WO2015126989A1 - Compositions and methods for treating neutropenia - Google Patents

Compositions and methods for treating neutropenia Download PDF

Info

Publication number
WO2015126989A1
WO2015126989A1 PCT/US2015/016447 US2015016447W WO2015126989A1 WO 2015126989 A1 WO2015126989 A1 WO 2015126989A1 US 2015016447 W US2015016447 W US 2015016447W WO 2015126989 A1 WO2015126989 A1 WO 2015126989A1
Authority
WO
WIPO (PCT)
Prior art keywords
csf
mcg
analog
subject
days
Prior art date
Application number
PCT/US2015/016447
Other languages
English (en)
French (fr)
Inventor
Lingtao Wu
Original Assignee
Children's Hospital Los Angeles
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 Children's Hospital Los Angeles filed Critical Children's Hospital Los Angeles
Priority to KR1020167025068A priority Critical patent/KR20160113302A/ko
Priority to AU2015219038A priority patent/AU2015219038B2/en
Priority to JP2016552579A priority patent/JP2017508737A/ja
Priority to US15/119,339 priority patent/US10286039B2/en
Priority to CN201580009151.6A priority patent/CN106413701A/zh
Priority to RU2016136833A priority patent/RU2016136833A/ru
Priority to EP15751576.8A priority patent/EP3107533A4/en
Priority to MX2016010699A priority patent/MX2016010699A/es
Priority to CA2937340A priority patent/CA2937340C/en
Publication of WO2015126989A1 publication Critical patent/WO2015126989A1/en
Priority to ZA2016/05109A priority patent/ZA201605109B/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • 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
    • A61K9/0012Galenical 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
    • A61K9/0021Intradermal administration, e.g. through microneedle arrays, needleless injectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • C07K14/535Granulocyte CSF; Granulocyte-macrophage CSF
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0642Granulocytes, e.g. basopils, eosinophils, neutrophils, mast cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/84Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving inorganic compounds or pH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the invention relates to compositions, methods and kits for treating a condition with retinoid agonists and granulocyte colony- stimulating factor (G-CSF or GCSF).
  • G-CSF granulocyte colony- stimulating factor
  • the invention also relates to compositions, methods and kits for treating a condition with ex vivo modified cells (such as granulocytes [for example, neutrophils, eosinophils and basophils] derived from hematopoietic stem cells [HSCs]) for cell therapies.
  • ex vivo modified cells such as granulocytes [for example, neutrophils, eosinophils and basophils] derived from hematopoietic stem cells [HSCs]
  • the condition includes but is not limited to various forms of neutropenia.
  • Neutrophils the most common granulocytes, constitute up to 70% of circulating leukocytes that primarily defend against pathogen infections.
  • Cancer chemotherapy-induced neutropenia is a hematological disorder marked with a significant decrease in the number of neutrophils in the bloodstream, leading to susceptibility to microbial infections. About 15- 40% of cancer patients require treatment delay and/or dose reduction because of chemotherapy-induced neutropenia. Mortality rate due to neutropenia is about 5% in patients with solid tumors and 11% in some hematological malignancies.
  • GCSF hematopoietic stem cells
  • chemotherapy comprises the majority of costs for both febrile neutropenia (33.5%) and non-febrile neutropenia (40.6%) patients.
  • the estimated cost for febrile neutropenia hospitalization (FNH) only for 2015 is about $2.1 billion.
  • This cost does not include the cost for treatment of non-febrile neutropenia, relapsed cancer patients requiring new chemotherapy, congenital neutropenia, idiopathic severe chronic neutropenia, cyclic neutropenia, and radiation-induced neutropenia.
  • neutropenia still remains a devastating issue for cancer patients, with substantial morbidity, mortality, and healthcare cost.
  • alternative modes of treatment are urgently needed for these neutropenic patients.
  • compositions, methods and kits that capitalize on the synergistic effect of retinoid agonist (e.g., Am80) with G-CSF on regeneration of mature neutrophils against infection and infection-related mortality.
  • Various embodiments of the present invention provide a method of treating, preventing, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the method may comprise or may consist of: providing a retinoid agonist; providing a G-CSF or an analog thereof; and administering a therapeutically effective amount of the retinoid agonist and the G-CSF or the analog thereof to the subject, thereby treating, preventing, reducing the severity of and/or slowing the progression of the condition in the subject.
  • the retinoid agonist and the G-CSF or the analog thereof may be in one composition or separate compositions.
  • the method may further comprise providing and administering a chemotherapeutic and/or an antimicrobial agent to the subject.
  • compositions comprising a retinoid agonist and a G-CSF or an analog thereof.
  • the composition may further comprise a chemotherapeutic and/or an antimicrobial agent.
  • kits for treating, preventing, reducing the severity of and/or slowing the progression of a condition in a subject comprises: a quantity of retinoid agonist; a quantity of G-CSF or an analog thereof; and instructions for using the retinoid agonist and the G-CSF or the analog thereof to treat, prevent, reduce the severity of and/or slow the progression of the condition in the subject.
  • the kit may further comprise a chemotherapeutic and/or an antimicrobial agent and instructions of using the chemotherapeutic and/or the antimicrobial agent to treat, prevent, reduce the severity of and/or slow the progression of the condition in the subject.
  • Various embodiments of the present invention provide a method of generating mature granulocytes.
  • the method includes providing a cell (e.g., a HSC, a bone marrow granulocytic progenitor cell and a hematopoietic CD34+ cell) and stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof, thereby generating mature granulocytes.
  • a cell e.g., a HSC, a bone marrow granulocytic progenitor cell and a hematopoietic CD34+ cell
  • stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof, thereby generating mature granulocytes.
  • Various embodiments of the present invention further provide a composition comprising regenerated mature granulocytes.
  • the composition further comprises a retinoid agonist and a G-CSF or an analog thereof.
  • Various embodiments of the present invention provide a method of treating, preventing, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the method includes providing a cell (e.g., a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34+ cell), stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof, thereby generating granulocytes and administering the generated granulocytes to the subject, thereby treating the condition in the subject.
  • the granulocytes are neutrophils.
  • generating granulocytes is stimulating formation of granulocytes.
  • Various embodiments of the present invention provide a method of treating, preventing, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the method includes providing a cell (e.g., a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34+ cell), stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof and administering the stimulated cell to the subject, thereby treating the condition in the subject.
  • a cell e.g., a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34+ cell
  • retinoid agonist examples include but are not limited to tamibarotene (Am80, retinobenzoic acid, Amnoid, Tamibaro), CH55, ITYA (ITYA-01115), Am580, BD4, or NRX195183 (also referred to as AGN195183), or their functional equivalents, analogs, or derivatives.
  • G-CSF examples include but are not limited to a wild type G-CSF, a recombinant G-CSF, a G-CSF monomer or dimer, a recombinant human G-CSF (rhG-CSF) dimer, a G-CSF mutant, a G-CSF fusion protein, a G-CSF fragment, a modified G-CSF polypeptide, a PEGylated G-CSF, a glycosylated G-CSF, and a G-CSF modified with Y-shaped branched polyethylene glycol (YPEG-G-CSF) at a specific lysine (Lys 17).
  • YPEG-G-CSF Y-shaped branched polyethylene glycol
  • compositions, methods and kits of the present invention find utility in the treatment of various conditions, including but not limited to various forms of neutropenia and neutropenia-related conditions.
  • the compositions, methods and kits of the present invention may be used in conjunction with cancer therapies (e.g., chemotherapy and radiation therapy) and/or treatments of microbial infections.
  • Various embodiments of the present invention provide methods for determining the efficacy of treatment in a subject in need thereof.
  • the methods include providing a sample from a subject, wherein the subject has been administered an effective a retinoid agonist and an effective amount of G-CSF, assaying the levels production of reactive oxygen species (ROS) and determining that the treatment is efficacious if the ROS production is higher than that of a reference sample or determining that the treatment is not efficacious if the ROS production is same as the reference sample or lower relative to the reference sample.
  • ROS reactive oxygen species
  • Figure 1 depicts, in accordance with various embodiments of the invention, phosphorylation regulation of cell cycle, RA target gene expression, and general transcription by both free CAK and TFIIH-containing CAK.
  • Figure 2 depicts, in accordance with various embodiments of the invention, intrinsically programmed cleavage of MAT 1 protein into M30 and pM9 fragments decreases CAK phosphorylation of RARa, leading to granulopoiesis underlying balanced myelopoietic expansion and differentiation.
  • CMP common myeloid progenitors
  • GMP granulocyte/monocyte progenitors
  • Gra granulocytes.
  • Figure 3 depicts, in accordance with various embodiments of the invention, as compared to RA, Am80 selectively activates RARa to induce a novel transcription response for stimulating both myeloid expansion and differentiation. Ribbon up-arrow indicates increased expression and ribbon down-arrow indicates decreased expression.
  • Figure 4 depicts, in accordance with various embodiments of the invention, a comparison of chemotherapy induced neutropenia in Human (left panel) and Mouse (right) to establish that the mouse model resembles human neutropenia.
  • Figure 5 depicts, in accordance with an embodiment of the invention, EC50 test show that human equivalent low and medium doses of Am80 and Am80-GCSF combination effectively promote recovery of neutrophils at neutrophil-decrease stage.
  • Figure 6 depicts, in accordance with various embodiments of the invention, that neutrophils induced by medium dose of Am80 in neutropenic mice at neutrophil-decrease stage display greater bactericidal activity than those induced by G-CSF or Am80-GCSF.
  • Figure 7 depicts, in accordance with various embodiments of the invention, neutrophils induced by low dose of Am80 in neutropenic mice at neutrophil-decrease stage display greater bactericidal activity than those induced by GCSF or Am80-GCSF.
  • Figure 8 depicts, in accordance with various embodiments of the invention, neutrophils induced by lower dose of Am80-GCSF combination in neutropenic mice at neutrophil-recovery stage display greater bactericidal activity than those induced by Am80 or GCSF.
  • Figure 9 depicts, in accordance with an embodiment of the invention, that survival mouse models demonstrate that low doses of Am80-GCSF reduce infection-related mortality of neutropenic mice.
