US20220281828A1 - Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease - Google Patents

Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease Download PDF

Info

Publication number
US20220281828A1
US20220281828A1 US17/555,786 US202117555786A US2022281828A1 US 20220281828 A1 US20220281828 A1 US 20220281828A1 US 202117555786 A US202117555786 A US 202117555786A US 2022281828 A1 US2022281828 A1 US 2022281828A1
Authority
US
United States
Prior art keywords
cells
reg
optionally substituted
jak2
aji
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US17/555,786
Inventor
Brian Betts
Said M. Sebti
Harshani Lawrence
Nicholas Lawrence
Claudio Anasetti
Joseph Pidala
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
H Lee Moffitt Cancer Center and Research Institute Inc
Original Assignee
H Lee Moffitt Cancer Center and Research Institute Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by H Lee Moffitt Cancer Center and Research Institute Inc filed Critical H Lee Moffitt Cancer Center and Research Institute Inc
Priority to US17/555,786 priority Critical patent/US20220281828A1/en
Publication of US20220281828A1 publication Critical patent/US20220281828A1/en
Assigned to H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUTE, INC. reassignment H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BETTS, BRIAN, PIDALA, Joseph, LAWRENCE, HARSHANI, LAWRENCE, NICHOLAS, ANASETTI, CLAUDIO, SEBTI, SAID M.
Priority to US18/098,990 priority patent/US20230150948A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the subject matter disclosed herein relates generally to graft versus host disease (GVHD). More specifically, the subject matter disclosed herein relates to inhibitors of Aurora kinase and JAK2 and their use in preventing or treating GVHD.
  • GVHD graft versus host disease
  • GVHD graft-versus-host disease
  • allogeneic hematopoietic cell transplantation alloHCT.
  • CNI calcineurin inhibitors
  • T reg regulatory T-cell differentiation and function
  • CD28 costimulation contributes to T-cell alloreactivity and GVHD.
  • GVHD in rodents is ameliorated by transplantation of CD28 negative compared to wild type T-cells (P. Tan et al., Induction of alloantigen-specific hyporesponsiveness in human T lymphocytes by blocking interaction of CD28 with its natural ligand B7/BB1 . J. Experi. Med. 177, 165 (1993); X. Z. Yu et al., CD28-specific antibody prevents graft versus-host disease in mice. J. Immunol. 164, 4564 (2000)). Blockade of ligand interactions between CD80/CD86 and CD28 with neutralizing antibody also reduces murine GVHD.
  • CD28 signal transduction activates mTOR and Aurora kinase in T-cells (J. Song et al., The kinases aurora B and mTOR regulate the G1-S cell cycle progression of T lymphocytes. Nature Immunol. 8, 64 (2007)).
  • mTOR is a known pharmacologic target in GVHD prophylaxis (C. Cutler et al., Tacrolimus/sirolimus vs tacrolimus/methotrexate as GVHD prophylaxis after matched, related donor allogeneic HCT. Blood 124, 1372 (2014); J.
  • IL-6 receptor signaling polarizes T H 1 and T H 17 cells that are effectors in GVHD, and impairs T regs that modulate GVHD (B. C. Betts et al., Janus kinase-2 inhibition induces durable tolerance to alloantigen by human dendritic cell-stimulated T cells yet preserves immunity to recall antigen. Blood 118, 5330 (2011); B. C. Betts, A. Veerapathran, J. Pidala, X. Z. Yu, C. Anasetti, STAT5 polarization promotes iT regs and suppresses human T-cell alloresponses while preserving CTL capacity. J. Leukocyte Biol. 95, 205 (2014); J.
  • JAK2 inhibition conversely polarizes natural T reg responses, and inhibits T H 1 and T H 17 development in vitro.
  • selective blockade of JAK2 alone does not provide lasting protection in murine GVHD. This observation is distinct from JAK1/JAK2 inhibition, where co-blockade of JAK1 acts broadly to reduce GVHD ( Blood 123, 3832 (2014)) as well as beneficial anti-viral CTL (S.
  • compositions and methods for treating GVHD address these and other needs.
  • the disclosed subject matter in one aspect, relates to compounds, compositions and methods of making and using compounds and compositions.
  • the disclosed subject matter relates to reducing the risk of, preventing, or treating graft versus host disease (GVHD) in a subject.
  • the subject matter disclosed herein relates to inhibitors of Aurora kinase A and JAK2.
  • the disclosed subject matter relates to concurrent inhibition of Aurora kinase A and JAK2 and their use in reducing the risk of, preventing, or treating GVHD.
  • GVHD can be attributed to a solid organ transplant, tissue graft, or a cellular transplant.
  • the methods described herein can include administering to a subject at risk of developing or having GVHD, a composition comprising a compound of the following formula
  • composition can include a compound of the following formula:
  • composition can include a compound having one of the following formulas:
  • R 3 is 2-Cl, and n is 1.
  • R 4 is 4-COOH, and m is 1.
  • R 4 is 4-CONH 2 , and m is 1.
  • R 3 is 2-Cl, n is 1, m is 1, and R 4 is COOH, COR 5 , CONH 2 , CONR 5 , or CONSO 2 R 5 , wherein R 5 is C 1 -C 6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl.
  • R 3 is H, m is 1, and R 4 is COOH, COR 5 , CONH 2 , CONR 5 , or CONSO 2 R 5 , wherein R 5 is C 1 -C 6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl.
  • R 1 is Cl
  • R 2 is H
  • R 3 is H
  • m is 1
  • R 4 is 4-CONH 2 .
  • n is 0.
  • compositions described herein can be administered at a dose of about 0.1 mg/kg to about 100 mg/kg.
  • FIGS. 1A-1E show synergistic immune suppression with combined inhibition of Aurora kinase A and JAK2.
  • T-cells were stimulated with DCs (DC:T-cell ratio 1:30) exposed to either TG101348 (JAK2 inhibitor), alisertib (Aurora kinase A inhibitor), or both at a fixed ratio of 1:5 respectively at varying concentrations once on day 0.
  • Proliferation was determined by fluorescence assay on day 5, with % proliferation based on DMSO control.
  • bar graphs depict mean gated CD3 + Tcell STAT3 (target of JAK2) or H3 Ser10 (target of Aurora) phosphorylation ⁇ SD following 5-day allogeneic DC stimulation treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (unpaired t-test). Representative contour plots are shown with phosphorylation based on isotype control.
  • a bar graph shows mean T-cell viability ⁇ SD by LIVE/DEAD Yellow exclusion from 4 independent experiments (unpaired t-test). *P ⁇ 0.05.
  • FIGS. 2A-2I show concurrent blockade of Aurora kinase A and JAK2 selectively suppresses alloreactive T conv , while sparing responder T regs .
  • T-cells were stimulated with DCs (DC:Tcell ratio 1:30) and treated with bisanilinopyrimidine (I) or DMSO once on day 0.
  • bar graphs show percent means of CD4 + alloreactive T conv (CD25 + , CD127 + ), CD4 + T reg (CD25 + , CD127 ⁇ ), and T reg :allo T conv ratio ⁇ SD from 5 independent experiments on day 5 of culture (unpaired t-test).
  • FIG. 2E box plots depict mean T reg versus T conv proliferation by Cell Trace Violet dilution ⁇ SD among identically treated alloMLRs exposed to bisanilinopyrimidine (I) or DMSO from 3 independent experiments (unpaired t-test).
  • FIG. 2F is a representative histogram that shows proliferation in each T-cell compartment with respect to bisanilinopyrimidine (I) or DMSO treatment.
  • FIG. 2G a bar graph shows mean gated CD4 + T-cell STAT5 phosphorylation ⁇ SD following brief IL-2 stimulation treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (ANOVA).
  • FIG. 2H a histogram depicts intracellular Foxp3 expression among CD4 + , CD25 + , CD127 ⁇ T regs following 5-day DCallostimulation exposed to bisanilinopyrimidine (I) or DMSO. Data representative of 5 experiments.
  • FIG. 1 a bar graph shows mean gated CD4 + T-cell STAT5 phosphorylation ⁇ SD following brief IL-2 stimulation treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (ANOVA).
  • FIG. 2H a histogram depicts intracellular Foxp3 expression among CD4 + , CD25 + , CD127 ⁇ T regs following 5-day DCallostimulation exposed to bisanilinopyrimidine (I) or DMSO. Data representative of 5 experiments
  • AlloMLRs (DC:T-cell ratio 1:30) were treated with alisertib (Aurora kinase A inhibitor), TG101348 (JAK2 inhibitor), a combination of both, or DMSO once on day 0.
  • FIGS. 3A-3E show dual blockade of Aurora kinase A and JAK2 selectively increases the ratio of iT reg to alloreactive T conv .
  • a representative contour plots show T reg (CD25 + , CD127 ⁇ ) depletion of na ⁇ ve CD4 + T-cell responders at outset of alloMLR (DC:T-cell ration 1:30), followed by induction of iT reg versus allo T conv (CD25 + , CD127 + ) after 5 days of culture exposed to AJI-214 or DMSO.
  • bar graphs show mean frequency of CD4 + iT reg and allo T conv in 5-day allogeneic co-cultures treated with bisanilinopyrimidine (I) or DMSO ⁇ SD from 4 independent experiments (unpaired t-test).
  • a bar graph depicts triplicate mean % demethylation of Foxp3 ⁇ SEM among iT regs in alloMLRs treated with bisanilinopyrimidine (I) or DMSO from 4 independent experiments (unpaired t-test).
  • FIG. 3B-3C bar graphs show mean frequency of CD4 + iT reg and allo T conv in 5-day allogeneic co-cultures treated with bisanilinopyrimidine (I) or DMSO ⁇ SD from 4 independent experiments (unpaired t-test).
  • bar graphs show replicate means of absolute numbers of iT reg , allo T conv , and the ratio of iT reg :allo T conv ⁇ SD from 5-day MLRs treated with Alisertib 1.75 ⁇ M, TG101348 350 nM, a combination of both, bisanilinopyrimidine (I) 750 nM, or DMSO control (ANOVA). Data are from one representative experiment of 2 performed in triplicate.
  • FIGS. 4A-4B show combined inhibition of Aurora kinase A and JAK2 enhances antigen-specific iT reg suppressive potency.
  • FIG. 4A the suppressive capacity of sorted, DC-allostimulated iT regs previously exposed to bisanilinopyrimidine (I) or DMSO was tested at different ratios of iT reg to self T-cell responders stimulated by original allogeneic DCs (DC:responder T-cell ratio 1:30) in fresh alloMLRs. No additional bisanilinopyrimidine (I) or DMSO was added.
  • the bar graph shows triplicate means of % proliferation ⁇ SEM based on 3H-thymidine incorporation on day 6 from 3 independent experiments (ANOVA).
  • FIGS. 5A-5J show targeting Aurora kinase A and JAK2 increases CD39 expression and ATP scavenging among iT reg .
  • contour plots show the CD4 + iT reg and non-T reg gating strategy after 5-day alloMLR treated with bisanilinopyrimidine (I) (750 nM) or DMSO.
  • FIGS. 5B-5D CD39 expression (% and geometric MFI) was increased by among iT reg generated in the presence of bisanilinopyrimidine (I) (750 nM).
  • the bar graphs show mean data ⁇ SD from 3 independent experiments (unpaired t-test).
  • FIG. 5A contour plots show the CD4 + iT reg and non-T reg gating strategy after 5-day alloMLR treated with bisanilinopyrimidine (I) (750 nM) or DMSO.
  • FIGS. 5B-5D CD39 expression (% and geometric MFI) was increased by among iT reg generated in the presence
  • FIGS. 5G-5H bar graphs depict mean fold MFI of LAG3 and CTLA4 ⁇ SD on iT regs harvested from alloMLRs treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (unpaired t-test).
  • FIGS. 6A-6B show Aurora kinase A inhibition does not impair T H 17 differentiation.
  • Isolated na ⁇ ve CD4 + T-cells were stimulated with allogeneic DCs for 5 days with alisertib, AJI-214, TG101348, or DMSO. Media was supplemented with IL-6, TGF-beta, and anti-IFN- ⁇ mAb to promote RORgammaT expression.
  • FIGS. 7A-7H show blockade of Aurora kinase A and JAK2 reduces xenogeneic GVHD and preserves anti-tumor CTL function.
  • AlloMLR DC:T-cell ratio 1:30
  • II bisanilinopyrimidine
  • DMSO DMSO
  • Proliferation was determined by colorimetric assay on day 5 (ANOVA).
  • IC 50 200 nM.
  • Graph shows triplicate means ⁇ SD from 1 representative experiment of 2.
  • FIG. 7B a representative histogram shows bisanilinopyrimidine (II) inhibits CD3 + T-cell STAT3 phosphorylation in vitro.
  • mice received human PBMCs (30 ⁇ 10 6 cells), with bisanilinopyrimidine (II) (50 mg/kg daily) or vehicle administered from day 0 to day +14.
  • FIG. 7C a representative histogram shows H3 ser10 phosphorylation in human CD3 + T-cells harvested at day +14 among each group.
  • FIG. 7G a graph depicts mean specific lysis ⁇ SD by human CTL generated in vivo using NSG mice transplanted with human PBMCs and irradiated U937 cells (10 7 ) on days 0 and +7. Results shown are from 1 of 2 independent experiments, using a total of 7 mice per arm.
  • FIGS. 8A-8K show targeting Aurora kinase A and JAK2 increases the proportion of T reg to allo T conv in vivo, and reduces T-cell homing to recipient livers.
  • Xenotransplanted NSG mice were treated with bisanilinopyrimidine (II) (50 mg/kg daily) or vehicle daily starting at day 0, then euthanized on day +14.
  • Recipient spleens and livers were harvested, and tissue-resident T-cells were evaluated.
  • Dot plots show absolute number of total spleen cells ( FIG. 8A ), human CD3 + T-cells ( FIG. 8B ), % CD4 + , CD25 + , CD127 ⁇ , Foxp3 + T reg ( FIG.
  • FIG. 8C representative contour plots show the % CD4 + T reg and % CD4 + alloreactive T conv residing in spleens of bisanilinopyrimidine (II)- or vehicle-treated mice at day +14.
  • FIG. 8G the representative histograms show corresponding expression of Foxp3 within the CD4+T regs .
  • FIGS. 9A-9H show synergistic immune suppression with combined inhibition of Aurora kinase A and JAK2.
  • Human T cells were stimulated with DCs (DC/T cell ratio of 1:30) exposed to TG101348 (JAK2 inhibitor), alisertib (Aurora kinase A inhibitor), or both TG101348 and alisertib at a fixed ratio of 1:5, respectively, at varying concentrations once on day 0.
  • Proliferation was determined by fluorescence assay on day 5, with % proliferation based on dimethyl sulfoxide (DMSO) control.
  • DMSO dimethyl sulfoxide
  • Graph depicts combination synergy (IC 50 values for TG101348 and alisertib were 350 nM and 1.75 mM, respectively), showing one representative independent experiment of two performed with triplicate technical replicates.
  • the CI was calculated using the Chou-Talalay method. AlloMLR (DC/T cell ratio of 1:30) treated with AJI-214 ( FIG. 9B ), AJI-100 ( FIG. 9C ) (dual JAK2/Aurora kinase A inhibitors), or DMSO once on day 0 is shown.
  • IC 50 values for AJI-214 and AJI-100 were 100 and 200 nM, respectively.
  • Graph shows average triplicate means ⁇ SEM from two to three independent experiments [analysis of variance (ANOVA)].
  • FIG. 9D a bar graph depicts T cell proliferation when exposed to alisertib (1.75 mM), TG101348 (350 nM), a combination of alisertib and TG101348 (combo), AJI-214 (750 nM), or AJI-100 (750 nM) in alloMLRs. Means ⁇ SEM from four independent experiments (ANOVA) are shown using triplicate technical replicates.
  • FIGS. 9E-9H bar graphs depict the mean gated CD3 + T cell H3Ser10 (target of Aurora) or STAT3 (target of JAK2) phosphorylation ⁇ SD from three independent experiments after stimulation with allogeneic DCs (5 days) or IL-6 (15 min), respectively (ANOVA). Representative contour plots show H3Ser10 and STAT3 phosphorylation, respectively.
  • FIGS. 10A-10H show immunosuppressive effect of Aurora kinase A/JAK2 blockade on responder T conv and T H subsets.
  • T cells were stimulated with allogeneic DCs (DC/T cell ratio of 1:30) and treated with kinase inhibitors or DMSO once on day 0.
  • Bar graphs show replicate mean absolute numbers of activated CD4 + , CD25 + , CD127 + or activated CD8 + , CD25 + T conv ⁇ SEM at day +5 from six independent experiments (ANOVA and paired t test).
  • FIG. 10A-10H show immunosuppressive effect of Aurora kinase A/JAK2 blockade on responder T conv and T H subsets.
  • FIG. 10A-10B T cells were stimulated with allogeneic DCs (DC/T cell ratio of 1:30) and treated with kinase inhibitors or DMSO once on day 0.
  • Bar graphs show replicate mean absolute numbers of activated CD4 + , CD
  • FIG. 10C a bar graph shows the mean number of IL-17 spots per well ⁇ SD from triplicate technical replicates among DC-allostimulated CD4 + T cells. One of three representative experiments is shown.
  • FIG. 10D a bar graph shows mean % CD4 + , IFN- ⁇ + T cells ⁇ SEM at day +5 of alloMLR from five independent experiments with technical replicates performed in triplicate.
  • FIGS. 10C a bar graph shows the mean number of IL-17 spots per well ⁇ SD from triplicate technical replicates among DC-allostimulated CD4 + T cells.
  • FIG. 10D a bar graph shows mean % CD4 + , IFN- ⁇ + T cells ⁇ SEM at day +5 of alloMLR from five independent experiments with technical replicates performed in triplicate.
  • FIG. 10E-10F representative contour plots show T reg (CD25 + and CD127 ⁇ ) depletion of CD4 + T cell responders at outset of alloMLR (DC/T cell ratio of 1:30), followed by induction of iT reg (CD127 ⁇ , CD25 + , and Foxp3 + ) after 5 days of culture exposed to kinase inhibitors or DMSO.
  • FIG. 10G a bar graph shows mean absolute numbers of iT regs ⁇ SEM from seven independent experiments performed with two to three technical replicates (ANOVA and paired t test).
  • FIG. 10H a representative histograms depict pSTAT5 expression among IL-2-stimulated CD3 + T cells while exposed to kinase inhibitors or DMSO.
  • Geometric mean fluorescence intensity (MFI) of pSTAT5 is shown along the right margin.
  • MFI mean fluorescence intensity
  • FIGS. 11A-11C show combined inhibition of Aurora kinase A and JAK2 enhances antigen-specific iT reg -suppressive potency.
  • a Bar graph depicts mean % demethylation of Foxp3 ⁇ SEM among iT regs in alloMLRs treated with AJI-214 (750 nM) or DMSO from four independent experiments using triplicate technical replicates.
  • FIG. 11B the suppressive capacity of sorted, DC-allostimulated iT regs previously exposed to AJI-214 or DMSO was tested at different ratios of iT reg to T cell responders stimulated by fresh allogeneic DCs (DC/responder T cell ratio of 1:30) in alloMLRs.
  • Bar graph shows means of % proliferation ⁇ SEM based on [ 3 H]thymidine incorporation on day 6 from three independent experiments with triplicate technical replicates (ANOVA).
  • ANOVA triplicate technical replicates
  • FIG. 11C the potency of iT regs generated in the presence of alisertib (1.75 mM), TG101348 (350 nM), a combination of alisertib and TG101348, or DMSO was tested in standard suppression assays. No additional small-molecule inhibitors or DMSO was added.
  • Bar graph shows means of % proliferation ⁇ SD based on [ 3 H]thymidine incorporation on day 6 (paired t test). Data are from one representative experiment of two performed using triplicate technical replicates. *P ⁇ 0.05.
  • FIGS. 12A-12J show targeting Aurora kinase A and JAK2 increases CD39 expression and ATP scavenging among iT reg .
  • contour plots show the CD4 + iT reg and non-T reg gating strategy after 5-day alloMLR treated with AJI-214 (750 nM) or DMSO.
  • CD39 density [geometric MFI (gMFI)] is increased by among iT reg generated in the presence of AJI-214 (750 nM). Bar graphs show mean data ⁇ SD from three independent experiments (paired t test). In FIG.
  • FIGS. 12G-12H bar graphs depict mean fold MFI of LAG3 and CTLA4 ⁇ SD on iT regs harvested from alloMLRs treated with AJI-214 or DMSO from three independent experiments.
  • FIG. 12I-12J bar graphs show mean concentrations of IL-10 and TGF-0 ⁇ SEM among PMA (phorbol 12-myristate 13-acetate)/ionomycin-stimulated iT regs previously exposed to AJI-214 or DMSO during coculture from four independent experiments using three technical replicates.
  • PMA phorbol 12-myristate 13-acetate
  • FIGS. 13A-13G show Blockade of Aurora kinase A and JAK2 reduces xenogeneic GVHD and preserves the in vivo generation of potent antitumor CTL.
  • NSG mice received human PBMCs (30 ⁇ 10 6 cells) by intraperitoneal injection, with alisertib (30 mg/kg daily), TG101348 (45 mg/kg twice a day), a combination of alisertib and TG101348, or vehicle administered by oral gavage from day 0 to day +14.
  • FIG. 13A percent survival is shown among the four groups (log-rank test).
  • FIG. 13A percent survival is shown among the four groups (log-rank test).
  • FIG. 13E representative contour plots show expression of H3Ser 10 and STAT3 phosphorylation among human CD3 + T cells harvested from recipient spleens at day +14.
  • FIG. 13G a graph depicts mean specific lysis ⁇ SD by human CD8 + CTL generated in vivo using NSG mice transplanted with human PBMCs and vaccinated with irradiated U937 cells (1 ⁇ 10 7 ) on days 0 and +7.
  • Results shown are from one of two independent experiments, using a total of seven mice per group.
  • FIGS. 14A-14M show targeting Aurora kinase A and JAK2 increases the proportion of T reg to activated T conv and reduces T H 17 and T H 1 cells in xenotransplanted recipient mice.
  • xenotransplanted NSG mice were treated with AJI-100 (50 mg/kg) or vehicle daily starting at day 0 and then euthanized on day +14. Recipient spleens, livers, and lungs were harvested, and tissue-resident T cells were evaluated.
  • Bar graphs show replicate mean absolute number of human CD4 + T cells ( FIG. 14A ), CD4 + T regs ( FIG. 14B ), CD4 + activated T conv (CD25 + and CD127 + ) ( FIG.
  • FIG. 14C representative contour plots show the % CD4 + T reg and % CD4 + activated allo-T conv residing in spleens of AJI-100-treated or vehicle-treated mice at day +14.
  • the representative histograms show corresponding expression of Foxp3 within the CD4 + T regs .
  • FIG. 14F a bar graph shows the replicate mean number of IL-17 spots per well ⁇ SEM among human lymphocytes harvested from recipient spleens at day +14 (Mann-Whitney).
  • FIG. 14E representative contour plots show the % CD4 + T reg and % CD4 + activated allo-T conv residing in spleens of AJI-100-treated or vehicle-treated mice at day +14.
  • the representative histograms show corresponding expression of Foxp3 within the CD4 + T regs .
  • FIG. 14F a bar graph shows the replicate mean number of IL-17 spots per well ⁇ SEM among human lymphocytes harvested from recipient sple
  • FIG. 14G-14H Bar graph and representative contour plots depict the amount of CD4+, IFN- ⁇ + T cells ⁇ SEM from AJI-100 or vehicle-treated mice at day +14 (Mann-Whitney).
  • FIG. 14I sections of recipient livers (top) and lung (bottom) show that AJI-100 significantly reduces GVHD damage in recipient target organs, compared to vehicle control.
  • FIG. 14J-14K bar graphs depict the mean GVHD pathology scores ⁇ SEM for host liver and lung at day +14.
  • a composition includes mixtures of two or more such compositions
  • an inhibitor includes mixtures of two or more such inhibitors
  • the kinase includes mixtures of two or more such kinase, and the like.
  • reduce or other forms of the word, such as “reducing” or “reduction,” is meant lowering of an event or characteristic (e.g., the risk of having GVHD). It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to.
  • prevent or other forms of the word, such as “preventing” or “prevention,” is meant to stop a particular event or characteristic, to stabilize or delay the development or progression of a particular event or characteristic, or to minimize the chances that a particular event or characteristic will occur. Prevent does not require comparison to a control as it is typically more absolute than, for example, reduce. As used herein, something could be reduced but not prevented, but something that is reduced could also be prevented. Likewise, something could be prevented but not reduced, but something that is prevented could also be reduced. It is understood that where reduce or prevent are used, unless specifically indicated otherwise, the use of the other word is also expressly disclosed.
  • treatment refers to obtaining beneficial or desired clinical results.
  • beneficial or desired clinical results include, but are not limited to, any one or more of: alleviation of one or more symptoms (such as GVHD), diminishment of extent of GVHD, stabilized (i.e., not worsening) state of GVHD, preventing or delaying occurrence or recurrence of GVHD, delay or slowing of GVHD progression, and amelioration of the GVHD state.
  • patient preferably refers to a human in need of treatment for any purpose, and more preferably a human in need of such a treatment to treat GVHD.
  • patient can also refer to non-human animals, preferably mammals such as dogs, cats, horses, cows, pigs, sheep and non-human primates, among others, that are in need of treatment.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • references in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed.
  • X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the mixture.
  • a weight percent (wt. %) of a component is based on the total weight of the formulation or composition in which the component is included.
  • the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds.
  • Illustrative substituents include, for example, those described below.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms, such as nitrogen can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms.
  • substitution or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • aliphatic refers to a non-aromatic hydrocarbon group and includes branched and unbranched, alkyl, alkenyl, or alkynyl groups.
  • alkyl as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like.
  • the alkyl group can also be substituted or unsubstituted.
  • the alkyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below
  • alkoxy as used herein is an alkyl group bound through a single, terminal ether linkage; that is, an “alkoxy” group can be defined as —OA 1 where A 1 is alkyl as defined above.
  • alkenyl as used herein is a hydrocarbon group of from 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon double bond.
  • Asymmetric structures such as (A 1 A 2 )C ⁇ C(A 3 A 4 ) are intended to include both the E and Z isomers. This may be presumed in structural formulae herein wherein an asymmetric alkene is present, or it may be explicitly indicated by the bond symbol C ⁇ C.
  • the alkenyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described
  • alkynyl as used herein is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond.
  • the alkynyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • aryl as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, phenoxybenzene, and the like.
  • heteroaryl is defined as a group that contains an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus.
  • non-heteroaryl which is included in the term “aryl,” defines a group that contains an aromatic group that does not contain a heteroatom. The aryl and heteroaryl group can be substituted or unsubstituted.
  • the aryl and heteroaryl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • the term “biaryl” is a specific type of aryl group and is included in the definition of aryl. Biaryl refers to two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl.
  • cycloalkyl as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms.
  • examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.
  • heterocycloalkyl is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus.
  • the cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted.
  • the cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • cycloalkenyl as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms and containing at least one double bound, i.e., C ⁇ C.
  • cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like.
  • heterocycloalkenyl is a type of cycloalkenyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus.
  • the cycloalkenyl group and heterocycloalkenyl group can be substituted or unsubstituted.
  • the cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • cyclic group is used herein to refer to either aryl groups, non-aryl groups (i.e., cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl groups), or both. Cyclic groups have one or more ring systems that can be substituted or unsubstituted. A cyclic group can contain one or more aryl groups, one or more non-aryl groups, or one or more aryl groups and one or more non-aryl groups.
  • aldehyde as used herein is represented by the formula —C(O)H. Throughout this specification “C(O)” is a short hand notation for C ⁇ O.
  • amine or “amino” as used herein are represented by the formula NA 1 A 2 A 3 , where A 1 , A 2 , and A 3 can be, independently, hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • carboxylic acid as used herein is represented by the formula —C(O)OH.
  • a “carboxylate” as used herein is represented by the formula —C(O)O—.
  • esters as used herein is represented by the formula —OC(O)A 1 or —C(O)OA 1 , where A 1 can be an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • ether as used herein is represented by the formula A 1 OA 2 , where A 1 and A 2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • ketone as used herein is represented by the formula A 1 C(O)A 2 , where A 1 and A 2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • halide refers to the halogens fluorine, chlorine, bromine, and iodine.
  • hydroxyl as used herein is represented by the formula —OH.
  • nitro as used herein is represented by the formula —NO 2 .
  • cyano as used herein is represented by the formula —CN
  • zido as used herein is represented by the formula —N 3 .
  • sulfonyl is used herein to refer to the sulfo-oxo group represented by the formula —S(O) 2 A 1 , where A 1 can be hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • sulfoxide is used herein to refer to the sulfo-oxo group represented by the formula —OS(O) 2 A 1 , where A 1 can be hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • sulfonylamino or “sulfonamide” as used herein is represented by the formula —S(O) 2 NH 2 .
  • thiol as used herein is represented by the formula —SH.
  • Such chiral centers may be of either the (R-) or (S-) configuration.
  • the compounds provided herein may either be enantiomerically pure, or be diastereomeric or enantiomeric mixtures. It is to be understood that the chiral centers of the compounds provided herein may undergo epimerization in vivo. As such, one of skill in the art will recognize that administration of a compound in its (R-) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its (S-) form.
  • substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), nuclear magnetic resonance (NMR), gel electrophoresis, high performance liquid chromatography (HPLC) and mass spectrometry (MS), gas-chromatography mass spectrometry (GC-MS), and similar, used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detectably alter the physical and chemical properties, such as enzymatic and biological activities, of the substance.
  • TLC thin layer chromatography
  • NMR nuclear magnetic resonance
  • HPLC high performance liquid chromatography
  • MS mass spectrometry
  • GC-MS gas-chromatography mass spectrometry
  • a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer, diastereomer, and meso compound, and a mixture of isomers, such as a racemic or scalemic mixture.
  • a “pharmaceutically acceptable” component is one that is suitable for use with humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable salt” refers to a salt that is pharmaceutically acceptable and has the desired pharmacological properties. Such salts include those that may be formed where acidic protons present in the compounds are capable of reacting with inorganic or organic bases. Suitable inorganic salts include those formed with the alkali metals, e.g., sodium, potassium, magnesium, calcium, and aluminum. Suitable organic salts include those formed with organic bases such as the amine bases, e.g., ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like.
  • Such salts also include acid addition salts formed with inorganic acids (e.g., hydrochloric and hydrobromic acids) and organic acids (e.g., acetic acid, citric acid, maleic acid, and the alkane- and arene-sulfonic acids such as methanesulfonic acid and benzenesulfonic acid).
  • inorganic acids e.g., hydrochloric and hydrobromic acids
  • organic acids e.g., acetic acid, citric acid, maleic acid, and the alkane- and arene-sulfonic acids such as methanesulfonic acid and benzenesulfonic acid.
  • a pharmaceutically acceptable salt may be a mono-acid-mono-salt or a di-salt; similarly, where there are more than two acidic groups present, some or all of such groups can be converted into salts.
  • “Pharmaceutically acceptable excipient” refers to an excipient that is conventionally useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use. Such excipients can be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.
  • a “pharmaceutically acceptable carrier” is a carrier, such as a solvent, suspending agent or vehicle, for delivering the disclosed compounds to the patient.
  • the carrier can be liquid or solid and is selected with the planned manner of administration in mind.
  • Liposomes are also a pharmaceutical carrier.
  • carrier includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
  • terapéuticaally effective amount means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence. An effective amount can be administered in one or more doses.
  • Effective amounts of a compound or composition described herein for treating a mammalian subject can include about 0.1 to about 1000 mg/Kg of body weight of the subject/day, such as from about 1 to about 100 mg/Kg/day, especially from about 10 to about 100 mg/Kg/day.
  • the doses can be acute or chronic.
  • a broad range of disclosed composition dosages are believed to be both safe and effective.
  • the compounds can concurrently block Aurora kinase A and JAK2 signal.
  • These disclosed compounds can be used in various compositions to reduce the risk of developing, prevent, or treat GVHD in a subject.
  • These disclosed compounds can be used in various compositions to synergistically suppress alloreactive human T-cells in vitro, prevents xenogeneic graft-versus-host disease (GVHD) without impairing anti-tumor responses, and promotes the development and suppressive potency of CD39 + inducible T reg .
  • GVHD xenogeneic graft-versus-host disease
  • the disclosed compounds have the chemical structure shown in Formula I.
  • R 1 is F
  • R 2 is hydrogen
  • R 3 is 2-Cl
  • R 1 is Cl or F
  • R 2 is hydrogen
  • R 3 is hydrogen or 2-Cl
  • R 4 is 4-CONH 2 .
  • the disclosed compounds have the chemical structure shown in Formula II.
  • R 1 -R 4 and m and n are as defined above for Formula I.
  • R 1 is selected from the group consisting of H, Cl, F, Br, I, C 1 -C 6 alkyl, CN, NO 2 , and NH 2 .
  • R 2 is selected from the group consisting of H, F, and Cl.
  • each R 3 is selected, independently, from the group consisting of Cl, Br, F, COOH, CF 3 , CN, phenyl, OCH 3 , COR 5 , CONH 2 , CONR 5 , and COONH 2 .
  • n is 0, there is no R 3 .
  • each R 4 is selected, independently, from the group consisting of H, COOH, CONH 2 , CONR 5 , SO 2 NH 2 , CONSO 2 R 5 , tetrazole, 4-morpholine, and COR 5 .
  • Each R 5 is selected, independently, from the group consisting of optionally substituted C 1 -C 6 alkyl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heteroalkyl. In some examples R 5 is an unsubstituted substituent.
  • n is 0-5 (e.g., 0, 1, 2, 3, 4, or 5) and m is 1-5 (e.g., 1, 2, 3, 4, or 5).
  • R 1 is F
  • R 2 is H
  • m is 3
  • R 4 is 3,5-di-F, 4-OH.
  • Pharmaceutically acceptable salts of these compounds are also disclosed.
  • the compound has Formula II, wherein R 3 is 2-Cl, and n is 1. In other examples, the compound has Formula II, n is 0 and there is no R 3 . In other examples, the compound has Formula II, n is 2 and one R 3 is an ortho-Cl and the other R 3 is a para hydroxyl, methoxyl, or cyano group. In other examples, the compound has Formula II, wherein R 4 is 4-CONH 2 , and m is 1.
  • the compound has Formula II, wherein R 4 is 4-CONHR 5 , and m is 1
  • the compound has Formula II, wherein R 3 is 2-Cl, n is 1, m is 1, and R 4 is COOH, COR 5 , CONH 2 , CONR 5 , or CONSO 2 R 5 , wherein R 5 is C 1 -C 6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl.
  • the compound has Formula II, wherein R 3 is absent, n is 0, m is 1, and R 4 is COOH, COR 5 , CONH 2 , CONR 5 , or CONSO 2 R 5 , wherein R 5 is C 1 -C 6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl.
  • the disclosed compounds can have the following Formula IIIA or IIIB:
  • R 1 is selected from the group consisting of H, Cl, F, Br, I, CH 3 and NH 2 ;
  • R 2 is selected from the group consisting of H, F, and Cl.
  • R 3 is selected from the group consisting of 2-Cl, 2-Br, 2-F, 2-COOH, 2-CF 3 , 2-CN, 2-phenyl, 2-OCH 3 , 2-COONH 2 , 4-COOH, and 4-OCH 3 .
  • R 4 is selected from the group consisting of H, COOH, 2-CONH 2 , 4-CONH 2 , SO 2 NH 2 , tetrazole, and 4-morpholine.
  • R 1 , R 2 , R 4 , and m are as defined herein.
  • R 1 and R 2 together form a fused cycloalkyl, cycloheteroalkyl, aryl or heteraryl group. In other examples, R 1 and R 2 together form a fused furan. In other examples, R 1 and R 2 together form a fused cyclopentyl or fused cyclohexyl. In other examples, R 1 and R 2 together form a fused phenyl.
  • R 1 is C 1-8 alkyl or heteroalkyl. In other examples, R 1 is methyl, ethyl, or trifluoromethyl. In other examples, R 1 is chloro, bromo, or fluoro. In other examples, R 1 is CO 2 C 1-8 alkyl, CO 2 H, CO 2 NH 2 , or CO 2 NHC 1-8 alkyl.
  • R 2 is C 1 _ 8 alkyl or heteroalkyl. In other examples, R 2 is hydrogen.
  • R 3 there can be from 1 to 5 different substituents R 3 , i.e., n can be 1 to 5, though preferable n can be 1 to 3. In some examples, there is no R 3 substituent, i.e., n is 0.
  • R 3 is SO 2 NH 2 , SO 2 NHR 5 , or NHSO 2 R 5 , wherein R 5 is C 1 -C 6 alkyl or C 1 -C 6 cycloalkyl optionally substituted with C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, hydroxyl, or halide.
  • R 3 is NHC(O)R 5 , wherein R 5 is C 1 -C 6 alkyl or C 1 -C 6 cycloalkyl optionally substituted with C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, cycloalky, cycloheteroalkyl, hydroxyl, or halide.
  • R 3 is C 1 -C 6 alkyl or C 1 -C 6 cycloalkyl.
  • R 3 is C 1 -C 6 alkoxyl.
  • R 3 is halide.
  • n is 2 and each R 3 is selected from the group consisting of C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, halide, SO 2 NH 2 , SO 2 NHR 5 , and NHSO 2 R 5 , wherein R 5 is C 1 -C 6 alkyl or C 1 -C 6 cycloalkyl optionally substituted with C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, hydroxyl, or halide.
  • n is 2 and each R 3 is selected from the group consisting of C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, and halide.
  • n is 3 and each R 3 is selected from the group consisting of C 1 -C 6 alkyl, C 1 -C 6 alkoxyl, and halide. In other examples, n is 2 and each R 3 together form a fused hetercycloalkyl.
  • R 1 is F and R 2 is H.
  • R 4 is C(O)NHR 5 .
  • R 6 is C 1-8 alkyl. In other examples, R 6 is methyl. In other examples, R 6 is hydrogen.
  • R 7 is chloro, bromo, or fluoro. In other examples, R 7 is hydrogen.
  • X is N.
  • L is CH 2 (O) or C(O)NH.
  • n and m are both 1. In other examples, n is 0.
  • R 8 is oxo and p is 1. In other examples R 8 is oxo and p is 2.
  • the compound can be one of the following compounds.
  • the compound can have Formula IIIC:
  • m is 1 and R 4 is selected from the group consisting of COOH, 2-CONH 2 , 4-CONH 2 , SO 2 NH 2 , tetrazole, and 4-morpholine.
  • R 1 is Cl, F, Br, or I.
  • R 2 is H.
  • the compound is:
  • R 3 and n are as defined in Formula I, and R 10 is hydrogen, C 1 -C 6 alkyl, or C 1 -C 6 alkyl-OH.
  • Scheme I describes the general synthetic route used for preparation of dianilinipyrimidine (1) from readily available building blocks.
  • the 2,4-dichloropyrimidine was initially reacted with the requisite commercially available anilines with the method predominantly using isopropanol as the solvent, with reflux heating to obtain the required analog.
  • the method can include administering to the subject an effective amount of a compound or composition as disclosed herein.
  • the methods can further include administering a second compound or composition, such as, for example, an immunosuppressant.
  • an effective amount of one or more compounds or compositions disclosed herein is administered to a patient at risk of developing or have GVHD and who is in need of treatment thereof.
  • the patient can be a human or other mammal, such as a primate (monkey, chimpanzee, ape, etc.), dog, cat, cow, pig, or horse, or other animals at risk of developing or have GVHD.
  • GVHD may be due to a transplatation procedure involving the implantation of immunogenic tissue including but are not limited to, solid organ transplants (such as heart, kidney, and liver), tissue grafts (such as skin, intestine, pancreas, cornea, gonad, bone, and cartilage), and cellular transplants (such as cells from pancreas, brain and nervous tissue, muscle, skin, bone, cartilage, and liver).
  • tissue transplants such as heart, kidney, and liver
  • tissue grafts such as skin, intestine, pancreas, cornea, gonad, bone, and cartilage
  • cellular transplants such as cells from pancreas, brain and nervous tissue, muscle, skin, bone, cartilage, and liver.
  • organ rejection is an obstacle to complete recovery.
  • the individual's immune system recognizes antigens (HLA or minor H antigens) on the implanted tissue as foreign and mounts an immune response against it, which injures and destroys the implanted tissue.
  • HLA antigen
  • methods of treating or preventing cancer in a subject comprising administering to the subject an effective amount of a compound or composition as disclosed herein.
  • the methods can further comprise administering a second compound or composition, such as, for example, anticancer agents or anti-inflammatory agents. Additionally, the method can further comprise administering an effective amount of ionizing radiation to the subject.
  • Methods of killing a tumor cell comprise contacting a tumor cell with an effective amount of a compound or composition as disclosed herein.
  • the methods can further include administering a second compound or composition (e.g., an anticancer agent or an anti-inflammatory agent) or administering an effective amount of ionizing radiation to the subject.
  • a second compound or composition e.g., an anticancer agent or an anti-inflammatory agent
