
  • t-AML newly diagnosed therapy-related AML
  • AML-MRC myelodysplasia-related changes
  • CPX-351 showed a significant improvement in overall survival in these patients compared to the standard 7+3 therapy.
  • results from earlier clinical studies also showed favorable results for treatment of other leukemias or hematological disorders such as myelodysplastic syndromes (MDS) and certain lymphoblastic leukemias
  • HMAs hypomethylating agents
  • LDAC low dose cytarabine
  • LIT low-intensity therapy
  • the present invention overcomes deficiencies in the prior art by providing a low-intensity therapy (LIT) of CPX-351 that is suitable for administration to patients who are ineligible for standard intensive chemotherapy (ISICT patients).
  • LIT low-intensity therapy
  • CPX-351 LIT provides a myelosuppressive profile that allows it to be combined with other agents shown effective in treating leukemias or other hematological proliferative disorders, such as myelodysplastic syndrome (MDS).
  • MDS myelodysplastic syndrome
  • a method to treat cancer or a hematologic proliferative disorder in an ISICT subject comprising administering to said subject a pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin, i.e. , CPX-351, wherein the cytarabine and daunorubicin are stably associated with one or more delivery vehicles. Encapsulation in these delivery vehicles allows two or more agents to be delivered to the disease site in a coordinated fashion, thereby assuring that the agents will be present at the disease site at a non-antagonistic ratio.
  • the agents are co-encapsulated in delivery vehicles, or are separately encapsulated in delivery vehicles administered such that non-antagonistic ratios are maintained at the disease site.
  • the pharmacokinetics (PK) of the composition are controlled by the delivery vehicles themselves such that coordinated delivery is achieved (provided that the PK of the delivery systems are comparable).
  • the pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is CPX-351, described in U.S. Patent Nos.
  • said pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is administered once or twice a week. In one embodiment, said pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is administered continuously for up to 12 months, or up to 24 months.
  • said pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is administered at a cytarabine dose of less than 32 mg/m 2 /day or less than 24 mg/m 2 /day.
  • a method to treat a hematologic proliferative disorder in an ISICT subject comprising administering to said patient a low-intensity CPX-351 treatment regimen.
  • a low-intensity treatment of CPX-351 comprises administering CPX-351 intravenously in 30 minutes to 3 hours.
  • CPX-351 is administered intravenously in 90 minutes or less.
  • CPX- 351 is administered intravenously in about 90 minutes.
  • CPX-351 is administered once or twice a week.
  • CPX-351 is administered at a cytarabine dose of less than 32 mg/m 2 /day or less than 24 mg/m 2 /day.
  • CPX- 351 is administered at a cytarabine dose of less than 24 mg/m 2 /day.
  • One embodiment of the present invention is a method to treat a hematologic proliferative disorder in a subject ineligible for standard intensive chemotherapy, wherein the cancer or hematologic proliferative disorder is an advanced hematologic cancer.
  • the advanced hematologic cancer is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML) or acute promyelocytic leukemia (APL).
  • the cancer or hematologic proliferative disorder is a hematologic proliferative disorder.
  • the hematologic proliferative disorder is myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN).
  • the subject ineligible for standard intensive chemotherapy has never undergone a previous anti-cancer regimen. In some embodiments, the subject ineligible for standard intensive chemotherapy has not previously received intensive chemotherapy. In some embodiments, the subject ineligible for standard intensive chemotherapy has previously undergone at least one anti-cancer regimen.
  • a method to treat a hematologic proliferative disorder in an ISICT subject comprising administering to said ISICT patient a low-dose CPX-351 treatment, wherein said composition is administered prior to said patient receiving any chemotherapy.
  • treatment of said subjects ineligible for standard intensive chemotherapy is administered on an outpatient basis or as a home-based treatment.
  • a method to reduce the toxicity of cytarabine/anthracycline therapy administered to a patient ineligible for standard intensive chemotherapy by administering to said patient a low-intensity CPX-351 treatment is measured as a reduction in non-hematopoietic toxicities such as, for example, mucositis or alopecia.
  • a reduction in such toxicities can in turn lead to a reduction in hospitalization, supportive care, morbidity and/or a reduction in induction mortality particularly in patients over 60 years of age and more specifically in patients over the age of 75.
  • a method to treat a post-remission hematologic proliferative disorder in an ISICT subject comprising administering to said patient a low-dose CPX-351 treatment, wherein said composition is administered less than 18 months, or less than 12 months, or less than 6 months after one or more initial treatments.
  • said one or more initial treatments comprises a hypomethylating agent or low-dose cytosine arabinoside.
  • a method to treat hematologic proliferative disorder in an ISICT subject comprising administering to said patient a low- intensity CPX-351 treatment, wherein said treatment provides a therapeutic effect which is greater than that achieved with the current standard of care for said hematologic proliferative disorder in an ISICT patient.
  • the therapeutic effect is measured as an increase in complete remission rate.
  • the therapeutic effect is measured as prolongation of complete remission duration and/or prolongation of time to progression and/or prolongation of survival.
  • a method to improve the safety of cytarabine/anthracycline treatments in an ISICT patient population comprising administering CPX-351 less than 3 times in one week and wherein the CPX-351 dose is less than 24 mg/m 2 /day.
  • the improved safety is measured as a reduction in non-hematopoietic toxicities.
  • the non-hematopoietic toxicities are mucositis and/or alopecia.
  • the method can include further administering a hypomethylating agent, or other agent shown effective in treating leukemias.
  • a method of the invention further comprises administering an agent approved for use in a hematological malignancy or disorder.
  • the agent approved for use in a hematological malignancy or disorder can be selected from, for example but not limited to, mylotarg, midostaurin, venetoclax or idhifa.
  • a method of treating a hematologic proliferative disorder in a subject ineligible for intensive chemotherapy comprising administering to said patient a low-intensity CPX-351 treatment wherein CPX-351 is administered intravenously at a cytarabine dose of less than 32mg/m 2 /day in less than 3 hours and wherein CPX-351 is administered no more than two times per week.
  • treatment is administered as a first line therapy.
  • a first line therapy method of treating a hematologic proliferative disorder in a subject ineligible for intensive chemotherapy comprising administering to said patient a low-intensity CPX- 351 treatment wherein CPX-351 is administered intravenously at a cytarabine dose of less than 32 mg/m 2 /day in less than 3 hours and wherein CPX-351 is administered no more than two times per week.
  • FIGURE 1A is schematic of a model used for PK/PD myelosuppression.
  • FIGURES 1B and 1C show a summary of Population PK and PK-PD Models.
  • FIGURE 1D shows a schematic representation of the modeling strategy
  • FIGURES 2A and 2B show the Visual Predictive Check Stratified by
  • FIGURE 3 A shows Platelet Count vs .Time During CPX-351 Treatment
  • FIGURES 3B and 3C shows the GOF (goodness-of-fit) Plots: Final CPX-351
  • FIGURE 4 shows the VPC (Visual Predictive Check): Final CPX-351 PK-PD
  • FIGURE 5 shows the ANC vs. Time During CPX-351 Treatment.
  • FIGURES 6A and 6B show the GOF (goodness-of-fit) Plots: Base CPX-351
  • FIGURE 7 shows the VPC (Visual Predictive Checks): Final CPX-351 PK-
  • FIGURE 8 shows the duration of ANC for a given dose range of CPX-351
  • FIGURES 9A and 9B show the duration of platelet count for a given dose range of CPX-351
  • FIGURES 10A and 10B show the first time of different platelet count using different dosing regimen.
  • FIGURE 11 show the first time of different ANC using different dosing regimen.
  • FIGURES 12 and 13 show the change in IC50 for the CTG-2226 and CTG-
  • PK pharmacokinetic
  • PD pharmacodynamic
  • CMT compartment
  • Ap peripheral amount of drug
  • Vp peripheral volume of distribution
  • CLd distribution clearance
  • ko 0-order rate constant
  • Ac central amount of drug
  • Vc central volume of distribution
  • CL clearance
  • GCSF granulocyte colony-stimulating factor
  • SC subcutaneous
  • F bioavailability
  • k a first-order absorption rate constant
  • D dose
  • CL clearance
  • Ki m first-order rate constant for internalization of GCSF receptor complexed with filgrastim or pegfilgrastim
  • ko dissociation constant
  • IV intravenous
  • ANC absolute neutrophil count.
  • weekly or monthly schedule such as an Induction treatment/therapy or Consolidation treatment/therapy
  • Specifically“low dose CPX-351 treatment” may be a lower dose and/or weekly or 28-day dosing schedule that has less dosages than that used in Study 301.
  • Vyxeos® Jazz Pharmaceuticals
  • cytarabine:daunorubicin liposome injection was based on 1) defining a non-antagonistic ratio of the two active moieties, cytarabine and daunorubicin, using cell-based screening assays and 2) designing a liposomal drug carrier to maintain this ratio after intravenous administration. This ratio was not based on the empirically-derived regimens currently used for cytarabine and anthracyclines.
  • the present invention overcomes deficiencies in the prior art by providing a low-intensity therapy (LIT) of CPX-351 that is suitable for administration to patients who are ineligible for standard intensive chemotherapy (ICT).
  • LIT low-intensity therapy
  • ICT standard intensive chemotherapy
  • A“treatment cycle” is a specific protocol that specifies dosage times and amounts for CPX-351 and, if appropriate, an additional agent.
  • a treatment cycle may employ 2-3 administrations of CPX-351 on certain days with specified dosages, for example, on days 1 and 3, or 1 and 5, or 1 and 8, or 1 and 15, or on days 1, 2 and 5, or 1,5, and 8, or 1, 5 and 15 or 1, 8 and 15.
  • the specific days may vary.
  • ISICT patients are critical populations to demonstrate benefit given the high unmet medical need. Population sizes are significant and yet the treatments available today still do not provide adequate improvements in overall survival. CPX-351 represents a promising new tool for improving efficacy while maintaining safety for these patients.
  • ICT intracranial pressure
  • factors for example, physical performance, comorbid conditions and cognitive function (Pettit and Odenike; Front. Oncol. (2015) 5:280; which is incorporated herein by reference).
  • the therapeutic aim for ISICT patients is to reduce disease burden, extend patient life, and improve their quality of life.
  • the CPX-351 LIT provides a myelosuppressive profile that allows it to be safely combined with other agents effective in treating these otherwise ineligible patients or effective in treating leukemias. These may, for example, be hypomethylation agents or BCL-2 inhibitors.
  • AML a myelosuppressive profile that allows it to be safely combined with other agents effective in treating these otherwise ineligible patients or effective in treating leukemias. These may, for example, be hypomethylation agents or BCL-2 inhibitors.
  • Acute Myeloid Leukemia is a clonal expansion of undifferentiated myeloid precursors that causes an impairment of new blood cell generation.
  • the median age of diagnosis 67 years.
  • Clinical manifestations include fever, fatigue, bone pain, pallor, easy bruising.
  • Diagnosis is based on having >20% myeloid blasts in the bone marrow.
  • Disease characteristics including cytogenetic and molecular genetic findings are major prognostic factors.
  • Patient characteristics (age, co-morbidities) also play a role in prognosis, since they influence the ability to undergo treatment.