  • Figure 10 depicts, in accordance with various embodiments of the invention, the molecular signaling of Am80.
  • Figure 11 depicts, in accordance with various embodiments of the invention, that the combination of Am80 and G-CSF promotes greater ROS production than Am80 or G-CSF alone while inhibiting leukemic growth.
  • Figure 12 depicts, in accordance with various embodiments of the invention, that the combination of Am80 and GCSF promotes greater ROS production than Am80 or G-CSF alone in normal hematopoietic CD34 + precursor cells.
  • FIG. 13 depicts, in accordance with various embodiments of the invention, G-CSF treatment promotes the growth of non-APL (acute promyelocytic leukemia) AML patient blasts compared to those treated with Am80 or ATRA.
  • non-APL acute promyelocytic leukemia
  • Figure 14 depicts, in accordance with various embodiments of the invention, that Am80-GCSF significantly promotes ROS production in peripheral blood mononuclear cells isolated from healthy human donors.
  • Figure 15 depicts, in accordance with various embodiments of the invention, that Am80-GCSF promotes significantly greater ROS production in healthy bone marrow (BM) cells or peripheral blood (PB) mononuclear cells obtained from acute myeloid leukemia patients.
  • BM bone marrow
  • PB peripheral blood
  • the term “comprising” or “comprises” is used in reference to compositions, methods, and respective component(s) thereof, that are useful to an embodiment, yet open to the inclusion of unspecified elements, whether useful or not. It will be understood by those within the art that, in general, terms used herein are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.).
  • the terms “treat,” “treatment,” “treating,” or “amelioration” when used in reference to a disease, disorder or medical condition refer to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent, reverse, alleviate, ameliorate, inhibit, lessen, slow down or stop the progression or severity of a symptom or condition.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease, disorder or medical condition is reduced or halted.
  • treatment includes not just the improvement of symptoms or markers, but also a cessation or at least slowing of progress or worsening of symptoms that would be expected in the absence of treatment. Also, “treatment” may mean to pursue or obtain beneficial results, or lower the chances of the individual developing the condition even if the treatment is ultimately unsuccessful. Those in need of treatment include those already with the condition as well as those prone to have the condition or those in whom the condition is to be prevented.
  • “Beneficial results” or “desired results” may include, but are in no way limited to, lessening or alleviating the severity of the disease condition, preventing the disease condition from worsening, curing the disease condition, preventing the disease condition from developing, lowering the chances of a patient developing the disease condition, decreasing morbidity and mortality, and prolonging a patient's life or life expectancy.
  • "beneficial results” or “desired results” may be alleviation of one or more symptom(s), diminishment of extent of the deficit, stabilized (i.e., not worsening) state of neutropenia, delay or slowing of neutropenia, and amelioration or palliation of symptoms associated with neutropenia.
  • administering refers to the placement an agent as disclosed herein into a subject by a method or route which results in at least partial localization of the agents at a desired site.
  • Neutropenia refers to a granulocyte disorder characterized by abnormally low levels of neutrophils in the blood. Neutropenia may be due to decreased production of white blood cells (for example, due to, including but not limited to therapeutic agents that affect the bone marrow, hereditary/congenital disorders that affect the bone marrow, alcoholism, hypersplenism, hyperthyroidism, Lupus, aplastic anemia, cancer (particularly blood cancers), radiation therapy, Vitamin B12, folate or copper deficiency and/or exposure to pesticides).
  • Neutropenia may also be due to destruction of white blood cells (for example, due to, including but not limited to viral infection, acute bacterial infections, certain autoimmune diseases, chemotherapy treatments and/or therapeutic agents). Neutropenia may also be due to marginalization, sequestration and/or migration of white blood cells (for example, due to, including but not limited to, hemodialysis, malaria and/or bacterial infections). Certain medications such as flecainide, phenytoin, indomethacin, propylthiouracil, carbimazole, chlorpromazine, trimethoprim/sulfamethoxazole (cotrimoxazole), clozapine, ticlodipine and certain anti-psychotic medications may also result in neutropenia.
  • Certain medications such as flecainide, phenytoin, indomethacin, propylthiouracil, carbimazole, chlorpromazine, trimethoprim/sulfamethoxazole (cotrimoxazole), clo
  • the methods and compositions of the invention may be used to treat, inhibit, reduce the severity of and/or promote prophylaxis of neutropenia resulting from any of the above causes.
  • the methods and compositions of the invention may also be used to treat, inhibit, reduce the severity of and/or promote prophylaxis of neutropenia-related conditions such as bacterial, fungal, viral, parasitic infections as well as radiation damage of innate immunity that result from any of the above causes of neutropenia by treating, inhibiting, reducing the severity of and/or promoting prophylaxis of neutropenia.
  • sample or "biological sample” as used herein denotes a sample taken or isolated from a biological organism, e.g., a tumor sample from a subject.
  • exemplary biological samples include, but are not limited to, a biofluid sample; serum; plasma; urine; saliva; a tumor sample; a tumor biopsy and/or tissue sample etc.
  • the term also includes a mixture of the above-mentioned samples.
  • sample also includes untreated or pretreated (or pre-processed) biological samples.
  • a sample can comprise one or more cells from the subject.
  • a sample can be a tumor cell sample, e.g. the sample can comprise cancerous cells, cells from a tumor, and/or a tumor biopsy.
  • a "subject” means a human or animal. Usually the animal is a vertebrate such as a primate, rodent, domestic animal or game animal. Primates include chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, and canine species, e.g., dog, fox, wolf. The terms, "patient”, “individual” and “subject” are used interchangeably herein.
  • the subject is mammal.
  • the mammal can be a human, non-human primate, mouse, rat, dog, cat, horse, or cow, but are not limited to these examples.
  • the methods described herein can be used to treat domesticated animals and/or pets.
  • mammal refers to any member of the class Mammalia, including, without limitation, humans and nonhuman primates such as chimpanzees and other apes and monkey species; farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs, and the like.
  • the term does not denote a particular age or sex. Thus, adult and newborn subjects, as well as fetuses, whether male or female, are intended to be included within the scope of this term.
  • Neurotropenia-induction or "neutrophil decrease” stage as used herein refers to cancer chemotherapy-induced neutropenia and coincides with a decrease in peripheral blood (PB) absolute neutrophil count (ANC) as shown in Fig. 4, left panel human days 0 - 12 and right panel mouse days 0 - 3.
  • PB peripheral blood
  • ANC absolute neutrophil count
  • Neurotropenia-recovery or "Neutrophil-recovery” or “neutrophil induction” stage as used herein refers to a recovery in PB neutrophil numbers (ANC) following the nadir or trough. See Fig. 4 days 12-24 (left panel) and days 3-9 (right panel).
  • a "subject” can be one who has been previously diagnosed with or identified as suffering from or having a condition (e.g., neutropenia or neutropenia-related disorders) in need of treatment or one or more complications related to the condition, and optionally, have already undergone treatment for the condition or the one or more complications related to the condition.
  • a subject can also be one who has not been previously diagnosed as having a condition or one or more complications related to the condition.
  • a subject can be one who exhibits one or more risk factors for a condition or one or more complications related to the condition or a subject who does not exhibit risk factors.
  • a "subject in need" of treatment for a particular condition can be a subject suspected of having that condition, diagnosed as having that condition, already treated or being treated for that condition, not treated for that condition, or at risk of developing that condition.
  • statically significant refers to statistical evidence that there is a difference. It is defined as the probability of making a decision to reject the null hypothesis when the null hypothesis is actually true. The decision is often made using the p- value.
  • G-CSF Granulocyte colony-stimulating factor
  • GCSF Granulocyte colony-stimulating factor
  • Retinoic acid enhances self-renewal of hematopoietic stem cells (HSC) through retinoic acid receptor gamma (RARy) activation while promoting differentiation of committed myeloid progenitors through RARa activation.
  • Am-80 is an all-trans retinoic acid (ATRA or RA) agonist designed to selectively binding RARa but not RARy. This selective property of Am80 can sufficiently promote granulocytic differentiation.
  • Am-80 tamibarotene enhances modest myelopoietic expansion while inducing mature neutrophils superior to G-CSF in combating bacterial infection. This effect arises from Am80-induced selective activation of transcription factor RARa to induce novel selective expression of RA target genes via inhibiting RARa phosphorylation, which is mediated by the CAK complex regulating both cell cycle and general transcription.
  • Am-80 may more effectively promote neutrophil differentiation than regenerating neutrophils from hematopoietic stem cells (HSC); b) Am80-induced significantly higher neutrophil bactericidal activity at the earlier developmental stage of mouse neutropenia (neutrophil- decrease stage) results from Am80-promoted differentiation of existing PB and BM granulocytic precursors into mature neutrophils; c) with low doses of Am80-GCSF treatment, Am80 can effectively differentiate GCSF-regenerated large numbers of granulocytic precursors into mature neutrophils against microbial infection; and d) Am80-GCSF combination is superior to GCSF in regeneration of mature neutrophils capable of reducing infection and infection-related mortality of neutropenic mice.
  • HSC hematopoietic stem cells
  • retinoid agonist and G-CSF as an effective therapy to treat cancer chemotherapy-induced neutropenia and beyond, including but not limited to treatment of congenital neutropenia (e.g., Kostmann syndrome, cyclic neutropenia, and Chediak Higashi).
  • congenital neutropenia e.g., Kostmann syndrome, cyclic neutropenia, and Chediak Higashi.
  • retinoid agonist and G-CSF for ex vivo granulocyte generation, particularly, neutrophils, for transfusion therapy to reduce the duration of neutropenia, and HSC transplantation of AML patients and as a radioprotective therapy against acute radiation syndrome.
  • the present invention provides a method of treating, preventing, reducing the likelihood of developing, reducing the severity of, promoting prophylaxis of and/or slowing the progression of a condition in a subject.
  • the method may comprise or may consist of: providing a retinoid agonist; providing a G-CSF or an analog thereof; and administering a therapeutically effective amount of the retinoid agonist and the G-CSF or the analog thereof to the subject, thereby treating, preventing, reducing the likelihood of developing, reducing the severity of, promoting prophylaxis of and/or slowing the progression of the condition in the subject.