  • an effective amount of one or more compounds or compositions disclosed herein is administered to a patient having an oncological disorder and who is in need of treatment thereof.
  • the disclosed methods can optionally include identifying a patient who is or can be in need of treatment of an oncological disorder.
  • the patient can be a human or other mammal, such as a primate (monkey, chimpanzee, ape, etc.), dog, cat, cow, pig, or horse, or other animals having an oncological disorder.
  • Oncological disorders include, but are not limited to, cancer and/or tumors of the anus, bile duct, bladder, bone, bone marrow, bowel (including colon and rectum), breast, eye, gall bladder, kidney, mouth, larynx, esophagus, stomach, testis, cervix, head, neck, ovary, lung, mesothelioma, neuroendocrine, penis, skin, spinal cord, thyroid, vagina, vulva, uterus, liver, muscle, pancreas, prostate, blood cells (including lymphocytes and other immune system cells), and brain.
  • cancer and/or tumors of the anus include, but are not limited to, cancer and/or tumors of the anus, bile duct, bladder, bone, bone marrow, bowel (including colon and rectum), breast, eye, gall bladder, kidney, mouth, larynx, esophagus, stomach, testis, cervix, head, neck, ovary, lung, mes
  • Specific cancers contemplated for treatment include carcinomas, Karposi's sarcoma, melanoma, mesothelioma, soft tissue sarcoma, pancreatic cancer, lung cancer, leukemia (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myeloid, and other), and lymphoma (Hodgkin's and non-Hodgkin's), and multiple myeloma.
  • the disclosed compounds can be administered in combination with pharmaceutical formulations. Appropriate doses will be readily appreciated by those skilled in the art.
  • administration means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment.
  • a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a cytotoxic agent, etc.)
  • administration and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
  • the disclosed compounds can be formulated in a physiologically- or pharmaceutically-acceptable form and administered by any suitable route known in the art including, for example, oral, nasal, rectal, topical, and parenteral routes of administration.
  • parenteral includes subcutaneous, intradermal, intravenous, intramuscular, intraperitoneal, and intrasternal administration, such as by injection.
  • Administration of the disclosed compounds or compositions can be a single administration, or at continuous or distinct intervals as can be readily determined by a person skilled in the art.
  • the compounds disclosed herein, and compositions comprising them can also be administered utilizing liposome technology, slow release capsules, implantable pumps, and biodegradable containers. These delivery methods can, advantageously, provide a uniform dosage over an extended period of time.
  • the compounds can also be administered in their salt derivative forms or crystalline forms.
  • the compounds disclosed herein can be formulated according to known methods for preparing pharmaceutically acceptable compositions. Formulations are described in detail in a number of sources which are well known and readily available to those skilled in the art. For example, Remington's Pharmaceutical Science by E. W. Martin (1995) describes formulations that can be used in connection with the disclosed methods. In general, the compounds disclosed herein can be formulated such that an effective amount of the compound is combined with a suitable carrier in order to facilitate effective administration of the compound.
  • the compositions used can also be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspension, suppositories, injectable and infusible solutions, and sprays.
  • compositions also preferably include conventional pharmaceutically-acceptable carriers and diluents which are known to those skilled in the art.
  • carriers or diluents for use with the compounds include ethanol, dimethyl sulfoxide, glycerol, alumina, starch, saline, and equivalent carriers and diluents.
  • compositions disclosed herein can advantageously comprise between about 0.1% and 99%, and especially, 1 and 15% by weight of the total of one or more of the subject compounds based on the weight of the total composition including carrier or diluent.
  • Formulations suitable for administration include, for example, aqueous sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions, which can include suspending agents and thickening agents.
  • the formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze dried (lyophilized) condition requiring only the condition of the sterile liquid carrier, for example, water for injections, prior to use.
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powder, granules, tablets, etc. It should be understood that in addition to the ingredients particularly mentioned above, the compositions disclosed herein can include other agents conventional in the art having regard to the type of formulation in question.
  • Compounds disclosed herein, and compositions comprising them can be delivered to a cell either through direct contact with the cell or via a carrier means.
  • Carrier means for delivering compounds and compositions to cells are known in the art and include, for example, encapsulating the composition in a liposome moiety.
  • Another means for delivery of compounds and compositions disclosed herein to a cell comprises attaching the compounds to a protein or nucleic acid that is targeted for delivery to the target cell.
  • U.S. Pat. No. 6,960,648 and U.S. Application Publication Nos. 20030032594 and 20020120100 disclose amino acid sequences that can be coupled to another composition and that allows the composition to be translocated across biological membranes.
  • compositions for transporting biological moieties across cell membranes for intracellular delivery can also be incorporated into polymers, examples of which include poly (D-L lactide-co-glycolide) polymer for intracranial tumors; poly [bis(p-carboxyphenoxy) propane:sebacic acid] in a 20:80 molar ratio (as used in GLIADEL); chondroitin; chitin; and chitosan.
  • the compounds disclosed herein can be administered to a patient at risk of developing GVHD or in need of treatment in combination with other known treatments for GVHD. These other substances or treatments can be given at the same as or at different times from the compounds disclosed herein.
  • the compounds disclosed herein can be used in combination with an immunosuppressive agent such as IMUREKTM (azathioprine sodium), brequinar sodium, SPANIDINTM (gusperimus trihydrochloride, also known as deoxyspergualin), mizoribine (also known as bredinin), CELLCEPTTM (mycophenolate mofetil), NEORALTM (Cyclosporin A; also marketed as a different formulation under the trademark SANDIMMUNETM), PROGRAFTM (tacrolimus, also known as FK-506), RAPIMMUNETM (sirolimus, also known as rapamycin), leflunomide (also known as HWA-486), ZENAPAXTM, glucocortcoids, such as prednisolone and its derivatives, corticosteroids, antibodies such as orthoclone (OKT3), cyclophosphamide, methotrexate, 6-mercaptopurine, vincristine, antithymyocyte
  • a conventional immunosuppressant drug such as those above, may thus be administered in an amount substantially less (e.g. 20% to 50% of the standard dose) than when the compound is administered alone.
  • the compounds described herein can be administered at regular intervals over a time period of at least 2 weeks.
  • Compounds and compositions disclosed herein can be locally administered at one or more anatomical sites, such as sites of a transplant, optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent.
  • a pharmaceutically acceptable carrier such as an inert diluent
  • Compounds and compositions disclosed herein can be systemically administered, such as intravenously or orally, optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent, or an assimilable edible carrier for oral delivery. They can be enclosed in hard or soft shell gelatin capsules, can be compressed into tablets, or can be incorporated directly with the food of the patient's diet.
  • the active compound can be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, aerosol sprays, and the like.
  • the tablets, troches, pills, capsules, and the like can also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring can be added.
  • a liquid carrier such as a vegetable oil or a polyethylene glycol.
  • any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed.
  • the active compound can be incorporated into sustained-release preparations and devices.
  • compositions disclosed herein can be administered intravenously, intramuscularly, or intraperitoneally by infusion or injection.
  • Solutions of the active agent or its salts can be prepared in water, optionally mixed with a nontoxic surfactant.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient, which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes.
  • the ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage.
  • the liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants.
  • the prevention of the action of microorganisms can be brought about by various other antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, buffers or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the inclusion of agents that delay absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating a compound and/or agent disclosed herein in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filter sterilization.
  • the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • compounds and agents disclosed herein can be applied in as a liquid or solid. However, it will generally be desirable to administer them topically to the skin as compositions, in combination with a dermatologically acceptable carrier, which can be a solid or a liquid.
  • a dermatologically acceptable carrier which can be a solid or a liquid.
  • Compounds and agents and compositions disclosed herein can be applied topically to a subject's skin to reduce the size (and can include complete removal) of malignant or benign growths, or to treat an infection site.
  • Compounds and agents disclosed herein can be applied directly to the growth or infection site.
  • the compounds and agents are applied to the growth or infection site in a formulation such as an ointment, cream, lotion, solution, tincture, or the like.
  • Drug delivery systems for delivery of pharmacological substances to dermal lesions can also be used, such as that described in U.S. Pat. No. 5,167,649.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like.
  • Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants.
  • Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use.
  • the resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers, for example.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user.
  • Examples of useful dermatological compositions which can be used to deliver a compound to the skin are disclosed in U.S. Pat. Nos. 4,608,392; 4,992,478; 4,559,157; and 4,820,508.
  • Useful dosages of the compounds and agents and pharmaceutical compositions disclosed herein can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • compositions that comprise a compound disclosed herein in combination with a pharmaceutically acceptable carrier.
  • Pharmaceutical compositions adapted for oral, topical or parenteral administration, comprising an amount of a compound constitute a preferred aspect.
  • the dose administered to a patient, particularly a human should be sufficient to achieve a therapeutic response in the patient over a reasonable time frame, without lethal toxicity, and preferably causing no more than an acceptable level of side effects or morbidity.
  • dosage will depend upon a variety of factors including the condition (health) of the subject, the body weight of the subject, kind of concurrent treatment, if any, frequency of treatment, therapeutic ratio, as well as the severity and stage of the pathological condition.
  • Kits for practicing the methods of the invention are further provided.
  • kit any manufacture (e.g., a package or a container) comprising at least one reagent, e.g., anyone of the compounds described in Table 1.
  • the kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention. Additionally, the kits may contain a package insert describing the kit and methods for its use. Any or all of the kit reagents may be provided within containers that protect them from the external environment, such as in sealed containers or pouches.
  • compositions disclosed herein can comprise between about 0.1% and 45%, and especially, 1 and 15%, by weight of the total of one or more of the compounds based on the weight of the total composition including carrier or diluents.
  • dosage levels of the administered active ingredients can be: intravenous, 0.01 to about 20 mg/kg; intraperitoneal, 0.01 to about 100 mg/kg; subcutaneous, 0.01 to about 100 mg/kg; intramuscular, 0.01 to about 100 mg/kg; orally 0.01 to about 200 mg/kg, and preferably about 1 to 100 mg/kg; intranasal instillation, 0.01 to about 20 mg/kg; and aerosol, 0.01 to about 20 mg/kg of animal (body) weight.
  • kits that comprise a composition comprising a compound disclosed herein in one or more containers.
  • the disclosed kits can optionally include pharmaceutically acceptable carriers and/or diluents.
  • a kit includes one or more other components, adjuncts, or adjuvants as described herein.
  • a kit includes one or more immunosuppressant agents, such as those agents described herein.
  • a kit includes instructions or packaging materials that describe how to administer a compound or composition of the kit.
  • Containers of the kit can be of any suitable material, e.g., glass, plastic, metal, etc., and of any suitable size, shape, or configuration.
  • a compound and/or agent disclosed herein is provided in the kit as a solid, such as a tablet, pill, or powder form.
  • a compound and/or agent disclosed herein is provided in the kit as a liquid or solution.
  • the kit comprises an ampoule or syringe containing a compound and/or agent disclosed herein in liquid or solution form.
  • Monoclonal antibodies and flow cytometry Fluorochrome-conjugated mouse anti-human monoclonal antibodies included anti-CD3, CD4, CD25, CD39, CD107a, CD127, CTLA4, Foxp3, LAG3, phosphorylated STAT3 Y705, phosphorylated STAT5 Y694, and phosphorylated H3 serine 10 (BD Biosciences, San Jose, Calif.; eBioscience San Jose, Calif.; Cell Signaling Technology, Boston, Mass.). LIVE/DEAD Fixable Yellow Dead Cell Stain (Life Technologies, Grand Island, N.Y. USA) was used to determine viability. Live events were acquired on a FACSCalibur or LSRII flow cytometer (FlowJo software, ver. 7.6.4; TreeStar, Ashland, Oreg.).
  • MTS Aqueous One Solution Cell Proliferation Assay
  • T-cells were cultured with allogeneic DCs (DC:T-cell ratio of 1:30) for 5 days in RPMI/10% pooled human serum, with alisertib (1.75 ⁇ M), TG101348 (350 nM), a combination of both inhibitors, AJI-214 (750 nM), AJI-100 (750 nM), or DMSO control added once on day 0. After 5 days, T cells were then harvested and directly fixed (Cytofix, BD Biosciences) for 10 min at 37° C.
  • the T cells were permeabilized with icecold methanol (90%, v/v) for at least 20 min at ⁇ 20° C. The cells were stained for expression of CD3 and pH3Ser 10 .
  • STAT3 and STAT5 phosphorylation As indicated, T cells were serum-starved in RPMI treated with DMSO diluent control, alisertib, TG101348, a combination of both inhibitors, AJI-214, or AJI-100 for 4 hours.
  • IL-6-induced pSTAT3 or IL-2-induced pSTAT5 (Y694) was measured by flow cytometry.
  • NSG mice After transplantation and treatment with either AJI-100 or vehicle control, human T cells were isolated from recipient mouse spleens at day +14, stained for pH3Ser 10 or pSTAT3 (+IL-6 stimulation), and analyzed as described.
  • Activated CD4+T conv were characterized by expression of CD25 and CD127, and activated CD8 + T conv (CD3 + and CD4 ⁇ ) were identified by CD25 expression.
  • the absolute number of CD4 + and CD8 + T conv was calculated by flow cytometry using CountBright beads (Life Technologies).
  • T H 1 cells were characterized by expression of CD3, CD4, and intracellular IFN- ⁇ (after an additional 4 to 5 hours of stimulation with PMA/ionomycin).
  • purified CD4 + T cells were used as opposed to bulk T cells.
  • iT reg differentiation and potency iT regs were generated as previously described in the presence of alisertib (1.75 mM), TG101348 (350 nM), both alisertib and TG101348, AJI-214 (750 nM), AJI-100 (750 nM), or DMSO. On day 5, iT regs were isolated and washed to minimize drug carry-over as reported. The T cells were harvested and surface-stained on day 5 for CD3, CD4, CD25, and CD127, followed by fixation and permeabilization (eBioscience) and Foxp3 staining. The absolute number of iT reg was calculated by flow cytometry using CountBright beads (Life Technologies).
  • the purified iT regs were titrated against alloMLRs consisting of responder CD4+CD25 ⁇ T cells (5 ⁇ 10 4 ) from the iT regs donor and fresh allogeneic DCs (1.6 ⁇ 10 3 ) to determine suppressive potency.
  • T cell proliferation was determined by pulsing cells with [ 3 H]thymidine (1 mCi per well). Surface expression of CD39 and LAG3 was evaluated on the iT regs .
  • iT regs production of CTLA4 was assessed by intracellular staining after a 5-hour treatment of PMA/ionomycin, with GolgiStop added during the last 4 hours of incubation.
  • iT regs synthesis of IL-10 and TGF- ⁇ was quantified from supernatants using multiplex cytokine assays after PMA/ionomycin stimulation.
  • ATP Hydrolysis assay iT regs generated in the presence of AJI-241 (750 nM) or DMSO were plated in V-bottom 96 well plates in serum free media at a concentration of 75,000 cells per 100 ⁇ L. ARL67156 (125 ⁇ M) was added or not as indicated. A fixed dose of ATP (50 ⁇ M) was added to the cells and incubated at 37° C. for 45 minutes. ATP consumption was measured by a luminescence assay per the manufacturer's instructions (Promega, CellTiter-Glo Luminescent Cell Viability Assay) and read by a spectrofluorimeter. Percent consumption was calculated as (luminescence of test supernatant/luminescence of 50 ⁇ M ATP cell-free control supernatant) ⁇ 100.
  • Foxp3 TSDR demethylation analysis Foxp3 TSDR demethylation was analyzed among magnetic bead-purified (Miltenyi), allostimulated bisanilinopyrimidine (I)- and DMSO-treated iT regs .
  • the primer selection, procedure for amplifying methylation and demethylation specific TSDR products, genomic DNA isolation, bisulfite conversion, and qPCR were performed as previously reported.
  • IL-6 1 ⁇ 10 5 IU/mL
  • TGF- ⁇ 4 ng/mL
  • anti-IFN- ⁇ monoclonal antibody 10 mg/ml; eBioscience
  • Xenogeneic GVHD model NOD scid gamma (NSG) mice (male or female, 6-24 weeks old) were purchased from Jackson Laboratory (Bar Harbor, Me., USA) and raised per an IACUC-approved protocol in adherence to the NIH Guide for the Care and Use of Laboratory Animals.
  • mice received either (i) alisertib (30 mg/kg daily), TG101348 (45 mg/kg twice a day), a combination of alisertib and TG101348, or vehicle (methylcellulose) by oral gavage or (ii) AJI-100 (50 mg/kg daily) or vehicle (50% polyethylene glycol, 15% 2-hydroxypropyl-b-cyclodextrin, and 10% DMSO in sterile saline) ip from day 0 to day +14.
  • mice were monitored for GVHD clinical scores (K. R. Cooke et al., An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood 88, 3230 (1996)), weight, and premoribund status. As indicated, mice were euthanized on day +14 to study recipient spleen T conv , T regs , T H 1, T H 17, B cells, and T-cell signal transduction.
  • Human CD4 + T reg (CD25 + , CD127 ⁇ , Foxp3 + ), T conv (CD25 + , CD127 + ), T H 1, T H 17, and CD19 + B cells residing in recipient spleens were quantified by flow cytometry.
  • T-cell pH3 ser10 and PSTAT3 were evaluated by flow cytometry.
  • IL-17 ELISPOTs were performed using isolated human T cells from recipient mouse spleen as described above. Tissue samples were prepared, stained (Ventana Medical Systems, Arlington, Ariz.), and imaged (Vista, Calif., USA) to identify human T-cells as previously described. All vertebrate animal work was IACUC-approved.
  • mice were transplanted with human PBMCs as described above, and treated with either AJI-100 (50 mg/kg daily) or vehicle control. Additionally, recipient mice received an inoculum of irradiated U937 cells (ATCC, 10 7 /mouse) on days 0 and +7. Mice were euthanized between days +10 to +12, spleens were harvested, and human CD8 + T-cells were isolated by magnetic bead separation. Fresh U937 target cells were labeled with Calcein-AM for 30 minutes, washed, and then cultured with the purified CD8 + T-cells at varying effector to target ratios for 4 hours at 37° C.
  • human PBMCs were cultured with irradiated U937 cells at a ratio of 1:1 in the presence of bisanilinopyrimidine (I) 750 nM or DMSO for 10 days. Cultures were replenished with media and inhibitors on days +3 and +7, and fresh irradiated U937 cells added on day +7. On day +10, the cells were harvested and CD8 T-cells were isolated by magnetic bead separation (Miltenyi).
  • the tumor lysis assay was performed as described using fresh, Calcein-AM labeled U937 targets.
  • the dose-effect curve for each drug alone was determined based on experimental observations using the median-effect principle and then compared to the effect achieved with a combination of the two drugs to derive a CI value. For this analysis,
  • the bisanilinopyrimidine bisanilinopyrimidine (I) was prepared using a method previously reported (Lawrence, et al., Development of o-chlorophenyl substituted pyrimidines as exceptionally potent aurora kinase inhibitors. J. Med. Chem. 2012, 55, 7392-416.).
  • the bisanilinopyrimidine bisanilinopyrimidine (II) was prepared using a two step route (Scheme I) to prepare other 2,4-dianilinopyrimidines (1). Reaction of 2,4-dichloro-5-fluoropyrimidine with aniline provided the intermediate 1. Intermediate 1 was reacted further with 4-aminobenzamide to give the required bisanilinopyrimidine (II), with HPLC purity >99%.
  • Bisanilinopyrimidine (I) is a potent inhibitor of Aurora kinase A and JAK2 that was designed and synthesized at Moffitt Cancer Center (H. R. Lawrence et al., Development of o-chlorophenyl substituted pyrimidines as exceptionally potent aurora kinase inhibitors. J. Med. Chem. 55, 7392 (2012)).
  • Bisanilinopyrimidine (I) exerted significant suppression over Tcells in alloMLRs, with single agent efficacy at nanomolar concentrations ( FIG. 1B ).
  • the viability of AJI-214 (750 nM) or DMSO treated T-cells was similar after 5 days of co-culture ( FIG. 1E ).
  • Bisanilinopyrimidine (I) demonstrated minimal loss of iT regs and significantly reduced allo T conv compared to DMSO, increasing the iT reg :allo T conv ratio ( FIG. 3E ).
  • bisanilinopyrimidine (I)- or DMSO-treated iT reg were cultured with self alloresponders targeting fresh allogeneic DCs.
  • the bisanilinopyrimidine (I)-treated iT reg not only demonstrated intact suppressive function, their potency was significantly increased by approximately 30% compared to DMSO-treated iT reg ( FIG. 4A ).
  • iT regs were generated as described in the presence of alisertib, TG101348, a combination of both, or DMSO.
  • Aurora kinase A inhibition with alisertib demonstrated superior suppressive capacity compared with either DMSO- or TG101348-exposed iT reg ( FIG. 4B ).
  • the combination of alisertib with TG101348 was similar to alisertib alone ( FIG. 4B ).
  • CD39 blockade significantly weakened the T-cell inhibition by bisanilinopyrimidine (I) ( FIG. 5F ), supporting that CD39+iT regs contribute to the immune suppressive effects of bisanilinopyrimidine (I).
  • FIGS. 5G-5J no difference in their expression of LAG3, CTLA4, or production of IL-10 or TGF-beta after exposure to bisanilinopyrimidine (I) or DMSO.
  • Bisanilinopyrimidine (II) is an Aurora kinase A/JAK2 inhibitor (Moffitt Cancer Center) that differs from bisanilinopyrimidine (I) by only a chlorine to hydrogen substitution at the ortho position of its phenyl ring enhancing its solubility.
  • the bisanilinopyrimidine (II) and bisanilinopyrimidine (I) analogues both inhibit Aurora kinase A and JAK2 with similar potency.
  • bisanilinopyrimidine (I) reduced responder T-cell proliferation in alloMLRs at nanomolar concentrations (IC 50 200 nM, FIG. 7A ).
  • mice were treated with bisanilinopyrimidine (II) at 50 mg/kg daily i.p. or vehicle control from day 0 to day +14. JAK2 inhibition was confirmed in vitro, where human T-cells stimulated with IL-6 expressed less STAT3 phosphorylation with bisanilinopyrimidine (II) exposure compared with DMSO ( FIG. 7B ). Harvested human T-cells from the mice at day +14 showed less H3 ser10 phosphorylation with bisanilinopyrimidine (II) compared to vehicle, confirming Aurora inhibition ( FIG. 7C ).
  • the vehicle control treated mice developed acute xenogeneic GVHD (including fur loss, skin changes, weight loss, and kyphosis) by the third week of the transplant with a median survival of 34 days ( FIGS. 7D-7E ).
  • the overall survival of the bisanilinopyrimidine (II)-treated mice was 71.4% during the 56 days of observation, while none of the vehicle-treated mice survived past day +45 ( FIG. 7F ).
  • the average GVHD clinical scores were ⁇ 2 among the surviving bisanilinopyrimidine (II)-treated mice at day +56 ( FIG. 7E ), where 60% showed limited fur/skin changes without significant weight loss ( FIGS. 7D-7E ).
  • FIGS. 8H-8J T reg frequency in the liver was similar among both treatment groups, however ( FIG. 8K ).
  • T-cell costimulation and cytokine activation independently contribute to GVHD, but control of donor alloresponses is incomplete when targeting either pathway alone. It has been disclosed herein that GVHD prevention with intact GVL can be accomplished by dual inhibition of Aurora kinase A and JAK2, respectively attenuating CD28 costimulation and IL-6-mediated signal transduction. Concurrent blockade of Aurora kinase A and JAK2 yields synergistic immune suppression over human allogeneic T-cells in vitro, preserves iT reg differentiation, and significantly enhances iT reg suppressive potency. These characteristics are distinct from CNI-based GVHD prophylaxis, which abrogates TCR function and indiscriminately suppresses donor T-cells.
  • Blockade of Aurora kinase A or JAK2 induces pathway-specific effects on developing iT reg and T H 17. It was observed that the dual pathway inhibitor, bisanilinopyrimidine (I), significantly increased the suppressive potency of allo-antigen specific iT reg . The data supports that the enhanced iT reg potency is largely a function of Aurora kinase A inhibition. While JAK2 blockade with TG101348 improved iT reg function compared with DMSO, iT regs previously exposed to alisertib profoundly eliminated T conv proliferation.
  • alisertib was unable to prevent RORgammaT expression in na ⁇ ve CD4 + T-cells responding to allo-antigen unlike AJI-214 or TG101348.
  • IL-6 receptor signal transduction facilitates T H 17 development
  • the data confirm that JAK2 blockade with bisanilinopyrimidine (I) or TG101348 significantly restrains T H 17 differentiation.
  • CD39 + T regs correlate with clinical outcomes in autoimmune diseases (R. S. Peres et al., Low expression of CD39 on regulatory T cells as a biomarker for resistance to methotrexate therapy in rheumatoid arthritis. Proc. Natl. Acad. Sci. USA 112, 2509 (2015); A. Thiolat et al., Interleukin-6 receptor blockade enhances CD39 + regulatory T cell development in rheumatoid arthritis and in experimental arthritis. Arthritis & Rheumatol. 66, 273 (2014)). A decrease in CD39 + T regs is associated with methotrexate-failure among rheumatoid arthritis (RA) patients.
  • RA rheumatoid arthritis
  • IL-6 neutralization with tocilizumab increases CD39 + T regs in a similar group of RA patients (38). It was verified that bisanilinopyrimidine (I)-treated iT reg degraded extracellular ATP more efficiently than those exposed to DMSO. This response was impaired by blocking CD39 with the inhibitor ARL67156. Additionally, neutralization of CD39 activity significantly attenuated the immune suppressive effect of dual Aurora kinase A/JAK2 inhibition in nT reg depleted alloMLRs. This data supports that increased CD39 expression is relevant to the enhanced iT reg function mediated by bisanilinopyrimidine (I).
  • Bisanilinopyrimidine (II) differs from bisanilinopyrimidine (I) by a single chlorine to hydrogen substitution on its phenyl ring to facilitate in vivo solubility for mouse studies, but the compounds are otherwise chemically and functionally similar.
  • Target inhibition was confirmed as bisanilinopyrimidine (II) and bisanilinopyrimidine (I) significantly reduced the phosphorylation of both STAT3 and H3 ser10 in human T-cells.
  • combined Aurora kinase A/JAK2 blockade permitted IL-2-induced STAT5 activation in T-cells, compared with resting, unstimulated controls.
  • the selective inhibition of Aurora kinase A and JAK2 paired with preserved common gamma chain cytokine signaling establishes a platform to control alloreactivity while maintaining antigen-specific T reg and CTL responses.
  • CNI-free GVHD prophylaxis is an important concept in improving patient outcomes after clinical transplantation.
  • the challenges of CNI-based GVHD prevention are clear; as CNIs offer incomplete protection from severe GVHD and render the donor immune system poorly equipped to counter post-transplant relapse.
  • the novel concept described here may represent a translatable CNI-free approach at GVHD prevention.
  • CNI-free GVHD prophylaxis strategies include T-cell depletion of the allograft (M. C. Pasquini et al., Comparative outcomes of donor graft CD34 + selection and immune suppressive therapy as graft-versus-host disease prophylaxis for patients with acute myeloid leukemia in complete remission undergoing HLA-matched sibling allogeneic hematopoietic cell transplantation. J. Clinical Oncol. 30, 3194 (2012)) or the use of post-transplant cyclophosphamide (C. G.
  • Allogeneic mixed leukocyte reactions are standard assays used to assess human T cell proliferation against polyclonal or antigen-specific stimuli.
  • alloMLRs consisting of human T cells and allogeneicmonocyte-derived dendritic cells (DCs)
  • the JAK2 inhibitor TG101348 reduced alloreactive T cell proliferation at concentrations of 350 nM and greater as previously reported ( FIG. 9A ).
  • the Aurora kinase A inhibitor alisertib suppressed the proliferative response of T cells in alloMLRs with a median inhibitory concentration (IC 50 ) of 10 mM ( FIG. 9A ).
  • Synergistic suppression of T cells allostimulated by DCs was achieved when TG101348 and alisertib were added together at a ratio of 1:5, respectively, with a calculated combination index (CI) of ⁇ 1 using the Chou-Talalay method ( FIG. 9A ).
  • the observed IC 50 of the combination correlated with 350 nM TG101348 and 1.75 mM alisertib ( FIG. 9A ).
  • the chemical analogs AJI-214 and AJI-100 were designed and synthesized at the Moffitt Cancer Center and shown to inhibit Aurora kinase A and JAK2 with similar potency.
  • AJI-100 differs from AJI-214 by a single chlorine to hydrogen substitution at the ortho position of its phenyl ring, enhancing its solubility, hence its preferred use in vivo ( FIGS. 9B-9C ). Because AJI-100 is tolerated in mouse models, a kinase target screen was performed on AJI-100 to verify its activity against Aurora kinase A and JAK2 among a panel of 140 kinases. Aurora kinase A and JAK2 were among the top three kinases inhibited by AJI-100.
  • AJI-100 also inhibits 5′ AMP activated protein kinase (AMPK) and exhibits slightly more potent suppression of Aurora kinase B than alisertib.
  • AMPK 5′ AMP activated protein kinase
  • AJI-214 and AJI-100 exerted significant suppression of T cells in alloMLRs, with single-agent efficacy at nanomolar concentrations (P ⁇ 0.05; FIGS. 9B-9C ).
  • the AJI analogs suppressed alloreactive T cell proliferation similar to the potency of alisertib (1.75 mM) and TG101348 (350 nM) combined ( FIG. 9D ).
  • AJI-214 and AJI-100 also exhibited similar target inhibition of Aurora kinase A and JAK2 signal transduction in human T cells, reducing the phosphorylation of histone 3 serine 10 (pH3Ser 10 ) and STAT3 (pSTAT3) Y705, respectively ( FIGS. 9E-9H ).
  • alisertib only inhibited pH3Ser 10 ( FIGS. 9E-9F )
  • TG101348 only inhibited pSTAT3 ( FIGS. 9G-9H ).
  • DMSO alisertib, TG101348, a combination of alisertib and TG101348, AJI-214, or AJI-100 was added to allogeneic cocultures of DC-stimulated T cells.
  • Activated CD4 + T conv were identified as coexpressing CD25 and CD127, and the latter assisted in excluding T regs from the analysis.
  • iT regs were identified as CD4 + , CD127 ⁇ , CD25 + , and Foxp3 + .
  • iT reg conversion from na ⁇ ve CD4 + precursors was variably reduced by all of the compounds compared to DMSO ( FIGS. 10F-10G ), and Aurora A inhibition appeared to exert greater iT reg impairment than JAK2 inhibition ( FIG. 10G ).
  • IL-2-induced STAT5 phosphorylation which is required for T reg development, remained intact among T cells treated with alisertib, TG101348, a combination of alisertib and TG101348, or the AJI analogs compared to DMSO ( FIG. 10H ).
  • AJI-214 and AJI-100 were confirmed to exhibit identical suppressive potency in regard to Aurora A and JAK2 signal transduction and human T cell proliferation assays. Therefore, AJI-214 was used as the representative bispecific analog for additional iT reg -based in vitro mechanistic tests. Given that iT regs are derived from phenotypically plastic na ⁇ ve CD4 + T cells, it was confirmed that demethylated Foxp3 T re g-specific demethylated region (TSDR) was similar among AJI-214-exposed and DMSO-exposed iT regs ( FIG. 11A ).
  • TSDR demethylated Foxp3 T re g-specific demethylated region
  • AJI-214-treated or DMSO-treated iT reg were cultured with autologous T cells targeting fresh allogeneic DCs.
  • Aurora kinase A versus JAK2 blockade contributed to this enhanced suppression by the iT regs was explored.
  • Antigen-specific iT regs were generated from CD25-depleted CD4 + T cells in the presence of alisertib, TG101348, a combination of alisertib and TG101348, or DMSO.
  • the combination of alisertib with TG101348 was similar to alisertib alone ( FIG. 11C ).
  • the mechanism supporting the increased iT reg function observed with AJI-214 was investigated.
  • ARL67156 was added to alloMLRs consisting of natural T reg -depleted CD4 + T cell responders with AJI-214 or DMSO. This eliminated potential interference from CD39 + natural T reg within the allogeneic coculture and ensured that the only T regs present in the system were induced. Moreover, ARL67156 would primarily affect the iT reg as T conv express negligible amounts of CD39.
  • iT reg suppression no difference in their expression of LAG3 and CTLA4 or production of IL-10 or transforming growth factor- ⁇ (TGF- ⁇ ) was found after exposure to AJI-214 or DMSO ( FIG. 12G-12J ).
  • a xenogeneic GVHD model was used to investigate the in vivo efficacy of dual Aurora kinase A/JAK2 blockade to specifically evaluate effects on human immune responses.
  • Recipient NOD nonobese diabetic
  • NSG scid g mice
  • PBMCs peripheral blood mononuclear cells
  • the drug combination significantly delayed the onset and severity of GVHD, compared to vehicle or TG101348 alone (P ⁇ 0.0001 and P 0.0001, respectively; FIGS. 13A-13B ).
  • There was also a suggestion toward an improved median survival with the drug combination compared to alisertib (50.5 versus 41 days; P not significant).
  • the bispecific inhibitor AJI-100 was used to test the in vivo efficacy of single agent blockade of Aurora A and JAK2 as GVHD prevention.
  • AJI-100 offers identical on-target inhibition and immunosuppressive properties as AJI-214 but exhibits superior bioavailability. Compared to using the combination of alisertib and TG101348, AJI-100 had the advantage of being given once daily by intraperitoneal injection and avoided the need for sustained gavage dosing. Additionally, the single bispecific compound provided a pharmacologically cleaner approach by eliminating the variability in pharmacokinetics between the two drugs in combination. The recipient mice were transplanted with human cell as described. AJI-100 (50 mg/kg) or vehicle control was administered daily by intraperitoneal injection from days 0 to +14.
  • On target inhibition of Aurora A and JAK2 was confirmed among human T cells harvested from recipient spleens at day +14.
  • An established method was used to generate human antitumor CTL in vivo and then test their specific killing.
  • CD8 + CTLs were generated in xenotransplanted mice receiving AJI-100 or vehicle control, where an inoculum of irradiated U937 cells was administered on day 0 and day +7. Unvaccinated, xenotransplanted mice served as negative control. Despite its immunosuppressive activity, AJI-100 did not inhibit CTL generation because CD8 + CTL from AJI-100-treated and vehicle-treated mice demonstrated similarly enhanced killing capacity against U937 targets in vitro, compared to unvaccinated controls ( FIG. 13G ). These data support that although AJI-100 significantly reduces GVHD, it also preserves antitumor CTL responses.
  • AJI-100 also exerted a suppressive effect on CD8 + T cell and CD19 + B cell reconstitution as determined by absolute numbers compared to vehicle.
  • the frequencies of CD4 + and CD8 + T cells and CD19 + B cells were similar among AJI-100-treated and vehicle-treated mice.
  • mice Using the nondiagnostic mastectomy skin from consenting donors, a 1 ⁇ 1 cm skin graft was transplanted onto immunodeficient NSG mice dorsally. After 30 days of healing, 5 ⁇ 10 6 peripheral blood mononuclear cells (PBMC) from an HLA-disparate random donor was given by i.p. injection. A cohort of mice only received a skin graft without PBMCs, as negative rejection controls. The transplanted mice were treated with AJI-100 (50 mg/kg), a dual JAK2/Aurora A kinase inhibitor, or vehicle daily by i.p. injection. Mice were humanely euthanized on day +21 to assess human anti-human skin rejection pathology.
  • PBMC peripheral blood mononuclear cells
  • T cell costimulation and cytokine activation independently contribute to GVHD, but control of donor alloresponses is incomplete when targeting either pathway alone.
  • GVHD prevention can be accomplished by dual inhibition of Aurora kinase A and JAK2, attenuating CD28 costimulation and IL-6-mediated signal transduction, respectively, without ablating potential antitumor CTL responses.
  • Concurrent blockade of Aurora kinase A and JAK2 yields synergistic immunosuppression of human allogeneic T cells in vitro, significantly enhances iT reg -suppressive potency, and enhances the ratio of T regs to activated T conv in vivo.
  • CNI-based GVHD prophylaxis which inhibits TCR function and indiscriminately suppresses donor T cells.
  • the lack of selectivity by CNIs results in a failure to achieve donor immune tolerance toward the host and mitigates the graft-versus-leukemia (GVL) potential of the allograft.
  • VTL graft-versus-leukemia
  • Inhibition of Aurora kinase A or JAK2 activity individually suppressed human T cell proliferation in alloMLRs, and synergy was achieved in vitro with simultaneous blockade of both signal transduction pathways.
  • a xenogeneic model was used to study human T cell responses in vivo after Aurora kinase A and JAK2 blockade, understanding that the lack of recipient conditioning does differ from clinical practice in allogeneic HCT. It was shown that alisertib combined with TG101348 significantly delays GVHD. However, the combination of inhibitors appears additive at best in vivo and does not completely eliminate GVHD. The bispecific inhibitor AJI-100 significantly reduced GVHD and improved survival compared to vehicle control. It was surmised that the apparent enhanced in vivo activity of AJI-100 compared to alisertib plus TG101348 may be due to inherent kinase selectivity.
  • JAK2 blockade is capable of restraining STAT3 phosphorylation and resultant T H 17 differentiation. Additionally, JAK2 inhibition appears to exhibit less inhibition of iT regs compared to Aurora kinase A blockade. Last, inhibition of JAK2, Aurora kinase A, or both JAK2 and Aurora kinase A equally impaired the T H 1 response in vitro. Selective inhibition of Aurora kinase A and JAK2 paired with preserved common gamma-chain cytokine signaling establishes a platform to control alloreactivity while permitting antigen-specific T reg and CTL responses. However, there are several limitations of this study that deserve further consideration.
  • the xenogeneic model is well suited to test whether concurrent Aurora kinase A/JAK2 inhibition can prevent GVHD mediated by human cells in vivo, it does not entirely replicate human GVHD pathogenesis.
  • the recipient mice do not receive transplant conditioning, unlike human patients, and this may affect GVHD target-organ injury, host antigen presentation, and the production of relevant cytokines such as IL-6.
  • Our work demonstrates that AJI-100 permits the generation and function of antitumor CTL, but it is important to recognize that such experiments are supportive and not definitive in assessing whether the bispecific inhibitor preserves GVL in vivo.
  • small-molecule inhibitors can exhibit off-target inhibition, as observed with AJI-100 and its suppression of AMPK.
  • CNI-free GVHD prophylaxis is an important concept in improving patient outcomes after clinical transplantation.
  • the challenges of CNI-based GVHD prevention are clear because CNIs offer incomplete protection from severe GVHD and render the donor immune system poorly equipped to counter posttransplant relapse.
  • the concept described here may represent a translatable CNI free approach at GVHD prevention.
  • the bispecific inhibitor AJI-100 is an attractive alternative because it does not require ex vivo allograft modification or the need to expose freshly infused donor stem cells to potent alkylators. Hence, further investigation of dual Aurora kinase A/JAK2 inhibition is merited to promote selective control of donor immune responses after alloHCT.