  • optimal disease treatment for fit patients is based on intensive chemotherapy, divided into induction and consolidation phases. Differential diagnosis includes other hematologic malignancies and MDS.
  • Myelodysplastic syndromes are a family of rare disorders in which the bone marrow fails to make enough healthy red blood cells, white blood cells or platelets. This is caused by bone marrow producing lots of underdeveloped, or immature, cells that have an abnormal shape, size or look. These are called blast cells. Most experts agree that MDS is a form of blood and bone marrow cancer and can be difficult to diagnose.
  • Chemotherapy-induced thrombocytopenia is a common and serious complication that is associated with an increased risk of bleeding and is often accompanied by chemotherapy dose adjustments, which may compromise treatment outcomes. Platelet transfusions are a mainstay for treatment of chemotherapy-induced thrombocytopenia.
  • Chemotherapy-induced neutropenia is a serious adverse event that is associated with an increased risk of life-threatening infection and often leads to chemotherapy dose reductions and/or treatment delays, which may lead to poor treatment outcomes.
  • Endogenous granulocyte colony-stimulating factor (GCSF) is the primary regulator of neutrophil production, and recombinant GCSF agents are commonly used to treat CIN.
  • Standard dose CPX-351 as currently approved does not cover patients with AML considered ineligible for intensive chemotherapy (ICT), or some patients with MDS.
  • the current approach to treating patients ineligible for ICT consists of lower intensity treatments (e.g. Low dose cytarabine (LDAC) or HMA alone or in various combinations with novel agents.
  • LDAC Low dose cytarabine
  • HMA HMA alone or in various combinations with novel agents.
  • Primary intent is to minimize toxicity while achieving acceptable response rate translating into improvement of Quality-of-Life, reduction of transfusion requirements and possibly extension of survival.
  • CPX-351 is biologically active in high-risk patient populations, including secondary AML with prior history of MDS and those patients with MDS related karyotype (see phase 3 study referenced herein). CPX-351 improved survival compared to the standard 7+3 therapy in patients with AML who progressed from MDS and who were previously untreated with HMA.
  • CPX-351 showed superior median overall survival of 12.5 months compared with 5.95 months with 7+3 including patients with prior HMA exposure. Thus there is therefore reason to believe that lower intensity CPX-351 could be a more effective backbone combination partner when compared to LDAC or HMA.
  • PK-PD modeling is used to determine a starting dose and/or dosing regimen for CPX-351 in patients considered ineligible for ICT.
  • previously developed population PK models (such as those described in Qi W, et al. Blood. 2017; 130, Abstract 5064) are used to generate patient-specific cytarabine and daunorubicin concentration-time profiles in patients with AML, acute lymphocytic leukemia (ALL), or myelodysplastic syndrome (MDS) following CPX-351 administration.
  • ALL acute lymphocytic leukemia
  • MDS myelodysplastic syndrome
  • a population PK-PD model of chemotherapy- induced thrombocytopenia following CPX-351 administration that incorporates the effect of concurrent platelet transfusions.
  • patient covariates are used to describe variability in chemotherapy-induced thrombocytopenia.
  • a population PK-PD model of chemotherapy- induced neutropenia (CIN) following CPX-351 administration that incorporates the effect of concurrent GCSF therapy.
  • patient covariates are used to describe variability in CIN.
  • an R Shiny application utilizing the population PK and PK-PD models for simulation of platelet dynamics following CPX-351 or 7+3 administration with or without intermittent platelet transfusions.
  • an R Shiny application utilizing the population PK and PK-PD models for simulation of neutrophil dynamics following CPX-351 administration with or without GCSF in various dosing regimens.
  • PK-PD Pharmacokinetic-Pharmacodynamic
  • PK Data Collection Intensive sampling on Days 1 and 5 • PD Data Collection: Pre-dose; on Days 1, 3, 5, 7, 10 ⁇ 2, and 14 ⁇ 2; and then weekly ( ⁇ 2 days) until whichever occurred last: (1) Day 42, (2) peripheral blood count recovery, or (3) removal from study
  • Table 1 shows the population PK parameters for Cytarabine and Daunorubicin following CPX-351 administration.
  • PK pharmacokinetic
  • CL clearance
  • BSA body surface area
  • CV coefficient of variation
  • Vc central volume of distribution
  • DOSEMGC cytarabine dose in mg
  • CLd distribution clearance
  • Vp peripheral volume of distribution
  • DOSEMGD daunombicin dose in mg.
  • Population PK-PD analysis was conducted using NONMEM version 7.3 via implementation of the first-order conditional estimation method with h-e interaction.
  • Previously developed population PK models for CPX-351 and non-liposomal cytarabine and daunorubicin were used to generate patient-specific cytarabine and daunorubicin concentration-time profiles (Qi W, et al. Blood. 20l7;l30. Abstract 5064 incorporated herein by reference).
  • 2-compartment disposition models were used to describe drug PK following CPX-351 administration.
  • Platelet count versus time data were described by a modified maturation PK-PD model proposed by Friberg et al (Friberg LE, et al. J Clin Oncol. 2002;20(24):47l3-472l ; incorporated herein by reference). Inhibition of platelet proliferation by CPX-351 and/or “7+3” was driven by a sigmoidal maximum inhibition (I m ax) function of the sum of the molar concentrations of cytarabine and daunorubicin. Interindividual and interoccasion variability were estimated for select structural PK-PD model parameters using exponential error models.
  • Baseline Patient Demographics were: Weight, height, age, body mass index, body surface area, sex, race, and ethnicity.
  • Baseline Clinical Laboratory Measures were: Albumin, alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, total bilirubin, white blood cell count, absolute neutrophil count, platelet count, and creatinine clearance.
  • Disease- related Indices were: Cancer type and Eastern Cooperative Oncology Group performance status.
  • PK pharmacokinetic
  • PD pharmacodynamic
  • SD standard deviation
  • BSA body surface area
  • BMI body mass index
  • CLcr creatinine clearance
  • ALT alanine transaminase
  • AST aspartate transaminase
  • ALP alkaline phosphatase
  • WBC white blood cell count
  • ANC absolute neutrophil count.
  • GCSF granulocyte colony stimulating factor
  • ECOG Eastern Cooperative Oncology Group
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndrome
  • CR complete remission
  • CRi complete remission with incomplete neutrophil or platelet recovery.
  • NB for patients who were thrombocytopenic (platelet count ⁇ 50 x 10 9 /L) prior to the first treatment cycle, data from Cycle 1 were excluded from analysis, and data from subsequent cycles were only included if the platelet count returned to >50 x 10 9 /L prior to treatment. However, for the purposes of covariate evaluation, the baseline platelet count from Cycle 1 was used, even if the value was ⁇ 50 x 10 9 /L.
  • the early analysis dataset consisted of 2,023 platelet counts from 137 patients.
  • the mean (standard deviation) number of platelet transfusions per person was 12 (9).
  • the model fit improved significantly (P ⁇ 0.00001) when bolus inputs to circulating platelets were incorporated to account for platelet transfusions. No potential covariates met criteria for inclusion.
  • PD pharmacodynamic
  • SEM standard error of the mean
  • Circo baseline circulating platelet count
  • MTT mean transit time (4/k tr )
  • I max maximum inhibition of platelet proliferation
  • IC composite concentration (cytarabine + daunorubicin) at which inhibition is 50% of I max
  • y Hill coefficient for sigmoidal I max function
  • gt feedback function exponent.
  • the median time to observe the first platelet count ⁇ 0.5 x 109/L was 6.4 days after CPX-351 treatment and 5.8 days after 7+3, while the median time to an observed platelet count ⁇ 20 x 109/L was 10.8 days and 8.9 days, respectively.
  • the median duration with platelet counts ⁇ 20 x 109/L was longer with CPX-351 (18 days) versus 7+3 (8 days), and the median duration of platelet counts platelet count ⁇ 50 x 109/L was 22 days and 15 days, respectively.
  • Figure 2A shows Visual Predictive Check Stratified by Treatment Cycle for the
  • a maturation PK-PD model was developed to characterize the effect of CPX-351 and 7+3 on platelets in patients with AML, ALL, or MDS.
  • CPX-351 achieved a rapid and substantial suppression of platelet counts, with an estimated IC50 of 0.324 mM.
  • the model accounted for the confounding nature of platelet transfusions on platelet dynamics during chemotherapy-induced thrombocytopenia.
  • the final model was successfully embedded in an R Shiny application that can be utilized to evaluate the temporal events of thrombocytopenia following administration of various CPX-351 or 7+3 dosing regimens with intermittent platelet transfusions. These are used to design an appropriate treatment cycle for LiT administration of CPX-351.
  • PK pharmacokinetic
  • GCSF granulocyte colony-stimulating factor
  • Fi bioavailability of first-order absorption process
  • k a first-order absorption rate constant
  • F 2 bioavailability of zero-order absorption process
  • D 2 bioavailability of zero-order absorption process
  • D 2 bioavailability of zero-order absorption process
  • K D equilibrium dissociation constant for GCSF and GCSF receptor
  • CV coefficient of variation
  • CL clearance
  • WTKG body weight in kg
  • V c central volume of distribution
  • ki remember t first-order rate constant for internalization of GCSF receptor complexed with filgrastim or pegfilgrastim.
  • Population PK-PD analysis was conducted using NONMEM version 7.3 via implementation of the first-order conditional estimation method with h-e interaction.
  • Previously developed population PK models for CPX-351 or 7+3 (2-compartment disposition) were used to generate patient-specific cytarabine and daunorubicin concentration-time profiles.
  • Previously reported population PK models for GCSF agents (1- compartment with target-mediated drug disposition) were used to predict patient-specific concentration-time profiles following exogenous GCSF administration based on individual dosing histories and population mean PK parameters.
  • Inhibition of neutrophil proliferation by CPX-351 was driven by an I max function of the sum of the molar concentrations of cytarabine and daunorubicin. Interindividual variability was estimated for select structural PK-PD model parameters using exponential error models. Previously reported population PK models for GCSF agents (1 compartment with target- mediated drug disposition) were used to predict patient-specific concentration-time profiles following exogenous GCSF administration (filgrastim and/or pegfilgrastim) based on individual dosing histories and population mean PK parameters.
  • Baseline Patient Demographics were: Weight, height, age, body mass index, body surface area, sex, race, and ethnicity.
  • Baseline Clinical Laboratory Measures were: Albumin, alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, total bilirubin, white blood cell count, absolute neutrophil count, platelet count, and creatinine clearance.
  • Disease -related Indices were: Cancer type and Eastern Cooperative Oncology Group performance status.
  • SD standard deviation
  • BSA body surface area
  • ANC absolute neutrophil count
  • GCSF granulocyte colony-stimulating factor
  • ECOG Eastern Cooperative Oncology Group
  • ALL acute lymphocytic leukemia
  • AML acute myeloid leukemia.
  • the early analysis dataset consisted of 1,797 ANC observations from 129 patients, of whom 43 (33.3%) received GCSF therapy at least once during the study. Incorporation of the stimulatory effect of exogenous GCSF on neutrophil proliferation and maturation significantly improved the model fit (P ⁇ 0.00001). During covariate evaluation, a strong inverse correlation was observed between mean transit time and baseline ANC ( P ⁇ 0.00001); no other potential covariates met the criteria for inclusion. In the late stages of model development, I m ax was estimated to be 1.05 and subsequently fixed at 1, which produced a minor increase of 4.5 units in the minimum value of objective function.