  • the retinoid agonist and the G-CSF or the analog thereof are in one composition. In other embodiments, the retinoid agonist and the G-CSF or the analog thereof are in separate compositions.
  • the condition is neutropenia or microbial infection.
  • the microbial infection is bacterial, viral, fungal or parasitic infection.
  • the neutropenia is chemotherapy-induced neutropenia, congenital neutropenia, idiopathic neutropenia, cyclic neutropenia, autoimmune neutropenia; or radiation neutropenia.
  • the present invention provides a method of treating, preventing, reducing the likelihood of developing, reducing the severity of, promoting prophylaxis of and/or slowing the progression of neutropenia in a subject.
  • the method may comprise or may consist of: providing a retinoid agonist; providing a G-CSF or an analog thereof; and administering a therapeutically effective amount of the retinoid agonist and the G-CSF or the analog thereof to the subject, thereby treating, preventing, reducing the likelihood of developing, reducing the severity of, promoting prophylaxis of and/or slowing the progression of the condition in the subject.
  • the retinoid agonist and the G-CSF or the analog thereof are in one composition.
  • the retinoid agonist and the G-CSF or the analog thereof are in separate compositions.
  • neutropenia is cancer chemotherapy induced neutropenia (CCIN).
  • infectious diseases are caused by infectious bacteria.
  • infectious bacteria include: Helicobacterpyloris, Borelia burgdorferi, Legionella pneumophilia, Mycobacteria sps (such as M. tuberculosis, M. avium, M. intracellulare, M. kansaii, M.
  • compositions and methods described herein are contemplated for use in treating infections with these bacterial agents.
  • Other infectious organisms include: Plasmodium falciparum and Toxoplasma gondii.
  • the compositions and methods described herein are contemplated for use in treating infections with these agents.
  • infectious diseases may be caused by viral infections.
  • infectious viruses include: Retro viridae (for example, HIV); Picornaviridae (for example, polio viruses, hepatitis A virus; enteroviruses, human coxsackie viruses, rhinoviruses, echoviruses); Calciviridae (such as strains that cause gastroenteritis); Togaviridae (for example, equine encephalitis viruses, rubella viruses); Flaviridae (for example, dengue viruses, encephalitis viruses, yellow fever viruses); Coronaviridae (for example, coronaviruses); Rhabdoviridae (for example, vesicular stomatitis viruses, rabies viruses); Filoviridae (for example, ebola viruses); Paramyxoviridae (for example, parainfluenza viruses, mumps virus, measles virus, respiratory syncytial virus); Orthomyxoviridae (for example,
  • Examples of fungal infections that may be treated with the compositions and methods described herein include but are not limited to: aspergillosis; thrush (caused by Candida albicans); cryptococcosis (caused by Cryptococcus); and histoplasmosis.
  • examples of infectious fungi include, but are not limited to, Cryptococcus neoformans, Histoplasma capsulatum, Coccidioides immitis, Blastomyces dermatitidis, Chlamydia trachomatis, Candida albicans, and Aspergillus spp.
  • the compositions and methods described herein are contemplated for use in treating infections with these fungal agents.
  • the subject is a human.
  • the subject is a mammalian subject including but not limited to human, monkey, ape, dog, cat, cow, horse, goat, pig, rabbit, mouse and rat.
  • the subject has consistent microbial infection including but not limited to bacterial, viral, fungal and parasitic infections.
  • the retinoid agonist is tamibarotene (Am80, retinobenzoic acid, AMNOID, TAMIBARO), CH55, ITYA (ITYA-01115), Am580, BD4, or NRX195183 (also referred to as AGN195183), or their functional equivalents, analogs, or derivatives, or a combination thereof.
  • the retinoid agonist is a RARa-specific agonist.
  • the retinoid agonist is Am80, or a functional equivalent, analog, derivative or salt of Am80.
  • the G-CSF or the analog thereof can be from any source, e.g., rat, mouse, guinea pig, dog, cat, rabbit, pig, cow, horse, goat, donkey or human.
  • examples of the G-CSF or the analog thereof include but are not limited to a wild type G-CSF, a recombinant G-CSF, a G-CSF monomer or dimer, a recombinant human G-CSF (rhG-CSF) dimer, a G-CSF mutant, a G-CSF fusion protein, a G- CSF fragment, a modified G-CSF polypeptide, a PEGylated G-CSF, a glycosylated G-CSF, and a G-CSF modified with Y-shaped branched polyethylene glycol (YPEG-G-CSF) at a specific lysine (Lys 17).
  • YPEG-G-CSF Y-shaped branched polyethylene glyco
  • G-CSFs and G-CSF equivalents, analogs, derivatives, variants or fragments are functional molecules that possess a biological activity substantially similar to or even better than wild type G-CSFs. Additional information can be found in, for example, US 8557546 (Recombinant human G-CSF dimer and use thereof for the treatment of neurological diseases); US 8530417 (Y-shaped polyethylene glycol modified G-CSF, the preparation and use thereof); US 8507221 (Process for the expression of peptides of interest using GCSF as a fusion partner); US 8058398 (Modified G-CSF polypeptide); US 7655766 (Compositions comprising positional isomers of PEGylated G-CSF); and US 7402304 (Methods of using G-CSF analog compositions), which are incorporated herein by reference in their entirety as though fully set forth.
  • Typical dosages of an effective amount of the retinoid agonist or the G-CSF or the analog thereof can be in the ranges recommended by the manufacturer where known therapeutic compounds are used, and also as indicated to the skilled artisan by the in vitro responses in cells or in vivo responses in animal models. Such dosages typically can be reduced by up to about an order of magnitude in concentration or amount without losing relevant biological activity.
  • the actual dosage can depend upon the judgment of the physician, the condition of the patient, and the effectiveness of the therapeutic method based, for example, on the in vitro responsiveness of relevant cultured cells or histocultured tissue sample, or the responses observed in the appropriate animal models.
  • the retinoid agonist or the G-CSF or the analog thereof may be administered once a day (SID/QD), twice a day (BID), three times a day (TID), four times a day (QID), or more, so as to administer an effective amount of the retinoid agonist and the G-CSF or the analog thereof to the subject, where the effective amount is any one or more of the doses described herein.
  • the retinoid agonist is administered at about 0.001 to 0.01 mg/kg, 0.01 to 0.1 mg/kg, 0.1 to 0.5 mg/kg, 0.5 to 5 mg/kg, 5 to 10 mg/kg, 10 to 20 mg/kg, 20 to 50 mg/kg, 50 to 100 mg/kg, 100 to 200 mg/kg, 200 to 300 mg/kg, 300 to 400 mg/kg, 400 to 500 mg/kg, 500 to 600 mg/kg, 600 to 700mg/kg, 700 to 800mg/kg, 800 to 900mg/kg, or 900 to 1000 mg/kg.
  • the retinoid agonist is administered 1-3 times per day or 1-7 times per week.
  • the retinoid agonist is administered for about 1-10 days, 10-20 days, 20-30 days, 30-40 days, 40-50 days, 50-60 days, 60-70 days, 70-80 days, 80-90 days, 90-100 days, 1-6 months, 6-12 months, or 1-5 years.
  • the retinoid agonist is Am80, or a functional equivalent, analog, derivative or salt of Am80.
  • “mg/kg” refers to mg per kg body weight of the subject.
  • the retinoid agonist is administered to a human.
  • the effective amount of the retinoid agonist is any one or more of about 0.01 to 0.05 ⁇ g/kg/day, 0.05-0. ⁇ g/kg/day, 0.1 to 0 ⁇ g/kg/day, 0.5 to 5 ⁇ g/kg/day, 5 to 10 ⁇ g/kg/day, 10 to 20 ⁇ g/kg/day, 20 to 50 ⁇ g/kg/day, 50 to 100 ⁇ g/kg/day, 100 to 150 ⁇ g/kg/day, 150 to 200 ⁇ g/kg/day, 200 to 250 ⁇ g/kg/day, 250 to 300 ⁇ g/kg/day, 300 to 350 ⁇ g/kg/day, 350 to 400 ⁇ g/kg/day, 400 to 500 ⁇ g/kg/day, 500 to 600 ⁇ g/kg/day, 600 to 700 ⁇ g/kg/day, 700 to 800 ⁇ g/kg/day, 800 to 900 ⁇ g/kg/day, 900 to 1000 ⁇ g/kg/day, 0.01 to 0.05
  • lmg/kg/day 0.1 to 0.5mg/kg/day, 0.5 to 1 mg/kg/day, 1 to 5 mg/kg/day, 5 to 10 mg/kg/day, 10 to 15 mg/kg/day, 15 to 20 mg/kg/day, 20 to 50 mg/kg/day, 50 to 100 mg/kg/day, 100 to 200 mg/kg/day, 200 to 300 mg/kg/day, 300 to 400 mg/kg/day, 400 to 500 mg/kg/day, 500 to 600 mg/kg/day, 600 to 700mg/kg/day, 700 to 800mg/kg/day, 800 to 900mg/kg/day, 900 to 1000 mg/kg/day or a combination thereof.
  • ' ⁇ g/kg/day" or "mg/kg/day” refers to ⁇ g or mg per kg body weight of the subject per day.
  • the G-CSF or the analog thereof is administered at about 0.01 to 0.1 mcg/kg, 0.1 to 0.5 mcg/kg, 0.5 to 5 mcg/kg, 5 to 10 mcg/kg, 10 to 20 mcg/kg, 20 to 50 mcg/kg, 50 to 100 mcg/kg, 100 to 200 mcg/kg, 200 to 300 mcg/kg, 300 to 400 mcg/kg, 400 to 500 mcg/kg, 500 to 600 mcg/kg, 600 to 700mcg/kg, 700 to 800mcg/kg, 800 to 900mcg/kg, or 900 to 1000 mcg/kg.
  • the G-CSF or the analog thereof is administered 1-3 times per day or 1-7 times per week. Still in some embodiments, the G- CSF or the analog thereof is administered for about 1-10 days, 10-20 days, 20-30 days, 30-40 days, 40-50 days, 50-60 days, 60-70 days, 70-80 days, 80-90 days, 90-100 days, 1-6 months, 6-12 months, or 1-5 years.
  • mcg/kg refers to meg per kg body weight of the subject.
  • the G-CSF or the analog thereof is administered to a human. [0070]
  • regimen doses may be converted between mouse and human. Table 1 provides an exemplar chart of conversions.
  • the patients receive 6 to 9 mg/m 2 Am80 daily (oral) for maximum 56 days without interval.
  • neutropenia congenital, idiopathic or cyclic
  • chemotherapy-induced neutropenia receive 4-69 mcg/kg/day (7-20days) G-CSF via subcutaneous injection.
  • the retinoid agonist and the G-CSF or the analog thereof may be administered at neutrophil-decrease stage of a condition (i.e., when the subject has not developed the condition but is likely to or in the process to develop the condition).
  • the retinoid agonist and the G-CSF or the analog thereof may be administered at the treatment stage of a condition (i.e., when the subject has already developed the condition).
  • the target condition is neutropenia and the subject is a cancer patient who has been prescribed with a chemotherapy or radiotherapy.
  • the patient may be treated with the methods described herein when the patient has not yet developed neutropenia, or is in the process of developing neutropenia, or has already developed neutropenia.
  • the retinoid agonist and the G-CSF or the analog thereof may be administered using the appropriate modes of administration, for instance, the modes of administration recommended by the manufacturer for each of the retinoid agonist and the G-CSF or the analog thereof.
  • various routes may be utilized to administer the retinoid agonist and the G-CSF or the analog thereof of the claimed methods, including but not limited to aerosol, nasal, oral, transmucosal, transdermal, parenteral, implantable pump, continuous infusion, topical application, capsules and/or injections.
  • the retinoid agonist is administered intravenously, intramuscularly, subcutaneously, intraperitoneally, orally or via inhalation.
  • the G-CSF or the analog thereof is administered intravenously, intramuscularly, subcutaneously, intraperitoneally, orally or via inhalation.
  • the retinoid agonist and the G-CSF or the analog thereof may be administered via the same or separate routes.
  • the retinoid agonist and the G-CSF or the analog thereof are administered concurrently or sequentially.
  • the retinoid agonist is administered before, during or after administering the G-CSF or the analog thereof.
  • the retinoid agonist and/or the G-CSF or the analog thereof are administered with food or without food.
  • the retinoid agonist e.g., Am80