Abstract

Disclosed herein are compounds and methods for reducing the risk of developing, preventing, or treating graft versus host disease (GVHD) in a subject. The compounds can concurrently block Aurora kinase A and JAK2 signal transduction which synergistically suppresses alloreactive human T-cells in vitro, prevents xenogeneic graft-versus-host disease without impairing anti-tumor responses, and promotes the development and suppressive potency of CD39+ inducible Treg. In certain aspects, disclosed are compounds of Formula I-V.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of priority to U.S. Provisional Application 62/307,030, filed Mar. 11, 2016, which is incorporated by reference herein.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
  • This invention was made with Government Support under Grant No. K08 HL11654701A1 and Grant No. P30-CA076292 awarded by the National Institutes of Health. The Government has certain rights in the invention.
  • FIELD
  • The subject matter disclosed herein relates generally to graft versus host disease (GVHD). More specifically, the subject matter disclosed herein relates to inhibitors of Aurora kinase and JAK2 and their use in preventing or treating GVHD.
  • BACKGROUND
  • Graft-versus-host disease (GVHD) is a leading cause of non-relapse mortality after allogeneic hematopoietic cell transplantation (alloHCT). Broadly acting calcineurin inhibitors (CNI) are often used to prevent GVHD, but exert undesirable antagonistic effects on T-cell receptor signaling and regulatory T-cell (Treg) differentiation and function (M. Vaeth et al., Selective NFAT targeting in T cells ameliorates GvHD while maintaining antitumor activity. Proc. Natl. Acad. Sci. USA 112, 1125 (2015); K. Singh et al., Superiority of rapamycin over tacrolimus in preserving nonhuman primate Treg half-life and phenotype after adoptive transfer. Am. J. Transplant.: 14, 2691 (2014); R. Zeiser et al., Inhibition of CD4+CD25+ regulatory T-cell function by calcineurindependent interleukin-2 production. Blood 108, 390 (2006)). The lack of immune selectivity by CNIs places the alloHCT recipient at risk for opportunistic infections and relapse of their underlying hematologic malignancy. An alternative approach at GVHD prevention is to concurrently target CD28 costimulation and IL-6 receptor activation of T-cells by inhibiting key signal transduction molecules in these pathways.
  • CD28 costimulation contributes to T-cell alloreactivity and GVHD. GVHD in rodents is ameliorated by transplantation of CD28 negative compared to wild type T-cells (P. Tan et al., Induction of alloantigen-specific hyporesponsiveness in human T lymphocytes by blocking interaction of CD28 with its natural ligand B7/BB1. J. Experi. Med. 177, 165 (1993); X. Z. Yu et al., CD28-specific antibody prevents graft versus-host disease in mice. J. Immunol. 164, 4564 (2000)). Blockade of ligand interactions between CD80/CD86 and CD28 with neutralizing antibody also reduces murine GVHD. CD28 signal transduction activates mTOR and Aurora kinase in T-cells (J. Song et al., The kinases aurora B and mTOR regulate the G1-S cell cycle progression of T lymphocytes. Nature Immunol. 8, 64 (2007)). mTOR is a known pharmacologic target in GVHD prophylaxis (C. Cutler et al., Tacrolimus/sirolimus vs tacrolimus/methotrexate as GVHD prophylaxis after matched, related donor allogeneic HCT. Blood 124, 1372 (2014); J. Pidala et al., Prolonged sirolimus administration after allogeneic hematopoietic cell transplantation is associated with decreased risk for moderate-severe chronic graft vs. host disease. Haematologica (2015); J. Pidala et al., A randomized phase II study to evaluate tacrolimus in combination with sirolimus or methotrexate after allogeneic hematopoietic cell transplantation. Haematologica 97, 1882 (2012)), and its blockade is selectively toxic to Tconv compared to Treg (R. Zeiser et al., Differential impact of mammalian target of rapamycin inhibition on CD4+CD25+Foxp3+ regulatory T cells compared with conventional CD4+ T cells. Blood 111, 453 (2008)). However, GVHD prevention with sirolimus, an mTOR inhibitor, is inadequate if not combined with a CNI (L. Johnston et al., Sirolimus and mycophenolate mofetil as GVHD prophylaxis in myeloablative, matched-related donor hematopoietic cell transplantation. Bone Marrow Transplant. 47, 581 (2012)). Aurora kinase isoforms ubiquitously regulate mitotic progression and cellular polarity in human cells (M. Carmena, W. C. Earnshaw, The cellular geography of aurora kinases. Nature Rev. Mol. Cell Biol. 4, 842 (2003); S. M. Lens et al., Shared and separate functions of polo-like kinases and aurora kinases in cancer. Nature Rev. Cancer 10, 825 (2010)), but also mediate T-cell costimulation. Aurora kinase is able to activate substrates required for T-cell proliferation that are shared with mTOR, and is only partially curtailed by sirolimus. Complete inhibition of Aurora activity requires direct blockade of the molecule or targeting upstream phosphatidylinositol-3-OH kinase (PI(3)K). Increased Aurora kinase A expression was recently correlated with acute GVHD in human recipients of alloHCT, as well as experimental models studying murine and nonhuman primate (NHP) hosts. Accordingly, treatment with sirolimus did not control Aurora activity in the transplanted NHPs. Moreover, pharmacologic blockade of this novel pathway with alisertib, an Aurora kinase A inhibitor, significantly delayed the onset of GVHD in mice. These data revealed that Aurora kinase A alone does not fully control alloreactivity (S. N. Furlan et al., Transcriptome analysis of GVHD reveals aurora kinase A as a targetable pathway for disease prevention. Sci. Translational Med. 7, 315ra191 (2015)).
  • IL-6 receptor signaling polarizes T H1 and TH17 cells that are effectors in GVHD, and impairs Tregs that modulate GVHD (B. C. Betts et al., Janus kinase-2 inhibition induces durable tolerance to alloantigen by human dendritic cell-stimulated T cells yet preserves immunity to recall antigen. Blood 118, 5330 (2011); B. C. Betts, A. Veerapathran, J. Pidala, X. Z. Yu, C. Anasetti, STAT5 polarization promotes iTregs and suppresses human T-cell alloresponses while preserving CTL capacity. J. Leukocyte Biol. 95, 205 (2014); J. Choi et al., Pharmacologic blockade of JAK1/JAK2 reduces GvHD and preserves the graft-versus-leukemia effect. PloS one 9, e109799 (2014); A. Laurence et al., STAT3 transcription factor promotes instability of nTreg cells and limits generation of iTreg cells during acute murine graft-versus-host disease. Immunity 37, 209 (2012); R. Zeiser et al., Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multi-center survey. Leukemia, (2015)). IL-6 activates JAK2 and leads to downstream phosphorylation of STAT3 (B. C. Betts et al., Anti-IL6-receptor-alpha (tocilizumab) does not inhibit human monocyte-derived dendritic cell maturation or alloreactive T-cell responses. Blood 118, 5340 (2011)). It was observed that CD4+ T-cell JAK2 activity by IL-6 is increased among alloHCT recipients who later develop GVHD (B. C. Betts et al., CD4+ T cell STAT3 phosphorylation precedes acute GVHD, and subsequent TH17 tissue invasion correlates with GVHD severity and therapeutic response. J. Leukocyte Biol., (2015)). Anti-IL-6 receptor antibody combined with a CNI ameliorates human GVHD, but it does not influence T H1, TH17, or Treg differentiation (G. A. Kennedy et al., Addition of interleukin-6 inhibition with tocilizumab to standard graft-versus-host disease prophylaxis after allogeneic stem-cell transplantation: a phase 1/2 trial. The Lancet. Oncology 15, 1451 (2014)). JAK2 inhibition conversely polarizes natural Treg responses, and inhibits T H1 and TH17 development in vitro. However, selective blockade of JAK2 alone does not provide lasting protection in murine GVHD. This observation is distinct from JAK1/JAK2 inhibition, where co-blockade of JAK1 acts broadly to reduce GVHD (Blood 123, 3832 (2014)) as well as beneficial anti-viral CTL (S. Spoerl et al., Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood 122, 3843 (2013); A. Heine, P. Brossart, D. Wolf, Ruxolitinib is a potent immunosuppressive compound: is it time for anti-infective prophylaxis? Blood 122, 1192 (2013)). These data show that JAK2 activation selective inhibition is insufficient to completely prevent GVHD despite favorable immune effects.
  • Thus there is a need for new compositions and methods for treating GVHD. The compositions and methods disclosed herein address these and other needs.
  • SUMMARY
  • In accordance with the purposes of the disclosed materials and methods, as embodied and broadly described herein, the disclosed subject matter, in one aspect, relates to compounds, compositions and methods of making and using compounds and compositions. In specific aspects, the disclosed subject matter relates to reducing the risk of, preventing, or treating graft versus host disease (GVHD) in a subject. More specifically, the subject matter disclosed herein relates to inhibitors of Aurora kinase A and JAK2. In more specific examples, the disclosed subject matter relates to concurrent inhibition of Aurora kinase A and JAK2 and their use in reducing the risk of, preventing, or treating GVHD. GVHD can be attributed to a solid organ transplant, tissue graft, or a cellular transplant.
  • The methods described herein can include administering to a subject at risk of developing or having GVHD, a composition comprising a compound of the following formula
  • Figure US20220281828A1-20220908-C00003
  • wherein
    • R1 is selected from the group consisting of H, Cl, F, Br, I, CN, NO2, NH2, CF3, CO2H, CO2NH2, CO2NHR5, CO2R5, C(O)R5, C(O)NH2, C(O)NHR5, and C1-C6 alkyl optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R2 is selected from the group consisting of H, OH, CN, NO2, NH2, optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, wherein optionally substituted substituents are optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • or R1 and R2 together form a fused cycloalkyl, cycloheteroalkyl, aryl, or heteraryl group;
    • each R3 is selected, independently, from the group consisting of SO2NH2, SO2NHR5, NHSO2R5, NHCO2R5, NHC(O)R5, NHCONHR5, F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted cycloheteroaryl, and optionally substituted fused cycloheteroalkyl, wherein optionally substituted substituents are optionally substituted with sulfonyl;
    • each R4 is selected, independently, from the group consisting of F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, COOH, C(O)NH2, C(O)R5, C(O)NHR5, CH2C(O)R5, SO2NH2, SO2NHR5, CONHSO2R5, optionally substituted phenyl, optionally substituted OPhenyl, tetrazole, piperadinyl, piperazinyl, and morpholinyl, wherein optionally substituted substituents are optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • each R5 is selected, independently, from optionally substituted C1-C6 alkyl, optionally substituted C1-C6 cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, and optionally substituted heteroalkyl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, cycloalkyl, cycloheteroalkyl, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R7 is selected from the group consisting of H, C1-C6 alkyl, C1-C6 alkoxyl, halide, hydroxyl, cyano, nitro, and amino;
    • n is 0-5; and
    • m is 1-5,
      or a pharmaceutically acceptable salt thereof.
  • In certain aspects, the composition can include a compound of the following formula:
  • Figure US20220281828A1-20220908-C00004
  • wherein
    • R1 is selected from the group consisting of H, Cl, F, Br, I, C1-C6 alkyl, CN, NO2, and NH2;
    • R2 is selected from the group consisting of H, F, and Cl;
    • each R3 is selected, independently, from the group consisting of Cl, Br, F, COOH, CF3, CN, phenyl, OCH3, COR5, CONH2, CONR5, and COONH2; and
    • each R4 is selected, independently, from the group consisting of H, COOH, CONH2, CONR5, SO2NH2, CONSO2R5, tetrazole, 4-morpholine, and COR5;
    • each R5 is selected, independently, from the group consisting of optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heteroalkyl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, cycloalkyl, cycloheteroalkyl, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • n is 0-5; and
    • m is 1-5,
      or a pharmaceutically acceptable salt thereof.
  • In certain aspects, the composition can include a compound having one of the following formulas:
  • Figure US20220281828A1-20220908-C00005
  • wherein
    • R1 is selected from the group consisting of H, Cl, F, Br, I, CH3 and NH2;
    • R2 is selected from the group consisting of H, F, and Cl;
    • R3 is selected from the group consisting of 2-Cl, 2-Br, 2-F, 2-COOH, 2-CF3, 2-CN, 2-phenyl, 2-OCH3, 2-COONH2, 4-COOH, and 4-OCH3; and
    • R4 is selected from the group consisting of H, COOH, 2-CONH2, 4-CONH2, SO2NH2, tetrazole, and 4-morpholine.
  • In certain examples of Formulas I, II, and IIIA, R3 is 2-Cl, and n is 1. In some examples, R4 is 4-COOH, and m is 1. In other examples, R4 is 4-CONH2, and m is 1. In some examples, R3 is 2-Cl, n is 1, m is 1, and R4 is COOH, COR5, CONH2, CONR5, or CONSO2R5, wherein R5 is C1-C6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl. In other examples, R3 is H, m is 1, and R4 is COOH, COR5, CONH2, CONR5, or CONSO2R5, wherein R5 is C1-C6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl. In a preferred example, R1 is Cl, R2 is H, R3 is H, m is 1, and R4 is 4-CONH2. In other examples n is 0.
  • The compositions described herein can be administered at a dose of about 0.1 mg/kg to about 100 mg/kg.
  • Additional advantages will be set forth in part in the description that follows, and in part will be obvious from the description, or may be learned by practice of the aspects described below. The advantages described below will be realized and attained by means of the elements and combinations particularly pointed out in the appended claims. It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive.
  • BRIEF DESCRIPTION OF THE FIGURES
  • The accompanying figures, which are incorporated in and constitute a part of this specification, illustrate several aspects described below.
  • FIGS. 1A-1E show synergistic immune suppression with combined inhibition of Aurora kinase A and JAK2. In FIG. 1A, T-cells were stimulated with DCs (DC:T-cell ratio 1:30) exposed to either TG101348 (JAK2 inhibitor), alisertib (Aurora kinase A inhibitor), or both at a fixed ratio of 1:5 respectively at varying concentrations once on day 0. Proliferation was determined by fluorescence assay on day 5, with % proliferation based on DMSO control. Graph depicts combination synergy (IC50=TG101348 350 nM and alisertib 1.75 μM), showing 1 representative independent experiment of 2 performed in triplicate. Combination index (CI) calculated per Chou and Talalay method. In FIG. 1B, AlloMLR (DC:T-cell ratio 1:30) was treated with bisanilinopyrimidine (I) (dual JAK2/Aurora kinase A inhibitor) or DMSO once on day 0. Proliferation was determined by colorimetric assay on day 5, with % proliferation based on DMSO control. IC50=100 nM. Graph shows average triplicate means±SEM from 3 independent experiments (ANOVA). In FIGS. 1C-1D, bar graphs depict mean gated CD3+ Tcell STAT3 (target of JAK2) or H3 Ser10 (target of Aurora) phosphorylation±SD following 5-day allogeneic DC stimulation treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (unpaired t-test). Representative contour plots are shown with phosphorylation based on isotype control. In FIG. 1E, a bar graph shows mean T-cell viability±SD by LIVE/DEAD Yellow exclusion from 4 independent experiments (unpaired t-test). *P<0.05.
  • FIGS. 2A-2I show concurrent blockade of Aurora kinase A and JAK2 selectively suppresses alloreactive Tconv, while sparing responder Tregs. T-cells were stimulated with DCs (DC:Tcell ratio 1:30) and treated with bisanilinopyrimidine (I) or DMSO once on day 0. In FIGS. 2A-2D, bar graphs show percent means of CD4+ alloreactive Tconv (CD25+, CD127+), CD4+ Treg (CD25+, CD127), and Treg:allo Tconv ratio±SD from 5 independent experiments on day 5 of culture (unpaired t-test). Representative contour plots show CD4+ alloreactive Tconv (CD25+, CD127+) and CD4+Treg (CD25+, CD127-) populations exposed to bisanilinopyrimidine (I) or DMSO after 5 days of DCallostimulation. In FIG. 2E, box plots depict mean Treg versus Tconv proliferation by Cell Trace Violet dilution±SD among identically treated alloMLRs exposed to bisanilinopyrimidine (I) or DMSO from 3 independent experiments (unpaired t-test). FIG. 2F is a representative histogram that shows proliferation in each T-cell compartment with respect to bisanilinopyrimidine (I) or DMSO treatment. In FIG. 2G, a bar graph shows mean gated CD4+T-cell STAT5 phosphorylation±SD following brief IL-2 stimulation treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (ANOVA). In FIG. 2H, a histogram depicts intracellular Foxp3 expression among CD4+, CD25+, CD127 Tregs following 5-day DCallostimulation exposed to bisanilinopyrimidine (I) or DMSO. Data representative of 5 experiments. In FIG. 2I, AlloMLRs (DC:T-cell ratio 1:30) were treated with alisertib (Aurora kinase A inhibitor), TG101348 (JAK2 inhibitor), a combination of both, or DMSO once on day 0. Representative contour plots show CD4+ alloreactive Tconv (CD25+, CD127+) and CD4+ Treg (CD25+, CD127) populations exposed to either inhibitor, the combination, or DMSO after 5 days of DCallostimulation. Data are from one representative experiment of 2. NS=not significant, *P<0.05, **P=0.001-0.01.
  • FIGS. 3A-3E show dual blockade of Aurora kinase A and JAK2 selectively increases the ratio of iTreg to alloreactive Tconv. In FIG. 3A, a representative contour plots show Treg (CD25+, CD127) depletion of naïve CD4+ T-cell responders at outset of alloMLR (DC:T-cell ration 1:30), followed by induction of iTreg versus allo Tconv (CD25+, CD127+) after 5 days of culture exposed to AJI-214 or DMSO. In FIGS. 3B-3C, bar graphs show mean frequency of CD4+ iTreg and allo Tconv in 5-day allogeneic co-cultures treated with bisanilinopyrimidine (I) or DMSO±SD from 4 independent experiments (unpaired t-test). In FIG. 3D, a bar graph depicts triplicate mean % demethylation of Foxp3±SEM among iTregs in alloMLRs treated with bisanilinopyrimidine (I) or DMSO from 4 independent experiments (unpaired t-test). In FIG. 3E, bar graphs show replicate means of absolute numbers of iTreg, allo Tconv, and the ratio of iTreg:allo Tconv±SD from 5-day MLRs treated with Alisertib 1.75 μM, TG101348 350 nM, a combination of both, bisanilinopyrimidine (I) 750 nM, or DMSO control (ANOVA). Data are from one representative experiment of 2 performed in triplicate.
  • FIGS. 4A-4B show combined inhibition of Aurora kinase A and JAK2 enhances antigen-specific iTreg suppressive potency. In FIG. 4A, the suppressive capacity of sorted, DC-allostimulated iTregs previously exposed to bisanilinopyrimidine (I) or DMSO was tested at different ratios of iTreg to self T-cell responders stimulated by original allogeneic DCs (DC:responder T-cell ratio 1:30) in fresh alloMLRs. No additional bisanilinopyrimidine (I) or DMSO was added. The bar graph shows triplicate means of % proliferation±SEM based on 3H-thymidine incorporation on day 6 from 3 independent experiments (ANOVA). In FIG. 4B, the potency of iTregs generated in the presence of alisertib (Aurora kinase A inhibitor), TG101348 (JAK2 inhibitor), a combination of both, or DMSO was tested in standard suppression assays. No additional small molecule inhibitors or DMSO was added. The bar graph shows triplicate means of % proliferation±SD based on 3H-thymidine incorporation on day 6 (unpaired t-test). Data are from one representative experiment of 2 performed in triplicate.
  • FIGS. 5A-5J show targeting Aurora kinase A and JAK2 increases CD39 expression and ATP scavenging among iTreg. In FIG. 5A, contour plots show the CD4+ iTreg and non-Treg gating strategy after 5-day alloMLR treated with bisanilinopyrimidine (I) (750 nM) or DMSO. In FIGS. 5B-5D, CD39 expression (% and geometric MFI) was increased by among iTreg generated in the presence of bisanilinopyrimidine (I) (750 nM). The bar graphs show mean data±SD from 3 independent experiments (unpaired t-test). In FIG. 5E, bar graphs show replicate means of ATP consumption±SD after stimulating 75,000 iTreg with 50 μM of ATP for 45 minutes. ATP measured by luminescence assay. Data are from one representative experiment of 2 performed in triplicate (unpaired t-test). In FIG. 5F, AlloMLRs of naïve, Treg-depleted CD4+ T-cells, and allogeneic DCs (DC:T-cell ratio 1:30) were treated with either bisanilinopyrimidine (I), ARL67156 (CD39 inhibitor), both, or DMSO. Proliferation was determined by colorimetric assay on day 5, with % proliferation based on DMSO control. Graph shows average triplicate means±SEM from 5 independent experiments (unpaired t-test). In FIGS. 5G-5H, bar graphs depict mean fold MFI of LAG3 and CTLA4±SD on iTregs harvested from alloMLRs treated with bisanilinopyrimidine (I) or DMSO from 3 independent experiments (unpaired t-test). In FIGS. 5I-5J, bar graphs show triplicate mean concentrations of IL-10 and TGF-beta±SEM among PMA/ionomycin-stimulated iTregs previously exposed to bisanilinopyrimidine (I) or DMSO during co-culture from 4 independent experiments (unpaired t-test). *P<0.05, **P=0.001-0.01, ****P<0.0001.
  • FIGS. 6A-6B show Aurora kinase A inhibition does not impair TH17 differentiation. Isolated naïve CD4+ T-cells were stimulated with allogeneic DCs for 5 days with alisertib, AJI-214, TG101348, or DMSO. Media was supplemented with IL-6, TGF-beta, and anti-IFN-γmAb to promote RORgammaT expression. The bar graphs depict replicate means of relative RORgammaT expression±SEM among drug- and control-treated CD4+ T-cells (unpaired t-test). Data are from one representative experiment of 2 performed in triplicate for each figure. *P<0.05, **P=0.001-0.01.
  • FIGS. 7A-7H show blockade of Aurora kinase A and JAK2 reduces xenogeneic GVHD and preserves anti-tumor CTL function. In FIG. 7A, AlloMLR (DC:T-cell ratio 1:30) was treated with bisanilinopyrimidine (II) (dual Aurora kinase A/JAK2 inhibitor) or DMSO once on day 0. Proliferation was determined by colorimetric assay on day 5 (ANOVA). IC50=200 nM. Graph shows triplicate means±SD from 1 representative experiment of 2. In FIG. 7B, a representative histogram shows bisanilinopyrimidine (II) inhibits CD3+ T-cell STAT3 phosphorylation in vitro. Data are from one representative experiment of 2. NSG mice received human PBMCs (30×106 cells), with bisanilinopyrimidine (II) (50 mg/kg daily) or vehicle administered from day 0 to day +14. In FIG. 7C, a representative histogram shows H3 ser10 phosphorylation in human CD3+ T-cells harvested at day +14 among each group. In FIGS. 7D-7F, graphs show mean % weight change±SEM, GVHD scores±SEM (unpaired t-test), and survival of pooled data from 2 independent experiments (long-rank test). n=8 mice per each group. In FIG. 7G, a graph depicts mean specific lysis±SD by human CTL generated in vivo using NSG mice transplanted with human PBMCs and irradiated U937 cells (107) on days 0 and +7. Results shown are from 1 of 2 independent experiments, using a total of 7 mice per arm. In FIG. 7H, a graph shows mean specific lysis±SD of CD8+ CTL generated in vitro while exposed to bisanilinopyrimidine (I) or DMSO for 10 days. Results shown are from 1 of 2 independent experiments. U937 lysis was measured by released fluorescence after 4 hours. **P=0.001-0.01, ***P=0.0001-0.001.
  • FIGS. 8A-8K show targeting Aurora kinase A and JAK2 increases the proportion of Treg to allo Tconv in vivo, and reduces T-cell homing to recipient livers. Xenotransplanted NSG mice were treated with bisanilinopyrimidine (II) (50 mg/kg daily) or vehicle daily starting at day 0, then euthanized on day +14. Recipient spleens and livers were harvested, and tissue-resident T-cells were evaluated. Dot plots show absolute number of total spleen cells (FIG. 8A), human CD3+ T-cells (FIG. 8B), % CD4+, CD25+, CD127, Foxp3+ Treg (FIG. 8C), % CD4+, CD25+, CD127+ allo Tconv (FIG. 8D), and the ratio of Treg to allo Tconv (FIG. 8E) (unpaired t-test). In FIG. 8F, representative contour plots show the % CD4+ Treg and % CD4+ alloreactive Tconv residing in spleens of bisanilinopyrimidine (II)- or vehicle-treated mice at day +14. FIG. 8G, the representative histograms show corresponding expression of Foxp3 within the CD4+Tregs. In FIGS. 8H-8I, sections of recipient livers show bisanilinopyrimidine (II) significantly reduces the amount of tissue-resident human T-cells, compared to vehicle control (red=CD3). Dot plots show the number of human T-cells per high power field (FIG. 8J) and % CD4+ cell expressing Foxp3 (FIG. 8K) in the livers at day +14 (unpaired t-test). Pooled data from 2 independent experiments. n=7-8 mice per each group. NS=not significant, *P<0.05, ***P=0.0001-0.001.
  • FIGS. 9A-9H show synergistic immune suppression with combined inhibition of Aurora kinase A and JAK2. In FIG. 9A, Human T cells were stimulated with DCs (DC/T cell ratio of 1:30) exposed to TG101348 (JAK2 inhibitor), alisertib (Aurora kinase A inhibitor), or both TG101348 and alisertib at a fixed ratio of 1:5, respectively, at varying concentrations once on day 0. Proliferation was determined by fluorescence assay on day 5, with % proliferation based on dimethyl sulfoxide (DMSO) control. Graph depicts combination synergy (IC50 values for TG101348 and alisertib were 350 nM and 1.75 mM, respectively), showing one representative independent experiment of two performed with triplicate technical replicates. The CI was calculated using the Chou-Talalay method. AlloMLR (DC/T cell ratio of 1:30) treated with AJI-214 (FIG. 9B), AJI-100 (FIG. 9C) (dual JAK2/Aurora kinase A inhibitors), or DMSO once on day 0 is shown. IC50 values for AJI-214 and AJI-100 were 100 and 200 nM, respectively. Graph shows average triplicate means±SEM from two to three independent experiments [analysis of variance (ANOVA)]. In FIG. 9D, a bar graph depicts T cell proliferation when exposed to alisertib (1.75 mM), TG101348 (350 nM), a combination of alisertib and TG101348 (combo), AJI-214 (750 nM), or AJI-100 (750 nM) in alloMLRs. Means±SEM from four independent experiments (ANOVA) are shown using triplicate technical replicates. In FIGS. 9E-9H, bar graphs depict the mean gated CD3+ T cell H3Ser10 (target of Aurora) or STAT3 (target of JAK2) phosphorylation±SD from three independent experiments after stimulation with allogeneic DCs (5 days) or IL-6 (15 min), respectively (ANOVA). Representative contour plots show H3Ser10 and STAT3 phosphorylation, respectively.
  • FIGS. 10A-10H show immunosuppressive effect of Aurora kinase A/JAK2 blockade on responder Tconv and TH subsets. In FIG. 10A-10B, T cells were stimulated with allogeneic DCs (DC/T cell ratio of 1:30) and treated with kinase inhibitors or DMSO once on day 0. Bar graphs show replicate mean absolute numbers of activated CD4+, CD25+, CD127+ or activated CD8+, CD25+ Tconv±SEM at day +5 from six independent experiments (ANOVA and paired t test). In FIG. 10C, a bar graph shows the mean number of IL-17 spots per well±SD from triplicate technical replicates among DC-allostimulated CD4+ T cells. One of three representative experiments is shown. In FIG. 10D, a bar graph shows mean % CD4+, IFN-γ+ T cells±SEM at day +5 of alloMLR from five independent experiments with technical replicates performed in triplicate. In FIGS. 10E-10F, representative contour plots show Treg (CD25+ and CD127) depletion of CD4+ T cell responders at outset of alloMLR (DC/T cell ratio of 1:30), followed by induction of iTreg (CD127, CD25+, and Foxp3+) after 5 days of culture exposed to kinase inhibitors or DMSO. In FIG. 10G, a bar graph shows mean absolute numbers of iTregs±SEM from seven independent experiments performed with two to three technical replicates (ANOVA and paired t test). In FIG. 10H, a representative histograms depict pSTAT5 expression among IL-2-stimulated CD3+ T cells while exposed to kinase inhibitors or DMSO. Geometric mean fluorescence intensity (MFI) of pSTAT5 is shown along the right margin. One of three representative experiments is shown. *P<0.05, **P=0.001 to 0.01. Alisertib (1.75 mM), TG101348 (350 nM), AJI-214 (750 nM), and AJI-100 (750 nM).
  • FIGS. 11A-11C show combined inhibition of Aurora kinase A and JAK2 enhances antigen-specific iTreg-suppressive potency. In FIG. 11A, a Bar graph depicts mean % demethylation of Foxp3±SEM among iTregs in alloMLRs treated with AJI-214 (750 nM) or DMSO from four independent experiments using triplicate technical replicates. In FIG. 11B, the suppressive capacity of sorted, DC-allostimulated iTregs previously exposed to AJI-214 or DMSO was tested at different ratios of iTreg to T cell responders stimulated by fresh allogeneic DCs (DC/responder T cell ratio of 1:30) in alloMLRs. No additional AJI-214 or DMSO was added. Bar graph shows means of % proliferation±SEM based on [3H]thymidine incorporation on day 6 from three independent experiments with triplicate technical replicates (ANOVA). In FIG. 11C, the potency of iTregs generated in the presence of alisertib (1.75 mM), TG101348 (350 nM), a combination of alisertib and TG101348, or DMSO was tested in standard suppression assays. No additional small-molecule inhibitors or DMSO was added. Bar graph shows means of % proliferation±SD based on [3H]thymidine incorporation on day 6 (paired t test). Data are from one representative experiment of two performed using triplicate technical replicates. *P<0.05.