  • PK pharmacokinetic
  • PD pharmacodynamic
  • SEM standard error of the mean
  • Circo baseline ANC
  • MTT mean transit time (4/k tr )
  • ANC absolute neutrophil count
  • I max maximum inhibition of neutrophil proliferation
  • IC 50 composite concentration (cytarabine + daunorubicin) at which inhibition is 50% of I max
  • y T feedback function exponent
  • Stimi slope parameter for stimulation of neutrophil proliferation by the fraction of GCSF receptors bound to exogenous GCSF
  • Stini2 slope parameter for stimulation of neutrophil maturation by the fraction of GCSF receptors bound to exogenous GCSF
  • w 2 interindividual variability
  • CV coefficient of variation
  • s 2 residual variability
  • SD standard deviation.
  • Figure 2B shows a visual predictive check stratified by treatment cycle for the final population PK-PD model. It is based on 100 simulated datasets showed reasonably good agreement between the median and the lOth and 90th percentiles of the observed and simulated ANC over time since the start of the treatment cycle.
  • the R Shiny application was used to simulate 200 patients undergoing chemotherapy with 100 units/m2 of CPX-351 via 90-minute IV infusion at 0, 48, and 96 hours ( Figure 1C). Without GCSF therapy, 172 (86%) patients recovered to ANC >1.0 x 109/L within the 96-day simulation period, and the median time from first CPX-351 dose to recovery of ANC >1.0 x 109/L was 788 hours (32.8 days).
  • a maturation PK-PD model was developed to characterize the effect of CPX-351 and 7+3 on ANC in patients with AML or ALL or MDS.
  • the model accounted for the confounding nature of concurrent GCSF therapy on neutrophil dynamics during CIN.
  • the final model was successfully embedded in an R Shiny application that can be utilized to evaluate the temporal events of neutropenia following administration of CPX-351 or 7+3 and GCSF therapy in various dosing regimens.
  • the myelosuppressive effects of CPX-351 were different from 7+3 in terms of duration of myelosuppression and time to neutrophil count ⁇ 0.5 x 109/L.
  • the median time for initial detection of myelosuppression with CPX-351 was 1 to 2 days later than with 7+3. Additionally, the median duration of myelosuppressive effects was longer with CPX-351 than with 7+3.
  • the duration of ANC ⁇ 500/uL increases as dose increases. Depending on regimen, the duration can be different for a given dose.
  • the duration ANC suppression after Vyxeos standard Day 1, 3, & 5 regimen or 7+3 treatment is also shown in the figure, where Vyxeos showed a longer duration of ANC suppression than 7+3. This longer suppression is consistent with clinical observations.
  • FIG. 9 A and 9B show the period of platelet count below 50, 000 or 20,000 /uL after administration of CPX-351.
  • the duration of platelet suppression increases as dose increases. Depending on regimen, the duration can be different for a given dose.
  • the duration platelet suppression after Vyxeos standard Day 1, 3, & 5 regimen or 7+3 treatment is also shown in the figure, where Vyxeos showed a longer duration of ANC suppression than 7+3. This longer suppression is consistent with clinical observations.
  • Vyxeos Day 1, 3, 5 regimen produced most longest suppression period ( ⁇ 20,000/uL) than other regimens.
  • Simulation outcome for platelet count this data appears to be the first time to observe a platelet count below a certain threshold level that is not related to regimen. Rather, it is related to the drug properties and dose.
  • Vyxeos treatment results in a later observation than 7+3 treatment.
  • the median start-time of observing platelet count ⁇ 20,000/uL approaches 258 h (Day 10 - 11 post the first dose).
  • the median start-time of observing platelet count ⁇ 50, 000/uL approaches 154 h (Day 6 - 7 post the first dose).
  • Venetoclax various drug:drug combinations were tested in 15 Passage 1 (Pl) models of human Acute Myeloid Leukemia (AML) and assayed for cell viability.
  • the study type and duration used was an ex-vivo cell killing assay with primary leukapharesis-derived AML cells using a 96-well plate format, 90 wells, 6-day incubation, Cell Titer Glo viability endpoint with ICso.
  • AML Models used were CTG-2226, CTG-2227, CTG-2228, CTG-2231 to
  • Cytarabine (5000nM) concentration was used as a positive control; Media + vehicle were used as negative control. Plates were kept in 37°C/5% C02 incubator. Media was not changed during the 6-day incubation period with test agents. On day 6, cell viability was tested using Cell Titer Glo assay.
  • Cell Titer Glo Assay Plates were removed from incubator and equilibrated to room temperature up to 30 minutes. lOOul of Cell Titer Glo was added to wells and mixed for 2 minutes by keeping the plates on plate rocker. The plates were incubated at room temperature for 10 minutes to stabilize luminescent signal. Luminescence was recorded using Tecan plate reader.
  • the primary objectives are to determine the maximum tolerated dose (MTD) of the combination of CPX-351 and venetoclax when administered to subjects with newly diagnosed Acute Myeloid Leukemia (AML) who are ineligible for intensive chemotherapy (ICT). In addition, the safety of this combination is to be determined.
  • MTD maximum tolerated dose
  • Secondary objections are to perform initial assessments of efficacy, including complete remission (CR), complete remission with incomplete hematologic recovery (CRi), composite complete remission rate (CRc: CR + CRi), overall response rate (ORR; CR + CRi + partial response [PR]) and assessment of Minimal Residual Disease (MRD) status (negative / positive) in subjects with documented CR or Cri.
  • CR complete remission
  • CRi complete remission with incomplete hematologic recovery
  • CRc composite complete remission rate
  • ORR CR + CRi + partial response [PR]
  • MRD Minimal Residual Disease
  • the study is also to determine the pharmacokinetics (PK) of CPX-351 and venetoclax when given in combination.
  • the study will comprise 2 phases: a Dose Escalation Phase and an Expansion
  • CPX-351 in which all subjects will receive a combination of CPX-351 and venetoclax.
  • a 3 + 3 design will be employed and enroll up to 24 subjects with newly diagnosed AML who are ineligible for standard ICT using stepwise dose escalations of CPX-351 in combination with 400 mg venetoclax in each cohort to determine the MTD for the combination.
  • the Expansion Phase will commence with 20 additional subjects treated with the combination to determine an initial response rate based on both morphologic assessment and MRD.
  • Subjects will be followed for safety (1 month after End of Treatment) and every 2 months ( ⁇ 2 weeks) for survival (up to 1 year after the first administration of study treatment). Each subject will be evaluated for response at the end of each cycle. Those subjects with CR, CRi or PR will be offered up to a total of 4 cycles of CPX-351 and venetoclax, as deemed appropriate by the treating physician. Subjects with NR (no response) after 2 cycles will stop treatment. Subjects who have completed 4 cycles of treatment will be managed at the investigator’s discretion, as per institutional guidelines.
  • CPX-351 will be administered on Days 1 and 3, and venetoclax will be administered on Days 2 through 21.
  • venetoclax will be administered at the study site during the infusion of CPX-351 to permit PK assessment.
  • Blood collection for PK studies will be conducted during the first and second cycles of therapy for all dose cohorts in the Dose Escalation Phase. Blood collection for sparse PK sampling will also be performed in the Expansion Phase for subjects receiving their Day 3 and Day 4 care at the study site. During Cycle 1 Day 3 of the Expansion Phase, venetoclax will be administered at the study site during the infusion of CPX-351 to permit PK assessment.
  • Dose Escalation and Definition ofMTD The Dose Escalation Phase will employ a 3 + 3 design to determine the DLTs and MTD as follows. The first 3 subjects will be treated at Dose Level 1. If this is deemed safe (see specifics below), then dose escalations will proceed as follows:
  • Dose escalation algorithm If none of the first 3 evaluable subjects at a dose level experience a DLT, then the next 3 subjects will be treated at the next dose level (dose escalation).! ⁇ 1 out of the first 3 evaluable subjects experiences a DLT, then an additional 3 subjects will be treated at the same dose (for a total of 6 subjects at a dose level). If 1 out the 6 total evaluable subjects at a dose level experiences a DLT, then the next 3 subjects will be treated at the next dose level (dose escalation). If 2 or more of the first 3 evaluable subjects at a dose level experience a DLT, then no additional subjects will be treated at this or higher dose level. If 2 or more of the 6 total evaluable subjects experience a DLT, then no additional subjects will be treated at this or higher dose level.
  • MTD determination algorithm If > 2 out of 3 or > 2 out of 6 evaluable subjects experience a DLT at Dose Level 1 (starting dose), then the study will stop and no MTD will be determined. Otherwise, the MTD will be determined as the highest dose level at which either 0 out of 3 or 1 out of 6 evaluable subjects experiences a DLT. If this is Dose Level 4, then the maximum administered dose will be Dose Level 4, and for the purposes of this study, Dose Level 4 will be considered the MTD.
  • the DLT observation period will be Days 1 to 49 after starting treatment, with a minimum observation period of 28 days.
  • Subjects who are eligible to progress to a second cycle, or who have transitioned to an alternative therapy may have an abbreviated DLT observation period (ie, less than 49 days).
  • Subjects who do not complete a full cycle of therapy due to disease-related mortality or who withdraw from the study for reasons unrelated to drug effects, and have not experienced a DLT will not be evaluable for DLTs. These subjects will be replaced.
  • a regular teleconference, occurring approximately every 3 weeks, will be held among the jazz medical and clinical team and study investigators to review safety data (hereafter referred to as the Safety Assessment Committee [SAC]).
  • the SAC will also meet at the completion of each dosing cohort to determine whether 1) dose escalation will proceed with the next cohort; 2) the current dose level requires additional assessment; 3) the MTD has been reached; or 4) whether the study will be stopped.
  • a venetoclax dose ramp-up will occur on Days 2 through 4, followed by treatment at the target dose on Days 5 to 21.
  • venetoclax will be administered at the full target dose during Days 2 through 21.
  • CHF Congestive Heart Failure
  • LVEF left ventricular ejection fraction
  • Subject must have a white blood cell count ⁇ 25 x l0 9 /L. (Note: subjects who have undergone hydroxyurea administration or leukapheresis for therapeutic cytoreduction will be considered eligible).
  • Subject has favorable risk cytogenetics ((t8;2l), inv(l6), t(l6;l6) or t 15; 17) karyotype abnormalities) as categorized by the National Comprehensive Cancer Network (NCCN) Guidelines Version 2.2014 for AML • Subject had an antecedent myeloproliferative neoplasm (MPN) including myelofibrosis, essential thrombocytosis, polycythemia vera, or chronic myelogenous leukemia (CML) with or without BCR-ABL 1 translocation and AML with BRC-ABL 1 translocation.
  • MPN myeloproliferative neoplasm
  • CML chronic myelogenous leukemia
  • APL acute promyelocytic leukemia
  • HIV Human Immunodeficiency Virus
  • HBV hepatitis B virus
  • HCV hepatitis C virus
  • CPX-351 is provided as a sterile, preservative-free, purple, lyophilized cake in a single-use vial. Each vial of CPX-351 contains 44 mg daunorubicin and 100 mg cytarabine. After reconstitution (but before final dilution) each mL contains 2.2 mg daunorubicin and 5 mg cytarabine. CPX-35l is administered as an IV infusion over approximately 90 minutes. Venetoclax is available in 3 strengths and is self-administered as an oral dose once daily with a meal and water:
  • the 100 mg tablet is provided as an oblong, biconvex shaped, pale yellow film -coated tablet debossed with“V” on one side and“100” on the other side.
  • the 50 mg tablet is provided as an oblong, biconvex shaped, beige film-coated tablet debossed with“V” on one side and“50” on the other side.
  • the 10 mg tablet is provided as a round, biconvex shaped, pale yellow film -coated tablet debossed with“V” on one side and“10” on the other side.