  • the G-CSF or the analog thereof may be administered, for example, daily, weekly, biweekly, every fortnight and/or monthly at the aforementioned dosages.
  • the retinoid agonist may be administered, for example, daily, weekly, biweekly, every fortnight and/or monthly, at the aforementioned dosages
  • the G-CSF or the analog thereof may be administered, for example, daily at the aforementioned dosages.
  • each of the retinoid agonist and the G-CSF or the analog thereof may be administered daily, weekly, biweekly, every fortnight and/or monthly, wherein the retinoid agnoist is administered at the aforementioned dosages on a day different than the day on which the G- CSF or the analog thereof is administered at the aforementioned dosages.
  • the method may further comprise providing and administering a chemotherapeutic to the subject.
  • the retinoid agonist, the G-CSF or the analog thereof, and the chemotherapeutic are administered concurrently or sequentially.
  • the retinoid agonist and/or the G-CSF or the analog is administered before, during or after administering the chemotherapeutic.
  • the retinoid agonist, the G-CSF or the analog thereof, and the chemotherapeutic are in one composition or separate compositions.
  • chemotherapeutic agents include but are not limited to Actinomycin, Alitretinoin, All-trans retinoic acid, Azacitidine, Azathioprine, Bevacizumab, Bexatotene, Bleomycin, Bortezomib, Carboplatin, Capecitabine, Cetuximab, Cisplatin, Chlorambucil, Cyclophosphamide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxorubicin, Epirubicin, Epothilone, Erlotinib, Etoposide, Fluorouracil, Gefitinib, Gemcitabine, Hydroxyurea, Idarubicin, Imatinib, Ipilimumab, Irinotecan, Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone, Ocrelizumab, Ofatumumab
  • the method may further comprise providing and administering an antimicrobial agent to the subject.
  • the retinoid agonist, the G-CSF or the analog thereof, and the antimicrobial agent are administered concurrently or sequentially.
  • the retinoid agonist and/or the G-CSF or the analog is administered before, during or after administering the antimicrobial agent.
  • the retinoid agonist, the G-CSF or the analog thereof, and the antimicrobial agent are in one composition or separate compositions.
  • the antimicrobial agent may be an antibacterial, antiviral, antifungal, or antiparasitic agent, or a combination thereof.
  • the present invention provides compositions comprising a retinoid agonist and a G-CSF or an analog thereof.
  • the retinoid agonist and G-CSF are in the same composition.
  • the retinoid againist and G- CSF are in separate compositions.
  • the compositions may be used for treating, preventing, reducing the likelihood of having, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the condition may be neutropenia or microbial infection.
  • compositions may be used for stimulating a cell (including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34+ cell) to generate granulocytes, and particularly, neutrophils.
  • a cell including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34+ cell
  • a cell including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34+ cell
  • the retinoid agonist is tamibarotene (AM 80, retinobenzoic acid, Amnoid, Tamibaro), CH55, ITYA (ITYA-01115), Am580, BD4, or NRX195183 (also referred to as AGN195183), or their functional equivalents, analogs, or derivatives, or a combination thereof.
  • the retinoid agonist is a RARa-specific agonist.
  • the retinoid agonist is Am80, or a functional equivalent, analog, derivative or salt of Am80.
  • the retinoid agonist in the composition is provided in a mg per kilogram weight of the subject; for example, about 0.001 to 0.01 mg/kg, 0.01 to 0.1 mg/kg, 0.1 to 0.5 mg/kg, 0.5 to 5 mg/kg, 5 to 10 mg/kg, 10 to 20 mg/kg, 20 to 50 mg/kg, 50 to 100 mg/kg, 100 to 200 mg/kg, 200 to 300 mg/kg, 300 to 400 mg/kg, 400 to 500 mg/kg, 500 to 600 mg/kg, 600 to 700mg/kg, 700 to 800mg/kg, 800 to 900mg/kg, or 900 to 1000 mg/kg.
  • the retinoid agonist is Am80, or a functional equivalent, analog, derivative or salt of Am80.
  • the composition is administered to a human.
  • the G-CSF or the analog thereof in the composition is provided in a meg per kg weight of the subject; for example, about 0.01 to 0.1 mcg/kg, 0.1 to 0.5 mcg/kg, 0.5 to 5 mcg/kg, 5 to 10 mcg/kg, 10 to 20 mcg/kg, 20 to 50 mcg/kg, 50 to 100 mcg/kg, 100 to 200 mcg/kg, 200 to 300 mcg/kg, 300 to 400 mcg/kg, 400 to 500 mcg/kg, 500 to 600 mcg/kg, 600 to 700mcg/kg, 700 to 800mcg/kg, 800 to 900mcg/kg, or 900 to 1000 mcg/kg.
  • the composition is administered to a human.
  • compositions further comprise a chemotherapeutic.
  • compositions furthers comprise an antimicrobial agent.
  • the compositions are formulated for intravenous, intramuscular, subcutaneous, intraperitoneal, oral or via inhalation administration.
  • the retinoid agonist and/or the G-CSF or the analog thereof useful in the treatment of disease in mammals will often be prepared substantially free of naturally-occurring immunoglobulins or other biological molecules.
  • Preferred retinoid agonists and/or G-CSFs or analogs thereof will also exhibit minimal toxicity when administered to a mammal.
  • the pharmaceutical compositions according to the invention can contain any pharmaceutically acceptable excipient.
  • “Pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use. Such excipients may be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.
  • excipients include but are not limited to starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents, wetting agents, emulsifiers, coloring agents, release agents, coating agents, sweetening agents, flavoring agents, perfuming agents, preservatives, antioxidants, plasticizers, gelling agents, thickeners, hardeners, setting agents, suspending agents, surfactants, humectants, carriers, stabilizers, and combinations thereof.
  • the pharmaceutical compositions according to the invention may be formulated for delivery via any route of administration.
  • Route of administration may refer to any administration pathway known in the art, including but not limited to aerosol, nasal, oral, transmucosal, transdermal, parenteral, enteral, topical or local.
  • Parenteral refers to a route of administration that is generally associated with injection, including intraorbital, infusion, intraarterial, intracapsular, intracardiac, intradermal, intramuscular, intraperitoneal, intrapulmonary, intraspinal, intrasternal, intrathecal, intrauterine, intravenous, subarachnoid, subcapsular, subcutaneous, transmucosal, or transtracheal.
  • the compositions may be in the form of solutions or suspensions for infusion or for injection, or as lyophilized powders. Via the parenteral route, the compositions may be in the form of solutions or suspensions for infusion or for injection.
  • the pharmaceutical compositions can be in the form of tablets, gel capsules, sugar-coated tablets, syrups, suspensions, solutions, powders, granules, emulsions, microspheres or nanospheres or lipid vesicles or polymer vesicles allowing controlled release.
  • the compositions are administered by injection. Methods for these administrations are known to one skilled in the art.
  • the composition is administered 1-3 times per day or 1-7 times per week. In various embodiments, the composition is administered for about 1-10 days, 10-20 days, 20-30 days, 30-40 days, 40-50 days, 50-60 days, 60-70 days, 70-80 days, 80-90 days, 90-100 days, 1-6 months, 6-12 months, or 1-5 years. In accordance with the invention, the composition may be formulated for intravenous, intramuscular, subcutaneous, intraperitoneal, oral or via inhalation administration.
  • the composition may be administered once a day (SID/QD), twice a day (BID), three times a day (TID), four times a day (QID), or more, so as to administer an effective amount of the retinoid agonist and the G-CSF or the analog thereof to the subject, where the effective amount is any one or more of the doses described herein.
  • the pharmaceutical compositions according to the invention can contain any pharmaceutically acceptable carrier.
  • “Pharmaceutically acceptable carrier” as used herein refers to a pharmaceutically acceptable material, composition, or vehicle that is involved in carrying or transporting a compound of interest from one tissue, organ, or portion of the body to another tissue, organ, or portion of the body.
  • the carrier may be a liquid or solid filler, diluent, excipient, solvent, or encapsulating material, or a combination thereof.
  • Each component of the carrier must be “pharmaceutically acceptable” in that it must be compatible with the other ingredients of the formulation. It must also be suitable for use in contact with any tissues or organs with which it may come in contact, meaning that it must not carry a risk of toxicity, irritation, allergic response, immunogenicity, or any other complication that excessively outweighs its therapeutic benefits.
  • compositions according to the invention can also be encapsulated, tableted or prepared in an emulsion or syrup for oral administration.
  • Pharmaceutically acceptable solid or liquid carriers may be added to enhance or stabilize the composition, or to facilitate preparation of the composition.
  • Liquid carriers include syrup, peanut oil, olive oil, glycerin, saline, alcohols and water.
  • Solid carriers include starch, lactose, calcium sulfate, dihydrate, terra alba, magnesium stearate or stearic acid, talc, pectin, acacia, agar or gelatin.
  • the carrier may also include a sustained release material such as glyceryl monostearate or glyceryl distearate, alone or with a wax.
  • the pharmaceutical preparations are made following the conventional techniques of pharmacy involving milling, mixing, granulation, and compressing, when necessary, for tablet forms; or milling, mixing and filling for hard gelatin capsule forms.
  • a liquid carrier When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion or an aqueous or non-aqueous suspension.
  • Such a liquid formulation may be administered directly p.o. or filled into a soft gelatin capsule.
  • the pharmaceutical compositions according to the invention may be delivered in a therapeutically effective amount.
  • the precise therapeutically effective amount is that amount of the composition that will yield the most effective results in terms of efficacy of treatment in a given subject. This amount will vary depending upon a variety of factors, including but not limited to the characteristics of the therapeutic compound (including activity, pharmacokinetics, pharmacodynamics, and bioavailability), the physiological condition of the subject (including age, sex, disease type and stage, general physical condition, responsiveness to a given dosage, and type of medication), the nature of the pharmaceutically acceptable carrier or carriers in the formulation, and the route of administration.
  • formulants may be added to the composition.
  • a liquid formulation may be preferred.
  • these formulants may include oils, polymers, vitamins, carbohydrates, amino acids, salts, buffers, albumin, surfactants, bulking agents or combinations thereof.
  • Carbohydrate formulants include sugar or sugar alcohols such as monosaccharides, disaccharides, or polysaccharides, or water soluble glucans.
  • the saccharides or glucans can include fructose, dextrose, lactose, glucose, mannose, sorbose, xylose, maltose, sucrose, dextran, pullulan, dextrin, alpha and beta cyclodextrin, soluble starch, hydroxethyl starch and carboxymethylcellulose, or mixtures thereof.
  • “Sugar alcohol” is defined as a C4 to C8 hydrocarbon having an -OH group and includes galactitol, inositol, mannitol, xylitol, sorbitol, glycerol, and arabitol. These sugars or sugar alcohols mentioned above may be used individually or in combination. There is no fixed limit to amount used as long as the sugar or sugar alcohol is soluble in the aqueous preparation. In one embodiment, the sugar or sugar alcohol concentration is between 1.0 w/v % and 7.0 w/v %, more preferable between 2.0 and 6.0 w/v %.