  • FIGS. 12A-12J show targeting Aurora kinase A and JAK2 increases CD39 expression and ATP scavenging among iTreg. In FIG. 12A, contour plots show the CD4+ iTreg and non-Treg gating strategy after 5-day alloMLR treated with AJI-214 (750 nM) or DMSO. In FIGS. 12B-12D, CD39 density [geometric MFI (gMFI)] is increased by among iTreg generated in the presence of AJI-214 (750 nM). Bar graphs show mean data±SD from three independent experiments (paired t test). In FIG. 12E, bar graphs show replicate means of ATP consumption±SD after stimulating 75,000 iTregs with 50 mM ATP for 45 min. ATP was measured by luminescence assay. Data are from one representative experiment of two performed using triplicate technical replicates (paired t test). In FIG. 12F, AlloMLRs of naïve, Treg-depleted CD4+ T cells and allogeneic DCs (DC/T cell ratio of 1:30) were treated with AJI-214, ARL67156 (CD39 inhibitor), both AJI-214 and ARL67156, or DMSO. Proliferation was determined by colorimetric assay on day 5, with % proliferation based on DMSO control. Graph shows means±SEM from five independent experiments using three technical replicates (paired t test). In FIGS. 12G-12H, bar graphs depict mean fold MFI of LAG3 and CTLA4±SD on iTregs harvested from alloMLRs treated with AJI-214 or DMSO from three independent experiments. In FIG. 12I-12J, bar graphs show mean concentrations of IL-10 and TGF-0±SEM among PMA (phorbol 12-myristate 13-acetate)/ionomycin-stimulated iTregs previously exposed to AJI-214 or DMSO during coculture from four independent experiments using three technical replicates. *P<0.05, **P=0.001 to 0.01, ****P<0.0001.
  • FIGS. 13A-13G show Blockade of Aurora kinase A and JAK2 reduces xenogeneic GVHD and preserves the in vivo generation of potent antitumor CTL. NSG mice received human PBMCs (30×106 cells) by intraperitoneal injection, with alisertib (30 mg/kg daily), TG101348 (45 mg/kg twice a day), a combination of alisertib and TG101348, or vehicle administered by oral gavage from day 0 to day +14. In FIG. 13A, percent survival is shown among the four groups (log-rank test). In FIG. 13B, a graph shows mean GVHD clinical scores±SEM for each group of mice (P=0.02 at day +30, vehicle versus combo, Mann-Whitney). Pooled data are from two independent experiments. n=7 to 8 mice per group. NSG mice were transplanted with human PBMCs as described, with AJI-100 (50 mg/kg daily) or vehicle administered ip from day 0 to day +14. In FIGS. 13C-13D, percent survival (log-rank test) and mean GVHD clinical scores±SEM (Mann-Whitney) are demonstrated. Pooled data are from two independent experiments. n=8 mice per group. In FIG. 13E, representative contour plots show expression of H3Ser10 and STAT3 phosphorylation among human CD3+ T cells harvested from recipient spleens at day +14. In FIG. 13F, a bar graph shows the mean % pH3Ser10+ and % pSTAT3+ T cells±SEM among AJI-100-treated and vehicle-treated mice at day +14 (n=6 mice per group, two independent experiments, Mann-Whitney). In FIG. 13G, a graph depicts mean specific lysis±SD by human CD8+ CTL generated in vivo using NSG mice transplanted with human PBMCs and vaccinated with irradiated U937 cells (1×107) on days 0 and +7. Results shown are from one of two independent experiments, using a total of seven mice per group. U937 lysis was measured by released fluorescence after 4 hours (vehicle versus AJI-100, not significant, Mann-Whitney). *P<0.05, **P=0.001 to 0.01, ***P=0.0001 to 0.001, ****P<0.0001.
  • FIGS. 14A-14M show targeting Aurora kinase A and JAK2 increases the proportion of Treg to activated Tconv and reduces TH17 and T H1 cells in xenotransplanted recipient mice. In FIGS. 14A-14D, xenotransplanted NSG mice were treated with AJI-100 (50 mg/kg) or vehicle daily starting at day 0 and then euthanized on day +14. Recipient spleens, livers, and lungs were harvested, and tissue-resident T cells were evaluated. Bar graphs show replicate mean absolute number of human CD4+ T cells (FIG. 14A), CD4+ Tregs (FIG. 14B), CD4+ activated Tconv (CD25+ and CD127+) (FIG. 14C), and the ratio of Treg to activated Tconv (FIG. 14D)±SEM (Mann-Whitney). In FIG. 14E, representative contour plots show the % CD4+ Treg and % CD4+ activated allo-Tconv residing in spleens of AJI-100-treated or vehicle-treated mice at day +14. The representative histograms show corresponding expression of Foxp3 within the CD4+ Tregs. In FIG. 14F, a bar graph shows the replicate mean number of IL-17 spots per well±SEM among human lymphocytes harvested from recipient spleens at day +14 (Mann-Whitney). In FIG. 14G-14H, Bar graph and representative contour plots depict the amount of CD4+, IFN-γ+ T cells±SEM from AJI-100 or vehicle-treated mice at day +14 (Mann-Whitney). In FIG. 14I, sections of recipient livers (top) and lung (bottom) show that AJI-100 significantly reduces GVHD damage in recipient target organs, compared to vehicle control. In FIG. 14J-14K bar graphs depict the mean GVHD pathology scores±SEM for host liver and lung at day +14. In FIG. 14L-14M, bar graphs shows that the mean number of human CD3+ T cells±SEM [per high-power field (HPF)] infiltrating liver or lung at day +14 is significantly reduced by AJI-100 compared to vehicle (Mann-Whitney). Pooled data are from at least two independent experiments. n=6 to 14 mice per group. *P<0.05, **P=0.001 to 0.01, ****P<0.0001.
  • FIGS. 15A-15C are micrographs of representative H&E stained human, xenogenic shin grafts at day +21. Mice were treated with AJI-100. Mice receiving skin alone and no PBMC's (allogenic peripheral blood mononuclear cells) were considered as non-rejection control (n=1 experiment, 12 mice).
  • DETAILED DESCRIPTION
  • The materials, compounds, compositions, and methods described herein may be understood more readily by reference to the following detailed description of specific aspects of the disclosed subject matter, the Figures, and the Examples included therein.
  • Before the present materials, compounds, compositions, and methods are disclosed and described, it is to be understood that the aspects described below are not limited to specific synthetic methods or specific reagents, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular aspects only and is not intended to be limiting.
  • Also, throughout this specification, various publications are referenced. The disclosures of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art to which the disclosed matter pertains. The references disclosed are also individually and specifically incorporated by reference herein for the material contained in them that is discussed in the sentence in which the reference is relied upon.
  • General Definitions
  • In this specification and in the claims that follow, reference will be made to a number of terms, which shall be defined to have the following meanings:
  • Throughout the specification and claims the word “comprise” and other forms of the word, such as “comprising” and “comprises,” means including but not limited to, and is not intended to exclude, for example, other additives, components, integers, or steps.
  • As used in the description and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a composition” includes mixtures of two or more such compositions, reference to “an inhibitor” includes mixtures of two or more such inhibitors, reference to “the kinase” includes mixtures of two or more such kinase, and the like.
  • “Optional” or “optionally” means that the subsequently described event or circumstance can or cannot occur, and that the description includes instances where the event or circumstance occurs and instances where it does not.
  • Notwithstanding that the numerical ranges and parameters setting forth the broad scope of the disclosure are approximations, the numerical values set forth in the specific examples are reported as precisely as possible. Any numerical value, however, inherently contain certain errors necessarily resulting from the standard deviation found in their respective testing measurements. Furthermore, when numerical ranges of varying scope are set forth herein, it is contemplated that any combination of these values inclusive of the recited values may be used. Further, ranges can be expressed herein as from “about” one particular value, and/or to “about” another particular value. When such a range is expressed, another aspect includes from the one particular value and/or to the other particular value. Similarly, when values are expressed as approximations, by use of the antecedent “about,” it will be understood that the particular value forms another aspect. It will be further understood that the endpoints of each of the ranges are significant both in relation to the other endpoint, and independently of the other endpoint. Unless stated otherwise, the term “about” means within 5% (e.g., within 2% or 1%) of the particular value modified by the term “about.”
  • By “reduce” or other forms of the word, such as “reducing” or “reduction,” is meant lowering of an event or characteristic (e.g., the risk of having GVHD). It is understood that this is typically in relation to some standard or expected value, in other words it is relative, but that it is not always necessary for the standard or relative value to be referred to.
  • By “prevent” or other forms of the word, such as “preventing” or “prevention,” is meant to stop a particular event or characteristic, to stabilize or delay the development or progression of a particular event or characteristic, or to minimize the chances that a particular event or characteristic will occur. Prevent does not require comparison to a control as it is typically more absolute than, for example, reduce. As used herein, something could be reduced but not prevented, but something that is reduced could also be prevented. Likewise, something could be prevented but not reduced, but something that is prevented could also be reduced. It is understood that where reduce or prevent are used, unless specifically indicated otherwise, the use of the other word is also expressly disclosed.
  • As used herein, “treatment” refers to obtaining beneficial or desired clinical results. Beneficial or desired clinical results include, but are not limited to, any one or more of: alleviation of one or more symptoms (such as GVHD), diminishment of extent of GVHD, stabilized (i.e., not worsening) state of GVHD, preventing or delaying occurrence or recurrence of GVHD, delay or slowing of GVHD progression, and amelioration of the GVHD state.
  • The term “patient” preferably refers to a human in need of treatment for any purpose, and more preferably a human in need of such a treatment to treat GVHD. However, the term “patient” can also refer to non-human animals, preferably mammals such as dogs, cats, horses, cows, pigs, sheep and non-human primates, among others, that are in need of treatment.
  • Chemical Definitions
  • As used herein, the term “composition” is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • References in the specification and concluding claims to parts by weight of a particular element or component in a composition denotes the weight relationship between the element or component and any other elements or components in the composition or article for which a part by weight is expressed. Thus, in a mixture containing 2 parts by weight of component X and 5 parts by weight component Y, X and Y are present at a weight ratio of 2:5, and are present in such ratio regardless of whether additional components are contained in the mixture.
  • A weight percent (wt. %) of a component, unless specifically stated to the contrary, is based on the total weight of the formulation or composition in which the component is included.
  • As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, and aromatic and nonaromatic substituents of organic compounds. Illustrative substituents include, for example, those described below. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this disclosure, the heteroatoms, such as nitrogen, can have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valencies of the heteroatoms. This disclosure is not intended to be limited in any manner by the permissible substituents of organic compounds. Also, the terms “substitution” or “substituted with” include the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., a compound that does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc.
  • The term “aliphatic” as used herein refers to a non-aromatic hydrocarbon group and includes branched and unbranched, alkyl, alkenyl, or alkynyl groups.
  • The term “alkyl” as used herein is a branched or unbranched saturated hydrocarbon group of 1 to 24 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, t-butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, dodecyl, tetradecyl, hexadecyl, eicosyl, tetracosyl, and the like. The alkyl group can also be substituted or unsubstituted. The alkyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • The symbols An is used herein as merely a generic substituent in the definitions below.
  • The term “alkoxy” as used herein is an alkyl group bound through a single, terminal ether linkage; that is, an “alkoxy” group can be defined as —OA1 where A1 is alkyl as defined above.
  • The term “alkenyl” as used herein is a hydrocarbon group of from 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon double bond. Asymmetric structures such as (A1A2)C═C(A3A4) are intended to include both the E and Z isomers. This may be presumed in structural formulae herein wherein an asymmetric alkene is present, or it may be explicitly indicated by the bond symbol C═C. The alkenyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • The term “alkynyl” as used herein is a hydrocarbon group of 2 to 24 carbon atoms with a structural formula containing at least one carbon-carbon triple bond. The alkynyl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol, as described below.
  • The term “aryl” as used herein is a group that contains any carbon-based aromatic group including, but not limited to, benzene, naphthalene, phenyl, biphenyl, phenoxybenzene, and the like. The term “heteroaryl” is defined as a group that contains an aromatic group that has at least one heteroatom incorporated within the ring of the aromatic group. Examples of heteroatoms include, but are not limited to, nitrogen, oxygen, sulfur, and phosphorus. The term “non-heteroaryl,” which is included in the term “aryl,” defines a group that contains an aromatic group that does not contain a heteroatom. The aryl and heteroaryl group can be substituted or unsubstituted. The aryl and heteroaryl group can be substituted with one or more groups including, but not limited to, alkyl, halogenated alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein. The term “biaryl” is a specific type of aryl group and is included in the definition of aryl. Biaryl refers to two aryl groups that are bound together via a fused ring structure, as in naphthalene, or are attached via one or more carbon-carbon bonds, as in biphenyl.
  • The term “cycloalkyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc. The term “heterocycloalkyl” is a cycloalkyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkyl group and heterocycloalkyl group can be substituted or unsubstituted. The cycloalkyl group and heterocycloalkyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • The term “cycloalkenyl” as used herein is a non-aromatic carbon-based ring composed of at least three carbon atoms and containing at least one double bound, i.e., C═C. Examples of cycloalkenyl groups include, but are not limited to, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like. The term “heterocycloalkenyl” is a type of cycloalkenyl group as defined above where at least one of the carbon atoms of the ring is substituted with a heteroatom such as, but not limited to, nitrogen, oxygen, sulfur, or phosphorus. The cycloalkenyl group and heterocycloalkenyl group can be substituted or unsubstituted. The cycloalkenyl group and heterocycloalkenyl group can be substituted with one or more groups including, but not limited to, alkyl, alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol as described herein.
  • The term “cyclic group” is used herein to refer to either aryl groups, non-aryl groups (i.e., cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl groups), or both. Cyclic groups have one or more ring systems that can be substituted or unsubstituted. A cyclic group can contain one or more aryl groups, one or more non-aryl groups, or one or more aryl groups and one or more non-aryl groups.
  • The term “aldehyde” as used herein is represented by the formula —C(O)H. Throughout this specification “C(O)” is a short hand notation for C═O.
  • The terms “amine” or “amino” as used herein are represented by the formula NA1A2A3, where A1, A2, and A3 can be, independently, hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • The term “carboxylic acid” as used herein is represented by the formula —C(O)OH. A “carboxylate” as used herein is represented by the formula —C(O)O—.
  • The term “ester” as used herein is represented by the formula —OC(O)A1 or —C(O)OA1, where A1 can be an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • The term “ether” as used herein is represented by the formula A1OA2, where A1 and A2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • The term “ketone” as used herein is represented by the formula A1C(O)A2, where A1 and A2 can be, independently, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • The term “halide” as used herein refers to the halogens fluorine, chlorine, bromine, and iodine.
  • The term “hydroxyl” as used herein is represented by the formula —OH.
  • The term “nitro” as used herein is represented by the formula —NO2.
  • The term “cyano” as used herein is represented by the formula —CN The term “azido” as used herein is represented by the formula —N3.
  • The term “oxo” as used herein is represented by ═O.
  • The term “sulfonyl” is used herein to refer to the sulfo-oxo group represented by the formula —S(O)2A1, where A1 can be hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above. The term “sulfoxide” is used herein to refer to the sulfo-oxo group represented by the formula —OS(O)2A1, where A1 can be hydrogen, an alkyl, halogenated alkyl, alkenyl, alkynyl, aryl, heteroaryl, cycloalkyl, cycloalkenyl, heterocycloalkyl, or heterocycloalkenyl group described above.
  • The term “sulfonylamino” or “sulfonamide” as used herein is represented by the formula —S(O)2NH2.
  • The term “thiol” as used herein is represented by the formula —SH.
  • It is to be understood that the compounds provided herein may contain chiral centers.
  • Such chiral centers may be of either the (R-) or (S-) configuration. The compounds provided herein may either be enantiomerically pure, or be diastereomeric or enantiomeric mixtures. It is to be understood that the chiral centers of the compounds provided herein may undergo epimerization in vivo. As such, one of skill in the art will recognize that administration of a compound in its (R-) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its (S-) form.
  • As used herein, substantially pure means sufficiently homogeneous to appear free of readily detectable impurities as determined by standard methods of analysis, such as thin layer chromatography (TLC), nuclear magnetic resonance (NMR), gel electrophoresis, high performance liquid chromatography (HPLC) and mass spectrometry (MS), gas-chromatography mass spectrometry (GC-MS), and similar, used by those of skill in the art to assess such purity, or sufficiently pure such that further purification would not detectably alter the physical and chemical properties, such as enzymatic and biological activities, of the substance. Both traditional and modern methods for purification of the compounds to produce substantially chemically pure compounds are known to those of skill in the art. A substantially chemically pure compound may, however, be a mixture of stereoisomers.
  • Unless stated to the contrary, a formula with chemical bonds shown only as solid lines and not as wedges or dashed lines contemplates each possible isomer, e.g., each enantiomer, diastereomer, and meso compound, and a mixture of isomers, such as a racemic or scalemic mixture.
  • A “pharmaceutically acceptable” component is one that is suitable for use with humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response) commensurate with a reasonable benefit/risk ratio.
  • “Pharmaceutically acceptable salt” refers to a salt that is pharmaceutically acceptable and has the desired pharmacological properties. Such salts include those that may be formed where acidic protons present in the compounds are capable of reacting with inorganic or organic bases. Suitable inorganic salts include those formed with the alkali metals, e.g., sodium, potassium, magnesium, calcium, and aluminum. Suitable organic salts include those formed with organic bases such as the amine bases, e.g., ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the like. Such salts also include acid addition salts formed with inorganic acids (e.g., hydrochloric and hydrobromic acids) and organic acids (e.g., acetic acid, citric acid, maleic acid, and the alkane- and arene-sulfonic acids such as methanesulfonic acid and benzenesulfonic acid). When two acidic groups are present, a pharmaceutically acceptable salt may be a mono-acid-mono-salt or a di-salt; similarly, where there are more than two acidic groups present, some or all of such groups can be converted into salts.
  • “Pharmaceutically acceptable excipient” refers to an excipient that is conventionally useful in preparing a pharmaceutical composition that is generally safe, non-toxic, and desirable, and includes excipients that are acceptable for veterinary use as well as for human pharmaceutical use. Such excipients can be solid, liquid, semisolid, or, in the case of an aerosol composition, gaseous.
  • A “pharmaceutically acceptable carrier” is a carrier, such as a solvent, suspending agent or vehicle, for delivering the disclosed compounds to the patient. The carrier can be liquid or solid and is selected with the planned manner of administration in mind. Liposomes are also a pharmaceutical carrier. As used herein, “carrier” includes any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated.
  • The term “therapeutically effective amount” as used herein means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence. An effective amount can be administered in one or more doses.
  • Effective amounts of a compound or composition described herein for treating a mammalian subject can include about 0.1 to about 1000 mg/Kg of body weight of the subject/day, such as from about 1 to about 100 mg/Kg/day, especially from about 10 to about 100 mg/Kg/day. The doses can be acute or chronic. A broad range of disclosed composition dosages are believed to be both safe and effective.
  • Reference will now be made in detail to specific aspects of the disclosed materials, compounds, compositions, articles, and methods, examples of which are illustrated in the accompanying Examples and Figures.
  • Compounds
  • Disclosed are compounds that are Aurora kinase inhibitors, e.g., Aurora A, B, and/or C kinase inhibitors and Janus kinase 2 (JAK2) inhibitors. The compounds can concurrently block Aurora kinase A and JAK2 signal. These disclosed compounds can be used in various compositions to reduce the risk of developing, prevent, or treat GVHD in a subject. These disclosed compounds can be used in various compositions to synergistically suppress alloreactive human T-cells in vitro, prevents xenogeneic graft-versus-host disease (GVHD) without impairing anti-tumor responses, and promotes the development and suppressive potency of CD39+ inducible Treg.
  • In certain embodiments, the disclosed compounds have the chemical structure shown in Formula I.
  • Figure US20220281828A1-20220908-C00006
  • wherein
    • R1 is selected from the group consisting of H, Cl, F, Br, I, CN, NO2, NH2, CF3, CO2H, CO2NH2, CO2NHR5, CO2R5, C(O)R5, C(O)NH2, C(O)NHR5, and C1-C6 alkyl optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R2 is selected from the group consisting of H, OH, CN, NO2, NH2, optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, wherein optionally substituted substituents are optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • or R1 and R2 together form a fused cycloalkyl, cycloheteroalkyl, aryl, or heteraryl group;
    • each R3 is selected, independently, from the group consisting of SO2NH2, SO2NHR5, NHSO2R5, NHCO2R5, NHC(O)R5, NHCONHR5, F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted cycloheteroaryl, and optionally substituted fused cycloheteroalkyl, wherein optionally substituted substituents are optionally substituted with sulfonyl;
    • each R4 is selected, independently, from the group consisting of F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, COOH, C(O)NH2, C(O)R5, C(O)NHR5, CH2C(O)R5, SO2NH2, SO2NHR5, CONHSO2R5, optionally substituted phenyl, optionally substituted OPhenyl, tetrazole, piperadinyl, piperazinyl, and morpholinyl, wherein optionally substituted substituents are optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • each R5 is selected, independently, from the group consisting of optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heteroalkyl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, cycloalkyl, cycloheteroalkyl, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R7 is selected from the group consisting of H, C1-C6 alkyl, C1-C6 alkoxyl, halide, hydroxyl, cyano, nitro, and amino;
    • n is 0-5; and
    • m is 1-5,
      or a pharmaceutically acceptable salt thereof.
  • Thus, in the disclosed compounds there can be from none to 5 different substituents R3 and from 1 to 5 different substituents R4. Pharmaceutically acceptable salts of these compounds are also disclosed. In some examples, R1 is F, R2 is hydrogen, and R3 is 2-Cl. In one specific example, R1 is Cl or F, R2 is hydrogen, R3 is hydrogen or 2-Cl, and R4 is 4-CONH2.
  • In certain embodiments, the disclosed compounds have the chemical structure shown in Formula II.
  • Figure US20220281828A1-20220908-C00007
  • wherein R1-R4 and m and n are as defined above for Formula I.
  • In some examples of Formula II, R1 is selected from the group consisting of H, Cl, F, Br, I, C1-C6 alkyl, CN, NO2, and NH2. Also in Formula II, R2 is selected from the group consisting of H, F, and Cl.
  • Additionally in Formula II, each R3 is selected, independently, from the group consisting of Cl, Br, F, COOH, CF3, CN, phenyl, OCH3, COR5, CONH2, CONR5, and COONH2. When n is 0, there is no R3.
  • Further in Formula II, each R4 is selected, independently, from the group consisting of H, COOH, CONH2, CONR5, SO2NH2, CONSO2R5, tetrazole, 4-morpholine, and COR5. Each R5 is selected, independently, from the group consisting of optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heteroalkyl. In some examples R5 is an unsubstituted substituent.
  • Still further in Formula II, n is 0-5 (e.g., 0, 1, 2, 3, 4, or 5) and m is 1-5 (e.g., 1, 2, 3, 4, or 5). Thus, in the disclosed compounds there can be from none to 5 different substituents R3 and from 1 to 5 different substituents R4. In certain examples of Formula II, R1 is F, R2 is H, m is 3 and R4 is 3,5-di-F, 4-OH. Pharmaceutically acceptable salts of these compounds are also disclosed.
  • In certain preferred aspects, the compound has Formula II, wherein R3 is 2-Cl, and n is 1. In other examples, the compound has Formula II, n is 0 and there is no R3. In other examples, the compound has Formula II, n is 2 and one R3 is an ortho-Cl and the other R3 is a para hydroxyl, methoxyl, or cyano group. In other examples, the compound has Formula II, wherein R4 is 4-CONH2, and m is 1. In other examples, the compound has Formula II, wherein R4 is 4-CONHR5, and m is 1 In still other examples, the compound has Formula II, wherein R3 is 2-Cl, n is 1, m is 1, and R4 is COOH, COR5, CONH2, CONR5, or CONSO2R5, wherein R5 is C1-C6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl. In other examples, the compound has Formula II, wherein R3 is absent, n is 0, m is 1, and R4 is COOH, COR5, CONH2, CONR5, or CONSO2R5, wherein R5 is C1-C6 alkyl, cycloalkyl, heteroaryl, or heteroalkyl.
  • Still further, the disclosed compounds can have the following Formula IIIA or IIIB:
  • Figure US20220281828A1-20220908-C00008
  • wherein
    • R1 is selected from the group consisting of H, Cl, F, Br, I, CH3 and NH2;
    • R2 is selected from the group consisting of H, F, and Cl;
    • R3 is selected from the group consisting of 2-Cl, 2-Br, 2-F, 2-COOH, 2-CF3, 2-CN, 2-phenyl, 2-OCH3, 2-COONH2, 4-COOH, and 4-OCH3; and
    • R4 is selected from the group consisting of H, COOH, 2-CONH2, 4-CONH2, SO2NH2, tetrazole, and 4-morpholine.
  • In Formula IIIA or IIIB, R1 is selected from the group consisting of H, Cl, F, Br, I, CH3 and NH2; R2 is selected from the group consisting of H, F, and Cl.
  • Also in Formula IIIA, R3 is selected from the group consisting of 2-Cl, 2-Br, 2-F, 2-COOH, 2-CF3, 2-CN, 2-phenyl, 2-OCH3, 2-COONH2, 4-COOH, and 4-OCH3.
  • Additionally in Formula IIIA or IIIB, R4 is selected from the group consisting of H, COOH, 2-CONH2, 4-CONH2, SO2NH2, tetrazole, and 4-morpholine.
  • Still further, the disclosed compounds can have the following Formula IV:
  • Figure US20220281828A1-20220908-C00009
  • In Formula IV, R1, R2, R4, and m are as defined herein.
  • In other examples, disclosed herein are compounds of Formula IA.
  • Figure US20220281828A1-20220908-C00010
  • wherein
    • X is CH or N;
    • R1 is selected from the group consisting of H, Cl, F, Br, I, CN, NO2, NH2, CF3, CO2H, CO2NH2, CO2NHR5, CO2R5, C(O)R5, C(O)NH2, C(O)NHR5, and C1-C6 alkyl optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R2 is H, OH, CN, NO2, NH2, optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl; wherein optionally substituted substituents are optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • or R1 and R2 together form a fused cycloalkyl, cycloheteroalkyl, aryl or heteraryl group;