Abstract

The invention relates to compositions and methods for treating patients with hematological proliferative disorders who are ineligible for treatment with standard intensive chemotherapy, using low intensity treatment with CPX-351, a liposomal composition of daunorubicin and cytarabine.

Description

LOW-INTENSITY TREATMENT OF HEMATOLOGICAL DISORDERS
Cross-Reference to Related Applications
[0001] This application claims priority of U.S. Provisional Application No. 62/736,393, filed 25 September 2018 and U.S. Provisional Application No. 62/772,372, filed 28 November 2018, the disclosures of which are herein incorporated by reference in its entirety.
Technical Field
[0002] The invention relates to compositions and methods for treating patients who are ineligible for treatment with conventional chemotherapy. More particularly, the invention concerns the treatment of hematological conditions that are susceptible to treatment with combinations of cytidine analogues and anthracyclines.
Background Art
[0003] Combination chemotherapies comprising cytidine analogues and anthracycline agents have been well studied for treatment against various cancers or hematologic proliferative disorders. Drug cocktails of the cytidine analogue, cytarabine, and an anthracycline such as daunorubicin demonstrate some efficacy in patients with hematologic malignancies. See, e.g. , Tallum et al., Blood 106:2243 (2005).
[0004] Cytarabine (cytosine arabinoside, Ara-C or 1 -b-D-arabinofuranosylcytosinc) is a cell cycle phase-specific antineoplastic agent, affecting cells predominantly during the S- phase of cell division. Intracellularly, cytarabine is converted into cytarabine-5’ -triphosphate (ara-CTP), which is the active metabolite. The mechanism of action is not completely understood, but it appears that ara-CTP acts primarily through inhibition of DNA polymerase. Incorporation into DNA and RNA may also contribute to cytarabine cytotoxicity. Cytarabine is cytotoxic to a wide variety of proliferating mammalian cells in culture.
[0005] Daunorubicin hydrochloride is the hydrochloride salt of an anthracycline cytotoxic antibiotic produced by a strain of Streptomyces coeruleorubidus . Daunorubicin has antimitotic and cytotoxic activity through a number of proposed mechanisms of action. Daunorubicin forms complexes with DNA by intercalation between base pairs. It inhibits topoisomerase II activity by stabilizing the DNA-topoisomerase II complex, preventing the religation portion of the ligation-religation reaction that topoisomerase II catalyzes. Single strand and double strand DNA breaks result. Daunorubicin may also inhibit polymerase activity, affect regulation of gene expression, and produce free radical damage to DNA. Daunorubicin possesses an antitumor effect against a wide spectrum of animal tumors, either grafted or spontaneous.
[0006] Since 1973, cytarabine combined with an anthracy cline has been the standard first-line therapy for acute myelogenous leukemia (AML), against which other regimens are compared. Until recently, the standard of care for AML has been a combination of cytarabine and daunorubicin administered in the classic“7 + 3” regimen with cytarabine administered for 7 consecutive days and daunorubicin for the first 3 of those 7 consecutive days. Addition of other agents such as 6-thioguanine or etoposide and changes in the dose or schedule of administration have been studied to improve outcomes, but while incremental gains have been made, the 40-year old use of an anthracycline and cytarabine remained the basis for standard induction treatment in AML.
[0007] In 2017, Vyxeos® (also known as“CPX-351” and used interchangeably here), a fixed-dose liposomal combination of cytarabine and daunorubicin at a 5: 1 ratio which delivers the administered ratio to the patient over time was approved by the FDA for the treatment of adults with two types of high-risk acute myeloid leukemia (AML): newly diagnosed therapy-related AML (t-AML) or AML with myelodysplasia-related changes (AML-MRC). CPX-351 showed a significant improvement in overall survival in these patients compared to the standard 7+3 therapy. Results from earlier clinical studies also showed favorable results for treatment of other leukemias or hematological disorders such as myelodysplastic syndromes (MDS) and certain lymphoblastic leukemias/lymphomas.
[0008] Unfortunately, due to age and/or co-morbidities, many patients with hematological malignancies are ineligible to receive standard intensive chemotherapy (ICT), i.e., ISICT patients or subjects, including approximately 50% of AML patients. The standard therapies for these ISICT patients are hypomethylating agents (HMAs) and low dose cytarabine (LDAC). However, the single agent activity of HMAs and LDAC is discouraging with response rates less than about 20% and a median overall survival of only 7-10 months.
[0009] Recognizing that there remains a need to achieve improved efficacy for ISICT patients, the present inventors have identified a low-intensity therapy (LIT) of CPX-351 for use in these patients who are ineligible for standard intensive chemotherapy. Furthermore, this LIT therapy provides a myelosuppressive profile that allows it to be combined with other less-intense agents suitable for these patient populations. Disclosure of the Invention
[0010] The present invention overcomes deficiencies in the prior art by providing a low-intensity therapy (LIT) of CPX-351 that is suitable for administration to patients who are ineligible for standard intensive chemotherapy (ISICT patients). In addition, the CPX-351 LIT provides a myelosuppressive profile that allows it to be combined with other agents shown effective in treating leukemias or other hematological proliferative disorders, such as myelodysplastic syndrome (MDS).
[0011] Provided herein is a method to treat cancer or a hematologic proliferative disorder in an ISICT subject, said method comprising administering to said subject a pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin, i.e. , CPX-351, wherein the cytarabine and daunorubicin are stably associated with one or more delivery vehicles. Encapsulation in these delivery vehicles allows two or more agents to be delivered to the disease site in a coordinated fashion, thereby assuring that the agents will be present at the disease site at a non-antagonistic ratio. This result will be achieved whether the agents are co-encapsulated in delivery vehicles, or are separately encapsulated in delivery vehicles administered such that non-antagonistic ratios are maintained at the disease site. The pharmacokinetics (PK) of the composition are controlled by the delivery vehicles themselves such that coordinated delivery is achieved (provided that the PK of the delivery systems are comparable). In a specific embodiment, the pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is CPX-351, described in U.S. Patent Nos. 7,238,367; 7,744,921 ; 7,850,990; 8,022,279; 8,092,828; 8,431,806; 8,518,437; 9,271,931 ; 10,028,912; and 10,166,184; all of which are incorporated herein by reference in their entireties for any purpose.
[0012] In one embodiment, said pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is administered once or twice a week. In one embodiment, said pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is administered continuously for up to 12 months, or up to 24 months.
[0013] In one embodiment, said pharmaceutical composition comprising a fixed, 5: 1 molar ratio of cytarabine and daunorubicin is administered at a cytarabine dose of less than 32 mg/m2/day or less than 24 mg/m2/day. [0014] In certain embodiments, provided is a method to treat a hematologic proliferative disorder in an ISICT subject, said method comprising administering to said patient a low-intensity CPX-351 treatment regimen.
[0015] In one embodiment, a low-intensity treatment of CPX-351 comprises administering CPX-351 intravenously in 30 minutes to 3 hours. In specific embodiments, CPX-351 is administered intravenously in 90 minutes or less. In specific embodiments, CPX- 351 is administered intravenously in about 90 minutes. In certain embodiments, CPX-351 is administered once or twice a week. In certain other embodiments, CPX-351 is administered at a cytarabine dose of less than 32 mg/m2/day or less than 24 mg/m2/day. Preferably, CPX- 351 is administered at a cytarabine dose of less than 24 mg/m2/day.
[0016] One embodiment of the present invention is a method to treat a hematologic proliferative disorder in a subject ineligible for standard intensive chemotherapy, wherein the cancer or hematologic proliferative disorder is an advanced hematologic cancer. In some embodiments, the advanced hematologic cancer is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML) or acute promyelocytic leukemia (APL). In other embodiments, the cancer or hematologic proliferative disorder is a hematologic proliferative disorder. In specific embodiments the hematologic proliferative disorder is myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN).
[0017] In some embodiments, the subject ineligible for standard intensive chemotherapy has never undergone a previous anti-cancer regimen. In some embodiments, the subject ineligible for standard intensive chemotherapy has not previously received intensive chemotherapy. In some embodiments, the subject ineligible for standard intensive chemotherapy has previously undergone at least one anti-cancer regimen.
[0018] In one embodiment, provided is a method to treat a hematologic proliferative disorder in an ISICT subject, said method comprising administering to said ISICT patient a low-dose CPX-351 treatment, wherein said composition is administered prior to said patient receiving any chemotherapy.
[0019] In another embodiment, treatment of said subjects ineligible for standard intensive chemotherapy is administered on an outpatient basis or as a home-based treatment.
[0020] In another embodiment, provided is a method to reduce the toxicity of cytarabine/anthracycline therapy administered to a patient ineligible for standard intensive chemotherapy by administering to said patient a low-intensity CPX-351 treatment. In some embodiments, reduction in toxicity is measured as a reduction in non-hematopoietic toxicities such as, for example, mucositis or alopecia. A reduction in such toxicities can in turn lead to a reduction in hospitalization, supportive care, morbidity and/or a reduction in induction mortality particularly in patients over 60 years of age and more specifically in patients over the age of 75.
[0021] In another embodiment, provided is a method to treat a post-remission hematologic proliferative disorder in an ISICT subject, said method comprising administering to said patient a low-dose CPX-351 treatment, wherein said composition is administered less than 18 months, or less than 12 months, or less than 6 months after one or more initial treatments. In certain embodiments, said one or more initial treatments comprises a hypomethylating agent or low-dose cytosine arabinoside.
[0022] In another embodiment, provided is a method to treat hematologic proliferative disorder in an ISICT subject, said method comprising administering to said patient a low- intensity CPX-351 treatment, wherein said treatment provides a therapeutic effect which is greater than that achieved with the current standard of care for said hematologic proliferative disorder in an ISICT patient. In some embodiments, the therapeutic effect is measured as an increase in complete remission rate. In other embodiments, the therapeutic effect is measured as prolongation of complete remission duration and/or prolongation of time to progression and/or prolongation of survival.
[0023] In another embodiment, provided is method to improve the safety of cytarabine/anthracycline treatments in an ISICT patient population, said method comprising administering CPX-351 less than 3 times in one week and wherein the CPX-351 dose is less than 24 mg/m2/day. In some embodiments, the improved safety is measured as a reduction in non-hematopoietic toxicities. In specific embodiments, the non-hematopoietic toxicities are mucositis and/or alopecia.
[0024] In each of the embodiments of the invention the method can include further administering a hypomethylating agent, or other agent shown effective in treating leukemias.
[0025] In specific embodiments, a method of the invention further comprises administering an agent approved for use in a hematological malignancy or disorder. The agent approved for use in a hematological malignancy or disorder can be selected from, for example but not limited to, mylotarg, midostaurin, venetoclax or idhifa.
[0026] In additional embodiments, provided herein is a method of treating a hematologic proliferative disorder in a subject ineligible for intensive chemotherapy, said method comprising administering to said patient a low-intensity CPX-351 treatment wherein CPX-351 is administered intravenously at a cytarabine dose of less than 32mg/m2/day in less than 3 hours and wherein CPX-351 is administered no more than two times per week. In a further embodiment, treatment is administered as a first line therapy.
[0027] In certain embodiments, provided herein is a first line therapy method of treating a hematologic proliferative disorder in a subject ineligible for intensive chemotherapy, said method comprising administering to said patient a low-intensity CPX- 351 treatment wherein CPX-351 is administered intravenously at a cytarabine dose of less than 32 mg/m2/day in less than 3 hours and wherein CPX-351 is administered no more than two times per week.
Brief Description of the Drawings
[0028] FIGURE 1A is schematic of a model used for PK/PD myelosuppression.
[0029] FIGURES 1B and 1C show a summary of Population PK and PK-PD Models.
[0030] FIGURE 1D shows a schematic representation of the modeling strategy
[0031] FIGURES 2A and 2B show the Visual Predictive Check Stratified by
Treatment Cycle for the Final Population PK-PD Model
[0032] FIGURE 3 A shows Platelet Count vs .Time During CPX-351 Treatment
[0033] FIGURES 3B and 3C shows the GOF (goodness-of-fit) Plots: Final CPX-351
PK-PD Model for Platelets
[0034] FIGURE 4 shows the VPC (Visual Predictive Check): Final CPX-351 PK-PD
Model for Platelets
[0035] FIGURE 5 shows the ANC vs. Time During CPX-351 Treatment.
[0036] FIGURES 6A and 6B show the GOF (goodness-of-fit) Plots: Base CPX-351
PK-PD Model for Neutrophils.
[0037] FIGURE 7 shows the VPC (Visual Predictive Checks): Final CPX-351 PK-
PD Model for Neutrophils
[0038] FIGURE 8 shows the duration of ANC for a given dose range of CPX-351
[0039] FIGURES 9A and 9B show the duration of platelet count for a given dose range of CPX-351
[0040] FIGURES 10A and 10B show the first time of different platelet count using different dosing regimen.
[0041] FIGURE 11 show the first time of different ANC using different dosing regimen.
[0042] FIGURES 12 and 13 show the change in IC50 for the CTG-2226 and CTG-
2233, cell lines respectively. Modes of Carrying Out the Invention
[0043] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which this invention belongs. All patents, applications, published applications and other publications referred to herein are incorporated by reference in their entirety. If a definition set forth in this section is contrary to or otherwise inconsistent with a definition set forth in the patents, applications, published applications and other publications that are herein incorporated by reference, the definition set forth in this section prevails over the definition that is incorporated herein by reference.
Definitions
[0044] As used herein,“a” or“an” means“at least one” or“one or more.”
[0045] Abbreviations used herein include: PK, pharmacokinetic; PD, pharmacodynamic; CMT, compartment; Ap, peripheral amount of drug; Vp, peripheral volume of distribution; CLd, distribution clearance; ko, 0-order rate constant; Ac, central amount of drug; Vc, central volume of distribution; CL, clearance; GCSF, granulocyte colony-stimulating factor; SC, subcutaneous; F, bioavailability; ka, first-order absorption rate constant; D, dose; CL, clearance; KD, equilibrium dissociation constant for GCSF and GCSF receptor R, accumulation ratio; Kim, first-order rate constant for internalization of GCSF receptor complexed with filgrastim or pegfilgrastim; ko, dissociation constant; IV, intravenous; ANC, absolute neutrophil count.