  • Amino acids formulants include levorotary (L) forms of carnitine, arginine, and betaine; however, other amino acids may be added.
  • polymers as formulants include polyvinylpyrrolidone (PVP) with an average molecular weight between 2,000 and 3,000, or polyethylene glycol (PEG) with an average molecular weight between 3,000 and 5,000.
  • PVP polyvinylpyrrolidone
  • PEG polyethylene glycol
  • a buffer in the composition it is also preferred to use a buffer in the composition to minimize pH changes in the solution before lyophilization or after reconstitution.
  • Most any physiological buffer may be used including but not limited to citrate, phosphate, succinate, and glutamate buffers or mixtures thereof.
  • the concentration is from 0.01 to 0.3 molar.
  • Surfactants that can be added to the formulation are shown in EP Nos. 270,799 and 268,110.
  • liposome Another drug delivery system for increasing circulatory half-life is the liposome.
  • Methods of preparing liposome delivery systems are discussed in Gabizon et al., Cancer Research (1982) 42:4734; Cafiso, Biochem Biophys Acta (1981) 649: 129; and Szoka, Ann Rev Biophys Eng (1980) 9:467.
  • Other drug delivery systems are known in the art and are described in, e.g., Poznansky et al., Drug Delivery Systems (R. L. Juliano, ed., Oxford, N.Y. 1980), pp. 253-315; M. L. Poznansky, Pharm Revs (1984) 36:277.
  • the liquid pharmaceutical composition may be lyophilized to prevent degradation and to preserve sterility.
  • Methods for lyophilizing liquid compositions are known to those of ordinary skill in the art.
  • the composition may be reconstituted with a sterile diluent (Ringer's solution, distilled water, or sterile saline, for example) which may include additional ingredients.
  • a sterile diluent Finger's solution, distilled water, or sterile saline, for example
  • the composition is administered to subjects using those methods that are known to those skilled in the art.
  • compositions of the invention may be sterilized by conventional, well-known sterilization techniques.
  • the resulting solutions may be packaged for use or filtered under aseptic conditions and lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • the compositions may contain pharmaceutically-acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, and stabilizers (e.g., 1-20% maltose, etc.).
  • the present invention provides a method of generating granulocytes, and in particular, neutrophils.
  • the method comprises: providing a cell (including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34 + cell); and stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof, thereby generating granulocytes, and in particular, neutrophils.
  • the method may further comprise culturing the stimulated cell and/or the generated granulocytes, and in particular, neutrophils.
  • the method further comprises isolating the stimulated cell and/or the generated granulocytes, and in particular, neutrophils. In accordance with various embodiments of the invention, the method further comprises administering the stimulated cell and/or the generated granulocytes, and in particular, neutrophils to a subject who desires a treatment of a condition. In various embodiments, the invention provides a kit of generating granulocytes, and in particular, neutrophils.
  • the kit comprises: a quantify of a retinoid agonist; a quantify of G-CSF or an analog thereof; and instructions for using the retinoid agonist and the G-CSF or the analog thereof to stimulate a cell (including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34 + cell) to generate granulocytes, and in particular, neutrophils.
  • a cell including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34 + cell
  • the invention provides a composition comprising the generated granulocytes, or in particular, neutrophils according to the described method and/or using the described kit.
  • the invention provides a composition comprising the stimulated cell according to the described method and/or using the described kit.
  • these compositions further comprise a retinoid agonist and a G-CSF or an analog thereof.
  • these compositions further comprise a pharmaceutically acceptable excipient and/or carrier.
  • these compositions may be formulated for administration to a subject via various routes including but not limited to transfusion and transplantation.
  • the invention provides a method of treating, preventing, reducing the severity of and/or slowing the progression of a condition in a subject by administering at least one of these described compositions to the subject.
  • the subject may be a patient with neutropenia, and a composition comprising the generated granulocytes, or in particular, neutrophils may be transfused into the patient.
  • the subject may be an AML patient, and a composition comprising the stimulated HSCs and/or bone marrow cells may be transplanted to the patient.
  • the present invention provides a method of treating, preventing, reducing the likelihood of having, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the method comprises: providing a cell (including but not limited to a HSC, a bone marrow granulocytic progenitor cell, and a hematopoietic CD34 + cell); stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof, thereby generating granulocytes, or in particular, neutrophils; and administering the generated granulocytes, or in particular, neutrophils to the subject, thereby treating the condition in the subject.
  • the condition is neutropenia or AML.
  • the generated granulocytes, or in particular, neutrophils are administered to the subject via transfusion or transplantation.
  • the present invention provides a method of treating, preventing, reducing the likelihood of having, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the method comprises: providing a cell (including but not limited to a HSC, a bone marrow cell, a CD34 + cell, and a stem cell); stimulating the cell with a retinoid agonist and a G-CSF or an analog thereof; and administering the stimulated cell to the subject, thereby treating the condition in the subject.
  • the condition is neutropenia or AML.
  • the stimulated cell is administered to the subject via transfusion or transplantation.
  • the present invention provides a kit for treating, preventing, reducing the severity of and/or slowing the progression of a condition in a subject.
  • the kit comprises: a quantify of a retinoid agonist; a quantify of G-CSF or an analog thereof; and instructions for using the retinoid agonist and the G-CSF or the analog thereof to treat, prevent, reduce the severity of and/or slow the progression of the condition in the subject.
  • the retinoid agonist is tamibarotene (Am80, retinobenzoic acid, Amnoid, Tamibaro), CH55, ITYA (ITYA-01115), Am580, BD4, or NRX195183 (also referred to as AGN195183), or their functional equivalents, analogs, or derivatives, or a combination thereof.
  • the retinoid agonist is a RARa-specific agonist.
  • the retinoid agonist is Am80, or a functional equivalent, analog, derivative or salt of Am80.
  • the G-CSF or the analog thereof can be from any source, e.g., rat, mouse, guinea pig, dog, cat, rabbit, pig, cow, horse, goat, donkey or human.
  • examples of the G-CSF or the analog thereof include but are not limited to a wild type G-CSF, a recombinant G-CSF, a G-CSF monomer or dimer, a recombinant human G-CSF (rhG-CSF) dimer, a G-CSF mutant, a G-CSF fusion protein, a G- CSF fragment, a modified G-CSF polypeptide, a PEGylated G-CSF, a glycosylated G-CSF, and a G-CSF modified with Y-shaped branched polyethylene glycol (YPEG-G-CSF) at a specific lysine (Lys 17).
  • YPEG-G-CSF Y-shaped branched polyethylene glyco
  • G-CSFs and G-CSF equivalents, analogs, derivatives, variants or fragments are functional molecules that possess a biological activity substantially similar to or even better than wild type G-CSFs. Additional information can be found in, for example, US 8557546 (Recombinant human G-CSF dimer and use thereof for the treatment of neurological diseases); US 8530417 (Y-shaped polyethylene glycol modified G-CSF, the preparation and use thereof); US 8507221 (Process for the expression of peptides of interest using GCSF as a fusion partner); US 8058398 (Modified G-CSF polypeptide); US 7655766 (Compositions comprising positional isomers of PEGylated G-CSF); and US 7402304 (Methods of using G-CSF analog compositions), which are incorporated herein by reference in their entirety as though fully set forth.
  • the kit is an assemblage of materials or components, including at least one of the inventive compositions.
  • the kit contains a composition including a drug delivery molecule complexed with a therapeutic agent, as described above.
  • the kit is configured particularly for the purpose of treating mammalian subjects. In another embodiment, the kit is configured particularly for the purpose of treating human subjects. In further embodiments, the kit is configured for veterinary applications, treating subjects such as, but not limited to, farm animals, domestic animals, and laboratory animals.
  • Instructions for use may be included in the kit.
  • “Instructions for use” typically include a tangible expression describing the technique to be employed in using the components of the kit to affect a desired outcome.
  • the kit also contains other useful components, such as, diluents, buffers, pharmaceutically acceptable carriers, syringes, catheters, applicators, pipetting or measuring tools, bandaging materials or other useful paraphernalia as will be readily recognized by those of skill in the art.
  • the materials or components assembled in the kit can be provided to the practitioner stored in any convenient and suitable ways that preserve their operability and utility.
  • the components can be in dissolved, dehydrated, or lyophilized form; they can be provided at room, refrigerated or frozen temperatures.
  • the components are typically contained in suitable packaging material(s).
  • packaging material refers to one or more physical structures used to house the contents of the kit, such as inventive compositions and the like.
  • the packaging material is constructed by well-known methods, preferably to provide a sterile, contaminant-free environment.
  • the term "package” refers to a suitable solid matrix or material such as glass, plastic, paper, foil, and the like, capable of holding the individual kit components.
  • a package can be a glass vial used to contain suitable quantities of a composition as described herein.
  • the packaging material generally has an external label which indicates the contents and/or purpose of the kit and/or its components.
  • the subjects in need of the methods, compositions, and kits described herein are subjects with decreased white blood cell production resulting from, including but not limited to, medication that affects the bone marrow (such as cancer drugs, antipsychotic drugs, anticonvulsant drugs), hereditary and/or congenital disorders that affect the bone marrow, patients undergoing radiation therapy, vitamin B12 deficiency, folic acid deficiency or a combination thereof.
  • medication that affects the bone marrow such as cancer drugs, antipsychotic drugs, anticonvulsant drugs
  • hereditary and/or congenital disorders that affect the bone marrow
  • patients undergoing radiation therapy vitamin B12 deficiency
  • folic acid deficiency or a combination thereof.