    • each R3 is selected, independently, from the group consisting of SO2NH2, SO2NHR5, NHSO2R5, NHCO2R5, NHC(O)R5, NHCONHR5, F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted cycloheteroaryl, and optionally substituted fused cycloheteroalkyl, wherein optionally substituted substituents are optionally substituted with sulfonyl;
    • each R5 is selected, independently, from optionally substituted C1-C6 alkyl, optionally substituted C1-C6 cycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, and optionally substituted heteroalkyl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R6 is selected from the group consisting of H, C1-C6 alkyl, C1-C6 alkyl-OH, CO2R5, CO2H, and CO2NHR5;
    • R7 is selected from the group consisting of H, C1-C6 alkyl, C1-C6 alkoxyl, halide, hydroxyl, cyano, nitro, and amino;
    • R8 is OH or =0;
    • n is 0-5; and
    • p is 1 or 2
      or a pharmaceutically acceptable salt thereof.
  • In other examples, disclosed herein are compounds of Formula IB.
  • Figure US20220281828A1-20220908-C00011
  • wherein
    • X is N or CH;
    • R1 is selected from the group consisting of H, Cl, F, Br, I, CN, NO2, NH2, CF3, CO2H, CO2NH2, CO2NHR5, CO2R5, C(O)R5, C(O)NH2, C(O)NHR5, and C1-C6 alkyl optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R2 is selected from the group consisting of H, OH, CN, NO2, NH2, optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, cycloalkyl, aryl, or heteroaryl substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • or R1 and R2 together form a fused cycloalkyl, cycloheteroalkyl, aryl or heteraryl group;
    • each R3 is selected, independently, from the group consisting of SO2NH2, SO2NHR5, NHSO2R5, NHCO2R5, NHC(O)R5, NHCONHR5, F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted cycloheteroaryl, and optionally substituted fused cycloheteroalkyl, wherein optionally substituted substituents are optionally substituted with sulfonyl;
    • each R5 is selected, independently, from the group consisting of optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heteroalkyl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, cycloalkyl, cycloheteroalkyl, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R6 is selected from the group consisting of H, C1-C6 alkyl, CO2R5, CO2H, and CO2NHR5;
    • R7 is selected from the group consisting of H, C1-C6 alkyl, C1-C6 alkoxyl, halide, hydroxyl, cyano, nitro, and amino;
    • R8 is OH or =0;
    • n is 0-5; and
    • p is 1 or 2
      or a pharmaceutically acceptable salt thereof.
  • In other examples, disclosed herein are compounds of Formula V.
  • Figure US20220281828A1-20220908-C00012
  • wherein
    • X is N or CH;
    • L is selected from the group consisting of O, S, C1-4alkyl, C(O)NH, NHC(O), CH2C(O), C(O)CH2, CH2CH2C(O), CH2C(O)CH2, CH2C(O)NH, and NH(CO)CH2; R1 is selected from the group consisting of H, Cl, F, Br, I, CN, NO2, NH2, CF3, CO2H, CO2NH2, CO2NHR5, CO2R5, C(O)R5, C(O)NH2, C(O)NHR5, and C1-C6 alkyl optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R2 is selected from the group consisting of H, OH, CN, NO2, NH2, optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted aryl, and optionally substituted heteroaryl, wherein optionally substituted substituents are optionally substituted with alkoxy, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • or R1 and R2 together form a fused cycloalkyl, cycloheteroalkyl, aryl, or heteraryl group;
    • each R3 is selected, independently, from the group consisting of SO2NH2, SO2NHR5, NHSO2R5, NHCO2R5, NHC(O)R5, NHCONHR5, F, Cl, Br, I, NO2, optionally substituted C1-C6 alkyl, optionally substituted C1-C6 alkoxy, optionally substituted cycloheteroaryl, and optionally substituted fused cycloheteroalkyl, wherein optionally substituted substituents are optionally substituted with sulfonyl;
    • each R5 is selected, independently, from the group consisting of optionally substituted C1-C6 alkyl, optionally substituted cycloalkyl, optionally substituted heteroaryl, and optionally substituted heteroalkyl, wherein optionally substituted substituents are optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, cycloalkyl, cycloheteroalkyl, alkenyl, alkynyl, aryl, heteroaryl, aldehyde, amino, carboxylic acid, ester, ether, halide, hydroxy, ketone, nitro, oxo, silyl, sulfo-oxo, sulfonyl, sulfone, sulfoxide, or thiol;
    • R6 is selected from the group consisting of H, C1-C6 alkyl, CO2R5, CO2H, and CO2NHR5;
    • R7 is selected from the group consisting of H, C1-C6 alkyl, C1-C6 alkoxyl, halide, hydroxyl, cyano, nitro, and amino;
    • R8 is OH or =0;
    • n is 0-5; and
    • p is 1 or 2
      or a pharmaceutically acceptable salt thereof.
  • In certain specific examples of Formula I-V, R1 and R2 together form a fused cycloalkyl, cycloheteroalkyl, aryl or heteraryl group. In other examples, R1 and R2 together form a fused furan. In other examples, R1 and R2 together form a fused cyclopentyl or fused cyclohexyl. In other examples, R1 and R2 together form a fused phenyl.
  • In certain specific examples of Formula I-V, R1 is C1-8 alkyl or heteroalkyl. In other examples, R1 is methyl, ethyl, or trifluoromethyl. In other examples, R1 is chloro, bromo, or fluoro. In other examples, R1 is CO2C1-8alkyl, CO2H, CO2NH2, or CO2NHC1-8 alkyl.
  • In certain specific examples of Formula I-V, R2 is C1_8 alkyl or heteroalkyl. In other examples, R2 is hydrogen.
  • In the disclosed compounds there can be from 1 to 5 different substituents R3, i.e., n can be 1 to 5, though preferable n can be 1 to 3. In some examples, there is no R3 substituent, i.e., n is 0. In specific examples, R3 is SO2NH2, SO2NHR5, or NHSO2R5, wherein R5 is C1-C6 alkyl or C1-C6 cycloalkyl optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, hydroxyl, or halide. In other examples, R3 is NHC(O)R5, wherein R5 is C1-C6 alkyl or C1-C6 cycloalkyl optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, cycloalky, cycloheteroalkyl, hydroxyl, or halide. In other examples, R3 is C1-C6 alkyl or C1-C6 cycloalkyl. In other examples, R3 is C1-C6 alkoxyl. In other examples, R3 is halide. In other examples, n is 2 and each R3 is selected from the group consisting of C1-C6 alkyl, C1-C6 alkoxyl, halide, SO2NH2, SO2NHR5, and NHSO2R5, wherein R5 is C1-C6 alkyl or C1-C6 cycloalkyl optionally substituted with C1-C6 alkyl, C1-C6 alkoxyl, hydroxyl, or halide. In other examples, n is 2 and each R3 is selected from the group consisting of C1-C6 alkyl, C1-C6 alkoxyl, and halide. In other examples, n is 3 and each R3 is selected from the group consisting of C1-C6 alkyl, C1-C6 alkoxyl, and halide. In other examples, n is 2 and each R3 together form a fused hetercycloalkyl.
  • In certain specific examples of the above formula, R1 is F and R2 is H.
  • In certain specific examples, R4 is C(O)NHR5.
  • In certain specific examples, R6 is C1-8 alkyl. In other examples, R6 is methyl. In other examples, R6 is hydrogen.
  • In certain specific examples, R7 is chloro, bromo, or fluoro. In other examples, R7 is hydrogen.
  • In some examples, X is N.
  • In specific examples, L is CH2(O) or C(O)NH.
  • In specific examples, n and m are both 1. In other examples, n is 0.
  • In specific examples R8 is oxo and p is 1. In other examples R8 is oxo and p is 2.
  • Pharmaceutically acceptable salts of these compounds are also disclosed.
  • Specific examples of compounds having Formula I, IA, IB, II, III, IV, and V are disclosed herein are in Tables 1-4.
  • TABLE 1
    Bisanilinipyrimidine analogs.
    Figure US20220281828A1-20220908-C00013
    Cmpd
    Entry ID # R1 R2 R3 R4
     1 1 H H ortho-COOH para-COOH
     2 3a H H ortho-COOH ortho-CONH 2
     3 3b H H ortho-COOH H
     4 3c H H ortho-COOH para-morpholine
     5 3d H H ortho-COOH ortho-COOH
     6 3e H H ortho-COOMe para-COOMe
     7 3f H H H H
     8 3g H H ortho-COONH2 para-CONH2
     9 3h H H H para-COOH
    10 3i H H para-COOH para-COOH
    11 3j CH3 H ortho-COOH para-COOH
    12 3k H CH3 ortho-COOH para-COOH
    13 3l H H ortho-Cl para-COOH
    14 3m F H ortho-COOH All H
    15 3n F H ortho-COOH para-COOH
    16 3o F H ortho-Cl para-COOH
    17 3p F H ortho-Cl H
    18 3q H H ortho-COOH meta-COOH
    19 3r F H ortho-COOH meta-COOH
    20 4c Cl Cl ortho-COOH para-COOH
    21 4d Cl H ortho-COOH para-COOH
    22 4a NH2 H ortho-COOH para-COOH
    23 4b H NH2 ortho-COOH para-COOH
  • TABLE 2
    Bisanilinipyrimidine analogs.
    Figure US20220281828A1-20220908-C00014
    Cmpd
    Entry ID # R1 R2 R3 R4 R5
    24 6a H H ortho-F para-COOH H
    25 6b H H ortho-CF3 All H H
    26 6c H H 2-Cl-4-F para-COOH H
    27 6d H H ortho-OCF3 para-COOH H
    28 6e H H ortho-OMe para-COOH H
    29 6f H H ortho-OMe All H H
    30 6g H H ortho-CN All H H
    31 6h H H ortho-CF3 para-COOH H
    31 6i H H ortho-Br para-COOH H
    33 6j H H ortho-Cl para- H
    CH2—COOH
    34 6k H H Ortho-Cl para-COOH, H
    meta-OH
    35 6l H H Ortho-F All H H
    36 6m H H Ortho-I para-COOH H
    37 6n H H Ortho-CN para-COOH H
    38 6o H H Ortho-Cl meta-COOH H
    39 6p H H ortho-Cl para-CONH2 H
    40 6q H H ortho-phenyl para-COOH H
    41 6r H H ortho-Cl para-COOH CH3
    42 6s H H ortho-Cl para-COOH CH3—CH2
    43 6t F H ortho-Cl meta-COOH H
  • TABLE 3
    Bisanilinipyrimidine analogs.
    Entry Compound ID
    44 9a (RE1-043)  
    Figure US20220281828A1-20220908-C00015
    45 9b (Re1-032)  
    Figure US20220281828A1-20220908-C00016
    46 9c (RE1-031)  
    Figure US20220281828A1-20220908-C00017
    47 9d (RE1-025)  
    Figure US20220281828A1-20220908-C00018
    48 9e (RE1-039)  
    Figure US20220281828A1-20220908-C00019
    49 9f (RE1-019)  
    Figure US20220281828A1-20220908-C00020
    50 9g (HM5-018-2)  
    Figure US20220281828A1-20220908-C00021
    51 9h (YL5-146-4)  
    Figure US20220281828A1-20220908-C00022
    52 6p (YL5-145)  
    Figure US20220281828A1-20220908-C00023
    53 9i (YL5-146-3)  
    Figure US20220281828A1-20220908-C00024
    54 9j (HM6-007-1)  
    Figure US20220281828A1-20220908-C00025
    55 9k (HM6-020-2)  
    Figure US20220281828A1-20220908-C00026
    56 9l (HM4-153-2)  
    Figure US20220281828A1-20220908-C00027
    57 9m (HM6-021-4)  
    Figure US20220281828A1-20220908-C00028
    58 9n (HM6-029-1)  
    Figure US20220281828A1-20220908-C00029
    59 13a (SO2-162)  
    Figure US20220281828A1-20220908-C00030
    60 13b (SO3-033)  
    Figure US20220281828A1-20220908-C00031
    61 13c (SO3-036)  
    Figure US20220281828A1-20220908-C00032
    62 13d (SO3-035)  
    Figure US20220281828A1-20220908-C00033
  • TABLE 4
    Bisanilinipyrimidine analogs.
    Figure US20220281828A1-20220908-C00034
    Cmpd
    CompoundID # R1 R2 R3 R4
    RK2-014 14 H H meta-CF3 para-COOH
    RK2-017-01 15 H H meta-CF3 para-CONH2
    RK2-037 16 H H meta-CF3 meta-
    isobutyramide
    RK2-025 17 H H meta-CF3 meta-CF3
    RK2-015-03 18 H H meta-CF3 ortho-COOH
    RK2-017-02 19 H H meta-CF3 meta-CONH2
    RK2-053 20 H H meta-CF3 meta-acetamide
    RK2-015-02 21 H H meta-CF3 meta-COOH
    RK2-056 22 H H meta-CF3 meta-butyramide
    RK2-046-02 23 H H meta-CF3 meta-
    propionamide
    RK2-013 24 H H meta-CF3 meta-
    cyclopropane
    carboxamide
    RK2-015-01 25 H H meta-CF3 All H
    RK2-046-01 26 H H meta-CF3 meta-tbutyl-
    carboxyamide
    RK2-044 27 H H meta-CF3 meta-
    cyclopentyl-
    carboxamide
    RK2-052 28 H H meta-CF3 meta-isobutyl-
    carboxamide
    RK2-043 29 H H meta-CF3 meta-(4-
    chlorobenzyl)
    carboxamide
    RK2-049 30 H H meta-CF3 meta-benzyl-
    carboxamide
    YL5-048 31 H Me2N H All H
    YL5-050 32 H Me2N ortho- All H
    COOH
    YL5-068 33 NH2 H ortho- para-COOC2H5
    COOH
    YL5-146-5 34 H H ortho-Cl para-OCH3
    YL5-080 35 Me H ortho- para-COOH
    COOH
  • In further example, the compound can be one of the following compounds.
  • Figure US20220281828A1-20220908-C00035
  • In still further examples, the compound can have Formula IIIC:
  • Figure US20220281828A1-20220908-C00036
  • wherein
    • R1 is selected from the group consisting of H, Cl, F, Br, I, C1-C6 alkyl, CN, NO2, and NH2;
    • R2 is selected from the group consisting of H, F, and Cl; and
    • each R4 is selected, independently, from the group consisting of H, COOH, CONH2, CONR5, SO2NH2, CONSO2R5, tetrazole, 4-morpholine, and COR5;
    • each R5 is selected, independently, from the group consisting of C1-C6 alkyl, cycloalkyl, heteroaryl, and heteroalkyl; and
    • m is 1-5,
  • or a pharmaceutically acceptable salt thereof.
  • In specific examples of Formula IIIC, m is 1 and R4 is selected from the group consisting of COOH, 2-CONH2, 4-CONH2, SO2NH2, tetrazole, and 4-morpholine. In other examples, R1 is Cl, F, Br, or I. In further examples, R2 is H. In still further examples, the compound is:
  • Figure US20220281828A1-20220908-C00037
  • Further specific examples, include
  • Figure US20220281828A1-20220908-C00038
  • wherein R3 and n are as defined in Formula I, and R10 is hydrogen, C1-C6 alkyl, or C1-C6 alkyl-OH.
  • Scheme I describes the general synthetic route used for preparation of dianilinipyrimidine (1) from readily available building blocks. The 2,4-dichloropyrimidine was initially reacted with the requisite commercially available anilines with the method predominantly using isopropanol as the solvent, with reflux heating to obtain the required analog.
  • Method
  • Further provided herein are methods of reducing the risk of developing, preventing, or treating graft versus host disease (GVHD) in a subject. The method can include administering to the subject an effective amount of a compound or composition as disclosed herein. The methods can further include administering a second compound or composition, such as, for example, an immunosuppressant.
  • Also disclosed are methods for treating GVHD in a patient. In one embodiment, an effective amount of one or more compounds or compositions disclosed herein is administered to a patient at risk of developing or have GVHD and who is in need of treatment thereof. The patient can be a human or other mammal, such as a primate (monkey, chimpanzee, ape, etc.), dog, cat, cow, pig, or horse, or other animals at risk of developing or have GVHD. GVHD may be due to a transplatation procedure involving the implantation of immunogenic tissue including but are not limited to, solid organ transplants (such as heart, kidney, and liver), tissue grafts (such as skin, intestine, pancreas, cornea, gonad, bone, and cartilage), and cellular transplants (such as cells from pancreas, brain and nervous tissue, muscle, skin, bone, cartilage, and liver). In such procedures, organ rejection is an obstacle to complete recovery. The individual's immune system recognizes antigens (HLA or minor H antigens) on the implanted tissue as foreign and mounts an immune response against it, which injures and destroys the implanted tissue.
  • Further provided herein are methods of treating or preventing cancer in a subject, comprising administering to the subject an effective amount of a compound or composition as disclosed herein. The methods can further comprise administering a second compound or composition, such as, for example, anticancer agents or anti-inflammatory agents. Additionally, the method can further comprise administering an effective amount of ionizing radiation to the subject.
  • Methods of killing a tumor cell are also provided herein. The methods comprise contacting a tumor cell with an effective amount of a compound or composition as disclosed herein. The methods can further include administering a second compound or composition (e.g., an anticancer agent or an anti-inflammatory agent) or administering an effective amount of ionizing radiation to the subject.
  • Also provided herein are methods of radiotherapy of tumors, comprising contacting the tumor with an effective amount of a compound or composition as disclosed herein and irradiating the tumor with an effective amount of ionizing radiation.
  • Also disclosed are methods for treating oncological disorders in a patient. In one embodiment, an effective amount of one or more compounds or compositions disclosed herein is administered to a patient having an oncological disorder and who is in need of treatment thereof. The disclosed methods can optionally include identifying a patient who is or can be in need of treatment of an oncological disorder. The patient can be a human or other mammal, such as a primate (monkey, chimpanzee, ape, etc.), dog, cat, cow, pig, or horse, or other animals having an oncological disorder. Oncological disorders include, but are not limited to, cancer and/or tumors of the anus, bile duct, bladder, bone, bone marrow, bowel (including colon and rectum), breast, eye, gall bladder, kidney, mouth, larynx, esophagus, stomach, testis, cervix, head, neck, ovary, lung, mesothelioma, neuroendocrine, penis, skin, spinal cord, thyroid, vagina, vulva, uterus, liver, muscle, pancreas, prostate, blood cells (including lymphocytes and other immune system cells), and brain. Specific cancers contemplated for treatment include carcinomas, Karposi's sarcoma, melanoma, mesothelioma, soft tissue sarcoma, pancreatic cancer, lung cancer, leukemia (acute lymphoblastic, acute myeloid, chronic lymphocytic, chronic myeloid, and other), and lymphoma (Hodgkin's and non-Hodgkin's), and multiple myeloma.
  • Administration
  • The disclosed compounds can be administered in combination with pharmaceutical formulations. Appropriate doses will be readily appreciated by those skilled in the art.
  • The term “administration” and variants thereof (e.g., “administering” a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g., a cytotoxic agent, etc.), “administration” and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
  • In vivo application of the disclosed compounds, and compositions containing them, can be accomplished by any suitable method and technique presently or prospectively known to those skilled in the art. For example, the disclosed compounds can be formulated in a physiologically- or pharmaceutically-acceptable form and administered by any suitable route known in the art including, for example, oral, nasal, rectal, topical, and parenteral routes of administration. As used herein, the term parenteral includes subcutaneous, intradermal, intravenous, intramuscular, intraperitoneal, and intrasternal administration, such as by injection. Administration of the disclosed compounds or compositions can be a single administration, or at continuous or distinct intervals as can be readily determined by a person skilled in the art.
  • The compounds disclosed herein, and compositions comprising them, can also be administered utilizing liposome technology, slow release capsules, implantable pumps, and biodegradable containers. These delivery methods can, advantageously, provide a uniform dosage over an extended period of time. The compounds can also be administered in their salt derivative forms or crystalline forms.
  • The compounds disclosed herein can be formulated according to known methods for preparing pharmaceutically acceptable compositions. Formulations are described in detail in a number of sources which are well known and readily available to those skilled in the art. For example, Remington's Pharmaceutical Science by E. W. Martin (1995) describes formulations that can be used in connection with the disclosed methods. In general, the compounds disclosed herein can be formulated such that an effective amount of the compound is combined with a suitable carrier in order to facilitate effective administration of the compound. The compositions used can also be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspension, suppositories, injectable and infusible solutions, and sprays. The preferred form depends on the intended mode of administration and therapeutic application. The compositions also preferably include conventional pharmaceutically-acceptable carriers and diluents which are known to those skilled in the art. Examples of carriers or diluents for use with the compounds include ethanol, dimethyl sulfoxide, glycerol, alumina, starch, saline, and equivalent carriers and diluents. To provide for the administration of such dosages for the desired therapeutic treatment, compositions disclosed herein can advantageously comprise between about 0.1% and 99%, and especially, 1 and 15% by weight of the total of one or more of the subject compounds based on the weight of the total composition including carrier or diluent.
  • Formulations suitable for administration include, for example, aqueous sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous sterile suspensions, which can include suspending agents and thickening agents. The formulations can be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and can be stored in a freeze dried (lyophilized) condition requiring only the condition of the sterile liquid carrier, for example, water for injections, prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powder, granules, tablets, etc. It should be understood that in addition to the ingredients particularly mentioned above, the compositions disclosed herein can include other agents conventional in the art having regard to the type of formulation in question.
  • Compounds disclosed herein, and compositions comprising them, can be delivered to a cell either through direct contact with the cell or via a carrier means. Carrier means for delivering compounds and compositions to cells are known in the art and include, for example, encapsulating the composition in a liposome moiety. Another means for delivery of compounds and compositions disclosed herein to a cell comprises attaching the compounds to a protein or nucleic acid that is targeted for delivery to the target cell. U.S. Pat. No. 6,960,648 and U.S. Application Publication Nos. 20030032594 and 20020120100 disclose amino acid sequences that can be coupled to another composition and that allows the composition to be translocated across biological membranes. U.S. Application Publication No. 20020035243 also describes compositions for transporting biological moieties across cell membranes for intracellular delivery. Compounds can also be incorporated into polymers, examples of which include poly (D-L lactide-co-glycolide) polymer for intracranial tumors; poly [bis(p-carboxyphenoxy) propane:sebacic acid] in a 20:80 molar ratio (as used in GLIADEL); chondroitin; chitin; and chitosan.
  • For the treatment of GVHD, the compounds disclosed herein can be administered to a patient at risk of developing GVHD or in need of treatment in combination with other known treatments for GVHD. These other substances or treatments can be given at the same as or at different times from the compounds disclosed herein. For example, the compounds disclosed herein can be used in combination with an immunosuppressive agent such as IMUREK™ (azathioprine sodium), brequinar sodium, SPANIDIN™ (gusperimus trihydrochloride, also known as deoxyspergualin), mizoribine (also known as bredinin), CELLCEPT™ (mycophenolate mofetil), NEORAL™ (Cyclosporin A; also marketed as a different formulation under the trademark SANDIMMUNE™), PROGRAF™ (tacrolimus, also known as FK-506), RAPIMMUNE™ (sirolimus, also known as rapamycin), leflunomide (also known as HWA-486), ZENAPAX™, glucocortcoids, such as prednisolone and its derivatives, corticosteroids, antibodies such as orthoclone (OKT3), cyclophosphamide, methotrexate, 6-mercaptopurine, vincristine, antithymyocyte globulins, such as thymoglobulins; an Aurora A inhibitor; or a Janus kinase 2 inhibitor. A conventional immunosuppressant drug, such as those above, may thus be administered in an amount substantially less (e.g. 20% to 50% of the standard dose) than when the compound is administered alone. The compounds described herein can be administered at regular intervals over a time period of at least 2 weeks.
  • Therapeutic application of compounds and/or compositions containing them can be accomplished by any suitable therapeutic method and technique presently or prospectively known to those skilled in the art. Further, compounds and compositions disclosed herein have use as starting materials or intermediates for the preparation of other useful compounds and compositions.
  • Compounds and compositions disclosed herein can be locally administered at one or more anatomical sites, such as sites of a transplant, optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent. Compounds and compositions disclosed herein can be systemically administered, such as intravenously or orally, optionally in combination with a pharmaceutically acceptable carrier such as an inert diluent, or an assimilable edible carrier for oral delivery. They can be enclosed in hard or soft shell gelatin capsules, can be compressed into tablets, or can be incorporated directly with the food of the patient's diet. For oral therapeutic administration, the active compound can be combined with one or more excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, aerosol sprays, and the like.
  • The tablets, troches, pills, capsules, and the like can also contain the following: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, fructose, lactose or aspartame or a flavoring agent such as peppermint, oil of wintergreen, or cherry flavoring can be added. When the unit dosage form is a capsule, it can contain, in addition to materials of the above type, a liquid carrier, such as a vegetable oil or a polyethylene glycol. Various other materials can be present as coatings or to otherwise modify the physical form of the solid unit dosage form. For instance, tablets, pills, or capsules can be coated with gelatin, wax, shellac, or sugar and the like. A syrup or elixir can contain the active compound, sucrose or fructose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavoring such as cherry or orange flavor. Of course, any material used in preparing any unit dosage form should be pharmaceutically acceptable and substantially non-toxic in the amounts employed. In addition, the active compound can be incorporated into sustained-release preparations and devices.
  • Compounds and compositions disclosed herein, including pharmaceutically acceptable salts, hydrates, or analogs thereof, can be administered intravenously, intramuscularly, or intraperitoneally by infusion or injection. Solutions of the active agent or its salts can be prepared in water, optionally mixed with a nontoxic surfactant. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, triacetin, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations can contain a preservative to prevent the growth of microorganisms.
  • The pharmaceutical dosage forms suitable for injection or infusion can include sterile aqueous solutions or dispersions or sterile powders comprising the active ingredient, which are adapted for the extemporaneous preparation of sterile injectable or infusible solutions or dispersions, optionally encapsulated in liposomes. The ultimate dosage form should be sterile, fluid and stable under the conditions of manufacture and storage. The liquid carrier or vehicle can be a solvent or liquid dispersion medium comprising, for example, water, ethanol, a polyol (for example, glycerol, propylene glycol, liquid polyethylene glycols, and the like), vegetable oils, nontoxic glyceryl esters, and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the formation of liposomes, by the maintenance of the required particle size in the case of dispersions or by the use of surfactants. Optionally, the prevention of the action of microorganisms can be brought about by various other antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, buffers or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the inclusion of agents that delay absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating a compound and/or agent disclosed herein in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filter sterilization. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and the freeze drying techniques, which yield a powder of the active ingredient plus any additional desired ingredient present in the previously sterile-filtered solutions.
  • For topical administration, compounds and agents disclosed herein can be applied in as a liquid or solid. However, it will generally be desirable to administer them topically to the skin as compositions, in combination with a dermatologically acceptable carrier, which can be a solid or a liquid. Compounds and agents and compositions disclosed herein can be applied topically to a subject's skin to reduce the size (and can include complete removal) of malignant or benign growths, or to treat an infection site. Compounds and agents disclosed herein can be applied directly to the growth or infection site. Preferably, the compounds and agents are applied to the growth or infection site in a formulation such as an ointment, cream, lotion, solution, tincture, or the like. Drug delivery systems for delivery of pharmacological substances to dermal lesions can also be used, such as that described in U.S. Pat. No. 5,167,649.
  • Useful solid carriers include finely divided solids such as talc, clay, microcrystalline cellulose, silica, alumina and the like. Useful liquid carriers include water, alcohols or glycols or water-alcohol/glycol blends, in which the compounds can be dissolved or dispersed at effective levels, optionally with the aid of non-toxic surfactants. Adjuvants such as fragrances and additional antimicrobial agents can be added to optimize the properties for a given use. The resultant liquid compositions can be applied from absorbent pads, used to impregnate bandages and other dressings, or sprayed onto the affected area using pump-type or aerosol sprayers, for example.