[0046] By‘‘low-intensity therapy”, “low intensity treatment, “LIT”, or“low dose treatment” or“low dose therapy”, it is meant to include both a lower individual dose and/or less dosages in the dosing schedule. (For example, weekly or monthly schedule, such as an Induction treatment/therapy or Consolidation treatment/therapy) which is lower and/or less intense than a similar therapy used in cancer patients who may qualify for ICT is also included. Specifically“low dose CPX-351 treatment” may be a lower dose and/or weekly or 28-day dosing schedule that has less dosages than that used in Study 301.
[0047] The development of Vyxeos® (Jazz Pharmaceuticals) (CPX-351), cytarabine:daunorubicin liposome injection was based on 1) defining a non-antagonistic ratio of the two active moieties, cytarabine and daunorubicin, using cell-based screening assays and 2) designing a liposomal drug carrier to maintain this ratio after intravenous administration. This ratio was not based on the empirically-derived regimens currently used for cytarabine and anthracyclines. [0048] The present invention overcomes deficiencies in the prior art by providing a low-intensity therapy (LIT) of CPX-351 that is suitable for administration to patients who are ineligible for standard intensive chemotherapy (ICT). ISICT patients are defined as either: -1) > 75 years of age, or
-2) > 18 to 74 years of age and fulfill at least 1 criteria associated with lack of fitness for intensive induction chemotherapy as follows:
-Eastern Cooperative Oncology Group (ECOG) Performance status of 2 to 3;
-Cardiac history of Congestive Heart Failure (CHF) requiring treatment of Ejection Fraction < 50% or chronic stable angina
-Diffusing Capacity of the Lung for Carbon Monoxide (DLCO) < 65% or Forced Expiratory Volume in 1 second (FEV1) < 65%;
-Creatinine clearance > 30 mL/min to < 45 mL/min;
-Moderate hepatic impairment with total bilirubin > 1.5 to < 3.0 x Upper Limit of Normal (ULN); or,
- Other comorbidity that the physician judges to be incompatible with conventional intensive chemotherapy.
[0049] A“treatment cycle” is a specific protocol that specifies dosage times and amounts for CPX-351 and, if appropriate, an additional agent. For example, a treatment cycle may employ 2-3 administrations of CPX-351 on certain days with specified dosages, for example, on days 1 and 3, or 1 and 5, or 1 and 8, or 1 and 15, or on days 1, 2 and 5, or 1,5, and 8, or 1, 5 and 15 or 1, 8 and 15. The specific days may vary.
[0050] ISICT patients are critical populations to demonstrate benefit given the high unmet medical need. Population sizes are significant and yet the treatments available today still do not provide adequate improvements in overall survival. CPX-351 represents a promising new tool for improving efficacy while maintaining safety for these patients.
[0051] A number of factors are used to determine eligibility for ICT including, for example, physical performance, comorbid conditions and cognitive function (Pettit and Odenike; Front. Oncol. (2015) 5:280; which is incorporated herein by reference). The therapeutic aim for ISICT patients is to reduce disease burden, extend patient life, and improve their quality of life.
[0052] In addition, the CPX-351 LIT provides a myelosuppressive profile that allows it to be safely combined with other agents effective in treating these otherwise ineligible patients or effective in treating leukemias. These may, for example, be hypomethylation agents or BCL-2 inhibitors. AML
[0053] Acute Myeloid Leukemia is a clonal expansion of undifferentiated myeloid precursors that causes an impairment of new blood cell generation. The median age of diagnosis 67 years. Clinical manifestations include fever, fatigue, bone pain, pallor, easy bruising. Diagnosis is based on having >20% myeloid blasts in the bone marrow. Disease characteristics including cytogenetic and molecular genetic findings are major prognostic factors. Patient characteristics (age, co-morbidities) also play a role in prognosis, since they influence the ability to undergo treatment. Currently, optimal disease treatment for fit patients is based on intensive chemotherapy, divided into induction and consolidation phases. Differential diagnosis includes other hematologic malignancies and MDS.
MDS
[0054] Myelodysplastic syndromes are a family of rare disorders in which the bone marrow fails to make enough healthy red blood cells, white blood cells or platelets. This is caused by bone marrow producing lots of underdeveloped, or immature, cells that have an abnormal shape, size or look. These are called blast cells. Most experts agree that MDS is a form of blood and bone marrow cancer and can be difficult to diagnose.
CPX-351 LIT Monotherapy or Combination Therapy
[0055] In a phase 3 trial in older adults (aged 60-75 years) with newly diagnosed high-risk/secondary AML, CPX-351 significantly prolonged overall survival versus standard 7+3 cytarabine/daunorubicin (9.56 vs 5.95 months; hazard ratio = 0.69; 1-sided P = 0.003), and the safety profile of CPX-351 was generally consistent with the known profile of the 7+3 regimen (Lancet JE, et al. J Clin Oncol. 2018;36(26):2684-2692; which is incorporated herein by reference).
[0056] Chemotherapy-induced thrombocytopenia is a common and serious complication that is associated with an increased risk of bleeding and is often accompanied by chemotherapy dose adjustments, which may compromise treatment outcomes. Platelet transfusions are a mainstay for treatment of chemotherapy-induced thrombocytopenia. Chemotherapy-induced neutropenia (CIN) is a serious adverse event that is associated with an increased risk of life-threatening infection and often leads to chemotherapy dose reductions and/or treatment delays, which may lead to poor treatment outcomes. Endogenous granulocyte colony-stimulating factor (GCSF) is the primary regulator of neutrophil production, and recombinant GCSF agents are commonly used to treat CIN.
[0057] Standard dose CPX-351 as currently approved does not cover patients with AML considered ineligible for intensive chemotherapy (ICT), or some patients with MDS. The current approach to treating patients ineligible for ICT consists of lower intensity treatments (e.g. Low dose cytarabine (LDAC) or HMA alone or in various combinations with novel agents. Primary intent is to minimize toxicity while achieving acceptable response rate translating into improvement of Quality-of-Life, reduction of transfusion requirements and possibly extension of survival.
[0058] Lower intensity combinations can achieve significant response rates in vulnerable patient populations. Whereas response rates with LDAC or HMAs single agents are modest, significantly higher and more durable responses can be seen in combinations as recently demonstrated with lower intensity therapy plus Venetoclax (DiNardo et al., Blood (2015) 126:327; incorporated herein by reference.)
[0059] CPX-351 is biologically active in high-risk patient populations, including secondary AML with prior history of MDS and those patients with MDS related karyotype (see phase 3 study referenced herein). CPX-351 improved survival compared to the standard 7+3 therapy in patients with AML who progressed from MDS and who were previously untreated with HMA.
[0060] Focusing on AML patients with 20-29% blasts (“oligoblastic AML”, in the past also referred to as“RAEBT”, a subtype of advanced MDS), CPX-351 showed superior median overall survival of 12.5 months compared with 5.95 months with 7+3 including patients with prior HMA exposure. Thus there is therefore reason to believe that lower intensity CPX-351 could be a more effective backbone combination partner when compared to LDAC or HMA.
[0061] When treating AML patients not eligible for standard ICT, subjects for use with low-intensity treatment of CPX-351 should have histological confirmation of AML by World Health Organization (WHO) criteria.
[0062] In one embodiment, PK-PD modeling is used to determine a starting dose and/or dosing regimen for CPX-351 in patients considered ineligible for ICT. In one embodiment, previously developed population PK models (such as those described in Qi W, et al. Blood. 2017; 130, Abstract 5064) are used to generate patient-specific cytarabine and daunorubicin concentration-time profiles in patients with AML, acute lymphocytic leukemia (ALL), or myelodysplastic syndrome (MDS) following CPX-351 administration.
[0063] In one embodiment, provided is a population PK-PD model of chemotherapy- induced thrombocytopenia following CPX-351 administration that incorporates the effect of concurrent platelet transfusions. In certain embodiments, patient covariates are used to describe variability in chemotherapy-induced thrombocytopenia. [0064] In one embodiment, provided is a population PK-PD model of chemotherapy- induced neutropenia (CIN) following CPX-351 administration that incorporates the effect of concurrent GCSF therapy. In certain embodiments, patient covariates are used to describe variability in CIN.
[0065] In one embodiment, provided is an R Shiny application utilizing the population PK and PK-PD models for simulation of platelet dynamics following CPX-351 or 7+3 administration with or without intermittent platelet transfusions. In another embodiment, provided is an R Shiny application utilizing the population PK and PK-PD models for simulation of neutrophil dynamics following CPX-351 administration with or without GCSF in various dosing regimens.
[0066] The following examples are offered to illustrate but not to limit the invention.
Example 1
Population Pharmacokinetic-Pharmacodynamic (PK-PD) Modeling of 7+3 or CPX-351- induced Thrombocytopenia
[0067] The following CPX-351 studies were used for PK-PD Modeling:
[0068] Study 101
• Phase 1, open-label, dose-escalation study of CPX-351 in 48 adults with relapsed/refractory AML, ALL, or MDS
• CPX-351 Dosages: 3 to 134 units/m2 (1 unit = 1 mg of cytarabine and 0.44 mg of daunorubicin) via 90-minute intravenous (IV) infusion on Days 1, 3, and 5
• PK Data Collection: Intensive sampling on Days 1, 3, and 5
• PD Data Collection: Pre-dose; on Days 1, 2, 3, 4, 5, 7, 14, 21, 28, 35, 42, and 56; and at follow-up (30 days after study discontinuation)
[0069] Study 206
• Phase 2, open-label study of CPX-351 in 26 adults with AML, ALL, or MDS
• CPX-351 dosages
o Induction: 100 units/m2 (100 mg/m2 cytarabine + 44 mg/m2 daunorubicin) via 90- minute IV infusion on Days 1, 3, and 5 (2nd induction: Days 1 and 3)
o Consolidation: 65 units/m2 (65 mg/m2 cytarabine + 29 mg/m2 daunorubicin) via 90- minute IV infusion on Days 1 and 3
• PK Data Collection: Intensive sampling on Days 1 and 5 • PD Data Collection: Pre-dose; on Days 1, 3, 5, 7, 10 ± 2, and 14 ± 2; and then weekly (±2 days) until whichever occurred last: (1) Day 42, (2) peripheral blood count recovery, or (3) removal from study
[0070] Study 301
• Phase 3, multicenter, randomized trial in older adults with newly diagnosed high- risk/secondary AML to assess CPX-351 (n = 153) versus 7+3 (n = 156)
• CPX-351 Dosages: Same as for Study 206
• PK Data Collection: Sparse sampling throughout first week
• PD Data Collection: Same as for Study 206
[0071] Table 1 shows the population PK parameters for Cytarabine and Daunorubicin following CPX-351 administration.
Table 1. Initial Population PK Parameters for Cytarabine and Daunorubicin Following CPX-351 Administration
Interindividual
Parameter Population mean equation variability
Figure imgf000014_0001
Residual 0.154 (log error)
variability
PK, pharmacokinetic; CL, clearance; BSA, body surface area; CV, coefficient of variation; Vc, central volume of distribution; DOSEMGC, cytarabine dose in mg; CLd, distribution clearance; Vp, peripheral volume of distribution; FORM2, CPX-351 formulation (1 for frozen, 0 for lyophilized); DOSEMGD, daunombicin dose in mg.
[0072] Population PK-PD analysis was conducted using NONMEM version 7.3 via implementation of the first-order conditional estimation method with h-e interaction. Previously developed population PK models for CPX-351 and non-liposomal cytarabine and daunorubicin were used to generate patient-specific cytarabine and daunorubicin concentration-time profiles (Qi W, et al. Blood. 20l7;l30. Abstract 5064 incorporated herein by reference). For both cytarabine and daunorubicin, 2-compartment disposition models were used to describe drug PK following CPX-351 administration. In the population PK-PD model of chemotherapy-induced thrombocytopenia, data from Cycle 1 were excluded if patients had a platelet count <50 x l09/L prior to the first treatment cycle; data from subsequent cycles were only included if the platelet count returned to >50 x l09/L prior to treatment.
[0073] Platelet count versus time data were described by a modified maturation PK-PD model proposed by Friberg et al (Friberg LE, et al. J Clin Oncol. 2002;20(24):47l3-472l ; incorporated herein by reference). Inhibition of platelet proliferation by CPX-351 and/or “7+3” was driven by a sigmoidal maximum inhibition (Imax) function of the sum of the molar concentrations of cytarabine and daunorubicin. Interindividual and interoccasion variability were estimated for select structural PK-PD model parameters using exponential error models.
[0074] The effect of each platelet transfusion on platelet dynamics was incorporated as a bolus input of 35 x l09/L into the circulating platelet pool, as this was the expected rise in platelet counts immediately post-transfusion (McCullough J. Semin Hematol. 20l0;47(3):235-242; incorporated herein by reference) (see Figure 1). Information regarding actual platelet dose administered was not standardized or always provided, so the typical value for platelet transfusion bioavailability was fixed at 1 and interindividual variability in platelet transfusion bioavailability was estimated to allow for interindividual variability in the expected increase in platelet count following a platelet transfusion.
[0075] A graphical screening procedure was conducted to examine the relationship between patient covariates and key PK-PD model parameters, followed by stepwise forward selection (a = 0.01) and backward elimination (a = 0.001) to evaluate covariate effects. Baseline Patient Demographics were: Weight, height, age, body mass index, body surface area, sex, race, and ethnicity. Baseline Clinical Laboratory Measures were: Albumin, alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, total bilirubin, white blood cell count, absolute neutrophil count, platelet count, and creatinine clearance. Disease- related Indices were: Cancer type and Eastern Cooperative Oncology Group performance status.
[0076] An R Shiny application was developed in conjunction with the mrgsolve package (Metrum Research Group) so that the population PK-PD model could be used to simulate neutrophil or platelet dynamics following administration of various CPX-351 and “7+3” dosing regimens (including as administered in Study 301) with or without intermittent platelet transfusions. Simulations were conducted in 200 patients with characteristics similar to the trial population to evaluate the temporal events of myelosuppression in the absence of platelet transfusion or GCSF administration (see below).
[0077] Relevant patient demographics were generated with distributions that were comparable to those in the analysis dataset. Results are shown in Table 2 below.
Table 2. Demographic Data for PK/PD Modeling
Figure imgf000016_0001
Figure imgf000017_0001
PK, pharmacokinetic; PD, pharmacodynamic; SD, standard deviation; BSA, body surface area; BMI, body mass index; CLcr, creatinine clearance; ALT, alanine transaminase; AST, aspartate transaminase; ALP, alkaline phosphatase; WBC, white blood cell count; ANC, absolute neutrophil count.
Table 2A. Demographic and Disease Status
Figure imgf000017_0002
Figure imgf000018_0001
GCSF, granulocyte colony stimulating factor; ECOG, Eastern Cooperative Oncology Group; ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; MDS, myelodysplastic syndrome; CR, complete remission; CRi, complete remission with incomplete neutrophil or platelet recovery. NB: for patients who were thrombocytopenic (platelet count <50 x 109/L) prior to the first treatment cycle, data from Cycle 1 were excluded from analysis, and data from subsequent cycles were only included if the platelet count returned to >50 x 109/L prior to treatment. However, for the purposes of covariate evaluation, the baseline platelet count from Cycle 1 was used, even if the value was <50 x 109/L.