  • the subjects in need of the methods, compositions, and kits described herein are subjects with damaged, destroyed and/or reduced amounts of white blood cells due to, including but not limited to, acute bacterial infections, autoimmune disorders (such as systemic lupus erythematosus), use of sulfonamide medications, or a combination thereof.
  • the subjects in need of the methods, compositions, and kits described herein are subjects undergoing sequestration and/or migration of white blood cells (such as neutrophils) due to, including but not limited to, hemodialysis, malaria, bacterial infections or a combination thereof.
  • white blood cells such as neutrophils
  • the methods, compositions, and kits described herein may be used in conjunction with other therapies including but not limited to chemotherapy and/or radiation therapy.
  • Chemotherapy and/or radiation therapy often reduce the number of white blood cells, resulting in neutropenia.
  • Applying the methods, compositions, and kits of the invention concurrently or sequentially with the chemotherapy and/or radiation therapy may prevent, inhibit and/or reduce the severity of neutropenia.
  • applying the methods, compositions, and kits of the invention concurrently or sequentially with anticonvulsant and/or antipsychotic drugs may prevent, inhibit and/or reduce the severity of neutropenia resulting from the use of said drugs.
  • applying the methods, compositions, and kits of the invention concurrently or sequentially with therapeutic agents used to treat microbial infections (e.g., bacterial, fungal, viral and parasitic infections) and/or autoimmune diseases or radiation-induced neutropenia may prevent, inhibit and/or reduce the severity of neutropenia that may occur due to microbial infections and/or autoimmune diseases and/or due to the therapeutic agents that may be used to treat microbial infections and/or autoimmune diseases.
  • microbial infections e.g., bacterial, fungal, viral and parasitic infections
  • autoimmune diseases or radiation-induced neutropenia may prevent, inhibit and/or reduce the severity of neutropenia that may occur due to microbial infections and/or autoimmune diseases and/or due to the therapeutic agents that may be used to treat microbial infections and/or autoimmune diseases.
  • ROS production bioassays may be used to assess efficacy of combination treatment methods described herein, based on neutrophil microbicidal function against both bacterial and fungal infections.
  • Current clinical endpoints for neutropenia therapy include neutrophil counts and the incidence of febrile neutropenia, neither of which provide information on the neutrophils' ability to fight microorganisms.
  • the ROS production bioassay permits monitoring of neutrophil microbicidal activity based on reference values of neutrophil ROS production in response to either bacterial or fungal stimuli.
  • Such established baseline values of neutrophil ROS production deliver a rapid functional clinical endpoint critical to identifying the optimal dosing and schedule of Am80 as an add-on therapy to GCSF.
  • the methods include providing a sample from a subject, wherein the subject has been administered an effective a retinoid agonist and an effective amount of G-CSF, assaying the levels production of reactive oxygen species (ROS) and determining that the treatment is efficacious if the ROS production is higher than that of a reference sample or determining that the treatment is not efficacious if the ROS production is same as the reference sample or lower relative to the reference sample.
  • ROS reactive oxygen species
  • the subject in whom the efficacy is to be evaluated is undergoing chemotherapy treatment or has undergone chemotherapy treatment (with or without GCSF. In some embodiments, the subject in whom the efficacy is to be evaluated is undergoing treatment for neutropenia or has undergone treatment of neutropenia.
  • the sample is blood, peripheral blood, bone marrow cells, plasma, tissue or a combination thereof. In various embodiments, the sample is obtained before, during or after treatment for neutropenia.
  • ROS production may be determined as described in Babior, Blood 1999, vol. 93, page: 1464; Dahlgren et al, J Immunol Methods, 1999, Vol.
  • ROS production is measured using stimulators that mimic bacterial and fungal infections, for example fMLP, PMA, ZAS or a combination thereof.
  • the reference value is based on the amount/level of production of reactive oxygen species (ROS).
  • ROS reactive oxygen species
  • the reference value is based on the ROS production in bone marrow cells from healthy donors.
  • the reference value is based on the ROS production in peripheral blood mononuclear cell from healthy donors.
  • the reference value is based on the ROS production in healthy bone marrow cells from subjects that have or had acute myeloid leukemia.
  • the reference value is based on the ROS production in healthy peripheral blood cells from subjects that have or had acute myeloid leukemia. (What other cell types?
  • the reference value is the amount/level of ROS production in a sample obtained from the subject from a different (for example, an earlier) time point, such as during diagnosis of neutropenia, before treatment of neturopenia, after treatment for neutropenia or a combination thereof.
  • the amount/level of ROS production in the subject (for example, the subject that is being treated for neutropenia) compared to the reference value is increased by at least or about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100%.
  • the amount/level of ROS production in the subject is increased by at least or about 1-fold, 2-fold, 3-fold, 4-fold, 5-fold, 10-fold, 15-fold, 20-fold, 25-fold, 30- fold, 35-fold, 40-fold, 45-fold, 50-fold, 55-fold, 60-fold, 65-fold, 70-fold, 75-fold, 80-fold, 85-fold, 90-fold, 95-fold, 100-fold or a combination thereof.
  • Example 1 Myelopoietic expansion vs. myeloipoietic differentiation induced by selective activity of Am80
  • Such pan-action of RA exerts potent effects on inhibiting proliferation while promoting differentiation by activating RARa, - ⁇ , - ⁇ subtypes in various normal and tumor tissues.
  • Am80 is a retinoid agonist designed to selectively binding RARa but not RARy thus avoiding the side effects induced by RA-activated RARy in the treatment of APL patients.
  • Am80 promotes a modest myeloid expansion while significantly enhancing differentiation of neutrophils against bacterial infection, which is in contrast to G-CSF that profoundly enhances myeloid expansion while failing to induce effective neutrophil differentiation.
  • RARa is a substrate for cyclin-dependent kinase-activating kinase (CAK) complexes consisting of CDK7, cyclin H, and MAT1 proteins.
  • CAK exists in cells either as a free CAK to regulate cell cycle progression by phosphorylation-activation of different CDKs, or as a kinase subunit of the TFIIH complex where it mediates transcription through phosphorylation of the RNA polymerase II C-terminal domain (RNA PII).
  • RNA PII RNA polymerase II C-terminal domain
  • Am80-induced myelopoietic expansion underlies an altered transcription expression pattern of RA target genes, as shown by decreased levels of cell cycle inhibitor p21 Cip/Kip but increased expression of neutrophil effector CD 18, secondary granule LL-37, and the myeloid-specific transcription factor CCAAT/ enhancer binding protein- epsilon (C/ ⁇ ).
  • This Am80-induced selective expression of RA target genes is distinct from RA stimuli and is associated with effective development of neutrophil bactericidal functions, significant MAT1 fragmentation, decreased RARa phosphorylation, and markedly higher production of reactive oxygen species (ROS) in either normal or malignant hematopoietic precursors, as compared to G-CSF or RA.
  • ROS reactive oxygen species
  • cancer chemotherapy may be used to induce neutropenia.
  • the median duration of severe neutropenia induced by cancer chemotherapy is 3-6 days in human (ANC ⁇ 0.5 x 10 9 /L) and 2-3 days in mouse, whereas GCSF can shorten neutropenic duration in both human and mouse by increasing number of neutrophils (Fig. 4).
  • Example 4 Human equivalent low and medium doses of Am80 and Am80-GCSF combination effectively promote recovery of neutrophils at neutrophil-decrease stage
  • Example 5 Neutrophils induced by medium dose of Am80 in neutropenic mice at neutrophil- decrease stage display greater bactericidal activity than those induced by G-CSF or Am80- GCSF
  • FIG. 6B Vetscan analysis of WBC and neutrophils in PB. Except blank mice, neutrophil induction was not observed in all other groups (Fig. 6C-E). Bactericidal activities of neutrophils were determined 3 hr (Fig. 6C) and 16 hrs post-infection (Fig. 6D) in PB as well as in heart (panel E), by using blood agar analysis of viable extracellular bacteria. Because neutrophil induction was not instigated by bacterial infection in GCSF or Am80 or Am80-GCSF mice, it indicates that significantly increased neutrophil bactericidal activity in Am80 mice results from Am80-promoted differentiation of existing granulocytic precursors into mature neutrophils at the earlier developmental stage of moue neutropenia. *: P ⁇ 0.05; **: P ⁇ 0.01; ***: P ⁇ 0.005. A+G: Am80-GCSF.
  • neutrophil induction was not observed in all other groups (Figur 7C- E). Bactericidal activities of neutrophils were determined 3 hr (panel C) and 16 hrs postinfection (panel D) in PB as well as in spleen (panel E), using blood agar analysis of viable extracellular bacteria. Because neutrophil induction was not instigated by bacterial infection in GCSF or Am80 or Am80-GCSF mice, it indicates that significantly increased neutrophil bactericidal activity in Am80 mice results from Am80-promoted differentiation of existing granulocytic precursors into mature neutrophils at the earlier developmental stage of moue neutropenia. *: P ⁇ 0.05; **: P ⁇ 0.01; P ⁇ 0.005. Neutrophils induced by a lower dose of Am80 in neutropenic mice at the prevention stage display a greater bactericidal activity than those induced by G-CSF or Am80+GCSF.
  • Example 7 Use of lower dose with addition of Am80-GCSF group at neutrophil-recovery stage
  • PB peripheral blood
  • Example 8 Use of lower dose of Am80-GCSF combination significantly reduces infection- related mortality of neutropenic mice enduring continual bacteremia
  • Example 10 Regimen effects in leukemic cells and CD34+ cells
  • Am80-GCSF inhibits NB4 cell proliferation (Fig. 11 A) while significantly promoting granulocytic differentiation (Fig. 1 IB) and ROS production at day 6 in the presence of either Formyl-Methionyl-Leucyl-Phenylalanine (f-Met-Leu-Phe; fMLP) (Fig. 11C) or in the presence of PMA (Fig. 11D). *, P ⁇ 0.04 at least. G-CSF fails to induce ROS production in NB4 cells in the presence of either fMLP or PMA.
  • Am80-GCSF induces myelopoietic expansion similar to GCSF while promoting significantly greater neutrophil differentiation than Am80 or GCSF (Fig. 12A, 12B) and significantly higher ROS production at day 5 in the presence of either fMLP (Fig. 12C) or in the presence of PMA (Fig. 12D) compared to Am80 or GCSF alone. *, P ⁇ 0.02 at least.
  • Example 11 AML patient blasts following Am80 or GCSF or ATRA treatment
  • FIG. 13 A Proliferation of PB primary blasts from acute myeloid leukemia patients was assayed (Fig. 13 A) and quantified P ⁇ 0.05 (Fig. 13B). Granulocytic morphology of AML PB primary blasts was analyzed (Fig. 13C) and quantified; Am80 vs. G-CSF, P ⁇ 4.0E-8; Am80 vs. control, P ⁇ 6.02E-7 (Fig. 13D).
  • Example 12 Am80-GCSF promotes ROS production in peripheral blood mononuclear cells and in bone marrow (BM) cells
  • BM Bone marrow
  • PB peripheral blood
  • AML acute myeloid leukemia
  • ROS production was assayed in the presence of either fMLP (Fig. 15A(ii), 15B(ii) and 15C(ii)) or PMA (Fig. 15A(iii), 15B(iii) and 15C(iii)).
  • Am80-GCSF promotes significantly greater ROS production compared to Am80 or GCSF alone

