EP4164653A1 - Morphische formen von trilaciclib und verfahren zur herstellung davon - Google Patents

Morphische formen von trilaciclib und verfahren zur herstellung davon

Info

Publication number
EP4164653A1
EP4164653A1 EP21826036.2A EP21826036A EP4164653A1 EP 4164653 A1 EP4164653 A1 EP 4164653A1 EP 21826036 A EP21826036 A EP 21826036A EP 4164653 A1 EP4164653 A1 EP 4164653A1
Authority
EP
European Patent Office
Prior art keywords
compound
pattern
patient
pharmaceutical composition
cancer
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.)
Pending
Application number
EP21826036.2A
Other languages
English (en)
French (fr)
Inventor
Stephen Schneider
Alexander Smith
Hannah S. WHITE
Jay Copeland Strum
Jaroslaw Mazurek
Sara WU
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.)
G1 Therapeutics Inc
Original Assignee
G1 Therapeutics 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
Priority claimed from US17/097,854 external-priority patent/US10988479B1/en
Application filed by G1 Therapeutics Inc filed Critical G1 Therapeutics Inc
Publication of EP4164653A1 publication Critical patent/EP4164653A1/de
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/22Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains four or more hetero rings

Definitions

  • This invention provides advantageous isolated morphic forms of trilaciclib which is 2'- ((5-(4-methylpiperazin-l-yl)pyridin-2-yl)amino)-7',8'-dihydro-6'H-spiro[cyclohexane-l,9'- pyrazino[r,2':l,5]pyrrolo[2,3-d]pyrimidin]-6'-one, for example in the form of a free base or a pharmaceutically acceptable salt thereof, for example a dihydrochloride salt, or a dihydrochloride, dihydrate.
  • Compound 1 dihydrochloride is the active ingredient in COSELATM which was approved for commercial sale by the US Food and Drug Administration on February 12, 2021, for the reduction of chemotherapy- induced myelosuppression when administered prior to platinum/etoposide-containing regimens or topotecan-containing regimens for extensive-stage small cell lung cancer.
  • Compound 1 is referred to as “G1T28” or “trilaciclib.” It transiently arrests normal cells to prevent chemotherapy-induced myelosuppression and may improve anti-tumor efficacy. It can be used, for example, in patients with small cell lung cancer (SCLC) receiving topotecan chemotherapy, as well as in combination with etoposide and carboplatin in SCLC. It is also useful, for example, in combination with carboplatin, etoposide, and atezolizumab (a PD-L1 inhibitor) in small cell lung cancer (SCLC). Based on myelopreservation data in patients with SCLC, trilaciclib was granted Breakthrough Therapy Designation (BTD) from the U.S.
  • BTD Breakthrough Therapy Designation
  • FDA Food and Drug Administration
  • Pattern 1 is a highly crystalline form of Compound 1 as a dihydrochloride. Pattern 1 exhibits superior stability when compared to other morphic forms. For example, in competitive slurry experiments (see Example 10) Pattern 1 is the predominantly formed morphic structure of Compound 1 as a dihydrochloride. Pattern 1 also has advantageous properties in the manufacture of Compound 1 as a dihydrochloride. For example, Pattern 1 can be made on large scale via crystallization of Compound 1 in a heated HC1 solution. Pattern 1 can also be formed from other morphic or amorphic forms of Compound 1 via recrystallization.
  • Pattern 1 can be in the form of a dihydrochloride, dihydrate. Pattern 1 typically, initially, forms a dihydrochloride, dihydrate and even after being dried may eventually revert to a dihydrochloride, dihydrate when exposed to air. Regardless of water content Pattern 1 can maintain its representative XRPD peaks as described in more detail below. For example, if the morphic form of Compound 1 is prepared as described herein to form Pattern 1 and then dried, the representative XRPD peaks will remain the same before and after drying.
  • Compound 1 is the approved drug trilaciclib which is sold under the brand name COSELATM and is used in certain embodiments as a myelopreservation agent to protect healthy cells, notably hematopoietic cells, during chemotherapy. It is intended to be administered by intravenous injection for fast access to the blood stream just prior to administration of chemotherapy, typically also by IV injection.
  • Compound 1 free base is not very soluble in water, and also practically insoluble in DMSO. Further, it becomes less soluble as the pH increases. Blood typically has a pH of 7.35-7.45, which is mildly basic. Phosphate buffered saline typically has a pH of 7.2-7.4. Compound 1 free base is not very soluble at this pH, and thus multiple problems can occur.
  • a significant amount of Compound 1 is required to achieve a therapeutic effect.
  • a typical dose is 240 mg/m 2 and can be estimated to be about a 300 mg dose for a normal adult. Because of the lack of solubility in water, it would take a dilute solution in a lot of fluid to provide this dosage of free base Compound 1, which would have to be given over a long period of time, in contradiction to how it should be delivered.
  • the Compound 1 IV is typically administered over about 30 minutes (20-60 minutes), which means that the drug has to be concentrated, not dilute, in the IV fluid.
  • the free base Compound 1 is injected into blood, there is a real possibility of at least some of it falling out of solution because of the fact that blood is mildly basic. This can cause problems at the site of injection caused by drug deposition and a loss of activity.
  • an IV solution of Compound 1 for injection to cancer patients to preserve healthy cells or for an antineoplastic use can be accomplished by administering it as a dihydrochloride.
  • This enables the intended therapeutic effect because it can be delivered quickly in a concentrated form directly to the blood stream.
  • the IV solution is prepared from a lyophilized powder that comprises Compound 1 dihydrochloride, mannitol, and citric acid.
  • the IV solution of Compound 1 is prepared from a solid pharmaceutical composition which has optionally been formulated into a composition for lyophilization and then dried.
  • this composition includes Compound 1 in the form of a dihydrochloride, for example Compound 1 dihydrochloride Pattern 1.
  • the composition may also include one or more appropriate excipients for example a bulking agent, buffer, and/or one or more pH adjustment agents.
  • the bulking agent is an appropriate sugar such as mannitol.
  • the buffer is an appropriate weak acid or weak base with one or more acidic or basic units.
  • the buffer is a weak acid with two or more acidic units, for example citric acid.
  • the composition includes NaOH, HC1, and or NaCl as pH adjustment agents (NaOH and/or HC1) or the residual salt from pH adjustment (NaCl).
  • a lyophilized Compound 1 dihydrochloride composition which comprises mannitol and citric acid can be prepared that is crystalline and readily characterized by X-ray powder diffraction (XRPD).
  • the lyophilized Compound 1 dihydrochloride composition comprises about 250 to 350 mg, about 275 to 325 mg or more particularly about 300 mg of Compound 1 wherein the 300 mg of weight corresponds to the free base (i.e. about 349 mg of Compound 1 as a dihydrochloride).
  • Compound 1 in the lyophilized composition is a crystalline dihydrochloride in the form characterized by Pattern 1.
  • Compound 1 within the lyophilized composition is in the form of a different pharmaceutically acceptable salt30%, 40% or a mixture of pharmaceutically acceptable salts. In certain embodiments Compound 1 within the lyophilized composition is in the form of a different pattern or a mixture of patterns. In certain embodiments the lyophilized Compound 1 composition comprises at least about 10%, 20%, 30%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% Compound 1 dihydrochloride Pattern 1 relative to other forms of Compound 1 in the composition. In certain embodiments the lyophilized Compound 1 composition comprises at least about 20%, 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75% or 80% Compound 1 dihydrochloride Pattern 1 relative to other components of the lyophilized composition.
  • the lyophilized Compound 1 dihydrochloride composition described in Example 28 contains mannitol and citric acid and is characterized by the XRPD pattern shown in FIG. 100. Other appropriate excipients can be added or replace mannitol and/or citric acid as appropriate to achieve the desired effect.
  • the Compound 1 dihydrochloride composition is lyophilized, in other embodiments it is dried using another technique.
  • the Compound 1 dihydrochloride composition comprises mannitol, for example at least about 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mg of mannitol or about 200 to 400 mg, 225 to 375 mg, 250 to 350 mg, 275 to 325 mg, or 300 mg of mannitol.
  • the Compound 1 dihydrochloride composition comprises citric acid, for example at least about 10, 15, 20, 25,
  • the Compound 1 dihydrochloride composition comprises one or more additional salts formed from the adjustment of the pH of the bulk solution which is dried to form the composition, for example sodium chloride and or various sodium salts of citric acid.
  • the lyophilized Compound 1 dihydrochloride composition includes about 349 mg of Compound 1 Pattern 1 dihydrochloride (equivalent to about 300 mg of trilaciclib free base), about 50-100 mg, about 60-80 mg, or more particularly about 76 mg of citric acid monohydrate, and about 280-320 mg, for example 300 mg of mannitol.
  • the lyophilized Compound 1 composition can be reconstituted in an appropriate solvent for example water, which can include other excipients, for example sodium chloride or dextrose.
  • the lyophilized Compound 1 composition is reconstituted in at least about 10, 11, 12, 13, 14, 15, 16, 17, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mL of at least about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2,
  • the lyophilized Compound 1 composition is reconstituted in about 14 to 25, 15 to 24, 16 to 23, 17 to 22, 17.5 to 21.5, 18 to 21, 18.5 to 20.5,
  • the lyophilized Compound 1 composition is reconstituted in at least about 10, 11, 12, 13, 14, 15, 16, 17, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mL of at least about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5,
  • the lyophilized Compound 1 composition is reconstituted in about 14 to 25, 15 to 24, 16 to 23, 17 to 22, 17.5 to 21.5, 18 to 21, 18.5 to 20.5, 19 to 20, or 19.5 mL of about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10% dextrose solution, for example about 5% dextrose solution.
  • isolated Compound 1 Pattern 1 is used in the manufacture of a lyophilized form that is then formulated with a suitable solvent such as phosphate buffered saline for administration to a patient, for example, by intravenous delivery. In another embodiment, it can be formulated into a parenteral dosage form. This dosage form can be used, for example, in subcutaneous administration.
  • Compound 1 Pattern 1 is a dihydrochloride morphic form.
  • Compound 1 Pattern 1 as a dihydrochloride can be used to treat any condition that is responsive to trilaciclib, including for myelopreservation or as an antineoplastic agent.
  • Compound 1 Pattern l is a dihydrochloride, dihydrate morphic form.
  • Compound 1 Pattern 1 as a dihydrochloride, dihydrate can be used to treat any condition that is responsive to trilaciclib, including for myelopreservation or as an antineoplastic agent.
  • Pattern 2 is also highly crystalline forms of Compound 1 as a dihydrochloride.
  • Pattern 4 is a crystalline solvate of Compound 1 as a dihydrochloride with acetonitrile.
  • Pattern 5 is a crystalline form of Compound 1 as a dihydrochloride.
  • Pattern 6 is a metastable form of Compound 1 as a dihydrochloride.
  • Pattern 11 is a crystalline form of Compound 1 as a dihydrochloride, hemi-hydrate.
  • Pattern 8 and Pattern 10 are crystalline forms of Compound 1 as a free base.
  • Pattern 9 is a crystalline form of Compound 1 as a free base which may be either a solvate or hydrate.
  • Pattern 1 is the most stable morphic form of Compound 1 as a dihydrochloride and formed by competitive slurry experiments with either Pattern 2 or Pattern 3.
  • Compound 1 is typically administered to the patient in an intravenous formulation.
  • the solid forms of trilaciclib can be important because they may be used to achieve desired manufacturing purity and/or performance, and may also be important to solid state shelf life prior to formulation, or in a solid dosage form for therapeutic delivery.
  • Compound 1 (trilaciclib) Given the therapeutic importance of Compound 1 (trilaciclib) to medical therapy for patients suffering from a proliferative disorder such as a tumor or cancer, it is beneficial to provide this advantageous solid form of the compound that contributes to the success or performance of the compound during manufacture, storage, formulation, and/or administration.
  • FIG. 1 is an XRPD Diffractogram of the crystalline Pattern 1.
  • the crystal structure was obtained using the method described in Example 1 and the peaks are shown in Table 1.
  • the x- axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 2 is a PLM Imaging of the crystalline Pattern 1. The image was obtained using the method described in Example 5. Pattern 1 material appeared highly crystalline under PLM imaging with the non-polarized material shown on the left, and the polarized material shown on the right.
  • FIG. 3 is athermogravimetric/differential thermal (TG/DTA) thermogram of crystalline Pattern 1.
  • the TG/DTA thermogram of Pattern 1 was obtained as described in Example 6.
  • the initial mass loss of ca. 6 % from the onset of heating related to the loss of surface moisture.
  • Two mass losses of ca. 7 % were observed from ca. 200°C peaking at 218°C and 300°C peaking at 343°C.
  • FIG. 4 is a Differential Scanning Calorimetry (DSC) thermogram of the crystalline Pattern 1.
  • the DSC thermogram of crystalline Pattern 1 was obtained as described in Example 7.
  • DSC analysis showed 2 broad endotherms ca. 65°C peaking at 130°C and 245°C peaking at 277°C.
  • a sharp melting endotherm was observed from an onset of 330°C peaking at 341°C.
  • FIG. 5. is a Dynamic Vapor Sorption (DVS) analysis showing the results from a moisture sorption experiment of Pattern 1.
  • DVS analysis of Pattern 1 was obtained as described in Example 3. The material was found to be stable and the XRPD analysis of a dried sample at the conclusion of the experiment confirmed Pattern 1.
  • Pattern 1 adsorbed 3.00 wt% at 40% RH (relative humidity) and 4.00 wt% at 90% RH.
  • the x-axis is relative humidity measured as a percent and the y-axis is weight of water of the material measured as a percent.
  • FIG. 6 is a Dynamic Vapor Sorption (DVS) Kinetic Plot of Pattern 1.
  • DVS of Pattern 1 was obtained as described in Example 3. The sample was subjected to a ramping profile from 40 - 90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (dm/dt 0.004%, minimum step length 30 minutes, maximum step length 500 minutes) at 25°C. After completion of the sorption cycle, the sample was dried using the same procedure to 0% RH and then a second sorption cycle back to 40% RH. Two cycles were performed. The material appeared hygroscopic by DVS with a mass increase of 4 % between 0% and 90 % RH. During the desorption cycles the material dehydrates.
  • FIG. 7 is a comparison of XRPD diffractograms of Pattern 1 and Pattern 1 post-DVS.
  • the diffractogram of Pattern 1 was obtained as described in Example 1; and post-DVS of Pattern 1 was obtained as described in Example 11.
  • the material appeared hygroscopic by DVS with a mass increase of 4 % between 0% and 90 % RH. During the desorption cycles the material dehydrates.
  • XRPD analysis post-DVS showed no change in Pattern 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 8 is a comparison of XRPD diffractograms of Pattern 1 before solvent solubility studies and the Pattern 1 material that results from various solvent solubility studies.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 9 is a comparison of XRPD diffractograms of Pattern 1 before solvent solubility studies and the Pattern 1 material that results from various solvent solubility studies.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 10 is a comparison of XRPD diffractograms of Pattern 2 resulting from various solvent solubility studies.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 11 is a comparison of XRPD diffractograms of Pattern 2 resulting from various solvent solubility studies.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 12 is a comparison of XRPD diffractograms of Pattern 3 resulting from various solvent solubility studies.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 13 is a comparison of XRPD diffractograms of Pattern 1 and Pattern 4 which resulted from the solubility study in MeCN.
  • the diffractograms were obtained as described in Example 1 and Example 4 and the studies were conducted as described in Example 13.
  • the x- axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 14 is a comparison of XRPD diffractograms Pattern 2 and Pattern 5 which resulted from various solvent systems.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 15 is a comparison of XRPD diffractograms Pattern 2 and Pattern 6 which resulted from various solvent systems.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 16 is a comparison of XRPD diffractograms of Pattern 1 before and after maturation experiments in various solvents systems.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 17 is a comparison of XRPD diffractograms of Pattern 2 before and after maturation experiments in various solvents systems.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 18 is a comparison of XRPD diffractograms of Pattern 2 before and after maturation experiments in various solvents systems.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 19 is a comparison of XRPD diffractograms of Pattern 3 before and after a maturation experiments in isopropyl alcohol and ethanol.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 20 is a comparison of XRPD diffractograms of Pattern 4 before and after a maturation experiment in acetonitrile.
  • the diffractograms were obtained as described in Example 4 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 21 is a comparison of XRPD diffractograms of Pattern 5 before and after a maturation experiment in methyl isobutyl ketone.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 22 is a comparison of XRPD diffractograms resulting from maturation experiments in various solvent systems.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 23 is a comparison of XRPD Pattern 1 diffractogram and diffractograms resulting from maturation experiments in various solvent systems after drying.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 24 is a comparison of XRPD Pattern 1 diffractogram and diffractograms resulting from maturation experiments in various solvent systems after drying.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 25 is a comparison of XRPD Pattern 2 diffractogram and diffractograms resulting from maturation experiments in various solvent systems after drying.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 26 is a comparison of XRPD Pattern 3 diffractogram and diffractograms resulting from maturation experiments in various solvent systems after drying.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 27 is a comparison of XRPD Pattern 3 diffractogram, Pattern 4 diffractogram and diffractograms resulting from maturation experiments in various solvent systems after drying.
  • the diffractograms were obtained as described in Example 1 and Example 4 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 28 is a comparison of XRPD Pattern 3 diffractogram, Pattern 6 diffractogram and the diffractogram resulting from maturation in heptane after drying.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 29 is a comparison of XRPD Pattern 5 and diffractograms resulting from maturation in various solvents after drying.
  • the diffractograms were obtained as described in Example 1 and the studies were conducted as described in Example 21.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 30 is a comparison of XRPD diffractogram of Pattern 1 and solids that were left after evaporation experiments.
  • the diffractograms were obtained as described in Example 1.
  • the solids were obtained as described in Example 13. Samples were left uncapped and placed in a cupboard to allow evaporation.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 31 is a comparison of XRPD diffractogram of Pattern 3 and solids that were left after evaporation experiments.
  • the diffractograms were obtained as described in Example 1.
  • the solids were obtained as described in Example 13. Samples were left uncapped and placed in a cupboard to allow evaporation.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 32 is a comparison of XRPD diffractogram of Pattern 3 and the solid that resulted from the addition of methyl /er/-butyl ether (t-BME) to 2,2,2-trifluoroethanol.
  • the diffractograms were obtained as described in Example 1.
  • the solids were obtained as described in Example 13.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 33 is a thermogravimetric/differential thermal (TG/DTA) thermogram of Pattern
  • the TG/DTA thermogram of Pattern 2 was obtained as described in Example 6.
  • Two mass losses of ca. 7 % were observed from ca. 222°C peaking at 239°C, 318 °C peaking at 340°C, and 345 °C peaking at 325°C.
  • the x-axis in Temp (°C) and the y- axis is TG (%).
  • FIG. 34 is a thermogravimetric/differential thermal (TG/DTA) thermogram of Pattern
  • the TG/DTA thermogram of Pattern 3 was obtained as described in Example 6.
  • Two mass losses of ca. 6.6 % were observed from ca. 222°C peaking at 229°C, 311 °C peaking at 317°C, and 343°C peaking at 345°C.
  • the x-axis in Temp (°C) and the y-axis is TG (%).
  • FIG. 35 is a thermogravimetric/differential thermal (TG/DTA) thermogram of Pattern 5 small scale material.
  • the TG/DTA thermogram of Pattern 5 was obtained as described in Example 6.
  • Three mass losses of ca. 7.3 % were observed from ca. 206°C peaking at 216°C, 284°C peaking at 321°C and 343 °C peaking at 345°C.
  • the x-axis in Temp (°C) and the y-axis is TG (%).
  • FIG. 36 is a comparison of XRPD patterns for small scale and scale-up materials of Pattern 2 and Pattern 6.
  • Pattern 2 synthesis was scaled it produced a mixture of Pattern 6 and Pattern 2 as shown in the XRPD diffractograms.
  • the diffractograms were obtained as described Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 37 is a PLM image of the mixture of Pattern 2 and Pattern 6 that resulted from the scaleup attempt.
  • the PLM imaging of Pattern 2/6 mixture was obtained as described in Example 5.
  • Pattern 2/6 scale-up material appeared highly crystalline under PLM imaging with the non-polarized material shown on the left, and the polarized material shown on the right.
  • FIG. 38 is a thermogravimetric/differential thermal (TG/DTA) thermogram of the mixture of Pattern 2 and Pattern 6 that resulted from the scaleup attempt.
  • Two mass losses of ca. 6.3 % were observed from ca. 211°C and 310 °C peaking at 321°C.
  • the x-axis in Temp (°C) and the y-axis is TG (%).
  • FIG. 39 is a differential scanning calorimetry (DSC) thermogram of the mixture of Pattern 2 and Pattern 6 that resulted from the scaleup attempt.
  • the DSC thermogram of Pattern 2/6 mixture was obtained as described in Example 7.
  • DSC analysis showed 2 broad endotherms ca. 55.9°C peaking at 131°C and 267 °C peaking at 295°C. A sharp melting endotherm was observed from an onset of 336 °C.
  • FIG. 40 is a gravimetric vapor sorption (GVS) isotherm plot of the mixture of Pattern 2 and Pattern 6 that resulted from the scaleup attempt.
  • the GVS isotherm plot of Pattern 2/6 mixture was obtained as described in Example 2.
  • the mixture of pattern 2 and pattern 6 adsorbed 4.00% wt% at 40% RH (relative humidity) and 7.00% wt% at 90% RH.
  • the x-axis is relative humidity measured as a percent and the y-axis is weight of water of the material measured as a percent.
  • FIG. 41 is a gravimetric vapor sorption (GVS) kinetic plot of the mixture of Pattern 2 and Pattern 6 that resulted from the scaleup attempt.
  • the GVS isotherm plot of Pattern 2/6 mixture was obtained as described in Example 2.
  • the sample was subjected to a ramping profile from 40 - 90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (dm/dt 0.004%, minimum step length 550 minutes, maximum step length 900 minutes) at 25°C.
  • RH relative humidity
  • the sample was dried using the same procedure to 0% RH and then a second sorption cycle back to 40% RH. Two cycles were performed.
  • the material appeared hygroscopic by DVS with a mass increase of 8 % between 0% and 90 % RH. During the desorption cycles the material dehydrates.
  • FIG. 42 is a comparison of XRPD diffractograms of Pattern 1, Pattern 2 scale up and Pattern 2 scale up post-DVS.
  • the diffractograms were obtained as described in Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 43 is a comparison of XRPD diffractograms of Pattern 3 small scale and Pattern 3 scaled in dry and damp form.
  • the diffractogram were obtained as described in Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 44 is a PLM image of the Pattern 3 scale-up material. Pattern 3 scale-up material appeared highly crystalline under PLM imaging as described in Example 5. The non-polarized material shown on the left, and the polarized material shown on the right.
  • FIG. 45 is a TG/DTA thermogram of the Pattern 3 scale up material.
  • the TG/DTA thermogram of Pattern 3 scale-up was obtained as described in Example 6.
  • the TG/DTA shows two peaks at 199°C and 345°C.
  • the initial mass loss of ca. 8 % from the onset of heating related to the loss of surface moisture.
  • Two mass losses of ca. 7 % were observed from ca. 199°C and 343°C peaking at 345°C.
  • the x-axis in Temp (°C) and the y-axis is TG (%).
  • FIG. 46 is a differential scanning calorimetry (DSC) thermogram of the Pattern 3 scale- up material.
  • the DSC thermogram of Pattern 3 was obtained as described in Example 7.
  • DSC analysis showed 3 broad endotherms ca. 168°C peaking at 189°C, 251°C peaking at 264°Cand 272 °C peaking at 296°C.
  • a sharp melting endotherm was observed from an onset of 337 °C peaking at 343 °C.
  • FIG. 47 is a DVS isotherm plot of the Pattern 3 scale-up material.
  • the DVS isotherm plot of Pattern 3 was obtained as described in Example 3.
  • the material appeared hygroscopic by DVS with a mass increase of 4 % between 0% and 90 % RH. During the desorption cycles the material dehydrates.
  • FIG. 48 is a DVS kinetic plot of the Pattern 3 scale-up material.
  • the DVS kinetic plot of Pattern 3 was obtained as described in Example 3.
  • the sample was subjected to a ramping profile from 40 - 90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (change in mass 0.004%, minimum step length 650 minutes, maximum step length 1050 minutes) at 25°C.
  • RH relative humidity
  • the sample was dried using the same procedure to 0% RH and then a second sorption cycle back to 40% RH. Two cycles were performed.
  • the material appeared hygroscopic by DVS with a mass increase of 4 % between 0% and 90 % RH. During the desorption cycles the material dehydrates.
  • FIG. 49 is a comparison of XRPD diffractograms of the Pattern 3 scale-up material before and after DVS.
  • the diffractograms were obtained as described in Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 50 is a comparison of the XRPD diffractograms of Pattern 5 from small scale and scale up experiments.
  • the diffractogram were obtained as described in Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 51 is a comparison of the XRPD diffractograms of Pattern 5 from small scale and scale up experiments after repeating the process.
  • the diffractogram were obtained as described in Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 52 is a comparison of XRPD diffractograms of Pattern 2 in various weeklong stability studies.
  • the diffractograms were obtained as described in Example 1.
  • the stability study compares Pattern 2, and Pattern 2 after 1 week at ambient temperature and at 80°C, respectively.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts. Repeat Scale Up.
  • FIG. 53 is a comparison of XRPD diffractograms of Pattern 2 in various weeklong stability studies.
  • the diffractograms were obtained as described in Example 1.
  • the stability study compares Pattern 2, Pattern 2 scale-up and Pattern 2 at 40°C.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts. Repeat Scale Up.
  • FIG. 54 is a comparison of XRPD diffractograms of Pattern 3 in various weeklong stability studies.
  • the diffractograms were obtained as described in Example 1.
  • the stability study compares Pattern 3, and Pattern 3 after 1 week at ambient temperature, 40°C and at 80°C, respectively.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 55 is a comparison of XRPD diffractograms resulting from competitive slurry experiments of Pattern 1 mixed with Pattern 2 and Pattern 3.
  • the diffractograms were obtained as described in Example 1 after the procedure form Example 11 was pursued.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 56 is a comparison of XRPD diffractograms resulting from competitive slurry experiments of Pattern 1 mixed with Pattern 2 and Pattern 3.
  • the diffractograms were obtained as described in Example 1 after the procedure form Example 11 was pursued.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 57 is a comparison of XRPD diffractograms resulting from competitive slurry experiments of Pattern 1 mixed with Pattern 2 and Pattern 3.
  • the diffractograms were obtained as described in Example 1 after the procedure form Example 11 was pursued.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 58 is a comparison of XRPD diffractograms resulting from competitive slurry experiments of Pattern 1 mixed with Pattern 2 and Pattern 3.
  • the diffractograms were obtained as described in Example 1 after the procedure form Example 11 was pursued.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 59 is a comparison of XRPD diffractograms resulting from competitive slurry experiments of Pattern 1 mixed with Pattern 2 and Pattern 3.