[0078] The early analysis dataset consisted of 2,023 platelet counts from 137 patients.
Most patients (n = 135; 98.5%) received >1 platelet transfusion during the study.
The mean (standard deviation) number of platelet transfusions per person was 12 (9). The model fit improved significantly (P <0.00001) when bolus inputs to circulating platelets were incorporated to account for platelet transfusions. No potential covariates met criteria for inclusion.
Table 3. Model Fitted PD Parameters for Platelets
Figure imgf000018_0002
PD, pharmacodynamic; SEM, standard error of the mean; Circo, baseline circulating platelet count; MTT, mean transit time (4/ktr); Imax, maximum inhibition of platelet proliferation; IC , composite concentration (cytarabine + daunorubicin) at which inhibition is 50% of Imax; y, Hill coefficient for sigmoidal Imax function; gt, feedback function exponent.
[0079] The model successfully captured observed data for CPX-351 and 7+3, with good precision on parameter estimates. In fact, most parameters in the final PK-PD model were estimated with excellent precision (<35% standard error of the mean [SEM]).
[0080] No covariates affecting the PD parameters of CPX-351 were identified, but body weight was identified as a covariate affecting the IC50 of 7+3. The population mean for baseline circulating platelet numbers (CircO) was similar for CPX-351 and 7+3, while the population mean for mean transit time (MTT) was slightly shorter for CPX-351. 7+3 was more potent than CPX-351, albeit the plasma concentrations of cytarabine and daunorubicin with CPX-351 were far greater than with 7+3.
[0081] The median time to observe the first platelet count <0.5 x 109/L was 6.4 days after CPX-351 treatment and 5.8 days after 7+3, while the median time to an observed platelet count <20 x 109/L was 10.8 days and 8.9 days, respectively. The median duration with platelet counts <20 x 109/L was longer with CPX-351 (18 days) versus 7+3 (8 days), and the median duration of platelet counts platelet count <50 x 109/L was 22 days and 15 days, respectively. These results are summarized in Table 3 A below.
Table 3A. Model-simulated Platelet Parameters After CPX-351 or 7+3 Treatment
Figure imgf000019_0001
[0082] Figure 2A shows Visual Predictive Check Stratified by Treatment Cycle for the
Final Population PK-PD Model.
[0083] A visual predictive check based on 100 simulated datasets showed reasonably good agreement between the median and the lOth and 90th percentiles of the observed and simulated platelet counts over time since the start of the treatment cycle. For the final dataset, the R Shiny application was used to simulate 200 patients undergoing chemotherapy with either 100 units/m2 of CPX-351 via 90-minute IV infusion at 0, 48, and 96 hours (Figure 1B) or with 7+3 as administered in Study 301. Without platelet transfusions, 186 (93.0%) patients reached incomplete recovery (platelet count >50 x 109/L) within the 96-day simulation period, and the median time from first CPX-351 dose to recovery of platelet count >50 x 109/L was 618 hours (25.8 days). With intermittent platelet transfusions (6 transfusions every 96 hours starting at 240 hours), 191 (95.5%) patients reached incomplete recovery (platelet count >50 x 109/L) within the 96-day simulation period, and the median time from first CPX-351 dose to recovery of platelet count >50 x 109/L was 528 hours (22.0 days). [0084] Conclusions: a maturation PK-PD model was developed to characterize the effect of CPX-351 and 7+3 on platelets in patients with AML, ALL, or MDS. CPX-351 achieved a rapid and substantial suppression of platelet counts, with an estimated IC50 of 0.324 mM. The model accounted for the confounding nature of platelet transfusions on platelet dynamics during chemotherapy-induced thrombocytopenia. The final model was successfully embedded in an R Shiny application that can be utilized to evaluate the temporal events of thrombocytopenia following administration of various CPX-351 or 7+3 dosing regimens with intermittent platelet transfusions. These are used to design an appropriate treatment cycle for LiT administration of CPX-351.
Example 2
Modeling of Chemotherapy-induced Neutropenia (ON ) in Patients Treated With CPX-351 and“7+3”
[0085] The CPX-351 and 7+3 studies and Cytarabine/Daunorubicin modeling was performed as in Example 1.
[0086] Results shown below:
Table 4. Population PK Parameters for GCSF Agents
Population mean Interindividual
Parameter equation variability Reference
Figure imgf000020_0001
Wiczhng et
Fi 0.105
al.5
ka,h_1 Wiczhng et
0.403
al.5
F2 0.586 Wiczhng et al.
D2, h 6.6 Wiczhng et al. KD, nM 0.0237 0.527 (72.6% CV) Melhem et al. CL, L/h 0.833*(WTKG/70)°·641 0.138 (37.1% CV) Melhem et al. Vc, L 3.12*(WTKG/70)°943 0.080 (28.2% CV) Melhem et al.
Pegfilgraslim
Figure imgf000020_0002
Fi 0.646 0.194 (44.0% CV) Melhem et al. ka, h 1 0.0188 0.051 (22.5% CV) Melhem et al
KD, nM 0.0959 0.527 (72.6% CV) Melhem et al.
CL, L/h 0.362*(WTKG/70)a641 0.138 (37.1% CV) Melhem et al.
Vc, L 5.76*(WTKG/70)°943 0.080 (28.2% CV) Melhem et al.
Filgrastim or pegfilgraslim
Figure imgf000020_0003
kint, h 1 0.113 0.325 (57.0% CV) Melhem et al.
Figure imgf000020_0004
Table 4. Population PK Parameters for GCSF Agents
Population mean Interindividual
Parameter equation variability Reference
PK, pharmacokinetic; GCSF, granulocyte colony-stimulating factor; Fi, bioavailability of first-order absorption process; ka, first-order absorption rate constant; F2, bioavailability of zero-order absorption process; D2,m, duration of the zero-order absorption process; KD, equilibrium dissociation constant for GCSF and GCSF receptor; CV, coefficient of variation; CL, clearance; WTKG, body weight in kg; Vc, central volume of distribution; ki„t, first-order rate constant for internalization of GCSF receptor complexed with filgrastim or pegfilgrastim.
[0087] Population PK-PD analysis was conducted using NONMEM version 7.3 via implementation of the first-order conditional estimation method with h-e interaction. Previously developed population PK models for CPX-351 or 7+3 (2-compartment disposition) were used to generate patient-specific cytarabine and daunorubicin concentration-time profiles. Previously reported population PK models for GCSF agents (1- compartment with target-mediated drug disposition) were used to predict patient-specific concentration-time profiles following exogenous GCSF administration based on individual dosing histories and population mean PK parameters.
[0088] In the population PK-PD model for chemotherapy-induced neutropenia (CIN), data from Cycle 1 were excluded if patients had an absolute neutrophil count (ANC) < 1.0 x l09/L prior to the first treatment cycle; data from subsequent cycles were only included if the ANC returned to >l.0xl09/L prior to treatment. ANC versus time data were described by a modified maturation PK-PD model proposed by Friberg et al. Stimulation of neutrophil proliferation and maturation by exogenous granulocyte colony stimulating factor (GCSF) agents was driven by the fraction of GCSF receptors bound to exogenous GCSF. Inhibition of neutrophil proliferation by CPX-351 (or 7+3) was driven by an Imax function of the sum of the molar concentrations of cytarabine and daunorubicin. Interindividual variability was estimated for select structural PK-PD model parameters using exponential error models. Previously reported population PK models for GCSF agents (1 compartment with target- mediated drug disposition) were used to predict patient-specific concentration-time profiles following exogenous GCSF administration (filgrastim and/or pegfilgrastim) based on individual dosing histories and population mean PK parameters. A graphical screening procedure was first conducted to examine the relationship between patient covariates and key PK-PD model parameters, followed by stepwise forward selection (a = 0.01) and backward elimination (a = 0.001) to evaluate covariate effects. Baseline Patient Demographics were: Weight, height, age, body mass index, body surface area, sex, race, and ethnicity. Baseline Clinical Laboratory Measures were: Albumin, alkaline phosphatase, alanine aminotransferase, aspartate aminotransferase, total bilirubin, white blood cell count, absolute neutrophil count, platelet count, and creatinine clearance. Disease -related Indices were: Cancer type and Eastern Cooperative Oncology Group performance status. An R Shiny application was developed in conjunction with the mrgsolve package (Metrum Research Group) so the population PK-PD model could be used to simulate neutrophil (or platelet) dynamics following administration of CPX-351 (or 7+3) and GCSF in various dosing regimens; relevant patient demographics were generated with distributions that were comparable to those in the analysis dataset.
[0089] Results:
Table 5. Summary Statistics of Selected Baseline Patient Demographics and Clinical Laboratory Measures in the Final Analysis Dataset
Mea SD Medi M M
Variable n %
n an in ax
Age, y 129 66.1 9.1 67.0 23 81
129 82.4 20.4 78.7 39 15 Weight, kg
.5 6
129 1.96 0.28 1.94 1 2
BSA, m2
3 8
129 67.1 61.0 43.0 2 28
Platelet count, 109/L
9
128 4.42 7.57 1.98 0a 59
ANC, 109/L
.4
Figure imgf000022_0001
SD, standard deviation; BSA, body surface area; ANC, absolute neutrophil count; GCSF, granulocyte colony-stimulating factor; ECOG, Eastern Cooperative Oncology Group; ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia.
aFor patients who were neutropenic (ANC <1.0 x 109/L) prior to the first treatment cycle, data from Cycle 1 were excluded from analysis, and data from subsequent cycles were only included if the ANC returned to >1.0 x l09/L prior to treatment. However, for the purposes of covariate evaluation, the baseline ANC from Cycle 1 was used, even if the value was <1.0 x l09/L.
[0090] The early analysis dataset consisted of 1,797 ANC observations from 129 patients, of whom 43 (33.3%) received GCSF therapy at least once during the study. Incorporation of the stimulatory effect of exogenous GCSF on neutrophil proliferation and maturation significantly improved the model fit (P <0.00001). During covariate evaluation, a strong inverse correlation was observed between mean transit time and baseline ANC ( P <0.00001); no other potential covariates met the criteria for inclusion. In the late stages of model development, Imax was estimated to be 1.05 and subsequently fixed at 1, which produced a minor increase of 4.5 units in the minimum value of objective function.
Table 6. Model Fitted PD Parameters for Neutrophils
Figure imgf000023_0001
PK, pharmacokinetic; PD, pharmacodynamic; SEM, standard error of the mean; Circo, baseline ANC; MTT, mean transit time (4/ktr); ANC, absolute neutrophil count; Imax, maximum inhibition of neutrophil proliferation; IC50, composite concentration (cytarabine + daunorubicin) at which inhibition is 50% of Imax; yT, feedback function exponent; Stimi, slope parameter for stimulation of neutrophil proliferation by the fraction of GCSF receptors bound to exogenous GCSF; Stini2, slope parameter for stimulation of neutrophil maturation by the fraction of GCSF receptors bound to exogenous GCSF; w2, interindividual variability; CV, coefficient of variation; s2, residual variability; SD, standard deviation.
[0091] Parameters in the PK-PD model were estimated with high precision (<26% standard error of the mean). The population mean for Circo before treatment was estimated to be similar for CPX-351 and 7+3, while the population mean for MTT of maturation was slightly longer with CPX-351 versus 7+3. 7+3 was more potent than CPX-351. Including GCSF effect in the model significantly improved the model fit.
[0092] The median time to observe the first blood neutrophil count <0.5 x 109/L was later following CPX-351 (8.3 days) versus 7+3 (7.4 days) treatment. The median duration with neutrophil counts <0.5 x 109/L was longer with CPX-351 (23 days) than with 7+3 (14 days). The median lowest neutrophil counts were well below 0.2 x 109/L for both CPX-351 and 7+3. These results are summarized in Table 6A below.
Table 6A. Model-simulated Neutrophil Parameters After CPX-351 or 7+3 Treatment
Figure imgf000024_0001
[0093] Figure 2B shows a visual predictive check stratified by treatment cycle for the final population PK-PD model. It is based on 100 simulated datasets showed reasonably good agreement between the median and the lOth and 90th percentiles of the observed and simulated ANC over time since the start of the treatment cycle. The R Shiny application was used to simulate 200 patients undergoing chemotherapy with 100 units/m2 of CPX-351 via 90-minute IV infusion at 0, 48, and 96 hours (Figure 1C). Without GCSF therapy, 172 (86%) patients recovered to ANC >1.0 x 109/L within the 96-day simulation period, and the median time from first CPX-351 dose to recovery of ANC >1.0 x 109/L was 788 hours (32.8 days). With GCSF therapy (480 pg filgrastim administered subcutaneously at 240, 360, and 480 hours), 176 (88%) patients recovered to ANC >1.0 x 109/L within the 96-day simulation period, and the median time from first CPX-351 dose to recovery of ANC >1.0 x 109/L was 730 hours (30.4 days).
[0094] Conclusions: a maturation PK-PD model was developed to characterize the effect of CPX-351 and 7+3 on ANC in patients with AML or ALL or MDS. CPX-351 achieved a rapid and maximal suppression of ANC (Imax = 1) and had an estimated IC50 of 24.9 mM. The model accounted for the confounding nature of concurrent GCSF therapy on neutrophil dynamics during CIN. The final model was successfully embedded in an R Shiny application that can be utilized to evaluate the temporal events of neutropenia following administration of CPX-351 or 7+3 and GCSF therapy in various dosing regimens. The myelosuppressive effects of CPX-351 were different from 7+3 in terms of duration of myelosuppression and time to neutrophil count <0.5 x 109/L. The median time for initial detection of myelosuppression with CPX-351 was 1 to 2 days later than with 7+3. Additionally, the median duration of myelosuppressive effects was longer with CPX-351 than with 7+3. These results may have implications for the clinical monitoring scheme of patients with AML who receive CPX-351 therapy. These are used to design an appropriate treatment cycle for LIT administration of CPX-351.
Example 3
Simulation of Alternative Dose-Regimen for CPX-351
[0095] Simulation of myelosuppression profile for lower dose/regimen scenarios:
• Day 1+5; various doses < 100 units/m2
• Day 1+8; various doses < 100 units/m2
• Day 1+8+15; various doses < l00 units/m2
• Other schemes, various doses < 100 units/m2
[0096] The PK/PD modeling described in Examples 1 and 2 is used to further examine the cell cycle time and Vyxeos treatment effect.
[0097] Simulation Outcomes for Absolute Neutrophil Count (ANC): Simulations were conducted using previous developed mechanistic PK/PD model for absolute neutrophil count. Figure 8 shows the period of ANC below 500/uL after administration of CPX-351.
The duration of ANC<500/uL increases as dose increases. Depending on regimen, the duration can be different for a given dose. The duration ANC suppression after Vyxeos standard Day 1, 3, & 5 regimen or 7+3 treatment is also shown in the figure, where Vyxeos showed a longer duration of ANC suppression than 7+3. This longer suppression is consistent with clinical observations.
