WO2018085574A2 - Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof - Google Patents

Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof Download PDF

Info

Publication number
WO2018085574A2
WO2018085574A2 PCT/US2017/059770 US2017059770W WO2018085574A2 WO 2018085574 A2 WO2018085574 A2 WO 2018085574A2 US 2017059770 W US2017059770 W US 2017059770W WO 2018085574 A2 WO2018085574 A2 WO 2018085574A2
Authority
WO
WIPO (PCT)
Prior art keywords
agent
vla
interacts
inhibitor
chemokine
Prior art date
Application number
PCT/US2017/059770
Other languages
French (fr)
Other versions
WO2018085574A3 (en
Inventor
John F. DIPERSIO
Darja KARPOVA
Original Assignee
Washington University
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 Washington University filed Critical Washington University
Publication of WO2018085574A2 publication Critical patent/WO2018085574A2/en
Publication of WO2018085574A3 publication Critical patent/WO2018085574A3/en
Priority to US16/401,950 priority Critical patent/US20190328706A1/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems

Definitions

  • the present disclosure relates to the fields of pharmaceuticals, medicine and cell biology. More specifically, it relates to pharmaceutical agents such as VLA-4 inhibitor and a first and second agent which interacts with a chemokine used in combination to enhance the collection of hematopoietic stem cells.
  • Hematopoietic stem cell transplantation is the major curative therapy available for many hematological diseases including hematological cancers.
  • HSCT is used to facilitate repopulation of healthy bone marrow and immune system cells after a high-dose chemotherapy treatment for cancers including but not limited to Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia.
  • cancers including but not limited to Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia.
  • hematopoietic stem/progenitor cells are collected from the patient's blood, harvested, frozen and then stored while the patient receives high-dose chemotherapy and/or radiation therapy.
  • HSPCs hematopoietic stem/progenitor cells
  • intravenous infusion of a minimum number of 2x106 CD34+ stem cells/kg body weight l is often needed; however, a dose of 5x106 CD34+ cells/kg is considered preferable for early and long term multilineage engraftment.
  • the stem cells for hematopoietic stem cell transplants are often harvested from peripheral blood. Due to the low amount of these cells in circulating peripheral blood, the stem cells often must be stimulated to increase the quantity in the peripheral blood, a process which can take almost a week. Even then, the collection is still done over several days to achieve sufficient concentrations of the stem cells for transplantation. This greatly increases the cost of the transplant and results in a significant burden on the patient.
  • cytokines such as granulocyte- colony forming unit (G-CSF)
  • immunostimulants such as plerixafor
  • the present disclosure provides methods using a compound which includes VLA-4 antagonists in combination with a first and second agent which interacts with a chemokine (such as CXCR2 agonist and a CXCR4 inhibitor) including methods of use and methods of treatment therewith. Also, provided herein are compositions comprising these three drugs.
  • a chemokine such as CXCR2 agonist and a CXCR4 inhibitor
  • FIG. 1 depicts a graph showing CFC mobilization by firategrast.
  • B6 mice were treated with AMD3100 (5mg/kg sc), firategrast (100 mg/kg IV), and G-CSF (250 mg/kg/d ⁇ 5 days) alone or in combination.
  • FIG. 2 depicts a graph showing CFC mobilization by firategrast + Gro- ⁇ .
  • FIGS. 3A & 3B show HSPC mobilization and transplantation.
  • FIG. 3B B6 (CD45.2+) mice were treated and blood was collected 0.5 h post-injection.
  • the frequency of repopulating units (RU) was calculated based on the contribution within the B-cell lymphoid and myeloid fraction 20 weeks after transplant. * P ⁇ 0.05, ** P ⁇ 0.01 .
  • FIG. 4A shows the results of the colony forming cell (CFC) assay with the integrin antagonist administered in combination with Gropt.
  • the top graph shows the number of colonies formed as a function of time while the bottom graph shows the number of CFC units at a specific time point for each mouse. These results show the results for Example Compound 35 (light gray) and Example Compound 36 (dark gray).
  • FIG. 4B shows the results of the colony forming cell (CFC) assay with only the VLA-4 inhibitor.
  • the top graph shows the number of colonies formed as a function of time while the bottom graph shows the number of CFC units at a specific time point for each mouse. These results show the results for Example Compound 35 (light gray) and Example Compound 36 (dark gray).
  • FIG. 7A shows the study design.
  • FIGS. 8A-8C show the results of order of administration of the agents.
  • FIG. 8A shows that similar results were obtained when the VLA-4 inhibitor was administered before tGro- ⁇ . Similar results were obtained for the opposite combination wherein tGro- ⁇ was administered before the VLA-4 inhibitor (FIG. 8B).
  • *** P ⁇ 0.001 The highest CFU-C was seen when the agents were administered simultaneously as shown in FIG. 8C.
  • *** P ⁇ 0.001 ** P ⁇ 0.001 t-test with Holm-Sidak correlation, n 4-5 and conducted in BALB/C mice.
  • FIG. 10 shows the combination of a VLA-4 inhibitor and tGro- ⁇ results in similar mobilization regardless of the presence of other factors which result in poor stem cell mobilization such as diabetes.
  • FIG. 11 A-11 B graphically depicts the effects of the triple
  • FIG. 12A-12E shows independence of VLA4 and CXCR4 blockade induced mobilization.
  • FIG. 12A-12B graphically depicts the CFU of vehicle treated and anti-vla-4 treated CXCR4 +/+ and CXCR4 -/- mice.
  • FIG. 12C shows the CFU in wt or cxcr4 f/f mice treated with AMD3100, GroB, CWHM-823 or control.
  • FIG. 12D-12E show LSK/CFU-C per mL of peripheral blood.
  • FIG. 13A-13C show the effects of the triple combination of combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist.
  • FIG. 13A shows increased CFU-C per mL
  • FIG. 13B shows increased WBC per mL
  • FIG. 13C shows increased NE per mL of peripheral blood in triple therapy treated mice compared to the other conditions.
  • compositions and methods comprising a first agent which interacts with a chemokine, such as a CXCR2 agonist, a second agent which interacts with a chemokine, such as, a CXCR4 inhibitor, and a compound that act as integrin antagonists or inhibitor, such as ⁇ 4 ⁇ 1 integrin (VLA-4) antagonists as well as compositions thereof.
  • a chemokine such as a CXCR2 agonist
  • a second agent which interacts with a chemokine
  • a compound that act as integrin antagonists or inhibitor such as ⁇ 4 ⁇ 1 integrin (VLA-4) antagonists
  • VLA-4 ⁇ 4 ⁇ 1 integrin
  • compositions described herein may be used to stimulate progenitor and/or stem cells and result in such stimulation in a shorter amount of time relative to either agent alone or other known agents or combinations.
  • These compositions may also have the added advantage that they result in the mobilization in higher numbers, begin mobilization in a shorter period of time, over a more prolonged period of time, or mobilize increased numbers of early progenitor and/or stem cells, LSK-SLAM cells, CFU-C cells, or other progenitor and/or stem cells which are competent to achieve a successful engraftment into the patient.
  • compositions may be used in improving the harvest of
  • hematopoietic stem cells or progenitor cells are described in more detail below.
  • the compounds used in the compositions of the present disclosure include the compounds described in the Examples and claims listed below. All the synthesis methods described above can be further modified and optimized using the principles and techniques of organic chemistry as applied by a person skilled in the art. Such principles and techniques are taught, for example, in March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure (2007), which is incorporated by reference herein.
  • Compounds employed in methods of the disclosure may contain one or more asymmetrically-substituted carbon or nitrogen atoms, and may be isolated in optically active or racemic form. Thus, all chiral, diastereomeric, racemic form, epimeric form, and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. Compounds may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. In some embodiments, a single diastereomer is obtained.
  • the chiral centers of the compounds of the present disclosure can have the S or the R configuration, as defined by the lUPAC 1974 Recommendations. For example, mixtures of stereoisomers may be separated using the techniques taught in the
  • Atoms making up the compounds of the present disclosure are intended to include all isotopic forms of such atoms.
  • Compounds of the present disclosure include those with one or more atoms that have been isotopically modified or enriched, in particular those with pharmaceutically acceptable isotopes or those useful for pharmaceutically research.
  • Isotopes include those atoms having the same atomic number but different mass numbers.
  • isotopes of hydrogen include deuterium and tritium
  • isotopes of carbon include 13 C and 14 C.
  • one or more carbon atom(s) of a compound of the present disclosure may be replaced by a silicon atom(s).
  • one or more oxygen atom(s) of a compound of the present disclosure may be replaced by a sulfur or selenium atom(s).
  • Compounds of the present disclosure may also exist in prodrug form. Since prodrugs are known to enhance numerous desirable qualities of
  • the compounds employed in some methods of the disclosure may, if desired, be delivered in prodrug form.
  • the disclosure contemplates prodrugs of compounds of the present disclosure as well as methods of delivering prodrugs.
  • Prodrugs of the compounds employed in the disclosure may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound.
  • prodrugs include, for example, compounds described herein in which a hydroxy, amino, or carboxy group is bonded to any group that, when the prodrug is administered to a subject, cleaves to form a hydroxy, amino, or carboxylic acid, respectively. Additional details regarding prodrugs may be found in Smith and Williams, 1988, the entire contents of which are hereby incorporated by reference. Smith and Williams Introduction to the Principles of Drug Design, Smith, H. J.; Wright, 2nd ed., London (1988).
  • any salt of this disclosure is not critical, so long as the salt, as a whole, is pharmacologically acceptable.
  • Compounds useful in the disclosure which are amines may be administered or prepared in the forms of their acid addition salts or metal complexes thereof.
  • Suitable acid addition salts include salts of inorganic acids that are biocompatible, including HCI, HBr, sulfuric, phosphoric and the like, as well as organic acids such as acetic, propionic, butyric and the like, as well as acids containing more than one carboxyl group, such as oxalic, glutaric, adipic and the like.
  • Compounds useful in the disclosure that are carboxylic acids or otherwise acidic may be administered or prepared in forms of salts formed from inorganic or organic bases that are physiologically compatible.
  • these compounds may be prepared in the forms of their sodium, potassium, calcium, or magnesium salts as appropriate or may be salts with organic bases such as caffeine or ethylamine.
  • organic bases such as caffeine or ethylamine.
  • These compounds also may be in the form of metal complexes. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are presented in Handbook of Pharmaceutical Salts: Properties, and Use (2002), which is incorporated herein by reference.
  • the compounds of the present disclosure include those that have been further modified to comprise substituents that are convertible to hydrogen in vivo.
  • hydrolyzable groups such as acyl groups, groups having an oxycarbonyl group, amino acid residues, peptide residues, o-nitrophenylsulfenyl, trimethylsilyl, tetrahydropyranyl, diphenylphosphinyl, and the like.
  • acyl groups include formyl, acetyl, trifluoroacetyl, and the like.
  • groups having an oxycarbonyl group include ethoxycarbonyl, fe/f-butoxycarbonyl (-C(0)OC(CH 3 ) 3 , Boc), benzyloxycarbonyl, p-methoxybenzyloxycarbonyl,
  • Suitable amino acid residues include, but are not limited to, residues of Gly (glycine), Ala (alanine), Arg (arginine), Asn (asparagine), Asp (aspartic acid), Cys (cysteine), Glu (glutamic acid), His (histidine), lie (isoleucine), Leu (leucine), Lys (lysine), Met (methionine), Phe
  • amino acid residues also include amino acid residues that are protected with a protecting group.
  • suitable protecting groups include those typically employed in peptide synthesis, including acyl groups (such as formyl and acetyl), arylm ethoxycarbonyl groups (such as benzyloxycarbonyl and p-nitrobenzyloxycarbonyl), fe/f-butoxycarbonyl groups (-C(0)OC(CH 3 ) 3 , Boc), and the like.
  • Suitable peptide residues include peptide residues comprising two to five amino acid residues. The residues of these amino acids or peptides can be present in stereochemical configurations of the D-form, the L-form or mixtures thereof. In addition, the amino acid or peptide residue may have an
  • Suitable amino acid residues having an asymmetric carbon atom include residues of Ala, Leu, Phe, Trp, Nva, Val, Met, Ser, Lys, Thr and Tyr.
  • Peptide residues having an asymmetric carbon atom include peptide residues having one or more constituent amino acid residues having an asymmetric carbon atom.
  • suitable amino acid protecting groups include those typically employed in peptide synthesis, including acyl groups (such as formyl and acetyl), arylmethoxycarbonyl groups (such as benzyloxycarbonyl and p-nitrobenzyloxycarbonyl), fe/f-butoxycarbonyl groups (-C(0)OC(CH 3 ) 3 ), and the like.
  • substituents "convertible to hydrogen in vivo" include reductively eliminable
  • hydrogenolyzable groups include, but are not limited to, arylsulfonyl groups (such as o- toluenesulfonyl); methyl groups substituted with phenyl or benzyloxy (such as benzyl, trityl and benzyloxymethyl); arylmethoxycarbonyl groups (such as benzyloxycarbonyl and o-methoxy-benzyloxycarbonyl); and haloethoxycarbonyl groups (such as ⁇ , ⁇ , ⁇ - trichloroethoxycarbonyl and ⁇ -iodoethoxycarbonyl).
  • arylsulfonyl groups such as o- toluenesulfonyl
  • methyl groups substituted with phenyl or benzyloxy such as benzyl, trityl and benzyloxymethyl
  • arylmethoxycarbonyl groups such as benzyloxycarbonyl and o-
  • Compounds of the disclosure may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. , higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the indications stated herein or otherwise.
  • a better pharmacokinetic profile e.g. , higher oral bioavailability and/or lower clearance
  • the present disclosure relates to compositions containing one or more VLA-4 inhibitors and one or more agents which interacts with a chemokine such as a CXCR2 agonist, a CXCR4 inhibitor, or G-CSF.
  • a chemokine such as a CXCR2 agonist, a CXCR4 inhibitor, or G-CSF.
  • the present disclosure relates to compositions containing one or more VLA-4 inhibitors and at least two agents which interact with a chemokine.
  • the present disclosure relates to compositions containing one or more VLA-4 inhibitors, at least one CXCR4 inhibitor and at least one CXCR2 agonist.
  • These compositions may further comprise an excipient such as solvent or diluent which renders the composition suitable for administration via injection.
  • these compositions may be formulated independently and then administered simultaneously to a patient.
  • these compositions are formulated with additional therapeutic agents or excipients.
  • these compositions consists substantially of, consists essentially of, or consists of one or more VLA-4 inhibitors, one or more agents which interact with a chemokine, and one or more excipients.
  • Each of the compositions described herein contain a pharmaceutically effective amount of each of these agents combined.
  • the compositions may contain a
  • the pharmaceutically effective combined amount results when each agent is present in an amount such that the effect of the combination results in increased activity relative to a similar amount of a single agent. In some embodiments, the effect of the combination results in an additive increase in activity. In some embodiments, the effect of the combination results in synergistic activity.
  • compositions may be used in a variety of indications such as the mobilization of hematopoietic stem cells or progenitor cells.
  • indications include elevating the number of progenitor and/or stem cells which are circulating in the patient especially elevating the number of these cells in the peripheral blood of a patient.
  • compositions may be used to treat a patient with cancer including sensitizing the patient to a chemotherapy and/or radiotherapy, for the treatment of hematopoietic cancer such as leukemias, myelomas, or lymphoma, or for the harvesting of hematopoietic progenitor and/or stem cells which may be transplanted into a patient who has impaired production of hematopoietic progenitor and/or stem cells.
  • a patient may have impaired production of hematopoietic progenitor and/or stem cells resulting from a high dose of chemotherapy, radiotherapy, another therapeutic agent, or a genetic abnormality.
  • compositions described herein may be used to mobilize pre-cancerous or cancerous cells from the bone marrow into the peripheral blood.
  • the mobilization of pre-cancerous or cancerous cells from the bone marrow is used to potentiate or increase the effects of a standard cancer therapy such as a chemotherapeutic and/or radiotherapy.
  • compositions may be used in the manufacture of
  • compositions and methods described herein may include one or more additional agents that are therapeutically or nutritionally useful such as antibiotics, vitamins, herbal extracts, anti-inflammatories, glucose, antipyretics, analgesics, cyclophosphamide, recombinant stem cell factor (Stemgen®), granulocyte- macrophage colony stimulating factor (GM-CSF) (such as Leukine®, and Leucomax®), ETRX-101 , TLK 199/TILENTRATM, lnterleukin-1 (IL-1 ), lnterleukin-3 (IL-3), lnterleukin-8 (IL-8), PIXY-321 (GM-CSF/IL-3 fusion protein), macrophage inflammatory protein, thrombopoietin, or a similar agent.
  • the compositions may contain one or more agents prevents microbial growth to increase the storage of the composition. Such agents may be an anti-parasitic, an antifungal, an antibiotic
  • compositions may further comprise one or more chemotherapeutic agents.
  • VLA-4 inhibitors may contain one or more VLA-4 inhibitors or VLA-4 antagonists.
  • VLA-4 inhibitor and VLA-4 antagonist are used interchangeably in the disclosure.
  • Some non-limiting examples of VLA-4 inhibitors which may be used in the compositions and methods described herein include antibodies, such as humanized monoclonal antibody against a4, natalizumab (Antegren®) and small molecules such as those described in U.S. Pat. No.
  • VLA-4 inhibitor An example of a VLA-4 inhibitor that may be used herein is BIO5192 (also known as AMD15057) disclosed in PCT publication WO 01/12186, which is incorporated herein by reference. Alternatively, analogs of BIO5192, such as BI0121 1 , may be used. In other embodiments, the VLA-4 inhibitor is firategrast or a harmaceutically acceptable salt thereof. Firategrast is a compound of the formula:
  • Ri is hydrogen, aminocarbonyl, carboxy, cyano, halo, hydroxy, -SF 5 , alkyl(c ⁇ 8), substituted alkyl( C ⁇ 8), alkoxy( C ⁇ 8), substituted alkoxy( C ⁇ 8), or -Y-i -R a ; wherein:
  • Yi is alkanediyl(c ⁇ 8) or substituted alkanediyl ( c ⁇ 8);
  • R a is alkoxy ( c ⁇ i2), acyloxy ( c ⁇ i2), or a substituted version of either of these groups;
  • X is a covalent bond or -0-
  • n 0 or 1 ;
  • n 1 , 2, 3, 4, 5, 6, 7, or 8;
  • R b is hydrogen, alkyl C ⁇ 8), or substituted alkyl (C ⁇ 8);
  • R 2 is hydrogen, aminocarbonyl, carboxy, cyano, halo, hydroxy, -SF 5 , alkyl ( c ⁇ 8), substituted alkyl( C ⁇ 8), alkoxy ( c ⁇ 8), substituted alkoxy ( c ⁇ 8), or -Y2 ⁇ R C ; wherein:
  • Y 2 is alkanediyl(c ⁇ 8) or substituted alkanediyl (C ⁇ 8); and R c is alkoxy (C ⁇ i2), acyloxy (C ⁇ i2), or a substituted version of either of these groups;
  • Xi is hydroxy, alkoxy ( c ⁇ 8), substituted alkoxy ( c ⁇ 8), cycloalkoxy( C ⁇ 8), substituted cycloalkoxy(c ⁇ 8), alkenyloxV(c ⁇ 8), substituted alkenyloxV(c ⁇ 8), aryloxy(c ⁇ 8), substituted aryloxy ( c ⁇ 8), aralkoxy ( c ⁇ 8), substituted aralkoxy ( c ⁇ 8), or a substituent convertible in vivo to hydrogen; and
  • R 3 and R 4 are each independently hydrogen, hydroxy, alkoxy (C ⁇ 8) or substituted alkoxy (C ⁇ 8);
  • R 5 is hydrogen, -CH(ORd)R e , or -C(0)Rf, wherein:
  • Rd is hydrogen, alkyl C ⁇ 8), or substituted alkyl (C ⁇ 8);
  • R e and R f are each independently alkyl (C ⁇ 8) or substituted alkyl (C ⁇ 8);
  • Z is a group of the formula:
  • p is O, 1 , 2, or 3;
  • R 6 is hydrogen or -C(0)X2; wherein:
  • X2 is amino, hydroxy, alkoxV(c ⁇ 8), substituted alkoxV(c ⁇ 8), cycloalkoxy(c ⁇ 8), substituted cycloalkoxy (C ⁇ 8), alkenyloxy (C ⁇ 8), substituted alkenyloxy (C ⁇ 8), aryloxy (C ⁇ 8), substituted aryloxy (C ⁇ 8), aralkyloxy (C ⁇ 8), substituted aralkyloxy( C ⁇ 8), alkylaminO( C ⁇ 8), substituted alkylaminO(c ⁇ 8), dialkylaminO( C ⁇ 8), substituted dialkylaminO( C ⁇ 8), cycloalkylaminO(c ⁇ 8), substituted cycloalkylamino ( c ⁇ 8), alkenylaminO(c ⁇ 8), substituted alkenylaminO( C ⁇ 8), arylamino ( c ⁇ 8), substituted arylaminO( C ⁇ 8),
  • R 9 is hydrogen, alkyl ( c ⁇ 8), or substituted alkyl ( c ⁇ 8); or
  • W is hydrogen, cyano, halo, hydroxy, or -C(0)X3, wherein:
  • X 3 is amino, hydroxy, alkoxy ( c ⁇ 8), substituted alkoxy ( c ⁇ 8),
  • aralkylaminO(c ⁇ 8) substituted aralkylaminO( C ⁇ 8), or a substituent convertible in vivo to hydrogen
  • R-io and Rn are each independently hydrogen or halo
  • chemokines includes chemokines, cytokines, chemokine receptors, or an agent which modulates the activity of these molecules such as a fragment, an antibody, or a small organic molecule.
  • agent which modulates the activity of these molecules such as a fragment, an antibody, or a small organic molecule.
  • interacts with means that the agent binds with a chemokine in a manner that modulates the activity of said chemokine, for example, by reducing, inhibiting, increasing or activating the activity of the chemokine.
  • the present disclosure relates to compositions which modulate the activity of a CXC chemokine receptor such as CXCR2 or CXCR4.
  • the present methods and compositions comprise at least two agents which interact with a chemokine.
  • the present methods and compositions comprise a first agent comprising aCXCR2 agonist and a second agent comprising a CXCR4 inhibitor.
  • the methods and compositions comprise a VLA-4 inhibitor or VLA-4 antagonist, a CXCR4 inhibitor and a CXCR2 agonist.
  • the methods and compositions comprise a VLA-4 inhibitor or VLA-4 antagonist, AMD3100 and Grop.
  • CXCR2 agonists include any molecule that activates the CXCR2 receptor. Such molecules include chemokines, cytokines, agonist antibodies or biologically active fragments thereof, or small organic molecules. Some non-limiting examples of chemokines acting via the CXCR2 receptor include, but are not limited to Grop, Groa, Groy, GCP-2 (granulocyte chemo-attractant protein 2), IL-8, NAP-2
  • ENA-78 epidermal-cell derived neutrophil activating protein 78
  • MGSA MGSA
  • the CXCR2 agonists used in the methods and compositions described herein are Grop and modified forms thereof.
  • a recombinant /V-terminal 4-amino acid truncated form of the human chemokine Grop also known as SB-251353 or garnocestim or Gropt
  • SB-251353 or garnocestim or Gropt a recombinant /V-terminal 4-amino acid truncated form of the human chemokine Grop
  • Chemokines such as the SB-251353, Groa, Grop, and Groy are further discussed in WO 94/29341 ; WO 97/15594; WO 97/15595; WO 99/26645; WO 02/02132; U.S. Pat. No. 6,080,398; U.S. Pat. No. 6,399,053; and U.S. Pat. No. 6,447,766, which are incorporated herein by reference.
  • the "Grop”, "Grop protein”, or “Grop chemokine” class includes Grop itself as well as modified forms of Grop. These modified forms include, but are not limited to, truncated, multimerized, amino-acid substituted, modified with amino-acid deletions and/or insertions, or combinations thereof. "Modified forms of Grop” includes truncated forms such as those described in U.S. patents 6,447,766; 6,399,053;
  • Modified forms of Grop are multimeric forms of Grop such as dimers, trimers, tetramers, or other versions containing multiple proteins or modified proteins.
  • modified forms include modified forms of Grop with truncation of between 2 to about 8 amino acids at the amino terminus of the mature protein, truncation of between about 2 to about 10 amino acids at the carboxy terminus of the mature protein, or multimeric forms of the modified and/or truncated proteins, e.g. , dimers, trimers, tetramers and other aggregated forms.
  • truncated forms of Grop may include SB-251353 which consists of amino acids 5-73 and forms thereof where amino acid 69 is deamidated.
  • Another specific CXCR2 receptor agonist that may be used in the compositions and methods described herein is SB-251353, a basic, heparin-binding protein with a molecular mass of approximately 7500 Da (King et al., J Immunol 2000; 164: 3774-3782, Hepburn et al., Journal of Pharmacology and Experimental
  • compositions and methods described herein may comprise one or more CXCR4 inhibitors.
  • CXCR4 inhibitors include AMD3100 (plerixafor), AMD3465, CTCE-0214, CTCE-9908, CP-1221 (linear peptides, cyclic peptides, natural amino-acids, unnatural amino acids, and
  • peptidomimetic compounds T140 and analogs, 4F-benzoyl-TN24003, KRH-1 120, KRH-1636, KRH-2731 , polyphemusin analogue, ALX40-4C, or a CXCR4 inhibitors described in WO 01/85196, WO 99/50461 , WO 01/94420, WO 03/090512, US
  • compositions or methods described herein may be comprise G-CSF. It is contemplated that any suitable source of G-CSF may be employed.
  • the G- CSF used in the compositions or methods may be either recombinant or purified using known techniques and includes, but is not limited to, Neupogen® filgrastim (Amgen), Neutrogin®/Granocyte® lenograstim (Chugai Pharmaceuticals), and Neulasta® pegylated filgrastim (Amgen). Additionally, biologically active fragments, variants, derivatives or fusion proteins may also be employed provided these agents retain the ability to mobilize progenitor or stem cells.
  • compositions comprising compounds described above. These compounds and pharmaceutical compositions may be used to improve the harvest of hematopoietic stem cells or progenitor cells. Additionally, the compounds or compositions may be used to elevate the circulation of hematopoietic progenitor and/or stem cells, improve the collection of hematopoietic stem cells or progenitor cells for a transfusion, increase the sensitization of an anti-cancer therapy such as a chemotherapeutic or radiotherapy, or mobilize pre-cancerous or cancerous cells into the peripheral blood which may increase their sensitivity to an anti-cancer therapy.
  • an anti-cancer therapy such as a chemotherapeutic or radiotherapy
  • Hematopoietic stem cell transplant is used to facilitate repopulation of healthy bone marrow and immune system cells after high-dose chemotherapy treatment for cancers such as Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia.
  • cancers such as Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia.
  • hematopoietic stem/progenitor cells are used to facilitate repopulation of healthy bone marrow and immune system cells after high-dose chemotherapy treatment for cancers such as Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia.
  • hematopoietic stem/progenitor cells are used to facilitate repopulation of healthy bone marrow and immune system cells after high-dose chemotherapy treatment for cancers such as Hodgkin's and non-Hodgkin's lymphoma,
  • HSPCs are collected from the patient's blood, harvested, frozen and then stored while the patient receives high-dose chemotherapy and/or radiation therapy.
  • Successful HSCT requires the intravenous infusion of a minimum number of 2 ⁇ 10 6 CD34+ stem cells/kg body weight; however, a dose of 5 ⁇ 10 6 CD34+ cells/kg is considered preferable for early and long term multilineage engraftment.
  • G-CSF granulocyte colony-stimulating factor
  • VLA-4 the integrin ⁇ 4 ⁇ 1
  • BM bone marrow
  • HSPC mobilization has been achieved by disrupting the integrin ⁇ 4 ⁇ 1 ⁇ / ⁇ -1 axis with antibodies against integrin ⁇ 4 ⁇ 1 or VCAM-1.
  • Preclinical mouse studies in the DiPersio laboratory have shown that administration of the small molecule inhibitor of integrin ⁇ 4 ⁇ 1 , BI05192, results in the rapid and reversible mobilization of HSPCs into the peripheral circulation with maximum mobilization occurring within 30 to 60 minutes and returning to baseline within 4 hours.
  • a superior treatment could be envisioned wherein a patient receives an integrin ⁇ 4 ⁇ 1 antagonist to continually inhibit integrin ⁇ 4 ⁇ 1 over the course of ⁇ 4 hours (average duration of CD34+ stem cell apheresis procedures), maximizing the mobilization of HSPCs that can be collected by apheresis during the same day of treatment.
  • BI05192 is a potent small molecule inhibitor of integrin ⁇ 4 ⁇ 1 and has demonstrated efficacy in mobilizing HSPCs in mice.
  • BI05192 has poor aqueous solubility, bioavailability, and pharmacokinetic properties and therefore has not been developed clinically.
  • a simpler, more soluble integrin ⁇ 4 ⁇ 1 antagonist is firategrast. Firategrast has been tested in clinical trials for the treatment of multiple sclerosis and has demonstrated efficacy in mobilizing HSPCs in mice but to a significantly lesser extent than BI05192 and at much higher doses.
  • development of integrin ⁇ 4 ⁇ 1 antagonist which exhibit improved properties would greatly improve the clinical use of this particular combination.
  • compositions or methods may also have the added advantage that the compositions or methods result in the mobilization in higher numbers, begin mobilization in a shorter period of time, over a more prolonged period of time, or mobilize increased numbers of early progenitor and/or stem cells, LSK-SLAM cells, CFU-C cells, or other progenitor and/or stem cells which are competent to achieve a successful engraftment into the patient.
  • the number of progenitor and/or stem cells mobilized when using the combination or methods described herein may be at least about 1 .2-fold, at least about 1 .5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7-fold, at least about 7.5-fold, at least about 8-fold, at least about 8.5-fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 1 1 -fold, at least about 12-fold, at least about 13-fold, at least about 14-fold, or at least about 15-fold greater then when using a single agent alone.
  • the number of early progenitor and/or stem cells (e.g. LSK-SLAM cells) mobilized when using the combination of at least one VLA-4 inhibitor and at least one CXCR2 agonist is at least about 1 .2-fold, at least about 1 .5- fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7-fold, at least about 7.5-fold, at least about 8-fold, at least about 8.5-fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 1 1 -fold, at least about 12-fold, at least about 13-fold, at least about 14-fold, at least about 15-fold, at least about 16-fold, at least about 17-fold, at least about 18-fold, at least about 19-fold
  • this disclosure provides methods of inhibiting or antagonizing VLA-4 using one or more of the compounds disclosed herein, as well as pharmaceutical compositions thereof containing one or more VLA-4 antagonists in the presence of two or more agents that interact with a chemokine, such as Gro- ⁇ ,
  • AMD3100 AMD3100, or a derivatives thereof.
  • the therapeutic methods described herein may be used to enhance or elevate the circulation of hematopoietic progenitor and/or stem cells. These therapeutic methods may be used to improve stem cell transplantation, tissue repair, improve the efficacy of cancer therapy, or other situations in which in vivo stimulation of hematopoiesis is desirable.