  • the diffractograms were obtained as described in Example 1 after the procedure form Example 11 was pursued.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 60 is a comparison of XRPD diffractograms of Pattern 7 prior to and after thermodynamic solubility experiments.
  • the thermodynamic solubility experiment is described in Example 22.
  • the diffractograms were obtained using the procedure from Example 1.
  • the x- axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 61 is an XRPD diffractogram of Pattern 7.
  • the diffractogram of Pattern 7 was obtained as described in Example 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 62 is a PLM image of Pattern 7 at 200x Magnification.
  • the PLM image of Pattern 7 was obtained as described in Example 5.
  • the birefringent sample under polarized light is shown on the bottom and the Non-Polarized Light of the sample is shown on the top.
  • FIG. 63 is a TG/DTA thermogram of Pattern 7.
  • the TG/DTA thermogram of Pattern 7 was obtained as described in Example 6.
  • the thermogram shows a 6.2 % weight loss between 150 and 340 °C and sample decomposition above 350 °C.
  • the thermogram also shows that the sample melted noted with onset at 340 °C and a peak at 345 °C.
  • the x-axis is Temp measured in degrees Celsius and the y-axis is intensity measured in DTAuV on the left and TG % on the right.
  • FIG. 64 is a DSC first heat thermogram of Pattern 7.
  • the DSC thermogram of Pattern 7 was obtained as described in Example 7.
  • the thermogram shows the sample melting event at 335 °C and a peak at 338 °C (consistent with TG/DTA data) during first heat.
  • the x-axis is Temp measured in degrees Celsius and the y-axis is intensity measured in DSC mW.
  • FIG. 65 is a DSC cool thermogram of Pattern 7.
  • the DSC thermogram of Pattern 7 was obtained as described in Example 7.
  • the thermogram shows no significant thermal events in the cool cycle.
  • the x-axis is Temp measured in degrees Celsius and the y-axis is intensity measured in DSC mW.
  • FIG. 66 is a DSC second heat thermogram of the crystalline Pattern 7.
  • the DSC thermogram of Pattern 7 was obtained as described in Example 7.
  • the second heat contained the melting event with onset 333 °C and a peak at 339 °C. Possible incomplete melting during the initial heat cycle.
  • the x-axis is Temp measured in degrees Celsius and the y-axis is intensity measured in DSC mW.
  • FIG. 67 is a comparison of XRPD diffractograms from pH solubility studies of Pattern 7 at various pHs.
  • the material resulting from pH 4 was identified as Pattern 8.
  • the diffractograms were obtained as described in Example 1 and the pH experiment was as described in Example 16.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 68 is a comparison of XRPD diffractograms from pH solubility studies of Pattern 7 at various pHs.
  • the diffractogram of Pattern 7 was obtained as described in Example 1 and Example 16.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 69 is a comparison of XRPD diffractograms from pH solubility studies of Pattern 7 at various pH solvent systems.
  • the diffractogram of Pattern 7 was obtained as described in Example 1 and Example 16.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 70 is a comparison of XRPD diffractograms that resulted from crystallization set 1.
  • the crystallizations were conducted as described in Example 17.
  • the comparison shows the Pattern 7 and forms resulting from various concentrations in various pH solvent systems.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 71 is a comparison of XRPD diffractograms that resulted from crystallization sets 2 and 3. The crystallizations were conducted as described in Example 18 and Example 19. The comparison shows the Pattern 7 and forms resulting from various concentrations in various pH solvent systems. The x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 72 is a comparison of XRPD diffractograms that resulted from crystallization sets 2 and 3. The crystallizations were conducted as described in Example 18 and Example 19. The comparison shows the Pattern 7 and forms resulting from various concentrations in various pH solvent systems. The x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 73 is a comparison of XRPD diffractograms that resulted from crystallization set 4. The crystallizations were conducted as described in Example 20. The comparison shows the Pattern 7 and forms resulting from various concentrations in various pH solvent systems. The x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 74 is a PLM image corresponding to the material obtained from experiment 1 from Example 23. The polarized sample is shown in the bottom row and the non-polarized light sample is shown in the top row.
  • FIG. 75 is a PLM image corresponding to the material obtained from experiment 2 from Example 23.
  • the PLM image was obtained as described in Example 5.
  • the polarized sample is shown on the right and the non-polarized light sample is shown on the left.
  • FIG. 76 is a PLM image corresponding to the material obtained from experiment 3 from Example 23.
  • the PLM image was obtained as described in Example 5.
  • the polarized sample is shown on the right and the non-polarized light sample is shown on the left.
  • FIG. 77 is a PLM image corresponding to the material obtained from experiment 4 from Example 23.
  • the PLM image was obtained as described in Example 5.
  • the polarized sample is shown in the bottom row and the non-polarized light sample is shown in the top row.
  • FIG. 78 is a PLM image corresponding to the material obtained from experiment 5 from Example 23.
  • the PLM image was obtained as described in Example 5.
  • the polarized sample is shown in the bottom row and the Non-Polarized Light sample is shown in the top row.
  • FIG. 79 is a PLM image corresponding to the material obtained from experiment 7 from Example 23.
  • the PLM image was obtained as described in Example 5.
  • the polarized sample is shown on the right and the non-polarized Light sample is shown on the left.
  • FIG. 80 is a PLM image corresponding to the material obtained from experiment 6 from Example 23.
  • the PLM image was obtained as described in Example 5.
  • the polarized sample is shown in the bottom row and the Non-Polarized Light sample is shown in the top row.
  • FIG. 81 is a PLM image corresponding to the material obtained from experiment 8 from Example 23.
  • the polarized sample is shown on the right and the non-polarized Light sample is shown on the left.
  • FIG. 82 is a PLM image corresponding to the material obtained from experiment 9 from Example 23. The PLM image was obtained as described in Example 5.
  • the polarized sample is shown on the right and the non-polarized Light sample is shown on the left.
  • FIG. 83 is a PLM image corresponding to the material obtained from experiment 10 from Example 23.
  • the polarized sample is shown in the bottom row and the Non-Polarized Light sample is shown in the top row.
  • FIG. 84 is a PLM image corresponding to the material obtained from experiment 11 from Example 23. The polarized sample is shown on the right and the non-polarized Light sample is shown on the left.
  • FIG. 85 is a PLM image corresponding to the material obtained from experiment 12 from Example 23. This material was identified as Pattern 9. The polarized sample is shown in the bottom row and the Non-Polarized Light sample is shown in the top row.
  • FIG. 86 is a PLM Images of the crystalline Pattern 10 at 200x Magnification.
  • the PLM imaging of crystalline Pattern 7 was obtained as described in Example 5.
  • the polarized sample is shown in the bottom row and the Non-Polarized Light sample is shown in the top row.
  • FIG. 87 is a TG/DTA thermogram of Pattern 9.
  • the TG/DTA thermogram of Pattern 9 was obtained as described in Example 6.
  • Pattern 9 exhibited a large weight loss of 24.3 % from the onset of heating up to 150°C.
  • Pattern 9 exhibited a second mass loss of 15 wt.% between 75 °C and 154 °C.
  • the DT trace identified two endothermic events associated with the mass losses at the first shown onset at 37°C peaking at 66.2°C; the second onset at 122°C peaking at 128°C; and the third and final endothermic event in the DT trace (sample melt) was observed with an onset of 326 °C and a peak at 330 °C.
  • the x-axis is Temp measured in degrees Celsius and the y-axis is intensity measured in DTAuV on the left and TG % on the right.
  • FIG. 88 is a TG/DTA thermogram of Pattern 10.
  • the TG/DTA thermogram was obtained as described in Example 6.
  • the thermogram shows showed minimal weight loss of 0.1 % weight loss between 150 and 340 °C from onset of heating.
  • the sample showed degradation above 320 °C.
  • the thermogram also shows two peaks, with the first onset at 199 and peaking at 206; and the second onset at 327 °C peaking at 331 °C.
  • the x-axis is Temp measured in degrees Celsius and the y-axis is intensity measured in DTAuV on the left and TG % on the right.
  • FIG. 89 is an XRPD Diffractogram of the crystalline Pattern 8.
  • the crystal structure was obtained using the method described in Example 1 and the peaks are shown in Table 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 90 is the crystal structure of Pattern 11.
  • the crystal structure was solved as described in Example 25. Non-hydrogen atoms are shown with thermal ellipsoids set at 50 % probability level.
  • FIG. 91 is the crystal structure of Pattern 11 with hydrogen bonding depicted.
  • the crystal structure was solved as described in Example 25. Non-hydrogen atoms are shown with thermal ellipsoids set at 50 % probability level.
  • FIG. 92 is the crystal structure of Pattern 11 with the view set to the unit cell a axis.
  • the crystal structure was solved as described in Example 25. Non-hydrogen atoms are shown with thermal ellipsoids set at 50 % probability level.
  • FIG. 93 is the crystal structure of Pattern 11 with the view set to the unit cell b axis.
  • the crystal structure was solved as described in Example 25. Non-hydrogen atoms are shown with thermal ellipsoids set at 50 % probability level.
  • FIG. 94 is the crystal structure of Pattern 11 with the view set to the unit cell c axis.
  • the crystal structure was solved as described in Example 25. Non-hydrogen atoms are shown with thermal ellipsoids set at 50 % probability level.
  • FIG. 95 is the crystal structure of Pattern 11 with the view set to the void view of the a axis.
  • the crystal structure was solved as described in Example 25. Non -hydrogen atoms are shown with thermal ellipsoids set at 50 % probability level.
  • FIG. 96 is a comparison of the simulated XRPD pattern of Pattern 11 and the XRPD pattern of Pattern 1.
  • the x-axis is 2Theta measured in degrees and the y-axis is intensity measured in counts.
  • FIG. 97 is the crystal structure of Pattern 1 resulting from the x-ray crystallography discussed in Example 27.
  • FIG. 98 is the crystal structure of Pattern 1 resulting from the x-ray crystallography discussed in Example 27 where the hydrogen bonds are shown explicitly.
  • FIG. 99 is an overlay of the experimentally determined XRPD diffractogram of Pattern 1 with the simulated diffractogram that corresponds to the crystal structure shown in FIG. 97. The differences between these two lines is shown on the bottom line. As the bottom line is nearly flat the simulation strongly suggests that the crystal structure corresponds to the XRPD pattern.
  • FIG. 100 is a XRPD diffractogram of a lyophilized Compound 1 dihydrochloride composition which comprises mannitol and citric acid. The crystallization was conducted as described in Example 28 and Table 42. The x-axis is 2Theta measured in degrees and the y- axis is intensity measured in counts.
  • the drug ritonavir is active in one morphic form and inactive in another form, and the inactive form is the more stable.
  • Compound 1 is the approved drug trilaciclib which is sold under the brand name COSELATM and is used in certain embodiments as a myelopreservation agent to protect healthy cells, notably hematopoietic cells, during chemotherapy. It is intended to be administered by intravenous injection for fast access to the blood stream just prior to administration of chemotherapy, typically also by IV injection.
  • Compound 1 free base is not very soluble in water, and also practically insoluble in DMSO. Further, it becomes less soluble as the pH increases. Blood typically has a pH of 7.35-7.45, which is mildly basic. Phosphate buffered saline typically has a pH of 7.2-7.4. Compound 1 is not very soluble at this pH, and thus multiple problems can occur.
  • a significant amount of Compound 1 is required to achieve a therapeutic effect.
  • a typical dose is 240 mg/m 2 and can be estimated to be about a 300 mg dose for a normal adult. Because of the lack of solubility in water, it would take a dilute solution in a lot of fluid to provide this dosage of free base Compound 1, which would have to be given over a long period of time, in contradiction to how it should be delivered.
  • the Compound 1 IV is typically administered over about 30 minutes (20-60 minutes), which means that the drug has to be concentrated, not dilute, in the IV fluid.
  • the free base Compound 1 is injected into blood, there is a real possibility of at least some of it falling out of solution because of the fact that blood is mildly basic. This can cause problems at the site of injection caused by drug deposition and a loss of activity.
  • an IV solution of Compound 1 for injection to cancer patients to preserve healthy cells or for an antineoplastic can be accomplished by administering it as a dihydrochloride or a dihydrochloride, dihydrate.
  • the IV solution is prepared from a lyophilized powder that comprises Compound 1 dihydrochloride, mannitol, and citric acid.
  • the IV solution of Compound 1 is prepared from a solid pharmaceutical composition which has optionally been dried by lyophilization.
  • this composition includes Compound 1 in the form of a dihydrochloride, for example Compound 1 dihydrochloride Pattern 1.
  • the composition may also include appropriate excipients for example a bulking agent, buffer, and/or one or more pH adjustment agents.
  • the bulking agent is an appropriate sugar such as mannitol.
  • the buffer is an appropriate weak acid or weak base with one or more acidic or basic sites.
  • the buffer is a weak acid with two or more acidic sites, for example citric acid.
  • the composition includes NaOH, HC1, and or NaCl as pH adjustment agents (NaOH and/or HC1) or the residual salt from pH adjustment (NaCl).
  • a lyophilized Compound 1 dihydrochloride composition which comprises mannitol and citric acid is crystalline and readily characterized by X-ray powder diffraction (XRPD).
  • the lyophilized Compound 1 dihydrochloride composition comprises about 250 to 350 mg, about 275 to 325 mg or more particularly about 300 mg of Compound 1 wherein the 300 mg of weight corresponds to the free base.
  • Compound 1 in the lyophilized composition is a crystalline dihydrochloride in the form characterized by Pattern 1 herein.
  • Compound 1 within the lyophilized composition is in the form of a different pharmaceutically acceptable salt or a mixture of pharmaceutically acceptable salts.
  • Compound 1 within the lyophilized composition is in the form of a different pattern or a mixture of patterns.
  • the lyophilized Compound 1 composition comprises at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% Compound 1 dihydrochloride Pattern 1 relative to other forms of Compound 1.
  • the lyophilized Compound 1 composition comprises at least about 20%, 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75% or 80% Compound 1 dihydrochloride Pattern 1 relative to other components of the lyophilized composition.
  • the lyophilized Compound 1 dihydrochloride composition described in Example 28 contains mannitol and citric acid and is characterized by the XRPD pattern shown in FIG. 100. Other appropriate excipients can be added or replace mannitol and/or citric acid as appropriate to achieve the desired effect.
  • the Compound 1 dihydrochloride composition is lyophilized. In other embodiments it is dried using another technique.
  • the Compound 1 dihydrochloride composition comprises mannitol, for example at least about 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mg of mannitol or about 200 to 400 mg, 225 to 375 mg, 250 to 350 mg, 275 to 325 mg, or 300 mg of mannitol, or at least about 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mg of mannitol, or at most about 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mg of
  • the Compound 1 dihydrochloride composition comprises one or more additional salts formed from the adjustment of the pH of the bulk solution which is dried
  • the lyophilized Compound 1 dihydrochloride composition includes about 349 mg of Compound 1 Pattern 1 dihydrochloride (equivalent to about 300 mg of trilaciclib free base), about 50-100 mg, about 60-80 mg, or more particularly about 76 mg of citric acid monohydrate, and about 280-320 mg, for example 300 mg, of mannitol.
  • the lyophilized Compound 1 composition can be reconstituted in an appropriate solvent for example water, which can include other excipients, for example sodium chloride or dextrose.
  • the lyophilized Compound 1 composition is reconstituted in at least about 10, 11, 12, 13, 14, 15, 16, 17, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mL of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2% sodium chloride solution, for example about 0.9% sodium chloride solution.
  • the lyophilized Compound 1 composition is reconstituted in at least about 14 to 25, 15 to 24, 16 to 23, 17 to 22, 17.5 to 21.5, 18 to 21, 18.5 to 20.5, 19 to 20, or 19.5 mL of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2% sodium chloride solution, for example about 0.9% sodium chloride solution.
  • the lyophilized Compound 1 composition is reconstituted in at least about 10, 11, 12, 13, 14, 15, 16, 17, 18.5, 19, 19.5, 20, 20.5, 21, 21.5, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mL of at least about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10% dextrose solution, for example about 5% dextrose solution.
  • the lyophilized Compound 1 composition is reconstituted in about 14 to 25, 15 to 24, 16 to 23, 17 to 22, 17.5 to 21.5, 18 to 21, 18.5 to 20.5, 19 to 20, or 19.5 mL of about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10% dextrose solution, for example about 5% dextrose solution.
  • Pattern 1, Pattern 2, and Pattern 3 are highly crystalline forms of Compound 1 as a dihydrochloride.
  • Pattern 1 is a useful dihydrochloride, dihydrate form of Compound 1.
  • Pattern 4 is a crystalline solvate of Compound 1 as a dihydrochloride with acetonitrile.
  • Pattern 5 is a crystalline form of Compound 1 as a dihydrochloride.
  • Pattern 6 is a metastable form of Compound 1 as a dihydrochloride.
  • Pattern 7 is a highly crystalline form of Compound 1 as a free base.
  • Pattern 8 and Pattern 10 are crystalline forms of Compound 1 as a free base.
  • Pattern 9 is a crystalline form of Compound 1 as a free base which may be either a solvate or hydrate.
  • Pattern 11 is a crystalline form of Compound 1 as a dihydrochloride, hemi-hydrate.
  • Pattern 1 dihydrochloride, dihydrate is the most stable morphic form of Compound 1 as a dihydrochloride and can be formed by competitive slurry experiments with either Pattern 2 or Pattern 3.
  • Pattern 1 regardless of its water content is the most stable morphic form of Compound 1 as a dihydrochloride and can be formed by competitive slurry experiments with either Pattern 2 or Pattern 3.
  • Pattern 7 is the most stable morphic form of Compound 1 as a free base.
  • Pattern 1 dihydrochloride, dihydrate has multiple therapeutic advantages over the other morphic forms of Compound 1. For example, Pattern 1 dihydrochloride, dihydrate is more easily used in an intravenous solution. Additionally, higher concentrations of Pattern 1 dihydrochloride, dihydrate can be achieved in water and/or DMSO.
  • Pattern 1 dihydrochloride, dihydrate has multiple manufacturing advantages over the other morphic forms of Compound 1.
  • Pattern 1 dihydrochloride, dihydrate typically has increased shelf stability, thermodynamic stability, and/or solubility in water than other morphic forms of Compound 1.
  • the manufacture of Pattern 1 dihydrochloride, dihydrate is reproducibly more scalable than the manufacture of other morphic forms of Compound 1.
  • isolated Pattern 1 is used in the manufacture of a lyophilized form that is then formulated with a suitable solvent such as phosphate buffered saline for administration to a patient, for example, by intravenous delivery.
  • a suitable solvent such as phosphate buffered saline
  • it can be formulated into a parenteral dosage form.
  • This dosage form can be used, for example, in subcutaneous administration.
  • isolated Pattern 2 is used in the manufacture of a lyophilized form that is then formulated with a suitable solvent such as phosphate buffered saline for administration to a patient, for example, by intravenous delivery.
  • a suitable solvent such as phosphate buffered saline
  • it can be formulated into a parenteral dosage form. This dosage form can be used, for example, in subcutaneous administration.
  • isolated Pattern 3 is used in the manufacture of a lyophilized form that is then formulated with a suitable solvent such as phosphate buffered saline for administration to a patient, for example, by intravenous delivery.
  • a suitable solvent such as phosphate buffered saline
  • it can be formulated into a parenteral dosage form. This dosage form can be used, for example, in subcutaneous administration.
  • isolated Pattern 7 is used in the manufacture of a lyophilized form that is then formulated with a suitable solvent such as phosphate buffered saline for administration to a patient, for example, by intravenous delivery.
  • a suitable solvent such as phosphate buffered saline
  • it can be formulated into a parenteral dosage form. This dosage form can be used, for example, in subcutaneous administration.
  • Pattern 1 An isolated morphic form of Compound 1 as a dihydrochloride that has been designated Pattern 1 is provided in this invention.
  • Pattern 1 can be in the form of a dihydrochloride, dihydrate. Pattern 1 is typically, initially, in the form of a dihydrochloride, dihydrate after being formed and even after being dried may eventually revert to a dihydrochloride, dihydrate when exposed to air. Regardless of water content Pattern 1 can maintain its representative XRPD peaks as described in more detail below. For example, if the morphic form of Compound 1 is prepared as described herein to form Pattern 1 and then dried, the representative XRPD peaks will remain the same before and after drying.
  • Pattern 1 can be dissolved in a composition for lyophilization and after drying, reforms into Pattern 1.
  • Compound 1 Pattern l is a dihydrochloride, dihydrate morphic form. In certain embodiments, the invention provides the composition of Compound 1 as a dihydrochloride, dihydrate. In certain embodiments, the invention provides the composition of Compound 1 Pattern 1 as a dihydrochloride salt dihydrate.
  • Compound 1 Pattern 1 dihydrochloride, dihydrate is more stable than other hydrate forms of Compound 1. In another embodiment, Compound 1 Pattern 1 dihydrochloride, dihydrate may be more stable than non-hydrate forms of Compound 1.
  • Compound 1 Pattern 1 as a dihydrochloride, dihydrate can be advantageous to the anhydrous, monohydrate, or even trihydrate versions of Compound 1 Pattern 1.
  • the dihydrate version of Compound 1 Pattern 1 dihydrochloride has increased shelf stability, thermodynamic stability, and/or solubility in water than the anhydrous, monohydrate, or even trihydrate versions of Compound 1 Pattern 1.
  • the manufacture of Pattern 1 dihydrochloride, dihydrate is cheaper, faster, and/or more scalable than the manufacture of the anhydrous, monohydrate, or even trihydrate versions Compound 1 Pattern 1.
  • Compound 1 Pattern 1 is characterized by an XRPD pattern that is substantially similar to that set forth in FIG. 1.
  • Compound 1 Pattern 1 optionally as a dihydrate is characterized by an XRPD pattern comprising at least three 2theta values selected from about 9.6 ⁇ 0.2°, about 21.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 24.0 ⁇ 0.2°, about 26.1 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • Compound 1 Pattern 1, optionally as a dihydrate is characterized by an XRPD pattern comprising a peak with a 2theta value of about 9.6 ⁇ 0.2°.
  • Compound 1 Pattern 1 is characterized as having a 6% ⁇ 3% weight loss on the onset of heating the sample. In certain embodiments, Compound 1 Pattern 1, optionally as a dihydrate, is characterized as having a 7% ⁇ 3% weight loss between about 200°C and about 230°C. In certain embodiments, Compound 1 Pattern 1 is characterized as having a second 7% ⁇ 3% weight loss between about 300°C and about 380°C.
  • Compound 1 Pattern 1 can be prepared using selective crystallization.
  • the method can be carried out by treating a solution comprising a suitable solvent(s) and Compound 1 as a dihydrochloride optionally in the presence of one or more seeds comprising Compound 1 Pattern 1 to conditions that provide for the crystallization of Compound 1 Pattern 1. If the solvent used is water or contains water then the resulting Compound 1 Pattern 1 is typically a dihydrochloride, dihydrate.
  • the selective crystallization can be carried out in any suitable solvent. For example, it can be carried out in an aprotic solvent or a mixture thereof. Examples of aprotic solvents that can be used include tetrahydrofuran, dichloromethane, nitromethane, and dioxane.
  • Compound 1 Pattern 1 is prepared from a protic solvent.
  • protic solvents examples include water, methanol, and ethanol.
  • Compound 1 Pattern 1 is prepared from a mixture of an aprotic solvent and a protic solvent, such as for example a mixture of acetonitrile and water.
  • the selective crystallization can be carried out at, for example, a temperature in the range of about 20°C to about 65°C. In another embodiment the selective crystallization can be carried out at, for example, a temperature in the range of about 35°C to about 45°C or at about 40°C.
  • Compound 1 Pattern 1 is produced by crystallization or recrystallization in an acidic solution.
  • Compound 1 Pattern 1 can be produced in an aqueous hydrochloric acid solution.
  • Compound 1 Pattern 1 is produced by heating Compound 1 to about 80°C and agitating for at least 30 minutes. The resulting solution is filtered into a reactor pre heated to about 80°C, and the reaction mixture is cooled to about 70°C. Preheated (about 70°C) purified water is added and the reaction mixture is cooled to below 20°C with agitation. Compound 1 Pattern 1 can be isolated by filtration, washed with purified water and acetone, and dried under vacuum at an elevated temperature.
  • Compound 1 Pattern 1 is sieved using a mill.
  • the sieved Compound 1 Pattern 1 material is formulated for parenteral administration to a patient.
  • Compound 1 Pattern 1 is characterized by an XRPD pattern comprising at least 2 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 23.9 ⁇ 0.2° and about 26.1 ⁇ 0.2°.
  • Compound 1 Pattern 1 is characterized by an XRPD pattern comprising at least 3 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 23.9 ⁇ 0.2° and about 26.1 ⁇ 0.2°.
  • Compound 1 Pattern 1 is characterized by an XRPD pattern comprising at least 4 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 23.9 ⁇ 0.2° and about 26.1 ⁇ 0.2°.
  • Compound 1 Pattern 1 is characterized by an XRPD pattern comprising at least 5 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 23.9 ⁇ 0.2° and about 26.1 ⁇ 0.2°.
  • Compound 1 Pattern 1, optionally as a dihydrate is characterized by an XRPD pattern comprising the 2theta values selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 23.9 ⁇ 0.2° and about 26.1 ⁇ 0.2°.
  • Compound 1 Pattern 1 is characterized by an XRPD pattern comprising all or at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20° 2theta values within ⁇ 0.4° 2theta of the peaks selected from: a. 9.6, 21.3, 19.8, 12.2, 24.0, 26.1, 19.3, 17.6, 28.6, 15.3, 23.2, 20.3, 22.7, 27.1, 21.0, 17.0, 10.1, 16.0, 26.8, 31.4, 13.8, 32.3, 32.9, 20.7, 18.7, and 27.6 °20; or b.
  • Compound 1 Pattern 1, optionally as a dihydrate is characterized by an XRPD pattern described above and is further characterized by having no peaks greater than 15% relative intensity in between 4 and 9° 20. In one embodiment Compound 1 Pattern 1, optionally as a dihydrate, is characterized by an XRPD pattern described above and is further characterized by having no peaks greater than 10% relative intensity in between 4 and 9° 20. In one embodiment Compound 1 Pattern 1, optionally as a dihydrate, is characterized by an XRPD pattern described above and is further characterized by having no peaks greater than 5% relative intensity in between 4 and 9 °20. In one embodiment, Compound 1 Pattern 1, optionally as a dihydrate, is characterized by an XRPD pattern described above and is further characterized by having no peaks greater than 3% relative intensity in between 4 and 9 °20.
  • Compound 1 Pattern 1 is used in the preparation of amorphous Compound 1 as a dihydrochloride.
  • the amorphous Compound 1 as a dihydrochloride can be formed by lyophilization or spray drying of Compound 1 Pattern 1, optionally as a dihydrate.
  • This amorphous dihydrochloride may possess higher purity than if it were directly prepared by known synthetic methods.
  • an advantage of first preparing Compound 1 Pattern 1, optionally as a dihydrate, and then converting it to an amorphic material may be that the total shelf life of the product is increased as a result.
  • Compound 1 Pattern 1 is used in the preparation of amorphous Compound 1 as a mono-HCl salt.
  • the amorphous Compound 1 as a mono-HCl salt can be formed by dissolving Compound 1 Pattern 1 and modifying the pH of the solution followed by removal of solvent.
  • This amorphous mono-HCl salt may possess higher purity than if it was directly prepared by known synthetic methods.
  • an advantage of first preparing Compound 1 Pattern 1, optionally as a dihydrate, and then converting it to an amorphic material may be that the total shelf life of the product is increased.
  • Compound 1 Pattern 1 is used in the preparation of amorphous Compound 1 as a free base.
  • This amorphous free base may possess higher purity than if it was directly prepared by known synthetic methods.
  • an advantage of first preparing Compound 1 Pattern 1, optionally as a dihydrate, and then converting it to an amorphic material may be that the total shelf life of the product is increased as a result.
  • Compound 1 Pattern 1 is used in the preparation of a liquid solution suitable for intravenous administration of Compound 1.
  • This liquid solution of Compound 1 may possess higher purity than if it was directly prepared by known synthetic methods.
  • an advantage of first preparing Compound 1 Pattern 1, optionally as a dihydrate, and then converting it to a liquid solution may be that the total shelf life of the product is increased as a result.
  • Compound 1 Pattern 1, optionally as a dihydrate is lyophilized.
  • Compound 1 Pattern 1, optionally as a dihydrate can optionally be mixed with one or more appropriate excipients before or after lyophilization.
  • the invention provides a solid lyophilized composition comprising Compound 1 Pattern 1, optionally as a dihydrate.
  • the invention provides solid lyophilized composition comprising Compound 1 Pattern 1, optionally as a dihydrate, mannitol, and citric acid.
  • the invention provides solid lyophilized composition comprising Compound 1 Pattern 1 dihydrochloride, dihydrate, mannitol, and citric acid.
  • a crystalline compound is provided of structure: which is a dihydrochloride, optionally as a dihydrate.
  • the crystalline compound of embodiment 1 characterized by an X-ray powder diffraction (XRPD) pattern comprising at least three 2theta values selected from 9.6 ⁇ 0.2°, 21.3 ⁇ 0.2°, 19.8 ⁇ 0.2°, 12.2 ⁇ 0.2°, 24.0 ⁇ 0.2°, 26.1 ⁇ 0.2°, 19.3 ⁇ 0.2°, 17.6 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • XRPD X-ray powder diffraction
  • a lyophilized powder prepared from the crystalline compound of embodiment 1 is provided.
  • a pharmaceutical composition comprising the crystalline compound of embodiment 1 and a pharmaceutically acceptable excipient is provided.
  • composition of embodiment 15 comprising about 200 milligrams to about 600 milligrams of the crystalline compound of embodiment 1.
  • composition of embodiment 15 comprising about 300 milligrams of the crystalline compound of embodiment 1.
  • the pharmaceutical composition of embodiment 15 comprising a dose of about 150 mg/m 2 to about 350 mg/m 2 of the crystalline compound of embodiment 1. 0.
  • the pharmaceutical composition of embodiment 15 further comprising about 300 mg of mannitol and about 76 mg of citric acid.
  • the pharmaceutical composition of embodiment 15 comprising a dose of about 240 mg/m 2 of the crystalline compound of embodiment 1.
  • a pharmaceutically acceptable reconstituted solution of the lyophilized powder of embodiment 8 is provided.
  • the pharmaceutical solution of embodiment 23 comprising a dose of about 150 mg/m 2 to about 350 mg/m 2 of the lyophilized powder of embodiment 8.
  • composition of embodiment 23 further comprising about 300 mg of mannitol and about 76 mg of citric acid.
  • composition of embodiment 23 further comprising sodium hydroxide or hydrochloric acid.
  • a lyophilized powder comprising a compound of structure: which is a dihydrochloride, which optionally includes hydrate.
  • a crystalline compound of structure which is a dihydrochloride is provided.
  • the crystalline compound of embodiment 1 characterized by an X-ray powder diffraction (XRPD) pattern comprising at least three 2theta values selected from 9.6 ⁇ 0.2°, 21.3 ⁇ 0.2°, 19.8 ⁇ 0.2°, 12.2 ⁇ 0.2°, 24.0 ⁇ 0.2°, 26.1 ⁇ 0.2°, 19.3 ⁇ 0.2°, 17.6 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • XRPD X-ray powder diffraction
  • composition comprising the crystalline compound of any one of embodiments 1-7, mannitol and citric acid.
  • a pharmaceutical composition comprising the crystalline compound of embodiments 1-7 and a pharmaceutically acceptable excipient.
  • composition of embodiment 9, wherein the pharmaceutically acceptable excipient comprises mannitol.
  • composition of embodiment 9, wherein the pharmaceutically acceptable excipient comprises citric acid.
  • composition of embodiment 9, wherein the pharmaceutically acceptable excipient comprises mannitol and citric acid.
  • composition of embodiment 9 comprising about 200-600 milligrams of the crystalline compound.
  • composition of embodiment 9 comprising about 300 milligrams of the crystalline compound.
  • composition of embodiment 9 comprising about 349 milligrams of the crystalline compound.
  • composition of embodiment 10 comprising about 250-350 mg of mannitol.
  • the pharmaceutical composition of embodiment 10 comprising about 300 mg of mannitol. 18.
  • the pharmaceutical composition of embodiment 11 comprising about 50-100 mg of citric acid.
  • composition of embodiment 11 comprising about 76 mg of citric acid.
  • composition of embodiment 12 comprising about 250-350 mg of mannitol.
  • composition of embodiment 12 comprising about 300 mg of mannitol.
  • composition of embodiment 12 comprising about 50-100 mg of citric acid.
  • composition of embodiment 12 comprising about 76 mg of citric acid.
  • composition of embodiment 12 comprising about 300 mg of mannitol and about 76 mg of citric acid.
  • composition of embodiment 24 comprising about 300 milligrams of the crystalline compound.
  • composition of embodiment 12 comprising a dose of about 150 mg/m 2 to about 350 mg/m 2 of the crystalline compound.
  • composition of embodiment 12 comprising a dose of about 240 mg/m 2 of the crystalline compound.
  • a pharmaceutical composition formed by dissolving the pharmaceutical composition of any one of embodiments 1-27 in a solvent. 29. The pharmaceutical composition of embodiment 28, wherein the solvent is water.
  • composition of embodiment 30, wherein the sodium chloride concentration is between about 0.1 and 2%.
  • a method of treating a disorder mediated by CDK4 or CDK6 comprising administering an effective amount of a compound of pharmaceutical composition of any one of embodiments 1-37 to a patient in need thereof.
  • a method of treating a cancer comprising administering an effective amount of a compound of pharmaceutical composition of any one of embodiments 1-37 to a patient in need thereof.
  • a method of reducing the effect of chemotherapy on healthy cells in a patient being treated for cancer or abnormal cell proliferation comprising administering to the subject an effective amount of at least one chemotherapeutic agent and an effective amount of a compound of pharmaceutical composition of any one of embodiments 1-37.
  • Compound 1 Pattern 1 in the form of a dihydrochloride, dihydrate in a formulation is provided.
  • the formulation comprises about 300-400 mg, about 350-400 mg, about 360-380 mg, or more particularly about 373 mg, of Compound 1 Pattern 1 dihydrochloride, dihydrate, about 50-100, about 60-80, or more particularly about 76 mg of citric acid monohydrate, and about 250-350 mg, about 280-320 mg, or more particularly about 300 mg of mannitol, and optionally sodium hydroxide and/or hydrochloric acid to adjust the pH as desired.
  • 373 mg of the dihydrochloride, dihydrate will provide 300 mg of Trilaiclib free base.
  • the formulation is lyophilized after Compound 1 Pattern 1 in the form of a dihydrochloride, dihydrate is mixed with citric acid and mannitol.
  • the formulation comprises lyophilized Compound 1 Pattern 1 in the form of a dihydrochloride, dihydrate mixed with citric acid and mannitol, wherein the citric acid and mannitol are added after Compound 1 Pattern 1 in the form of a dihydrochloride, dihydrate is lyophilized.
  • Compound 1 Pattern 1 in the form of a dihydrochloride in a formulation is provided.
  • the formulation comprises about 300-400 mg, about 350-400 mg, or about 360-380 mg, or more particularly about 373 mg, of Compound 1 Pattern 1 dihydrochloride, dihydrate, about 50-100, about 60-80, or more particularly about 76 mg of citric acid monohydrate, and about 250-350 mg, about 280-320 mg or more particularly about 300 mg of mannitol, and optionally sodium hydroxide and/or hydrochloric acid to adjust the pH as desired for reconstitution.
  • this formulation is sterile.
  • the formulation is lyophilized after Compound 1 Pattern 1 in the form of a dihydrochloride, dihydrate is mixed with citric acid and mannitol.
  • the formulation comprises lyophilized Compound 1 Pattern 1 in the form of a dihydrochloride mixed with citric acid and mannitol, wherein the citric acid and mannitol are added after Compound 1 Pattern 1 in the form of a dihydrochloride, dihydrate is lyophilized.
  • a formulation wherein the formulation is prepared by lyophilizing about 300-400 mg, about 350-400 mg, or about 360-380 mg, or more particularly about 373 mg, of Compound 1 Pattern 1 dihydrochloride, dihydrate, about 50-100, about 60-80, or more particularly about 76 mg citric acid monohydrate, and about 250-350 mg, about 280-320 mg or more particularly about 300 mg of mannitol, and optionally sodium hydroxide and/or hydrochloric acid to adjust the pH as desired.
  • the formulation results from lyophilizing Compound 1 Pattern 1 after it is mixed with about 50- 100 mg, about 60-80 mg, or more particularly about 76 mg of citric acid monohydrate, and about 280-320 mg, or more particularly about 300 mg of mannitol.
  • any of the formulations described above can be reconstituted with about 5-100, 5-50, 10-30, about 10-25, or more particularly about 19.5 mL of sterile water, phosphate buffered saline, dilute sugar, or another saline solution.
  • the reconstitution solution is a sterile sodium chloride solution, for example, comprising about 0.9% NaCl or a sterile sugar solution, for example, of about 5% dextrose solution.
  • the resulting reconstituted solution will have any amount of trilaciclib needed for the intended purpose, such as for example, between about 5-50 mg/mL, about 10-25 mg/ml or even about 15 mg/mL of trilaciclib.
  • the resulting solution is then diluted prior to parenteral administration, such as intravenous delivery.
  • Non-limiting examples of typical quality standards and pharmaceutical function are provided in the table below.
  • NF National Formulary
  • USP United States Pharmacopoeia
  • Ph .Eur. European Pharmacopoeia
  • N/A not applicable
  • q.s. quantity sufficient a
  • the target weight of solution filled into vials prior to lyophilization is 12.252 g (12 mL).
  • b Amount in terms of free base (equivalent to 373 mg of Compound 1 Pattern 1 dihydrochloride, dihydrate).
  • c May be used if necessary, to adjust the bulk solution pH.
  • d Used to dissolve components then removed during lyophilization.
  • e Nitrogen is used as an inert gas at the end of the lyophilization process for vacuum adjustment.
  • the resulting formulation can be supplied in any appropriate container, for example in a 20 mL (Type 1) clear glass vial, sealed with a 20 mm gray chlorobutyl rubber stopper and secured with a 20 mm aluminum overseal with a plastic flip-off seal.
  • a 20 mL (Type 1) clear glass vial sealed with a 20 mm gray chlorobutyl rubber stopper and secured with a 20 mm aluminum overseal with a plastic flip-off seal.
  • the solid formulation is reconstituted with at least about 5-100, 5-50, 10-30, about 10-25, or more particularly about 19.5 mL of about 0.9% NaCl or a sterile sugar solution, for example, of about 5% dextrose solution, sodium hydroxide and/or hydrochloric acid is added to adjust pH, and then the reconstituted solution is diluted to the appropriate dose for administration to a human in need thereof, for example at a dosage of about 200-300 mg/m 2 , for example 240 mg/m 2 .
  • an “active agent” is a compound (including a compound disclosed herein), element, or mixture that when administered to a patient, alone or in combination with another compound, element, or mixture, confers, directly or indirectly, a physiological effect on the patient.
  • the indirect physiological effect may occur via a metabolite or other indirect mechanism.
  • “Deuteration” and “deuterated” means that a hydrogen is replaced by a deuterium such that the deuterium exists over natural abundance and is thus “enriched”.
  • An enrichment of 50% means that rather than hydrogen at the specified position the deuterium content is 50%.
  • the term “enriched” as used herein does not mean percentage enriched over natural abundance.
  • the enrichment of deuterium in the specified position of the compound described herein is at least 90%.
  • a “dosage form” means a unit of administration of an active agent.
  • dosage forms include tablets, capsules, injections, suspensions, liquids, intravenous fluids, emulsions, creams, ointments, suppositories, inhalable forms, transdermal forms, and the like.
  • “Pharmaceutical compositions” are compositions comprising at least one active agent, such as a compound or salt of one of the active compounds disclosed herein, and at least one other substance, such as a carrier. Pharmaceutical compositions optionally contain more than one active agent.
  • “Pharmaceutical combinations” or “combination therapy” refers to the administration of at least two active agents, and in one embodiment, three or four or more active agents which may be combined in a single dosage form or provided together in separate dosage forms optionally with instructions that the active agents are to be used together to treat a disorder.
  • “Pharmaceutically acceptable salts” includes derivatives of the disclosed compounds in which the parent compound is modified by making inorganic and organic, suitably non-toxic, acid or base addition salts thereof.
  • the salts of the present compounds can be synthesized from a parent compound that contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • the pharmaceutically acceptable salt can be in the form of a pure crystal, or single morphic form, or can be used in non-crystalline or amorphic, glassy, or vitreous form, or a mixture thereof.
  • the active compound can be provided in the form of a solvate.
  • Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like.
  • the pharmaceutically acceptable salts include the conventional non-toxic salts and the quaternary ammonium salts of the parent compound formed, for example, from non-toxic inorganic or organic acids.
  • conventional non-toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; and the salts prepared from organic acids such as acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC-(CH2)n-COOH where n is 0-4, and the like.
  • inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, n
  • carrier means a diluent, excipient, or vehicle with which an active compound is provided.
  • a “pharmaceutically acceptable excipient” means an excipient that is useful in preparing a pharmaceutical composition/combination that is generally safe, is sufficiently non toxic, and neither biologically nor otherwise undesirable.
  • a “pharmaceutically acceptable excipient” as used in the present application includes both one and more than one such excipient.
  • a “patient” or “host” is a human or non-human animal, including, but not limited to, simian, avian, feline, canine, bovine, equine or porcine in need of medical treatment.
  • Medical treatment can include treatment of an existing condition, such as a disease or disorder, or a prophylactic or diagnostic treatment.
  • the patient or host is a human patient.
  • the patient such as a host can be treated for any condition that is responsive to trilaciclib, including for myelopreservation or as an antineoplastic agent.
  • isolated refers to the material in substantially pure form.
  • An isolated compound does not have another component that materially affects the properties of the compound.
  • an isolated form is at least 50, 60, 70, 80, 90, 95, 98 or 99% pure.
  • a formulation wherein the formulation is prepared by mixing Pattern 1 Compound 1 with excipients for lyophilization.
  • the formulation results from lyophilizing Compound 1 Pattern 1 before mixing with about 50-100 mg, about 60-80 mg, or more particularly about 76 mg of citric acid monohydrate, and about 250-350 mg, about 280-320 mg or more particularly about 300 mg of mannitol, and optionally sodium hydroxide and/or hydrochloric acid to adjust the pH as desired.
  • This formulation can be reconstituted with sterile water, and in non-limiting embodiments, about 10-30, or more particularly about 5-100, 5-50, 10-30, about 10-25, or more particularly about 19.5 mL of sterile water, phosphate buffered saline, dilute sugar, or another saline solution.
  • the reconstitution solution is a sterile sodium chloride solution, for example, comprising about 0.9% NaCl or a sterile sugar solution, for example, of about 5% dextrose solution.
  • the resulting reconstituted solution will have any amount of trilaciclib needed for the intended purpose, such as for example, between about 5- 50 mg/mL, about 10-25 mg/ml or even about 15 mg/mL of trilaciclib.
  • the resulting solution is then diluted prior to parenteral administration, such as intravenous delivery.
  • the isolated Compound 1 Pattern 1 described herein, or an alternative salt, isotopic analog, or prodrug thereof, can be administered in an effective amount to a host to treat any of the disorders described herein using any suitable approach which achieves the desired therapeutic result.
  • the amount and timing of the isolated Compound 1 Pattern 1 administered will, of course, be dependent on the host being treated, the instructions of the supervising medical specialist, on the time course of the exposure, on the manner of administration, on the pharmacokinetic properties of the particular active compound, and on the judgment of the prescribing physician.
  • the dosages given below are a guideline and the physician can titrate doses of the compound to achieve the treatment that the physician considers appropriate for the host.
  • the physician can balance a variety of factors such as age and weight of the host, presence of preexisting disease, as well as presence of other diseases.
  • the pharmaceutical composition may be formulated as any pharmaceutically useful form, and typically a parenteral formulation, such as an intravenous, intramuscular, subcutaneous, or intradermal formulation.
  • parenteral formulation such as an intravenous, intramuscular, subcutaneous, or intradermal formulation.
  • Other alternative formulations include oral, transdermal, or intranasal formulations.
  • the pharmaceutical composition may be in the form of an I.V. bag a vial for injection, a pill, a capsule, a tablet, a transdermal patch, a subcutaneous patch, a dry powder, an inhalation formulation, in a medical device, suppository, buccal, or sublingual formulation.
  • Some dosage forms, such as tablets and capsules are subdivided into suitably sized unit doses containing appropriate quantities of the active components, e.g., an effective amount to achieve the desired purpose.
  • the pharmaceutical composition is in a dosage form that contains from at least about 50 mg/m 2 to about 800 mg/m 2 , from about 100 mg/m 2 to about 600 mg/m 2 , from about 100 mg/m 2 to about 500 mg/m 2 , from about 100 mg/m 2 to about 400 mg/m 2 , from about 100 mg/m 2 to about 350 mg/m 2 , from about 150 mg/m 2 to about 350 mg/m 2 , from about 200 mg/m 2 to about 350 mg/m 2 , or from about 200 mg/m 2 to about 300 mg/m 2 .
  • the pharmaceutical composition is in a dosage form that contains about 240 mg/m 2 .
  • the therapeutically effective dosage of the isolated Compound 1 Pattern 1 described herein will be determined by the health care practitioner depending on the condition, size and age of the patient as well as the route of delivery. In one non-limited embodiment, a dosage from about 0.1 to about 200 mg/kg has therapeutic efficacy, with all weights being calculated based upon the weight of the active compound.
  • the dosage may be the amount of the isolated Compound 1 Pattern 1 needed to provide a serum concentration of the active compound of up to about 10 nM, 50 nM, 100 nM, 200 nM, 300 nM, 400 nM, 500 nM, 600 nM, 700 nM, 800 nM, 900 nM, 1 mM, 5 mM, 10 pM, 20 pM, 30 pM, or 40 pM.
  • the pharmaceutical composition is in a dosage form that contains from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg of the active compound and optionally from about 0.1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 100 mg to about 800 mg, or from about 200 mg to about 600 mg, for example from about 300 mg to about 400 mg of the isolated Compound 1 Pattern 1, measured alternatively either as the free base or its salt, in a unit dosage form, for example for parenteral deliver such as an I.V. bag.
  • Examples are dosage forms with at least 5, 10, 15, 20, 25, 50, 100, 200, 250, 300, 400, 500, 600, 700, or 750 mg of active compound, or its salt.
  • the pharmaceutical composition may also include a molar ratio of the isolated Compound 1 Pattern 1 and an additional active agent, in a ratio that achieves the desired results.
  • the isolated Compound 1 Pattern 1 disclosed herein or used as described herein may be administered parenterally, intravaneously, orally, topically, by inhalation or spray, sublingually, via implant, including ocular implant, transdermally, via buccal administration, rectally, intramuscular, inhalation, intra-aortal, intracranial, subdermal, intraperitioneal, subcutaneous, transnasal, sublingual, or rectal or by other means, in dosage unit formulations containing conventional pharmaceutically acceptable carriers.
  • a parenteral dosage form can be made from Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate.
  • This dosage form can include any pharmaceutically acceptable excipient, for example a liquid excipient.
  • liquid excipients include phosphate buffered saline, unbuffered or buffered saline (e.g. NaCl solution without buffering agents), a sugar solution (e.g. a solution of dextrose, or a combination thereof as desired.
  • phosphate buffered saline unbuffered or buffered saline (e.g. NaCl solution without buffering agents)
  • a sugar solution e.g. a solution of dextrose, or a combination thereof as desired.
  • This concentrated solution of Compound 1 may then be diluted with the same or different liquid excipient to the appropriate dose for treating a human in need thereof.
  • the pH of the solution (either before or after dilution) is adjusted with a pH adjusting reagent (for example HC1 or NaOH).
  • a pH adjusting reagent for example HC1 or NaOH.
  • an additional therapeutic or non-therapeutic agent is added to the solution before administration or to improve shelf life.
  • an oral administration can be in any desired form in which the isolated Compound 1 Pattern 1 is stable as a solid.
  • the isolated Compound 1 Pattern 1 is delivered in a solid microparticle or nanoparticle.
  • the isolated Compound 1 Pattern 1 may be in the form of a plurality of solid particles or droplets having any desired particle size, and for example, from about 0.01, 0.1 or 0.5 to about 5, 10, 20 or more microns, and optionally from about 1 to about 2 microns.
  • the isolated Compound 1 Pattern 1 as disclosed in the present invention has good pharmacokinetic and pharmacodynamics properties, for instance when administered by the oral or intravenous routes.
  • the pharmaceutical formulations can comprise the isolated Compound 1 Pattern 1 described herein or an alternative pharmaceutically acceptable salt thereof, in any pharmaceutically acceptable carrier.
  • Carriers include excipients and diluents and must be of sufficiently high purity and sufficiently low toxicity to render them suitable for administration to the patient being treated.
  • the carrier can be inert or it can possess pharmaceutical benefits of its own.
  • the amount of carrier employed in conjunction with the compound is sufficient to provide a practical quantity of material for administration per unit dose of the compound.
  • Classes of carriers include, but are not limited to binders, buffering agents, coloring agents, diluents, disintegrants, emulsifiers, flavorants, glidents, lubricants, preservatives, stabilizers, surfactants, tableting agents, and wetting agents.
  • Some carriers may be listed in more than one class, for example vegetable oil may be used as a lubricant in some formulations and a diluent in others.
  • Exemplary pharmaceutically acceptable carriers include sugars, starches, celluloses, powdered tragacanth, malt, gelatin; talc, and vegetable oils.
  • Optional active agents may be included in a pharmaceutical composition, which do not substantially interfere with the activity of the compound of the present invention.
  • the pharmaceutical compositions can be in the form of solid form or a semi-solid dosage form that the isolated Compound 1 Pattern 1 is stable in, such as, for example, tablets, suppositories, pills, capsules, powders, or the like, preferably in unit dosage form suitable for single administration of a precise dosage.
  • the compositions will include an effective amount of the selected drug in combination with a pharmaceutically acceptable carrier and, in addition, can include other pharmaceutical agents, adjuvants, diluents, buffers, and the like.
  • compositions of the disclosure can be administered as pharmaceutical formulations including those suitable for oral (including buccal and sub-lingual), rectal, nasal, topical, pulmonary, vaginal administration or in a form suitable for administration by inhalation or insufflation.
  • the preferred manner of administration is oral using a convenient daily dosage regimen which can be adjusted according to the degree of affliction.
  • conventional nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talc, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • permeation enhancer excipients including polymers such as: poly cations (chitosan and its quaternary ammonium derivatives, poly-L- arginine, aminated gelatin); polyanions (A-carboxymethyl chitosan, poly-acrylic acid); and, thiolated polymers (carboxymethyl cellulose-cysteine, polycarbophil-cysteine, chitosan- thiobutylamidine, chitosan-thioglycolic acid, chitosan-glutathione conjugates).
  • polymers such as: poly cations (chitosan and its quaternary ammonium derivatives, poly-L- arginine, aminated gelatin); polyanions (A-carboxymethyl chitosan, poly-acrylic acid); and, thiolated polymers (carboxymethyl cellulose-cysteine, polycarbophil-cysteine, chitosan- thiobutyl
  • the composition will generally take the form of a tablet or capsule. Tablets and capsules are preferred oral administration forms. Tablets and capsules for oral use can include one or more commonly used carriers such as lactose and com starch. Lubricating agents, such as magnesium stearate, are also typically added.
  • the compositions of the disclosure can be combined with an oral, non-toxic, pharmaceutically acceptable, inert carrier such as lactose, starch, sucrose, glucose, methyl cellulose, magnesium stearate, dicalcium phosphate, calcium sulfate, mannitol, sorbitol and the like.
  • suitable binders, lubricants, disintegrating agents, and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • the pharmaceutical formulations can contain other additives, such as pH-adjusting additives.
  • useful pH-adjusting agents include acids, such as hydrochloric acid, bases or buffers, such as sodium lactate, sodium acetate, sodium phosphate, sodium citrate, sodium borate, or sodium gluconate.
  • the formulations can contain antimicrobial preservatives.
  • Useful antimicrobial preservatives include methylparaben, propylparaben, and benzyl alcohol. An antimicrobial preservative is typically employed when the formulations is placed in a vial designed for multi-dose use.
  • the pharmaceutical formulations described herein can be lyophilized using techniques well known in the art.
  • a pharmaceutical composition can take the form of a tablet, pill, capsule, powder, and the like.
  • Tablets containing various excipients such as sodium citrate, calcium carbonate and calcium phosphate may be employed along with various disintegrants such as starch (e.g., potato or tapioca starch) and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • disintegrants such as starch (e.g., potato or tapioca starch) and certain complex silicates, together with binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • binding agents such as polyvinylpyrrolidone, sucrose, gelatin and acacia.
  • lubricating agents such as magnesium stearate, sodium lauryl sulfate, and talc are often very useful for tableting purposes.
  • Solid compositions of a similar type may be employed as fillers in soft and hard-
  • compositions also are provided which provide a controlled release of a compound described herein, including through the use of a degradable polymer, as known in the art.
  • pharmaceutically acceptable salts refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with hosts (e.g., human hosts) without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the presently disclosed host matter.
  • Compound 1 Pattern 1 is not a HC1 salt, but is instead a salt described below.
  • the additional therapeutic agent described in the Combination Section below is administered as a pharmaceutically acceptable salt, for example, a salt described below.
  • salts refers to the relatively non-toxic, inorganic and organic acid addition salts of the presently disclosed compounds. These salts can be prepared during the final isolation and purification of the compounds or by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed.
  • Basic compounds are capable of forming a wide variety of different salts with various inorganic and organic acids. Acid addition salts of the basic compounds are prepared by contacting the free base form with a sufficient amount of the desired acid to produce the salt in the conventional manner.
  • the free base form can be regenerated by contacting the salt form with a base and isolating the free base in the conventional manner.
  • the free base forms may differ from their respective salt forms in certain physical properties such as solubility in polar solvents.
  • Pharmaceutically acceptable base addition salts may be formed with metals or amines, such as alkali and alkaline earth metal hydroxides, or of organic amines. Examples of metals used as cations, include, but are not limited to, sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines include, but are not limited to, N,N'- dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, N- methylglucamine, and procaine.
  • the base addition salts of acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner.
  • the free acid form can be regenerated by contacting the salt form with an acid and isolating the free acid in a conventional manner.
  • the free acid forms may differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents.
  • Salts can be prepared from inorganic acids sulfate, pyrosulfate, bisulfate, sulfite, bisulfite, nitrate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, chloride, bromide, iodide such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydriodic, phosphorus, and the like.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, nitrate, acetate, oxalate, valerate, oleate, palmitate, stearate, laurate, borate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate mesylate, glucoheptonate, lactobionate, laurylsulphonate and isethionate salts, and the like.
  • Salts can also be prepared from organic acids, such as aliphatic mono- and dicarboxylic acids, phenyl -substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and the like.
  • organic acids such as aliphatic mono- and dicarboxylic acids, phenyl -substituted alkanoic acids, hydroxy alkanoic acids, alkanedioic acids, aromatic acids, aliphatic and aromatic sulfonic acids, etc. and the like.
  • Representative salts include acetate, propionate, caprylate, isobutyrate, oxalate, malonate, succinate, suberate, sebacate, fumarate, maleate, mandelate, benzoate, chlorobenzoate, methylbenzoate, dinitrobenzoate, phthalate, benzenesulfonate, toluenesulfonate, phenyl acetate, citrate, lactate, maleate, tartrate, methanesulfonate, and the like.
  • Pharmaceutically acceptable salts can include cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Also contemplated are the salts of amino acids such as arginate, gluconate, galacturonate, and the like. See, for example, Berge et ah, J. Pharm. Sci., 1977, 66, 1-19, which is incorporated herein by reference.
  • Formulations suitable for rectal administration are typically presented as unit dose suppositories. These may be prepared by admixing the active disclosed compound with one or more conventional solid carriers, for example, cocoa butter, and then shaping the resulting mixture.
  • Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil, which maintain the stability of the isolated Compound 1 Pattern 1.
  • Carriers which may be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof.
  • Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Formulations suitable for transdermal administration may also be delivered by iontophoresis (see, for example, Pharmaceutical Research 3 (6):318 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound.
  • microneedle patches or devices are provided for delivery of drugs across or into biological tissue, particularly the skin. The microneedle patches or devices permit drug delivery at clinically relevant rates across or into skin or other tissue barriers, with minimal or no damage, pain, or irritation to the tissue.
  • Formulations suitable for administration to the lungs can be delivered by a wide range of passive breath driven and active power driven single/-multiple dose dry powder inhalers (DPI).
  • DPI dry powder inhalers
  • the devices most commonly used for respiratory delivery include nebulizers, metered- dose inhalers, and dry powder inhalers.
  • nebulizers include jet nebulizers, ultrasonic nebulizers, and vibrating mesh nebulizers. Selection of a suitable lung delivery device depends on parameters, such as nature of the drug and its formulation, the site of action, and pathophysiology of the lung.
  • an IV solution of Compound 1 for injection to cancer patients to preserve healthy cells or for an antineoplastic can be accomplished by administering it as a dihydrochloride or a dihydrochloride, dihydrate.
  • the IV solution is prepared from a lyophilized powder that comprises Compound 1 dihydrochloride, mannitol, and citric acid.
  • the IV solution of Compound 1 is prepared from a solid pharmaceutical composition which has optionally been dried by lyophilization.
  • this composition includes Compound 1 in the form of a dihydrochloride, for example Compound 1 dihydrochloride Pattern 1.
  • the composition may also include appropriate excipients for example a bulking agent, buffer, and/or one or more pH adjustment agents.
  • the bulking agent is an appropriate sugar such as mannitol.
  • the buffer is an appropriate weak acid or weak base with one or more acidic or basic sites.
  • the buffer is a weak acid with two or more acidic sites, for example citric acid.
  • the composition includes NaOH, HC1, and or NaCl as pH adjustment agents (NaOH and/or HC1) or the residual salt from pH adjustment (NaCl).
  • a lyophilized Compound 1 dihydrochloride composition which comprises mannitol and citric acid is crystalline and readily characterized by X-ray powder diffraction (XRPD).
  • the lyophilized Compound 1 dihydrochloride composition comprises about 250 to 350 mg, about 275 to 325 mg or more particularly about 300 mg of Compound 1 wherein the 300 mg of weight corresponds to the free base.
  • Compound 1 in the lyophilized composition is a crystalline dihydrochloride in the form characterized by Pattern 1 herein.
  • Compound 1 within the lyophilized composition is in the form of a different pharmaceutically acceptable salt or a mixture of pharmaceutically acceptable salts.
  • Compound 1 within the lyophilized composition is in the form of a different pattern or a mixture of patterns.
  • the lyophilized Compound 1 composition comprises at least about 1%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% Compound 1 dihydrochloride Pattern 1 relative to other forms of Compound 1.
  • the lyophilized Compound 1 dihydrochloride composition described in Example 28 contains mannitol and citric acid and is characterized by the XRPD pattern shown in FIG. 100.
  • the Compound 1 dihydrochloride composition is lyophilized, in other embodiments it is dried using another technique.
  • the Compound 1 dihydrochloride composition comprises mannitol, for example at least about 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mg of mannitol or about 200 to 400 mg, 225 to 375 mg, 250 to 350 mg, 275 to 325 mg, or 300 mg of mannitol, or at least about 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, or 400 mg of mannitol
  • the Compound 1 dihydrochloride composition comprises one or more additional salts formed from the adjustment of the pH of the bulk solution which is dried to
  • the lyophilized Compound 1 dihydrochloride composition includes about 349 mg of Compound 1 Pattern 1 dihydrochloride (equivalent to about 300 mg of trilaciclib free base), about 50-100 mg, about 60-80 mg, or more particularly about 76 mg of citric acid monohydrate, and about 280-320 mg, for example 300 mg, of mannitol.
  • isolated Compound 1 Pattern 1 is used in the manufacture of a lyophilized form that is then formulated with a suitable solvent such as phosphate buffered saline for administration to a patient, for example, by intravenous delivery. In another embodiment, it can be formulated into a parenteral dosage form. This dosage form can be used, for example, in subcutaneous administration.
  • the morphic form of Compound 1, for example Compound 1 dihydrochloride, dihydrate is used to produce an intravenous formulation as a myelopreservation agent or alternatively when given along with chemotherapy to treat cancer optionally along with the standard of care for the cancer being treated.
  • the standard of care for treating a particular cancer includes the use of therapies that are approved by a regulatory agency, for example the U.S. Food and Drug Administration (FDA), European Medicines Agency (EMA), or the Chinese National Medical Products Administration (NMPA) for the cancer being treated.
  • FDA U.S. Food and Drug Administration
  • EMA European Medicines Agency
  • NMPA Chinese National Medical Products Administration
  • a pharmaceutical formulation as described herein is selected and managed by the patient’s healthcare practitioner.
  • a method of treating a CDK4/6 mediated disorder for example any of the disorders or methods described below, wherein the patient is administered an IV solution comprising Compound 1, mannitol, and citric acid, wherein the IV solution is formed by reconstituting a lyophilized Compound 1 dihydrochloride composition comprising mannitol and citric acid.
  • the lyophilized composition comprises Compound 1 dihydrochloride Pattern 1.
  • Compound 1 Pattern 1 dihydrochloride, dihydrate or an intravenous solution prepared from it is administered on each day of a treatment cycle on which chemotherapy is administered.
  • the intravenous solution is also administered on day 1, day 2, and/or day 3 of the treatment cycle (for example on day 1 or day 1 and day 2 of the treatment cycle).
  • the intravenous solution is administered on day 1 and day 8 of a 21 -day treatment cycle, and as non-limiting examples, in combination with gemcitabine, carboplatin, or topotecan.
  • the isolated Compound 1 Pattern 1 of the present invention decreases the effect of chemotherapeutic agent toxicity on CDK4/6 replication dependent healthy cells, such as hematopoietic stem cells and hematopoietic progenitor cells (together referred to as HSPCs), and/or renal epithelial cells, in subjects, typically humans, that will be, are being, or have been exposed to the chemotherapeutic agent (typically a DNA-damaging agent).
  • CDK4/6 replication dependent healthy cells such as hematopoietic stem cells and hematopoietic progenitor cells (together referred to as HSPCs), and/or renal epithelial cells, in subjects, typically humans, that will be, are being, or have been exposed to the chemotherapeutic agent (typically a DNA-damaging agent).
  • the subject has been exposed to a chemotherapeutic agent, and, using the intravenous solution described herein, the subject’s CDK4/6-replication dependent healthy cells are placed in G1 arrest following exposure in order to mitigate, for example, DNA damage.
  • the compound is administered at least 1 ⁇ 2 hour, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 18 hours, at least 20 hours or more post chemotherapeutic agent exposure.
  • Compound 1 Pattern 1 dihydrochloride, dihydrate or an intravenous solution prepared from it is administered at least 1 ⁇ 2 hour, at least 1 hour, at least 2 hours, at least 3 hours, at least 4 hours, at least 5 hours, at least 6 hours, at least 7 hours, at least 8 hours, at least 10 hours, at least 12 hours, at least 14 hours, at least 16 hours, at least 18 hours, at least 20 hours or more prior to chemotherapeutic agent exposure.
  • Compound 1 Pattern 1 dihydrochloride, dihydrate or an intravenous solution prepared from it is administered about 4 hours before chemotherapy.
  • an intravenous solution prepared from Compound 1 Pattern 1 dihydrochloride, dihydrate is administered in combination with a chemotherapeutic agent including but not limited to a treatment regimen wherein the chemotherapeutic agent is administered: on day 1-3 every 21 days; on days 1-3 every 28 days; on day 1 every 3 weeks; on day 1, day 8, and day 15 every 28 days, on day 1 and day 8 every 28 days; on days 1 and 8 every 21 days; on days 1-5 every 21 days; 1 day a week for 6-8 weeks; on days 1, 22, and 43; days 1 and 2 weekly; days 1-4 and 22-25; days 1-4, 22-25, and 43-46; and similar type- regimens, wherein the CDK4/6-replication dependent cells are arrested at the G1 phase during chemotherapeutic agent exposure.
  • a chemotherapeutic agent including but not limited to a treatment regimen wherein the chemotherapeutic agent is administered: on day 1-3 every 21 days; on days 1-3 every 28 days; on day 1 every 3 weeks; on day 1, day 8, and day
  • the isolated Compound 1 Pattern 1 can allow for dose intensification (e.g., more therapy can be given in a fixed period of time) in medically related chemotherapies, which will translate to better efficacy. Therefore, the presently disclosed methods can result in chemotherapy regimens that are less toxic and more effective.
  • the use of the isolated Compound 1 Pattern 1 described herein may result in reduced or substantially limited off-target effects, for example, related to inhibition of kinases other than CDK4 and/or CDK6 such as CDK2. Furthermore, in certain embodiments, the use of the isolated Compound 1 Pattern 1 described herein should not induce cell cycle arrest in CDK4/6 replication independent cells.
  • the use of the isolated Compound 1 Pattern 1 described herein reduces the risk of undesirable off-target effects including, but not limited to, long term toxicity, anti-oxidant effects, and estrogenic effects.
  • Anti -oxidant effects can be determined by standard assays known in the art. For example, a compound with no significant anti-oxidant effects a compound that does not significantly scavenge free-radicals, such as oxygen radicals.
  • the anti-oxidant effects of a compound can be compared to a compound with known anti oxidant activity, such as genistein.
  • a compound with no significant anti-oxidant activity can be one that has less than about 2, 3, 5, 10, 30, or 100 fold anti-oxidant activity relative to genistein.
  • Estrogenic activities can also be determined via known assays.
  • a non- estrogenic compound is one that does not significantly bind and activate the estrogen receptor.
  • a compound that is substantially limited estrogenic effects can be one that has less than about 2, 3, 5, 10, 20, or 100-fold estrogenic activity relative to a compound with estrogenic activity, e.g., genistein.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it may be administered parenterally, for example, intravenously, to a patient prior to administration of an immune-response inducing chemotherapy such as an ICD-inducing chemotherapy.
  • Compound 1 is administered up to about 24 hours or less, or up to about 20, 15, 10, 5, or 4 hours or less for example about 30-60 minutes or less, prior to administration of the chemotherapy.
  • Compound 1 is administered approximately about 22 to 26 hours before administration of the chemotherapy, and again about 4 hours or less, for example about 30-60 minutes or less, prior to administration of the chemotherapy.
  • the dose of Compound 1 administered is between about 180 and about 280 mg/m 2 .
  • the dose is up to about 100, 125, 150, 180, 185, 190, 195, 200, 205, 210, 215, 220, 225, 230, 235, 240, 245, 250, 255, 260, 265, 270, 275, or 280 mg/m 2 or any dose in between these numbers as determined desirable by the healthcare practitioner.
  • the dose is about 240 mg/m 2 .
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered to the subject prior to treatment with the chemotherapeutic agent such that its concentration reaches peak serum levels before or during treatment with the chemotherapeutic agent, allowing for the inhibition of proliferation of immune effector cells, thus protecting them from the harmful effects of chemotherapy.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered concomitantly, or closely thereto, with the chemotherapeutic agent exposure.
  • the CDK4/6 inhibitor described herein can be administered following exposure to the chemotherapeutic agent if desired to mitigate immune effector cell damage associated with chemotherapeutic agent exposure.
  • an aqueous solution produced form Compound 1 Pattern 1 is administered to a patient in need thereof.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered to the subject less than about 24 hours, about 20 hours, about 16 hours, about 12 hours, about 8 hours, about 4 hours, about 2.5 hours, about 2 hours, about 1 hour, about 1 ⁇ 2 hour or less prior to treatment with the chemotherapeutic agent.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered about 1 ⁇ 2 hour prior to administration of the chemotherapeutic agent.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered to the subject twice before administration of the chemotherapy.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered between about 18 and 28 hours before the administration of the chemotherapy, and then once again at less than about 4 hours, about 2.5 hours, about 2 hours, about 1 hour, about 1 ⁇ 2 hour or less prior to treatment with the chemotherapeutic agent.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered between about 22 and 26 hours prior to administration of the chemotherapeutic agent and again about 1 ⁇ 2 hour or less prior to administration of the chemotherapeutic agent.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered prior to or concomitantly with the administration of a chemotherapeutic agent, wherein the chemotherapeutic agent is administered: for example, on day 1-3 every 21 days; on days 1-3 every 28 days; on day 1 every 3 weeks; on day 1, day 8, and day 15 every 28 days, on day 1 and day 8 every 28 days; on days 1 and day 8 every 21 days; on days 1-5 every 21 days; 1 day a week for 6-8 weeks; on days 1, 22, and 43; days 1 and 2 weekly; days 1-4 and 22-25; days 1- 4; days 22-25, and days 43-46; and similar type chemotherapeutic regimens.
  • a chemotherapeutic agent wherein the chemotherapeutic agent is administered: for example, on day 1-3 every 21 days; on days 1-3 every 28 days; on day 1 every 3 weeks; on day 1, day 8, and day 15 every 28 days, on day 1 and day 8 every 28 days;
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered prior to or concomitantly with at least one administration of the chemotherapeutic agent during a chemotherapeutic treatment regimen.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered prior to or concomitantly with one or more administrations of the chemotherapeutic agent during a chemotherapeutic treatment regimen.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered prior to or concomitantly with each administration of the chemotherapeutic agent during a chemotherapeutic treatment regimen.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered prior to or concomitantly with each administration of a chemotherapeutic agent for example during a standard chemotherapeutic protocol such as, for example, a 21-day cycle. Following cessation of the standard chemotherapeutic protocol, Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it can be further administered alone in a maintenance dose.
  • a chemotherapeutic agent for example during a standard chemotherapeutic protocol such as, for example, a 21-day cycle.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is further administered once a week for at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 26, 52, 104 weeks, or longer.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered once every 21 days following the cessation of the chemotherapeutic protocol.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is a fast-acting, short half-life CDK4/6 inhibitor.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered with a chemotherapy agent in a maintenance therapy treatment regimen following cessation of the standard chemotherapeutic protocol.
  • Maintenance therapy can comprise either continuation of an agent given as part of the first-line or previous regimen (continuation maintenance) or treatment with a new agent (switch maintenance).
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is further administered in a maintenance-type therapeutic regimen, wherein Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered in combination with a reduced maintenance dose of chemotherapy at a regular dosing interval for example but not limited to, once a week, once every two weeks, once every three weeks, once a month, once every six weeks, once every two months, once every three months, or once every six months following the completion of the initial chemotherapy treatment.
  • a regular dosing interval for example but not limited to, once a week, once every two weeks, once every three weeks, once a month, once every six weeks, once every two months, once every three months, or once every six months following the completion of the initial chemotherapy treatment.
  • Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered with the same agent used in the previous phase of chemotherapy treatment. In some embodiments, Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is administered with a different chemotherapy agent than was used in the previous phase of chemotherapy treatment.
  • Standard cancer chemotherapy can promote tumor immunity in two major ways: (i) inducing immunogenic cell death as part of its intended therapeutic effect; and (ii) disrupting strategies that tumors use to evade the immune response.
  • a large body of data demonstrates that some chemotherapy drugs at their standard dose and schedule mediate their antitumor effect, at least in part, by inducing immunogenic cell death (see, e.g., Emens et ah, Chemotherapy: friend of foe to cancer vaccines? Curr Opin Mol Ther 2001;3:77-84; Vanmeerbeek et al., Trial Watch: Chemotherapy-Induced Immunogenic Cell Death in Immuni- Oncology. Oncoimmunology Vol. 9, No. 1 2020:el703449, both incorporated by reference herein).
  • Immunogenic cell death is a type of cell death characterized by, for example, cell surface translocation of calreticulin (CRT), extracellular release of ATP and high mobility group box 1 (HMBG1), and stimulation of type I interferon (IFN) responses.
  • CRT cell surface translocation of calreticulin
  • HMBG1 high mobility group box 1
  • IFN type I interferon
  • ICD in cancer cells may prime an anticancer immune response.
  • a variety of chemotherapeutic agents can induce ICD, as indicated by the alterations in tumor-infiltrating lymphocytes (TIL) abundance and composition.
  • DAMP damage-associated molecular pattern
  • ATP damage-associated molecular pattern
  • HMGB1 damage-associated molecular pattern
  • DCs dendritic cells
  • Cross-priming of CD8+ T-cells is triggered by mature DCs and gd T-cells in an IL-Ib and IL-17 dependent manner.
  • Primed CTLs then elicit a direct cytotoxic response to kill remaining tumor cells through the generation of IFN-g, perforin- 1 and granzyme B.
  • Compound 1 Pattern 1 for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it is used for myelopreservation in combination with ICD-inducing chemotherapy.
  • ICD-inducing chemotherapies for use in the present invention include alkylating agents such as cyclophosphamide, trabectedin, temozolomide, melphalan, dacarbazine, and oxaliplatin; antimetabolites such as methotrexate, mitroxantrone, gemcitabine, and 5- fluorouracil (5-FU); cytotoxic antibiotics such as bleomycin and anthracyclines, including doxorubicin, daunorubicin, epirubicin, idarubicin, and valrubicin; taxanes, such as paclitaxel, cabazitaxel, and docetaxel; topoisomerase inhibitors such as topotecan, irinotecan, and etoposide; platinum compounds such as carboplatin and cisplatin; anti -microtubule vinca alkaloid agents such as vinblastine, vincristine, vinorelbine, and vindesine
  • ICD- inducing chemotherapies include bortezomib, an inhibitor of the 26S proteasome subunit, mechlorethamine, diaziquone, mitomycin C, fludarabine and cytosine arabinoside.
  • the ICD-inducing chemotherapy is selected from idarubicin, epirubicin, doxorubicin, mitoxantrone, oxaliplatin, bortezomib, gemcitabine, and cyclophosphamide, and combinations thereof.
  • the chemotherapeutic administered is capable of inducing an immune-response may modulate tumor immunity by mechanisms distinct from immunogenic cell death.
  • Various chemotherapy drugs can modulate the activity of distinct immune cell subsets or the immune phenotype of tumor cells through enhancing antigen presentation, enhancing expression of costimulatory molecules including B7.1 (CD80) and B7.2 (CD86), downregulating checkpoint molecules such as programmed death-ligand 1 (PD-L1), or promoting tumor cell death through the fas, perforin, or Granzyme B pathways.
  • costimulatory molecules including B7.1 (CD80) and B7.2 (CD86)
  • PD-L1 programmed death-ligand 1
  • Chemotherapies that modulate tumor immunity may do so by: abrogating myeloid-derived suppressor cell (MDSC) activity, for example gemcitabine, 5-fluoruracil, cisplatin, and doxorubicin; abrogating Treg activity, for example cyclophosphamide, 5-fluorouracil; paclitaxel, cisplatin, and fludarabine; enhancement of T-cell cross priming, for example gemcitabine and anthracyclines such as doxorubicin, daunorubicin, epirubicin, valrubicin and idarubicin.; augmenting dendritic cell activation, for example anthracyclines, taxanes, cyclophosphamide, vinca alkaloids, methotrexate, and mitomycin C; promoting anti tumor CD4+ T-cell phenotype, for example cyclophosphamide and paclitaxel; and promoting tumor cell recognition and lysis, for example cyclo
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of an alkylating agent such as cyclophosphamide, trabectedin, temozolomide, melphalan, dacarbazine, or oxaliplatin; an antimetabolite such as methotrexate, mitroxantrone, gemcitabine, or 5-fluorouracil (5-FU); a cytotoxic antibiotic
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with the chemotherapy does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of cyclophosphamide.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with cyclophosphamide does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered-excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of a CDK 4/6 inhibitor with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of trabectedin.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with trabectedin does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of temozolomide.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it inhibitor in combination with temozolomide does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered-excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of melphalan.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of a CDK 4/6 inhibitor with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of dacarbazine.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of oxaliplatin.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of methotrexate.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of a CDK 4/6 inhibitor with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of 5-fluorouracil (5-FU).
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of gemcitabine.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of mitoxantrone.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of a CDK 4/6 inhibitor with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of doxorubicin.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with doxorubicin does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of daunorubicin.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of idarubicin
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of valrubicin.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor. In some embodiments, the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of a CDK 4/6 inhibitor with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of epirubicin.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor. In some embodiments, the patient has an altered-excluded immune tumor. In some embodiments, the patient has a cold tumor. In some embodiments, the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer. In some embodiments, the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it combination with an effective amount of bleomycin.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with bleomycin does not include administering an immune checkpoint inhibitor.
  • the patient has a hot immune tumor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered-excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of bortezomib.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with bortezomib does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.” In some embodiments, the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of paclitaxel.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with paclitaxel does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of docetaxel.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with docetaxel does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered-excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of cabazitaxel.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with cabazitaxel does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of a CDK 4/6 inhibitor with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of topotecan.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with topotecan does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of etoposide.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with etoposide does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of irinotecan.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with irinotecan does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of cisplatin.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with cisplatin does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of carboplatin.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with carboplatin does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of vinblastine.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with vinblastine does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of vincristine.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with vincristine does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of vinorelbine.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with vinorelbine does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of vindesine.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with vindesine does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of diaziquone.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with diaziquone does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of mechlorethamine.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with mechlorethamine does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered-excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of mitomycin C.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with mitomycin C does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of fludarabine.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with fludarabine does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered- excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • a method for selecting a patient or patient population for cancer therapy that includes the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it with chemotherapy in a manner that increases the progression free survival or overall survival of the patient or patient population is provided comprising, determining if the cancer has a surrounding microenvironment that is favorable to immune modulation, is immunogenically susceptible to CDK4/6 inhibitor treatment, or is immunogenic, and if so, administering to the patient an effective amount of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with an effective amount of cytosine arabinoside.
  • the administration of Compound 1 Pattern 1, for example Compound 1 Pattern 1 dihydrochloride, dihydrate, or a formulation produced from it in combination with cytosine arobinoside does not include administering an immune checkpoint inhibitor.
  • the patient has a tumor classified as immunogenic.
  • the patient has a hot immune tumor.
  • the patient has an altered-immunosuppressed immune tumor.
  • the patient has an altered-excluded immune tumor.
  • the patient has a cold tumor.
  • the patient has a tumor that is classified as a C2 “IFN-g Dominant” class cancer.
  • the patient has a tumor that is classified as a high “IFN-g Signature” or a high “Expanded Immune Signature.”
  • the patient has a tumor that is PD-L1 positive.
  • the patient to be treated has been determined to have a cancer having a surrounding microenvironment that is favorable to immune modulation, is immunogenic, or is immunogenically susceptible to CDK4/6 inhibitor treatment. Accordingly, provided the cancer fits into the category as described herein, the patient may be suitable for the described treatments.
  • the cancer to be treated is selected from the group consisting of breast cancer, including but not limited to estrogen receptor (ER)-positive breast cancer and triple negative breast cancer, non-small cell lung carcinoma, head and neck squamous cell cancer, classical Hodgkin lymphoma (cHL), diffuse large B-cell lymphoma, bladder cancer, primary mediastinal B-cell lymphoma (PBMCL), urothelial carcinoma, microsatellite instability -high (MSI-H) solid tumors, mismatch repair deficient (dMMR) solid tumor, gastric or gastroesophageal junction (GEJ) adenocarcinoma, squamous cell carcinoma of the esophagus, cervical cancer, endometrial cancer, cholangiocarcinoma, hepatocellular carcinoma, Merkel cell carcinoma, renal cell carcinoma, ovarian cancer, anal canal cancer, colorectal cancer, skin cutaneous melanoma and melanoma.
  • cHL central lymphoma
  • the patient is not administered a check point inhibitor.
  • the patient is administered a check point inhibitor.
  • Compound 1 Pattern 1 dihydrochloride, dihydrate can be used to treat an abnormal cellular proliferation disorder, inflammatory disorder, immune disorder, or autoimmune disorder.
  • a method of treating a proliferative disorder in a host, including a human comprising administering isolated Compound 1 Pattern 1, for example as a dihydrochloride, dihydrate, optionally in a pharmaceutically acceptable carrier.
  • disorders include tumors, cancers, disorders related to abnormal cellular proliferation, inflammatory disorders, immune disorders, and autoimmune disorders.
  • Compound 1 Pattern 1 is administered parentally. This administration can be daily or with treatment holidays for myelopreservation.
  • Compound 1 Pattern 1 is useful as a therapeutic agent in a dosage form when administered in an effective amount to a host, including a human, to treat a tumor, cancer (solid, non-solid, diffuse, hematological, etc.), abnormal cellular proliferation, immune disorder, inflammatory disorder, blood disorder, a myelo- or lymphoproliferative disorder such as B- or T-cell lymphomas, multiple myeloma, breast cancer, prostate cancer, AML, ALL, ACL, lung cancer, pancreatic cancer, colon cancer, skin cancer, melanoma, Waldenstrom’s macroglobulinemia, Wiskott-Aldrich syndrome, or a post-transplant lymphoproliferative disorder; an autoimmune disorder, for example, Lupus, Crohn’s Disease, Addison disease, Celiac disease, dermatomyositis, Graves’ disease, thyroiditis, multiple sclerosis, pernicious anemia, reactive arthritis, or type I diabetes; a disease of cardiologic malfunction, including hyperchol
  • proliferative disorders include, but are not limited to, benign growths, neoplasms, tumors, cancer (Rb positive or Rb negative), autoimmune disorders, inflammatory disorders graft-versus-host rejection, and fibrotic disorders.
  • Non-limiting examples of cancers that can be treated according to the present invention include, but are not limited to, acoustic neuroma, adenocarcinoma, adrenal gland cancer, anal cancer, angiosarcoma (e.g., lymphangiosarcoma, lymphangioendotheliosarcoma, hemangiosarcoma), appendix cancer, benign monoclonal gammopathy, biliary cancer (e.g., cholangiocarcinoma), bladder cancer, breast cancer (e.g., adenocarcinoma of the breast, papillary carcinoma of the breast, mammary cancer, medullary carcinoma of the breast), brain cancer (e.g., meningioma; glioma, e.g., astrocytoma, oligodendroglioma; medulloblastoma), bronchus cancer, carcinoid tumor, cervical cancer (e.g., cervical adenocarcinoma
  • Wilms tumor, renal cell carcinoma), liver cancer (e.g., hepatocellular cancer (HCC), malignant hepatoma), lung cancer (e.g., bronchogenic carcinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung), leiomyosarcoma (LMS), mastocytosis (e.g., systemic mastocytosis), myelodysplastic syndrome (MDS), mesothelioma, myeloproliferative disorder (MPD) (e.g., polycythemia Vera (PV), essential thrombocytosis (ET), agnogenic myeloid metaplasia (AMM) a.k.a.
  • HCC hepatocellular cancer
  • lung cancer e.g., bronchogenic carcinoma, small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), adenocarcinoma of the lung
  • myelofibrosis MF
  • chronic idiopathic myelofibrosis chronic neutrophilic leukemia (CNL), hypereosinophilic syndrome (HES)
  • neuroblastoma e.g., neurofibromatosis (NF) type 1 or type 2, schwannomatosis
  • neuroendocrine cancer e.g., gastroenteropancreatic neuroendoctrine tumor (GEP-NET), carcinoid tumor
  • osteosarcoma ovarian cancer (e.g., cystadenocarcinoma, ovarian embryonal carcinoma, ovarian adenocarcinoma), papillary adenocarcinoma, pancreatic cancer (e.g., pancreatic andenocarcinoma, intraductal papillary mucinous neoplasm (IPMN), Islet cell tumors), penile cancer (e.g., Paget’s disease of the penis and scrotum), pineal om
  • the invention includes the use of an effective amount of the isolated Compound 1 Pattern 1, or its pharmaceutically acceptable salt, prodrug or isotopic variant optionally in a pharmaceutical composition, to treat a host, typically a human, with a selected cancer, tumor, hyperproliferative condition or an inflammatory or immune disorder.
  • Compound 1 Pattern 1 is also active against T-cell proliferation. Given the paucity of drugs for T-cell cancers and abnormal proliferation, the identification of such uses represents a substantial improvement in the medical therapy for these diseases.
  • the disorder is myelodysplastic syndrome (MDS).
  • MDS myelodysplastic syndrome
  • the cancer is a hematopoietic cancer.
  • the hematopoietic cancer is a lymphoma.
  • the hematopoietic cancer is a leukemia.
  • the leukemia is acute myelocytic leukemia (AML).
  • the proliferative disorder is a myeloproliferative neoplasm.
  • the myeloproliferative neoplasm is primary myelofibrosis (PMF).
  • the cancer is a solid tumor.
  • a solid tumor refers to an abnormal mass of tissue that usually does not contain cysts or liquid areas. Different types of solid tumors are named for the type of cells that form them. Examples of classes of solid tumors include, but are not limited to, sarcomas, carcinomas, and lymphomas, as described above herein. Additional examples of solid tumors include, but are not limited to, squamous cell carcinoma, colon cancer, breast cancer, prostate cancer, lung cancer, liver cancer, pancreatic cancer, and melanoma.
  • condition treated with Compound 1 Pattern l is a disorder related to abnormal cellular proliferation.
  • Abnormal cellular proliferation notably hyperproliferation
  • Psoriasis is a benign disease of human skin generally characterized by plaques covered by thickened scales. The disease is caused by increased proliferation of epidermal cells of unknown cause. Chronic eczema is also associated with significant hyperproliferation of the epidermis.
  • Other diseases caused by hyperproliferation of skin cells include atopic dermatitis, lichen planus, warts, pemphigus vulgaris, actinic keratosis, basal cell carcinoma and squamous cell carcinoma.
  • hyperproliferative cell disorders include blood vessel proliferation disorders, fibrotic disorders, autoimmune disorders, graft-versus-host rejection, tumors and cancers.
  • Blood vessel proliferative disorders include angiogenic and vasculogenic disorders. Proliferation of smooth muscle cells in the course of development of plaques in vascular tissue cause, for example, restenosis, retinopathies and atherosclerosis. Both cell migration and cell proliferation play a role in the formation of atherosclerotic lesions.
  • Fibrotic disorders are often due to the abnormal formation of an extracellular matrix.
  • fibrotic disorders include hepatic cirrhosis and mesangial proliferative cell disorders.
  • Hepatic cirrhosis is characterized by the increase in extracellular matrix constituents resulting in the formation of a hepatic scar.
  • Hepatic cirrhosis can cause diseases such as cirrhosis of the liver.
  • An increased extracellular matrix resulting in a hepatic scar can also be caused by viral infection such as hepatitis. Lipocytes appear to play a major role in hepatic cirrhosis.
  • Mesangial disorders are brought about by abnormal proliferation of mesangial cells.
  • Mesangial hyperproliferative cell disorders include various human renal diseases, such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombotic micro angiopathy syndromes, transplant rejection, and glomerulopathies.
  • Rheumatoid arthritis is generally considered an autoimmune disease that is thought to be associated with activity of autoreactive T cells, and to be caused by autoantibodies produced against collagen and IgE.
  • Bechet CAD
  • ARDS acute respiratory distress syndrome
  • ischemic heart disease CAD
  • post dialysis syndrome CAD
  • leukemia CAD
  • acquired immune deficiency syndrome CAD
  • vasculitis lipid histiocytosis
  • septic shock inflammation in general.
  • a compound of the present invention and its pharmaceutically acceptable derivatives or pharmaceutically acceptable formulations containing these compounds are also useful in the prevention and treatment of HBV infections and other related conditions such as anti -HBV antibody positive and HBV-positive conditions, chronic liver inflammation caused by HBV, cirrhosis, acute hepatitis, fulminant hepatitis, chronic persistent hepatitis, and fatigue.
  • These compounds or formulations can also be used prophylactically to prevent or retard the progression of clinical illness in individuals who are anti-HBV antibody or HBV-antigen positive or who have been exposed to HBV.
  • the condition is associated with an immune response.
  • Cutaneous contact hypersensitivity and asthma are just two examples of immune responses that can be associated with significant morbidity.
  • Others include atopic dermatitis, eczema, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, ulceris, conjunctivitis, keratoconjunctivitis, ulcerative colitis, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, and drug eruptions.
  • immunologically mediated leukocyte infiltration In atopic dermatitis, and eczema in general, immunologically mediated leukocyte infiltration (particularly infiltration of mononuclear cells, lymphocytes, neutrophils, and eosinophils) into the skin importantly contributes to the pathogenesis of these diseases. Chronic eczema also is associated with significant hyperproliferation of the epidermis. Immunologically mediated leukocyte infiltration also occurs at sites other than the skin, such as in the airways in asthma and in the tear producing gland of the eye in keratoconjunctivitis sicca.
  • compounds of the present invention are used as topical agents in treating contact dermatitis, atopic dermatitis, eczematous dermatitis, psoriasis, Sjogren's Syndrome, including keratoconjunctivitis sicca secondary to Sjogren's Syndrome, alopecia areata, allergic responses due to arthropod bite reactions, Crohn's disease, aphthous ulcer, ulceris, conjunctivitis, keratoconjunctivitis, ulcerative colitis, asthma, allergic asthma, cutaneous lupus erythematosus, scleroderma, vaginitis, proctitis, and drug eruptions.
  • novel method may also be useful in reducing the infiltration of skin by malignant leukocytes in diseases such as mycosis fungoides.
  • These compounds can also be used to treat an aqueous- deficient dry eye state (such as immune mediated keratoconjunctivitis) in a patient suffering therefrom, by administering the compound topically to the eye.
  • neoplasia or “cancer” is used throughout the specification to refer to the pathological process that results in the formation and growth of a cancerous or malignant neoplasm, i.e., abnormal tissue (solid) or cells (non-solid) that grow by cellular proliferation, often more rapidly than normal and continues to grow after the stimuli that initiated the new growth cease.
  • malignant neoplasms show partial or complete lack of structural organization and functional coordination with the normal tissue and most invade surrounding tissues, can metastasize to several sites, are likely to recur after attempted removal and may cause the death of the patient unless adequately treated.
  • neoplasia is used to describe all cancerous disease states and embraces or encompasses the pathological process associated with malignant hematogenous, ascitic and solid tumors.
  • Exemplary cancers which may be treated by the present disclosed compounds either alone or in combination with at least one additional anti-cancer agent include squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, and renal cell carcinomas, cancer of the bladder, head, kidney, and neck, ; leukemias; benign and malignant lymphomas, particularly Burkitf s lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sar
  • Additional cancers which may be treated using the disclosed compounds according to the present invention include, for example, acute granulocytic leukemia, acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), adenocarcinoma, adenosarcoma, adrenal cancer, adrenocortical carcinoma, anal cancer, anaplastic astrocytoma, angiosarcoma, appendix cancer, astrocytoma, Basal cell carcinoma, B-Cell lymphoma, bile duct cancer, bladder cancer, bone cancer, bone marrow cancer, bowel cancer, brain cancer, brain stem glioma, breast cancer, triple (estrogen, progesterone and HER-2) negative breast cancer, double negative breast cancer (two of estrogen, progesterone and HER-2 are negative), single negative (one of estrogen, progesterone and HER-2 is negative), estrogen-receptor positive, HER2- negative breast cancer, estrogen receptor-negative breast cancer, estrogen receptor positive breast
  • a method of increasing BIM expression (e.g., BCLC2L11 expression) is provided to induce apoptosis in a cell comprising contacting a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof with the cell.
  • the method is an in vitro method.
  • the method is an in vivo method.
  • BCL2L11 expression is tightly regulated in a cell.
  • BCL2L11 encodes for BIM, a proapoptotic protein.
  • BCL2L11 is downregulated in many cancers and BIM is inhibited in many cancers, including chronic myelocytic leukemia (CML) and non-small cell lung cancer (NSCLC) and that suppression of BCL2L11 expression can confer resistance to tyrosine kinase inhibitors.
  • CML chronic myelocytic leukemia
  • NSCLC non-small cell lung cancer
  • a method of treating a condition associated with angiogenesis comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
  • a diabetic condition e.g., diabetic retinopathy
  • an inflammatory condition e.g., rheumatoid arthritis
  • macular degeneration e.g., obesity, atherosclerosis, or a proliferative disorder
  • the condition associated with angiogenesis is macular degeneration.
  • a method of treating macular degeneration comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
  • the condition associated with angiogenesis is obesity.
  • “obesity” and “obese” as used herein refers to class I obesity, class II obesity, class III obesity and pre-obesity (e.g., being “over-weight”) as defined by the World Health Organization.
  • a method of treating obesity comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
  • the condition associated with angiogenesis is atherosclerosis.
  • a method of treating atherosclerosis comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
  • the condition associated with angiogenesis is a proliferative disorder.
  • a method of treating a proliferative disorder comprising administering to a subject in need thereof a compound of the present invention or a pharmaceutically acceptable composition, salt, isotopic analog, or prodrug thereof.
  • the compound is administered at a dose of about 50 mg/m 2 to about 800 mg/m 2 , from about 100 mg/m 2 to about 600 mg/m 2 , from about 100 mg/m 2 to about 500 mg/m 2 , from about 100 mg/m 2 to about 400 mg/m 2 , from about 100 mg/m 2 to about 350 mg/m 2 , from about 150 mg/m 2 to about 350 mg/m 2 , from about 200 mg/m 2 to about 350 mg/m 2 , or from about 200 mg/m 2 to about 300 mg/m 2 .
  • Isolated Compound 1 Pattern 1 can be used in an effective amount alone or in combination with another compound of the present invention or another bioactive agent to treat a host such as a human with a disorder as described herein.
  • the isolated Compound 1 Pattern 1 described herein can be used in an effective amount alone or in combination with another compound of the present invention or another bioactive agent to treat a host such as a human with a disorder as described herein.
  • bioactive agent is used to describe an agent, other than the selected compound according to the present invention, which can be used in combination or alternation with a compound of the present invention to achieve a desired result of therapy.
  • the compound of the present invention and the bioactive agent are administered in a manner that they are active in vivo during overlapping time periods, for example, have time-period overlapping Cmax, Tmax, AUC or other pharmacokinetic parameter.
  • isolated Compound 1 Pattern 1 and the bioactive agent are administered to a host in need thereof that do not have overlapping pharmacokinetic parameter, however, one has a therapeutic impact on the therapeutic efficacy of the other.
  • the bioactive agent is an immune modulator, including but not limited to a checkpoint inhibitor, including as non-limiting examples, a PD- 1 inhibitor, PD-L1 inhibitor, PD-L2 inhibitor, CTLA-4 inhibitor, LAG-3 inhibitor, TIM-3 inhibitor, V-domain Ig suppressor of T-cell activation (VISTA) inhibitors, small molecule, peptide, nucleotide, or other inhibitor.
  • the immune modulator is an antibody, such as a monoclonal antibody.
  • PD-L1 inhibitors that block the interaction of PD-1 and PD-L1 by binding to the PD-L1 receptor, and in turn inhibits immune suppression, include for example, atezolizumab (Tecentriq), durvalumab (AstraZeneca and Medlmmune), KN035 (Alphamab), and BMS-936559 (Bristol-Myers Squibb).
  • CTLA-4 checkpoint inhibitors that bind to CTLA-4 and inhibits immune suppression include, but are not limited to, ipilimumab, tremelimumab (AstraZeneca and Medlmmune), AGEN1884 and AGEN2041 (Agenus).
  • LAG-3 checkpoint inhibitors include, but are not limited to, BMS-986016 (Bristol-Myers Squibb), GSK2831781 (GlaxoSmithKline), IMP321 (Prima BioMed), LAG525 (Novartis), and the dual PD-1 and LAG-3 inhibitor MGD013 (MacroGenics).
  • BMS-986016 Bristol-Myers Squibb
  • GSK2831781 GaxoSmithKline
  • IMP321 Primary BioMed
  • LAG525 Novartis
  • MGD013 Non-Genics
  • An example of a TIM-3 inhibitor is TSR-022 (Tesaro).
  • isolated Compound 1 Pattern 1 as described herein can be administered in an effective amount for the treatment of abnormal tissue of the female reproductive system such as breast, ovarian, endometrial, or uterine cancer, in combination or alternation with an effective amount of an estrogen inhibitor including but not limited to a SERM (selective estrogen receptor modulator), a SERD (selective estrogen receptor degrader), a complete estrogen receptor degrader, or another form of partial or complete estrogen antagonist or agonist.
  • SERM selective estrogen receptor modulator
  • SERD selective estrogen receptor degrader
  • Partial anti -estrogens like raloxifene and tamoxifen retain some estrogen like effects, including an estrogen-like stimulation of uterine growth, and also, in some cases, an estrogen-like action during breast cancer progression which actually stimulates tumor growth.
  • fulvestrant a complete anti -estrogen, is limited estrogen-like action on the uterus and is effective in tamoxifen-resistant tumors.
  • anti-estrogen compounds are provided in WO 2014/19176 assigned to Astra Zeneca, W02013/090921, WO 2014/203129, WO 2014/203132, and US2013/0178445 assigned to Olema Pharmaceuticals, and U.S. Patent Nos. 9,078,871, 8,853,423, and 8,703, 810, as well as US 2015/0005286, WO 2014/205136, and WO 2014/205138.
  • anti-estrogen compounds include: SERMS such as anordrin, adoxifene, broparestriol, chlorotrianisene, clomiphene citrate, cyclofenil, lasofoxifene, ormeloxifene, raloxifene, tamoxifen, toremifene, and fulvestrant; aromatase inhibitors such as aminoglutethimide, testolactone, anastrozole, exemestane, fadrozole, formestane, and letrozole; and antigonadotropins such as leuprorelin, cetrorelix, allylestrenol, chloromadinone acetate, cyproterone acetate, delmadinone acetate, dydrogesterone, medroxyprogesterone acetate, megestrol acetate, nomegestrol acetate, norethisterone acetate, progestrol acetate
  • estrogenic ligands that can be used according to the present invention are described in U.S. Patent Nos. 4,418,068; 5,478,847; 5,393,763; and 5,457,117, WO2011/156518, US Patent Nos. 8,455,534 and 8,299,112, U.S. Patent Nos. 9,078,871; 8,853,423; 8,703,810; US 2015/0005286; and WO 2014/205138,
  • the isolated Compound 1 Pattern 1 described herein can be administered in an effective amount for the treatment of abnormal tissue of the male reproductive system such as prostate or testicular cancer, in combination or alternation with an effective amount of an androgen (such as testosterone) inhibitor including but not limited to a selective androgen receptor modulator, a selective androgen receptor degrader, a complete androgen receptor degrader, or another form of partial or complete androgen antagonist.
  • an androgen such as testosterone
  • the prostate or testicular cancer is androgen-resistant.
  • anti-androgen compounds are provided in WO 2011/156518 and US Patent Nos. 8,455,534 and 8,299,112.
  • anti -androgen compounds include: enzalutamide, apalutamide, cyproterone acetate, chlormadinone acetate, spironolactone, canrenone, drospirenone, ketoconazole, topilutamide, abiraterone acetate, and cimetidine.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of abiraterone acetate (Zytiga) for the treatment of abnormal tissue of the male reproductive system.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of abiraterone acetate (Zytiga) for the treatment of prostate cancer.
  • Zytiga abiraterone acetate
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of enzalutamide for the treatment of prostate cancer.
  • the bioactive agent is an ALK inhibitor.
  • ALK inhibitors include but are not limited to Crizotinib, Alectinib, ceritinib, TAE684 (NVP- TAE684), GSK1838705A, AZD3463, ASP3026, PF-06463922, entrectinib (RXDX-101), and AP26113.
  • the bioactive agent is an EGFR inhibitor.
  • EGFR inhibitors include erlotinib (Tarceva), gefitinib (Iressa), afatinib (Gilotrif), rociletinib (CO- 1686), osimertinib (Tagrisso), olmutinib (Olita), naquotinib (ASP8273), soloartinib (EGF816), PF- 06747775 (Pfizer), icotinib (BPI-2009), neratinib (HKI-272; PB272); avitinib (ACOOIO), EAI045, tarloxotinib (TH-4000; PR-610), PF-06459988 (Pfizer), tesevatinib (XL647; EXEL- 7647; KD-019), transtinib, WZ-3146, WZ8040, CNX-2006,
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of afatinib dimaleate (Gilotrif) for the treatment of non small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of alectinib (Alecensa) for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ceritinib (Zykadia) for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of crizotinib (Xalkori) for the treatment of non-small cell lung cancer. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of osimertinib (Tagrisso) for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of brigatinib (Alunbrig) for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of lorlatinib for the treatment of non-small cell lung cancer.
  • the bioactive agent is an HER-2 inhibitor.
  • HER-2 inhibitors include trastuzumab, lapatinib, ado-trastuzumab emtansine, and pertuzumab.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of lapatinib ditosylate for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of lapatinib ditosylate for the treatment of HER2+ breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of PF7775 for the treatment of non-small cell lung cancer.
  • the bioactive agent is a CD20 inhibitor.
  • CD20 inhibitors include obinutuzumab, rituximab, fatumumab, ibritumomab, tositumomab, and ocrelizumab.
  • the bioactive agent is a JAK3 inhibitor.
  • JAK3 inhibitors include tasocitinib.
  • the bioactive agent is a BCL-2 inhibitor.
  • BCL-2 inhibitors include venetoclax, ABT-199 (4-[4-[[2-(4-Chlorophenyl)-4,4-dimethylcyclohex-l- en-l-yl]methyl]piperazin-l-yl]-N-[[3-nitro-4-[[(tetrahydro-2H-pyran-4- yl)methyl]amino]phenyl]sulfonyl]-2-[(lH- pyrrolo[2,3-b]pyridin-5-yl)oxy]benzamide), ABT- 737 (4-[4-[[2-(4-chlorophenyl)phenyl]methyl]piperazin-l-yl]-N-[4-[[(2R)-4-
  • a treatment regimen comprising the administration of Compound 1 Pattern lin combination with at least one additional chemotherapeutic agent.
  • the combinations disclosed herein can be administered for beneficial, additive, or synergistic effect in the treatment of abnormal cellular proliferative disorders.
  • the treatment regimen includes the administration of isolated Compound 1 Pattern 1 in combination with at least one kinase inhibitor.
  • the at least one kinase inhibitor is selected from a phosphoinositide 3 -kinase (PI3K) inhibitor, a Bruton’s tyrosine kinase (BTK) inhibitor, or a spleen tyrosine kinase (Syk) inhibitor, or a combination thereof.
  • PI3K phosphoinositide 3 -kinase
  • BTK Bruton’s tyrosine kinase
  • Syk spleen tyrosine kinase
  • PI3k inhibitors that may be used in the present invention are well known.
  • PI3 kinase inhibitors include but are not limited to Wortmannin, demethoxyviridin, perifosine, idelalisib, Pictilisib, Palomid 529, ZSTK474, PWT33597, CUDC-907, and AEZS-136, duvelisib, GS-9820, BKM120, GDC-0032 (Taselisib), (2-[4-[2-(2-Isopropyl-5-methyl-l,2,4- triazol-3 -yl)-5,6-dihydroimidazo[ 1 ,2-d] [ 1 ,4]benzoxazepin-9-yl]pyrazol- 1 -yl]-2- methylpropanamide), MLN-1117 ((2R)-l-Phenoxy-2-butanyl hydrogen (S)- methylphosphonate; or Methyl
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of alpelisib for the treatment of solid tumors.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of alpelisib for the treatment of abnormal tissue of the female reproductive system. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of alpelisib for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of copanlisib hydrochloride (Aliqopa) for the treatment of lymphoma.
  • Aliqopa copanlisib hydrochloride
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of copanlisib hydrochloride (Aliqopa) for the treatment of follicular lymphoma.
  • Aliqopa copanlisib hydrochloride
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of idelalisib (Zydelig) for the treatment of chronic lymphocytic leukemia.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of idelalisib (Zydelig) for the treatment of Non-Hodgkin lymphoma, including follicular B-cell non-Hodgkin lymphoma or small lymphocytic lymphoma.
  • Zydelig idelalisib
  • BTK inhibitors for use in the present invention are well known.
  • BTK inhibitors include ibrutinib (also known as PCI-32765)(ImbruvicaTM)(l-[(3R)-3-[4-amino-3- (4-phenoxy-phenyl)pyrazolo[3 ,4-d]pyrimidin- 1 -yljpiperidin- 1 -yl]prop-2-en- 1 -one), dianilinopyrimidine-based inhibitors such as AVL-101 and AVL-291/292 (N-(3-((5-fluoro-2- ((4-(2-methoxyethoxy)phenyl)amino)pyrimidin-4-yl)amino)phenyl)acrylamide) (Avila
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ibrutinib (Imbruvica) for the treatment of chronic lymphocytic leukemia.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ibrutinib (Imbruvica) for the treatment of lymphoma, including small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, or Waldenstrom macroglobulinemia.
  • Imbruvica an effective amount of ibrutinib for the treatment of lymphoma, including small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, or Waldenstrom macroglobulinemia.
  • Syk inhibitors for use in the present invention are well known, and include, for example, Cerdulatinib (4-(cyclopropylamino)-2-((4-(4-(ethylsulfonyl)piperazin-l- yl)phenyl)amino)pyrimidine-5-carboxamide), entospletinib (6-(lH-indazol-6-yl)-N-(4- morpholinophenyl)imidazo[l,2-a]pyrazin-8-amine), fostamatinib ([6-( ⁇ 5-Fluoro-2-[(3,4,5- trimethoxyphenyl)amino]-4-pyrimidinyl ⁇ amino)-2,2-dimethyl-3-oxo-2,3-dihydro-4H- pyrido[3,2-b][l,4]oxazin-4-yl]methyl dihydrogen phosphate), fostamatinib disodium salt (s
  • an effective amount of the isolated Compound 1 Pattern 1 is combined in a single dosage form with the Syk inhibitor.
  • the at least one additional chemotherapeutic agent is a protein cell death- 1 (PD-1) inhibitor.
  • PD-1 inhibitors are known in the art, and include, for example, nivolumab (BMS), pembrolizumab (Merck), pidilizumab (CureTech/Teva), AMP -244 (Amplimmune/GSK), BMS-936559 (BMS), and MEDI4736 (Roche/Genentech).
  • BMS protein cell death- 1
  • PD-1 inhibitors include, for example, nivolumab (BMS), pembrolizumab (Merck), pidilizumab (CureTech/Teva), AMP -244 (Amplimmune/GSK), BMS-936559 (BMS), and MEDI4736 (Roche/Genentech).
  • an effective amount of the isolated Compound 1 Pattern 1 is combined in a single dosage form with the PD-1 inhibitor.
  • the at least one additional chemotherapeutic agent is
  • BCL-2 inhibitors are known in the art, and include, for example, ABT-199 (4-[4-[[2-(4-Chlorophenyl)-4,4-dimethylcyclohex-l-en-l- yl]methyl]piperazin-l-yl]-N-[[3-nitro-4-[[(tetrahydro-2H-pyran-4- yl)methyl]amino]phenyl]sulfonyl]-2-[(lH- pyrrolo[2,3-b]pyridin-5-yl)oxy]benzamide), ABT- 737 (4-[4-[[2-(4-chlorophenyl)phenyl]methyl]piperazin-l-yl]-N-[4- [[(2R)-4-
  • a combination described herein can be further combined with an additional therapeutic to treat the cancer.
  • the second therapy can be an immunotherapy.
  • an effective amount of the isolated Compound 1 Pattern 1 can be conjugated to an antibody, radioactive agent, or other targeting agent that directs the compound to the diseased or abnormally proliferating cell.
  • the combination is used in combination with another pharmaceutical or a biologic agent (for example an antibody) to increase the efficacy of treatment with a combined or a synergistic approach.
  • combination can be used with T-cell vaccination, which typically involves immunization with inactivated autoreactive T cells to eliminate a cancer cell population as described herein.
  • the combination is used in combination with a bispecific T-cell Engager (BiTE), which is an antibody designed to simultaneously bind to specific antigens on endogenous T cells and cancer cells as described herein, linking the two types of cells.
  • BiTE bispecific T-cell Engager
  • the bioactive agent is a MEK inhibitor.
  • MEK inhibitors are well known, and include, for example, trametinib/GSK1120212 (N-(3- ⁇ 3-Cyclopropyl-5-[(2-fluoro- 4-iodophenyl)amino]-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydropyrido[4,3-d]pyrimidin- l(2H-yl ⁇ phenyl)acetamide), selumetinib (6-(4-bromo-2-chloroanilino)-7-fluoro-N-(2- hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide), pimasertib/AS703026/MSC
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of binimetinib for the treatment of melanoma, including BRAF-mutant melanoma and NRAS-mutant melanoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of cobimetinib (Cotellic) for the treatment of melanoma, including BRAF-mutant melanoma and NRAS-mutant melanoma.
  • cobimetinib Cotellic
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of binimetinib for the treatment of ovarian cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of selumetinib for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of selumetinib for the treatment of thyroid cancer. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of trametinib (Mekinist) for the treatment of thyroid cancer.
  • Mekinist trametinib
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of trametinib (Mekinist) for the treatment of melanoma.
  • Mekinist trametinib
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of trametinib (Mekinist) for the treatment of non-small cell lung cancer.
  • Mokinist trametinib
  • the bioactive agent is a Raf inhibitor.
  • Raf inhibitors include, for example, Vemurafmib (N-[3-[[5-(4-Chlorophenyl)-lH-pyrrolo[2,3-b]pyridin- 3-yl]carbonyl]-2,4-difluorophenyl]-l-propanesulfonamide), sorafenib tosylate (4-[4-[[4- chloro-3-(trifluoromethyl)phenyl]carbamoylamino]phenoxy]-N-methylpyridine-2- carboxamide;4-methylbenzenesulfonate), AZ628 (3-(2-cyanopropan-2-yl)-N-(4-methyl-3-(3- methyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)phenyl)benzamide), NVP-BHG712 (4- methyl-3-(l-methyl-6
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of dabrafenib (Tafmlar) for the treatment of thyroid cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of dabrafenib (Tafmlar) for the treatment of melanoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of dabrafenib (Tafmlar) for the treatment of non-small cell lung cancer. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of encorafenib for the treatment of melanoma, including B RAF -mutant melanoma.
  • the additional therapy is a monoclonal antibody (MAb).
  • MAbs stimulate an immune response that destroys cancer cells. Similar to the antibodies produced naturally by B cells, these MAbs “coat” the cancer cell surface, triggering its destruction by the immune system.
  • bevacizumab targets vascular endothelial growth factor(VEGF), a protein secreted by tumor cells and other cells in the tumor’s microenvironment that promotes the development of tumor blood vessels. When bound to bevacizumab, VEGF cannot interact with its cellular receptor, preventing the signaling that leads to the growth of new blood vessels.
  • VEGF vascular endothelial growth factor
  • cetuximab and panitumumab target the epidermal growth factor receptor (EGFR), and trastuzumab targets the human epidermal growth factor receptor 2 (HER-2).
  • MAbs that bind to cell surface growth factor receptors prevent the targeted receptors from sending their normal growth-promoting signals. They may also trigger apoptosis and activate the immune system to destroy tumor cells.
  • MAbs are the immunoconjugates. These MAbs, which are sometimes called immunotoxins or antibody-drug conjugates, consist of an antibody attached to a cell-killing substance, such as a plant or bacterial toxin, a chemotherapy drug, or a radioactive molecule. The antibody latches onto its specific antigen on the surface of a cancer cell, and the cell-killing substance is taken up by the cell. FDA-approved conjugated MAbs that work this way include ado-trastuzumab emtansine, which targets the HER-2 molecule to deliver the drug DM1, which inhibits cell proliferation, to HER-2 expressing metastatic breast cancer cells.
  • FDA-approved conjugated MAbs that work this way include ado-trastuzumab emtansine, which targets the HER-2 molecule to deliver the drug DM1, which inhibits cell proliferation, to HER-2 expressing metastatic breast cancer cells.
  • Immunotherapies with T cells engineered to recognize cancer cells via bispecific antibodies (bsAbs) or chimeric antigen receptors (CARs) are approaches with potential to ablate both dividing and non/slow-dividing subpopulations of cancer cells.
  • Bispecific antibodies by simultaneously recognizing target antigen and an activating receptor on the surface of an immune effector cell, offer an opportunity to redirect immune effector cells to kill cancer cells.
  • the other approach is the generation of chimeric antigen receptors by fusing extracellular antibodies to intracellular signaling domains. Chimeric antigen receptor-engineered T cells are able to specifically kill tumor cells in a MHC- independent way.
  • the combination can be administered to the subject in further combination with other chemotherapeutic agents. If convenient, the combination described herein can be administered at the same time as another chemotherapeutic agent, in order to simplify the treatment regimen. In some embodiments, the combination and the other chemotherapeutic can be provided in a single formulation.
  • the use of the compounds described herein is combined in a therapeutic regime with other agents.
  • agents may include, but are not limited to, tamoxifen, midazolam, letrozole, bortezomib, anastrozole, goserelin, an mTOR inhibitor, a PI3 kinase inhibitors, dual mTOR-PI3K inhibitors, MEK inhibitors, RAS inhibitors, ALK inhibitors, HSP inhibitors (for example, HSP70 and HSP 90 inhibitors, or a combination thereof), BCL-2 inhibitors, apopototic inducing compounds, ART inhibitors, including but not limited to, MK-2206, GSK690693, Perifosine, (KRX-0401), GDC-0068, Triciribine, AZD5363, Honokiol, PF-04691502, and ipatasertib, Miltefosine; PD-1 inhibitors including but not limited to, Nivolumab,
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ipatasertib for the treatment of breast cancer, including triple negative breast cancer.
  • the bioactive agent is an mTOR inhibitor.
  • mTOR inhibitors include but are not limited to vistusertib and rapamycin and its analogs, everolimus (Afmitor), temsirolimus, ridaforolimus, sirolimus, and deforolimus.
  • MEK inhibitors include but are not limited to tametinib/GSK1120212 (N-(3- ⁇ 3-Cyclopropyl-5-[(2- fluoro-4-iodophenyl)amino]-6,8-dimethyl-2,4,7-trioxo-3,4,6,7-tetrahydropyrido[4,3- d]pyrimidin-l(2H-yl ⁇ phenyl)acetamide), selumetinob (6-(4-bromo-2-chloroanilino)-7-fluoro- N-(2-hydroxyethoxy)-3-methylbenzimidazole-5-carboxamide), pimasertib/AS703026/MSC 1935369 ((S)-N-(2,3-dihydroxypropyl)-3-((2-fluoro-4- iodophenyl)amino)isonicotinamide), XL-518/GDC-0973 (l-( ⁇ (
  • the bioactive agent is a RAS inhibitor.
  • RAS inhibitors include but are not limited to Reolysin and siG12D LODER.
  • the bioactive agent is an ALK inhibitor.
  • ALK inhibitors include but are not limited to Crizotinib, AP26113, and LDK378.
  • the bioactive agent is a HSP inhibitor.
  • HSP inhibitors include but are not limited to Geldanamycin or 17-N-Allylamino-17-demethoxygeldanamycin (17AAG), and Radicicol.
  • a compound described herein is administered in combination with letrozole and/or tamoxifen.
  • Other chemotherapeutic agents that can be used in combination with the compounds described herein include, but are not limited to, chemotherapeutic agents that do not require cell cycle activity for their anti -neoplastic effect.
  • Additional bioactive compounds include, for example, everolimus, trabectedin, abraxane, TLK 286, AV-299, DN-101, pazopanib, GSK690693, RTA 744, ON 0910.Na, AZD 6244 (ARRY-142886), AMN-107, TKI-258, GSK461364, AZD 1152, enzastaurin, vandetanib, ARQ-197, MK-0457, MLN8054, PHA-739358, R-763, AT-9263, a FLT-3 inhibitor, a VEGFR inhibitor, an aurora kinase inhibitor, a PIK-1 modulator, an HD AC inhibitor, a c-MET inhibitor, a PARP inhibitor, a Cdk inhibitor, an IGFR-TK inhibitor, an anti- HGF antibody, a focal adhesion kinase inhibitor, a Map kinase (mek) inhibitor, a VEGF trap antibody, pemetrexed
  • an effective amount of the isolated Compound 1 Pattern 1 described herein can be combined with a PARP inhibitor selected from niraparib tosylate monohydrate (Zejula), olaparib (Lynparza), rucaparib camsylate (Rubraca), and talazoparib.
  • a PARP inhibitor selected from niraparib tosylate monohydrate (Zejula), olaparib (Lynparza), rucaparib camsylate (Rubraca), and talazoparib.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of niraparib tosylate monohydrate (Zejula) for the treatment of abnormal tissue of the female reproductive system, including ovarian epithelial cancer or fallopian tube cancer.
  • Zejula niraparib tosylate monohydrate
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of niraparib tosylate monohydrate (Zejula) for the treatment of peritoneal cancer.
  • Zejula niraparib tosylate monohydrate
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of olaparib (Lynparza) for the treatment of abnormal tissue of the female reproductive system, including breast cancer, ovarian cancer, ovarian epithelial cancer or fallopian tube cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of olaparib (Lynparza) for the treatment of BRAC1 or BRAC2-mutated breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of olaparib (Lynparza) for the treatment of HER2- breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of olaparib (Lynparza) for the treatment of peritoneal cancer. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of rucaparib camsylate (Rubraca) for the treatment of abnormal tissue of the female reproductive system, including breast cancer, ovarian cancer, ovarian epithelial cancer or fallopian tube cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of rucaparib camsylate (Rubraca) for the treatment of peritoneal cancer.
  • Rubraca rucaparib camsylate
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of talazoparib for the treatment of abnormal tissue of the female reproductive system, including breast cancer, ovarian cancer, ovarian epithelial cancer or fallopian tube cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of talazoparib for the treatment of BRAC1 or BRAC2- mutated breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of olaratumab for the treatment of soft tissue sarcoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of savolitinib for the treatment of adenocarcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of savolitinib for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of savolitinib for the treatment of renal cell carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of vistusertib for the treatment of advanced breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of vistusertib for the treatment of advanced breast cancer.
  • an effective amount of the isolated Compound 1 Pattern 1 described herein can be combined with a chemotherapeutic selected from, but are not limited to, Imatinib mesylate (Gleevac®), Dasatinib (Sprycel®), Nilotinib (Tasigna®), Bosutinib (Bosulif®), Trastuzumab (Herceptin®), Pertuzumab (PerjetaTM), Lapatinib (Tykerb®), Gefitinib (Iressa®), Erlotinib (Tarceva®), Cetuximab (Erbitux®), Panitumumab (Vectibix®), Vandetanib (Caprelsa®), Vemurafenib (Zelboraf®), Vorinostat (Zolinza®), Romidepsin (Istodax®), Bexarotene (Tagretin®), Alitretinoin (Panretin®),
  • an effective amount of the isolated Compound 1 Pattern 1 described herein can be combined with a CD4/6 inhibitor including abemaciclib (Versenio), palbociclib (Ibrance), or trilaciclib.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of abemaciclib (Versenio) for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of abemaciclib (Versenio) for the treatment of HR+ HER2- breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of palbociclib (Ibrance) for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of palbociclib (Ibrance) for the treatment of HR+ HER2- breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of palbociclib (Ibrance) for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of palbociclib (Ibrance) for the treatment of metastatic triple negative breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of palbociclib (Ibrance) for the treatment of small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of cabozantinib S-malate (CometriqTM) for the treatment of thyroid cancer. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of cabozantinib S-maleate (CometriqTM) for the treatment of renal cell carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of dasatinib (Sprycel) for the treatment of leukemia, including acute lymphoblastic leukemia or chronic myelogenous leukemia.
  • leukemia including acute lymphoblastic leukemia or chronic myelogenous leukemia.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of dasatinib (Sprycel) for the treatment of prostate cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of Erlotinib (Tarceva®) for the treatment of prostate cancer.
  • Tarceva® Erlotinib
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of Gefitinib (Iressa®) for the treatment of prostate cancer.
  • Gefitinib Iressa®
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of imatinib mesylate (Gleevec) for the treatment of leukemia, including acute lymphoblastic leukemia, chronic eosinophilic leukemia, hypereosinophilic syndrome, or chronic myelogenous leukemia.
  • leukemia including acute lymphoblastic leukemia, chronic eosinophilic leukemia, hypereosinophilic syndrome, or chronic myelogenous leukemia.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of trastuzumab (Herceptin) for the treatment of adenocarcinoma.
  • trastuzumab Herceptin
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of trastuzumab (Herceptin) for the treatment of breast cancer, including HER2+ breast cancer.
  • trastuzumab Herceptin
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of imatinib mesylate (Gleevec) for the treatment of tumors, including but not limited to dermatofibrosarcoma protuberans and gastrointestinal stromal tumors.
  • Tumors including but not limited to dermatofibrosarcoma protuberans and gastrointestinal stromal tumors.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of imatinib mesylate (Gleevec) for the treatment of myelodysplastic/myeloproliferative neoplasms. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of imatinib mesylate (Gleevec) for the treatment of systemic mastocytosis.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of nilotinib (Tasigna) for the treatment of chronic myelogenous leukemia, including Philadelphia chromosome positive chronic myeloid leukemia (Ph+ CML).
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of pazopanib hydrochloride (Votrient) for the treatment of renal cell carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of pazopanib hydrochloride (Votrient) for the treatment of soft tissue sarcoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of regorafenib (Stivarga) for the treatment of colorectal cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of regorafenib (Stivarga) for the treatment of gastrointestinal stromal tumor.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of regorafenib (Stivarga) for the treatment of hepatocellular carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of sorafenib Tosylate (Nexavar) for the treatment of carcinoma, including hepatocellular carcinoma or renal cell carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of sunitinib malate (Sutent) for the treatment of gastrointestinal stromal tumor.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of sunitinib malate (Sutent) for the treatment of pancreatic cancer. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of sunitinib malate (Sutent) for the treatment of renal cell carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of vemurafenib (Zelboraf) for the treatment of Erdheim- Chester disease.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of vemurafenib (Zelboraf) for the treatment of melanoma.
  • the additional therapeutic agent is an anti-inflammatory agent, a chemotherapeutic agent, a radiotherapeutic, additional therapeutic agents, or immunosuppressive agents.
  • Suitable chemotherapeutic agents include, but are not limited to, radioactive molecules, toxins, also referred to as cytotoxins or cytotoxic agents, which includes any agent that is detrimental to the viability of cells, agents, and liposomes or other vesicles containing chemotherapeutic compounds.
  • General anticancer pharmaceutical agents include: Vincristine (Oncovin®) or liposomal vincristine (Marqibo®), Daunorubicin (daunomycin or Cerubidine®) or doxorubicin (Adriamycin®), Cytarabine (cytosine arabinoside, ara-C, or Cytosar®),
  • L-asparaginase (Elspar®) or PEG-L-asparaginase (pegaspargase or Oncaspar®), Etoposide (VP- 16), Teniposide (Vumon®), 6-mercaptopurine (6-MP or Purinethol®), Methotrexate, Cyclophosphamide (Cytoxan®), Prednisone, Dexamethasone (Decadron), imatinib (Gleevec®), dasatinib (Sprycel®), nilotinib (Tasigna®), bosutinib (Bosulif®), and ponatinib (IclusigTM).
  • additional suitable chemotherapeutic agents include but are not limited to
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of bosutinib (Bosulif®) for the treatment of chronic myelogenous leukemia (CML).
  • Bosulif® bosutinib
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ponatinib hydrochloride (Iclusig) for the treatment of leukemia, including acute lymphoblastic leukemia and chronic myelogenous leukemia.
  • Iclusig ponatinib hydrochloride
  • Additional therapeutic agents that can be administered in combination with a compound disclosed herein can include bevacizumab, sutinib, sorafenib, 2-methoxyestradiol or 2ME2, fmasunate, vatalanib, vandetanib, aflibercept, volociximab, etaracizumab (MEDI-522), cilengitide, erlotinib, cetuximab, panitumumab, gefitinib, trastuzumab, dovitinib, figitumumab, atacicept, rituximab, alemtuzumab, aldesleukine, atlizumab, tocilizumab, temsirolimus, everolimus, lucatumumab, dacetuzumab, HLL1, huN901-DMl, atiprimod, natalizumab, bortezomib, carfilzomib
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of HR+, HER2- breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of pancreatic cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of gastrointestinal cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of renal cell carcinoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of everolimus (Afmitor) for the treatment of astrocytoma, including subependymal giant cell astrocytoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of fulvestrant (Faslodex) for the treatment of breast cancer.
  • fulvestrant Fraslodex
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of fulvestrant (Faslodex) for the treatment of HR+, HER2- breast cancer.
  • fulvestrant Fraslodex
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ramucirumab for the treatment of adenocarcinoma. In one embodiment, an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ramucirumab for the treatment of non-small cell lung cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ramucirumab for the treatment of colorectal cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ribociclib (Kisqali) for the treatment of breast cancer.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ribociclib (Kisqali) for the treatment of HR+ and HER2- breast cancer.
  • a compound described herein can be combined with at least one IDHl or IDH2 inhibitor.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of enasidenib mesylate (Idhifa) for the treatment of acute myeloid leukemia.
  • a compound described herein can be combined with at least one fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor.
  • FGFR fibroblast growth factor receptor
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of erdafitinib for the treatment of urothelial cancer, including metastatic urothelial cancer.
  • a compound described herein can be combined with at least one ERK inhibitor.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of SCH772984 for the treatment of melanoma, including B RAF -mutant melanoma or NRAS-mutant melanoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ulixertinib for the treatment of melanoma, including uveal melanoma.
  • an effective amount of Compound 1 Pattern 1 is administered in combination with an effective amount of ulixertinib for the treatment of pancreatic cancer.
  • a compound described herein can be combined with at least one immunosuppressive agent.
  • the immunosuppressive agent is preferably selected from the group consisting of a calcineurin inhibitor, e.g. a cyclosporin or an ascomycin, e.g. Cyclosporin A (NEORAL®), FK506 (tacrolimus), pimecrolimus, a mTOR inhibitor, e.g. rapamycin or a derivative thereof, e.g.
  • Sirolimus (RAPAMUNE®), Everolimus (Certican®), temsirolimus, zotarolimus, biolimus-7, biolimus-9, a rapalog, e.g., ridaforolimus, azathioprine, campath 1H, a SIP receptor modulator, e.g. fmgolimod or an analogue thereof, an anti-IL-8 antibody, mycophenolic acid or a salt thereof, e.g. sodium salt, or a prodrug thereof, e.g.
  • Mycophenolate Mofetil (CELLCEPT®), OKT3 (ORTHOCLONE OKT3®), Prednisone, ATGAM®, THYMOGLOBULIN®, Brequinar Sodium, OKT4, T10B9.A-3A, 33B3.1, 15-deoxyspergualin, tresperimus, Leflunomide ARAVA®, CTLAI-Ig, anti-CD25, anti-IL2R, Basiliximab (SIMULECT®), Daclizumab (ZENAPAX®), mizorbine, methotrexate, dexamethasone, ISAtx-247, SDZ ASM 981 (pimecrolimus, Elidel®), CTLA41g (Abatacept), belatacept, LFA31g courts etanercept (sold as Enbrel® by Immunex), adalimumab (Humira®), infliximab (Remicade®), an anti-LFA-1 antibody, natal
  • a compound described herein is administered to the subject prior to treatment with another chemotherapeutic agent, during treatment with another chemotherapeutic agent, after administration of another chemotherapeutic agent, or a combination thereof.
  • an effective amount of the isolated Compound 1 Pattern 1 can be administered to the subject such that the other chemotherapeutic agent can be administered either at higher doses (increased chemotherapeutic dose intensity) or more frequently (increased chemotherapeutic dose density).
  • Dose-dense chemotherapy is a chemotherapy treatment plan in which drugs are given with less time between treatments than in a standard chemotherapy treatment plan.
  • Chemotherapy dose intensity represents unit dose of chemotherapy administered per unit time. Dose intensity can be increased or decreased through altering dose administered, time interval of administration, or both.
  • the compounds described herein can be administered in a concerted regimen with another agent such as a non-DNA-damaging, targeted anti-neoplastic agent or a hematopoietic growth factor agent.
  • another agent such as a non-DNA-damaging, targeted anti-neoplastic agent or a hematopoietic growth factor agent.
  • hematopoietic growth factors can have serious side effects.
  • the use of the EPO family of growth factors has been associated with arterial hypertension, cerebral convulsions, hypertensive encephalopathy, thromboembolism, iron deficiency, influenza like syndromes and venous thrombosis.
  • the G-CSF family of growth factors has been associated with spleen enlargement and rupture, respiratory distress syndrome, allergic reactions and sickle cell complications.
  • G-CSF granulocyte colony stimulating factor
  • Neupogen filamentgrastin
  • Neulasta peg-filgrastin
  • lenograstin granulocyte- macrophage colony stimulating factor
  • GM-CSF granulocyte- macrophage colony stimulating factor
  • M-CSF macrophage colony stimulating factor
  • thrombopoietin megakaryocyte growth development factor (MGDF), for example sold as Romiplostim and Eltrombopag
  • IL interleukin
  • interleukin-3 interleukin-3
  • interleukin- 11 adipogenesis inhibiting factor or oprelvekin
  • SCF stem cell factor, steel factor, kit-ligand, or KL
  • erythropoietin EPO
  • an effective amount of the isolated Compound 1 Pattern 1 is administered prior to administration of the hematopoietic growth factor.
  • the hematopoietic growth factor administration is timed so that the compound’s effect on HSPCs has dissipated.
  • the growth factor is administered at least 20 hours after the administration of a compound described herein.
  • multiple doses of a compound described herein can be administered to the subject.
  • the subject can be given a single dose of a compound described herein.
  • a compound disclosed herein can be beneficially administered in combination with any therapeutic regimen entailing radiotherapy, chemotherapy, or other therapeutic agents.
  • the compounds disclosed herein can be beneficially administered in combination with therapeutic agents targeting auto-immune disorders.
  • XRPD analysis was carried out on a PANalytical X’pert pro, scanning the samples between 3 and 35° 20.
  • the material was gently grounded to release any agglomerates and loaded onto a multi-well plate with Kapton or Mylar polymer film to support the sample.
  • the above technique was used to generate the images in FIG. 1, FIG. 7, FIG. 8 through FIG. 19, FIG. 21 through FIG. 32, FIG. 36, FIG. 38, FIG. 42, FIG. 43, FIG. 49 through FIG. 59, FIG 61, FIG. 67 through FIG. 69, and FIG. 89.
  • Table 1 below provides the XRPD Peak List for Pattern 1 with >10% relative intensity peaks.
  • the XRPD performed on Pattern 1 exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 1 at about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.3 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 24.0 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°. Table 1. XRPD Peak List for Pattern 1
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 2 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 3 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 4 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 5 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 6 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 7 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 8 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 9 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising at least 10 peaks selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 1 and is characterized by an XRPD pattern comprising the 2theta values selected from about 9.6 ⁇ 0.2°, about 12.2 ⁇ 0.2°, about 15.3 ⁇ 0.2°, about 17.6 ⁇ 0.2°, about 19.3 ⁇ 0.2°, about 19.8 ⁇ 0.2°, about 21.2 ⁇ 0.2°, about 22.7 ⁇ 0.2°, about 23.9 ⁇ 0.2°, about 26.1 ⁇ 0.2°, and about 28.6 ⁇ 0.2°.
  • Table 2 below provides the results of the XRPD performed on Pattern 2.
  • the XRPD exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 2 at 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 4 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 5 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 6 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 7 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 8 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising at least 9 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • the morphic form is Pattern 2 and is characterized by an XRPD pattern comprising the 2theta values selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.3 ⁇ 0.2°, 17.2 ⁇ 0.2°, 18.2 ⁇ 0.2°, 19.8 ⁇ 0.2°, 20.4 ⁇ 0.2°, 20.6 ⁇ 0.2°, 26.4 ⁇ 0.2°, and 27.3 ⁇ 0.2°.
  • Table 4 below provides the results of the XRPD performed on Pattern 5. Significant peaks were observed in the XRPD on Pattern 5 at 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°, 16.2 ⁇ 0.2°, 26.5 ⁇ 0.2°, 27.4 ⁇ 0.2°, 27.8 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • Table 3 below provides the results of the XRPD performed on Pattern 3 scale-up.
  • the XRPD exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 3 scale-up at about 8.1 ⁇ 0.2°, about 14.8 ⁇ 0.2°, about 16.5 ⁇ 0.2°, about 18.2 ⁇ 0.2°, about 19.7 ⁇ 0.2°, about 24.8 ⁇ 0.2°, about
  • the morphic form is Pattern 3 is characterized by an XRPD pattern comprising at least 2 peaks selected from 8.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 16.5 ⁇ 0.2°, 18.1 ⁇ 0.2°, 19.7 ⁇ 0.2°, 24.84 ⁇ 0.2°, and 27.7 ⁇ 0.2°. In certain embodiments, the morphic form is Pattern 3 is characterized by an XRPD pattern comprising at least 3 peaks selected from 8.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 16.5 ⁇ 0.2°, 18.1 ⁇ 0.2°,
  • the morphic form is Pattern 3 is characterized by an XRPD pattern comprising at least 4 peaks selected from 8.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 16.5 ⁇ 0.2°, 18.1 ⁇ 0.2°,
  • the morphic form is Pattern 3 is characterized by an XRPD pattern comprising at least 5 peaks selected from 8.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 16.5 ⁇ 0.2°, 18.1 ⁇ 0.2°,
  • the morphic form is Pattern 3 is characterized by an XRPD pattern comprising at least 6 peaks selected from 8.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 16.5 ⁇ 0.2°, 18.1 ⁇ 0.2°,
  • the morphic form is Pattern 3 is characterized by an XRPD pattern comprising the 2theta values selected from 8.1 ⁇ 0.2°, 14.8 ⁇ 0.2°, 16.5 ⁇ 0.2°, 18.1 ⁇ 0.2°,
  • Table 4 below provides the results of the XRPD performed on Pattern 4.
  • the XRPD exhibited sharp peaks, indicating the sample was composed of crystalline material.
  • Tabulated below are peaks for Pattern 4 >10% relative intensity.
  • Significant peaks were observed in the XRPD of Pattern 4 at about 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°, 16.2 ⁇ 0.2°, 26.5 ⁇ 0.2°, 27.4 ⁇ 0.2°, 27.8 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°,
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°,
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 4 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°, 16.2 ⁇ 0.2°, 26.5 ⁇ 0.2°, 27.4 ⁇ 0.2°, 27.8 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 5 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°,
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 6 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°,
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 7 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°,
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising at least 8 peaks selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°, 16.2 ⁇ 0.2°, 26.5 ⁇ 0.2°, 27.4 ⁇ 0.2°, 27.8 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • the morphic form is Pattern 5 and is characterized by an XRPD pattern comprising the 2theta values selected from 6.3 ⁇ 0.2°, 6.8 ⁇ 0.2°, 8.1 ⁇ 0.2°, 11.2 ⁇ 0.2°, 16.2 ⁇ 0.2°, 26.5 ⁇ 0.2°, 27.4 ⁇ 0.2°, 27.8 ⁇ 0.2°, and 28.6 ⁇ 0.2°.
  • Table 5 below provides the results of the XRPD performed on Pattern 6 exhibited sharp peaks, indicating the sample was composed of crystalline material. Tabulated peak picks for Pattern 6 >10% relative intensity. Significant peaks were observed in the XRPD on Pattern 6 at about 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.26 ⁇ 0.2°, 26.3 ⁇ 0.2°, and 27.2 ⁇ 0.2°.
  • the morphic form is Pattern 6 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.26 ⁇ 0.2°, 26.3 ⁇ 0.2°, and 27.2 ⁇ 0.2°. In certain embodiments, the morphic form is Pattern 6 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.26 ⁇ 0.2°, 26.3 ⁇ 0.2°, and 27.2 ⁇ 0.2°.
  • the morphic form is Pattern 6 and is characterized by an XRPD pattern comprising at least 4 peaks selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.26 ⁇ 0.2°, 26.3 ⁇ 0.2°, and 27.2 ⁇ 0.2°.
  • the morphic form is Pattern 6 and is characterized by an XRPD pattern comprising the 2theta values selected from 6.7 ⁇ 0.2°, 11.1 ⁇ 0.2°, 16.26 ⁇ 0.2°, 26.3 ⁇ 0.2°, and 27.2 ⁇ 0.2°.
  • Table 6 below provides tabulated GVS data for Pattern 1 showing the difference in water vapor uptake between the sorption and desorption isotherms. The weight change during the sorption/desorption cycles were plotted, allowing for the hygroscopic nature of the sample to be determined.
  • the sample was subjected to a ramping profile from 40 - 90% relative humidity (RH) at 10% increments. In certain embodiments, at least two sorption cycles are performed.
  • three sorption cycles are performed.
  • Table 7 below provides the results of the GVS data for the mixture of Pattern 2 and Pattern 6, showing the difference in water vapor uptake between the sorption and desorption isotherms. The weight change during the sorption/desorption cycles were plotted, allowing for the hygroscopic nature of the sample to be determined. Table 7. GVS data of Pattern 2 and Pattern 6 mixture
  • the sample was subjected to a ramping profile from 40 - 90% relative humidity (RH) at 10% increments. In certain embodiments, at least two sorption cycles are performed.
  • three sorption cycles are performed.
  • Table 8 Provides results for the tabulated DVS data of Pattern 3 scale-up.
  • the material appeared hygroscopic by DVS with a mass increase of ca. 4 % at 90 % RH. During the desorption cycles the material loses mass, likely due to the loss of surface water present in the sample and the crystallization of amorphous content present in the sample. The material adsorbed 2.5 wt% moisture at 40% RH, 3.1 wt% moisture at 60% and 3.5 wt% at 90% RH.
  • At least two sorption cycles are performed. In certain embodiments, three sorption cycles are performed.
  • XRPD analysis was carried out on a PANalytical X’pert pro, scanning the samples between 3 and 35° 20.
  • the material was gently ground to release any agglomerates and loaded onto a multi-well plate with Kapton or Mylar polymer film to support the sample.
  • the above technique was used to generate the images in FIG. 13, FIG. 20 and FIG. 27.
  • Table 9 provides the results of the XRPD performed on Pattern 4 exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 4 at 6.1 ⁇ 0.2°, 15.0 ⁇ 0.2°, 27.5 ⁇ 0.2°, and 28.1 ⁇ 0.2°.
  • the morphic form is Pattern 4 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 6.1 ⁇ 0.2°, 15.0 ⁇ 0.2°, 27.5 ⁇ 0.2°, and 28.1 ⁇ 0.2°. In certain embodiments, the morphic form is Pattern 4 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 6.1 ⁇ 0.2°, 15.0 ⁇ 0.2°, 27.5 ⁇ 0.2°, and 28.1 ⁇ 0.2°.
  • the morphic form is Pattern 4 and is characterized by an XRPD pattern comprising the 2theta values selected from 6.1 ⁇ 0.2°, 15.0 ⁇ 0.2°, 27.5 ⁇ 0.2°, and 28.1 ⁇ 0.2°.
  • PLM imaging was carried out in the presence of crystallinity (birefringence). PLM imaging was determined using an Olympus BX50 polarizing microscope, equipped with a Motic camera and image capture software (Motic Images Plus 2.0). All images were recorded using the 20x objective, unless otherwise stated. The above technique was used to generate the images in FIG. 2, FIG. 37, FIG. 44, FIG. 62, and FIG. 74 through FIG. 86.
  • XRPD analysis was carried out on a PANalytical X’pert pro, scanning the samples between 3 and 35° 20.
  • the material was gently grounded to release any agglomerates and loaded onto a multi-well plate with Kapton or Mylar polymer film to support the sample.
  • the post-DVS samples were analyzed by XRPD. After DVS was performed, samples were analyzed by XRPD to confirm any changes in the material. The above technique was used to generate the images in FIG. 7, FIG. 42 and FIG. 49.
  • Example 11 Various solvent solubility systems
  • the solvent solubility experiment was carried out using various solvents to determine solid formation, as provided in Tables 11, 12 and 13.
  • a known volume aliquot typically 5 volumes
  • solvent was added to approximately 10 mg of amorphous Compound 1 as a dihydrochloride.
  • the mixture was checked for dissolution and where no dissolution was apparent, the mixture was heated to ca. 40 °C and checked again. This procedure was continued until dissolution was observed or until 100 volumes of solvent had been added. All observed solids were isolated and analyzed by XRPD.
  • solvents include, but are not limited to, 1,4 dioxane, methanol, ethanol, propanol, butanol, butanone, ethoxy ethanol, trifluoroethanol, methyl-THF, acetone, acetonitrile, anisole, chlorobenzene, DMSO, DMF, cumene, dichloromethane, ethyl acetate, ethylene glycol, heptane, isopropyl acetate, nitromethane, n-methylpyrrolidone, MIBK, t- BME, tetrahydrofuran, toluene, water and mixtures thereof.
  • the above technique was used to generate the images in FIG. 8, FIG. 9, FIG. 10, FIG. 11, FIG. 12, FIG. 14. Table 11. Approximate solubility results from solvent screen
  • the antisolvent addition experiments were carried out using selected anti-solvent (for example t-BME) layered over the material solution in each sample until precipitation was observed or 1 mL of anti-solvent was added.
  • the samples were stored in a fridge at ca. 5 °C to promote precipitation and any observed solids were collected and analyzed by XRPD, as provided in Tables 14 and 15.
  • Material solution were prepared using solvent systems listed in Example 13. The above technique was used to generate the images in FIG. 32.
  • Pattern 2 was scaled up via the addition of 100 pL aliquots of MEK to approximately 500 mg of amorphous Compound 1 as a di HC1 salt until a mobile slurry was formed. The slurry was then temperature cycled between ambient and 40 °C for approximately 72 hours. The sample was collected, and an aliquot of the slurry was analyzed by XRPD. The solid was then isolated and dried under vacuum at 40 °C for approximately 4 hours. The resulting material was a mixture of Pattern 2 and Pattern 6. Pattern 3 was scaled up via the addition of 100 pL aliquots of ethanol to approximately
  • the solid was then isolated and dried under vacuum at 40 °C for approximately 4 hours. This resulting material was then slurried in 10 mL of methyl isobutyl ketone for another twenty -four hours. An aliquot of the slurry was analyzed by XRPD. The solid was then isolated and dried under vacuum at 40 °C for approximately four hours. The dried material was analyzed by XRPD.
  • Pattern 7 solids were carried out on Pattern 7 solids at pH 2 to pH 13.
  • a total of 12 vials contained Pattern 7 (100 mg) and 500 pL of 66 % DMSO : 33 % THF (% v/v), in order to make a slurry.
  • the starting pH of each slurry was measured using a pH meter.
  • the pH of each vial was adjusted to the required pH using aliquots of acetic acid or 1M sodium hydroxide solution. If dissolution was observed at a selected pH, additional Pattern 7 solid was added to the vial, until a slurry persisted or a maximum of 500 mg had been added. The pH was then readjusted as required.
  • Pattern 7 Small scale crystallization trials were carried out on Pattern 7 with Set 1 to determine the effect of altering the starting concentration of Pattern 7 in DMSO/THF solvent systems, as provided in Tables 16 and 17.
  • Several concentrations of Pattern 7 in DMSO/THF were prepared, with starting concentrations of 100, 150 and 200 mg/mL respectively.
  • Samples were prepared in a volume of 66 % DMSO: 33 % THF (% v/v) and Pattern 7 (200 mg) was added as the appropriate starting concentration.
  • Acetic acid 100 pL aliquots was added at ambient temperature to each sample, until dissolution was achieved.
  • solid was crystallized from solution by using a) a 1 : 1 equivalent ratio of acetic acid to base (1M sodium hydroxide solution), or b) adjusting the final pH to 12 using 1M sodium hydroxide.
  • the pH of the solution was measured using a pH meter and adjusted to pH 12 using 1M sodium hydroxide solution.
  • the sample sets were stirred at ambient temperature for ca. 18 hours with agitation provided via a magnetic stirrer plate. After ca. 18 hours, stirring was stopped and the supernatants were filtered using 0.45 pm PVDF needle filters and syringes. The concentration of the supernatants was analyzed by HPLC.
  • Samples (1 and 2) was prepared by adding Pattern 7 (200 mg) with DMSO (3 mL) and THF (1.6 mL) for a slurry to be formed. The resulting slurries were heated to 70 °C using a temperature-controlled block. Acetic acid was added until dissolution was achieved. The experiments were held for 1 hour at 70 °C. After 1 hour, Sample 1 was added 1M sodium hydroxide solution in 100 pL aliquots, until the experiment reached pH 7. The experiment was then cooled from 70 °C to 25 °C at a rate of 0.25 °C/min. Sample 2 solution was cooled from 70 °C to 25 °C at a rate of 0.25 °C/min.
  • Samples (1 and 2) were prepared by adding Pattern 7 (200 mg) and 1 mL of 66 % DMSO : 33 % THF (% v/v) for a slurry to be formed. Acetic acid was added in 100 pL aliquots at ambient temperature until dissolution was noted. Sodium hydroxide solution (1M) was added in 100 pL aliquots until the pH reached pH 5. The solution was then seeded with 4 mg (2%) of Pattern 7 and the pH was adjusted further with 1M sodium hydroxide solution until pH 7 was achieved. Sample 1 was stirred at ambient temperature for ca.
  • the pH 4.3 solution was prepared by diluting glacial acetic acid (11.6 mL) to 100 mL in deionized water, then adding sodium acetate (1.07 g) and acetic acid solution (5.9 mL) preparing up to 500 mL with deionized water; then the pH was adjusted to 4.2.
  • the above technique was used to generate the images in FIG. 60.
  • a saturated solution of Compound 1 di-HCl was prepared in N,N’-dimethylacetamide at ambient temperature and the solution was then cooled from 60 °C to 25 °C at 0.1 °C per minute. The sample was left at ambient temperature until large crystals were obtained (roughly two weeks). During an attempt to carry out polarized light microscopy, the crystals re-dissolved upon agitation of the solution. The solution was placed in the fridge for one week to encourage crystal growth, but no crystals appeared. Finally, ca. 10 drops of acetone were added as anti solvent and crystals were yielded.
  • a suitable crystal of Pattern 11 was selected and mounted in a loop using paratone oil.
  • Data were collected using a Bruker D8Venture diffractometer equipped with a Photon III detector operating in shutterless mode at 100(2) K with Cu-Ka radiation (1.54178 A).
  • the structure was solved in the 01ex2 software package with the ShelXT (intrinsic phasing) structure solution program and refined with the ShelXL3 refinement package using Least Squares minimization.
  • Data were collected, solved and refined in the triclinic space-group P- 1
  • All non-hydrogen atoms were located in the Fourier map and their positions refined prior to describing the thermal movement of all non-hydrogen atoms anisotropically. Within the structure, two complete G1T28 dihydrochloride formula units were refined. All fully occupied hydrogen atoms were placed in calculated positions using a riding model with fixed Uiso at 1.2 times for all CH, CFh and NH groups and 1.5 times for all C3 ⁇ 4 groups. All partially occupied hydroxyl hydrogen atoms were located in the Fourier map and their positions and occupancy refined.
  • the hemihydrate of Compound 1 as a dihydrochloride was found to be extremely crystalline and possess unit cell dimensions as follows: This asymmetric unit contains two complete Compound 1 formula units and two chloride anions.
  • the solvent mask was calculated and 91.1 electrons were found within a volume of 341.0 A 3 in a void in the unit cell. This corresponds to 45.5 electrons per asymmetric unit and 22.75 electrons per G1T28 formula unit. This is consistent with the presence of one additional chloride anion and a water molecule at 50% occupancy per G1T28 molecule, totaling 44 electrons per asymmetric unit.
  • the solvent mask was removed in order to determine the location of the disordered chlorides which were found within channels intersecting the unit cell at the cell origins.
  • Packing within the structure was shown to be efficient whereby a moderate density of 1.342 g/cm 3 was noted.
  • a solvent mask was applied to a channel along the a-axis which contained the disordered solvent and counterion.
  • a discrete hydrogen bonding motif was noted in the asymmetric unit, as shown in FIG. 90.
  • Diffraction data were collected on XRD1 beamline (Lausi, et al, 2015), in its standard single crystal configuration.
  • the experimental setup consists in a Huber goniometer with k geometry, fully controllable from remote. Samples diffraction properties were characterized at 100K (nitrogen stream supplied through an Oxford Cryostream 700 - Oxford Cryosystems Ltd., Oxford, United Kingdom) through the rotating crystal method.
  • Data were acquired using a monochromatic wavelength of 0.6199 A, on a Pilatus 6M hybrid-pixel area detector (DECTRIS Ltd., Baden-Daettwil, Switzerland). Crystals were dipped in NHV oil (Jena Bioscience, Jena, Germany), mounted at RT on kapton loops (MiTeGen, Ithaca, USA) and flash frozen in liquid nitrogen. Diffraction properties of 15 different crystals have been assessed - all of them appear as thin yellow needles less than 50- 80 mm in the longest direction. Most of specimens diffracted around -0.9-1 A, but few of them diffracted up to -0.7-0.8 A.
  • the structure was solved using direct methods by SHELXT-2014/7 (Sheldrick, G. M., 2015a) and refined by least-square full matrix refinement using SHELXL-2014/7 (Sheldrick, G. M., 2015b). All H-atoms, except for those linked to N or O atoms, were included from the geometry. In the final cycles of refinement, the large peak -2 e/A3 was found nearby C1B atom. This was interpreted as partial disorder of this atom. The refinement revealed that disorder can be established as -0.9 and -0.1. This disorder affects the water molecule (01 W); therefore, to keep the model intact H1W1 was kept with AFIX card (AFIX 1) and thermal parameter at 150% of neighboring O atom.
  • AFIX 1 AFIX 1
  • the HR-XRPD data were collected on D8 Advance diffractometer using Cu Kal radiation (1.54056 A) with germanium monochromator at RT. Diffraction data were collected in the 20 range 2 - 41.5°. Detector scan on solid state LynxEye detector was performed using 0.016° per step with 5 sec/step scan speed. The samples were measured in 8 mm long glass capillary with 0.3 mm outer diameter.
  • the first step of calculation was taken by finding unit cell parameters using LSI-Index (Coelho, 2003; Coelho & Kern, 2005) indexing program.
  • the space group was selected on reflections condition and knowledge of the compound.
  • the cell parameters, purity as well as instrument parameters were refined using Whole Powder Pattern Decomposition method (Pawley, 1981).
  • the cell parameters were taken from the RT measurement while the atom positions and their thermal parameters from single crystal measurement file (cif) at 100K. During the refinement the following parameters were refined: cell constants; background; instrument geometry; zero shift; absorption.
  • FIG. 97 and FIG. 98 The resulting crystal structure is displayed in FIG. 97 and FIG. 98 as well as a simulation comparison in FIG. 99 which shows close agreement between the predicted and experimental diffractograms.
  • Compound 1 dihydrochloride crystallizes as yellow columnar crystals in the monoclinic centrosymmetric space group P21/c.
  • one dicationic API2+ two chloride anions and two water molecules were found (overall ratio 1 :2:2).
  • the molecule is charged on the most basic amine N atom (N2) as well as on the pyridine N atom (N10).
  • the molecule adopts an extended (unfolded) conformation.
  • the crystal is held by intermolecular H-bond interactions (see FIG. 98).
  • One of the chloride anions (CIA) interacts directly with the positively-charged N atom (N2).
  • the second chloride anion (C1B) acts as a bridge between the two water molecules (see FIG. 97).
  • this C1B interacts also through H-bond with another amine N atom (N14).
  • N14 another amine N atom
  • all water molecules act as donors as well as acceptors of H-bonds, while N atoms act preferably as donors only.
  • the only acceptor of H-bond is N16 atom and in this case this interaction is intramolecular.
  • XRPD analysis was carried out on a PANalytical X’pert pro, scanning the free base samples between 3 and 35° 2Q.
  • the material was gently grounded to release any agglomerates and loaded onto a multi-well plate with Kapton or Mylar polymer film to support the sample.
  • Table 37 below provides the XRPD Peak List for Pattern 7 with >10% relative intensity peaks.
  • the XRPD performed on Pattern 7 exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 7 at about 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • Table 37 XRPD Peak List for Pattern 7.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 4 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 5 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 6 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 7 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 8 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 9 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 10 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 11 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising at least 12 peaks selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 15.2 ⁇ 0.2°, 15.6 ⁇ 0.2°, 17.6 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 22.7 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 24.6 ⁇ 0.2°.
  • the morphic form is Pattern 7 and is characterized by an XRPD pattern comprising the 2theta values selected from 5.8 ⁇ 0.2°, 10.9 ⁇ 0.2°, 11.8 ⁇ 0.2°, 18.1 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 21.6 ⁇ 0.2°, 23.3 ⁇ 0.2°, and 25.6 ⁇ 0.2°.
  • Table 38 below provides the XRPD Peak List for Pattern 8 with >10% relative intensity peaks.
  • the XRPD performed on Pattern 8 exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 8 at about 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°, 21.6 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.0 ⁇ 0.2°, 23.5 ⁇ 0.2°, 23.8 ⁇ 0.2°, 24.2 ⁇ 0.2°, 25 ⁇ 0.2°, and 29.9 ⁇ 0.2°. Table 38. XRPD Peak List for Pattern 8.
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°, 21.6 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.0 ⁇ 0.2°, 23.5 ⁇ 0.2°, 23.8 ⁇ 0.2°, 24.2 ⁇ 0.2°, 25 ⁇ 0.2°, and 29.9 ⁇ 0.2°.
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°, 21.6 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.0 ⁇ 0.2°, 23.5 ⁇ 0.2°, 23.8 ⁇ 0.2°, 24.2 ⁇ 0.2°, 25 ⁇ 0.2°, and 29.9 ⁇ 0.2°.
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 5 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°, 21.6 ⁇ 0.2°, 21.9 ⁇ 0.2°, 23.0 ⁇ 0.2°, 23.5 ⁇ 0.2°, 23.8 ⁇ 0.2°, 24.2 ⁇ 0.2°, 25 ⁇ 0.2°, and 29.9 ⁇ 0.2°.
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 7 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 9 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 11 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 13 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 15 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 17 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising at least 19 peaks selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • the morphic form is Pattern 8 and is characterized by an XRPD pattern comprising the 2theta values selected from 4.0 ⁇ 0.2°, 7.4 ⁇ 0.2°, 10.8 ⁇ 0.2°, 15.8 ⁇ 0.2°, 16.4 ⁇ 0.2°, 17.3 ⁇ 0.2°, 18.5 ⁇ 0.2°, 18.7 ⁇ 0.2°, 19.3 ⁇ 0.2°, 19.6 ⁇ 0.2°, 20.0 ⁇ 0.2°, 20.9 ⁇ 0.2°,
  • Table 39 below provides the XRPD Peak List for Pattern 9 with >10% relative intensity peaks.
  • the XRPD performed on Pattern 9 exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 9 at about 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.0 ⁇ 0.2°, 22.5 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.7 ⁇ 0.2°, 25.0 ⁇ 0.2°, 25.2 ⁇ 0.2°, 25.6 ⁇ 0.2°, 26.9 ⁇ 0.2°, 28.2 ⁇ 0.2°, 29.2 ⁇ 0.2°, 29.6 ⁇ 0.2°, 29.7 ⁇ 0.2°, 29.8 ⁇ 0.2°, 32.5 ⁇ 0.2°, 32.6 ⁇ 0.2°, and 33.6 ⁇ 0.2°.
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.0 ⁇ 0.2°, 22.5 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.7 ⁇ 0.2°, 25.0 ⁇ 0.2°, 25.2 ⁇ 0.2°, 25.6 ⁇ 0.2°, 26.9 ⁇ 0.2°, 28.2 ⁇ 0.2°, 29.2 ⁇ 0.2°, 29.6 ⁇ 0.2°, 29.7 ⁇ 0.2°, 29.8 ⁇ 0.2°, 32.5 ⁇ 0.2°, 32.6 ⁇ 0.2°, and
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 3 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.0 ⁇ 0.2°, 22.5 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.7 ⁇ 0.2°, 25.0 ⁇ 0.2°, 25.2 ⁇ 0.2°, 25.6 ⁇ 0.2°, 26.9 ⁇ 0.2°, 28.2 ⁇ 0.2°, 29.2 ⁇ 0.2°, 29.6 ⁇ 0.2°, 29.7 ⁇ 0.2°, 29.8 ⁇ 0.2°, 32.5 ⁇ 0.2°, 32.6 ⁇ 0.2°, and
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 5 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.0 ⁇ 0.2°, 22.5 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.7 ⁇ 0.2°, 25.0 ⁇ 0.2°, 25.2 ⁇ 0.2°, 25.6 ⁇ 0.2°, 26.9 ⁇ 0.2°, 28.2 ⁇ 0.2°, 29.2 ⁇ 0.2°, 29.6 ⁇ 0.2°, 29.7 ⁇ 0.2°, 29.8 ⁇ 0.2°, 32.5 ⁇ 0.2°, 32.6 ⁇ 0.2°, and
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 7 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°,
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 9 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°,
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 11 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°,
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 13 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°,
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 15 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°,
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 17 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°,
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising at least 19 peaks selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.0 ⁇ 0.2°, 22.5 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.7 ⁇ 0.2°, 25.0 ⁇ 0.2°, 25.2 ⁇ 0.2°, 25.6 ⁇ 0.2°, 26.9 ⁇ 0.2°, 28.2 ⁇ 0.2°, 29.2 ⁇ 0.2°, 29.6 ⁇ 0.2°, 29.7 ⁇ 0.2°, 29.8 ⁇ 0.2°, 32.5 ⁇ 0.2°, 32.6 ⁇ 0.2°, and 33.6 ⁇ 0.2°.
  • the morphic form is Pattern 9 and is characterized by an XRPD pattern comprising the 2theta values selected from 11.4 ⁇ 0.2°, 11.5 ⁇ 0.2°, 12.8 ⁇ 0.2°, 16.3 ⁇ 0.2°, 16.9 ⁇ 0.2°, 19.0 ⁇ 0.2°, 22.5 ⁇ 0.2°, 24.0 ⁇ 0.2°, 24.7 ⁇ 0.2°, 25.0 ⁇ 0.2°, 25.2 ⁇ 0.2°, 25.6 ⁇ 0.2°, 26.9 ⁇ 0.2°, 28.2 ⁇ 0.2°, 29.2 ⁇ 0.2°, 29.6 ⁇ 0.2°, 29.7 ⁇ 0.2°, 29.8 ⁇ 0.2°, 32.5 ⁇ 0.2°, 32.6 ⁇ 0.2°, and 33.6 ⁇ 0.2°.
  • Table 40 below provides the XRPD Peak List for Pattern 10 with >10% relative intensity peaks.
  • the XRPD performed on Pattern 10 exhibited sharp peaks, indicating the sample was composed of crystalline material. Significant peaks were observed in the XRPD on Pattern 10 at about 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising at least 2 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 2
  • XRPD pattern comprising at least 3 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising at least 5 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising at least 7 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising at least 9 peaks selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • the morphic form is Pattern 10 and is characterized by an XRPD pattern comprising the 2theta values selected from 8.6 ⁇ 0.2°, 8.8 ⁇ 0.2°, 18.8 ⁇ 0.2°, 22.8 ⁇ 0.2°, 28.4 ⁇ 0.2°, 30.0 ⁇ 0.2°, 30.6 ⁇ 0.2°, 30.7 ⁇ 0.2°, 31.1 ⁇ 0.2°, 31.3 ⁇ 0.2°, and 33.4 ⁇ 0.2°.
  • NF National Formulary
  • USP United States Pharmacopoeia
  • Ph .Eur. European Pharmacopoeia
  • N/A not applicable
  • q.s. quantity sufficient a
  • the target weight of solution filled into vials prior to lyophilization is 12.252 g (12 mL).
  • b Amount in terms of free base (equivalent to 373 mg of Compound 1 Pattern 1 dihydrochloride, dihydrate).
  • c May be used if necessary, to adjust the bulk solution pH.
  • d Used to dissolve components then removed during lyophilization.
  • Nitrogen is used as an inert gas at the end of the lyophilization process for vacuum adjustment.
  • the lyophilized composition described in Table 41 can be prepared by mixing mannitol, and citric acid monohydrate in the mass ratios described in Table 41 in water.
  • the pH of the solution is adjusted with hydrochloric acid and sodium hydroxide as appropriate to achieve a pH between 3.5 and 5, typically to a pH between 4.1 and 4.3.
  • a representative kilogram scale manufacture of the lyophilized composition from table 41 is provided:
  • Mannitol (4 kg) was slowly added to a tank of Water for Injection (WFI) grade water (130 kg).
  • WFI Water for Injection
  • the mannitol dispensing container was rinsed with 500 mL of WFI and the material was added to the tank and mixed until the mannitol was completely dissolved.
  • citric acid monohydrate (1 kg) was added to the tank and the dispensing container was washed with 200 mL of WFI. The resulting solution was mixed until completely dissolved.
  • a sample of the solution was taken, and the pH was checked (2.26) and then was adjusted by adding 2800 mL of 5N NaOH slowly. The pH was checked again (6.56) and an additional 100 mL of 5N NaOH was added.
  • the filtered solution was then loaded into appropriately sized vials. Shelves of the lyophilization instrument were then precooled to 5°C and then the vials were placed on the shelves. The lyophilization was conducted with the following temperature and vacuum cycle: After the cycle was complete the lyophilization chamber was purged with nitrogen gas filtered through a 0.22 um sterile !yophi!ized vent filter. A lyophilized sample of this material was then analyzed by XRPD.
  • the XRPD analysis was carried out on a Bruker D8 advance powder X-ray diffraction system by placing the sample on the center of a zero-background holder coated with a thin layer of petroleum jelly, after which the sample was leveled with a glass slide.
  • the XRPD image is shown in FIG. 100.
  • Table 42 below provides a list of peaks with >10% relative intensity peaks for the lyophilized Compound 1 dihydrochloride composition which comprises mannitol and citric acid described above.
  • the XRPD performed on the lyophilized Compound 1 dihydrochloride composition exhibited sharp peaks, indicating that the sample was still crystalline after lyophilization.
  • Significant peaks were observed in the XRPD on lyophilized Compound 1 dihydrochloride composition at about 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the XRPD pattern exhibits several Compound 1 Pattern 1 peaks and shows that Compound 1 Pattern 1 is formed during the lyophilization process.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 2 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 3 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 4 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 5 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 6 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 7 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 8 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 9 peaks selected 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least 10 peaks selected from 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising at least the following peaks 9.8 ⁇ 0.4°, 18.9 ⁇ 0.4°, 19.5 ⁇ 0.4°, 20.5 ⁇ 0.4°, 21.2 ⁇ 0.4°, 22.2 ⁇ 0.4°, 23.5 ⁇ 0.4°, 24.7 ⁇ 0.4°, 25.4 ⁇ 0.4°, 31.8 ⁇ 0.4°, and 36.2 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising a peak at 9.8 ⁇ 0.4° 2theta.
  • the lyophilized Compound 1 dihydrochloride composition is characterized by an XRPD pattern comprising a peak at 20.5 ⁇ 0.4° 2theta.
  • ⁇ 0.4° 2theta is ⁇ 0.3° 2theta or ⁇ 0.2° 2theta.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP21826036.2A 2020-06-15 2021-06-15 Morphische formen von trilaciclib und verfahren zur herstellung davon Pending EP4164653A1 (de)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202063039372P 2020-06-15 2020-06-15
US17/097,854 US10988479B1 (en) 2020-06-15 2020-11-13 Morphic forms of trilaciclib and methods of manufacture thereof
US202163180578P 2021-04-27 2021-04-27
PCT/US2021/037462 WO2021257587A1 (en) 2020-06-15 2021-06-15 Morphic forms of trilaciclib and methods of manufacture thereof