[0098] Simulation outcome for platelet count: Simulations were conducted using previous developed mechanistic PK/PD model for platelet count. During simulation, no platelet infusion was given and the starting platelet count is around 10,000 /uL. Figures 9 A and 9B show the period of platelet count below 50, 000 or 20,000 /uL after administration of CPX-351. The duration of platelet suppression increases as dose increases. Depending on regimen, the duration can be different for a given dose. The duration platelet suppression after Vyxeos standard Day 1, 3, & 5 regimen or 7+3 treatment is also shown in the figure, where Vyxeos showed a longer duration of ANC suppression than 7+3. This longer suppression is consistent with clinical observations. Additionally, Vyxeos Day 1, 3, 5 regimen produced most longest suppression period (<20,000/uL) than other regimens. [0099] Simulation outcome for platelet count: this data appears to be the first time to observe a platelet count below a certain threshold level that is not related to regimen. Rather, it is related to the drug properties and dose. Vyxeos treatment results in a later observation than 7+3 treatment. With increase of dose of Vyxeos, the median start-time of observing platelet count <20,000/uL approaches 258 h (Day 10 - 11 post the first dose). With increase of dose of Vyxeos, the median start-time of observing platelet count<50, 000/uL approaches 154 h (Day 6 - 7 post the first dose). For 7+3, the median start-time of observing platelet count <20, 000/uL is around 200 h (Day 8 - 9 post start of the treatment). For 7+3, the median start-time of observing platelet count <50, 000/uL is around 139 h (Day 5 - 6 post start of the treatment). The results are shown in Figures 10A and 10B.
[0100] Simulation outcome for neutrophils: this data appears to be the first time to observe a ANC below a certain threshold related to the drug properties and dose. Vyxeos treatment results in a late-onset but prolonged neutropenia than 7+3 treatment. With increase of dose of Vyxeos, the median start-time of observing ANC<500/uL approaches 200 h (Day 8 - 9 post the first dose). For 7+3, the median start-time of observing ANC<500/uL is around 178 h (Day 7 - 8 post start of the treatment). The results are shown in Figure 11.
Example 4
CPX-351 and Venetoclax show Synergy in Ex Vivo Leukemia Models
[0101] In order to evaluate the cytotoxicity of CPX-351 in combination with
Venetoclax, various drug:drug combinations were tested in 15 Passage 1 (Pl) models of human Acute Myeloid Leukemia (AML) and assayed for cell viability.
[0102] The study type and duration used was an ex-vivo cell killing assay with primary leukapharesis-derived AML cells using a 96-well plate format, 90 wells, 6-day incubation, Cell Titer Glo viability endpoint with ICso.
[0103] AML Models used were CTG-2226, CTG-2227, CTG-2228, CTG-2231 to
CTG-2234, CTG-2236, CTG-2237, CTG-2238, CTG-2242, CTG-2243, CTG-2251, CTG- 2255, and CTG-2299. The IDH1/2 status for each cell line was noted and some were Wildtype while others were IDH1 or IDH2 mutant. AML cells were seeded at a density of 20,000 cells/lOOul per well in a 96 well plate in enriched media. Therapeutic agents were added to wells on Day 0 along with cell plating according to the experimental design in volume of 1 OOul/wcll bringing the total volume to 200ul/well. [0104] CPX-351 and Venetoclax were each tested alone (100 uM) as well as in combination as outlined in Table 7 below:
Table 7: CPX-351 + Venetoclax screening in AML cell models
Figure imgf000027_0001
[0105] Cytarabine (5000nM) concentration was used as a positive control; Media + vehicle were used as negative control. Plates were kept in 37°C/5% C02 incubator. Media was not changed during the 6-day incubation period with test agents. On day 6, cell viability was tested using Cell Titer Glo assay.
[0106] Cell Titer Glo Assay: Plates were removed from incubator and equilibrated to room temperature up to 30 minutes. lOOul of Cell Titer Glo was added to wells and mixed for 2 minutes by keeping the plates on plate rocker. The plates were incubated at room temperature for 10 minutes to stabilize luminescent signal. Luminescence was recorded using Tecan plate reader.
[0107] Data Analysis: data was reported as changes in RLU units to different drug concentrations as luminescence read is directly proportional to cell viability.
[0108] The results from first 7 cells all demonstrated strong synergy. This may be due to the high concentrations of venetoclax in the incubation and so testing of lower concentrations is in progress. The results from remaining 8 cell models demonstrated either synergistic or additive effects of combining Vyxeos and Venetoclax in 7 cell models, with only showing antagonistic effect. Figures 12 and 13 show the change in IC50 for the CTG- 2226 and CTG-2233, cell lines respectively. Example 5
Clinical Trial of CPX-351 and Venetoclax
[0109] A Phase lb trial of CPX-351 Lower Intensity Therapy (LIT) plus venetoclax is conducted as Lirst Line Treatment for Subjects with AML who are ineligible for Intensive Chemotherapy.
[0110] The primary objectives are to determine the maximum tolerated dose (MTD) of the combination of CPX-351 and venetoclax when administered to subjects with newly diagnosed Acute Myeloid Leukemia (AML) who are ineligible for intensive chemotherapy (ICT). In addition, the safety of this combination is to be determined.
[0111] Secondary objections are to perform initial assessments of efficacy, including complete remission (CR), complete remission with incomplete hematologic recovery (CRi), composite complete remission rate (CRc: CR + CRi), overall response rate (ORR; CR + CRi + partial response [PR]) and assessment of Minimal Residual Disease (MRD) status (negative / positive) in subjects with documented CR or Cri. The study is also to determine the pharmacokinetics (PK) of CPX-351 and venetoclax when given in combination.
[0112] The duration of remission (DOR) and overall survival (OS) and event-free survival (EPS) l-year after first administration of the study treatment will also be explored.
[0113] The study will comprise 2 phases: a Dose Escalation Phase and an Expansion
Phase, in which all subjects will receive a combination of CPX-351 and venetoclax. In the Dose Escalation Phase, a 3 + 3 design will be employed and enroll up to 24 subjects with newly diagnosed AML who are ineligible for standard ICT using stepwise dose escalations of CPX-351 in combination with 400 mg venetoclax in each cohort to determine the MTD for the combination.
[0114] After determining the MTD of the combination of CPX-351 and venetoclax, the Expansion Phase will commence with 20 additional subjects treated with the combination to determine an initial response rate based on both morphologic assessment and MRD.
Stopping rules for excessive toxicity and for futility will be included for the Expansion Phase.
[0115] Subjects will be followed for safety (1 month after End of Treatment) and every 2 months (± 2 weeks) for survival (up to 1 year after the first administration of study treatment). Each subject will be evaluated for response at the end of each cycle. Those subjects with CR, CRi or PR will be offered up to a total of 4 cycles of CPX-351 and venetoclax, as deemed appropriate by the treating physician. Subjects with NR (no response) after 2 cycles will stop treatment. Subjects who have completed 4 cycles of treatment will be managed at the investigator’s discretion, as per institutional guidelines.
[0116] In each cycle, CPX-351 will be administered on Days 1 and 3, and venetoclax will be administered on Days 2 through 21. During Cycle 2 Day 3 of the Dose Escalation Phase, venetoclax will be administered at the study site during the infusion of CPX-351 to permit PK assessment.
[0117] Blood collection for PK studies will be conducted during the first and second cycles of therapy for all dose cohorts in the Dose Escalation Phase. Blood collection for sparse PK sampling will also be performed in the Expansion Phase for subjects receiving their Day 3 and Day 4 care at the study site. During Cycle 1 Day 3 of the Expansion Phase, venetoclax will be administered at the study site during the infusion of CPX-351 to permit PK assessment. The
[0118] Dose Escalation and Definition ofMTD. The Dose Escalation Phase will employ a 3 + 3 design to determine the DLTs and MTD as follows. The first 3 subjects will be treated at Dose Level 1. If this is deemed safe (see specifics below), then dose escalations will proceed as follows:
[0119] Dose escalation algorithm: If none of the first 3 evaluable subjects at a dose level experience a DLT, then the next 3 subjects will be treated at the next dose level (dose escalation).!† 1 out of the first 3 evaluable subjects experiences a DLT, then an additional 3 subjects will be treated at the same dose (for a total of 6 subjects at a dose level). If 1 out the 6 total evaluable subjects at a dose level experiences a DLT, then the next 3 subjects will be treated at the next dose level (dose escalation). If 2 or more of the first 3 evaluable subjects at a dose level experience a DLT, then no additional subjects will be treated at this or higher dose level. If 2 or more of the 6 total evaluable subjects experience a DLT, then no additional subjects will be treated at this or higher dose level.
[0120] MTD determination algorithm: If > 2 out of 3 or > 2 out of 6 evaluable subjects experience a DLT at Dose Level 1 (starting dose), then the study will stop and no MTD will be determined. Otherwise, the MTD will be determined as the highest dose level at which either 0 out of 3 or 1 out of 6 evaluable subjects experiences a DLT. If this is Dose Level 4, then the maximum administered dose will be Dose Level 4, and for the purposes of this study, Dose Level 4 will be considered the MTD. The DLT observation period will be Days 1 to 49 after starting treatment, with a minimum observation period of 28 days. Subjects who are eligible to progress to a second cycle, or who have transitioned to an alternative therapy, may have an abbreviated DLT observation period (ie, less than 49 days). Subjects who do not complete a full cycle of therapy due to disease-related mortality or who withdraw from the study for reasons unrelated to drug effects, and have not experienced a DLT, will not be evaluable for DLTs. These subjects will be replaced. A regular teleconference, occurring approximately every 3 weeks, will be held among the Jazz medical and clinical team and study investigators to review safety data (hereafter referred to as the Safety Assessment Committee [SAC]). The SAC will also meet at the completion of each dosing cohort to determine whether 1) dose escalation will proceed with the next cohort; 2) the current dose level requires additional assessment; 3) the MTD has been reached; or 4) whether the study will be stopped. To mitigate the risk of potential tumor lysis syndrome, during the first cycle a venetoclax dose ramp-up will occur on Days 2 through 4, followed by treatment at the target dose on Days 5 to 21. For subsequent cycles, venetoclax will be administered at the full target dose during Days 2 through 21. d
[0121] Main Inclusion Criteria (full list provided in body of protocol)
• Subject must have newly diagnosed AML with histological confirmation by World
Health Organization (WHO) criteria
Definition of subjects who are ineligible for standard ICT:
• Each subject must meet the following criteria characterizing him / her as ineligible to receive ICT within 21 days prior to the first day of therapy to be enrolled in the study: o > 75 years of age
OR
o > 18 to 74 years of age and fulfilling at least 1 criteria associated with lack of fitness for ICT as follows:
- Eastern Cooperative Oncology Group (ECOG) Performance Status of 2 to 3;
- Cardiac history of Congestive Heart Failure (CHF) requiring treatment or left ventricular ejection fraction (LVEF) < 50%.
- Diffusing Capacity of the Lung for Carbon Monoxide (DLCO) < 65% or Forced Expiratory Volume in 1 second (FEV1) < 65%;
- Creatinine clearance (CrCl) > 30 mL/min to < 45 mL/min calculated by the
Cockcroft-Gault formula;
- Moderate hepatic impairment with total bilirubin > 1.5 to < 3. O x Upper Limit of Normal (ULN);
- Other comorbidity that the physician judges to be incompatible with conventional intensive chemotherapy which must be reviewed and approved by the study medical monitor before study enrollment.
[0122] Additional Criteria:
In addition, all subjects must meet the following criteria:
• If the subject is > 75 years of age, then ECOG Performance Status must be 0-2.
• Subject must have adequate renal function as demonstrated by a CrCl > 30 mL/min
(calculated by the Cockcroft Gault formula or measured by 24-hour urine collection). • Subject must have adequate liver function as demonstrated by:
o Aspartate aminotransferase (AST) < 3.0 x ULN*
o Alanine aminotransferase (ALT) < 3.0 x ULN*
o Bilirubin < 1.5 x ULN (subjects who are < 75 years of age may have bilirubin of < 3.0 x ULN)*
*Unless considered to be due to leukemic organ involvement.
• Female subjects must be either postmenopausal defined as:
o Age > 55 years with no menses for > 2 years without an alternative medical cause o OR
o Age < 55 years with no menses for > 12 months without an alternative medical cause AND a follicle-stimulating hormone level > 40 IU/L;
o OR
o Permanently surgical sterile (bilateral oophorectomy, bilateral salpingectomy or hysterectomy);
OR
• A woman of childbearing potential practicing at least 1 protocol specified method of birth control starting at Study Day 1 through at least 6 months after the last dose of study treatment.
• A woman of childbearing potential must have negative results for pregnancy test
performed:
o At Pretreatment with a serum sample obtained within 14 days prior to the first study treatment administration, and
o Prior to dosing with urine sample obtained on Cycle 1 Day 1, if it has been > 7 days since obtaining the serum pregnancy test results
o Subjects with borderline pregnancy tests at Pretreatment must have a serum
pregnancy test > 3 days later to document continued lack of a positive result.
• Male subjects who are sexually active, must agree, from Study Day 1 through at least 6 months after the last dose of study treatment, to practice protocol specified methods of contraception. Male subjects must agree to refrain from sperm donation from initial study treatment administration through at least 6 months after the last dose of study treatment.
• Subject must have a white blood cell count < 25 x l09/L. (Note: subjects who have undergone hydroxyurea administration or leukapheresis for therapeutic cytoreduction will be considered eligible).
[0123] Main Exclusion Criteria (full list provided in body of protocol)
Subjects who meet any of the following criteria will be excluded from the study:
• Subject has ECOG Performance status > 3, regardless of age.
• Subject has received any prior treatment for AML with the exception of hydroxyurea, which is allowed up until the initiation of therapy / first dose of CPX-351. (Note: Prior treatment for Myelodysplastic Syndrome is allowed except for use of cytarabine or daunorubicin.)
• Subject has favorable risk cytogenetics ((t8;2l), inv(l6), t(l6;l6) or t 15; 17) karyotype abnormalities) as categorized by the National Comprehensive Cancer Network (NCCN) Guidelines Version 2.2014 for AML • Subject had an antecedent myeloproliferative neoplasm (MPN) including myelofibrosis, essential thrombocytosis, polycythemia vera, or chronic myelogenous leukemia (CML) with or without BCR-ABL 1 translocation and AML with BRC-ABL 1 translocation.
• Subject has acute promyelocytic leukemia (APL).
• Subject has known Central Nervous System (CNS) involvement with AML.
• Subject has known Human Immunodeficiency Virus (HIV) infection (due to potential drug-drug interactions between antiretroviral medications and venetoclax). HIV testing will be performed at Pretreatment, if required per local guidelines or institutional standards.
Subject is known to be positive for hepatitis B virus (HBV), or hepatitis C virus (HCV) infection. (Inactive hepatitis carrier status or low viral hepatitis titer on antivirals
[nonexclusionary medications] are not excluded.)
[0124] Test Product, Dose and Mode of Administration:
[0125] CPX-351 is provided as a sterile, preservative-free, purple, lyophilized cake in a single-use vial. Each vial of CPX-351 contains 44 mg daunorubicin and 100 mg cytarabine. After reconstitution (but before final dilution) each mL contains 2.2 mg daunorubicin and 5 mg cytarabine.CPX-35l is administered as an IV infusion over approximately 90 minutes. Venetoclax is available in 3 strengths and is self-administered as an oral dose once daily with a meal and water:
• The 100 mg tablet is provided as an oblong, biconvex shaped, pale yellow film -coated tablet debossed with“V” on one side and“100” on the other side.
• The 50 mg tablet is provided as an oblong, biconvex shaped, beige film-coated tablet debossed with“V” on one side and“50” on the other side.
• The 10 mg tablet is provided as a round, biconvex shaped, pale yellow film -coated tablet debossed with“V” on one side and“10” on the other side.