  • the compositions or methods described herein wherein the VLA-4 inhibitor and agents which interact with a chemokine combine to act synergistically to induce rapid mobilization of progenitor and stem cells.
  • peak mobilization when these combined therapeutic agents are used, may occur at about 10 minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, or about 6 hours after administration of the combination.
  • these compositions or methods result in a composition which acts synergistically to induce rapid mobilization of progenitor and stem cells with peak mobilization at about 15 minutes after administration of the combination. In contrast, this mobilization is significantly shorter than the 4-5 days needed to achieve maximum mobilization using G-CSF.
  • compositions described herein may be used to increase the harvest of HSPCs for a variety of different applications. These compositions and methods may be used to treat a patient who requires a
  • compositions and methods may be used to treat a patient who does not require a transplantation.
  • the patient who needs a transplant of HSPCs requires either an allogenic, autologous, or tandem transplant of HSPCs.
  • the HSPCs may be used in either allogenic or autologous transplants.
  • the present methods and compositions described herein may be used to improve the circulation of cells to tissues which need repair. The increased circulation of HSPCs may be used to improve the repair of the target tissue in the patient.
  • HSPCs are harvested, these cells may be returned to the donor patient (autologous transplant) or may be donated to another patient that is sufficiently compatible to prevent rejection (allogeneic transplant).
  • autologous transplantation is in combination with radiation or chemotherapy in patients bearing tumors since the radiotherapeutic or
  • chemotherapeutic methods deplete the patient's normal cells.
  • the patients cells may be harvested prior to or during the therapeutic treatments,
  • Allogeneic recipients may receive the cells for the same purpose, or may have a condition that may be benefited by enhancing their hematopoietic systems.
  • the mobilized cells are collected from the donor by, for example, apheresis and then stored/cultured/expanded/fractionated as desired.
  • the compositions or methods described herein may result in the need for aphersis being eliminated.
  • compositions and methods described herein may be used to increase the circulation of pre-cancerous or cancerous cells out of the bone marrow into the peripheral blood.
  • these methods or compositions may be used to treat patients who have or are at risk of a hematopoietic malignancy such as lymphoma, myeloma, or leukemia.
  • the compositions or methods described herein may be administered or employed prior to, during, or subsequent to the anti-cancer therapy.
  • Two non-limiting examples of anticancer therapies that may be used in the methods described herein or conjunction with the compositions described herein include chemotherapeutic agents or radiotherapy.
  • compositions and methods described herein may be used to decrease inflammation which may result in increasing tissue repair.
  • the compositions or methods described herein may be used to treat graft versus host disease. Additionally, these compositions or methods may be used to treat diseases or disorders associated with cell adhesion-mediated inflammatory pathways.
  • cell adhesion-mediated inflammatory pathologies include asthma, multiple sclerosis, rheumatoid arthritis, atherosclerosis, and
  • compositions of these combinations may further comprise one or more non-toxic, pharmaceutically acceptable carriers and/or diluents and/or adjuvants (collectively referred to herein as "carrier" materials) and if desired other active ingredients.
  • carrier non-toxic, pharmaceutically acceptable carriers and/or diluents and/or adjuvants
  • these methods may be used to treat a blood disease or disorder such as sickle cell anemia or as a part of hematopoietic stem cell therapy to promote the development of stem cells.
  • the compound is administered as part of a pharmaceutical composition further comprising a
  • the compounds and/or pharmaceutical compositions thereof may be administered orally, parenterally, or by inhalation spray, or topically in unit dosage formulations containing conventional pharmaceutically acceptable carriers, adjuvants and vehicles.
  • parenteral as used herein includes, for example, subcutaneous, intravenous, intramuscular, intrasternal, infusion techniques or intraperitoneally.
  • the compounds of the present disclosure are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended.
  • Therapeutically effective doses of the compounds required to prevent or arrest the progress of or to treat a medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
  • the compounds described above can be used in the treatment of patients suffering from the above pathological conditions.
  • One skilled in the art will recognize that selection of the most appropriate compound of the disclosure is within the ability of one with ordinary skill in the art and will depend on a variety of factors including assessment of results obtained in standard assay and animal models.
  • the compounds provided herein may be used in a variety of biological, prophylactic or therapeutic areas, including those in wherein VLA-4 plays a role.
  • the compounds in a therapeutically effective amount are ordinarily combined with one or more excipients appropriate to the indicated route of
  • the compounds of the present disclosure are contemplated to be formulated in a manner ameniable to treatment of a veterinary patient as well as a human patient.
  • the veterinary patient may be an avian such as chicken, turkey, or duck, a companion animal such as a cat or dog, livestock animals such as a cow, horse, pig, or goat, zoo animals, and wild animals
  • the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and tableted or encapsulated for convenient administration.
  • the compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers.
  • Other excipients and modes of administration are well and widely known in the pharmaceutical art and may be adapted to the type of animal being treated. Description of potential
  • compositions described herein include those taught in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Company, Easton, Pa., which is incorporated herein by reference.
  • compositions useful in the present disclosure may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional pharmaceutical carriers and excipients such as preservatives, stabilizers, wetting agents, emulsifiers, buffers, etc.
  • the compounds of the present disclosure may be administered by a variety of methods, e.g., orally or by injection (e.g. subcutaneous, intravenous, intraperitoneal, etc.).
  • the active compounds may be coated in a material to protect the compound from the action of acids and other natural conditions which may inactivate the compound. They may also be administered by continuous perfusion/infusion of a disease or wound site.
  • the present methods or compositions may be administered such that the VLA-4 inhibitor is administered intravenously and the first and/ or second agent which interacts with a chemokine is administered subcutaneously.
  • the VLA-4 inhibitor and the first and/or second agent which interacts with a chemokine may be administered subcutaneously.
  • both the VLA-4 inhibitor and the first and/or second agent which interacts with a chemokine are administered subcutaneously in a single formulation.
  • the VLA-4 inhibitor and the first and/or second agent which interacts with a chemokine are administered as a single formulation subcutaneously or intravenously.
  • the protocols for administration to a particular patient may be further optimized by a skilled practitioner.
  • the therapeutic compound may be administered to a patient in an appropriate carrier, for example, liposomes, or a diluent.
  • suitable diluents include saline and aqueous buffer solutions.
  • Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes.
  • the therapeutic compound may also be administered parenterally, intraperitoneally, intramuscularly, intraarterially, intraspinally, or intracerebrally.
  • Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • compositions may be suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (such as, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin.
  • the compounds are formulated in suitable liquid form with excipients as required.
  • the compositions may contain liposomes or other suitable carriers.
  • the solution is made isotonic using standard preparations such as Hank's solution or other isotonic solutions.
  • Sterile injectable solutions can be prepared by incorporating the therapeutic compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the therapeutic compound into a sterile carrier which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the methods of preparation include vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., the therapeutic compound) plus any additional desired ingredient from a previously sterile- filtered solution thereof.
  • the therapeutic compound can be orally administered, for example, with an inert diluent or an assimilable edible carrier.
  • the therapeutic compound and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet.
  • the therapeutic compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • the percentage of the therapeutic compound in the compositions and preparations may, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic compound for the treatment of a selected condition in a patient.
  • the therapeutic compound may also be administered topically to the skin, eye, or mucosa. Alternatively, if local delivery to the lungs is desired the therapeutic compound may be administered by inhalation in a dry-powder or aerosol formulation. Alternatively, the therapeutic agents may be administered transdermally.
  • Active compounds are administered at a therapeutically effective dosage sufficient to treat a condition associated with a condition in a patient.
  • the efficacy of a compound can be evaluated in an animal model system that may be predictive of efficacy in treating the disease in a human or another animal, such as the model systems shown in the examples and drawings.
  • HED human equivalent dose
  • K m values for humans and various animals are well known.
  • the K m for an average 60 kg human (with a BSA of 1 .6 m 2 ) is 37, whereas a 20 kg child (BSA 0.8 m 2 ) would have a K m of 25.
  • K m for some relevant animal models are also well known, including: mice K m of 3 (given a weight of 0.02 kg and BSA of 0.007); hamster K m of 5 (given a weight of 0.08 kg and BSA of 0.02); rat K m of 6 (given a weight of 0.15 kg and BSA of 0.025) and monkey K m of 12 (given a weight of 3 kg and BSA of 0.24).
  • the actual dosage amount of a compound of the present disclosure or composition comprising a compound of the present disclosure administered to a subject may be determined by physical and physiological factors such as type of animal treated, age, sex, body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the subject and on the route of administration. These factors may be determined by a skilled artisan. The practitioner responsible for administration will typically determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject. The dosage may be adjusted by the individual physician in the event of any
  • An effective amount typically will vary from about 0.0001 mg/kg to about 1000 mg/kg, from about 0.001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 750 mg/kg, from about 0.001 mg/kg to about 50 mg/kg, from about 100 mg/kg to about 500 mg/kg, from about 1 .0 mg/kg to about 250 mg/kg, from about 10.0 mg/kg to about 150 mg/kg, or from about 1 .0 mg/kg to about 15 mg/kg in one or more dose administrations daily, for one or several days (depending of course of the mode of administration and the factors discussed above).
  • suitable dose ranges include 1 mg to 10000 mg per day, 100 mg to 10000 mg per day, 500 mg to 10000 mg per day, and 500 mg to 1000 mg per day. In some particular embodiments, the amount is less than 10,000 mg per day with a range of 750 mg to 9000 mg per day.
  • an agent which interacts with a chemokine may be administered in an amount from about 1 mg/kg to about 10 mg/kg, or about 1 mg/kg to about 5 mg/kg, or about 2.5 mg/kg.
  • the agents which interact with a chemokine may be Grop, a CXCR4 inhibitor or derivatives thereof.
  • the VLA-4 inhibitor may be administered in an amount from about 1 mg/kg to about 100 mg/kg, or about 1 mg/kg to about 50 mg/kg, or about 1 mg/kg to about 25 mg/kg, or about 1 mg/kg to about 15 mg/kg, or about 1 mg/kg to about 10 mg/kg, or about 1 mg/kg to about 5 mg/kg, or about 3 mg/kg.
  • a specific VLA-4 inhibitor such as a compound of formula I or a specific compound described in the examples such as firategrast or compound numbers 12, 21 , 22, 23, 24, 25, 26, 33, 34, 35, 36, 42, 43, 44, 45, or 49 may be administered in a range of about 1 mg/kg to about 200 mg/kg, or about 50 mg/kg to about 200 mg/kg, or about 50 mg/kg to about 100 mg/kg, or about 75 mg/kg to about 100 mg/kg, or about 100 mg/kg.
  • the effective amount may be less than 1 mg/kg/day, less than 500 mg/kg/day, less than 250 mg/kg/day, less than 100 mg/kg/day, less than 50 mg/kg/day, less than 25 mg/kg/day or less than 10 mg/kg/day. It may alternatively be in the range of 1 mg/kg/day to 200 mg/kg/day.
  • a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100
  • microgram/kg/body weight about 200 microgram/kg/body weight, about 350
  • microgram/kg/body weight about 500 microgram/kg/body weight, about 1
  • milligram/kg/body weight about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein.
  • a derivable range from the numbers listed herein a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc. , can be administered, based on the numbers described above.
  • a pharmaceutical composition of the present disclosure may comprise, for example, at least about 0.1 % of a compound of the present disclosure.
  • the compound of the present disclosure may comprise between about 1 % to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein.
  • Desired time intervals for delivery of multiple doses can be determined by one of ordinary skill in the art employing no more than routine experimentation. As an example, subjects may be administered two doses daily at approximately 12 hour intervals. In some
  • the agent is administered once a day.
  • the agent(s) may be administered on a routine schedule.
  • a routine schedule refers to a predetermined designated period of time.
  • the routine schedule may encompass periods of time which are identical or which differ in length, as long as the schedule is predetermined.
  • the routine schedule may involve administration twice a day, every day, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks there-between.
  • the predetermined routine schedule may involve administration on a twice daily basis for the first week, followed by a daily basis for several months, etc.
  • the disclosure provides that the agent(s) may taken orally and that the timing of which is or is not dependent upon food intake.
  • the agent can be taken every morning and/or every evening, regardless of when the subject has eaten or will eat.
  • the present disclosure relates to two or more agents used in combination with a VLA-4 antagonist. It is common in the field of mobilizing hematopoietic stem cells to combine therapeutic modalities.
  • the present disclosure describes combinations of VLA-4 antagonists with other therapeutic modalities as combination therapies to increase the mobilization of hematopoietic stem cells.
  • a VLA-4 antagonist and at least two other therapies. These therapies would be provided in a combined amount effective to achieve an increased activity.
  • This process may involve contacting the cells/subjects with the both agents/therapies at the same time, e.g. , using a single composition or pharmacological formulation that includes the agents, or by contacting the cell/subject with two or more distinct compositions or formulations, at the same time, wherein one composition includes the VLA-4 antagonist, one composition includes a first agent and another composition that includes a second agent.
  • the individual compounds in the compositions described herein may precede or follow the other compound treatment by time intervals ranging from seconds to days.
  • time intervals ranging from seconds to days.
  • the agents which interact with a chemokine may be administered about 10-15 minutes, about 5-10 minutes, or about 0-5 minutes prior to administration of the VLA-4 inhibitor.
  • the agents which interact with a chemokine may be administered from about 15 minutes, about 14 minutes, about 13 minutes, about 12 minutes, about 1 1 minutes, about 10 minutes, about 9 minutes, about 8 minutes, about 7 minutes, about 6 minutes, about 5 minutes, about 4 minutes, about 3 minutes, about 2 minutes, to about 1 minute, or any range derivable therein before the VLA-4 inhibitor.
  • the components may be administered at the same time.
  • the compositions and combination of agents used in the methods described herein may be administered as a single bolus dose, a dose over time such as an infusion, as in intravenous, subcutaneous, or transdermal administration, or in multiple dosages. If infusion is used, the combination may be infused for about 15 minutes to about 6 hours. In one embodiment, the infusion may occur for the duration of length of the apheresis. Additionally, the compositions or combination may be administered once daily for multiple days including from 1 to 4 days.
  • compositions or combinations may be any suitable compositions or combinations.
  • compositions or combinations described herein may be
  • HSPCs may be isolated about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, or about 8 hours following administration.
  • the agent may be a CXC chemokine, a CXC chemokine receptor, or a derivative thereof.
  • Some non-limiting examples of agent including Gro- ⁇ , truncated Gro- ⁇ (tGro- ⁇ ), plerixafor (AMD3100), a granulocyte-colony stimulating factor (G-CSF) such as filgrastim, PEG-filgrastim, or lenograstim, or an inhibitor of integrin ⁇ 9 ⁇ 1 such as BOP (/V-benzenesulfonyl)-L-prolyl-L-O-(1 -pyrrolidinylcarbonyl)tyrosine.
  • compositions or methods used herein may be administered with an anti-cancer therapy such as those described below.
  • the methods or compositions described herein may be used in conjunction with standard methods or variations as practiced by a person of ordinary skill in the art.
  • These anti- cancer agents may be administered prior to and/or concomitant with the compositions or methods described herein.
  • anti-cancer therapies which may be used herein include carmustine, etoposide, cytarabine, melphalan, cyclophosphamide, busulfan, thiotepa, bleomycin, platinum (cisplatin), cytarabine, cyclophosphamide, buside, Cytoxan, daunorubicin, doxorubicin, agent ara-C,
  • Revlimid® (thalidomide analog); ProchymalTM; Genasense° (oblimersen sodium);
  • GleevecTM Glivec® (imatinib); tamibarotene; nelarabine; gallium nitrate; PT-100;
  • chemotherapeutic agent refers to the use of drugs to treat cancer.
  • a “chemotherapeutic agent” is used to connote a compound or composition that is administered in the treatment of cancer. These agents or drugs are categorized by their mode of activity within a cell, for example, whether and at what stage they affect the cell cycle. Alternatively, an agent may be characterized based on its ability to directly crosslink DNA, to intercalate into DNA, or to induce chromosomal and mitotic aberrations by affecting nucleic acid synthesis. Most chemotherapeutic agents fall into the following categories: alkylating agents, antimetabolites, antitumor antibiotics, mitotic inhibitors, and nitrosoureas.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan,
  • improsulfan and piposulfan aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine,
  • cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1 -TM1 ); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine,
  • antibiotics such as the enediyne antibiotics ⁇ e.g., calicheamicin, especially calicheamicin ⁇ and
  • calicheamicin ⁇ - ⁇ dynemicin, including dynemicin A; uncialamycin and derivatives thereof; bisphosphonates, such as clodronate; an esperamicin; as well as
  • neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores include aclacinomysins, actinomycin, authrarnycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-l-norleucine, doxorubicin (including
  • morpholino-doxorubicin cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin
  • epirubicin esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalarnycin, olivomycins, peplomycin,
  • mitoguazone mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK polysaccharide complex); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol;
  • mitolactol pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., paclitaxel and docetaxel; chlorambucil; gemcitabine; 6-thioguanine;
  • mercaptopurine methotrexate
  • platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin
  • vinblastine platinum
  • platinum etoposide (VP-16)
  • ifosfamide ifosfamide
  • mitoxantrone vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g. , CPT-1 1 ); topoisomerase inhibitor RFS 2000; difluorometlhylornithine (DMFO); retinoids such as retinoic acid;
  • CDDP cisplatin
  • carboplatin carboplatin
  • procarbazine mechlorethamine
  • cyclophosphamide camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, paclitaxel, docetaxel, gemcitabien, navelbine, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin and methotrexate and
  • Radiotherapy also called radiation therapy, is the treatment of cancer and other diseases with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated by damaging their genetic material, making it impossible for these cells to continue to grow. Although radiation damages both cancer cells and normal cells, the latter are able to repair themselves and function properly.
  • Radiation therapy used according to the present disclsoure may include, but is not limited to, the use of ⁇ -rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells.
  • Other forms of DNA damaging factors are also present.
  • Dosage ranges for X-rays range from daily doses of 12.9 to 51 .6 mC/kg for prolonged periods of time (3 to 4 wk), to single doses of 0.516 to 1 .55 C/kg.
  • Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
  • Radiotherapy may comprise the use of radiolabeled antibodies to deliver doses of radiation directly to the cancer site (radioimmunotherapy).
  • Antibodies are highly specific proteins that are made by the body in response to the presence of antigens (substances recognized as foreign by the immune system). Some tumor cells contain specific antigens that trigger the production of tumor-specific antibodies. Large quantities of these antibodies can be made in the laboratory and attached to radioactive substances (a process known as radiolabeling). Once injected into the body, the antibodies actively seek out the cancer cells, which are destroyed by the cell-killing (cytotoxic) action of the radiation. This approach can minimize the risk of radiation damage to healthy cells.
  • Conformal radiotherapy uses the same radiotherapy machine, a linear accelerator, as the normal radiotherapy treatment but metal blocks are placed in the path of the x-ray beam to alter its shape to match that of the cancer. This ensures that a higher radiation dose is given to the tumor. Healthy surrounding cells and nearby structures receive a lower dose of radiation, so the possibility of side effects is reduced.
  • a device called a multi-leaf collimator has been developed and may be used as an alternative to the metal blocks.
  • the multi-leaf collimator consists of a number of metal sheets which are fixed to the linear accelerator. Each layer can be adjusted so that the radiotherapy beams can be shaped to the treatment area without the need for metal blocks. Precise positioning of the radiotherapy machine is very important for conformal radiotherapy treatment and a special scanning machine may be used to check the position of internal organs at the beginning of each treatment.
  • High-resolution intensity modulated radiotherapy also uses a multi- leaf collimator. During this treatment the layers of the multi-leaf collimator are moved while the treatment is being given. This method is likely to achieve even more precise shaping of the treatment beams and allows the dose of radiotherapy to be constant over the whole treatment area.
  • conformal radiotherapy and intensity modulated radiotherapy may reduce the side effects of radiotherapy treatment, it is possible that shaping the treatment area so precisely could stop microscopic cancer cells just outside the treatment area being destroyed. This means that the risk of the cancer coming back in the future may be higher with these
  • Radiosensitizers make the tumor cells more likely to be damaged, and radioprotectors protect normal tissues from the effects of radiation.
  • Hyperthermia the use of heat, is also being studied for its effectiveness in sensitizing tissue to radiation.
  • immunotherapeutics In the context of cancer treatment, immunotherapeutics, generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells.
  • Trastuzumab (HerceptinTM) is such an example.
  • the immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell.
  • the antibody alone may serve as an effector of therapy or it may recruit other cells to actually affect cell killing.
  • the antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent.
  • the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target.
  • Various effector cells include cytotoxic T cells and NK cells. The combination of therapeutic modalities, i.e. , direct cytotoxic activity and inhibition or reduction of ErbB2 would provide therapeutic benefit in the treatment of ErbB2 overexpressing cancers.
  • the tumor cell must bear some marker that is amenable to targeting, i.e. , is not present on the majority of other cells.
  • Common tumor markers include carcinoembryonic antigen, prostate specific antigen, urinary tumor associated antigen, fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and p155.
  • An alternative aspect of immunotherapy is to combine anticancer effects with immune stimulatory effects.
  • Immune stimulating molecules also exist including: cytokines such as IL-2, IL-4, IL-12, GM-CSF, ⁇ -IFN, chemokines such as MIP-1 , MCP-1 , IL-8 and growth factors such as FLT3 ligand.
  • cytokines such as IL-2, IL-4, IL-12, GM-CSF, ⁇ -IFN
  • chemokines such as MIP-1 , MCP-1 , IL-8 and growth factors such as FLT3 ligand.
  • immune stimulating molecules either as proteins or using gene delivery in combination with a tumor suppressor has been shown to enhance anti-tumor effects (Ju et al., 2000).
  • antibodies against any of these compounds may be used to target the anti-cancer agents discussed herein.
  • immunotherapies currently under investigation or in use are immune adjuvants e.g., Mycobacterium bovis, Plasmodium falciparum, dinitrochlorobenzene and aromatic compounds (U.S. Patents 5,801 ,005 and 5,739, 169; Hui and Hashimoto, 1998; Christodoulides, et ai, 1998), cytokine therapy, e.g., interferons ⁇ , ⁇ , and ⁇ ; IL-1 , GM-CSF and TNF (Bukowski, et ai , 1998; Davidson, et ai, 1998; Hellstrand, et ai, 1998) gene therapy, e.g., TNF, IL-1 , IL-2, p53 (Qin et ai, 1998; Austin-Ward and Villaseca, 1998; U.S.
  • immune adjuvants e.g., Mycobacterium bovis, Plasmodium falciparum, dinit
  • Patents 5,830,880 and 5,846,945) and monoclonal antibodies e.g., anti-ganglioside GM2, anti-HER-2, anti-p185 (Pietras, et al., 1998; Hanibuchi, et ai , 1998; U.S. Patent 5,824,31 1 ). It is contemplated that one or more anti-cancer therapies may be employed with the gene silencing therapies described herein.
  • an antigenic peptide, polypeptide or protein, or an autologous or allogenic tumor cell composition or "vaccine” is an antigenic peptide, polypeptide or protein, or an autologous or allogenic tumor cell composition or "vaccine”.
  • lymphokines such as IL-2 or transduced with genes for tumor necrosis, and readministered (Rosenberg, et al., 1988; 1989).
  • phosphate means -OP(O)(OH)O- or a deprotonated form thereof;
  • mercapto means -SH; and
  • sulfonyl means -S(O)2 ⁇ ; and
  • sulfinyl means -S(O)-.
  • the point of attachment is typically only identified in this manner for larger groups in order to assist the reader in unambiguously identifying a point of attachment.
  • the symbol "- ⁇ " means a single bond where the group attached to the thick end of the wedge is “out of the page.”
  • the symbol “ ⁇ " means a single bond where the group attached to the thick end of the wedge is “into the page”.
  • the symbol " > ⁇ " means a single bond where the geometry around a double bond (e.g., either E or Z) is undefined. Both options, as well as combinations thereof are therefore intended. Any undefined valency on an atom of a structure shown in this application implicitly represents a hydrogen atom bonded to that atom.
  • a bold dot on a carbon atom indicates that the hydrogen attached to that carbon is oriented out of the plane of the paper.
  • R may replace any hydrogen atom attached to any of the ring atoms, including a depicted, implied, or expressly defined hydrogen, so long as a stable structure is formed.
  • a group “R” is depicted as a "floating group” on a fused ring system, as for example in the formula:
  • R may replace any hydrogen attached to any of the ring atoms of either of the fused rings unless specified otherwise.
  • Replaceable hydrogens include depicted hydrogens (e.g. , the hydrogen attached to the nitrogen in the formula above), implied hydrogens (e.g., a hydrogen of the formula above that is not shown but understood to be present), expressly defined hydrogens, and optional hydrogens whose presence depends on the identity of a ring atom ⁇ e.g., a hydrogen attached to group X, when X equals -CH-), so long as a stable structure is formed.
  • R may reside on either the 5-membered or the 6-membered ring of the fused ring system.
  • the number of carbon atoms in the group or class is as indicated as follows: "Cn” defines the exact number (n) of carbon atoms in the group/class. "C ⁇ n” defines the maximum number (n) of carbon atoms that can be in the group/class, with the minimum number as small as possible for the group/class in question, e.g. , it is understood that the minimum number of carbon atoms in the group “alkenyl ( c ⁇ 8)” or the class “alkene ( c ⁇ 8)” is two. Compare with “alkoxy(c ⁇ io)", which designates alkoxy groups having from 1 to 10 carbon atoms.
  • Cn-n defines both the minimum (n) and maximum number ( ⁇ ') of carbon atoms in the group.
  • alkyl C 2-i o> designates those alkyl groups having from 2 to 10 carbon atoms. These carbon number indicators may precede or follow the chemical groups or class it modifies and it may or may not be enclosed in parenthesis, without signifying any change in meaning.
  • C5 olefin C5-olefin
  • olefin ⁇ s olefin ⁇ s
  • olefin C 5" are all synonymous. When any of the chemical groups or compound classes defined herein is modified by the term “substituted”, any carbon atom(s) in a moiety replacing a hydrogen atom is not counted. Thus methoxyhexyl, which has a total of seven carbon atoms, is an example of a substituted alkyl ( ci-6) .
  • saturated when used to modify a compound or chemical group means the compound or chemical group has no carbon-carbon double and no carbon-carbon triple bonds, except as noted below.
  • the term when used to modify an atom, it means that the atom is not part of any double or triple bond.