Publications (1)

Publication Number Publication Date
EP4164653A1 true EP4164653A1 (de) 2023-04-19

Family

ID=80326310

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21826036.2A Pending EP4164653A1 (de) 2020-06-15 2021-06-15 Morphische formen von trilaciclib und verfahren zur herstellung davon

Country Status (4)

Country Link
EP (1) EP4164653A1 (de)
CN (1) CN116157403A (de)
TW (1) TW202214650A (de)
WO (1) WO2021257587A1 (de)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230374028A1 (en) * 2020-10-08 2023-11-23 Teva Pharmaceuticals International Gmbh Solid state forms of trilaciclib and of trilaciclib salts
WO2023164255A1 (en) 2022-02-28 2023-08-31 Teva Pharmaceuticals International Gmbh Crystalline forms of trilaciclib and trilaciclib salts
WO2024009319A1 (en) * 2022-07-02 2024-01-11 Dr. Reddy's Laboratories Limited Liquid injectable compositions of trilaciclib

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE030714T2 (en) 2010-10-25 2017-05-29 G1 Therapeutics Inc CDK inhibitors
RS59790B1 (sr) * 2013-03-15 2020-02-28 G1 Therapeutics Inc Privremena zaštita normalnih ćelija tokom hemoterapije
US20160220569A1 (en) * 2015-02-03 2016-08-04 G1 Therapeutics, Inc. CDK4/6 Inhibitor Dosage Formulations For The Protection Of Hematopoietic Stem And Progenitor Cells During Chemotherapy
NZ749275A (en) 2016-07-01 2023-06-30 G1 Therapeutics Inc Synthesis of n-(heteroaryl)-pyrrolo[2,3-d]pyrimidin-2-amines
IL303038B1 (en) * 2016-12-05 2024-04-01 G1 Therapeutics Inc Preservation of immune response during chemotherapy regimens
RU2019142591A (ru) * 2017-06-29 2021-07-29 Г1 Терапьютикс, Инк. Морфологические формы g1t38 и способы их получения
US10988479B1 (en) * 2020-06-15 2021-04-27 G1 Therapeutics, Inc. Morphic forms of trilaciclib and methods of manufacture thereof

Also Published As

Publication number Publication date
CN116157403A (zh) 2023-05-23
TW202214650A (zh) 2022-04-16
WO2021257587A1 (en) 2021-12-23

Similar Documents

Publication Publication Date Title
US10654831B2 (en) Antiproliferative pyrimidine-based compounds
US20200239486A1 (en) Pyrimidine-based compounds for the treatment of cancer
US20210047328A1 (en) Pyrimidine-based antiproliferative agents
US10988479B1 (en) Morphic forms of trilaciclib and methods of manufacture thereof
US11261193B2 (en) Morphic forms of G1T38 and methods of manufacture thereof
US20210198256A1 (en) Compounds for the degradation of brd9 or mth1
EP4164653A1 (de) Morphische formen von trilaciclib und verfahren zur herstellung davon
WO2019222521A1 (en) Cdk inhibitors for the treatment of neoplastic disorders
CA3174245A1 (en) Therapeutics for the degradation of mutant braf
TW202328101A (zh) 用於標靶降解brd9之經選擇的化合物
AU2022280070A1 (en) Egfr degraders to treat cancer metastasis to the brain or cns
EA041689B1 (ru) Морфологические формы g1t38 и способы их получения
JP2024521791A (ja) 脳又はcnsへの癌転移を治療するためのegfrデグレーダー
CA3174290A1 (en) Selected compounds for targeted degradation of brd9
WO2024119111A2 (en) Morphic forms of a mutant braf degrader and methods of manufacture thereof
WO2023154417A1 (en) Morphic forms of cft7455 and methods of manufacture thereof
WO2024097980A1 (en) Ret-ldd protein inhibitors
CN117720516A (zh) 选择性brd9降解剂的有利晶型形式

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230104

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230524

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40091353

Country of ref document: HK

TPAC Observations filed by third parties

Free format text: ORIGINAL CODE: EPIDOSNTIPA