Claims

Claims
1. A pharmaceutical composition comprising CPX-351 as the sole active ingredient that supplies daunorubicin and cytarabine for use in a treatment cycle to treat a hematological proliferative disorder in a subject ineligible for standard intensive chemotherapy (an ISICT subject), which treatment cycle comprises administering said composition to deliver less than 300 mg/m2 of cytarabine over the course of the treatment cycle.
2. The composition of claim 1 wherein said treatment delivers less than 250 mg/m2 of cytarabine over the course of the treatment cycle.
3. The composition of claim 1 wherein said treatment cycle delivers daily dose(s) of less than or equal to 135 mg/m2 per day of cytarabine and consists of 2 days.
4. The composition of claim 1 wherein said composition is administered for no more than a total of 2 days at intervals of days 1 and 3 or days 1 and 5 or days 1 and 8.
5. The composition of claim 1 wherein said composition delivers daily dosages of less than 32 mg/m2 per day of cytarabine and the treatment cycle consists of 3 days or less.
6. The composition of claim 1 that is administered intravenously in less than 3 hours.
7. The composition of any of claims 1-5 wherein the protocol further includes administering an agent that has been shown effective to treat a leukemia.
8. The composition of claim 7 wherein said agent is a BCL-2 inhibitor or a hypomethylation agent.
9. The composition of claim 7 wherein said agent is mylotarg, midostaurin, venetocla or idhifa.
10. The composition of any of claims 1-5 wherein the hematologic proliferative disorder is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL) or acute promyelocytic leukemia (APL), myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN).
11. The composition of claim 7 wherein the hematologic proliferative disorder is AML, ALL, CLL, APL, MDS or MPN.
12. A method to treat a hematologic proliferative disorder in an ISICT subject, which method comprises administering to said subject a pharmaceutical composition comprising CPX-351 as the sole active ingredient that supplies daunorubicin and cytarabine in a treatment cycle that comprises delivering less than 300 mg/m2 of cytarabine over the course of the treatment cycle.
13. The method of claim 12 wherein said treatment delivers less than 250 mg/m2 of cytarabine over the course of the treatment cycle.
14. The method of claim 12 wherein said treatment cycle delivers daily dose(s) of less than or equal to 135 mg/m2 per day of cytarabine and consists of 2 days.
15. The method of claim 12 wherein said composition is administered for no more than a total of 2 days at intervals of days 1 and 3 or days 1 and 5 or days 1 and 8.
16. The method of claim 12 said composition delivers daily dosages of less than 32 mg/m2 per day of cytarabine and the treatment cycle consists of 3 days or less.
17. The method of claim 12 wherein the composition is administered intravenously in less than 3 hours.
18. The method of claim 12-17 wherein the protocol further includes administering an agent that has been shown effective to treat a leukemia.
19. The method of claim 12-17 wherein the hematologic proliferative disorder is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL) or acute promyelocytic leukemia (APL), myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN).
20. The method of claim 18 wherein the hematologic proliferative disorder is acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL) or acute promyelocytic leukemia (APL), myelodysplastic syndrome (MDS) or myeloproliferative neoplasm (MPN).
PCT/US2019/052952 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders WO2020068979A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
EP19864378.5A EP3856198A4 (en) 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders
MX2021003527A MX2021003527A (en) 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders.
JP2021540394A JP2022502498A (en) 2018-09-25 2019-09-25 Low intensity treatment for blood disorders
AU2019350759A AU2019350759A1 (en) 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders
KR1020217011132A KR20210065962A (en) 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders
CN201980063289.2A CN113164502A (en) 2018-09-25 2019-09-25 Hypo-intensive treatment of hematological disorders
BR112021005539-8A BR112021005539A2 (en) 2018-09-25 2019-09-25 low-intensity treatment of hematological disorders
CA3114002A CA3114002A1 (en) 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders
IL281729A IL281729A (en) 2018-09-25 2021-03-22 Cpx-351 low intensity treatment of hematological disorders
US17/338,561 US20210393665A1 (en) 2018-09-25 2021-06-03 Low-intensity treatment of hematological disorders

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201862736393P 2018-09-25 2018-09-25
US62/736,393 2018-09-25
US201862772372P 2018-11-28 2018-11-28
US62/772,372 2018-11-28

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US17279556 A-371-Of-International 2019-09-25
US17/338,561 Continuation US20210393665A1 (en) 2018-09-25 2021-06-03 Low-intensity treatment of hematological disorders

Publications (1)

Publication Number Publication Date
WO2020068979A1 true WO2020068979A1 (en) 2020-04-02

Family

ID=69950910

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/052952 WO2020068979A1 (en) 2018-09-25 2019-09-25 Low-intensity treatment of hematological disorders

Country Status (11)

Country Link
US (1) US20210393665A1 (en)
EP (1) EP3856198A4 (en)
JP (1) JP2022502498A (en)
KR (1) KR20210065962A (en)
CN (1) CN113164502A (en)
AU (1) AU2019350759A1 (en)
BR (1) BR112021005539A2 (en)
CA (1) CA3114002A1 (en)
IL (1) IL281729A (en)
MX (1) MX2021003527A (en)
WO (1) WO2020068979A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022233782A1 (en) 2021-05-03 2022-11-10 Lead Discovery Center Gmbh Composition comprising an inhibitor of mitochondrial transcription
US11980636B2 (en) 2021-11-18 2024-05-14 Jazz Pharmaceuticals Ireland Limited Treatment of hematological disorders

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3005076C (en) * 2015-11-11 2024-01-02 Celator Pharmaceuticals, Inc. Assays and methods for selecting a treatment regimen for a subject with leukemia

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2120568T3 (en) * 2007-02-16 2018-03-30 Celator Pharmaceuticals, Inc. Fixed drug ratios for treatment of hematopoietic cancers and proliferative disorders
CA3005076C (en) * 2015-11-11 2024-01-02 Celator Pharmaceuticals, Inc. Assays and methods for selecting a treatment regimen for a subject with leukemia
US20190151365A1 (en) * 2016-07-28 2019-05-23 Novartis Ag Combination therapies of chimeric antigen receptors and pd-1 inhibitors

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ALAN K. BURNETT, DONALD MILLIGAN, ARCHIE G. PRENTICE, ANTHONY H. GOLDSTONE, MARY F. MCMULLIN, ROBERT K. HILLS, KEITH WHEATLEY: "A Comparison Of Low-Dose Cytarabine And Hydroxyurea With Or Without All-Trans Retinoic Acid For Acute Myeloid Leukemia And High-Risk Myelodysplastic Syndrome In Patients Not Considered Fit For Intensive Treatment", CANCER, vol. 109, no. 6, 15 March 2007 (2007-03-15), pages 1114 - 1124, XP055701855, ISSN: 0008-543X, DOI: 10.1002/cncr.22496 *
ANDERSON MARY ANN, DENG JING, SEYMOUR JOHN F, TAM CONSTANTINE, KIM SU YOUNG, FEIN JOSHUA, YU LIJIAN, BROWN JENNIFER R, WESTERMAN D: "The BCL2 selective inhibitor venetoclax induces rapid onset apoptosis of CLL cells in patients via a TP53-independent mechanism", BLOOD, vol. 127, no. 25, 1 January 2016 (2016-01-01), pages 3215 - 3224, XP055701853, DOI: 10.1182/blood-2016-01-688796 *
ANDREW H. WEI ,STEPHEN A. STRICKLAND ,GAIL J. ROBOZ ,JING-ZHOU HOU ,WALTER FIEDLER ,TARA L. LIN ,GIOVANNI MARTINELLI ,ROLAND B. WA: "Updated Safety And Efficacy Results Of Phase 1/2 Study Of Venetoclax Plus Low-Dose Cytarabine In Treatment Naive Acute Myeloid Leukemia Patients Aged >65 Years And Unfit For Standard Induction Therapy(Abstract S473, Oral Presentation)", 24 June 2017 (2017-06-24), pages 1 - 2, XP055791775, Retrieved from the Internet <URL:https://library.ehaweb.org/eha/2017/22nd/181760/andrew.wei.updated.safety.and.efficacy.results.of.phase.1.2.study.of.html> [retrieved on 20191120] *
ANONYMOUS: "Liposome-Encapsulated Daunorubicin-Cytarabine And Gemtuzumab Ozogamicin In Treating Patients With Relapsed Or Regractory Myeloid Leukemia (AML) Or High Risk Myelodysplastic Syndrome", 13 September 2018 (2018-09-13), pages 1 - 5, XP055791782, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/history/NCT03672539?A=1&B=1&C=merged#StudyPageTop> [retrieved on 20191120] *
ANONYMOUS: "Liposome-Encapsulated Daunorubicin-Cytarabine And Venetoclax In Treating Participants With Relapsed, Refractory, on Untreated Acute Myeloid Leukemia (AML", 14 August 2017 (2017-08-14), pages 1 - 7, XP055791767, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/NCT03629171> [retrieved on 20191120] *
JEFFREY E LANCET, UY GEOFFREY L, CORTES JORGE E, NEWELL LAURA F, LIN TARA L, RITCHIE ELLEN K, STUART ROBERT K, STRICKLAND STEPHEN : "CPX-351 (Cytarabine And Daunorubicin) Liposome For Injection Versus Conventional Cytarabine Plus Daunorubicin In Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia", JOURNAL OF CLINICAL ONCOLOGY, vol. 36, no. 26, 10 September 2018 (2018-09-10), pages 2684 - 2692, XP055701861, DOI: 10.1200/JCO.2017.77.6112 *
LINU A. JACOB, S. APARNA, K. C. LAKSHMAIAH, D. LOKANATHA, GOVIND BABU, SURESH BABU, SANDHYA APPACHU: "Decitabine Compared With Low-Dose Cytarabine For The Treatment Of Older Patients With Newly Diagnosed Acute Myeloid Leukemia: A Pilot Study Of Safety, Efficacy, And Cost-Effectiveness", ADVANCES IN HEMATOLOGY, 1 January 2015 (2015-01-01), pages 1 - 6, XP055701854, ISSN: 1687-9104, DOI: 10.1155/2015/167029 *
See also references of EP3856198A4 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022233782A1 (en) 2021-05-03 2022-11-10 Lead Discovery Center Gmbh Composition comprising an inhibitor of mitochondrial transcription
US11980636B2 (en) 2021-11-18 2024-05-14 Jazz Pharmaceuticals Ireland Limited Treatment of hematological disorders

Also Published As

Publication number Publication date
EP3856198A1 (en) 2021-08-04
EP3856198A4 (en) 2022-06-22
KR20210065962A (en) 2021-06-04
CA3114002A1 (en) 2020-04-02
AU2019350759A1 (en) 2021-04-22
CN113164502A (en) 2021-07-23
BR112021005539A2 (en) 2021-06-29
US20210393665A1 (en) 2021-12-23
IL281729A (en) 2021-05-31
MX2021003527A (en) 2021-05-27
JP2022502498A (en) 2022-01-11

Similar Documents

Publication Publication Date Title
Blackstein et al. Gemcitabine as first-line therapy in patients with metastatic breast cancer: a phase II trial
Olmi et al. Locoregionally advanced carcinoma of the oropharynx: conventional radiotherapy vs. accelerated hyperfractionated radiotherapy vs. concomitant radiotherapy and chemotherapy—a multicenter randomized trial
Comella et al. Cisplatin, gemcitabine, and vinorelbine combination therapy in advanced non–small-cell lung cancer: a phase II randomized study of the Southern Italy Cooperative Oncology Group
JP2011505336A (en) Cytidine analogs for the treatment of myelodysplastic syndrome
Chua et al. Recent advances in management of small-cell lung cancer
US20210393665A1 (en) Low-intensity treatment of hematological disorders
Solal-Celigny et al. Age as the main prognostic factor in adult aggressive non-Hodgkin's lymphoma
Tomlinson et al. A phase II study of midostaurin and 5-azacitidine for untreated elderly and unfit patients with FLT3 wild-type acute myelogenous leukemia
Rosso et al. Etoposide versus etoposide plus high‐dose cisplatin in the management of advanced non‐small cell lung cancer: Results of a prospective randomized fonicap trial
Fountzilas et al. Postoperative dose-dense sequential chemotherapy with epirubicin, paclitaxel and CMF in patients with high-risk breast cancer: safety analysis of the Hellenic Cooperative Oncology Group randomized phase III trial HE 10/00
Marangolo et al. Dose and outcome: the hurdle of neutropenia
Fennelly et al. Simultaneous dose escalation and schedule intensification of carboplatin-based chemotherapy using peripheral blood progenitor cells and filgrastim: a phase I trial
US20220152074A1 (en) Treatment of hematological disorders
US11980636B2 (en) Treatment of hematological disorders
Ott et al. Phase II trial of dacarbazine and thalidomide for the treatment of metastatic melanoma
JP2016525530A (en) Decitabine combined with boraseltib for the treatment of acute myeloid leukemia and myelodysplastic syndrome
US20170173023A1 (en) Combination therapy with volasertib
Laurie et al. The impact of anemia on outcome of chemoradiation for limited small-cell lung cancer: a combined analysis of studies of the National Cancer Institute of Canada Clinical Trials Group
CN114650826A (en) Application of multi-target protein kinase inhibitor
Maharaj et al. Molecular remission using personalized low-dose immunotherapy with minimal toxicities for poor prognosis hematological and solid tumor cancers
Aglietta et al. Short term treatment with Escheria coli recombinant human granulocyte‐macrophage–colony stimulating factor prior to chemotherapy for Hodgkin disease
Drozd-Sokołowska et al. Outcomes of Jehovah’s Witnesses with hematological malignancies treated without transfusions—single center experience
Bhutani et al. A Clinical and Correlative Study of Elotuzumab, Carfilzomib, Lenalidomide, and Dexamethasone (Elo-KRd) for Lenalidomide Refractory Multiple Myeloma in First Relapse
Sun et al. EP12. 01-01 The Safety and Efficacy of Aumolertinib in Advanced NSCLC Patients with EGFRm Who were Intolerant to Osimertinib: A Retrospective Clinical Study
Chen et al. Severe lung injury induced by CD38 monoclonal antibody Daratumumab and bortezomib‐containing regimen in a patient with preexisting interstitial lung disease: a case report and literature review.

Legal Events

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

Ref document number: 19864378

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3114002

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021540394

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021005539

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217011132

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019350759

Country of ref document: AU

Date of ref document: 20190925

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019864378

Country of ref document: EP

Effective date: 20210426

ENP Entry into the national phase

Ref document number: 112021005539

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210323