  • substituted versions of saturated groups one or more carbon oxygen double bond or a carbon nitrogen double bond may be present. And when such a bond is present, then carbon-carbon double bonds that may occur as part of keto-enol tautomerism or imine/enamine tautomerism are not precluded.
  • saturated when used to modify a solution of a substance, it means that no more of that substance can dissolve in that solution.
  • aliphatic when used without the "substituted” modifier signifies that the compound or chemical group so modified is an acyclic or cyclic, but non-aromatic hydrocarbon compound or group.
  • the carbon atoms can be joined together in straight chains, branched chains, or non- aromatic rings (alicyclic).
  • Aliphatic compounds/groups can be saturated, that is joined by single carbon-carbon bonds (alkanes/alkyl), or unsaturated, with one or more carbon-carbon double bonds (alkenes/alkenyl) or with one or more carbon-carbon triple bonds (alkynes/alkynyl).
  • aromatic when used to modify a compound or a chemical group refers to a planar unsaturated ring of atoms with An +2 electrons in a fully conjugated cyclic ⁇ system.
  • alkyl when used without the "substituted” modifier refers to a monovalent saturated aliphatic group with a carbon atom as the point of
  • alkanediyl when used without the “substituted” modifier refers to a divalent saturated aliphatic group, with one or two saturated carbon atom(s) as the point(s) of attachment, a linear or branched acyclic structure, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen.
  • the groups -CH 2 - (methylene), -CH 2 CH 2 -, -CH 2 C(CH 3 ) 2 CH 2 -, and -CH 2 CH 2 CH 2 - are non-limiting examples of alkanediyl groups.
  • alkane refers to the class of compounds having the formula H-R, wherein R is alkyl as this term is defined above.
  • R is alkyl as this term is defined above.
  • substituted one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO 2 , -CO 2 H, -CO 2 CH 3 , -CN, -SH, -OCH 3 , -OCH 2 CH 3 , -C(O)CH 3 , -NHCH 3 , -NHCH 2 CH 3 , -N(CH 3 ) 2 , -C(O)NH 2 , -C(O)NHCH 3 , -C(O)N(CH 3 ) 2 ,
  • -OC(O)CH 3 -NHC(O)CH 3 , -S(O) 2 OH, or -S(O) 2 NH 2 .
  • the following groups are non- limiting examples of substituted alkyl groups: -CH 2 OH, -CH 2 CI, -CF 3 , -CH 2 CN,
  • haloalkyl is a subset of substituted alkyl, in which the hydrogen atom replacement is limited to halo (i.e. -F, -CI, -Br, or -I) such that no other atoms aside from carbon, hydrogen and halogen are present.
  • -CH 2 CI is a non-limiting example of a haloalkyl.
  • fluoroalkyl is a subset of substituted alkyl, in which the hydrogen atom replacement is limited to fluoro such that no other atoms aside from carbon, hydrogen and fluorine are present.
  • the groups -CH 2 F, -CF 3 , and -CH 2 CF 3 are non-limiting examples of fluoroalkyl groups.
  • cycloalkyi when used without the “substituted” modifier refers to a monovalent saturated aliphatic group with a carbon atom as the point of attachment, said carbon atom forming part of one or more non-aromatic ring structures, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen.
  • Non-limiting examples include: -CH(CH 2 )2 (cyclopropyl), cyclobutyl, cyclopentyl, or cyclohexyl (Cy).
  • cycloalkanediyl when used without the “substituted” modifier refers to a divalent saturated aliphatic group with two carbon atoms as points of attachment, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen.
  • the group is a non-limiting example of cycloalkanediyl group.
  • a "cycloalkane” refers to the class of compounds having the formula H-R, wherein R is cycloalkyi as this term is defined above.
  • alkenyl when used without the "substituted” modifier refers to an monovalent unsaturated aliphatic group with a carbon atom as the point of attachment, a linear or branched, acyclic structure, at least one nonaromatic carbon- carbon double bond, no carbon-carbon triple bonds, and no atoms other than carbon and hydrogen.
  • alkenediyl when used without the "substituted” modifier refers to a divalent unsaturated aliphatic group, with two carbon atoms as points of attachment, a linear or branched, a linear or branched acyclic structure, at least one nonaromatic carbon- carbon double bond, no carbon-carbon triple bonds, and no atoms other than carbon and hydrogen.
  • alkene and olefin are synonymous and refer to the class of compounds having the formula H-R, wherein R is alkenyl as this term is defined above.
  • terminal alkene and "a- olefin” are synonymous and refer to an alkene having just one carbon-carbon double bond, wherein that bond is part of a vinyl group at an end of the molecule.
  • substituted one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO 2 , -CO 2 H, -CO 2 CH 3 , -CN, -SH, -OCH3, -OCH 2 CH 3 , -C(O)CH 3 , -NHCH 3 , -NHCH 2 CH 3 , -N(CH 3 ) 2 ,
  • aryl when used without the "substituted” modifier refers to a monovalent unsaturated aromatic group with an aromatic carbon atom as the point of attachment, said carbon atom forming part of a one or more six-membered aromatic ring structure, wherein the ring atoms are all carbon, and wherein the group consists of no atoms other than carbon and hydrogen. If more than one ring is present, the rings may be fused or unfused. As used herein, the term does not preclude the presence of one or more alkyl or aralkyi groups (carbon number limitation permitting) attached to the first aromatic ring or any additional aromatic ring present.
  • aryl groups include phenyl (Ph), methylphenyl, (dimethyl)phenyl, -C 6 H 4 CH 2 CH 3
  • aromaticiyl when used without the "substituted” modifier refers to a divalent aromatic group with two aromatic carbon atoms as points of attachment, said carbon atoms forming part of one or more six-membered aromatic ring structure(s) wherein the ring atoms are all carbon, and wherein the monovalent group consists of no atoms other than carbon and hydrogen.
  • the term does not preclude the presence of one or more alkyl, aryl or aralkyi groups (carbon number limitation permitting) attached to the first aromatic ring or any additional aromatic ring present.
  • the rings may be fused or unfused.
  • Unfused rings may be connected via one or more of the following: a covalent bond, alkanediyl, or alkenediyl groups (carbon number limitation permitting).
  • alkanediyl or alkenediyl groups (carbon number limitation permitting).
  • arenediyl groups include:
  • An "arene” refers to the class of compounds having the formula H-R, wherein R is aryl as that term is defined above. Benzene and toluene are non- limiting examples of arenes. When any of these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO 2 , -CO 2 H, -CO 2 CH 3 , -CN, -SH, -OCH 3 , -OCH 2 CH 3 , -C(O)CH 3 , -NHCH 3 , -NHCH 2 CH 3 , -N(CH 3 ) 2 , -C(O)NH 2 , -C(O)NHCH 3 , -C(O)N(CH 3 ) 2 ,
  • aralkyl when used without the “substituted” modifier refers to the monovalent group -alkanediyl-aryl, in which the terms alkanediyl and aryl are each used in a manner consistent with the definitions provided above.
  • Non-limiting examples are: phenylmethyl (benzyl, Bn) and 2-phenyl-ethyl.
  • aralkyl When the term aralkyl is used with the "substituted" modifier one or more hydrogen atom from the alkanediyl and/or the aryl group has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO 2 , -CO 2 H, -CO 2 CH 3 , -CN, -SH, -OCH 3 , -OCH 2 CH 3 , -C(O)CH 3 , -NHCH 3 ,
  • Non-limiting examples of substituted aralkyls are: (3-chlorophenyl)-methyl, and 2-chloro-2-phenyl-eth-1 -yl.
  • acyl when used without the “substituted” modifier refers to the group -C(O)R, in which R is a hydrogen, alkyl, cycloalkyl, or aryl as those terms are defined above.
  • R is a hydrogen, alkyl, cycloalkyl, or aryl as those terms are defined above.
  • acyl groups are non-limiting examples of acyl groups.
  • a "thioacyl” is defined in an analogous manner, except that the oxygen atom of the group -C(0)R has been replaced with a sulfur atom, -C(S)R.
  • aldehyde corresponds to an alkyl group, as defined above, attached to a -CHO group.
  • one or more hydrogen atom (including a hydrogen atom directly attached to the carbon atom of the carbonyl or thiocarbonyl group, if any) has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO 2 , -CO 2 H, -CO 2 CH 3 , -CN, -SH, -OCH 3 , -OCH 2 CH 3 , -C(O)CH 3 , -NHCH 3 , -NHCH 2 CH 3 , -N(CH 3 ) 2 , -C(O)NH 2 , -C(O)NHCH 3 , -C(O)N(CH 3 ) 2 , -OC(O)CH 3 , -NHC(O)CH 3 , -S(O) 2 OH, or -S(O) 2 NH 2 .
  • the groups, -C(O)CH 2 CF 3 , -CO 2 H (carboxyl), -CO 2 CH 3 (methylcarboxyl), -CO 2 CH 2 CH 3 , -C(O)NH 2 (carbamoyl), and -CON(CH 3 ) 2 are non-limiting examples of substituted acyl groups.
  • alkoxy when used without the "substituted” modifier refers to the group -OR, in which R is an alkyl, as that term is defined above.
  • Non- limiting examples include: -OCH 3 (methoxy), -OCH 2 CH 3 (ethoxy), -OCH 2 CH 2 CH 3 , -OCH(CH 3 ) 2 (isopropoxy), -OC(CH 3 ) 3 (fe/f-butoxy), -OCH(CH 2 ) 2 , -O-cyclopentyl, and -O-cyclohexyl.
  • cycloalkoxy "alkenyloxy", “aryloxy”, “aralkoxy”
  • acyloxy when used without the “substituted” modifier, refers to groups, defined as -OR, in which R is cycloalkyl, alkenyl, aryl, aralkyl, and acyl, respectively.
  • alkylthio and “acylthio” when used without the “substituted” modifier refers to the group -SR, in which R is an alkyl and acyl, respectively.
  • alcohol corresponds to an alkane, as defined above, wherein at least one of the hydrogen atoms has been replaced with a hydroxy group.
  • ether corresponds to an alkane, as defined above, wherein at least one of the hydrogen atoms has been replaced with an alkoxy group.
  • alkylamino when used without the "substituted” modifier refers to the group -NHR, in which R is an alkyl, as that term is defined above. Non- limiting examples include: -NHCH 3 and -NHCH 2 CH 3 .
  • dialkylamino when used without the "substituted” modifier refers to the group -NRR', in which R and R' can be the same or different alkyl groups, or R and R' can be taken together to represent an alkanediyl.
  • dialkylamino groups include: -N(CH 3 ) 2 and
  • substituted modifier refers to groups, defined as -NHR, in which R is cycloalkyl, alkenyl, aryl, aralkyl, alkoxy, and alkylsulfonyl, respectively.
  • a non-limiting example of an arylamino group is -NHC6H5.
  • amido (acylamino), when used without the “substituted” modifier, refers to the group -NHR, in which R is acyl, as that term is defined above.
  • a non-limiting example of an amido group is -NHC(0)CH 3 .
  • R is an alkyl
  • one or more hydrogen atom attached to a carbon atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH 2 , -NO 2 , -CO 2 H, -CO 2 CH 3 , -CN, -SH, -OCH 3 , -OCH 2 CH 3 , -C(O)CH 3 , -NHCH 3 , -NHCH 2 CH 3 ,
  • -N(CH 3 ) 2 -C(O)NH 2 , -C(O)NHCH 3 , -C(O)N(CH 3 ) 2 , -OC(O)CH 3 , -NHC(O)CH 3 , -S(O) 2 OH, or -S(O) 2 NH 2 .
  • the groups -NHC(O)OCH 3 and -NHC(O)NHCH 3 are non- limiting examples of substituted amido groups.
  • alkylsulfonyl and “alkylsulfinyl” when used without the “substituted” modifier refers to the groups -S(O) 2 R and -S(O)R, respectively, in which R is an alkyl, as that term is defined above.
  • cycloalkylsulfonyl
  • alkenylsulfonyl alkynylsulfonyl
  • arylsulfonyl arylsulfonyl
  • aralkylsulfonyl heteroarylsulfonyl
  • heterocycloalkylsulfonyl and “heterocycloalkylsulfony are defined in an analogous manner.
  • An "active ingredient” (also referred to as an active compound, active substance, active agent, pharmaceutical agent, agent, biologically active molecule, or a therapeutic compound) is the ingredient in a pharmaceutical drug or a pesticide that is biologically active.
  • active pharmaceutical ingredient API
  • bulk active are also used in medicine, and the term active substance may be used for pesticide formulations.
  • any method that "comprises,” “has” or “includes” one or more steps is not limited to possessing only those one or more steps and also covers other unlisted steps.
  • the term “consisting substantially of” means that the composition contains at least 98% of the elements listed. Similarly, the term “consisting essentially of” means that the composition contains at least 99% of the elements listed.
  • derivative when used in the context of a protein, such as a cytokine or chemokine, refers to a protein which is a recombinant version, a fragment, a truncated version, or a chemically modified protein.
  • the chemically modified protein is a protein to which a secondary group such as a PEG chain
  • Effective amount when used in the context of treating a patient or subject with a compound means that amount of the compound which, when administered to a subject or patient for treating or preventing a disease, is an amount sufficient to effect such treatment or prevention of the disease.
  • An “excipient” is a pharmaceutically acceptable substance formulated along with the active ingredient(s) of a medication, pharmaceutical composition, formulation, or drug delivery system. Excipients may be used, for example, to stabilize the composition, to bulk up the composition (thus often referred to as “bulking agents,” “fillers,” or “diluents” when used for this purpose), or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption, reducing viscosity, or enhancing solubility.
  • Excipients include pharmaceutically acceptable versions of antiadherents, binders, coatings, colors, disintegrants, flavors, glidants, lubricants, preservatives, sorbents, sweeteners, and vehicles.
  • the main excipient that serves as a medium for conveying the active ingredient is usually called the vehicle. Excipients may also be used in the
  • an excipient will typically vary depending on the route of administration, the dosage form, the active ingredient, as well as other factors.
  • HSPCs refers to hematopoietic stem and progenitor cells. HSPCs are a combination of progenitor cells and stem cells.
  • hydrate when used as a modifier to a compound means that the compound has less than one (e.g., hemihydrate), one (e.g., monohydrate), or more than one (e.g., dihydrate) water molecules associated with each compound molecule, such as in solid forms of the compound.
  • IC50 refers to an inhibitory dose which is 50% of the maximum response obtained. This quantitative measure indicates how much of a particular active ingredient or other substance (inhibitor) is needed to inhibit a given biological, biochemical or chemical process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half.
  • An "isomer" of a first compound is a separate compound in which each molecule contains the same constituent atoms as the first compound, but where the configuration of those atoms in three dimensions differs.
  • the term "patient” or “subject” refers to a living organism, such as a human, monkey, cow, sheep, goat, dog, cat, mouse, rat, guinea pig, avain, or transgenic species thereof.
  • the patient or subject is a primate.
  • Non-limiting examples of human patients are adults, juveniles, infants and fetuses.
  • pharmaceutically acceptable refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable salts” means salts of compounds of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as
  • cyclopentanepropionic acid ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, heptanoic acid, hexanoic acid,
  • hydroxynaphthoic acid lactic acid, laurylsulfuric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, muconic acid, o-(4-hydroxybenzoyl)benzoic acid, oxalic acid, p-chlorobenzenesulfonic acid, phenyl-substituted alkanoic acids, propionic acid, p-toluenesulfonic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, tartaric acid, tertiarybutylacetic acid, trimethylacetic acid, and the like.
  • Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases.
  • Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide.
  • Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine,
  • a "pharmaceutically acceptable carrier,” “drug carrier,” or simply “carrier” is a pharmaceutically acceptable substance formulated along with the active ingredient medication that is involved in carrying, delivering and/or transporting a chemical agent. Drug carriers may be used to improve the delivery and the
  • Some drug carriers may increase the effectiveness of drug delivery to the specific target sites.
  • carriers include: liposomes, microspheres (e.g., made of poly(lactic-co- glycolic) acid), albumin microspheres, synthetic polymers, nanofibers, protein-DNA complexes, protein conjugates, erythrocytes, virosomes, and dendrimers.
  • a "pharmaceutical drug” (also referred to as a pharmaceutical, pharmaceutical preparation, pharmaceutical composition, pharmaceutical formulation, pharmaceutical product, medicinal product, medicine, medication, medicament, or simply a drug) is a compound or composition used to diagnose, cure, treat, or prevent disease.
  • An active ingredient (Al) (defined above) is the ingredient in a pharmaceutical drug or a pesticide that is biologically active.
  • active pharmaceutical ingredient (API) and bulk active are also used in medicine, and the term active substance may be used for pesticide formulations.
  • Some medications and pesticide products may contain more than one active ingredient.
  • the inactive ingredients are usually called excipients (defined above) in pharmaceutical contexts.
  • pre-malignant cells refers to cells that can form malignant hematopoietic or myeloid cells.
  • the malignant hematopoietic or myeloid cells are those which characterize the conditions of myeloma, leukemia, and lymphoma.
  • Particular forms of these diseases include acute myelitic leukemia (AML), acute lymphatic leukemia (ALL), multiple myeloma (MM), chronic myelogenous leukemia (CML), chronic lymphatic leukemia (CLL), hairy cell leukemia (HCL), acute promyelocytic leukemia (APL), and various lymphomas.
  • Prevention includes: (1 ) inhibiting the onset of a disease in a subject or patient which may be at risk and/or predisposed to the disease but does not yet experience or display any or all of the pathology or symptomatology of the disease, and/or (2) slowing the onset of the pathology or symptomatology of a disease in a subject or patient which may be at risk and/or predisposed to the disease but does not yet experience or display any or all of the pathology or symptomatology of the disease.
  • Prodrug means a compound that is convertible in vivo
  • the prodrug itself may or may not also have activity with respect to a given target protein.
  • a compound comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound.
  • Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, phosphates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis- -hydroxynaphthoate, gentisates, isethionates, di-p-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, esters of amino acids, and the like.
  • progenitor cells refers to cells that, in response to certain stimuli, can form differentiated hematopoietic or myeloid cells. The presence of progenitor cells can be assessed by the ability of the cells in a sample to form colony-forming units of various types, including, for example, CFU-GM (colony- forming units, granulocyte-macrophage); CFU-GEMM (colony-forming units,
  • BFU-E burst-forming units, erythroid
  • HPP-CFC high proliferative potential colony-forming cells
  • a “stereoisomer” or “optical isomer” is an isomer of a given compound in which the same atoms are bonded to the same other atoms, but where the configuration of those atoms in three dimensions differs. "Enantiomers” are
  • stereoisomers of a given compound that are mirror images of each other, like left and right hands.
  • “Diastereomers” are stereoisomers of a given compound that are not enantiomers.
  • Chiral molecules contain a chiral center, also referred to as a
  • stereocenter or stereogenic center which is any point, though not necessarily an atom, in a molecule bearing groups such that an interchanging of any two groups leads to a stereoisomer.
  • the chiral center is typically a carbon, phosphorus or sulfur atom, though it is also possible for other atoms to be stereocenters in organic and inorganic compounds.
  • a molecule can have multiple stereocenters, giving it many stereoisomers.
  • compounds whose stereoisomerism is due to tetrahedral stereogenic centers (e.g. , tetrahedral carbon), the total number of hypothetically possible
  • stereoisomers will not exceed 2 n , where n is the number of tetrahedral stereocenters. Molecules with symmetry frequently have fewer than the maximum possible number of stereoisomers.
  • a 50:50 mixture of enantiomers is referred to as a racemic mixture.
  • a mixture of enantiomers can be enantiomerically enriched so that one enantiomer is present in an amount greater than 50%.
  • enantiomers and/or diastereomers can be resolved or separated using techniques known in the art.
  • stereocenter or axis of chirality for which stereochemistry has not been defined, that stereocenter or axis of chirality can be present in its R form, S form, or as a mixture of the R and S forms, including racemic and non-racemic mixtures.
  • the phrase "substantially free from other stereoisomers” means that the composition contains ⁇ 15%, more preferably ⁇ 10%, even more preferably ⁇ 5%, or most preferably ⁇ 1 % of another stereoisomer(s).
  • stem cells are less differentiated forms of progenitor cells. Typically, such cells are positive for CD34, but stem cells do not have to contain this marker. While other types of cells such as endothelial cells and mast cells also may exhibit this marker, CD34 is considered an one marker of stem cell presence. CD34+ cells can be assayed using fluorescence activated cell sorting (FACS) and thus their presence can be assessed in a sample using this technique. In general, CD34+ cells are present only in low levels in the blood, but are present in large numbers in bone marrow. Additionally, the stem cells may be hematopoietic stem cells that express the SLAM and LSK markers.
  • FACS fluorescence activated cell sorting
  • hematopoietic stem cells may be LSK cells or LSK-SLAM cells, which are considered early hematopoietic stem cells.
  • LSK refers to Lin " Sca1 + c-Kit + and SLAM is signaling lymphocyte activation molecules or Lin " CD41 " CD48 " CD150 + .
  • Treatment includes (1 ) inhibiting a disease in a subject or patient experiencing or displaying the pathology or symptomatology of the disease (e.g., arresting further development of the pathology and/or symptomatology), (2) ameliorating a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease (e.g., reversing the pathology and/or symptomatology), and/or (3) effecting any measurable decrease in a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease.
  • inhibiting a disease in a subject or patient experiencing or displaying the pathology or symptomatology of the disease e.g., arresting further development of the pathology and/or symptomatology
  • ameliorating a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease e.g., reversing the pathology and/or symptomatology
  • CFC colony-forming cell
  • CPU-GM, BFU-E, and CFU-GEMM colony types will be enumerated following 1 week of culture at 37 °C.
  • CD34- c-kit+ Sca-1 + lineage marker- (CD34-KSL) cells are regarded as one of the populations that have the highest enrichment of HSPCs in adult mouse bone marrow. More recently, the SLAM family markers, CD150 and CD48, have been used to differentiate stem cells from more committed progenitor cells.
  • SLAM family markers, CD150 and CD48 have been used to differentiate stem cells from more committed progenitor cells.
  • primitive murine HSPC mobilization will be examined by flow cytometry enumeration of CD34-c-kit+Sca-1 +lineage marker-CD150+CD48- cell numbers following treatment with mobilization regimens.
  • the definitive assay for stem cell activity in a test sample is the complete and sustained (> 5 months) reconstitution of all hematopoietic lineages in irradiated recipients by transplanted HSPCs.
  • peripheral blood mononuclear cells from wild- type C57BL/6 (CD45.2+) mice mobilized by the regimens described herein will be mixed with 2x105 competitor bone marrow cells from C57BL/6J x B6.SJL-Ptprca Pepcb/BoyJ F1 (CD45.1 +/CD45.2+) mice and transplanted into lethally irradiated (1 100 cGy) recipients congenic at the CD45.1 + locus (B6.SJL-Ptprca Pepcb/BoyJ).
  • PBMCs peripheral blood mononuclear cells
  • At least 3 mobilized PBMC:competitor ratios (typically the number of PBMCs isolated from 0.5, 1 , and 1 .5 ml_ of blood) will be evaluated to establish the appropriate donor: competitor ratio. Secondary transplantations will be performed by intravenously injecting 106 unfractionated bone marrow cells from donors 5 months after transplantation into lethally irradiated (1 100 cGy) CD45.1 + secondary recipients.
  • Recipient mice C57BL/6 will be injected with 1 ⁇ 106 murine acute promyelocytic (APL) leukemia cells transduced with Click Beetle Red-GFPe.
  • APL acute promyelocytic
  • the treatments disclosed herein will be administered with and without chemotherapy and mouse survival and leukemia burden as measured by BLI and FACS (Gr-1 +/c-kit+/CD34+ leukemia cells) will be assessed. Both BLI and FACS measurements will be obtained on days +7, +14, +21 , +28, and +35.
  • mice Lethal irradiation of BALB/c recipients.
  • Recipient BALB/c mice are lethally irradiated with 925 cGy of total body irradiation on day -1 and then infused with 5x106 T cell depleted bone marrow cells (TCD-BM) containing 2x106 T cells from C57BL/6 mice on day 0.
  • TCD-BM 5x106 T cell depleted bone marrow cells
  • Mice are monitored for indications of GvHD (clinical score based on weight loss, hunched posture, activity, fur texture, diarrhea and skin integrity) and survival time. Signs of GvHD are typically seen within 2-3 weeks and all mice die by 4-5 weeks.
  • mice receiving TCD-BM only will be compared with the rate in mice receiving TCD-BM plus the different donor PBMC populations described herein.
  • Mice will be sacrificed on day 25 to collect and examine GvHD target organs (histology of liver, intestines, and spleen). Peripheral blood will also be collected on day 25 to determine the extent of donor cell engraftment and immune reconstitution (flow cytometry using antibodies against CD3, B220, CD4, CD8, FOXP3, H-2Kb, CD45.1 , and CD45.2).
  • B6D2F1 mice Lethal irradiation of B6D2F1 recipients.
  • Recipient B6D2F1 mice are lethally irradiated with 1 100 cGy (split doses, 3 hr apart) of total body irradiation on day - 1 and then infused with 1 ⁇ 10 T cell depleted splenocytes (TCD-SPL) containing 3x106 T cells from C57BL/6 mice on day 0.
  • TCD-SPL T cell depleted splenocytes
  • mice will be monitored for indications of GvHD and donor engraftment as described above.
  • Example 2 - VLA-4 antagonists enhance mobilization of HSPCs.
  • VLA-4 antagonist BI05192 results in a 30-fold increase in mobilization of HSPCs and combination of BI05192 with plerixafor gave a further 3-fold increase.
  • Treatment with a combination of BI05192, plerixafor, and G-CSF enhanced mobilization by 17-fold compared to G-CSF alone.
  • BI05192 also mobilized long-term repopulating cells that successfully engraft and expand in a multi- lineage manner in secondary transplant experiments. Similar mobilization studies were performed using the small molecule VLA-4 antagonist firategrast.
  • a combination under investigation uses firategrast with the CXCR2 agonist truncated Gro-beta (Gro- ⁇ , SB-251353).
  • Gro- ⁇ is a recombinant, N-terminal truncated form of Gro- ⁇ that binds to CXCR2 with greater potency than the full-length form of Gro- ⁇ . If both firategrast and Gro- ⁇ are given to mice within 5 minutes of each other and blood is collected 15 min post-administration, a significant synergy in colony- forming cell (CFC) mobilization is observed that persisted for >2 hours (FIG. 2).
  • CFC colony- forming cell
  • the level of CFC mobilization achieved with the firategrast and Gro- ⁇ regimen is greater than that of G-CSF used alone for 4 days.
  • Example 3 Combination of Grop or truncated Grop (Gropt) plus a VLA-4 small molecule inhibitor for hematopoietic stem cell mobilization and leukemia chemosensitization.
  • Example 3 Additional VLA-4 antagonists.
  • Table 1 includes the characterization of the VLA-4 antagonist described herein
  • Table 2 Inhibition of s VCAM-1 binding to human G2 ALL cells.
  • Ptprc a Pep3 b /BoyJ (CD45.1 ) mice were purchased from the Jackson Laboratory (Bar Harbor, ME, USA).
  • F1 -hybrid mice (CD45.1/2) were obtained through breeding CD45.2 and CD45.1 mice. Animals were housed at the Washington University Medical School vivarium under SPF conditions. All experiments were performed in accordance with the guidelines of the Washington University Animal Studies Committee and the institutional animal care and use committee (IACUC), in agreement with AAALAC guidelines.
  • mice were kept on antibiotic medication, sulfamethoxazole and trimethoprim, 0.5 and 0.1 mg/ml_ respectively, (Hi-Tech Pharmacal, Amityville, NY, USA) peroral in drinking water.
  • RhG-CSF (Neupogen®, Filgrastim, Amgen, Thousand Oaks, CA, USA) diluted in PBS was injected i.p. every 12 hrs at a dose of 100 g/kg for a total of 9 (day 5) doses.
  • CFC Colony forming cell
  • PB Peripheral blood
  • WBC cell counts
  • Dead cells were excluded from these assays by staining with 2 ⁇ g/mL 7-amino- actinomycin D (Molecular Probes, Eugene, OR) for 5 min prior to analysis. Appropriate isotype-matched negative controls were used to assess background fluorescence intensity. Samples were analyzed on a Beckman Coulter Gallios flow cytometer and data were analyzed using FlowJo software (TreeStar, Ashland, OR). Absolute numbers of immunophenotypically defined populations (e.g. LSK) were calculated by identifying CD45 positive cells as WBC from the corresponding cell count analysis. Statistical comparisons of flow cytometry data were performed using an unpaired two-tailed Student t-test (GraphPad Prism). P-values ⁇ 0.05 were considered significant.
  • Repopulating unit (RU) assay An RU assay was performed to directly compare the repopulating capacity of peripheral blood mobilized with different combinations of compounds.
  • Lethally irradiated CD45.2 hosts received transplants consisting of 6 ⁇ _ of blood from mobilized mice (CD45.1 +) together with 2.5x10 5 CD45.2+ BM competitor cells.
  • the repopulation unit assay shows that the mobilization methods described herein using a combination of a VLA4 antagonist and tGro- ⁇ result in mobilized stem cells which can be successfully transplanted in both primary and secondary recipients.
  • compositions which contained tGro- ⁇ showed MMP9 activity.
  • flow cytometry and real-time PCR it was determined that myeloid cells express both MMP9 and CXCR2 and may play a role in the mobilization of the desired stem cells. See FIG. 9B.
  • GR1 + myeloid cells were removed through the use of a GR1 + antibody, the mobilization of CFU-C was greatly decreased relative to a control antibodies (FIG. 9C). Without wishing to be bound by any theory, it is believed that GR1 + myeloid cells play a role in stem cell mobilization.
  • mice were inject mice as indicated above.
  • Peripheral blood was collected from the mice at baseline, 0.25, 0.5, 1 , 2 and 4 hours post-injection. Aliquots of blood were removed and stored for the CFU assay and Hemavet. On day +7 CFUs were counted.
  • the triple combination of combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist provided significantly improved results over the other conditions tested.
  • mice were administered simultaneous subcutaneous injection of; A) CWHM-823 (3 mg/kg) + t-Grop (2.5 mg/kg), B) AMD3100 (5 mg/kg) + t-Grop (2.5 mg/kg), or C) CWHM-823 (3 mg/kg) + AMD3100 (5 mg/kg) + t- Grop (2.5 mg/kg).
  • A) CWHM-823 (3 mg/kg) + t-Grop (2.5 mg/kg)
  • B) AMD3100 5 mg/kg) + t-Grop (2.5 mg/kg)
  • C) CWHM-823 (3 mg/kg) + AMD3100 (5 mg/kg) + t- Grop (2.5 mg/kg).
  • the triple combination surprisingly outperforms compositions with a VLA-4 inhibitor and CXCR2 agonist or CXCR4 inhibitor and CXCR2 agonist.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present disclosure provides methods of treating a patient comprising administering an agent which interacts with a chemokine such as G-CSF, plerixafor, or Gro-β and VLA-4 inhibitors. These methods may be used in the treatment of a condition which requires the collection of hematopoietic stem cells for transfusions or in chemotherapy.

Description

COMPOSITIONS COMPRISING AN INTEGRIN INHIBITOR AND AGENTS WHICH INTERACT WITH A CHEMOKINE AND METHODS OF USE THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
[0001 ] This application claims the benefit of priority to United States Provisional Application Nos. 62/416,462, filed on November 2, 2016, 62/430, 167, filed on December 5, 2016, the entire contents of which are hereby incorporated by reference.
GOVERNMENTAL RIGHTS
[0002] This invention was made with government support under
CA152329 awarded by the National Institute of Health. The government has certain rights in the invention.
FIELD OF THE INVENTION
[0003] The present disclosure relates to the fields of pharmaceuticals, medicine and cell biology. More specifically, it relates to pharmaceutical agents such as VLA-4 inhibitor and a first and second agent which interacts with a chemokine used in combination to enhance the collection of hematopoietic stem cells.
BACKGROUND OF THE INVENTION
[0004] Hematopoietic stem cell transplantation (HSCT) is the major curative therapy available for many hematological diseases including hematological cancers. In this technique, HSCT is used to facilitate repopulation of healthy bone marrow and immune system cells after a high-dose chemotherapy treatment for cancers including but not limited to Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia. In order to facilitate transplantation when the cells are need,
hematopoietic stem/progenitor cells (HSPCs) are collected from the patient's blood, harvested, frozen and then stored while the patient receives high-dose chemotherapy and/or radiation therapy. In order to achieve a successful transplantation, an
intravenous infusion of a minimum number of 2x106 CD34+ stem cells/kg body weight l is often needed; however, a dose of 5x106 CD34+ cells/kg is considered preferable for early and long term multilineage engraftment.
[0005] Currently, the stem cells for hematopoietic stem cell transplants are often harvested from peripheral blood. Due to the low amount of these cells in circulating peripheral blood, the stem cells often must be stimulated to increase the quantity in the peripheral blood, a process which can take almost a week. Even then, the collection is still done over several days to achieve sufficient concentrations of the stem cells for transplantation. This greatly increases the cost of the transplant and results in a significant burden on the patient. Currently, cytokines, such as granulocyte- colony forming unit (G-CSF), and immunostimulants, such as plerixafor, are used to increase the amount of hematopoietic stem cells in the peripheral blood but a single agent often results in insufficient mobilization of stem cells. Additional methods of harvesting hematopoietic stem cells have been developed which involve combining G- CSF with multiple other agents such as plerixafor or another cytokine. Unfortunately, even these combined therapies often fail to increase the concentrations to sufficient levels for transplantation in many patients even with multiple days of apheresis.
Furthermore, several of these agents, like plerixafor, are extremely expensive adding over $25,000 per patient relative to using G-CSF alone. Therefore, a need remains for better methods to harvest hematopoietic stem cells.
BRIEF SUMMARY OF THE INVENTION
[0006] The present disclosure provides methods using a compound which includes VLA-4 antagonists in combination with a first and second agent which interacts with a chemokine (such as CXCR2 agonist and a CXCR4 inhibitor) including methods of use and methods of treatment therewith. Also, provided herein are compositions comprising these three drugs.
[0007] Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description. Note that simply because a particular compound is ascribed to one particular generic formula does not mean that it cannot also belong to another generic formula.
BRIEF DESCRIPTION OF THE FIGURES
[0008] The application file contains at least one drawing executed in color. Copies of this patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
[0009] The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure. The disclosure may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
[0010] FIG. 1 depicts a graph showing CFC mobilization by firategrast. B6 mice were treated with AMD3100 (5mg/kg sc), firategrast (100 mg/kg IV), and G-CSF (250 mg/kg/d χ 5 days) alone or in combination. *P < 0.05, **P < 0.01 , ***P < 0.001 .
[001 1 ] FIG. 2 depicts a graph showing CFC mobilization by firategrast + Gro-βί. DBA/2 mice were treated with firategrast (100 mg/kg IV) and Gro-βί (2.5 mg/kg) alone or in combination, n = 5. **P < 0.01 , ***P < 0.001
[0012] FIGS. 3A & 3B show HSPC mobilization and transplantation. (FIG. 3A) DBA/2 mice (n=3/cohort) were treated and blood CFC and LSK numbers were determined 0.5 h post-injection. (FIG. 3B) B6 (CD45.2+) mice were treated and blood was collected 0.5 h post-injection. Mobilized blood (pooled from n=4/cohort) were co- transplanted with 2.5x105 competitor BM (CD45.1/2+) into lethally irradiated recipients (CD45.1 +, n=10/mobilizing agent). The frequency of repopulating units (RU) was calculated based on the contribution within the B-cell lymphoid and myeloid fraction 20 weeks after transplant. *P < 0.05, **P < 0.01 .
[0013] FIG. 4A shows the results of the colony forming cell (CFC) assay with the integrin antagonist administered in combination with Gropt. The top graph shows the number of colonies formed as a function of time while the bottom graph shows the number of CFC units at a specific time point for each mouse. These results show the results for Example Compound 35 (light gray) and Example Compound 36 (dark gray). FIG. 4B shows the results of the colony forming cell (CFC) assay with only the VLA-4 inhibitor. The top graph shows the number of colonies formed as a function of time while the bottom graph shows the number of CFC units at a specific time point for each mouse. These results show the results for Example Compound 35 (light gray) and Example Compound 36 (dark gray).
[0014] FIG. 5 shows the effects of blocking VLA4 with CXCR2 stimulation results in the mobilization of both LSK and CFU-C cells in DBA2/J mice, n = 3 ** P<0.01 **P<0.05 2-way ANOVA with Sidak post-hoc correction.
[0015] FIGS. 6A & 6B show pharmacokinetics (n = 3) (FIG. 6A) and pharmacodynamics (n = 4-6) (FIG. 6B) of different methods of administering the agents in DBA2/J mice.
[0016] FIGS. 7A & 7B show the results of repopulation assay show that the mobilized blood results in functional HPSC from either primary (n = 8-18) or secondary recipients (n = 5-10) in BALB/C mice. FIG. 7A shows the study design.
[0017] FIGS. 8A-8C show the results of order of administration of the agents. FIG. 8A shows that similar results were obtained when the VLA-4 inhibitor was administered before tGro-β. Similar results were obtained for the opposite combination wherein tGro-β was administered before the VLA-4 inhibitor (FIG. 8B). *** P<0.001 The highest CFU-C was seen when the agents were administered simultaneously as shown in FIG. 8C. *** P<0.001 ** P<0.001 t-test with Holm-Sidak correlation, n =4-5 and conducted in BALB/C mice.
[0018] FIGS. 9A-9H show that compositions containing tGro-β resulted in the induction of MMP9 proteolytic activity in blood (FIG. 9A-9C) in BALB/C mice (n = 3). Myeloid cells express both CXCR2 and MMP9 (FIG. 9D-9E) and may serve as a key cell type for HSPC mobilization in BALB/C mice, (flow cytometry n =4; real time PCR n = 3-9). FIG. 9F-9H shows that GR1 + myeloid cells are required to mobilize HSPCs in BALB/C mice, n = 7-9 * P<0.05. [0019] FIG. 10 shows the combination of a VLA-4 inhibitor and tGro-β results in similar mobilization regardless of the presence of other factors which result in poor stem cell mobilization such as diabetes.
[0020] FIG. 11 A-11 B graphically depicts the effects of the triple
combination of combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist.
[0021 ] FIG. 12A-12E shows independence of VLA4 and CXCR4 blockade induced mobilization. FIG. 12A-12B graphically depicts the CFU of vehicle treated and anti-vla-4 treated CXCR4 +/+ and CXCR4 -/- mice. FIG. 12C shows the CFU in wt or cxcr4 f/f mice treated with AMD3100, GroB, CWHM-823 or control. FIG. 12D-12E show LSK/CFU-C per mL of peripheral blood.
[0022] FIG. 13A-13C show the effects of the triple combination of combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist. FIG. 13A shows increased CFU-C per mL, FIG. 13B shows increased WBC per mL and FIG. 13C shows increased NE per mL of peripheral blood in triple therapy treated mice compared to the other conditions.
DETAILED DESCRIPTION OF THE INVENTION
[0023] Disclosed herein are compositions and methods comprising a first agent which interacts with a chemokine, such as a CXCR2 agonist, a second agent which interacts with a chemokine, such as, a CXCR4 inhibitor, and a compound that act as integrin antagonists or inhibitor, such as α4β1 integrin (VLA-4) antagonists as well as compositions thereof. These compositions may result in the mobilization of progenitor and/or stem cells from bone marrow to peripheral circulation. Additionally, provided herein are methods for the treatment and/or prevention of disease using these two therapeutic agents in combination. These compositions described herein may be used to stimulate progenitor and/or stem cells and result in such stimulation in a shorter amount of time relative to either agent alone or other known agents or combinations. These compositions may also have the added advantage that they result in the mobilization in higher numbers, begin mobilization in a shorter period of time, over a more prolonged period of time, or mobilize increased numbers of early progenitor and/or stem cells, LSK-SLAM cells, CFU-C cells, or other progenitor and/or stem cells which are competent to achieve a successful engraftment into the patient. In some
embodiments, these compositions may be used in improving the harvest of
hematopoietic stem cells or progenitor cells. These methods, compositions, or uses are described in more detail below.
I. Compounds and Synthetic Methods
[0024] In some embodiments, the compounds used in the compositions of the present disclosure include the compounds described in the Examples and claims listed below. All the synthesis methods described above can be further modified and optimized using the principles and techniques of organic chemistry as applied by a person skilled in the art. Such principles and techniques are taught, for example, in March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure (2007), which is incorporated by reference herein.
[0025] Compounds employed in methods of the disclosure may contain one or more asymmetrically-substituted carbon or nitrogen atoms, and may be isolated in optically active or racemic form. Thus, all chiral, diastereomeric, racemic form, epimeric form, and all geometric isomeric forms of a structure are intended, unless the specific stereochemistry or isomeric form is specifically indicated. Compounds may occur as racemates and racemic mixtures, single enantiomers, diastereomeric mixtures and individual diastereomers. In some embodiments, a single diastereomer is obtained. The chiral centers of the compounds of the present disclosure can have the S or the R configuration, as defined by the lUPAC 1974 Recommendations. For example, mixtures of stereoisomers may be separated using the techniques taught in the
Examples section below, as well as modifications thereof. Tautomeric forms are also included as well as pharmaceutically acceptable salts of such isomers and tautomers.
[0026] Atoms making up the compounds of the present disclosure are intended to include all isotopic forms of such atoms. Compounds of the present disclosure include those with one or more atoms that have been isotopically modified or enriched, in particular those with pharmaceutically acceptable isotopes or those useful for pharmaceutically research. Isotopes, as used herein, include those atoms having the same atomic number but different mass numbers. By way of general example and without limitation, isotopes of hydrogen include deuterium and tritium, and isotopes of carbon include 13C and 14C. Similarly, it is contemplated that one or more carbon atom(s) of a compound of the present disclosure may be replaced by a silicon atom(s). Furthermore, it is contemplated that one or more oxygen atom(s) of a compound of the present disclosure may be replaced by a sulfur or selenium atom(s).
[0027] Compounds of the present disclosure may also exist in prodrug form. Since prodrugs are known to enhance numerous desirable qualities of
pharmaceuticals (e.g. , solubility, bioavailability, manufacturing, etc.), the compounds employed in some methods of the disclosure may, if desired, be delivered in prodrug form. Thus, the disclosure contemplates prodrugs of compounds of the present disclosure as well as methods of delivering prodrugs. Prodrugs of the compounds employed in the disclosure may be prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Accordingly, prodrugs include, for example, compounds described herein in which a hydroxy, amino, or carboxy group is bonded to any group that, when the prodrug is administered to a subject, cleaves to form a hydroxy, amino, or carboxylic acid, respectively. Additional details regarding prodrugs may be found in Smith and Williams, 1988, the entire contents of which are hereby incorporated by reference. Smith and Williams Introduction to the Principles of Drug Design, Smith, H. J.; Wright, 2nd ed., London (1988).
[0028] It should be recognized that the particular anion or cation forming a part of any salt of this disclosure is not critical, so long as the salt, as a whole, is pharmacologically acceptable. Compounds useful in the disclosure which are amines, may be administered or prepared in the forms of their acid addition salts or metal complexes thereof. Suitable acid addition salts include salts of inorganic acids that are biocompatible, including HCI, HBr, sulfuric, phosphoric and the like, as well as organic acids such as acetic, propionic, butyric and the like, as well as acids containing more than one carboxyl group, such as oxalic, glutaric, adipic and the like. Compounds useful in the disclosure that are carboxylic acids or otherwise acidic may be administered or prepared in forms of salts formed from inorganic or organic bases that are physiologically compatible. Thus, these compounds may be prepared in the forms of their sodium, potassium, calcium, or magnesium salts as appropriate or may be salts with organic bases such as caffeine or ethylamine. These compounds also may be in the form of metal complexes. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are presented in Handbook of Pharmaceutical Salts: Properties, and Use (2002), which is incorporated herein by reference.
[0029] It should be further recognized that the compounds of the present disclosure include those that have been further modified to comprise substituents that are convertible to hydrogen in vivo. This includes those groups that may be convertible to a hydrogen atom by enzymological or chemical means including, but not limited to, hydrolysis and hydrogenolysis. Examples include hydrolyzable groups, such as acyl groups, groups having an oxycarbonyl group, amino acid residues, peptide residues, o-nitrophenylsulfenyl, trimethylsilyl, tetrahydropyranyl, diphenylphosphinyl, and the like. Examples of acyl groups include formyl, acetyl, trifluoroacetyl, and the like. Examples of groups having an oxycarbonyl group include ethoxycarbonyl, fe/f-butoxycarbonyl (-C(0)OC(CH3)3, Boc), benzyloxycarbonyl, p-methoxybenzyloxycarbonyl,
vinyloxycarbonyl, p-(p-toluenesulfonyl)ethoxycarbonyl, and the like. Suitable amino acid residues include, but are not limited to, residues of Gly (glycine), Ala (alanine), Arg (arginine), Asn (asparagine), Asp (aspartic acid), Cys (cysteine), Glu (glutamic acid), His (histidine), lie (isoleucine), Leu (leucine), Lys (lysine), Met (methionine), Phe
(phenylalanine), Pro (proline), Ser (serine), Thr (threonine), Trp (tryptophan), Tyr (tyrosine), Val (valine), Nva (norvaline), Hse (homoserine), 4-Hyp (4-hydroxyproline), 5-Hyl (5-hydroxylysine), Orn (ornithine) and β-Ala. Examples of suitable amino acid residues also include amino acid residues that are protected with a protecting group. Examples of suitable protecting groups include those typically employed in peptide synthesis, including acyl groups (such as formyl and acetyl), arylm ethoxycarbonyl groups (such as benzyloxycarbonyl and p-nitrobenzyloxycarbonyl), fe/f-butoxycarbonyl groups (-C(0)OC(CH3)3, Boc), and the like. Suitable peptide residues include peptide residues comprising two to five amino acid residues. The residues of these amino acids or peptides can be present in stereochemical configurations of the D-form, the L-form or mixtures thereof. In addition, the amino acid or peptide residue may have an
asymmetric carbon atom. Examples of suitable amino acid residues having an asymmetric carbon atom include residues of Ala, Leu, Phe, Trp, Nva, Val, Met, Ser, Lys, Thr and Tyr. Peptide residues having an asymmetric carbon atom include peptide residues having one or more constituent amino acid residues having an asymmetric carbon atom. Examples of suitable amino acid protecting groups include those typically employed in peptide synthesis, including acyl groups (such as formyl and acetyl), arylmethoxycarbonyl groups (such as benzyloxycarbonyl and p-nitrobenzyloxycarbonyl), fe/f-butoxycarbonyl groups (-C(0)OC(CH3)3), and the like. Other examples of substituents "convertible to hydrogen in vivo" include reductively eliminable
hydrogenolyzable groups. Examples of suitable reductively eliminable
hydrogenolyzable groups include, but are not limited to, arylsulfonyl groups (such as o- toluenesulfonyl); methyl groups substituted with phenyl or benzyloxy (such as benzyl, trityl and benzyloxymethyl); arylmethoxycarbonyl groups (such as benzyloxycarbonyl and o-methoxy-benzyloxycarbonyl); and haloethoxycarbonyl groups (such as β,β,β- trichloroethoxycarbonyl and β-iodoethoxycarbonyl).
[0030] Compounds of the disclosure may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. , higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the indications stated herein or otherwise.
[0031 ] In some aspects, the present disclosure relates to compositions containing one or more VLA-4 inhibitors and one or more agents which interacts with a chemokine such as a CXCR2 agonist, a CXCR4 inhibitor, or G-CSF. In some aspects, the present disclosure relates to compositions containing one or more VLA-4 inhibitors and at least two agents which interact with a chemokine. In some aspects, the present disclosure relates to compositions containing one or more VLA-4 inhibitors, at least one CXCR4 inhibitor and at least one CXCR2 agonist. These compositions may further comprise an excipient such as solvent or diluent which renders the composition suitable for administration via injection. In some embodiments, the components of these compositions may be formulated independently and then administered simultaneously to a patient. In other embodiments, these compositions are formulated with additional therapeutic agents or excipients. In other embodiments, these compositions consists substantially of, consists essentially of, or consists of one or more VLA-4 inhibitors, one or more agents which interact with a chemokine, and one or more excipients. Each of the compositions described herein contain a pharmaceutically effective amount of each of these agents combined. In particular, the compositions may contain a
pharmaceutically effective combined amount of a VLA-4 inhibitor, a CXCR4 inhibitor and a CXCR2 agonist. The pharmaceutically effective combined amount results when each agent is present in an amount such that the effect of the combination results in increased activity relative to a similar amount of a single agent. In some embodiments, the effect of the combination results in an additive increase in activity. In some embodiments, the effect of the combination results in synergistic activity.
[0032] These compositions may be used in a variety of indications such as the mobilization of hematopoietic stem cells or progenitor cells. These indications include elevating the number of progenitor and/or stem cells which are circulating in the patient especially elevating the number of these cells in the peripheral blood of a patient. Alternatively, these compositions may be used to treat a patient with cancer including sensitizing the patient to a chemotherapy and/or radiotherapy, for the treatment of hematopoietic cancer such as leukemias, myelomas, or lymphoma, or for the harvesting of hematopoietic progenitor and/or stem cells which may be transplanted into a patient who has impaired production of hematopoietic progenitor and/or stem cells. A patient may have impaired production of hematopoietic progenitor and/or stem cells resulting from a high dose of chemotherapy, radiotherapy, another therapeutic agent, or a genetic abnormality. Alternatively, the compositions described herein may be used to mobilize pre-cancerous or cancerous cells from the bone marrow into the peripheral blood. In some embodiments, the mobilization of pre-cancerous or cancerous cells from the bone marrow is used to potentiate or increase the effects of a standard cancer therapy such as a chemotherapeutic and/or radiotherapy.
Furthermore, each of these compositions may be used in the manufacture of
medicament for these indications.
[0033] The compositions and methods described herein may include one or more additional agents that are therapeutically or nutritionally useful such as antibiotics, vitamins, herbal extracts, anti-inflammatories, glucose, antipyretics, analgesics, cyclophosphamide, recombinant stem cell factor (Stemgen®), granulocyte- macrophage colony stimulating factor (GM-CSF) (such as Leukine®, and Leucomax®), ETRX-101 , TLK 199/TILENTRA™, lnterleukin-1 (IL-1 ), lnterleukin-3 (IL-3), lnterleukin-8 (IL-8), PIXY-321 (GM-CSF/IL-3 fusion protein), macrophage inflammatory protein, thrombopoietin, or a similar agent. Additionally, the compositions may contain one or more agents prevents microbial growth to increase the storage of the composition. Such agents may be an anti-parasitic, an antifungal, an antibiotic, or anti-viral.
Additionally, the compositions may further comprise one or more chemotherapeutic agents.
(a) VLA-4 Inhibitors
[0034] The methods and compositions used herein may contain one or more VLA-4 inhibitors or VLA-4 antagonists. The terms VLA-4 inhibitor and VLA-4 antagonist are used interchangeably in the disclosure. Some non-limiting examples of VLA-4 inhibitors which may be used in the compositions and methods described herein include antibodies, such as humanized monoclonal antibody against a4, natalizumab (Antegren®) and small molecules such as those described in U.S. Pat. No. 5,510,332; WO 06/023396; WO 97/03094; WO 97/02289; WO 96/40781 ; WO 96/22966; WO 96/20216; WO 96/01644; WO 96/06108; WO 95/15973; WO 96/31206; WO 06/010054; WO 05/087760; WO 01/12186; WO 99/37605; WO 01/51487; WO 03/01 1288; WO 02/14272; WO 01/32610; and EP 0842943, the entire contents of which are hereby incorporated by reference. An example of a VLA-4 inhibitor that may be used herein is BIO5192 (also known as AMD15057) disclosed in PCT publication WO 01/12186, which is incorporated herein by reference. Alternatively, analogs of BIO5192, such as BI0121 1 , may be used. In other embodiments, the VLA-4 inhibitor is firategrast or a harmaceutically acceptable salt thereof. Firategrast is a compound of the formula:
Figure imgf000013_0001
wherein:
Ri is hydrogen, aminocarbonyl, carboxy, cyano, halo, hydroxy, -SF5, alkyl(c<8), substituted alkyl(C≤8), alkoxy(C<8), substituted alkoxy(C<8), or -Y-i -Ra; wherein:
Yi is alkanediyl(c<8) or substituted alkanediyl(c<8); and
Ra is alkoxy(c<i2), acyloxy(c<i2), or a substituted version of either of these groups; or
-X(CH20)m-(CH2CH20)n-Rb; wherein:
X is a covalent bond or -0-;
m is 0 or 1 ;
n is 1 , 2, 3, 4, 5, 6, 7, or 8; and
Rb is hydrogen, alkyl C≤8), or substituted alkyl(C<8);
R2 is hydrogen, aminocarbonyl, carboxy, cyano, halo, hydroxy, -SF5, alkyl(c<8), substituted alkyl(C≤8), alkoxy(c<8), substituted alkoxy(c<8), or -Y2~RC; wherein:
Y2 is alkanediyl(c<8) or substituted alkanediyl(C<8); and Rc is alkoxy(C<i2), acyloxy(C<i2), or a substituted version of either of these groups;
Xi is hydroxy, alkoxy(c<8), substituted alkoxy(c<8), cycloalkoxy(C<8), substituted cycloalkoxy(c<8), alkenyloxV(c<8), substituted alkenyloxV(c<8), aryloxy(c<8), substituted aryloxy(c<8), aralkoxy(c<8), substituted aralkoxy(c<8), or a substituent convertible in vivo to hydrogen; and
R3 and R4 are each independently hydrogen, hydroxy, alkoxy(C<8) or substituted alkoxy(C<8);
R5 is hydrogen, -CH(ORd)Re, or -C(0)Rf, wherein:
Rd is hydrogen, alkyl C≤8), or substituted alkyl(C<8); and
Re and Rf are each independently alkyl(C<8) or substituted alkyl(C<8); and
Z is a group of the formula:
Figure imgf000014_0001
wherein:
p is O, 1 , 2, or 3;
R6 is hydrogen or -C(0)X2; wherein:
X2 is amino, hydroxy, alkoxV(c<8), substituted alkoxV(c<8), cycloalkoxy(c<8), substituted cycloalkoxy(C<8), alkenyloxy(C<8), substituted alkenyloxy(C<8), aryloxy(C<8), substituted aryloxy(C<8), aralkyloxy(C<8), substituted aralkyloxy(C<8), alkylaminO(C<8), substituted alkylaminO(c<8), dialkylaminO(C<8), substituted dialkylaminO(C<8), cycloalkylaminO(c<8), substituted cycloalkylamino(c<8), alkenylaminO(c<8), substituted alkenylaminO(C<8), arylamino(c<8), substituted arylaminO(C<8), aralkylaminO(C<8), substituted aralkylaminO(c<8), or a substituent convertible in vivo to hydrogen; R7 and R8 are each independently hydrogen, halo, haloalkyl(C substituted haloalkyl(C<8);
R9 is hydrogen, alkyl(c<8), or substituted alkyl(c<8); or
group of the formula:
Figure imgf000015_0001
wherein:
W is hydrogen, cyano, halo, hydroxy, or -C(0)X3, wherein:
X3 is amino, hydroxy, alkoxy(c<8), substituted alkoxy(c<8),
cycloalkoxy(c<8), substituted cycloalkoxy(C<8), alkenyloxV(C<8), substituted alkenyloxV(C<8), aryloxy(C<8), substituted aryloxy(C<8), aralkyloxy(c<8), substituted aralkyloxy(c<8), alkylaminO(c<8), substituted alkylaminO(C<8), dialkylaminO(C<8), substituted
dialkylaminO(c<8), cycloalkylaminO(C<8), substituted cycloalkylaminO(c<8), alkenylaminO(C<8), substituted alkenyl aminos), arylaminO(C<8), substituted arylaminO(C<8),
aralkylaminO(c<8), substituted aralkylaminO(C<8), or a substituent convertible in vivo to hydrogen;
R-io and Rn are each independently hydrogen or halo;
or a pharmaceutically acceptable salt thereof.
An Agent Which Interacts with a Chemokine
[0036] As used herein, the "agent which interacts with a chemokine:
includes chemokines, cytokines, chemokine receptors, or an agent which modulates the activity of these molecules such as a fragment, an antibody, or a small organic molecule. As used herein "interacts with" means that the agent binds with a chemokine in a manner that modulates the activity of said chemokine, for example, by reducing, inhibiting, increasing or activating the activity of the chemokine. In one embodiment, the present disclosure relates to compositions which modulate the activity of a CXC chemokine receptor such as CXCR2 or CXCR4. In some embodiments, the present methods and compositions comprise at least two agents which interact with a chemokine. In some embodiments, the present methods and compositions comprise a first agent comprising aCXCR2 agonist and a second agent comprising a CXCR4 inhibitor. In a preferred embodiment, the methods and compositions comprise a VLA-4 inhibitor or VLA-4 antagonist, a CXCR4 inhibitor and a CXCR2 agonist. In a particular embodiment, the methods and compositions comprise a VLA-4 inhibitor or VLA-4 antagonist, AMD3100 and Grop.
[0037] CXCR2 agonists include any molecule that activates the CXCR2 receptor. Such molecules include chemokines, cytokines, agonist antibodies or biologically active fragments thereof, or small organic molecules. Some non-limiting examples of chemokines acting via the CXCR2 receptor include, but are not limited to Grop, Groa, Groy, GCP-2 (granulocyte chemo-attractant protein 2), IL-8, NAP-2
(neutrophil activating peptide 2), ENA-78 (epithelial-cell derived neutrophil activating protein 78), and MGSA.
[0038] In some embodiments, the CXCR2 agonists used in the methods and compositions described herein are Grop and modified forms thereof. King et al., Blood 2001 ; 97: 1534-1542 have demonstrated that a recombinant /V-terminal 4-amino acid truncated form of the human chemokine Grop (also known as SB-251353 or garnocestim or Gropt) can mobilize progenitor cells after administration of SB-251353 in combination with G-CSF. This combination resulted in the mobilization of neutrophils and platelets during these studies. Chemokines such as the SB-251353, Groa, Grop, and Groy are further discussed in WO 94/29341 ; WO 97/15594; WO 97/15595; WO 99/26645; WO 02/02132; U.S. Pat. No. 6,080,398; U.S. Pat. No. 6,399,053; and U.S. Pat. No. 6,447,766, which are incorporated herein by reference.
[0039] The "Grop", "Grop protein", or "Grop chemokine" class includes Grop itself as well as modified forms of Grop. These modified forms include, but are not limited to, truncated, multimerized, amino-acid substituted, modified with amino-acid deletions and/or insertions, or combinations thereof. "Modified forms of Grop" includes truncated forms such as those described in U.S. patents 6,447,766; 6,399,053;
6,080,398; PCT publication 99/26645; PCT publication WO 97/15595; PCT publication WO 02/02132; PCT publication WO 97/15594; and PCT publication WO 94/29341 , which are incorporated herein by reference. "Modified forms of Grop" are multimeric forms of Grop such as dimers, trimers, tetramers, or other versions containing multiple proteins or modified proteins. Some non-limiting examples of "modified forms" include modified forms of Grop with truncation of between 2 to about 8 amino acids at the amino terminus of the mature protein, truncation of between about 2 to about 10 amino acids at the carboxy terminus of the mature protein, or multimeric forms of the modified and/or truncated proteins, e.g. , dimers, trimers, tetramers and other aggregated forms. Some non-limiting examples of truncated forms of Grop may include SB-251353 which consists of amino acids 5-73 and forms thereof where amino acid 69 is deamidated.
[0040] Another specific CXCR2 receptor agonist that may be used in the compositions and methods described herein is SB-251353, a basic, heparin-binding protein with a molecular mass of approximately 7500 Da (King et al., J Immunol 2000; 164: 3774-3782, Hepburn et al., Journal of Pharmacology and Experimental
Therapeutics 2001 ; 298: 886-893).
[0041 ] The compositions and methods described herein may comprise one or more CXCR4 inhibitors. Some non-limiting examples of CXCR4 inhibitors include AMD3100 (plerixafor), AMD3465, CTCE-0214, CTCE-9908, CP-1221 (linear peptides, cyclic peptides, natural amino-acids, unnatural amino acids, and
peptidomimetic compounds), T140 and analogs, 4F-benzoyl-TN24003, KRH-1 120, KRH-1636, KRH-2731 , polyphemusin analogue, ALX40-4C, or a CXCR4 inhibitors described in WO 01/85196, WO 99/50461 , WO 01/94420, WO 03/090512, US
2005/0059702, US 2005027767, US 2003/9229341 , US 5021409, US 6001826, and US 5583131 , each of which is incorporated by reference herein.
[0042] The compositions or methods described herein may be comprise G-CSF. It is contemplated that any suitable source of G-CSF may be employed. The G- CSF used in the compositions or methods may be either recombinant or purified using known techniques and includes, but is not limited to, Neupogen® filgrastim (Amgen), Neutrogin®/Granocyte® lenograstim (Chugai Pharmaceuticals), and Neulasta® pegylated filgrastim (Amgen). Additionally, biologically active fragments, variants, derivatives or fusion proteins may also be employed provided these agents retain the ability to mobilize progenitor or stem cells.
II. Biological Activity
[0043] It is another object of the disclosure to provide pharmaceutical compositions comprising compounds described above. These compounds and pharmaceutical compositions may be used to improve the harvest of hematopoietic stem cells or progenitor cells. Additionally, the compounds or compositions may be used to elevate the circulation of hematopoietic progenitor and/or stem cells, improve the collection of hematopoietic stem cells or progenitor cells for a transfusion, increase the sensitization of an anti-cancer therapy such as a chemotherapeutic or radiotherapy, or mobilize pre-cancerous or cancerous cells into the peripheral blood which may increase their sensitivity to an anti-cancer therapy.
[0044] Hematopoietic stem cell transplant (HSCT) is used to facilitate repopulation of healthy bone marrow and immune system cells after high-dose chemotherapy treatment for cancers such as Hodgkin's and non-Hodgkin's lymphoma, multiple myeloma, and leukemia. In HSCT, hematopoietic stem/progenitor cells
(HSPCs) are collected from the patient's blood, harvested, frozen and then stored while the patient receives high-dose chemotherapy and/or radiation therapy. Successful HSCT requires the intravenous infusion of a minimum number of 2 χ 106 CD34+ stem cells/kg body weight; however, a dose of 5 χ 106 CD34+ cells/kg is considered preferable for early and long term multilineage engraftment.
[0045] Stem cells harvested from peripheral blood are the most commonly used graft source in HSCT. While granulocyte colony-stimulating factor (G-CSF) is the most frequently used agent for stem cell mobilization, the use of G-CSF alone results in suboptimal stem cell yields in a significant proportion of patients. Plerixafor (AMD3100), a small molecule CXCR4 antagonist, in combination with G-CSF increases total CD34+ HSPCs compared to G-CSF alone and is FDA approved for stem cell mobilization in Non-Hodgkin's lymphoma and multiple myeloma. However, a significant disadvantage of plerixafor is cost, adding $25,567 per patient compared to G-CSF alone.
Furthermore, up to 24% of patients receiving plerixafor and G-CSF still fail to collect >2 χ 106 CD34+ cells/kg in 4 days of apheresis. Recent economic analysis has determined that reducing apheresis by 1 day has the potential to decrease medical costs by $6,600. Thus improved/alternative mobilizing agents and strategies are needed.
[0046] Mechanistic studies have shown that the integrin α4β1 (VLA-4) plays an important role in the retention of HSPCs within the bone marrow (BM) microenvironment. HSPC mobilization has been achieved by disrupting the integrin α4β1Λ/ΟΑΜ-1 axis with antibodies against integrin α4β1 or VCAM-1. Preclinical mouse studies in the DiPersio laboratory have shown that administration of the small molecule inhibitor of integrin α4β1 , BI05192, results in the rapid and reversible mobilization of HSPCs into the peripheral circulation with maximum mobilization occurring within 30 to 60 minutes and returning to baseline within 4 hours. A superior treatment could be envisioned wherein a patient receives an integrin α4β1 antagonist to continually inhibit integrin α4β1 over the course of ~4 hours (average duration of CD34+ stem cell apheresis procedures), maximizing the mobilization of HSPCs that can be collected by apheresis during the same day of treatment.
[0047] BI05192 is a potent small molecule inhibitor of integrin α4β1 and has demonstrated efficacy in mobilizing HSPCs in mice. However, BI05192 has poor aqueous solubility, bioavailability, and pharmacokinetic properties and therefore has not been developed clinically. A simpler, more soluble integrin α4β1 antagonist is firategrast. Firategrast has been tested in clinical trials for the treatment of multiple sclerosis and has demonstrated efficacy in mobilizing HSPCs in mice but to a significantly lesser extent than BI05192 and at much higher doses. Thus, development of integrin α4β1 antagonist which exhibit improved properties would greatly improve the clinical use of this particular combination.
[0048] Integrin antagonist compounds with high VLA-4 binding affinity and in vivo efficacy similar to B 105192 but with superior pharmacokinetic and
physiochemical properties are expected to increase mobilization of HSPCs alone or in a synergistic manner with G-CSF, plerixafor, truncated Gro-beta (Gro-βί) or other mobilization agents and thus remarkably improve the current HSCT regimen. These compositions or methods may also have the added advantage that the compositions or methods result in the mobilization in higher numbers, begin mobilization in a shorter period of time, over a more prolonged period of time, or mobilize increased numbers of early progenitor and/or stem cells, LSK-SLAM cells, CFU-C cells, or other progenitor and/or stem cells which are competent to achieve a successful engraftment into the patient. For example, the number of progenitor and/or stem cells mobilized when using the combination or methods described herein may be at least about 1 .2-fold, at least about 1 .5-fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7-fold, at least about 7.5-fold, at least about 8-fold, at least about 8.5-fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 1 1 -fold, at least about 12-fold, at least about 13-fold, at least about 14-fold, or at least about 15-fold greater then when using a single agent alone. Specifically, the number of early progenitor and/or stem cells (e.g. LSK-SLAM cells) mobilized when using the combination of at least one VLA-4 inhibitor and at least one CXCR2 agonist is at least about 1 .2-fold, at least about 1 .5- fold, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-fold, at least about 6.5-fold, at least about 7-fold, at least about 7.5-fold, at least about 8-fold, at least about 8.5-fold, at least about 9-fold, at least about 9.5-fold, at least about 10-fold, at least about 1 1 -fold, at least about 12-fold, at least about 13-fold, at least about 14-fold, at least about 15-fold, at least about 16-fold, at least about 17-fold, at least about 18-fold, at least about 19-fold, at least about 20-fold, at least about 21 -fold, at least about 22-fold, at least about 23-fold, at least about 24- fold, or at least about 25-fold greater then when using a single agent alone.
III. Therapeutic Methods
[0049] In one aspect, this disclosure provides methods of inhibiting or antagonizing VLA-4 using one or more of the compounds disclosed herein, as well as pharmaceutical compositions thereof containing one or more VLA-4 antagonists in the presence of two or more agents that interact with a chemokine, such as Gro-β,
AMD3100, or a derivatives thereof. The therapeutic methods described herein may be used to enhance or elevate the circulation of hematopoietic progenitor and/or stem cells. These therapeutic methods may be used to improve stem cell transplantation, tissue repair, improve the efficacy of cancer therapy, or other situations in which in vivo stimulation of hematopoiesis is desirable. The compositions or methods described herein wherein the VLA-4 inhibitor and agents which interact with a chemokine combine to act synergistically to induce rapid mobilization of progenitor and stem cells. For example, peak mobilization, when these combined therapeutic agents are used, may occur at about 10 minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, or about 6 hours after administration of the combination. In one embodiment, these compositions or methods result in a composition which acts synergistically to induce rapid mobilization of progenitor and stem cells with peak mobilization at about 15 minutes after administration of the combination. In contrast, this mobilization is significantly shorter than the 4-5 days needed to achieve maximum mobilization using G-CSF.
[0050] In one aspect, the methods and compositions described herein may be used to increase the harvest of HSPCs for a variety of different applications. These compositions and methods may be used to treat a patient who requires a
transplantation. Alternatively, the compositions and methods may be used to treat a patient who does not require a transplantation. The patient who needs a transplant of HSPCs requires either an allogenic, autologous, or tandem transplant of HSPCs. In some embodiments, the HSPCs may be used in either allogenic or autologous transplants. In another aspect, the present methods and compositions described herein may be used to improve the circulation of cells to tissues which need repair. The increased circulation of HSPCs may be used to improve the repair of the target tissue in the patient.
[0051 ] If the HSPCs are harvested, these cells may be returned to the donor patient (autologous transplant) or may be donated to another patient that is sufficiently compatible to prevent rejection (allogeneic transplant). One non-limiting application of autologous transplantation is in combination with radiation or chemotherapy in patients bearing tumors since the radiotherapeutic or
chemotherapeutic methods deplete the patient's normal cells. In this application, the patients cells may be harvested prior to or during the therapeutic treatments,
fractionated if necessary, cultured and optionally expanded, and then returned to the patient to restore the damaged immune system depleted by the therapy. Allogeneic recipients may receive the cells for the same purpose, or may have a condition that may be benefited by enhancing their hematopoietic systems. In a typical protocol for these types of transplants, the mobilized cells are collected from the donor by, for example, apheresis and then stored/cultured/expanded/fractionated as desired. In some embodiments, the compositions or methods described herein may result in the need for aphersis being eliminated.
[0052] In some aspects, the present compositions and methods described herein may be used to increase the circulation of pre-cancerous or cancerous cells out of the bone marrow into the peripheral blood. Without wishing to be bound by any theory, it is believed that the increasing the circulation of pre-cancerous or cancerous cells out of the bone marrow may increase the effectiveness of an anti-cancer therapy. In particular, these methods or compositions may be used to treat patients who have or are at risk of a hematopoietic malignancy such as lymphoma, myeloma, or leukemia. The compositions or methods described herein may be administered or employed prior to, during, or subsequent to the anti-cancer therapy. Two non-limiting examples of anticancer therapies that may be used in the methods described herein or conjunction with the compositions described herein include chemotherapeutic agents or radiotherapy.
[0053] In another aspect, the compositions and methods described herein may be used to decrease inflammation which may result in increasing tissue repair. Thus, the compositions or methods described herein may be used to treat graft versus host disease. Additionally, these compositions or methods may be used to treat diseases or disorders associated with cell adhesion-mediated inflammatory pathways. Some non-limiting examples of cell adhesion-mediated inflammatory pathologies include asthma, multiple sclerosis, rheumatoid arthritis, atherosclerosis, and
inflammatory bowel disease. [0054] The pharmaceutical compositions of these combinations may further comprise one or more non-toxic, pharmaceutically acceptable carriers and/or diluents and/or adjuvants (collectively referred to herein as "carrier" materials) and if desired other active ingredients. These methods may be used to treat a blood disease or disorder such as sickle cell anemia or as a part of hematopoietic stem cell therapy to promote the development of stem cells. In some embodiments, the compound is administered as part of a pharmaceutical composition further comprising a
pharmaceutically acceptable carrier. In some embodiments, the compounds and/or pharmaceutical compositions thereof may be administered orally, parenterally, or by inhalation spray, or topically in unit dosage formulations containing conventional pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used herein includes, for example, subcutaneous, intravenous, intramuscular, intrasternal, infusion techniques or intraperitoneally. In some embodiments, the compounds of the present disclosure are administered by any suitable route in the form of a pharmaceutical composition adapted to such a route, and in a dose effective for the treatment intended. Therapeutically effective doses of the compounds required to prevent or arrest the progress of or to treat a medical condition are readily ascertained by one of ordinary skill in the art using preclinical and clinical approaches familiar to the medicinal arts.
[0055] Based upon standard laboratory experimental techniques and procedures well known and appreciated by those skilled in the art, as well as
comparisons with compounds of known usefulness, the compounds described above can be used in the treatment of patients suffering from the above pathological conditions. One skilled in the art will recognize that selection of the most appropriate compound of the disclosure is within the ability of one with ordinary skill in the art and will depend on a variety of factors including assessment of results obtained in standard assay and animal models.
[0056] In several aspects of the present disclosure, the compounds provided herein may be used in a variety of biological, prophylactic or therapeutic areas, including those in wherein VLA-4 plays a role. IV. Pharmaceutical Formulations and Routes of Administration
[0057] For administration to an animal especially a mammal in need of such treatment, the compounds in a therapeutically effective amount are ordinarily combined with one or more excipients appropriate to the indicated route of
administration. The compounds of the present disclosure are contemplated to be formulated in a manner ameniable to treatment of a veterinary patient as well as a human patient. In some embodiments, the veterinary patient may be an avian such as chicken, turkey, or duck, a companion animal such as a cat or dog, livestock animals such as a cow, horse, pig, or goat, zoo animals, and wild animals The compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol, and tableted or encapsulated for convenient administration. Alternatively, the compounds may be dissolved in water, polyethylene glycol, propylene glycol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, sodium chloride, and/or various buffers. Other excipients and modes of administration are well and widely known in the pharmaceutical art and may be adapted to the type of animal being treated. Description of potential
administration routes which may be used to formulate the compositions described herein include those taught in Remington's Pharmaceutical Sciences, latest edition, Mack Publishing Company, Easton, Pa., which is incorporated herein by reference.
[0058] The pharmaceutical compositions useful in the present disclosure may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional pharmaceutical carriers and excipients such as preservatives, stabilizers, wetting agents, emulsifiers, buffers, etc.
[0059] The compounds of the present disclosure may be administered by a variety of methods, e.g., orally or by injection (e.g. subcutaneous, intravenous, intraperitoneal, etc.). Depending on the route of administration, the active compounds may be coated in a material to protect the compound from the action of acids and other natural conditions which may inactivate the compound. They may also be administered by continuous perfusion/infusion of a disease or wound site.
[0060] In one aspect, the present methods or compositions may be administered such that the VLA-4 inhibitor is administered intravenously and the first and/ or second agent which interacts with a chemokine is administered subcutaneously. Alternatively, the VLA-4 inhibitor and the first and/or second agent which interacts with a chemokine may be administered subcutaneously. In still another embodiment, both the VLA-4 inhibitor and the first and/or second agent which interacts with a chemokine are administered subcutaneously in a single formulation. In some embodiments, the VLA-4 inhibitor and the first and/or second agent which interacts with a chemokine are administered as a single formulation subcutaneously or intravenously. The protocols for administration to a particular patient may be further optimized by a skilled practitioner.
[0061 ] To administer the therapeutic compound by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. For example, the therapeutic compound may be administered to a patient in an appropriate carrier, for example, liposomes, or a diluent. Pharmaceutically acceptable diluents include saline and aqueous buffer solutions. Liposomes include water-in-oil-in-water CGF emulsions as well as conventional liposomes.
[0062] The therapeutic compound may also be administered parenterally, intraperitoneally, intramuscularly, intraarterially, intraspinally, or intracerebrally.
Dispersions can be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
[0063] Pharmaceutical compositions may be suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or
dispersion. In all cases, the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (such as, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it may be useful to include isotonic agents, for example, sugars, sodium chloride, or polyalcohols such as mannitol and sorbitol, in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate or gelatin. For intravenous or parenteral administration, the compounds are formulated in suitable liquid form with excipients as required. The compositions may contain liposomes or other suitable carriers. For injection intravenously, the solution is made isotonic using standard preparations such as Hank's solution or other isotonic solutions.
[0064] Sterile injectable solutions can be prepared by incorporating the therapeutic compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the therapeutic compound into a sterile carrier which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the methods of preparation include vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., the therapeutic compound) plus any additional desired ingredient from a previously sterile- filtered solution thereof.
[0065] The therapeutic compound can be orally administered, for example, with an inert diluent or an assimilable edible carrier. The therapeutic compound and other ingredients may also be enclosed in a hard or soft shell gelatin capsule, compressed into tablets, or incorporated directly into the subject's diet. For oral therapeutic administration, the therapeutic compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. The percentage of the therapeutic compound in the compositions and preparations may, of course, be varied. The amount of the therapeutic compound in such therapeutically useful compositions is such that a suitable dosage will be obtained.
[0066] It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the disclosure are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding such a therapeutic compound for the treatment of a selected condition in a patient.
[0067] The therapeutic compound may also be administered topically to the skin, eye, or mucosa. Alternatively, if local delivery to the lungs is desired the therapeutic compound may be administered by inhalation in a dry-powder or aerosol formulation. Alternatively, the therapeutic agents may be administered transdermally.
[0068] Active compounds are administered at a therapeutically effective dosage sufficient to treat a condition associated with a condition in a patient. For example, the efficacy of a compound can be evaluated in an animal model system that may be predictive of efficacy in treating the disease in a human or another animal, such as the model systems shown in the examples and drawings.
[0069] An effective dose range of a therapeutic can be extrapolated from effective doses determined in animal studies for a variety of different animals. In general a human equivalent dose (HED) in mg/kg can be calculated in accordance with the following formula (see, e.g., Reagan-Shaw et al., FASEB J., 22(3):659-661 , 2008, which is incorporated herein by reference): [0070] HED (mg/kg) = Animal dose (mg/kg) χ (Animal Km/Human Km) [0071 ] Use of the Km factors in conversion results in more accurate HED values, which are based on body surface area (BSA) rather than only on body mass. Km values for humans and various animals are well known. For example, the Km for an average 60 kg human (with a BSA of 1 .6 m2) is 37, whereas a 20 kg child (BSA 0.8 m2) would have a Km of 25. Km for some relevant animal models are also well known, including: mice Km of 3 (given a weight of 0.02 kg and BSA of 0.007); hamster Km of 5 (given a weight of 0.08 kg and BSA of 0.02); rat Km of 6 (given a weight of 0.15 kg and BSA of 0.025) and monkey Km of 12 (given a weight of 3 kg and BSA of 0.24).
[0072] Precise amounts of the therapeutic composition depend on the judgment of the practitioner and are peculiar to each individual. Nonetheless, a calculated HED dose provides a general guide. Other factors affecting the dose include the physical and clinical state of the patient, the route of administration, the intended goal of treatment and the potency, stability and toxicity of the particular therapeutic formulation.
[0073] The actual dosage amount of a compound of the present disclosure or composition comprising a compound of the present disclosure administered to a subject may be determined by physical and physiological factors such as type of animal treated, age, sex, body weight, severity of condition, the type of disease being treated, previous or concurrent therapeutic interventions, idiopathy of the subject and on the route of administration. These factors may be determined by a skilled artisan. The practitioner responsible for administration will typically determine the concentration of active ingredient(s) in a composition and appropriate dose(s) for the individual subject. The dosage may be adjusted by the individual physician in the event of any
complication.
[0074] An effective amount typically will vary from about 0.0001 mg/kg to about 1000 mg/kg, from about 0.001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 750 mg/kg, from about 0.001 mg/kg to about 50 mg/kg, from about 100 mg/kg to about 500 mg/kg, from about 1 .0 mg/kg to about 250 mg/kg, from about 10.0 mg/kg to about 150 mg/kg, or from about 1 .0 mg/kg to about 15 mg/kg in one or more dose administrations daily, for one or several days (depending of course of the mode of administration and the factors discussed above). Other suitable dose ranges include 1 mg to 10000 mg per day, 100 mg to 10000 mg per day, 500 mg to 10000 mg per day, and 500 mg to 1000 mg per day. In some particular embodiments, the amount is less than 10,000 mg per day with a range of 750 mg to 9000 mg per day.
[0075] In some embodiments, an agent which interacts with a chemokine may be administered in an amount from about 1 mg/kg to about 10 mg/kg, or about 1 mg/kg to about 5 mg/kg, or about 2.5 mg/kg. The agents which interact with a chemokine may be Grop, a CXCR4 inhibitor or derivatives thereof. Similarly, the VLA-4 inhibitor may be administered in an amount from about 1 mg/kg to about 100 mg/kg, or about 1 mg/kg to about 50 mg/kg, or about 1 mg/kg to about 25 mg/kg, or about 1 mg/kg to about 15 mg/kg, or about 1 mg/kg to about 10 mg/kg, or about 1 mg/kg to about 5 mg/kg, or about 3 mg/kg. In some embodiments, a specific VLA-4 inhibitor such as a compound of formula I or a specific compound described in the examples such as firategrast or compound numbers 12, 21 , 22, 23, 24, 25, 26, 33, 34, 35, 36, 42, 43, 44, 45, or 49 may be administered in a range of about 1 mg/kg to about 200 mg/kg, or about 50 mg/kg to about 200 mg/kg, or about 50 mg/kg to about 100 mg/kg, or about 75 mg/kg to about 100 mg/kg, or about 100 mg/kg.
[0076] The effective amount may be less than 1 mg/kg/day, less than 500 mg/kg/day, less than 250 mg/kg/day, less than 100 mg/kg/day, less than 50 mg/kg/day, less than 25 mg/kg/day or less than 10 mg/kg/day. It may alternatively be in the range of 1 mg/kg/day to 200 mg/kg/day.
[0077] In other non-limiting examples, a dose may also comprise from about 1 microgram/kg/body weight, about 5 microgram/kg/body weight, about 10 microgram/kg/body weight, about 50 microgram/kg/body weight, about 100
microgram/kg/body weight, about 200 microgram/kg/body weight, about 350
microgram/kg/body weight, about 500 microgram/kg/body weight, about 1
milligram/kg/body weight, about 5 milligram/kg/body weight, about 10 milligram/kg/body weight, about 50 milligram/kg/body weight, about 100 milligram/kg/body weight, about 200 milligram/kg/body weight, about 350 milligram/kg/body weight, about 500 milligram/kg/body weight, to about 1000 mg/kg/body weight or more per administration, and any range derivable therein. In non-limiting examples of a derivable range from the numbers listed herein, a range of about 5 mg/kg/body weight to about 100 mg/kg/body weight, about 5 microgram/kg/body weight to about 500 milligram/kg/body weight, etc. , can be administered, based on the numbers described above.
[0078] In certain embodiments, a pharmaceutical composition of the present disclosure may comprise, for example, at least about 0.1 % of a compound of the present disclosure. In other embodiments, the compound of the present disclosure may comprise between about 1 % to about 75% of the weight of the unit, or between about 25% to about 60%, for example, and any range derivable therein.
[0079] Single or multiple doses of the agents are contemplated. Desired time intervals for delivery of multiple doses can be determined by one of ordinary skill in the art employing no more than routine experimentation. As an example, subjects may be administered two doses daily at approximately 12 hour intervals. In some
embodiments, the agent is administered once a day.
[0080] The agent(s) may be administered on a routine schedule. As used herein a routine schedule refers to a predetermined designated period of time. The routine schedule may encompass periods of time which are identical or which differ in length, as long as the schedule is predetermined. For instance, the routine schedule may involve administration twice a day, every day, every two days, every three days, every four days, every five days, every six days, a weekly basis, a monthly basis or any set number of days or weeks there-between. Alternatively, the predetermined routine schedule may involve administration on a twice daily basis for the first week, followed by a daily basis for several months, etc. In other embodiments, the disclosure provides that the agent(s) may taken orally and that the timing of which is or is not dependent upon food intake. Thus, for example, the agent can be taken every morning and/or every evening, regardless of when the subject has eaten or will eat.
V. Combination Therapies
[0081 ] The present disclosure relates to two or more agents used in combination with a VLA-4 antagonist. It is common in the field of mobilizing hematopoietic stem cells to combine therapeutic modalities. The present disclosure describes combinations of VLA-4 antagonists with other therapeutic modalities as combination therapies to increase the mobilization of hematopoietic stem cells.
[0082] To increase the mobilization of hematopoietic stem cells using the methods and compositions of the present disclosure, one would generally administer to the subject with a VLA-4 antagonist and at least two other therapies. These therapies would be provided in a combined amount effective to achieve an increased activity. This process may involve contacting the cells/subjects with the both agents/therapies at the same time, e.g. , using a single composition or pharmacological formulation that includes the agents, or by contacting the cell/subject with two or more distinct compositions or formulations, at the same time, wherein one composition includes the VLA-4 antagonist, one composition includes a first agent and another composition that includes a second agent.
[0083] Alternatively, the individual compounds in the compositions described herein may precede or follow the other compound treatment by time intervals ranging from seconds to days. One would generally ensure that a significant period of time did not expire between the time of each delivery, such that the therapies would still be able to exert an advantageously combined effect on the cell/subject. In such instances, it is contemplated that one would administer the modalities within about 12- 24 hours of each other, within about 6-12 hours of each other, with a delay time of only about 1 -2 hours, or less than 1 hour. Additionally, the agents which interact with a chemokine may be administered about 10-15 minutes, about 5-10 minutes, or about 0-5 minutes prior to administration of the VLA-4 inhibitor. For example, the agents which interact with a chemokine may be administered from about 15 minutes, about 14 minutes, about 13 minutes, about 12 minutes, about 1 1 minutes, about 10 minutes, about 9 minutes, about 8 minutes, about 7 minutes, about 6 minutes, about 5 minutes, about 4 minutes, about 3 minutes, about 2 minutes, to about 1 minute, or any range derivable therein before the VLA-4 inhibitor. Alternatively, the components may be administered at the same time. [0084] The compositions and combination of agents used in the methods described herein may be administered as a single bolus dose, a dose over time such as an infusion, as in intravenous, subcutaneous, or transdermal administration, or in multiple dosages. If infusion is used, the combination may be infused for about 15 minutes to about 6 hours. In one embodiment, the infusion may occur for the duration of length of the apheresis. Additionally, the compositions or combination may be administered once daily for multiple days including from 1 to 4 days.
[0085] Furthermore, the compositions or combinations may be
administered to the patient for one day or less than one day and then HSPCs isolated from the patient. The compositions or combinations described herein may be
administered and then HSPCs may be isolated about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, or about 8 hours following administration.
[0086] It also is conceivable that more than one administration of either the compound or the other therapy will be desired. Various combinations may be employed, where a compound of the present disclosure is "A," and the other compound or therapy is "B," as exemplified below:
A/B/A B/A/B B/B/A A/A/B B/A/A A/B/B B/B/B/A B/B/A/B
A/A/B/B A/B/A/B A/B/B/A B/B/A/A B/A/B/A B/A/A/B B/B/B/A
A/A/A/B B/A/A/A A/B/A/A A/A/B/A A/B/B/B B/A/B/B B/B/A/B
[0087] Other combinations are also contemplated. In some aspects of the present disclosure, the agent may be a CXC chemokine, a CXC chemokine receptor, or a derivative thereof. Some non-limiting examples of agent including Gro-β, truncated Gro-β (tGro-β), plerixafor (AMD3100), a granulocyte-colony stimulating factor (G-CSF) such as filgrastim, PEG-filgrastim, or lenograstim, or an inhibitor of integrin α9β1 such as BOP (/V-benzenesulfonyl)-L-prolyl-L-O-(1 -pyrrolidinylcarbonyl)tyrosine.
[0088] In other embodiments, the compositions or methods used herein may be administered with an anti-cancer therapy such as those described below. The methods or compositions described herein may be used in conjunction with standard methods or variations as practiced by a person of ordinary skill in the art. These anti- cancer agents may be administered prior to and/or concomitant with the compositions or methods described herein. Some non-limiting examples of anti-cancer therapies which may be used herein include carmustine, etoposide, cytarabine, melphalan, cyclophosphamide, busulfan, thiotepa, bleomycin, platinum (cisplatin), cytarabine, cyclophosphamide, buside, Cytoxan, daunorubicin, doxorubicin, agent ara-C,
cyclosporin; Rituxan®; thalidomide; clofarabine; Velcade®; Antegren®; Ontak®;
Revlimid® (thalidomide analog); Prochymal™; Genasense° (oblimersen sodium);
Gleevec™; Glivec® (imatinib); tamibarotene; nelarabine; gallium nitrate; PT-100;
Bexxar®; Zevalin®; pixantrone; Onco-TCS; and agents that are topoisomerase inhibitors, or another specific anti-cancer therapy.
1. Chemotherapy
[0089] The term "chemotherapy" refers to the use of drugs to treat cancer. A "chemotherapeutic agent" is used to connote a compound or composition that is administered in the treatment of cancer. These agents or drugs are categorized by their mode of activity within a cell, for example, whether and at what stage they affect the cell cycle. Alternatively, an agent may be characterized based on its ability to directly crosslink DNA, to intercalate into DNA, or to induce chromosomal and mitotic aberrations by affecting nucleic acid synthesis. Most chemotherapeutic agents fall into the following categories: alkylating agents, antimetabolites, antitumor antibiotics, mitotic inhibitors, and nitrosoureas.