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Dermatology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Wood Science & Technology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Analytical Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
PCT/US2015/016447 2014-02-18 2015-02-18 Compositions and methods for treating neutropenia WO2015126989A1 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
KR1020167025068A KR20160113302A (ko) 2014-02-18 2015-02-18 호중구감소증을 치료하기 위한 조성물 및 방법
AU2015219038A AU2015219038B2 (en) 2014-02-18 2015-02-18 Compositions and methods for treating neutropenia
JP2016552579A JP2017508737A (ja) 2014-02-18 2015-02-18 好中球減少症の治療用の組成物及び方法
US15/119,339 US10286039B2 (en) 2014-02-18 2015-02-18 Compositions and methods for treating neutropenia
CN201580009151.6A CN106413701A (zh) 2014-02-18 2015-02-18 用于治疗中性粒细胞减少症的组合物和方法
RU2016136833A RU2016136833A (ru) 2014-02-18 2015-02-18 Композиции и способы для лечения нейтропении
EP15751576.8A EP3107533A4 (en) 2014-02-18 2015-02-18 Compositions and methods for treating neutropenia
MX2016010699A MX2016010699A (es) 2014-02-18 2015-02-18 Composiciones y metodos para tratar neutropenia.
CA2937340A CA2937340C (en) 2014-02-18 2015-02-18 Compositions and methods for treating neutropenia
ZA2016/05109A ZA201605109B (en) 2014-02-18 2016-07-21 Compositions and methods for treating neutropenia

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201461941233P 2014-02-18 2014-02-18
US61/941,233 2014-02-18

Publications (1)

Publication Number Publication Date
WO2015126989A1 true WO2015126989A1 (en) 2015-08-27

Family

ID=53878925

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/016447 WO2015126989A1 (en) 2014-02-18 2015-02-18 Compositions and methods for treating neutropenia

Country Status (11)

Country Link
US (1) US10286039B2 (US20070167479A1-20070719-C00034.png)
EP (1) EP3107533A4 (US20070167479A1-20070719-C00034.png)
JP (1) JP2017508737A (US20070167479A1-20070719-C00034.png)
KR (1) KR20160113302A (US20070167479A1-20070719-C00034.png)
CN (1) CN106413701A (US20070167479A1-20070719-C00034.png)
AU (1) AU2015219038B2 (US20070167479A1-20070719-C00034.png)
CA (1) CA2937340C (US20070167479A1-20070719-C00034.png)
MX (1) MX2016010699A (US20070167479A1-20070719-C00034.png)
RU (1) RU2016136833A (US20070167479A1-20070719-C00034.png)
WO (1) WO2015126989A1 (US20070167479A1-20070719-C00034.png)
ZA (1) ZA201605109B (US20070167479A1-20070719-C00034.png)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10286039B2 (en) 2014-02-18 2019-05-14 Children's Hospital Los Angeles Compositions and methods for treating neutropenia
US11116738B2 (en) 2012-06-07 2021-09-14 Children's Hospital Los Angeles Methods for treating neutropenia using retinoid agonists

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107130026B (zh) * 2017-05-15 2018-03-09 新疆医科大学第四附属医院 一种结直肠癌诊断和治疗效果评价相关的基因
EP3697404A4 (en) * 2017-10-18 2021-07-21 Versitech Limited COMPOSITIONS AND METHODS FOR BROAD SPECTRUM ANTIVIRAL THERAPY
CN112390731B (zh) * 2020-11-16 2022-11-25 成都大学 一种具有多靶点的维甲酸类衍生物及其制备方法与应用

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120201884A1 (en) * 2009-10-19 2012-08-09 LAlLA NUTRACEUTICALS Extracts, fractions and compositions comprising acetogenins and their applications
US20130165637A1 (en) * 2010-08-31 2013-06-27 Generon (Shanghai) Corporation Ltd. Use of g-csf dimer in the treatment of neutropenia
WO2013185105A1 (en) * 2012-06-07 2013-12-12 Children's Hospital Los Angeles Methods for treating neutropenia using retinoid agonists
US20140037600A1 (en) * 2011-02-08 2014-02-06 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5691196A (en) 1992-12-31 1997-11-25 American Cyanamid Company Recombinant nucleic acid containing a response element
US5965606A (en) 1995-12-29 1999-10-12 Allergan Sales, Inc. Methods of treatment with compounds having RAR.sub.α receptor specific or selective activity
EP0957918A1 (en) 1996-12-19 1999-11-24 American Cyanamid Company Method of treating or inhibiting neutropenia
WO2002020767A2 (en) 2000-09-08 2002-03-14 Massachusetts Institute Of Technology G-csf analog compositions and methods
US7119061B2 (en) 2002-11-18 2006-10-10 Vicuron Pharmaceuticals, Inc. Dalbavancin compositions for treatment of bacterial infections
AU2003902308A0 (en) 2003-05-14 2003-05-29 Diagnose It Pty Ltd A method and system for the monitoring of medical conditions
AU2004238146A1 (en) 2003-05-16 2004-11-25 Kyowa Hakko Kogyo Co., Ltd. Preventive and/or remedy for diseases accompanied by tissue destruction
EP1621191A1 (en) 2004-07-29 2006-02-01 Werner Bollag Treatment of inflammatory diseases by RXR Antagonists
JP2008509928A (ja) 2004-08-13 2008-04-03 アノーメド インコーポレイテッド 前駆/幹細胞を動員するためのケモカインの組み合わせ
MX2007006540A (es) 2004-12-02 2008-01-31 Venus Remedies Ltd Composiciones para combatir la resistencia a antibioticos mediada por beta-lactamasa usando inhibidores de beta-lactamasa utiles para inyeccion.
US20090111786A1 (en) 2004-12-03 2009-04-30 Glass Christopher K Compounds that Prevent Macrophage Apoptosis and Uses Thereof
CA2651487A1 (en) 2006-05-16 2007-11-29 Vitae Pharmaceuticals, Inc. Methods for treating chemotherapy and radiation therapy side effects
NZ580209A (en) 2007-03-30 2011-11-25 Tmrc Co Ltd Tamibarotene capsule preparation
WO2009070844A1 (en) 2007-12-06 2009-06-11 Csl Limited Method of inhibition of leukemic stem cells
EP2334814A4 (en) * 2008-09-05 2012-04-25 Solulink Biosciences Inc METHODS AND COMPOSITIONS FOR DIRECT DETECTION OF DNA INJURIES
RU2446173C1 (ru) 2010-08-13 2012-03-27 Зао "Биокад" Новый функционально активный, высокоочищенный стабильный конъюгат гранулоцитарного колониестимулирующего фактора (г-ксф) с полиэтиленгликолем с пролонгированным биологическим действием, пригодный для медицинского применения, и иммунобиологическое средство на его основе
WO2013169864A2 (en) 2012-05-08 2013-11-14 Lycera Corporation TETRAHYDRO[1,8]NAPHTHYRIDINE SULFONAMIDE AND RELATED COMPOUNDS FOR USE AS AGONISTS OF RORƴ AND THE TREATMENT OF DISEASE
WO2013182652A1 (en) 2012-06-06 2013-12-12 Fondazione Telethon Allosteric chaperones and uses thereof
DK3017533T3 (da) 2013-07-04 2020-12-07 Linak As Aktuatorsystem
US10286039B2 (en) 2014-02-18 2019-05-14 Children's Hospital Los Angeles Compositions and methods for treating neutropenia

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120201884A1 (en) * 2009-10-19 2012-08-09 LAlLA NUTRACEUTICALS Extracts, fractions and compositions comprising acetogenins and their applications
US20130165637A1 (en) * 2010-08-31 2013-06-27 Generon (Shanghai) Corporation Ltd. Use of g-csf dimer in the treatment of neutropenia
US20140037600A1 (en) * 2011-02-08 2014-02-06 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
WO2013185105A1 (en) * 2012-06-07 2013-12-12 Children's Hospital Los Angeles Methods for treating neutropenia using retinoid agonists

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP3107533A4 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11116738B2 (en) 2012-06-07 2021-09-14 Children's Hospital Los Angeles Methods for treating neutropenia using retinoid agonists
US10286039B2 (en) 2014-02-18 2019-05-14 Children's Hospital Los Angeles Compositions and methods for treating neutropenia

Also Published As

Publication number Publication date
RU2016136833A3 (US20070167479A1-20070719-C00034.png) 2018-10-19
EP3107533A1 (en) 2016-12-28
RU2016136833A (ru) 2018-03-22
AU2015219038A1 (en) 2016-08-04
JP2017508737A (ja) 2017-03-30
MX2016010699A (es) 2017-10-11
CN106413701A (zh) 2017-02-15
CA2937340A1 (en) 2015-08-27
US20170007672A1 (en) 2017-01-12
CA2937340C (en) 2022-06-14
KR20160113302A (ko) 2016-09-28
AU2015219038B2 (en) 2018-10-18
US10286039B2 (en) 2019-05-14
ZA201605109B (en) 2017-09-27
EP3107533A4 (en) 2017-10-18

Similar Documents

Publication Publication Date Title
JP7328187B2 (ja) がん及び感染症の治療方法並びに治療用組成物
US10286039B2 (en) Compositions and methods for treating neutropenia
AU2022202113A1 (en) Pharmaceutical compositions and methods
US20160263198A1 (en) Pharmaceutical compositions and methods
US11116738B2 (en) Methods for treating neutropenia using retinoid agonists
JP2019512495A (ja) 急性骨髄性白血病の治療のための併用療法
US20160082071A1 (en) Methods of treating myeloid leukemia
JP2016536300A (ja) Flt3変異を有する急性骨髄性白血病の治療方法
KR20230003314A (ko) 암 치료를 위한 제약 조성물 및 방법
US7632802B2 (en) S100 protein as neutrophil activator for alleviating neutropenia in cancer treatment
WO2013057592A2 (en) Treatment of sickle cell disease
WO2018017571A1 (en) Uses of il-12 as a replacement immunotherapeutic
US10646552B2 (en) Pharmaceutical compositions and methods
WO2023274105A1 (zh) 一种针对阿霉素心脏及系统毒性解毒的仿生纳米保护剂制备方法及应用
JP4460220B2 (ja) オリゴヌクレオチド組成物、および細胞の分化を誘導するためのオリゴヌクレオチド組成物の使用
KR20220082862A (ko) 혈액 악성 종양을 치료하기 위한 병용 요법
Schuster et al. Phase I trial of CG53135-05 to prevent mucositis in patients undergoing high-dose chemotherapy (HDCT) and autologous peripheral blood stem cell transplantation (PBSCT)
US20150031617A1 (en) Use of thymosin alpha for treatment of purulent rhinosinusitis
Hasegawa et al. Preemptive therapy with ganciclovir and short-course intravenous immunoglobulin does not prevent recurrent cytomegalovirus infection (CBMTG-102 study)

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: 15751576

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2937340

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015219038

Country of ref document: AU

Date of ref document: 20150218

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: A201608340

Country of ref document: UA

WWE Wipo information: entry into national phase

Ref document number: 15119339

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2016552579

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/010699

Country of ref document: MX

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: 112016018938

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20167025068

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2015751576

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015751576

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016136833

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016018938

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160816