  • Thickeners such as synthetic polymers, fatty acids, fatty acid salts and esters, fatty alcohols, modified celluloses or modified mineral materials can also be employed with liquid carriers to form spreadable pastes, gels, ointments, soaps, and the like, for application directly to the skin of the user. Examples of useful dermatological compositions which can be used to deliver a compound to the skin are disclosed in U.S. Pat. Nos. 4,608,392; 4,992,478; 4,559,157; and 4,820,508.
  • Useful dosages of the compounds and agents and pharmaceutical compositions disclosed herein can be determined by comparing their in vitro activity, and in vivo activity in animal models. Methods for the extrapolation of effective dosages in mice, and other animals, to humans are known to the art; for example, see U.S. Pat. No. 4,938,949.
  • Also disclosed are pharmaceutical compositions that comprise a compound disclosed herein in combination with a pharmaceutically acceptable carrier. Pharmaceutical compositions adapted for oral, topical or parenteral administration, comprising an amount of a compound constitute a preferred aspect. The dose administered to a patient, particularly a human, should be sufficient to achieve a therapeutic response in the patient over a reasonable time frame, without lethal toxicity, and preferably causing no more than an acceptable level of side effects or morbidity. One skilled in the art will recognize that dosage will depend upon a variety of factors including the condition (health) of the subject, the body weight of the subject, kind of concurrent treatment, if any, frequency of treatment, therapeutic ratio, as well as the severity and stage of the pathological condition.
  • Kits
  • Kits for practicing the methods of the invention are further provided. By “kit” is intended any manufacture (e.g., a package or a container) comprising at least one reagent, e.g., anyone of the compounds described in Table 1. The kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention. Additionally, the kits may contain a package insert describing the kit and methods for its use. Any or all of the kit reagents may be provided within containers that protect them from the external environment, such as in sealed containers or pouches.
  • To provide for the administration of such dosages for the desired therapeutic treatment, in some embodiments, pharmaceutical compositions disclosed herein can comprise between about 0.1% and 45%, and especially, 1 and 15%, by weight of the total of one or more of the compounds based on the weight of the total composition including carrier or diluents. Illustratively, dosage levels of the administered active ingredients can be: intravenous, 0.01 to about 20 mg/kg; intraperitoneal, 0.01 to about 100 mg/kg; subcutaneous, 0.01 to about 100 mg/kg; intramuscular, 0.01 to about 100 mg/kg; orally 0.01 to about 200 mg/kg, and preferably about 1 to 100 mg/kg; intranasal instillation, 0.01 to about 20 mg/kg; and aerosol, 0.01 to about 20 mg/kg of animal (body) weight.
  • Also disclosed are kits that comprise a composition comprising a compound disclosed herein in one or more containers. The disclosed kits can optionally include pharmaceutically acceptable carriers and/or diluents. In one embodiment, a kit includes one or more other components, adjuncts, or adjuvants as described herein. In another embodiment, a kit includes one or more immunosuppressant agents, such as those agents described herein. In one embodiment, a kit includes instructions or packaging materials that describe how to administer a compound or composition of the kit. Containers of the kit can be of any suitable material, e.g., glass, plastic, metal, etc., and of any suitable size, shape, or configuration. In one embodiment, a compound and/or agent disclosed herein is provided in the kit as a solid, such as a tablet, pill, or powder form. In another embodiment, a compound and/or agent disclosed herein is provided in the kit as a liquid or solution. In one embodiment, the kit comprises an ampoule or syringe containing a compound and/or agent disclosed herein in liquid or solution form.
  • Examples
  • The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how the compounds, compositions, articles, devices and/or methods claimed herein are made and evaluated, and are intended to be purely exemplary of the invention and are not intended to limit the scope of what the inventors regard as their invention. Efforts have been made to ensure accuracy with respect to numbers (e.g., amounts, temperature, etc.), but some errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in ° C. or is at ambient temperature, and pressure is at or near atmospheric.
  • Material and Methods:
  • Monoclonal antibodies and flow cytometry: Fluorochrome-conjugated mouse anti-human monoclonal antibodies included anti-CD3, CD4, CD25, CD39, CD107a, CD127, CTLA4, Foxp3, LAG3, phosphorylated STAT3 Y705, phosphorylated STAT5 Y694, and phosphorylated H3 serine 10 (BD Biosciences, San Jose, Calif.; eBioscience San Jose, Calif.; Cell Signaling Technology, Boston, Mass.). LIVE/DEAD Fixable Yellow Dead Cell Stain (Life Technologies, Grand Island, N.Y. USA) was used to determine viability. Live events were acquired on a FACSCalibur or LSRII flow cytometer (FlowJo software, ver. 7.6.4; TreeStar, Ashland, Oreg.).
  • Allogeneic mixed leukocyte reactions: Bulk donor T-cells were allostimulated with allogeneic DCs (DC:T-cell ratio of 1:30) as previously described (OneBlood, Tampa, Fla.)(15, 16, 20). For synergy assays, TG101348 (JAK2 inhibitor, Chemietek, Indianapolis, Ind.), Alisertib (Aurora kinase A inhibitor, Selleckchem, Houston, Tex.), or both TG101348 and Alisertib (ratio 1:5, respectively) were added once on day 0. bisanilinopyrimidine (I) (dual JAK2 and Aurora kinase A inhibitor(27, 29), Moffitt Chemical Biology Core, Tampa, Fla.) or DMSO was added once on day 0 at concentrations ranging from 0.078-2.5 μM. As indicated, alloMLRs consisting of Treg depleted, naïve CD4+ T-cells (Miltenyi Biotec Inc, San Diego, Calif.) were treated with a combination of ARL67156 (CD39 inhibitor, 125 μM, Sigma), AJI, or DMSO on day 0 to study the role of CD39 and ATP in this system. T-cell proliferation was measured on day 5 by a colorimetric assay (CellTiter 96 Aqueous One Solution Cell Proliferation Assay [MTS]; or CellTiter Blue, Promega, Madison, Wis.). Absorbance/optical density (OD) was analyzed at 490 nm or 590 nm, respectively. Proliferation (%)=(OD treated alloMLR−OD T-cells alone)/(OD DMSO alloMLR−OD T-cells alone)×100.
  • Protein phosphorylation in T-cells: T-cells were cultured with allogeneic DCs (DC:T-cell ratio of 1:30) for 5 days in RPMI/10% pooled human serum, with alisertib (1.75 μM), TG101348 (350 nM), a combination of both inhibitors, AJI-214 (750 nM), AJI-100 (750 nM), or DMSO control added once on day 0. After 5 days, T cells were then harvested and directly fixed (Cytofix, BD Biosciences) for 10 min at 37° C. After washing with phosphate-buffered saline, the T cells were permeabilized with icecold methanol (90%, v/v) for at least 20 min at −20° C. The cells were stained for expression of CD3 and pH3Ser10.
  • STAT3 and STAT5 phosphorylation: As indicated, T cells were serum-starved in RPMI treated with DMSO diluent control, alisertib, TG101348, a combination of both inhibitors, AJI-214, or AJI-100 for 4 hours. IL-6-induced pSTAT3 or IL-2-induced pSTAT5 (Y694) was measured by flow cytometry.
  • NSG mice: After transplantation and treatment with either AJI-100 or vehicle control, human T cells were isolated from recipient mouse spleens at day +14, stained for pH3Ser10 or pSTAT3 (+IL-6 stimulation), and analyzed as described.
  • Effect of dual pathway inhibition on effector CD4 T cell differentiation: Purified human T-cells were allostimulated with DCs at a DC:T-cell ratio of 1:30 in RPMI/10% pooled human serum. DMSO, alisertib (1.75 mM), TG101348 (350 nM), both alisertib and TG101348, AJI-214 (750 nM), or AJI-100 (750 nM) was added once on day 0. The T cells were harvested and surface-stained on day 5 for CD3, CD4, CD25, and CD127. Activated CD4+Tconv were characterized by expression of CD25 and CD127, and activated CD8+ Tconv (CD3+ and CD4) were identified by CD25 expression. The absolute number of CD4+ and CD8+ Tconv was calculated by flow cytometry using CountBright beads (Life Technologies). T H1 cells were characterized by expression of CD3, CD4, and intracellular IFN-γ (after an additional 4 to 5 hours of stimulation with PMA/ionomycin). For T H1 experiments, purified CD4+ T cells were used as opposed to bulk T cells.
  • iTreg differentiation and potency: iTregs were generated as previously described in the presence of alisertib (1.75 mM), TG101348 (350 nM), both alisertib and TG101348, AJI-214 (750 nM), AJI-100 (750 nM), or DMSO. On day 5, iTregs were isolated and washed to minimize drug carry-over as reported. The T cells were harvested and surface-stained on day 5 for CD3, CD4, CD25, and CD127, followed by fixation and permeabilization (eBioscience) and Foxp3 staining. The absolute number of iTreg was calculated by flow cytometry using CountBright beads (Life Technologies). The purified iTregs were titrated against alloMLRs consisting of responder CD4+CD25− T cells (5×104) from the iTregs donor and fresh allogeneic DCs (1.6×103) to determine suppressive potency. T cell proliferation was determined by pulsing cells with [3H]thymidine (1 mCi per well). Surface expression of CD39 and LAG3 was evaluated on the iTregs. iTregs production of CTLA4 was assessed by intracellular staining after a 5-hour treatment of PMA/ionomycin, with GolgiStop added during the last 4 hours of incubation. iTregs synthesis of IL-10 and TGF-β (Quansys Biosciences) was quantified from supernatants using multiplex cytokine assays after PMA/ionomycin stimulation.
  • ATP Hydrolysis assay: iTregs generated in the presence of AJI-241 (750 nM) or DMSO were plated in V-bottom 96 well plates in serum free media at a concentration of 75,000 cells per 100 μL. ARL67156 (125 μM) was added or not as indicated. A fixed dose of ATP (50 μM) was added to the cells and incubated at 37° C. for 45 minutes. ATP consumption was measured by a luminescence assay per the manufacturer's instructions (Promega, CellTiter-Glo Luminescent Cell Viability Assay) and read by a spectrofluorimeter. Percent consumption was calculated as (luminescence of test supernatant/luminescence of 50 μM ATP cell-free control supernatant)×100.
  • Foxp3 TSDR demethylation analysis: Foxp3 TSDR demethylation was analyzed among magnetic bead-purified (Miltenyi), allostimulated bisanilinopyrimidine (I)- and DMSO-treated iTregs. The primer selection, procedure for amplifying methylation and demethylation specific TSDR products, genomic DNA isolation, bisulfite conversion, and qPCR were performed as previously reported.
  • RORgammaT expression by RT-PCR: Naïve CD4+ T cells were purified and allostimulated (DC/T cell ratio of 1:30) as reported (B. C. Betts et al., CD4+ T cell STAT3 phosphorylation precedes acute GVHD, and subsequent TH17 tissue invasion correlates with GVHD severity and therapeutic response. J. Leukocyte Biol. (2015)). Alisertib (1.75 mM), TG101348 (350 nM), both alisertib and TG101348, AJI-214 (750 nM), AJI-100 (750 nM), or DMSO control was added once on day 0. Medium was supplemented with IL-6 (1×105 IU/mL), TGF-β (4 ng/mL; R&D Systems), and anti-IFN-γ monoclonal antibody (10 mg/ml; eBioscience) to polarize TH17. After 5 days, the T cells were harvested, washed, and then plated at 35,000 cells per well in an IL-17 ELISPOT plate (R&DSystems). The CD4+ T cells were stimulated with PMA/ionomycin, and the ELISPOT assay was performed according to the manufacturer's instructions.
  • Xenogeneic GVHD model: NOD scid gamma (NSG) mice (male or female, 6-24 weeks old) were purchased from Jackson Laboratory (Bar Harbor, Me., USA) and raised per an IACUC-approved protocol in adherence to the NIH Guide for the Care and Use of Laboratory Animals. Mice received either (i) alisertib (30 mg/kg daily), TG101348 (45 mg/kg twice a day), a combination of alisertib and TG101348, or vehicle (methylcellulose) by oral gavage or (ii) AJI-100 (50 mg/kg daily) or vehicle (50% polyethylene glycol, 15% 2-hydroxypropyl-b-cyclodextrin, and 10% DMSO in sterile saline) ip from day 0 to day +14.
  • Mice were monitored for GVHD clinical scores (K. R. Cooke et al., An experimental model of idiopathic pneumonia syndrome after bone marrow transplantation: I. The roles of minor H antigens and endotoxin. Blood 88, 3230 (1996)), weight, and premoribund status. As indicated, mice were euthanized on day +14 to study recipient spleen Tconv, Tregs, T H1, TH17, B cells, and T-cell signal transduction. Human CD4+ Treg (CD25+, CD127, Foxp3+), Tconv (CD25+, CD127+), T H1, TH17, and CD19+ B cells residing in recipient spleens were quantified by flow cytometry. T-cell pH3 ser10 and PSTAT3 were evaluated by flow cytometry. IL-17 ELISPOTs were performed using isolated human T cells from recipient mouse spleen as described above. Tissue samples were prepared, stained (Ventana Medical Systems, Tucson, Ariz.), and imaged (Vista, Calif., USA) to identify human T-cells as previously described. All vertebrate animal work was IACUC-approved.
  • CTL generation and tumor lysis assays: NSG mice were transplanted with human PBMCs as described above, and treated with either AJI-100 (50 mg/kg daily) or vehicle control. Additionally, recipient mice received an inoculum of irradiated U937 cells (ATCC, 107/mouse) on days 0 and +7. Mice were euthanized between days +10 to +12, spleens were harvested, and human CD8+ T-cells were isolated by magnetic bead separation. Fresh U937 target cells were labeled with Calcein-AM for 30 minutes, washed, and then cultured with the purified CD8+ T-cells at varying effector to target ratios for 4 hours at 37° C. No additional drugs were added during this final culture. The amount of supernatant fluorescence released by the target cells was measured using a spectrofluorimeter (485 nm excitation and 535 nm emission) (S. Neri, E. Mariani, A. Meneghetti, L. Cattini, A. Facchini, Calcein-acetyoxymethyl cytotoxicity assay: standardization of a method allowing additional analyses on recovered effector cells and supernatants. Clinical Diagnostic Lab. Immunol. 8, 1131 (2001)). Percent lysis was calculated as follows: [(test fluorescence−spontaneous fluorescence)/(maximum fluorescence−spontaneous fluorescence)]×100. For in vitro CTL generation, human PBMCs were cultured with irradiated U937 cells at a ratio of 1:1 in the presence of bisanilinopyrimidine (I) 750 nM or DMSO for 10 days. Cultures were replenished with media and inhibitors on days +3 and +7, and fresh irradiated U937 cells added on day +7. On day +10, the cells were harvested and CD8 T-cells were isolated by magnetic bead separation (Miltenyi).
  • The tumor lysis assay was performed as described using fresh, Calcein-AM labeled U937 targets.
  • Statistical Analysis: For comparisons of paired data sets, the paired t test was used. ANOVA was used for group comparisons. Survival comparisons were made using the log-rank test. The Mann-Whitney test was used for all others. The statistical analysis was conducted using Prism software version 5.04 (GraphPad). Statistical significance was defined by P<0.05 (two-tailed).
  • For drug combination experiments, the results were analyzed for synergistic, additive, or antagonistic effects using the combination index (CI) method developed by Chou and Talalay.
  • The dose-effect curve for each drug alone was determined based on experimental observations using the median-effect principle and then compared to the effect achieved with a combination of the two drugs to derive a CI value. For this analysis,
  • XLfit software (IDBS) was used to create log-log dose-fractional effect plots for each drug and combination and to regress a straight line through the points and was used to calculate the values of Dm and m for use in the median-effect equation as follows: fa/fu=(D/Dm)m, where D is the dose of the drug, Dm is the dose required for a 50% effect (analogous to IC50), fa and fu are the affected and unaffected fractions, respectively (fa=1−fu), and m is the exponent signifying the sigmoidicity of the dose-effect curve. The CI calculation chosen for the analysis of the drug combinations was the isobologram equation for mutually nonexclusive drugs with different modes of action as follows: CI=(D)1/(Dx)1+(D)2/(Dx)2+(D)1(D)2/(Dx)1(Dx)2, where (Dx)1 and (Dx)2 in the denominators are the concentrations for drug 1 and drug 2 alone that gave x % inhibition, whereas (D)1 and (D)2 in the numerators are the concentrations of drug 1 and drug 2 in combination that also inhibited x % (that is, isoeffective).
  • Synthesis of bisanilinopyrimidine (I) and bisanilinopyrimidine (II): The bisanilinopyrimidine bisanilinopyrimidine (I) was prepared using a method previously reported (Lawrence, et al., Development of o-chlorophenyl substituted pyrimidines as exceptionally potent aurora kinase inhibitors. J. Med. Chem. 2012, 55, 7392-416.). The bisanilinopyrimidine bisanilinopyrimidine (II) was prepared using a two step route (Scheme I) to prepare other 2,4-dianilinopyrimidines (1). Reaction of 2,4-dichloro-5-fluoropyrimidine with aniline provided the intermediate 1. Intermediate 1 was reacted further with 4-aminobenzamide to give the required bisanilinopyrimidine (II), with HPLC purity >99%.
  • Figure US20220281828A1-20220908-C00039
  • 2-Chloro-5-fluoro-N-phenylpyrimidin-4-amine (intermediate 1): To a solution of 5-fluoro-2,4-dichloropyrimidine (2.00 g, 11.98 mmol) and diethylisopropylamine (2.50 mL, 14.37 mmol) in isopropanol (12 mL) was added aniline (1.09 mL, 11.98 mmol). The mixture was stirred at reflux for 1 h. The reaction mixture was concentrated under reduced pressure. The residue was dissolved in dichloromethane (20 mL) and washed with water (10 mL) and brine (10 mL). The organic layer was dried (Na2SO4) and concentrated under reduced pressure. The resulting solid was triturated using EtOAc/hexanes to give the title compound as a white solid (1.35 g, 50%). Mp: 135-136° C. 1H NMR (400 MHz, DMSO-d6): δ 9.99 (s, 1H, disappeared on D2O shake), 8.30 (d, J=3.5 Hz, 1H), 7.66 (d, J=8.0 Hz, 2H), 7.37 (t, J=8.0 Hz, 2H), 7.13 (t, J=8.0 Hz, 1H). 19F NMR (376 MHz, DMSO-d6): δ −153.7 (s). HPLC-MS (ESI+): m/z 226.1 [40%, (M37Cl+H)+], 224.1 [100%, (M35Cl+H)+].
  • 5-Fluoro-N4-phenyl-N2-[4-(4-carboxamide)phenyl]pyrimidine-2,4-diamine (bisanilinopyrimidine (II)): A mixture of intermediate 1 (1.00 g, 4.47 mmol), 4-aminobenzamide (0.609 g, 4.47 mmol), and methanol (4.5 mL) was heated at 100° C. for 14 h. The reaction mixture was cooled to room temperature and the precipitate filtered and washed with MeOH (2×10 mL). The resulting solid was sonicated in saturated sodium bicarbonate solution (10 mL) for 2 min, then filtered, washed with water (3×20 mL), MeOH (2×10 mL), and dried to give bisanilinopyrimidine (II) as a white solid (1.07 g, 74%). Mp: 248-249° C. HPLC: 99.9% [tR=10.9 min, 50% MeOH, 50% water (with 0.1% TFA), 20 min]. 1H NMR (400 MHz, DMSO-d6): δ 9.51 (s, 1H, disappeared on D2O shake), 9.44 (s, 1H, disappeared on D2O shake), 8.14 (d, J=3.7 Hz, 1H), 7.77 (brs, 1H, disappeared on D2O shake), 7.76 (d, J=7.8 Hz, 2H), 7.72 (s, 4H), 7.36 (t, J=7.8 Hz, 2H), 7.12 (brs, 1H, disappeared on D2O shake), 7.10 (t, J=7.8 Hz, 1H). 19F NMR (376 MHz, DMSO-d6): δ −163.2 (s). HPLC-MS (ESI+): m/z 324.2 [100%, (M+H)+]. LC-MS (ESI+): 992.3 [20%, (3M+Na)+], 669.2 [50%, (2M+Na)+], 346.1 [30%, (M+Na)+], 324.1 [100%, (M+H)+]. HRMS (ESI+): m/z calcd for C17H14FN5O (M+H)+ 324.1255, found 324.1262.
  • Study 1:
  • Synergistic immune suppression with combined inhibition of Aurora kinase A and JAK2 Using allogeneic mixed leukocyte reactions (alloMLRs), the JAK2 inhibitor, TG101348, reduced alloreactive T-cell proliferation at concentrations of 350 nM and greater as previously reported(15) (FIG. 1A). The Aurora kinase A inhibitor, alisertib (M. G. Manfredi et al., Characterization of Alisertib (MLN8237), an investigational small molecule inhibitor of aurora A kinase using novel in vivo pharmacodynamic assays. Clinical Cancer research 17, 7614 (2011); H. Yang et al., Dual Aurora A and JAK2 kinase blockade effectively suppresses malignant transformation. Oncotarget 5, 2947 (2014)), suppressed the proliferative response of T-cells in alloMLRs at 625 nM (32.5% inhibition) with an IC50 of 10 NM (FIG. 1A). Synergistic suppression over T-cells allostimulated by dendritic cells (DC) was achieved when TG101348 and alisertib were added together at a ratio of 1:5, respectively, with a calculated combination index (CI) of <1 per the Chou and Talalay method (T. C. Chou, Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol. Rev. 58, 621 (2006)) (FIG. 1A). The observed IC50 of the combination correlated with 350 nM of TG101348 and 1.75 μM of alisertib (FIG. 1A).
  • Bisanilinopyrimidine (I) is a potent inhibitor of Aurora kinase A and JAK2 that was designed and synthesized at Moffitt Cancer Center (H. R. Lawrence et al., Development of o-chlorophenyl substituted pyrimidines as exceptionally potent aurora kinase inhibitors. J. Med. Chem. 55, 7392 (2012)). Bisanilinopyrimidine (I) exerted significant suppression over Tcells in alloMLRs, with single agent efficacy at nanomolar concentrations (FIG. 1B). Bisanilinopyrimidine (I) significantly decreased the activity of Aurora kinase A and JAK2 in DC-allostimulated T-cells, and reduced phosphorylation of histone 3 serine 10 (pH3 Ser10) and signal transducer and activator of transcription 3 (pSTAT3 Y705) respectively (FIGS. 1C-1D). The viability of AJI-214 (750 nM) or DMSO treated T-cells was similar after 5 days of co-culture (FIG. 1E).
  • Concurrent blockade of Aurora kinase A and JAK2 selectively impairs alloreactive Tconv, while sparing responder Tregs bisanilinopyrimidine (I) exerted dose-dependent inhibition of alloreactive CD4+ Tconv (CD25+, CD127+) (A. K. Heninger et al., IL-7 abrogates suppressive activity of human CD4+CD25+FOXP3+ regulatory T cells and allows expansion of alloreactive and autoreactive T cells. J. Immunol. 189, 5649 (2012); S. Samarasinghe et al., Functional characterization of alloreactive T cells identifies CD25 and CD71 as optimal targets for a clinically applicable allodepletion strategy. Blood 115, 396 (2010); S. Touil et al., Depletion of T regulatory cells through selection of CD127-positive cells results in a population enriched in memory T cells: implications for anti-tumor cell therapy. Haematologica 97, 1678 (2012)) in alloMLRs (FIG. 2A). The CD4+ Treg (CD25+, CD127) (W. Liu et al., CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ Treg cells. J. Experi. Med. 203, 1701 (2006); N. Seddiki et al., Expression of interleukin (IL)-2 and IL-7 receptors discriminates between human regulatory and activated T cells. J. Experi. Med. 203, 1693 (2006)) population was preserved even when co-cultures were exposed to 750 nM of bisanilinopyrimidine (I) (FIG. 2B). The high degree of selectivity demonstrated by dual inhibition of Aurora kinase A and JAK2 resulted in a significant increase in the Treg:allo Tconv ratio (FIG. 2C-2D). Cell Trace Violet dilution was used to study the effect of dual Aurora kinase A/JAK2 blockade on proliferation among the individual Treg and Tconv compartments. This verified the observed shifts in Treg:Tconv populations by bisanilinopyrimidine (I) exposure, where a significant decrease in Tconv proliferation occurred with bisanilinopyrimidine (I) at 750 nM (FIGS. 2E-2F). Treg proliferation was similar between bisanilinopyrimidine (I) and control (FIGS. 2E-2F). STAT5 activation remained functional in IL-2 stimulated CD4+ T-cells exposed to either DMSO or bisanilinopyrimidine (I) compared with unstimulated baseline controls, though the effect was blunted by bisanilinopyrimidine (I) (FIG. 2G). Moreover, CD4+, CD25+, CD127 Treg expression of Foxp3 was maintained in the presence of bisanilinopyrimidine (I) or DMSO (FIG. 2H).
  • To confirm the Treg-sparing effects were related to concurrent inhibition of JAK2 and Aurora kinase A, as opposed to a phenomenon of bisanilinopyrimidine (I) alone, similar allogeneic co-cultures were treated with either TG101348 (JAK2 inhibitor, 350 nM), alisertib (Aurora kinase A inhibitor, 1.75 NM), a combination of TG101348 and alisertib, or DMSO. These doses of TG101348 and alisertib were based on the synergy studies, where a 1:5 ratio of each respective compound was used concurrently. As observed with bisanilinopyrimidine (I), dual blockade of JAK2 and Aurora kinase A reduced the alloreactive Tconv population and spared the responder Tregs (FIG. 2I).
  • Dual inhibition of Aurora kinase A and JAK2 favors iTreg development and potency. The effects of dual Aurora A/JAK2 blockade on inducible Treg (iTreg) differentiation was studied. Naïve CD4+ T-cells (>99% pure) were depleted of natural Tregs and stimulated with allogeneic DCs for 5 days in the presence of bisanilinopyrimidine (I) or DMSO (FIG. 3A). Bisanilinopyrimidine (I) permitted iTreg differentiation, and significantly decreased the frequency of alloreactive Tconv (FIGS. 3B-3C). Given that iTregs are derived from phenotypically plastic naïve CD4+ T-cells, it was confirmed that demethylated Foxp3 TSDR was similar among bisanilinopyrimidine (I)- and DMSO-exposed iTreg (FIG. 3D). To determine the influence of Aurora A versus JAK2 inhibition on iTreg differentiation, co-cultures were treated with bisanilinopyrimidine (I) 750 nM, alisertib 1.75 μM, TG101348 350 nM, a combination of both, or DMSO. While alisertib, TG101348, and the combination of each all had less absolute numbers of iTregs and allo Tconv compared to DMSO, the suppressive effect on Tconv was greater resulting in incrementally larger Treg:allo Tconv ratios (FIG. 3E).
  • Bisanilinopyrimidine (I) demonstrated minimal loss of iTregs and significantly reduced allo Tconv compared to DMSO, increasing the iTreg:allo Tconv ratio (FIG. 3E). To study the influence of dual pathway inhibition on iTreg suppressive function, bisanilinopyrimidine (I)- or DMSO-treated iTreg were cultured with self alloresponders targeting fresh allogeneic DCs. The bisanilinopyrimidine (I)-treated iTreg not only demonstrated intact suppressive function, their potency was significantly increased by approximately 30% compared to DMSO-treated iTreg(FIG. 4A). It was then explored how Aurora kinase A versus JAK2 blockade contributed to this enhanced suppression by the iTregs. iTregs were generated as described in the presence of alisertib, TG101348, a combination of both, or DMSO. Interestingly, Aurora kinase A inhibition with alisertib demonstrated superior suppressive capacity compared with either DMSO- or TG101348-exposed iTreg (FIG. 4B). The combination of alisertib with TG101348 was similar to alisertib alone (FIG. 4B).
  • Also investigated was the mechanism supporting the increased iTreg function observed with bisanilinopyrimidine (I). Identified was a significant increase in the relative surface density of CD39, an ectonucleotidase that hydrolyzes ATP, among the bisanilinopyrimidine (I)-exposed iTreg compared with DMSO controls (FIGS. 5A-5D). CD39 expression on non-Treg CD4+ T-cells was minimal (M. Mandapathil et al., Generation and accumulation of immunosuppressive adenosine by human CD4+CD25highFOXP3+ regulatory T cells. J. Biol. Chem. 285, 7176 (2010)) (FIG. 5B). It was confirmed that the higher CD39 expression among the bisanilinopyrimidine (I) treated iTreg resulted in improved scavenging of extracellular ATP, compared to DMSO-treated iTreg (FIG. 5E). The enhanced hydrolysis of ATP by the AJI-treated iTregs was also significantly impaired by blocking the CD39 enzyme with ARL67156 (FIG. 5E). To determine the influence of CD39+ iTreg in the overall efficacy of dual Aurora A/JAK2 blockade, ARL67156 was added to alloMLRs consisting of natural Treg-depleted CD4+ T-cell responders with bisanilinopyrimidine (I) or DMSO. This eliminated potential interference from CD39+ natural Treg within the allogeneic co-culture. CD39 blockade significantly weakened the T-cell inhibition by bisanilinopyrimidine (I) (FIG. 5F), supporting that CD39+iTregs contribute to the immune suppressive effects of bisanilinopyrimidine (I). With regard to other modes of iTreg suppression, no difference in their expression of LAG3, CTLA4, or production of IL-10 or TGF-beta after exposure to bisanilinopyrimidine (I) or DMSO (FIGS. 5G-5J).
  • As previously reported JAK2 inhibition with TG101348 impairs TH17 production of IL-17, it was investigated how Aurora kinase A inhibition influenced RORgammaT expression. Alisertib exerted no effect on RORgammaT levels among alloresponder CD4+ T-cells, while JI-214 significantly decreased the expression of this key TH17-differentiation transcription factor compared with DMSO (FIG. 6A). It was confirmed that TG101348 significantly decreases RORgammaT expression in CD4+ alloresponders (FIG. 6B). These data suggest that bisanilinopyrimidine (I) supports enhanced iTreg potency primarily through Aurora kinase A blockade, while its suppression of TH17 polarization is a function of JAK2 blockade.
  • Targeting Aurora kinase A and JAK2 reduces xenogeneic GVHD and preserves GVL A xenogeneic GVHD model was used to investigate the in vivo efficacy of dual Aurora kinase A/JAK2 blockade while maintaining a focus on human immune responses. Recipient NSG mice were transplanted with 30×106 human PBMCs i.p. once on day 0. Bisanilinopyrimidine (I) is not suited for in vivo use, due to limited bioavailability. Bisanilinopyrimidine (II) is an Aurora kinase A/JAK2 inhibitor (Moffitt Cancer Center) that differs from bisanilinopyrimidine (I) by only a chlorine to hydrogen substitution at the ortho position of its phenyl ring enhancing its solubility. The bisanilinopyrimidine (II) and bisanilinopyrimidine (I) analogues both inhibit Aurora kinase A and JAK2 with similar potency. As observed with bisanilinopyrimidine (I), bisanilinopyrimidine (II) reduced responder T-cell proliferation in alloMLRs at nanomolar concentrations (IC 50 200 nM, FIG. 7A).