[0090] Examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan,
improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine,
triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC- 1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues);
cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1 -TM1 ); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine,
chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics {e.g., calicheamicin, especially calicheamicin γι and
calicheamicin ω-ι ; dynemicin, including dynemicin A; uncialamycin and derivatives thereof; bisphosphonates, such as clodronate; an esperamicin; as well as
neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores, aclacinomysins, actinomycin, authrarnycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-l-norleucine, doxorubicin (including
morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalarnycin, olivomycins, peplomycin,
potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, or zorubicin; anti-metabolites such as methotrexate and 5- fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti- adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as folinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins;
mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK polysaccharide complex); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol;
mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., paclitaxel and docetaxel; chlorambucil; gemcitabine; 6-thioguanine;
mercaptopurine; methotrexate; platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide;
mitoxantrone; vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g. , CPT-1 1 ); topoisomerase inhibitor RFS 2000; difluorometlhylornithine (DMFO); retinoids such as retinoic acid;
capecitabine; cisplatin (CDDP), carboplatin, procarbazine, mechlorethamine,
cyclophosphamide, camptothecin, ifosfamide, melphalan, chlorambucil, busulfan, nitrosurea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide (VP16), tamoxifen, raloxifene, estrogen receptor binding agents, taxol, paclitaxel, docetaxel, gemcitabien, navelbine, farnesyl-protein tansferase inhibitors, transplatinum, 5-fluorouracil, vincristin, vinblastin and methotrexate and
pharmaceutically acceptable salts, acids or derivatives of any of the above.
2. Radiotherapy
[0091 ] Radiotherapy, also called radiation therapy, is the treatment of cancer and other diseases with ionizing radiation. Ionizing radiation deposits energy that injures or destroys cells in the area being treated by damaging their genetic material, making it impossible for these cells to continue to grow. Although radiation damages both cancer cells and normal cells, the latter are able to repair themselves and function properly.
[0092] Radiation therapy used according to the present disclsoure may include, but is not limited to, the use of γ-rays, X-rays, and/or the directed delivery of radioisotopes to tumor cells. Other forms of DNA damaging factors are also
contemplated such as microwaves and UV-irradiation. It is most likely that all of these factors induce a broad range of damage on DNA, on the precursors of DNA, on the replication and repair of DNA, and on the assembly and maintenance of chromosomes. Dosage ranges for X-rays range from daily doses of 12.9 to 51 .6 mC/kg for prolonged periods of time (3 to 4 wk), to single doses of 0.516 to 1 .55 C/kg. Dosage ranges for radioisotopes vary widely, and depend on the half-life of the isotope, the strength and type of radiation emitted, and the uptake by the neoplastic cells.
[0093] Radiotherapy may comprise the use of radiolabeled antibodies to deliver doses of radiation directly to the cancer site (radioimmunotherapy). Antibodies are highly specific proteins that are made by the body in response to the presence of antigens (substances recognized as foreign by the immune system). Some tumor cells contain specific antigens that trigger the production of tumor-specific antibodies. Large quantities of these antibodies can be made in the laboratory and attached to radioactive substances (a process known as radiolabeling). Once injected into the body, the antibodies actively seek out the cancer cells, which are destroyed by the cell-killing (cytotoxic) action of the radiation. This approach can minimize the risk of radiation damage to healthy cells.
[0094] Conformal radiotherapy uses the same radiotherapy machine, a linear accelerator, as the normal radiotherapy treatment but metal blocks are placed in the path of the x-ray beam to alter its shape to match that of the cancer. This ensures that a higher radiation dose is given to the tumor. Healthy surrounding cells and nearby structures receive a lower dose of radiation, so the possibility of side effects is reduced. A device called a multi-leaf collimator has been developed and may be used as an alternative to the metal blocks. The multi-leaf collimator consists of a number of metal sheets which are fixed to the linear accelerator. Each layer can be adjusted so that the radiotherapy beams can be shaped to the treatment area without the need for metal blocks. Precise positioning of the radiotherapy machine is very important for conformal radiotherapy treatment and a special scanning machine may be used to check the position of internal organs at the beginning of each treatment.
[0095] High-resolution intensity modulated radiotherapy also uses a multi- leaf collimator. During this treatment the layers of the multi-leaf collimator are moved while the treatment is being given. This method is likely to achieve even more precise shaping of the treatment beams and allows the dose of radiotherapy to be constant over the whole treatment area. [0096] Although research studies have shown that conformal radiotherapy and intensity modulated radiotherapy may reduce the side effects of radiotherapy treatment, it is possible that shaping the treatment area so precisely could stop microscopic cancer cells just outside the treatment area being destroyed. This means that the risk of the cancer coming back in the future may be higher with these
specialized radiotherapy techniques.
[0097] Scientists also are looking for ways to increase the effectiveness of radiation therapy. Two types of investigational drugs are being studied for their effect on cells undergoing radiation. Radiosensitizers make the tumor cells more likely to be damaged, and radioprotectors protect normal tissues from the effects of radiation.
Hyperthermia, the use of heat, is also being studied for its effectiveness in sensitizing tissue to radiation.
3. Immunotherapy
[0098] In the context of cancer treatment, immunotherapeutics, generally, rely on the use of immune effector cells and molecules to target and destroy cancer cells. Trastuzumab (Herceptin™) is such an example. The immune effector may be, for example, an antibody specific for some marker on the surface of a tumor cell. The antibody alone may serve as an effector of therapy or it may recruit other cells to actually affect cell killing. The antibody also may be conjugated to a drug or toxin (chemotherapeutic, radionuclide, ricin A chain, cholera toxin, pertussis toxin, etc.) and serve merely as a targeting agent. Alternatively, the effector may be a lymphocyte carrying a surface molecule that interacts, either directly or indirectly, with a tumor cell target. Various effector cells include cytotoxic T cells and NK cells. The combination of therapeutic modalities, i.e. , direct cytotoxic activity and inhibition or reduction of ErbB2 would provide therapeutic benefit in the treatment of ErbB2 overexpressing cancers.
[0099] In one aspect of immunotherapy, the tumor cell must bear some marker that is amenable to targeting, i.e. , is not present on the majority of other cells. Many tumor markers exist and any of these may be suitable for targeting in the context of the present disclsoure. Common tumor markers include carcinoembryonic antigen, prostate specific antigen, urinary tumor associated antigen, fetal antigen, tyrosinase (p97), gp68, TAG-72, HMFG, Sialyl Lewis Antigen, MucA, MucB, PLAP, estrogen receptor, laminin receptor, erb B and p155. An alternative aspect of immunotherapy is to combine anticancer effects with immune stimulatory effects. Immune stimulating molecules also exist including: cytokines such as IL-2, IL-4, IL-12, GM-CSF, γ-IFN, chemokines such as MIP-1 , MCP-1 , IL-8 and growth factors such as FLT3 ligand.
Combining immune stimulating molecules, either as proteins or using gene delivery in combination with a tumor suppressor has been shown to enhance anti-tumor effects (Ju et al., 2000). Moreover, antibodies against any of these compounds may be used to target the anti-cancer agents discussed herein.
[0100] Examples of immunotherapies currently under investigation or in use are immune adjuvants e.g., Mycobacterium bovis, Plasmodium falciparum, dinitrochlorobenzene and aromatic compounds (U.S. Patents 5,801 ,005 and 5,739, 169; Hui and Hashimoto, 1998; Christodoulides, et ai, 1998), cytokine therapy, e.g., interferons α, β, and γ; IL-1 , GM-CSF and TNF (Bukowski, et ai , 1998; Davidson, et ai, 1998; Hellstrand, et ai, 1998) gene therapy, e.g., TNF, IL-1 , IL-2, p53 (Qin et ai, 1998; Austin-Ward and Villaseca, 1998; U.S. Patents 5,830,880 and 5,846,945) and monoclonal antibodies, e.g., anti-ganglioside GM2, anti-HER-2, anti-p185 (Pietras, et al., 1998; Hanibuchi, et ai , 1998; U.S. Patent 5,824,31 1 ). It is contemplated that one or more anti-cancer therapies may be employed with the gene silencing therapies described herein.
[0101 ] In active immunotherapy, an antigenic peptide, polypeptide or protein, or an autologous or allogenic tumor cell composition or "vaccine" is
administered, generally with a distinct bacterial adjuvant (Ravindranath and Morton, 1991 ; Morton, et ai, 1992; Mitchell, et ai, 1990; Mitchell, et ai , 1993).
[0102] In adoptive immunotherapy, the patient's circulating lymphocytes, or tumor infiltrated lymphocytes, are isolated in vitro, activated by lymphokines such as IL-2 or transduced with genes for tumor necrosis, and readministered (Rosenberg, et al., 1988; 1989).
VI. Definitions [0103] When used in the context of a chemical group: "hydrogen" means -H; "hydroxy" means -OH; "oxo" means =O; "carbonyl" means -C(=0)-; "carboxy" means -C(=O)OH (also written as -COOH or -CO2H); "halo" means independently -F, -CI, -Br or -I; "amino" means -NH2; "hydroxyamino" means -NHOH; "nitro" means -NO2; imino means =NH; "cyano" means -CN; "isocyanate" means -N=C=O; "azido" means -N3; in a monovalent context "phosphate" means -OP(O)(OH)2 or a
deprotonated form thereof; in a divalent context "phosphate" means -OP(O)(OH)O- or a deprotonated form thereof; "mercapto" means -SH; and "thio" means =S; "sulfonyl" means -S(O)2~; and "sulfinyl" means -S(O)-.
[0104] In the context of chemical formulas, the symbol "-" means a single bond, "=" means a double bond, and "≡" means triple bond. The symbol "— " represents an optional bond, which if present is either single or dou he symbol " represents a sin le bond double bond. Thus, the formula
Figure imgf000039_0001
covers, for example,
Figure imgf000039_0002
And it is understood that no one such ring atom forms part of more than one double bond. Furthermore, it is noted that the covalent bond symbol "-", when connecting one or two stereogenic atoms, does not indicate any preferred stereochemistry. Instead, it covers all stereoisomers as well as mixtures thereof. The symbol " ^ v " when drawn perpendicularly across a bond (e.g.,
Figure imgf000039_0003
attachment of the group. It is noted that the point of attachment is typically only identified in this manner for larger groups in order to assist the reader in unambiguously identifying a point of attachment. The symbol "-^ " means a single bond where the group attached to the thick end of the wedge is "out of the page." The symbol " Ί " means a single bond where the group attached to the thick end of the wedge is "into the page". The symbol " >ΛΛΛ " means a single bond where the geometry around a double bond (e.g., either E or Z) is undefined. Both options, as well as combinations thereof are therefore intended. Any undefined valency on an atom of a structure shown in this application implicitly represents a hydrogen atom bonded to that atom. A bold dot on a carbon atom indicates that the hydrogen attached to that carbon is oriented out of the plane of the paper.
[0105] When a group "R" is depicted as a "floating group" on a ring system, for example, in the formula:
Figure imgf000040_0001
[0106]
[0107] then R may replace any hydrogen atom attached to any of the ring atoms, including a depicted, implied, or expressly defined hydrogen, so long as a stable structure is formed. When a group "R" is depicted as a "floating group" on a fused ring system, as for example in the formula:
Figure imgf000040_0002
[0108]
[0109] then R may replace any hydrogen attached to any of the ring atoms of either of the fused rings unless specified otherwise. Replaceable hydrogens include depicted hydrogens (e.g. , the hydrogen attached to the nitrogen in the formula above), implied hydrogens (e.g., a hydrogen of the formula above that is not shown but understood to be present), expressly defined hydrogens, and optional hydrogens whose presence depends on the identity of a ring atom {e.g., a hydrogen attached to group X, when X equals -CH-), so long as a stable structure is formed. In the example depicted, R may reside on either the 5-membered or the 6-membered ring of the fused ring system. In the formula above, the subscript letter "y" immediately following the group "R" enclosed in parentheses, represents a numeric variable. Unless specified otherwise, this variable can be 0, 1 , 2, or any integer greater than 2, only limited by the maximum number of replaceable hydrogen atoms of the ring or ring system.
[01 10] For the chemical groups and compound classes, the number of carbon atoms in the group or class is as indicated as follows: "Cn" defines the exact number (n) of carbon atoms in the group/class. "C≤n" defines the maximum number (n) of carbon atoms that can be in the group/class, with the minimum number as small as possible for the group/class in question, e.g. , it is understood that the minimum number of carbon atoms in the group "alkenyl(c<8)" or the class "alkene(c<8)" is two. Compare with "alkoxy(c<io)", which designates alkoxy groups having from 1 to 10 carbon atoms. "Cn-n"' defines both the minimum (n) and maximum number (η') of carbon atoms in the group. Thus, "alkyl C2-i o>" designates those alkyl groups having from 2 to 10 carbon atoms. These carbon number indicators may precede or follow the chemical groups or class it modifies and it may or may not be enclosed in parenthesis, without signifying any change in meaning. Thus, the terms "C5 olefin", "C5-olefin", "olefin^s)", and
"olefinC5" are all synonymous. When any of the chemical groups or compound classes defined herein is modified by the term "substituted", any carbon atom(s) in a moiety replacing a hydrogen atom is not counted. Thus methoxyhexyl, which has a total of seven carbon atoms, is an example of a substituted alkyl(ci-6).
[01 1 1 ] The term "saturated" when used to modify a compound or chemical group means the compound or chemical group has no carbon-carbon double and no carbon-carbon triple bonds, except as noted below. When the term is used to modify an atom, it means that the atom is not part of any double or triple bond. In the case of substituted versions of saturated groups, one or more carbon oxygen double bond or a carbon nitrogen double bond may be present. And when such a bond is present, then carbon-carbon double bonds that may occur as part of keto-enol tautomerism or imine/enamine tautomerism are not precluded. When the term "saturated" is used to modify a solution of a substance, it means that no more of that substance can dissolve in that solution.
[01 12] The term "aliphatic" when used without the "substituted" modifier signifies that the compound or chemical group so modified is an acyclic or cyclic, but non-aromatic hydrocarbon compound or group. In aliphatic compounds/groups, the carbon atoms can be joined together in straight chains, branched chains, or non- aromatic rings (alicyclic). Aliphatic compounds/groups can be saturated, that is joined by single carbon-carbon bonds (alkanes/alkyl), or unsaturated, with one or more carbon-carbon double bonds (alkenes/alkenyl) or with one or more carbon-carbon triple bonds (alkynes/alkynyl). [01 13] The term "aromatic" when used to modify a compound or a chemical group refers to a planar unsaturated ring of atoms with An +2 electrons in a fully conjugated cyclic π system.
[01 14] The term "alkyl" when used without the "substituted" modifier refers to a monovalent saturated aliphatic group with a carbon atom as the point of
attachment, a linear or branched acyclic structure, and no atoms other than carbon and hydrogen. The groups -CH3 (Me), -CH2CH3 (Et), -CH2CH2CH3 (n-Pr or propyl), -CH(CH3)2 ( -Pr, 'Pr or isopropyl), -CH2CH2CH2CH3 (n-Bu), -CH(CH3)CH2CH3 (sec- butyl), -CH2CH(CH3)2 (isobutyl), -C(CH3)3 (fe/f-butyl, f-butyl, f-Bu or fBu), and
-CH2C(CH3)3 (neo-pentyl) are non-limiting examples of alkyl groups. The term
"alkanediyl" when used without the "substituted" modifier refers to a divalent saturated aliphatic group, with one or two saturated carbon atom(s) as the point(s) of attachment, a linear or branched acyclic structure, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen. The groups -CH2- (methylene), -CH2CH2-, -CH2C(CH3)2CH2-, and -CH2CH2CH2- are non-limiting examples of alkanediyl groups. The term "alkylidene" when used without the "substituted" modifier refers to the divalent group =CRR' in which R and R' are independently hydrogen or alkyl. Non-limiting examples of alkylidene groups include: =CH2, =CH(CH2CH3), and =C(CH3)2. An
"alkane" refers to the class of compounds having the formula H-R, wherein R is alkyl as this term is defined above. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2,
-OC(O)CH3, -NHC(O)CH3, -S(O)2OH, or -S(O)2NH2. The following groups are non- limiting examples of substituted alkyl groups: -CH2OH, -CH2CI, -CF3, -CH2CN,
-CH2C(O)OH, -CH2C(O)OCH3, -CH2C(O)NH2, -CH2C(O)CH3, -CH2OCH3,
-CH2OC(O)CH3, -CH2NH2, -CH2N(CH3)2, and -CH2CH2CI. The term "haloalkyl" is a subset of substituted alkyl, in which the hydrogen atom replacement is limited to halo (i.e. -F, -CI, -Br, or -I) such that no other atoms aside from carbon, hydrogen and halogen are present. The group, -CH2CI is a non-limiting example of a haloalkyl. The term "fluoroalkyl" is a subset of substituted alkyl, in which the hydrogen atom replacement is limited to fluoro such that no other atoms aside from carbon, hydrogen and fluorine are present. The groups -CH2F, -CF3, and -CH2CF3 are non-limiting examples of fluoroalkyl groups.
[01 15] The term "cycloalkyi" when used without the "substituted" modifier refers to a monovalent saturated aliphatic group with a carbon atom as the point of attachment, said carbon atom forming part of one or more non-aromatic ring structures, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen. Non-limiting examples include: -CH(CH2)2 (cyclopropyl), cyclobutyl, cyclopentyl, or cyclohexyl (Cy). The term "cycloalkanediyl" when used without the "substituted" modifier refers to a divalent saturated aliphatic group with two carbon atoms as points of attachment, no carbon-carbon double or triple bonds, and no atoms other than carbon and hydrogen. The group
Figure imgf000043_0001
is a non-limiting example of cycloalkanediyl group. A "cycloalkane" refers to the class of compounds having the formula H-R, wherein R is cycloalkyi as this term is defined above. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3, -NHC(O)CH3, -S(O)2OH, or -S(O)2NH2.
[01 16] The term "alkenyl" when used without the "substituted" modifier refers to an monovalent unsaturated aliphatic group with a carbon atom as the point of attachment, a linear or branched, acyclic structure, at least one nonaromatic carbon- carbon double bond, no carbon-carbon triple bonds, and no atoms other than carbon and hydrogen. Non-limiting examples include: -CH=CH2 (vinyl), -CH=CHCH3,
-CH=CHCH2CH3, -CH2CH=CH2 (allyl), -CH2CH=CHCH3, and -CH=CHCH=CH2. The term "alkenediyl" when used without the "substituted" modifier refers to a divalent unsaturated aliphatic group, with two carbon atoms as points of attachment, a linear or branched, a linear or branched acyclic structure, at least one nonaromatic carbon- carbon double bond, no carbon-carbon triple bonds, and no atoms other than carbon and hydrogen. The groups -CH=CH- -CH=C(CH3)CH2- -CH=CHCH2- and -CH2CH=CHCH2- are non-limiting examples of alkenediyl groups. It is noted that while the alkenediyl group is aliphatic, once connected at both ends, this group is not precluded from forming part of an aromatic structure. The terms "alkene" and "olefin" are synonymous and refer to the class of compounds having the formula H-R, wherein R is alkenyl as this term is defined above. Similarly the terms "terminal alkene" and "a- olefin" are synonymous and refer to an alkene having just one carbon-carbon double bond, wherein that bond is part of a vinyl group at an end of the molecule. When any of these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2,
-C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3, -NHC(O)CH3, -S(0)2OH, or -S(O)2NH2. The groups -CH=CHF, -CH=CHCI and -CH=CHBr are non-limiting examples of substituted alkenyl groups.
[01 17] The term "aryl" when used without the "substituted" modifier refers to a monovalent unsaturated aromatic group with an aromatic carbon atom as the point of attachment, said carbon atom forming part of a one or more six-membered aromatic ring structure, wherein the ring atoms are all carbon, and wherein the group consists of no atoms other than carbon and hydrogen. If more than one ring is present, the rings may be fused or unfused. As used herein, the term does not preclude the presence of one or more alkyl or aralkyi groups (carbon number limitation permitting) attached to the first aromatic ring or any additional aromatic ring present. Non-limiting examples of aryl groups include phenyl (Ph), methylphenyl, (dimethyl)phenyl, -C6H4CH2CH3
(ethylphenyl), naphthyl, and a monovalent group derived from biphenyl. The term "arenediyl" when used without the "substituted" modifier refers to a divalent aromatic group with two aromatic carbon atoms as points of attachment, said carbon atoms forming part of one or more six-membered aromatic ring structure(s) wherein the ring atoms are all carbon, and wherein the monovalent group consists of no atoms other than carbon and hydrogen. As used herein, the term does not preclude the presence of one or more alkyl, aryl or aralkyi groups (carbon number limitation permitting) attached to the first aromatic ring or any additional aromatic ring present. If more than one ring is present, the rings may be fused or unfused. Unfused rings may be connected via one or more of the following: a covalent bond, alkanediyl, or alkenediyl groups (carbon number limitation permitting). Non-limiting examples of arenediyl groups include:
Figure imgf000045_0001
[01 19] An "arene" refers to the class of compounds having the formula H-R, wherein R is aryl as that term is defined above. Benzene and toluene are non- limiting examples of arenes. When any of these terms are used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2,
-OC(O)CH3, -NHC(O)CH3, -S(O)2OH, or -S(O)2NH2.
[0120] The term "aralkyl" when used without the "substituted" modifier refers to the monovalent group -alkanediyl-aryl, in which the terms alkanediyl and aryl are each used in a manner consistent with the definitions provided above. Non-limiting examples are: phenylmethyl (benzyl, Bn) and 2-phenyl-ethyl. When the term aralkyl is used with the "substituted" modifier one or more hydrogen atom from the alkanediyl and/or the aryl group has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3,
-NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3,
-NHC(O)CH3, -S(O)2OH, or -S(O)2NH2. Non-limiting examples of substituted aralkyls are: (3-chlorophenyl)-methyl, and 2-chloro-2-phenyl-eth-1 -yl.
[0121 ] The term "acyl" when used without the "substituted" modifier refers to the group -C(O)R, in which R is a hydrogen, alkyl, cycloalkyl, or aryl as those terms are defined above. The groups, -CHO, -C(O)CH3 (acetyl, Ac), -C(O)CH2CH3,
-C(O)CH(CH3)2, -C(O)CH(CH2)2, -C(O)C6H5, and -C(O)C6H4CH3 are non-limiting examples of acyl groups. A "thioacyl" is defined in an analogous manner, except that the oxygen atom of the group -C(0)R has been replaced with a sulfur atom, -C(S)R. The term "aldehyde" corresponds to an alkyl group, as defined above, attached to a -CHO group. When any of these terms are used with the "substituted" modifier one or more hydrogen atom (including a hydrogen atom directly attached to the carbon atom of the carbonyl or thiocarbonyl group, if any) has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3, -NHC(O)CH3, -S(O)2OH, or -S(O)2NH2. The groups, -C(O)CH2CF3, -CO2H (carboxyl), -CO2CH3 (methylcarboxyl), -CO2CH2CH3, -C(O)NH2 (carbamoyl), and -CON(CH3)2, are non-limiting examples of substituted acyl groups.
[0122] The term "alkoxy" when used without the "substituted" modifier refers to the group -OR, in which R is an alkyl, as that term is defined above. Non- limiting examples include: -OCH3 (methoxy), -OCH2CH3 (ethoxy), -OCH2CH2CH3, -OCH(CH3)2 (isopropoxy), -OC(CH3)3 (fe/f-butoxy), -OCH(CH2)2, -O-cyclopentyl, and -O-cyclohexyl. The terms "cycloalkoxy", "alkenyloxy", "aryloxy", "aralkoxy", and
"acyloxy", when used without the "substituted" modifier, refers to groups, defined as -OR, in which R is cycloalkyl, alkenyl, aryl, aralkyl, and acyl, respectively. The term "alkylthio" and "acylthio" when used without the "substituted" modifier refers to the group -SR, in which R is an alkyl and acyl, respectively. The term "alcohol" corresponds to an alkane, as defined above, wherein at least one of the hydrogen atoms has been replaced with a hydroxy group. The term "ether" corresponds to an alkane, as defined above, wherein at least one of the hydrogen atoms has been replaced with an alkoxy group. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3,
-NHC(O)CH3, -S(O)2OH, or -S(O)2NH2.
[0123] The term "alkylamino" when used without the "substituted" modifier refers to the group -NHR, in which R is an alkyl, as that term is defined above. Non- limiting examples include: -NHCH3 and -NHCH2CH3. The term "dialkylamino" when used without the "substituted" modifier refers to the group -NRR', in which R and R' can be the same or different alkyl groups, or R and R' can be taken together to represent an alkanediyl. Non-limiting examples of dialkylamino groups include: -N(CH3)2 and
-N(CH3)(CH2CH3). The terms "cycloalkylamino", "alkenylamino", "arylamino",
"aralkylamino", "alkoxyamino", and "alkylsulfonylamino" when used without the
"substituted" modifier, refers to groups, defined as -NHR, in which R is cycloalkyl, alkenyl, aryl, aralkyl, alkoxy, and alkylsulfonyl, respectively. A non-limiting example of an arylamino group is -NHC6H5. The term "amido" (acylamino), when used without the "substituted" modifier, refers to the group -NHR, in which R is acyl, as that term is defined above. A non-limiting example of an amido group is -NHC(0)CH3. The term "alkylimino" when used without the "substituted" modifier refers to the divalent group =NR, in which R is an alkyl, as that term is defined above. When any of these terms is used with the "substituted" modifier one or more hydrogen atom attached to a carbon atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3,
-N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3, -NHC(O)CH3, -S(O)2OH, or -S(O)2NH2. The groups -NHC(O)OCH3 and -NHC(O)NHCH3 are non- limiting examples of substituted amido groups.
[0124] The terms "alkylsulfonyl" and "alkylsulfinyl" when used without the "substituted" modifier refers to the groups -S(O)2R and -S(O)R, respectively, in which R is an alkyl, as that term is defined above. The terms "cycloalkylsulfonyl",
"alkenylsulfonyl", "alkynylsulfonyl", "arylsulfonyl", "aralkylsulfonyl", "heteroarylsulfonyl", and "heterocycloalkylsulfony are defined in an analogous manner. When any of these terms is used with the "substituted" modifier one or more hydrogen atom has been independently replaced by -OH, -F, -CI, -Br, -I, -NH2, -NO2, -CO2H, -CO2CH3, -CN, -SH, -OCH3, -OCH2CH3, -C(O)CH3, -NHCH3, -NHCH2CH3, -N(CH3)2, -C(O)NH2, -C(O)NHCH3, -C(O)N(CH3)2, -OC(O)CH3, -NHC(O)CH3, -S(O)2OH, or -S(O)2NH2.
[0125] The use of the word "a" or "an," when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one."
[0126] Throughout this application, the term "about" is used to indicate that a value includes the inherent variation of error for the device, the method being employed to determine the value, or the variation that exists among the study subjects.
[0127] An "active ingredient" (Al) (also referred to as an active compound, active substance, active agent, pharmaceutical agent, agent, biologically active molecule, or a therapeutic compound) is the ingredient in a pharmaceutical drug or a pesticide that is biologically active. The similar terms active pharmaceutical ingredient (API) and bulk active are also used in medicine, and the term active substance may be used for pesticide formulations.
[0128] The terms "comprise," "have" and "include" are open-ended linking verbs. Any forms or tenses of one or more of these verbs, such as "comprises,"
"comprising," "has," "having," "includes" and "including," are also open-ended. For example, any method that "comprises," "has" or "includes" one or more steps is not limited to possessing only those one or more steps and also covers other unlisted steps. As used herein, the term "consisting substantially of" means that the composition contains at least 98% of the elements listed. Similarly, the term "consisting essentially of" means that the composition contains at least 99% of the elements listed.
[0129] The term "derivative" when used in the context of a protein, such as a cytokine or chemokine, refers to a protein which is a recombinant version, a fragment, a truncated version, or a chemically modified protein. In some aspects, the chemically modified protein is a protein to which a secondary group such as a PEG chain
(pegylation) or other solubilizing polymer has been added to the protein.
[0130] The term "effective," as that term is used in the specification and/or claims, means adequate to accomplish a desired, expected, or intended result.
"Effective amount," "Therapeutically effective amount" or "pharmaceutically effective amount" when used in the context of treating a patient or subject with a compound means that amount of the compound which, when administered to a subject or patient for treating or preventing a disease, is an amount sufficient to effect such treatment or prevention of the disease.
[0131 ] An "excipient" is a pharmaceutically acceptable substance formulated along with the active ingredient(s) of a medication, pharmaceutical composition, formulation, or drug delivery system. Excipients may be used, for example, to stabilize the composition, to bulk up the composition (thus often referred to as "bulking agents," "fillers," or "diluents" when used for this purpose), or to confer a therapeutic enhancement on the active ingredient in the final dosage form, such as facilitating drug absorption, reducing viscosity, or enhancing solubility. Excipients include pharmaceutically acceptable versions of antiadherents, binders, coatings, colors, disintegrants, flavors, glidants, lubricants, preservatives, sorbents, sweeteners, and vehicles. The main excipient that serves as a medium for conveying the active ingredient is usually called the vehicle. Excipients may also be used in the
manufacturing process, for example, to aid in the handling of the active substance, such as by facilitating powder flowability or non-stick properties, in addition to aiding in vitro stability such as prevention of denaturation or aggregation over the expected shelf life. The suitability of an excipient will typically vary depending on the route of administration, the dosage form, the active ingredient, as well as other factors.
[0132] As used herein, "HSPCs" refers to hematopoietic stem and progenitor cells. HSPCs are a combination of progenitor cells and stem cells.
[0133] The term "hydrate" when used as a modifier to a compound means that the compound has less than one (e.g., hemihydrate), one (e.g., monohydrate), or more than one (e.g., dihydrate) water molecules associated with each compound molecule, such as in solid forms of the compound.
[0134] As used herein, the term "IC50" refers to an inhibitory dose which is 50% of the maximum response obtained. This quantitative measure indicates how much of a particular active ingredient or other substance (inhibitor) is needed to inhibit a given biological, biochemical or chemical process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half. [0135] An "isomer" of a first compound is a separate compound in which each molecule contains the same constituent atoms as the first compound, but where the configuration of those atoms in three dimensions differs.
[0136] As used herein, the term "patient" or "subject" refers to a living organism, such as a human, monkey, cow, sheep, goat, dog, cat, mouse, rat, guinea pig, avain, or transgenic species thereof. In certain embodiments, the patient or subject is a primate. Non-limiting examples of human patients are adults, juveniles, infants and fetuses.
[0137] As generally used herein "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of human beings and animals without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio.
[0138] "Pharmaceutically acceptable salts" means salts of compounds of the present invention which are pharmaceutically acceptable, as defined above, and which possess the desired pharmacological activity. Such salts include acid addition salts formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or with organic acids such as
1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, 2-naphthalenesulfonic acid,
3- phenylpropionic acid, 4,4'-methylenebis(3-hydroxy-2-ene-1 -carboxylic acid),
4- methylbicyclo[2.2.2]oct-2-ene-1 -carboxylic acid, acetic acid, aliphatic mono- and dicarboxylic acids, aliphatic sulfuric acids, aromatic sulfuric acids, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, carbonic acid, cinnamic acid, citric acid,
cyclopentanepropionic acid, ethanesulfonic acid, fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, glycolic acid, heptanoic acid, hexanoic acid,
hydroxynaphthoic acid, lactic acid, laurylsulfuric acid, maleic acid, malic acid, malonic acid, mandelic acid, methanesulfonic acid, muconic acid, o-(4-hydroxybenzoyl)benzoic acid, oxalic acid, p-chlorobenzenesulfonic acid, phenyl-substituted alkanoic acids, propionic acid, p-toluenesulfonic acid, pyruvic acid, salicylic acid, stearic acid, succinic acid, tartaric acid, tertiarybutylacetic acid, trimethylacetic acid, and the like. Pharmaceutically acceptable salts also include base addition salts which may be formed when acidic protons present are capable of reacting with inorganic or organic bases. Acceptable inorganic bases include sodium hydroxide, sodium carbonate, potassium hydroxide, aluminum hydroxide and calcium hydroxide. Acceptable organic bases include ethanolamine, diethanolamine, triethanolamine, tromethamine,
/V-methylglucamine and the like. It should be recognized that the particular anion or cation forming a part of any salt of this invention is not critical, so long as the salt, as a whole, is pharmacologically acceptable. Additional examples of pharmaceutically acceptable salts and their methods of preparation and use are presented in Handbook of Pharmaceutical Salts: Properties, and Use (P. H. Stahl & C. G. Wermuth eds., Verlag Helvetica Chimica Acta, 2002).
[0139] A "pharmaceutically acceptable carrier," "drug carrier," or simply "carrier" is a pharmaceutically acceptable substance formulated along with the active ingredient medication that is involved in carrying, delivering and/or transporting a chemical agent. Drug carriers may be used to improve the delivery and the
effectiveness of drugs, including for example, controlled-release technology to modulate drug bioavailability, decrease drug metabolism, and/or reduce drug toxicity. Some drug carriers may increase the effectiveness of drug delivery to the specific target sites. Examples of carriers include: liposomes, microspheres (e.g., made of poly(lactic-co- glycolic) acid), albumin microspheres, synthetic polymers, nanofibers, protein-DNA complexes, protein conjugates, erythrocytes, virosomes, and dendrimers.
[0140] A "pharmaceutical drug" (also referred to as a pharmaceutical, pharmaceutical preparation, pharmaceutical composition, pharmaceutical formulation, pharmaceutical product, medicinal product, medicine, medication, medicament, or simply a drug) is a compound or composition used to diagnose, cure, treat, or prevent disease. An active ingredient (Al) (defined above) is the ingredient in a pharmaceutical drug or a pesticide that is biologically active. The similar terms active pharmaceutical ingredient (API) and bulk active are also used in medicine, and the term active substance may be used for pesticide formulations. Some medications and pesticide products may contain more than one active ingredient. In contrast with the active ingredients, the inactive ingredients are usually called excipients (defined above) in pharmaceutical contexts.
[0141 ] As used herein, the term "pre-malignant cells" refers to cells that can form malignant hematopoietic or myeloid cells. The malignant hematopoietic or myeloid cells are those which characterize the conditions of myeloma, leukemia, and lymphoma. Particular forms of these diseases include acute myelitic leukemia (AML), acute lymphatic leukemia (ALL), multiple myeloma (MM), chronic myelogenous leukemia (CML), chronic lymphatic leukemia (CLL), hairy cell leukemia (HCL), acute promyelocytic leukemia (APL), and various lymphomas.
[0142] "Prevention" or "preventing" includes: (1 ) inhibiting the onset of a disease in a subject or patient which may be at risk and/or predisposed to the disease but does not yet experience or display any or all of the pathology or symptomatology of the disease, and/or (2) slowing the onset of the pathology or symptomatology of a disease in a subject or patient which may be at risk and/or predisposed to the disease but does not yet experience or display any or all of the pathology or symptomatology of the disease.
[0143] "Prodrug" means a compound that is convertible in vivo
metabolically into an active ingredient according to the present invention. The prodrug itself may or may not also have activity with respect to a given target protein. For example, a compound comprising a hydroxy group may be administered as an ester that is converted by hydrolysis in vivo to the hydroxy compound. Suitable esters that may be converted in vivo into hydroxy compounds include acetates, citrates, lactates, phosphates, tartrates, malonates, oxalates, salicylates, propionates, succinates, fumarates, maleates, methylene-bis- -hydroxynaphthoate, gentisates, isethionates, di-p-toluoyltartrates, methanesulfonates, ethanesulfonates, benzenesulfonates, p-toluenesulfonates, cyclohexylsulfamates, quinates, esters of amino acids, and the like. Similarly, a compound comprising an amine group may be administered as an amide that is converted by hydrolysis in vivo to the amine compound. [0144] The term "progenitor cells" as used herein refers to cells that, in response to certain stimuli, can form differentiated hematopoietic or myeloid cells. The presence of progenitor cells can be assessed by the ability of the cells in a sample to form colony-forming units of various types, including, for example, CFU-GM (colony- forming units, granulocyte-macrophage); CFU-GEMM (colony-forming units,
multipotential); BFU-E (burst-forming units, erythroid); HPP-CFC (high proliferative potential colony-forming cells); or other types of differentiated colonies which can be obtained in culture using known protocols such as those described below.
[0145] A "stereoisomer" or "optical isomer" is an isomer of a given compound in which the same atoms are bonded to the same other atoms, but where the configuration of those atoms in three dimensions differs. "Enantiomers" are
stereoisomers of a given compound that are mirror images of each other, like left and right hands. "Diastereomers" are stereoisomers of a given compound that are not enantiomers. Chiral molecules contain a chiral center, also referred to as a
stereocenter or stereogenic center, which is any point, though not necessarily an atom, in a molecule bearing groups such that an interchanging of any two groups leads to a stereoisomer. In organic compounds, the chiral center is typically a carbon, phosphorus or sulfur atom, though it is also possible for other atoms to be stereocenters in organic and inorganic compounds. A molecule can have multiple stereocenters, giving it many stereoisomers. In compounds whose stereoisomerism is due to tetrahedral stereogenic centers (e.g. , tetrahedral carbon), the total number of hypothetically possible
stereoisomers will not exceed 2n, where n is the number of tetrahedral stereocenters. Molecules with symmetry frequently have fewer than the maximum possible number of stereoisomers. A 50:50 mixture of enantiomers is referred to as a racemic mixture. Alternatively, a mixture of enantiomers can be enantiomerically enriched so that one enantiomer is present in an amount greater than 50%. Typically, enantiomers and/or diastereomers can be resolved or separated using techniques known in the art. It is contemplated that that for any stereocenter or axis of chirality for which stereochemistry has not been defined, that stereocenter or axis of chirality can be present in its R form, S form, or as a mixture of the R and S forms, including racemic and non-racemic mixtures. As used herein, the phrase "substantially free from other stereoisomers" means that the composition contains < 15%, more preferably < 10%, even more preferably < 5%, or most preferably < 1 % of another stereoisomer(s).
[0146] "Stem cells", as used herein, are less differentiated forms of progenitor cells. Typically, such cells are positive for CD34, but stem cells do not have to contain this marker. While other types of cells such as endothelial cells and mast cells also may exhibit this marker, CD34 is considered an one marker of stem cell presence. CD34+ cells can be assayed using fluorescence activated cell sorting (FACS) and thus their presence can be assessed in a sample using this technique. In general, CD34+ cells are present only in low levels in the blood, but are present in large numbers in bone marrow. Additionally, the stem cells may be hematopoietic stem cells that express the SLAM and LSK markers. Specifically, hematopoietic stem cells may be LSK cells or LSK-SLAM cells, which are considered early hematopoietic stem cells. The nomenclature, LSK, refers to Lin"Sca1 +c-Kit+ and SLAM is signaling lymphocyte activation molecules or Lin"CD41 "CD48"CD150+.
[0147] "Treatment" or "treating" includes (1 ) inhibiting a disease in a subject or patient experiencing or displaying the pathology or symptomatology of the disease (e.g., arresting further development of the pathology and/or symptomatology), (2) ameliorating a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease (e.g., reversing the pathology and/or symptomatology), and/or (3) effecting any measurable decrease in a disease in a subject or patient that is experiencing or displaying the pathology or symptomatology of the disease.
[0148] The above definitions supersede any conflicting definition in any of the reference that is incorporated by reference herein. The fact that certain terms are defined, however, should not be considered as indicative that any term that is undefined is indefinite. Rather, all terms used are believed to describe the disclosure in terms such that one of ordinary skill can appreciate the scope and practice the present disclosure. EXAMPLES
[0149] The following examples are included to demonstrate various embodiments of the present disclosure. It should be appreciated by those of skill in the art that the techniques disclosed in the examples that follow represent techniques discovered by the inventors to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Example 1 - Methods and Materials
A. In vitro assays of HSPC mobilization.
[0150] Colony Assays. In short-term colony-forming cell (CFC) assays, test samples are cultured in a semi-solid matrix supplemented with nutrients and cytokines for ~1 week at 37 °C. During this culture period, CFC proliferate and produce discrete cell clusters or colonies of morphologically recognizable daughter cells that can be quantified by light microscopy. Here, mice will be treated with mobilization regimens and peripheral blood will be collected via the retroorbital sinus at the peak of
mobilization. After collection of blood, a complete blood count will be performed using a Hemavet 950FS cell counter (Drew Scientific) and CFC assays will be established in methylcellulose-containing Iscove modified Dulbecco medium supplemented with interleukin-3, interleukin-6, and stem cell factor (MethoCult3534; StemCell
Technologies). CPU-GM, BFU-E, and CFU-GEMM colony types will be enumerated following 1 week of culture at 37 °C.
[0151 ] Flow cytometric enumeration of murine HSPCs. Among the subsets that define HSPCs, CD34- c-kit+ Sca-1 + lineage marker- (CD34-KSL) cells are regarded as one of the populations that have the highest enrichment of HSPCs in adult mouse bone marrow. More recently, the SLAM family markers, CD150 and CD48, have been used to differentiate stem cells from more committed progenitor cells. Here, primitive murine HSPC mobilization will be examined by flow cytometry enumeration of CD34-c-kit+Sca-1 +lineage marker-CD150+CD48- cell numbers following treatment with mobilization regimens.
B. In vivo assays of HSPC mobilization.
[0152] Competitive repopulation assays. The definitive assay for stem cell activity in a test sample is the complete and sustained (> 5 months) reconstitution of all hematopoietic lineages in irradiated recipients by transplanted HSPCs. For competitive stem cell repopulation assays, peripheral blood mononuclear cells (PBMCs) from wild- type C57BL/6 (CD45.2+) mice mobilized by the regimens described herein will be mixed with 2x105 competitor bone marrow cells from C57BL/6J x B6.SJL-Ptprca Pepcb/BoyJ F1 (CD45.1 +/CD45.2+) mice and transplanted into lethally irradiated (1 100 cGy) recipients congenic at the CD45.1 + locus (B6.SJL-Ptprca Pepcb/BoyJ). At least 3 mobilized PBMC:competitor ratios (typically the number of PBMCs isolated from 0.5, 1 , and 1 .5 ml_ of blood) will be evaluated to establish the appropriate donor: competitor ratio. Secondary transplantations will be performed by intravenously injecting 106 unfractionated bone marrow cells from donors 5 months after transplantation into lethally irradiated (1 100 cGy) CD45.1 + secondary recipients.
C. In vitro assay of chemosensitization.
[0153] Stromal protections assay. The impact of bone marrow stroma on leukemia cell survival during treatments listed herein will be assessed. Mouse AML (APL) and human ALL (G2) cell lines will be incubated with the appropriate drugs and drug combinations ± chemotherapy ± stroma. BLI (bioluminescence) as well as standard MTT assays will be used to assess cell growth and survival.
D. In vivo assay of chemosensitization.
[0154] Recipient mice (C57BL/6) will be injected with 1 χ 106 murine acute promyelocytic (APL) leukemia cells transduced with Click Beetle Red-GFPe. In the absence of treatment all mice develop overt leukemia by day +20 and die from a rapidly fatal leukemia with leukocytosis and splenomegaly by day +35. The treatments disclosed herein will be administered with and without chemotherapy and mouse survival and leukemia burden as measured by BLI and FACS (Gr-1 +/c-kit+/CD34+ leukemia cells) will be assessed. Both BLI and FACS measurements will be obtained on days +7, +14, +21 , +28, and +35.
E. C57BL/6 into BALB/c transplant model.
[0155] Lethal irradiation of BALB/c recipients. Recipient BALB/c mice are lethally irradiated with 925 cGy of total body irradiation on day -1 and then infused with 5x106 T cell depleted bone marrow cells (TCD-BM) containing 2x106 T cells from C57BL/6 mice on day 0. Mice are monitored for indications of GvHD (clinical score based on weight loss, hunched posture, activity, fur texture, diarrhea and skin integrity) and survival time. Signs of GvHD are typically seen within 2-3 weeks and all mice die by 4-5 weeks. The rate of GvHD in irradiated mice receiving TCD-BM only (n= 15 mice) will be compared with the rate in mice receiving TCD-BM plus the different donor PBMC populations described herein. Mice will be sacrificed on day 25 to collect and examine GvHD target organs (histology of liver, intestines, and spleen). Peripheral blood will also be collected on day 25 to determine the extent of donor cell engraftment and immune reconstitution (flow cytometry using antibodies against CD3, B220, CD4, CD8, FOXP3, H-2Kb, CD45.1 , and CD45.2).
[0156] Sublethal irradiation of BALB/c recipients. Recipient BALB/c mice are sublethally irradiated with 500 cGy of total body irradiation on day and then infused with 5x106 T cell depleted bone marrow cells (TCD-BM) containing 2x106 T cells from C57BL/6 mice on day 0. Mice are monitored for indications of GvHD and donor engraftment as described above.
F. C57BL/6 into B6D2F1 transplant model.
[0157] Lethal irradiation of B6D2F1 recipients. Recipient B6D2F1 mice are lethally irradiated with 1 100 cGy (split doses, 3 hr apart) of total body irradiation on day - 1 and then infused with 1 χ10 T cell depleted splenocytes (TCD-SPL) containing 3x106 T cells from C57BL/6 mice on day 0. The rate of GvHD and donor engraftment in irradiated mice receiving TCD-SPL only (n= 15 mice) will be compared with the rate in mice receiving TCD-SPL plus the different purified T cell populations listed herein. Mice will be monitored for indications of GvHD and donor engraftment as described above. Example 2 - VLA-4 antagonists enhance mobilization of HSPCs. [0158] Treatment of mice with the VLA-4 antagonist BI05192 results in a 30-fold increase in mobilization of HSPCs and combination of BI05192 with plerixafor gave a further 3-fold increase. Treatment with a combination of BI05192, plerixafor, and G-CSF enhanced mobilization by 17-fold compared to G-CSF alone. BI05192 also mobilized long-term repopulating cells that successfully engraft and expand in a multi- lineage manner in secondary transplant experiments. Similar mobilization studies were performed using the small molecule VLA-4 antagonist firategrast. Similar results relative to BI05192 were obtained (FIG. 1 ). Although significant additive/synergistic effects were observed following combination therapy with firategrast and AMD3100 and/or G-CSF, 30-fold higher concentrations of firategrast were required to mimic the mobilization effects of BI05192 (100 mg/kg vs. 3 mg/kg, respectively). This decreased biological activity is presumably due to firategrast's ~30-fold lower potency for VLA-4.
[0159] A combination under investigation uses firategrast with the CXCR2 agonist truncated Gro-beta (Gro-βί, SB-251353). Gro-βί is a recombinant, N-terminal truncated form of Gro-β that binds to CXCR2 with greater potency than the full-length form of Gro-β. If both firategrast and Gro-βί are given to mice within 5 minutes of each other and blood is collected 15 min post-administration, a significant synergy in colony- forming cell (CFC) mobilization is observed that persisted for >2 hours (FIG. 2).
Remarkably, the level of CFC mobilization achieved with the firategrast and Gro-βί regimen is greater than that of G-CSF used alone for 4 days.
Example 3 - Combination of Grop or truncated Grop (Gropt) plus a VLA-4 small molecule inhibitor for hematopoietic stem cell mobilization and leukemia chemosensitization.
A. Mobilization of HSPCs with Gro- pt and firategrast
[0160] This experiment showed that the combination of Gro-βί (2.5 mg/kg, SC) and firategrast (VLA-4 inhibitor; 100 mg/kg, IV) synergistically mobilized murine HSPCs, as measured by a colony-forming unit (CFU) assay of mobilized peripheral blood. The combination of Gro-βί and firategrast mobilized 5.6-fold more murine CFUs (14,210 ± 843 CFU/mL blood) compared to when each agent was administered alone (Gro-pt: 2,520 ± 580 CFU/mL; firategrast: 2,480 ± 230 CFU/mL) in DBA/2 mice. This experiment was carried out again and similar results were obtained. In the second experiment, the combination of Gro-pt (2.5 mg/kg, SC) and firategrast (100 mg/kg, IV) mobilized 9.5-fold more murine CFUs (16,225 ± 3,600 CFUs) compared to when each agent was administered alone (Gro-pt: 1 ,710 ± 600 CFU/mL; firategrast: 1 ,205 ± 290 CFU/mL) in DBA/2 mice. To ensure the synergistic CFU mobilization that was observed with the combination of Gro-pt and firategrast in DBA/2 mice in the previous
experiments, the administration of these two agents was also tested in a different mouse strain. Specifically, the combination of Gro-pt (2.5 mg/kg, SC) and firategrast (100 mg/kg, IV) mobilized 7.3-fold more murine CFUs (4,600 ± 1 ,600 CFU/mL) compared to when each agent was administered alone (Gro-pt: 630 ± 390 CFU/mL; firategrast: 430 ± 130 CFU/mL) to C57BL/6 mice. Furthermore, the combination of Gro- pt and firategrast mobilized 18-fold more murine lineage-Sca-1 +c-kit+ (LSK) cells (72 LSK cells/ L blood), another measure of HSPCs, compared to when each agent was administered alone (Gro-pt: 4 LSK cells/pL; firategrast: 3 LSK cells/pL) to C57BL/6 mice.
Example 3 - Additional VLA-4 antagonists.
Preparation of Compounds
[0161 ] Table 1 below includes the characterization of the VLA-4 antagonist described herein
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
B. VCAM-1 Assay Results
[0162] Flow cytometry eel I -based assay. Compounds were tested for their ability to inhibit the binding of soluble VCAM-1 to human G2 acute lymphoblastic leukemia (ALL) cells. Briefly, G2 ALL cells are pre-incubated with increasing
concentrations (0.001 to 1000 nM) of compounds for 30 minutes. Soluble VCAM/Fc chimera protein (R&D systems) is then added to the mixture and the cells incubated for an additional 30 minutes. Afterwards, cells are washed and VCAM-1 is detected using PE-conjugated secondary mAbs. In each experiment, an aliquot of cells will be stained with isotype control mAbs to serve as a negative control. The percentage of VCAM-1 binding cells was then determined by flow cytometry.
Table 2: Inhibition of s VCAM-1 binding to human G2 ALL cells.
Figure imgf000070_0001
C. CFC Mobilization by Combination Therapies
[0163] Mice. DBA/2 J, C57BL/6J (CD45.2) and syngeneic B6.SJL-
Ptprca Pep3b/BoyJ (CD45.1 ) mice were purchased from the Jackson Laboratory (Bar Harbor, ME, USA). F1 -hybrid mice (CD45.1/2) were obtained through breeding CD45.2 and CD45.1 mice. Animals were housed at the Washington University Medical School vivarium under SPF conditions. All experiments were performed in accordance with the guidelines of the Washington University Animal Studies Committee and the institutional animal care and use committee (IACUC), in agreement with AAALAC guidelines.
Following lethal irradiation (1 χ 9.5 or 1 1 .0 Gy, using a 137Cesium source) and
transplantation, mice were kept on antibiotic medication, sulfamethoxazole and trimethoprim, 0.5 and 0.1 mg/ml_ respectively, (Hi-Tech Pharmacal, Amityville, NY, USA) peroral in drinking water.
[0164] HSPC mobilization. Recombinant human CXCL2 (Gro t; R&D systems) was reconstituted in sterile Ca+2/Mg+2-free phosphate buffered saline (PBS) and injected subcutaneously (SC) at a dose of 2.5 mg/kg. Firategrast (published structure: C27H27F2NO6; U.S. Patent Application No. 2014/051655) was dissolved in Ca+2/Mg+2-free PBS plus 1 % ethanol and injected intravenously at a dose of 100 mg/kg. RhG-CSF (Neupogen®, Filgrastim, Amgen, Thousand Oaks, CA, USA) diluted in PBS was injected i.p. every 12 hrs at a dose of 100 g/kg for a total of 9 (day 5) doses.
[0165] Colony forming cell (CFC) assay. Peripheral blood (PB) was drawn from the facial vein without anaesthesia into K/EDTA anti-coagulated tubes (Sarstedt AG & Co, Numbrecht, Germany). Red blood cells were removed from 25 μΙ_ aliquots of blood using hypotonic lysis (Ammonium-Chloride-Potassium, ACK buffer, 5- 10 min at RT) and samples were mixed with 2.5 ml_ methylcellulose media
supplemented with a cocktail of recombinant cytokines (MethoCult 3434; Stem Cell Technologies, Vancouver, BC, Canada). Cultures were plated in duplicate in 35 mm dishes and placed in a humidified chamber with 5% CO2 at 37 °C. After 7 d of culture, colonies containing at least 50 cells were counted using an inverted microscope in a blinded fashion.
[0166] Flow cytometric enumeration of murine HSPCs. Peripheral blood (PB) was drawn from the facial vein without anaesthesia into K/EDTA anti- coagulated tubes (Sarstedt AG & Co, Numbrecht, Germany). Cell counts (WBC) were determined using an automatic hemocytometer (Hemavet 950, Drew Scientific, Dallas, TX). Red blood cells were removed from 25 μΙ_ aliquots of blood using hypotonic lysis (Ammonium-Chloride-Potassium, ACK buffer, 5-10 min at RT). Samples were
resuspended in staining buffer (PBS supplemented with 0.5% bovine serum albumin and 2 mM EDTA) and incubated for 30 min at 4 °C with pre-titrated saturating dilutions of the following fluorochrome-labeled monoclonal antibodies (BD Biosciences, San Jose, CA; clone designated in parenthesis): Sca1 -PE (D7), CD1 17-BV421 (ACK2), lineage-APC (17A2, RB6-8C5, RA3-6B2, Ter-1 19, M/170) and CD45-A700 (30-F1 1 ). Dead cells were excluded from these assays by staining with 2 μg/mL 7-amino- actinomycin D (Molecular Probes, Eugene, OR) for 5 min prior to analysis. Appropriate isotype-matched negative controls were used to assess background fluorescence intensity. Samples were analyzed on a Beckman Coulter Gallios flow cytometer and data were analyzed using FlowJo software (TreeStar, Ashland, OR). Absolute numbers of immunophenotypically defined populations (e.g. LSK) were calculated by identifying CD45 positive cells as WBC from the corresponding cell count analysis. Statistical comparisons of flow cytometry data were performed using an unpaired two-tailed Student t-test (GraphPad Prism). P-values <0.05 were considered significant.
[0167] Repopulating unit (RU) assay. An RU assay was performed to directly compare the repopulating capacity of peripheral blood mobilized with different combinations of compounds. Lethally irradiated CD45.2 hosts received transplants consisting of 6 μΙ_ of blood from mobilized mice (CD45.1 +) together with 2.5x105 CD45.2+ BM competitor cells. After 20 weeks, blood graft-derived RUs were calculated for the recipient mice by the following formula: RU = (D*C)/(100-D) where D is the percentage of blood-derived B lymphocytes and myeloid cells and C is the number of RUs contained in the competitor BM fraction of the graft (C = 2.5 for 250,000 competitor BM cells). The results of these test are shown in FIG. 3 and FIG. 4.
Example 4 - Additional Studies of Combination of VLA-4 Inhibitors
[0168] Using the combination of VLA4 blockade with CXCR2 stimulation, both CFU-C and LSK cells were mobilized. See FIG. 5.
[0169] Additionally, the pharmacokinetics and pharmacodynamics of these combinations result in the fast mobilization of CFU-C per mL of peripheral blood. Three routes of administration were tested: subcutaneous, intraperitoneal, and intravenous administration. Subcutaneous and intraperitoneal showed higher CFU-C per mL of peripheral blood relative to intravenous administration and all routes of administration showed fast mobilization with the peak happening within 30 minutes of administration in DBA2/J mice. See FIGS. 6A & 6B.
[0170] As can be seen in FIGS. 7A & 7B, the repopulation unit assay shows that the mobilization methods described herein using a combination of a VLA4 antagonist and tGro-β result in mobilized stem cells which can be successfully transplanted in both primary and secondary recipients.
[0171 ] To determine if the order of administration matters, examples were the order of administration was varied. As can be seen in FIGS. 8A-8C, the highest number of colony forming units were obtained when the two agents were administered simultaneously. Without wishing to be bound by any theory, it is believed that CXCR2 stimulation primes the biological system for VLA4 blockage.
[0172] In order to determine the specific mechanism of action, studies were carried out to determine if MMP9 proteolytic activity was present in the
compositions. In FIG. 9A, compositions which contained tGro-β showed MMP9 activity. Using both flow cytometry and real-time PCR it was determined that myeloid cells express both MMP9 and CXCR2 and may play a role in the mobilization of the desired stem cells. See FIG. 9B. In an alternative example where GR1 + myeloid cells were removed through the use of a GR1 + antibody, the mobilization of CFU-C was greatly decreased relative to a control antibodies (FIG. 9C). Without wishing to be bound by any theory, it is believed that GR1 + myeloid cells play a role in stem cell mobilization.
[0173] Finally, studies were carried out to determine the effectiveness of the combination in conditions which are typically difficult to achieve mobilization such as diabetes. Using a streptozotocin induced diabetes model, the combination of a VLA-4 inhibitor and tGro-β resulted in similar mobilization regardless of the nature of the model animal (FIG. 10). Thus, the combinations described herein may be especially useful in conditions which are difficult to mobilize hematopoietic stem or progenitor cells.
Example 5 - Triple Combination Therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist
[0174] To test the mobilization efficacy of combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist BALB/c mice were treated with a variety of compositions. The mice were separated into seven groups: A) 3 mg/kg CWHM-823 (SC, n=5), B) 5 mg/kg AMD3100 (SC, n=5), C) 2.5 mg/kg tGrop (SC, n=5), D) 3 mg/kg CWHM-823 + 2.5 mg/kg tGrop (SC, n=5), E) 5 mg/kg AMD3100 + 2.5 mg/kg tGrop (SC, n=5), F) 5 mg/kg AMD3100 + 3 CWHM-823 (SC, n=5) and G) 3 mg/kg CWHM-823 + 5 mg/kg AMD3100 + 2.5 mg/kg tGrop (SC, n=5). On day 0, mice were inject mice as indicated above. Peripheral blood was collected from the mice at baseline, 0.25, 0.5, 1 , 2 and 4 hours post-injection. Aliquots of blood were removed and stored for the CFU assay and Hemavet. On day +7 CFUs were counted. As can be seen in FIG. 11A and FIG. 11 B, the triple combination of combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist provided significantly improved results over the other conditions tested.
[0175] To further test the combination therapy with VLA-4 inhibitor + CXCR4 inhibitor + CXCR2 agonist BL/6 mice were administered simultaneous subcutaneous injection of; A) CWHM-823 (3 mg/kg) + t-Grop (2.5 mg/kg), B) AMD3100 (5 mg/kg) + t-Grop (2.5 mg/kg), or C) CWHM-823 (3 mg/kg) + AMD3100 (5 mg/kg) + t- Grop (2.5 mg/kg). As can be seen in FIG. 13A-13B, the triple combination surprisingly outperforms compositions with a VLA-4 inhibitor and CXCR2 agonist or CXCR4 inhibitor and CXCR2 agonist.
[0176] All of the compounds, compositions, and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the disclosure may have focused on several embodiments or may have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations and modifications may be applied to the
compounds, compositions, and methods without departing from the spirit, scope, and concept of the invention. All variations and modifications apparent to those skilled in the art are deemed to be within the spirit, scope, and concept of the invention as defined by the appended claims. References
[0177] The following references to the extent that they provide exemplary procedural or other details supplementary to those set forth herein, are specifically incorporated herein by reference.
U.S. Patent Application No. 2014/0051655
Greene & Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley, 1999.
Handbook of Pharmaceutical Salts: Properties, and Use, Stahl and Wermuth (Eds.), Verlag Helvetica Chimica Acta, 2002.
March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, 2007.
Reagan-Shaw et ai, FASEB J., 22(3):659-661 , 2008