  • To study the concept of dual Aurora kinase A/JAK2 inhibition as GVHD prevention, mice were treated with bisanilinopyrimidine (II) at 50 mg/kg daily i.p. or vehicle control from day 0 to day +14. JAK2 inhibition was confirmed in vitro, where human T-cells stimulated with IL-6 expressed less STAT3 phosphorylation with bisanilinopyrimidine (II) exposure compared with DMSO (FIG. 7B). Harvested human T-cells from the mice at day +14 showed less H3 ser10 phosphorylation with bisanilinopyrimidine (II) compared to vehicle, confirming Aurora inhibition (FIG. 7C). The vehicle control treated mice developed acute xenogeneic GVHD (including fur loss, skin changes, weight loss, and kyphosis) by the third week of the transplant with a median survival of 34 days (FIGS. 7D-7E). The overall survival of the bisanilinopyrimidine (II)-treated mice was 71.4% during the 56 days of observation, while none of the vehicle-treated mice survived past day +45 (FIG. 7F). The average GVHD clinical scores were <2 among the surviving bisanilinopyrimidine (II)-treated mice at day +56 (FIG. 7E), where 60% showed limited fur/skin changes without significant weight loss (FIGS. 7D-7E). Conversely, bisanilinopyrimidine (II) and vehicle treated mice both facilitated the generation of U937-specific CTL in vivo and retained similar anti-tumor killing in vitro (FIG. 7G). It was confirmed that the bisanilinopyrimidine (I) analogue similarly allowed for CTL generation in vitro, and that CD8+ CTL remained functional in tumor lysis assays against U937 targets (FIG. 7H).
  • The absolute number of total spleen cells and human CD3+ T-cells within the spleens were similar among vehicle- versus bisanilinopyrimidine (II)-treated mice (FIGS. 8A-8B). bisanilinopyrimidine (II) significantly increased the relative amount of human Tregs in the spleen, while the percentage of CD4+ alloreactive Tconv was similar among both groups of transplanted mice (FIGS. 8C-8G). As such, bisanilinopyrimidine (II) significantly increased the ratio of human Treg to CD4+ alloreactive Tconv in the recipient spleens, compared with vehicle (FIG. 8E). GVHD pathology at day +14 was limited to the liver, where bisanilinopyrimidine (II) dramatically reduced human T-cell invasion (FIGS. 8H-8J). Treg frequency in the liver was similar among both treatment groups, however (FIG. 8K).
  • T-cell costimulation and cytokine activation independently contribute to GVHD, but control of donor alloresponses is incomplete when targeting either pathway alone. It has been disclosed herein that GVHD prevention with intact GVL can be accomplished by dual inhibition of Aurora kinase A and JAK2, respectively attenuating CD28 costimulation and IL-6-mediated signal transduction. Concurrent blockade of Aurora kinase A and JAK2 yields synergistic immune suppression over human allogeneic T-cells in vitro, preserves iTreg differentiation, and significantly enhances iTreg suppressive potency. These characteristics are distinct from CNI-based GVHD prophylaxis, which abrogates TCR function and indiscriminately suppresses donor T-cells. The lack of selectivity by CNIs results in a failure to achieve donor immune tolerance toward the host and negates the GVL potential of the allograft. Selective targeting of Aurora kinase A and JAK2 signal transduction controls fundamental aspects of T-cell allo-activation, without ceding TCR function required by Tregs and anti-tumor CTL.
  • Blockade of Aurora kinase A or JAK2 induces pathway-specific effects on developing iTreg and TH17. It was observed that the dual pathway inhibitor, bisanilinopyrimidine (I), significantly increased the suppressive potency of allo-antigen specific iTreg. The data supports that the enhanced iTreg potency is largely a function of Aurora kinase A inhibition. While JAK2 blockade with TG101348 improved iTreg function compared with DMSO, iTregs previously exposed to alisertib profoundly eliminated Tconv proliferation. On the other hand, alisertib was unable to prevent RORgammaT expression in naïve CD4+ T-cells responding to allo-antigen unlike AJI-214 or TG101348. Given that IL-6 receptor signal transduction facilitates TH17 development, the data confirm that JAK2 blockade with bisanilinopyrimidine (I) or TG101348 significantly restrains TH17 differentiation.
  • Modes of Treg suppression to understand how bisanilinopyrimidine (I) improved iTreg suppressive potency was investigated. iTreg production of the anti-inflammatory cytokines, IL-10 and TGF-beta, where comparable among bisanilinopyrimidine (I)- and DMSO-treated iTregs. Moreover, iTreg expression of CTLA4 and LAG3 was similar among each experimental condition. Interestingly, it was identified that bisanilinopyrimidine (I) significantly increased the surface density of CD39 expressed on the iTregs. CD39 is an ectonucleotidase that hydrolyzes extracellular ATP and reduces T-cell activation (M. Vukmanovic-Stejic et al., The kinetics of CD4+Foxp3+ T cell accumulation during a human cutaneous antigen-specific memory response in vivo. The J. Clinical Invest. 118, 3639 (2008)).
  • CD39+ Tregs correlate with clinical outcomes in autoimmune diseases (R. S. Peres et al., Low expression of CD39 on regulatory T cells as a biomarker for resistance to methotrexate therapy in rheumatoid arthritis. Proc. Natl. Acad. Sci. USA 112, 2509 (2015); A. Thiolat et al., Interleukin-6 receptor blockade enhances CD39+ regulatory T cell development in rheumatoid arthritis and in experimental arthritis. Arthritis & Rheumatol. 66, 273 (2014)). A decrease in CD39+ Tregs is associated with methotrexate-failure among rheumatoid arthritis (RA) patients. Alternatively, IL-6 neutralization with tocilizumab increases CD39+ Tregs in a similar group of RA patients (38). It was verified that bisanilinopyrimidine (I)-treated iTreg degraded extracellular ATP more efficiently than those exposed to DMSO. This response was impaired by blocking CD39 with the inhibitor ARL67156. Additionally, neutralization of CD39 activity significantly attenuated the immune suppressive effect of dual Aurora kinase A/JAK2 inhibition in nTreg depleted alloMLRs. This data supports that increased CD39 expression is relevant to the enhanced iTreg function mediated by bisanilinopyrimidine (I).
  • While inhibition of Aurora kinase A or JAK2 activity individually suppressed human Tcell proliferation in alloMLRs, synergy was achieved with simultaneous blockade of both signal transduction pathways. The bi-specific inhibitors, bisanilinopyrimidine (II) and bisanilinopyrimidine (I), demonstrated potent single agent activity at nanomolar concentrations. As published, bisanilinopyrimidine (II) and bisanilinopyrimidine (I) exhibit similar activity against Aurora kinase A and JAK2. Bisanilinopyrimidine (II) differs from bisanilinopyrimidine (I) by a single chlorine to hydrogen substitution on its phenyl ring to facilitate in vivo solubility for mouse studies, but the compounds are otherwise chemically and functionally similar.
  • Target inhibition was confirmed as bisanilinopyrimidine (II) and bisanilinopyrimidine (I) significantly reduced the phosphorylation of both STAT3 and H3 ser10 in human T-cells. Conversely, combined Aurora kinase A/JAK2 blockade permitted IL-2-induced STAT5 activation in T-cells, compared with resting, unstimulated controls. The selective inhibition of Aurora kinase A and JAK2 paired with preserved common gamma chain cytokine signaling establishes a platform to control alloreactivity while maintaining antigen-specific Treg and CTL responses. Accordingly, it was observed that bisanilinopyrimidine (II) significantly reduces GVHD, increases the proportion of Treg to allo Tconv, and preserves CTL generation and anti-tumor activity. CNI-free GVHD prophylaxis is an important concept in improving patient outcomes after clinical transplantation. The challenges of CNI-based GVHD prevention are clear; as CNIs offer incomplete protection from severe GVHD and render the donor immune system poorly equipped to counter post-transplant relapse. Given that targeting Aurora kinase A and JAK2 selectively eliminates alloreactive Tconv while sparing Tregs and tumor-specific CTL, the novel concept described here may represent a translatable CNI-free approach at GVHD prevention. A limited number of CNI-free GVHD prophylaxis strategies currently exist, and include T-cell depletion of the allograft (M. C. Pasquini et al., Comparative outcomes of donor graft CD34+ selection and immune suppressive therapy as graft-versus-host disease prophylaxis for patients with acute myeloid leukemia in complete remission undergoing HLA-matched sibling allogeneic hematopoietic cell transplantation. J. Clinical Oncol. 30, 3194 (2012)) or the use of post-transplant cyclophosphamide (C. G. Kanakry et al., Single-agent GVHD prophylaxis with posttransplantation cyclophosphamide after myeloablative, HLA-matched BMT for AML, ALL, and MDS. Blood 124, 3817 (2014)). The bispecific inhibitor, bisanilinopyrimidine (II), is an attractive alternative as it does not require ex vivo allograft modification or the need to expose freshly infused donor stem cells to potent alkylators. As such, further investigation of dual Aurora kinase A/JAK2 inhibition is merited to promote selective control over donor immune responses after alloHCT.
  • Study 2:
  • Synergistic Immunosuppression is Attainable with Combined Inhibition of Aurora Kinase a and JAK2.
  • Allogeneic mixed leukocyte reactions (alloMLRs) are standard assays used to assess human T cell proliferation against polyclonal or antigen-specific stimuli. In alloMLRs consisting of human T cells and allogeneicmonocyte-derived dendritic cells (DCs), the JAK2 inhibitor TG101348 reduced alloreactive T cell proliferation at concentrations of 350 nM and greater as previously reported (FIG. 9A). The Aurora kinase A inhibitor alisertib suppressed the proliferative response of T cells in alloMLRs with a median inhibitory concentration (IC50) of 10 mM (FIG. 9A). Synergistic suppression of T cells allostimulated by DCs was achieved when TG101348 and alisertib were added together at a ratio of 1:5, respectively, with a calculated combination index (CI) of <1 using the Chou-Talalay method (FIG. 9A). The observed IC50 of the combination correlated with 350 nM TG101348 and 1.75 mM alisertib (FIG. 9A). The chemical analogs AJI-214 and AJI-100 were designed and synthesized at the Moffitt Cancer Center and shown to inhibit Aurora kinase A and JAK2 with similar potency. AJI-100 differs from AJI-214 by a single chlorine to hydrogen substitution at the ortho position of its phenyl ring, enhancing its solubility, hence its preferred use in vivo (FIGS. 9B-9C). Because AJI-100 is tolerated in mouse models, a kinase target screen was performed on AJI-100 to verify its activity against Aurora kinase A and JAK2 among a panel of 140 kinases. Aurora kinase A and JAK2 were among the top three kinases inhibited by AJI-100. It was found that AJI-100 also inhibits 5′ AMP activated protein kinase (AMPK) and exhibits slightly more potent suppression of Aurora kinase B than alisertib. AJI-214 and AJI-100 exerted significant suppression of T cells in alloMLRs, with single-agent efficacy at nanomolar concentrations (P<0.05; FIGS. 9B-9C). Moreover, the AJI analogs suppressed alloreactive T cell proliferation similar to the potency of alisertib (1.75 mM) and TG101348 (350 nM) combined (FIG. 9D). AJI-214 and AJI-100 (750 nM for each) also exhibited similar target inhibition of Aurora kinase A and JAK2 signal transduction in human T cells, reducing the phosphorylation of histone 3 serine 10 (pH3Ser10) and STAT3 (pSTAT3) Y705, respectively (FIGS. 9E-9H). As expected, alisertib only inhibited pH3Ser10 (FIGS. 9E-9F), and TG101348 only inhibited pSTAT3 (FIGS. 9G-9H).
  • Concurrent Blockade of Aurora Kinase a and JAK2 is Immunosuppressive and Permits the Differentiation of Inducible Treg
  • DMSO, alisertib, TG101348, a combination of alisertib and TG101348, AJI-214, or AJI-100 was added to allogeneic cocultures of DC-stimulated T cells. Activated CD4+ Tconv were identified as coexpressing CD25 and CD127, and the latter assisted in excluding Tregs from the analysis. CD25 expression alone was used to identify activated CD8+ Tconv. Although all inhibitors suppressed the activated Tconv compared to DMSO, combined inhibition of Aurora A and JAK2 offered greater immunosuppression than either alisertib (P=0.007, FIG. 10A; P=0.002, FIG. 10B) or TG101348 alone (P=0.02, FIG. 10A; P=0.02, FIG. 10B). To quantify the effect of Aurora A/JAK2 blockade on TH17, IL-17 ELISPOTs was performed using DC-stimulated, purified CD4+ T cells in the presence of the compounds or DMSO. As predicted by the effect of each inhibitor on STAT3 phosphorylation, the JAK2-targeting compounds significantly reduced TH17, whereas alisertib had no effect (P<0.05; FIG. 10C). All of the inhibitors significantly decreased the frequency of interferon-γ+ (IFN-γ+) TH1 T cells among treated allogeneic cocultures (P<0.05; FIG. 10D). However, dual blockade of Aurora A and JAK2 did not offer increased suppression of T H1 compared to either inhibitor alone (FIG. 10D). Given that inhibition of Aurora A and JAK2 significantly reduced alloreactive Tconv, T H1, and TH17 cells, the effects of dual blockade on inducible Treg (iTreg) differentiation was then studied. Naïve CD4+ T cells (>99% pure; FIG. 10E) were depleted of natural Tregs and stimulated with allogeneic DCs for 5 days in the presence of DMSO, alisertib, TG101348, a combination of alisertib and TG101348, AJI-214, or AJI-100. The iTregs were identified as CD4+, CD127, CD25+, and Foxp3+. iTreg conversion from naïve CD4+ precursors was variably reduced by all of the compounds compared to DMSO (FIGS. 10F-10G), and Aurora A inhibition appeared to exert greater iTreg impairment than JAK2 inhibition (FIG. 10G). In contrast, IL-2-induced STAT5 phosphorylation, which is required for Treg development, remained intact among T cells treated with alisertib, TG101348, a combination of alisertib and TG101348, or the AJI analogs compared to DMSO (FIG. 10H).
  • Dual Inhibition of Aurora Kinase A and JAK2 Supports Potent CD39+iTreg
  • AJI-214 and AJI-100 were confirmed to exhibit identical suppressive potency in regard to Aurora A and JAK2 signal transduction and human T cell proliferation assays. Therefore, AJI-214 was used as the representative bispecific analog for additional iTreg-based in vitro mechanistic tests. Given that iTregs are derived from phenotypically plastic naïve CD4+ T cells, it was confirmed that demethylated Foxp3 Treg-specific demethylated region (TSDR) was similar among AJI-214-exposed and DMSO-exposed iTregs (FIG. 11A). To study the influence of dual pathway inhibition on iTreg-suppressive function, AJI-214-treated or DMSO-treated iTreg were cultured with autologous T cells targeting fresh allogeneic DCs. The AJI-214-treated iTregs demonstrated intact suppressive function, and its potency was significantly increased by about 30% compared to DMSO-treated iTregs (P=0.018; FIG. 11B). How Aurora kinase A versus JAK2 blockade contributed to this enhanced suppression by the iTregs was explored. Antigen-specific iTregs were generated from CD25-depleted CD4+ T cells in the presence of alisertib, TG101348, a combination of alisertib and TG101348, or DMSO. Aurora kinase A inhibition with alisertib demonstrated superior suppressive capacity compared with either DMSO-exposed (P=0.03) or TG101348-exposed (P=0.04) iTreg (FIG. 11C). The combination of alisertib with TG101348 was similar to alisertib alone (FIG. 11C). The mechanism supporting the increased iTreg function observed with AJI-214 was investigated. A significant increase in the cell surface density of CD39, an ectonucleotidase that hydrolyzes adenosine triphosphate (ATP), was identified among the AJI-214-exposed iTreg compared with DMSO controls (P=0.045; FIGS. 12A-12D). As reported by others, CD39 expression on non-Treg CD4+ T cells was minimal (FIGS. 12B-12D). The higher CD39 cell surface density was confirmed among the AJI-214-treated iTregs resulted in improved scavenging of extracellular ATP, compared to DMSO-treated iTregs (FIG. 12E). The enhanced hydrolysis of ATP by the AJI-214-treated iTregs was also significantly impaired by blocking the CD39 enzyme with ARL67156 (P<0.0001; FIG. 12E). To determine the influence of CD39+ iTreg in the overall efficacy of dual Aurora A/JAK2 blockade, ARL67156 was added to alloMLRs consisting of natural Treg-depleted CD4+ T cell responders with AJI-214 or DMSO. This eliminated potential interference from CD39+ natural Treg within the allogeneic coculture and ensured that the only Tregs present in the system were induced. Moreover, ARL67156 would primarily affect the iTreg as Tconv express negligible amounts of CD39. CD39 blockade significantly weakened the T cell inhibition by AJI-214 (P=0.037; FIG. 12F), supporting that CD39+ iTregs contribute to the immunosuppressive effects of AJI-214. With regard to other modes of iTreg suppression, no difference in their expression of LAG3 and CTLA4 or production of IL-10 or transforming growth factor-β (TGF-β) was found after exposure to AJI-214 or DMSO (FIG. 12G-12J).
  • Targeting Aurora Kinase a and JAK2 Reduces Xenogeneic GVHD and Preserves the Generation Antitumor CTL
  • A xenogeneic GVHD model was used to investigate the in vivo efficacy of dual Aurora kinase A/JAK2 blockade to specifically evaluate effects on human immune responses. Recipient NOD (nonobese diabetic) scid g (NSG) mice were transplanted with human peripheral blood mononuclear cells (PBMCs) (30×106 cells) intraperitoneally (ip) on day 0. Independent donors were used for each experiment. Whether combining individual inhibitors of Aurora A and JAK2 prevented acute xenogeneic GVHD was tested. Mice received alisertib (30 mg/kg daily), TG101348 (45 mg/kg twice a day), a combination of alisertib and TG101348, or methylcellulose vehicle from days 0 to +14 by oral gavage. The drug combination significantly delayed the onset and severity of GVHD, compared to vehicle or TG101348 alone (P<0.0001 and P=0.0001, respectively; FIGS. 13A-13B). There was also a suggestion toward an improved median survival with the drug combination compared to alisertib (50.5 versus 41 days; P=not significant). The bispecific inhibitor AJI-100 was used to test the in vivo efficacy of single agent blockade of Aurora A and JAK2 as GVHD prevention. As demonstrated, AJI-100 offers identical on-target inhibition and immunosuppressive properties as AJI-214 but exhibits superior bioavailability. Compared to using the combination of alisertib and TG101348, AJI-100 had the advantage of being given once daily by intraperitoneal injection and avoided the need for sustained gavage dosing. Additionally, the single bispecific compound provided a pharmacologically cleaner approach by eliminating the variability in pharmacokinetics between the two drugs in combination. The recipient mice were transplanted with human cell as described. AJI-100 (50 mg/kg) or vehicle control was administered daily by intraperitoneal injection from days 0 to +14. AJI-100 significantly improved the overall survival of the mice and reduced the severity of GVHD, compared to vehicle control (P=0.003; FIGS. 13C-13D). On target inhibition of Aurora A and JAK2 was confirmed among human T cells harvested from recipient spleens at day +14. AJI-100 significantly reduced the amount of pH3Ser10+ and pSTAT3+ T cells, respectively (P=0.027 and P=0.0098, respectively; FIGS. 13E-13F). An established method was used to generate human antitumor CTL in vivo and then test their specific killing. CD8+ CTLs were generated in xenotransplanted mice receiving AJI-100 or vehicle control, where an inoculum of irradiated U937 cells was administered on day 0 and day +7. Unvaccinated, xenotransplanted mice served as negative control. Despite its immunosuppressive activity, AJI-100 did not inhibit CTL generation because CD8+ CTL from AJI-100-treated and vehicle-treated mice demonstrated similarly enhanced killing capacity against U937 targets in vitro, compared to unvaccinated controls (FIG. 13G). These data support that although AJI-100 significantly reduces GVHD, it also preserves antitumor CTL responses.
  • AJI-100 Significantly Increases the Ratio of Treg to Activated Tconv while Eliminating TH17 and TH1 T Cells
  • Similar to its activity in vitro, AJI-100 suppressed the in vivo expansion of human T cells in the xenotransplanted mice. The absolute number of total CD4+ T cells (P<0.0001), Tregs (P=0.001), and activated CD4+ Tconv (P<0.0001) from recipient spleens at day +14 was all significantly reduced by AJI-100 compared to vehicle (FIGS. 14A-E). Activated Tconv were proportionally more reduced by AJI-100 compared to Treg (FIGS. 14B-14C). Therefore, the ratio of Treg to activated Tconv was significantly increased among mice treated with AJI-100 compared to vehicle (P=0.034; FIG. 14D). AJI-100 also significantly reduced the amount of spleen-resident human TH17 and TH1 T cells, compared to vehicle (P=0.002 for both; FIGS. 14F-14H). AJI-100 also exerted a suppressive effect on CD8+ T cell and CD19+ B cell reconstitution as determined by absolute numbers compared to vehicle. However, the frequencies of CD4+ and CD8+ T cells and CD19+ B cells were similar among AJI-100-treated and vehicle-treated mice. The primary host target organs affected by GVHD at day +14 in this xenogeneic model were liver and lung. GVHD severity within these organs was significantly reduced by AJI-100, compared to vehicle (P=0.043 and P=0.002, respectively; FIGS. 14I-14K). Immunohistochemistry demonstrated that the number of tissue-infiltrating, human CD3+ T cells in recipient liver and lung was also significantly decreased by AJI-100 treatment (P=0.006 and P=0.002, respectively; FIGS. 14L-14M).
  • AJI-100 Reduces Xenograft Rejection.
  • Using the nondiagnostic mastectomy skin from consenting donors, a 1×1 cm skin graft was transplanted onto immunodeficient NSG mice dorsally. After 30 days of healing, 5×106 peripheral blood mononuclear cells (PBMC) from an HLA-disparate random donor was given by i.p. injection. A cohort of mice only received a skin graft without PBMCs, as negative rejection controls. The transplanted mice were treated with AJI-100 (50 mg/kg), a dual JAK2/Aurora A kinase inhibitor, or vehicle daily by i.p. injection. Mice were humanely euthanized on day +21 to assess human anti-human skin rejection pathology. Representative H&E staining of the skin grafts show normal cutaneous histology in the no PBMC control (per the Bejarano scoring system, Am J Surg Pathol 28:670-675, 2004); demonstrated by an obvious dermal-epidermal junction, lack of perivascular or dermal infiltrates, and no lymphocytic exocytosis or hyperkeratosis (grade 0, FIG. 15C). Conversely, skin grafts from the mice who received Vehicle+PBMCs show diffuse lymphocytic infiltration of the dermis with exocytosis, a disrupted dermal-epidermal junction, and hyperkeratosis (grade III, FIG. 15B). Mice that received AJI-100+PBMCs exhibited very little graft damage, with mild dermal infiltration, a normal dermal-epidermal junction, and limited hyperkeratosis (grade I, FIG. 15C).
  • Results Summary
  • T cell costimulation and cytokine activation independently contribute to GVHD, but control of donor alloresponses is incomplete when targeting either pathway alone. Here, it is demonstrated that GVHD prevention can be accomplished by dual inhibition of Aurora kinase A and JAK2, attenuating CD28 costimulation and IL-6-mediated signal transduction, respectively, without ablating potential antitumor CTL responses. Concurrent blockade of Aurora kinase A and JAK2 yields synergistic immunosuppression of human allogeneic T cells in vitro, significantly enhances iTreg-suppressive potency, and enhances the ratio of Tregs to activated Tconv in vivo. These characteristics are distinct from CNI-based GVHD prophylaxis, which inhibits TCR function and indiscriminately suppresses donor T cells. The lack of selectivity by CNIs results in a failure to achieve donor immune tolerance toward the host and mitigates the graft-versus-leukemia (GVL) potential of the allograft. Inhibition of Aurora kinase A or JAK2 activity individually suppressed human T cell proliferation in alloMLRs, and synergy was achieved in vitro with simultaneous blockade of both signal transduction pathways. A xenogeneic model was used to study human T cell responses in vivo after Aurora kinase A and JAK2 blockade, understanding that the lack of recipient conditioning does differ from clinical practice in allogeneic HCT. It was shown that alisertib combined with TG101348 significantly delays GVHD. However, the combination of inhibitors appears additive at best in vivo and does not completely eliminate GVHD. The bispecific inhibitor AJI-100 significantly reduced GVHD and improved survival compared to vehicle control. It was surmised that the apparent enhanced in vivo activity of AJI-100 compared to alisertib plus TG101348 may be due to inherent kinase selectivity. The ratio of Aurora kinase A to kinase B inhibition by AJI-100 is greater than alisertib. This could contribute to enhanced impairment of T cell costimulation by AJI-100 and secondarily enhance efficacy. Although the role of AMPK in GVHD is unknown, mouse models of inflammatory colitis using AMPK-deficient T cells suggest that AMPK neutralization may have immunosuppressive properties as well. Therefore, off-target inhibition of AMPK by AJI-100 could be beneficial in controlling GVHD. However, the immunosuppressive effects of the combination of the JAK2 inhibitor TG101348 and the Aurora kinase A inhibitor alisertib coupled with the potent activity of AJI-100 suggests that the ability of AJI-100 to prevent GVHD is likely due to its dual JAK2/Aurora kinase A inhibitory activity. Blockade of Aurora kinase A or JAK2 induces pathway-specific effects on developing iTreg and TH17. First, blockade of Aurora kinase A and JAK2 permits the differentiation of highly suppressive, alloantigen-specific, CD39+ iTreg. Patients with rheumatoid arthritis lacking sufficient CD39+ Tregs experience greater rates of methotrexate failure and poor clinical outcomes, suggesting that dual Aurora kinase A/JAK2 inhibition may benefit other inflammatory conditions. Our data support that the enhanced iTreg potency is largely a function of Aurora kinase A inhibition because iTregs exposed to alisertib, an Aurora kinase A-specific inhibitor, eliminated Tconv proliferation. On the other hand, alisertib was unable to prevent TH17 differentiation among naïve CD4+ T cells responding to alloantigen. Given that IL-6 receptor signal transduction facilitates TH17 development, our data confirm that JAK2 blockade is capable of restraining STAT3 phosphorylation and resultant TH17 differentiation. Additionally, JAK2 inhibition appears to exhibit less inhibition of iTregs compared to Aurora kinase A blockade. Last, inhibition of JAK2, Aurora kinase A, or both JAK2 and Aurora kinase A equally impaired the T H1 response in vitro. Selective inhibition of Aurora kinase A and JAK2 paired with preserved common gamma-chain cytokine signaling establishes a platform to control alloreactivity while permitting antigen-specific Treg and CTL responses. However, there are several limitations of this study that deserve further consideration. Although the xenogeneic model is well suited to test whether concurrent Aurora kinase A/JAK2 inhibition can prevent GVHD mediated by human cells in vivo, it does not entirely replicate human GVHD pathogenesis. The recipient mice do not receive transplant conditioning, unlike human patients, and this may affect GVHD target-organ injury, host antigen presentation, and the production of relevant cytokines such as IL-6. Our work demonstrates that AJI-100 permits the generation and function of antitumor CTL, but it is important to recognize that such experiments are supportive and not definitive in assessing whether the bispecific inhibitor preserves GVL in vivo. Last, small-molecule inhibitors can exhibit off-target inhibition, as observed with AJI-100 and its suppression of AMPK. Unlike molecular knockout strategies, off-target effects by pharmacologic inhibitors may be immunologically relevant and should be considered when interpreting such data. CNI-free GVHD prophylaxis is an important concept in improving patient outcomes after clinical transplantation. The challenges of CNI-based GVHD prevention are clear because CNIs offer incomplete protection from severe GVHD and render the donor immune system poorly equipped to counter posttransplant relapse. Given that targeting Aurora kinase A and JAK2 significantly reduces activated Tconv while permitting Tregs and tumor-specific CTL, the concept described here may represent a translatable CNI free approach at GVHD prevention. A limited number of CNI-free GVHD prophylaxis strategies currently exist and include T cell depletion of the allograft or the use of posttransplant cyclophosphamide. The bispecific inhibitor AJI-100 is an attractive alternative because it does not require ex vivo allograft modification or the need to expose freshly infused donor stem cells to potent alkylators. Hence, further investigation of dual Aurora kinase A/JAK2 inhibition is merited to promote selective control of donor immune responses after alloHCT.
  • It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