Claims

CLAIMS What is claimed is:
1 . A composition comprising:
a) a VLA-4 inhibitor compound;
b) a first agent which interacts with one or more chemokines; and c) a second agent which interact with one or more chemokines.
2. The composition according to any one of claims 1 , wherein the first agent and second agent which interact with a chemokine are selected from the group consisting of: an agent which interacts with a C-X-C chemokine or a C-X-C chemokine receptor.
3. The composition of claim 2, wherein the first agent is a CXCR2 agonist.
4. The composition of either claim 2 or claim 3, wherein the second agent is a CXCR4 inhibitor.
5. The composition of according to claim 3, wherein the agent is Gro-β, or a
derivative of Gro-β.
6. The composition of claim 5, wherein the derivative of Gro-β is a truncated Gro- β.
7. The composition of claim 4, wherein the CXCR4 inhibitor is one or more of AMD3100 (plerixafor), AMD3465, CTCE-0214, CTCE-9908, CP-1221 (linear peptides, cyclic peptides, natural amino-acids, unnatural amino acids, and peptidomimetic compounds), T140 and analogs, 4F-benzoyl-TN24003, KRH- 1 120, KRH-1636, KRH-2731 , polyphemusin analogue, ALX40-4C, or combinations thereof.
8. The composition of claim 1 , wherein the first agent inhibits the activity of the chemokine and the second agent activates the activity of the chemokine.
9. A composition comprising:
a) a VLA-4 inhibitor compound;
b) a CXCR4 inhibitor; and
c) a CXCR2 agonist.
10. A pharmaceutical composition comprising:
(a) a composition according to any one of claims 1 -9; and
(b) an excipient.
1 1 . The pharmaceutical composition of claim 10, wherein the pharmaceutical
composition is formulated for oral administration, intraarterial administration, intraperitoneal administration, intravenous administration, or subcutaneous administration.
12. The pharmaceutical composition of claim 10, wherein the pharmaceutical
composition is formulated for administration via intravenous infusion.
13. The pharmaceutical composition of claim 10, wherein the pharmaceutical
composition is formulated for administration via subcutaneous injection.
14. The pharmaceutical composition according to any one of claims 10-13, wherein the composition consists substantially of:
(a) the VLA-4 inhibitor;
(b) the first agent which interacts with one or more chemokines; (c) the second agent which interacts with one or more chemokines; and
(d) the excipient.
15. The pharmaceutical composition of claim 14, wherein the composition consists essentially of:
(a) the VLA-4 inhibitor;
(b) the first agent which interacts with one or more chemokines;
(c) the second agent which interacts with one or more chemokines; and
(d) the excipient.
16. A method of treating a disease or disorder in a patient comprising administering to the patient a therapeutically effective combined amount of:
(a) a VLA-4 inhibitor compound;
(b) a first agent which interacts with one or more chemokines; and
(c) a second agent which interact with one or more chemokines.
17. The method of claim 1 , wherein the first agent inhibits the activity of the chemokine and the second agent activates the chemokine activity.
18. The method of claim 1 , wherein the first agent includes a CXCR4 inhibitor and the second agent includes a CXCR2 agonist.
19. The method of claim 16, wherein the disease or disorder is associated with integrin α4β1 .
20. The method of claim 16, wherein the disease or disorder is associated with hematopoietic stem cells.
21 . The method of claim 16, wherein the hematopoietic stem cells are LSK-SLAM cells.
22. The method of claim 16, wherein the disease or disorder is cancer or a therapy for cancer which results in a reduced blood cell count.
23. The method of claim 16, wherein the disease or disorder is a therapy for cancer which results in a reduced blood cell count.
24. The method of claim 23, wherein the therapy is chemotherapy or radiation therapy.
25. The method of claim 22, wherein the disease or disorder is cancer.
26. The method of claim 25, wherein the patient is also administered a
chemotherapy or a radiotherapy.
27. The method of claim 26, wherein the effective combined amount of an agent which interacts with a chemokine and a VLA-4 inhibitor results in improved efficacy of the chemotherapy or radiotherapy.
28. A method of inducing the mobilization of hematopoietic stem cells or progenitor cells comprising contacting the hematopoietic stem cells with an effective combined amount of:
(a) a VLA-4 inhibitor compound;
(b) a first agent which interacts with one or more chemokines; and
(c) a second agent which interact with one or more chemokines.
29. The method of claim 28, wherein the method is ex vivo.
30. The method of claim 28, wherein the method is in vitro.
31 . The method of claim 28, wherein the method is in vivo.
32. A method of improving the harvest of hematopoietic stem cells or progenitor cells comprising administering to a patient a therapeutically effective combined amount of:
(a) a VLA-4 inhibitor compound;
(b) a first agent which interacts with one or more chemokines; and
(c) a second agent which interact with one or more chemokines.
33. A method of transplanting hematopoietic stem cells or progenitor cells
comprising:
(A) administering to a first patient a therapeutically effective combined amount of:
(i) a VLA-4 inhibitor compound;
(ii) a first agent which interacts with one or more chemokines; and
(iii) a second agent which interact with one or more chemokines;
(B) collecting hematopoietic stem cells or progenitor cells from the first patient;
(C) transplanting the hematopoietic stem cells or progenitor cells to a second patient.
34. A method of transplanting hematopoietic stem cells or progenitor cells comprising transplanting the hematopoietic stem cells or progenitor cells collected from a first patient who has been administered a therapeutically effective combined amount of:
(a) a VLA-4 inhibitor compound;
(b) a first agent which interacts with one or more chemokines; and
(c) a second agent which interact with one or more chemokines;
to a second patient.
35. A method of transplanting to a patient hematopoietic stem cells or progenitor cells comprising:
(A) administering to a first patient a therapeutically effective combined amount of:
(i) a VLA-4 inhibitor compound;
(ii) a first agent which interacts with one or more chemokines; and
(iii) a second agent which interact with one or more chemokines;
(B) collecting hematopoietic stem cells or progenitor cells from the first patient;
(C) transplanting the hematopoietic stem cells or progenitor cells to a second patient.
36. A method of transplanting to a patient hematopoietic stem cells or progenitor cells comprising transplanting the hematopoietic stem cells or progenitor cells collected from the patient who has been administered a therapeutically effective combined amount of:
(a) a VLA-4 inhibitor compound;
(b) a first agent which interacts with one or more chemokines; and
(c) a second agent which interact with one or more chemokines.
37. The method of either claim 33 or claim 34, wherein the hematopoietic stem cells are collected from the patient before an event which results in a reduction of the amount of the first patient's hematopoietic stem cells or progenitor cells.
38. The method of either claim 35 or claim 36, wherein the hematopoietic stem cells or progenitor cells are transplanted after an event which results in a reduction of the amount of the patient's hematopoietic stem cells or progenitor cells.
39. The method of either claim 33 or claim 34, wherein the first patient is a
compatible hematopoietic stem cell donor.
40. A method of improving the effectiveness of a treatment of cancer in a patient administered a chemotherapy or a radiotherapy comprising:
(A) administering to the patient a therapeutically effective combined amount of:
(i) a VLA-4 inhibitor compound;
(ii) a first agent which interacts with one or more chemokines; and
(iii) a second agent which interact with one or more chemokines; and
(B) administered a chemotherapy or a radiotherapy to the patient.
41 . A method of improving the effectiveness of a treatment of cancer in patient who has been administered a chemotherapy or radiotherapy and a therapeutically effective combined amount of:
(a) a VLA-4 inhibitor compound;
(b) a first agent which interacts with one or more chemokines; and
(c) a second agent which interact with one or more chemokines;
42. The method of either claim 40 or claim 41 , wherein the chemotherapy or radiotherapy is administered simultaneously with the first and/or second agent which interacts with a chemokine and the VLA-4 inhibitor.
43. The method of either claim 40 or claim 41 , wherein the chemotherapy or radiotherapy is administered before the first and/or second agent which interacts with a chemokine and the VLA-4 inhibitor.
44. The method of either claim 40 or claim 41 , wherein the chemotherapy or radiotherapy is administered after the first and/or second agent which interacts with a chemokine and the VLA-4 inhibitor.
45. The method according to any one of claims 16-44, wherein the method comprises administering the first and/or second agent which interacts with a chemokine once.
46. The method according to any one of claims 16-44, wherein the method comprises administering the first and/or second agent which interacts with a chemokine two or more times.
47. The method according to any one of claims 16-46, wherein the method comprises administering the VLA-4 inhibitor once.
48. The method according to any one of claims 16-46, wherein the method comprises administering the VLA-4 inhibitor two or more times.
49. The method according to any one of claims 16-48, wherein the VLA-4 inhibitor and the first and second agent which interacts with a chemokine are administered simultaneously.
50. The method of claim 56, wherein the method comprises administering a composition comprising the first and second agent which interacts with a chemokine and VLA-4 inhibitor.
51 . The method according to any one of claims 16-48, wherein the method comprises administering the first and/or second agent which interacts with a chemokine before administering the VLA-4 inhibitor.
52. The method of claim 51 , wherein the first and/or second agent which interacts with a chemokine is administered from 15 minutes to 0 minutes before the VLA-4 inhibitor.
53. The method according to any one of claims 16-48, wherein the method comprises administering the first and/or second agent which interacts with a chemokine after administering the VLA-4 inhibitor.
54. The method according to any one of claims 16-53, wherein the first and/or second agent which interacts with a chemokine is administered subcutaneously and the VLA-4 inhibitor is administered intravenously.
55. The method according to any one of claims 16-53, wherein both the first and/or second agent which interacts with a chemokine and the VLA-4 inhibitor are
administered subcutaneously.
56. The method according to any one of claims 16-55, wherein the method produces effects equivalent to the sum of the effects of each of the first or second agent which interacts with a chemokine or VLA-4 inhibitor when administered independently.
57. The method according to any one of claims 16-56, wherein the method produces a synergistic effect relative to the effects of each of the first or second agent which interacts with a chemokine or VLA-4 inhibitor when administered independently.
58. The method according to any one of claims 30-56, wherein the hematopoietic stem cells or progenitor cells are LSK-SLAM cells.
59. The method according to any one of claims 16-56, wherein the first agent and second agent which interact with a chemokine are selected from the group consisting of: an agent which interacts with a C-X-C chemokine or a C-X-C chemokine receptor.
60. The method of claim 59, wherein the first agent is a CXCR2 agonist.
61 . The method of either claim 59 or claim 60, wherein the second agent is a CXCR4 inhibitor.
62. The method according to claim 60, wherein the agent is Gro-β, or a derivative of Gro-β.
63. The method of claim 62, wherein the derivative of Gro-β is a truncated Gro- β.
64. The method of claim 61 , wherein the CXCR4 inhibitor is one or more of
AMD3100 (plerixafor), AMD3465, CTCE-0214, CTCE-9908, CP-1221 (linear peptides, cyclic peptides, natural amino-acids, unnatural amino acids, and peptidomimetic compounds), T140 and analogs, 4F-benzoyl-TN24003, KRH- 1 120, KRH-1636, KRH-2731 , polyphemusin analogue, ALX40-4C, or
combinations thereof.
65. The method according to any one of claims 16-56, wherein the first agent inhibits the activity of the chemokine and the second agent activates the activity of the chemokine.
PCT/US2017/059770 2016-11-02 2017-11-02 Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof WO2018085574A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/401,950 US20190328706A1 (en) 2016-11-02 2019-05-02 Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662416462P 2016-11-02 2016-11-02
US62/416,462 2016-11-02
US201662430167P 2016-12-05 2016-12-05
US62/430,167 2016-12-05

Related Child Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/059733 Continuation-In-Part WO2018085552A1 (en) 2016-11-02 2017-11-02 Integrin antagonists

Publications (2)

Publication Number Publication Date
WO2018085574A2 true WO2018085574A2 (en) 2018-05-11
WO2018085574A3 WO2018085574A3 (en) 2018-07-12

Family

ID=62076173

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/059770 WO2018085574A2 (en) 2016-11-02 2017-11-02 Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof

Country Status (1)

Country Link
WO (1) WO2018085574A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11116760B2 (en) 2018-10-30 2021-09-14 Gilead Sciences, Inc. Quinoline derivatives
US11174256B2 (en) 2018-10-30 2021-11-16 Gilead Sciences, Inc. Imidazopyridine derivatives
US11179383B2 (en) 2018-10-30 2021-11-23 Gilead Sciences, Inc. Compounds for inhibition of α4β7 integrin
US11224600B2 (en) 2018-10-30 2022-01-18 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin
US11578069B2 (en) 2019-08-14 2023-02-14 Gilead Sciences, Inc. Compounds for inhibition of α4 β7 integrin

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008509928A (en) * 2004-08-13 2008-04-03 アノーメド インコーポレイテッド Chemokine combination to mobilize progenitor / stem cells
CN101495115A (en) * 2006-08-02 2009-07-29 健赞股份有限公司 Combination therapy
WO2013149064A1 (en) * 2012-03-30 2013-10-03 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Uridine diphosphate compounds as mobilizers of hematopoietic progenitor cells

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11116760B2 (en) 2018-10-30 2021-09-14 Gilead Sciences, Inc. Quinoline derivatives
US11174256B2 (en) 2018-10-30 2021-11-16 Gilead Sciences, Inc. Imidazopyridine derivatives
US11179383B2 (en) 2018-10-30 2021-11-23 Gilead Sciences, Inc. Compounds for inhibition of α4β7 integrin
US11224600B2 (en) 2018-10-30 2022-01-18 Gilead Sciences, Inc. Compounds for inhibition of alpha 4 beta 7 integrin
US11578069B2 (en) 2019-08-14 2023-02-14 Gilead Sciences, Inc. Compounds for inhibition of α4 β7 integrin

Also Published As

Publication number Publication date
WO2018085574A3 (en) 2018-07-12

Similar Documents

Publication Publication Date Title
WO2018085574A2 (en) Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof
US20220008468A1 (en) Anti-tumor t cell immunity induced by high dose radiation
Isidori et al. Phase II study of a single pegfilgrastim injection as an adjunct to chemotherapy to mobilize stem cells into the peripheral blood of pretreated lymphoma patients
US20170106003A1 (en) Anti-cancer effects of jak2 inhibitors in combination with thalidomide derivatives and glucocorticoids
US20100178271A1 (en) Combination Therapy
US10029000B2 (en) Uses of IL-12 in hematopoiesis
Morstyn et al. Stem cell factor is a potent synergistic factor in hematopoiesis
US20190328706A1 (en) Compositions comprising an integrin inhibitor and agents which interact with a chemokine and methods of use thereof
WO2018085578A1 (en) Integrin inhibitors in combination with an agent which interacts with a chemokine receptor
RU2351357C2 (en) Pharmaceutical composition used for stem cells mobilisation
US6693134B2 (en) Bicyclic aromatic chemokine receptor ligands
JP2012025765A (en) Use of il-12 in hematopoiesis
Elkordy et al. A phase I trial of recombinant human interleukin-1β (OCT-43) following high-dose chemotherapy and autologous bone marrow transplantation
US20230059785A1 (en) Methods for enhancing t cells using venetoclax
Wierenga et al. Peripheral blood stem cells differ from bone marrow stem cells in cell cycle status, repopulating potential, and sensitivity toward hyperthermic purging in mice mobilized with cyclophosphamide and granulocyte colony-stimulating factor
TW201121965A (en) Use of a quinolone derivative containing 7-(4-aminomethyl-3-oxime)pyrrolidine group that is capable of inducing granulocyte colony stimulating factor for treatment of neutropenia and recovery of hematopoiesis
Boyehansen et al. Hematopoietic growth factors for the treatment of myelodysplastic syndromes
JP2016501208A (en) Combination therapy with boraseltib
US20220118022A1 (en) Conditioning agents for use in allogeneic hematopoetic stem cell transplantation
US20110256236A1 (en) Enhancement of the Anti-Leukemic Effect of Thalidomide
Teicher et al. Interaction of interleukin‐11 with cytotoxic therapies in vitro against CEM cells and in vivo against EMT‐6 murine mammary carcinoma
Pelus et al. Chemokine axes in hematopoietic stem cell mobilization
Teicher et al. Lisofylline as an adjuvant to high dose cytotoxic therapy
WO2001028573A2 (en) Monocyte conditioned medium for cancer treatment
Campion et al. J. Nemunaitis, zyxwvutsrqponmlkji

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: 17866876

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17866876

Country of ref document: EP

Kind code of ref document: A2