Claims (6)

1-18. (canceled)
19. A compound comprising the formula IIIC:
Figure US20220281828A1-20220908-C00040
wherein
R1 is selected from the group consisting of H, Cl, F, Br, I, C1-C6 alkyl, CN, NO2, and NH2;
R2 is selected from the group consisting of H, F, and Cl; and
each R4 is selected, independently, from the group consisting of H, COOH, CONH2, CONR5, SO2NH2, CONSO2R5, tetrazole, 4-morpholine, and COR5;
each R5 is selected, independently, from the group consisting of C1-C6 alkyl, cycloalkyl, heteroaryl, and heteroalkyl; and
m is 1-5,
or a pharmaceutically acceptable salt thereof.
20. The compound of claim 19, wherein m is 1 and R4 is selected from the group consisting of COOH, 2-CONH2, 4-CONH2, SO2NH2, tetrazole, and 4-morpholine.
21. The compound of claim 19, wherein R1 is Cl, F, Br, or I and R2 is H.
22. The compound of claim 19, wherein the compound is:
Figure US20220281828A1-20220908-C00041
23-24. (canceled)
US17/555,786 2016-03-11 2021-12-20 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease Abandoned US20220281828A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US17/555,786 US20220281828A1 (en) 2016-03-11 2021-12-20 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease
US18/098,990 US20230150948A1 (en) 2016-03-11 2023-01-19 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201662307030P 2016-03-11 2016-03-11
PCT/US2017/022074 WO2017156527A1 (en) 2016-03-11 2017-03-13 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease
US201816083681A 2018-09-10 2018-09-10
US17/555,786 US20220281828A1 (en) 2016-03-11 2021-12-20 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2017/022074 Division WO2017156527A1 (en) 2016-03-11 2017-03-13 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease
US16/083,681 Division US11203576B2 (en) 2016-03-11 2017-03-13 Aurora kinase and Janus kinase inhibitors for prevention of graft versus host disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US18/098,990 Continuation US20230150948A1 (en) 2016-03-11 2023-01-19 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease

Publications (1)

Publication Number Publication Date
US20220281828A1 true US20220281828A1 (en) 2022-09-08

Family

ID=59790849

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/083,681 Active US11203576B2 (en) 2016-03-11 2017-03-13 Aurora kinase and Janus kinase inhibitors for prevention of graft versus host disease
US17/555,786 Abandoned US20220281828A1 (en) 2016-03-11 2021-12-20 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease
US18/098,990 Pending US20230150948A1 (en) 2016-03-11 2023-01-19 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/083,681 Active US11203576B2 (en) 2016-03-11 2017-03-13 Aurora kinase and Janus kinase inhibitors for prevention of graft versus host disease

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/098,990 Pending US20230150948A1 (en) 2016-03-11 2023-01-19 Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease

Country Status (2)

Country Link
US (3) US11203576B2 (en)
WO (1) WO2017156527A1 (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7109013B2 (en) 2019-03-19 2022-07-29 ボロノイ インコーポレイテッド Heteroaryl Derivatives, Methods for Producing Heteroaryl Derivatives, and Pharmaceutical Compositions Containing Heteroaryl Derivatives as Active Ingredients
WO2021003417A1 (en) 2019-07-03 2021-01-07 Sumitomo Dainippon Pharma Oncology, Inc. Tyrosine kinase non-receptor 1 (tnk1) inhibitors and uses thereof
WO2023056075A1 (en) * 2021-10-01 2023-04-06 The Regents Of The University Of Colorado, A Body Corporate Compositions and methods for reduced toxicity in transplantation using janus kinase (jak) inhibitors

Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6649608B2 (en) * 2000-03-01 2003-11-18 Astrazeneca Ab 2,4-di(hetero-)arylamino (oxy)-5-substituted pyrimidines as antineoplastic agents
WO2005013996A2 (en) * 2003-08-07 2005-02-17 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and uses as anti-proliferative agents
US20060063789A1 (en) * 2001-11-01 2006-03-23 Freyne Eddy Jean E Aminobenzamide derivatives as glycogen synthase kinase 3 beta inhibitors
US7153964B2 (en) * 2000-03-01 2006-12-26 Astrazeneca Ab Pyrimidine compounds
US7560458B2 (en) * 2003-07-16 2009-07-14 Janssen Pharmaceutica, N.V. Triazolopyrimidine derivatives as glycogen synthase kinase 3 inhibitors
US8044201B2 (en) * 2008-12-03 2011-10-25 The Scripps Research Institute Stem cell cultures
US8211929B2 (en) * 2004-12-30 2012-07-03 Exelixis, Inc. Pyrimidine derivatives as kinase modulators and method of use
WO2012135641A2 (en) * 2011-03-30 2012-10-04 H. Lee Moffitt Cancer Center And Research Institute Aurora kinase inhibitors and methods of making and using thereof

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4559157A (en) 1983-04-21 1985-12-17 Creative Products Resource Associates, Ltd. Cosmetic applicator useful for skin moisturizing
LU84979A1 (en) 1983-08-30 1985-04-24 Oreal COSMETIC OR PHARMACEUTICAL COMPOSITION IN AQUEOUS OR ANHYDROUS FORM WHOSE FATTY PHASE CONTAINS OLIGOMER POLYETHER AND NEW OLIGOMER POLYETHERS
US4820508A (en) 1987-06-23 1989-04-11 Neutrogena Corporation Skin protective composition
US4992478A (en) 1988-04-04 1991-02-12 Warner-Lambert Company Antiinflammatory skin moisturizing composition and method of preparing same
US5167649A (en) 1988-08-22 1992-12-01 Zook Gerald P Drug delivery system for the removal of dermal lesions
US4938949A (en) 1988-09-12 1990-07-03 University Of New York Treatment of damaged bone marrow and dosage units therefor
WO2000032235A1 (en) 1998-11-26 2000-06-08 Pentapharm Ag Transport system conjugate
US7033597B2 (en) 2000-10-13 2006-04-25 Université de Lausanne Intracellular delivery of biological effectors
JP4387669B2 (en) 2000-10-13 2009-12-16 ザイジェン エス.アー. Intracellular delivery of biological effectors with novel transporter peptide sequences
TWI329105B (en) 2002-02-01 2010-08-21 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds and their uses
WO2005016893A2 (en) * 2003-07-30 2005-02-24 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds for use in the treatment or prevention of autoimmune diseases
CA2566531A1 (en) * 2004-05-18 2005-12-15 Rigel Pharmaceuticals, Inc. Cycloalkyl substituted pyrimidinediamine compounds and their uses
WO2008118822A1 (en) * 2007-03-23 2008-10-02 Rigel Pharmaceuticals, Inc. Compositions and methods for inhibition of the jak pathway

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6649608B2 (en) * 2000-03-01 2003-11-18 Astrazeneca Ab 2,4-di(hetero-)arylamino (oxy)-5-substituted pyrimidines as antineoplastic agents
US7153964B2 (en) * 2000-03-01 2006-12-26 Astrazeneca Ab Pyrimidine compounds
US20060063789A1 (en) * 2001-11-01 2006-03-23 Freyne Eddy Jean E Aminobenzamide derivatives as glycogen synthase kinase 3 beta inhibitors
US7560458B2 (en) * 2003-07-16 2009-07-14 Janssen Pharmaceutica, N.V. Triazolopyrimidine derivatives as glycogen synthase kinase 3 inhibitors
WO2005013996A2 (en) * 2003-08-07 2005-02-17 Rigel Pharmaceuticals, Inc. 2,4-pyrimidinediamine compounds and uses as anti-proliferative agents
US8211929B2 (en) * 2004-12-30 2012-07-03 Exelixis, Inc. Pyrimidine derivatives as kinase modulators and method of use
US8044201B2 (en) * 2008-12-03 2011-10-25 The Scripps Research Institute Stem cell cultures
WO2012135641A2 (en) * 2011-03-30 2012-10-04 H. Lee Moffitt Cancer Center And Research Institute Aurora kinase inhibitors and methods of making and using thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Hu et al., Exploring the Scaffold Universe of Kinase Inhibitors, Journal of Medicinal Chemistry, Vol. 58, No. 1, pp. 315-332 (2015) *
Li et al., CAPLUS Abstract 163:427238 (2015). *
Lum et al., 2,5-diaminopyrimidines and 3,5-disubstituted azapurines as inhibitors of glycogen synthase kinase-3 (GSK-3), Bioorganic & Medicinal Chemistry Letters, Vol. 18, No. 12, pp. 3578-3581 (2008). *

Also Published As

Publication number Publication date
WO2017156527A1 (en) 2017-09-14
US11203576B2 (en) 2021-12-21
US20230150948A1 (en) 2023-05-18
US20190127335A1 (en) 2019-05-02

Similar Documents

Publication Publication Date Title
US20220281828A1 (en) Aurora kinase and janus kinase inhibitors for prevention of graft versus host disease
KR101740076B1 (en) Compositions and methods for inhibition of the jak pathway
US11833152B2 (en) JAK1 pathway inhibitors for the treatment of cytokine-related disorders
US20220000867A1 (en) Histone deacetylase 6 inhibition for enhancing t-cell function during anti-tumor response and tumor-peptide vaccination
KR20140081757A (en) Multiple myeloma treatment
MX2010014002A (en) Triazolopyridine jak inhibitor compounds and methods.
EP3468559B1 (en) Methods of use and pharmaceutical combinations of hdac inhibitors with bet inhibitors
JP6453441B2 (en) Protein phosphatase 2A inhibitor for the treatment of myelodysplastic syndrome
JP2008526861A (en) Drug combination therapy and pharmaceutical composition for treating inflammatory disorders
KR20140045411A (en) Use of cxcr4 antagonists
US20230190760A1 (en) Advantageous therapies for disorders mediated by ikaros or aiolos
CN109069461A (en) The treatment method of illness relevant to marrow source property inhibition cell
BR112021006033A2 (en) USE OF AN INHIBITOR OF A TRANSPORTER OF THE ENT FAMILY IN THE TREATMENT OF CANCER AND COMBINATION OF THE SAME WITH AN ADENOSINE RECEPTOR ANTAGONIST
US20090123389A1 (en) Methods for modulating Th17 cell development in the treatment and prevention of cellulite
Nüssler et al. Immunomodulatory agents in the laboratory and clinic
US11872220B2 (en) Methods and compositions for treating B-cell malignancies
JP2023506118A (en) Use of JAK1 inhibitors for the treatment of cutaneous lupus erythematosus and lichen planus (LP)
JP2020506879A (en) Heterocyclic compounds and uses thereof
KR20200091405A (en) IL-8 inhibitors for use in the treatment of some sarcomas
US11952328B2 (en) Selective ship inhibitors for treating disease
US20230285556A1 (en) Methods and compositions for treating cancer
CA3196283A1 (en) Csf1r kinase inhibitor and use thereof
JP2021534216A (en) Combination of natural killer cells with cyclophosphamide compounds for cancer treatment
JP2020176145A (en) Combination treatment for hematological cancers
WO2020041344A1 (en) Methods and compositions for drugs to treat ophthalmic diseases

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: H. LEE MOFFITT CANCER CENTER AND RESEARCH INSTITUTE, INC., FLORIDA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BETTS, BRIAN;SEBTI, SAID M.;LAWRENCE, HARSHANI;AND OTHERS;SIGNING DATES FROM 20190418 TO 20201102;REEL/FRAME:061154/0760

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION