SOLID FORMS COMPRISING 3-(4-AMINO-1-OXO-1,3-DIHYDRO-ISOINDOL-2-YL)-PIPERIDINE-2,6-DIONE AND A COFORMER, COMPOSITIONS AND METHODS OF USE THEREOF

Provided herein are solid forms comprising (a) 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione (Lenalidomide) and (b) a coformer. Pharmaceutical compositions comprising the solid forms and methods for treating, preventing and managing various disorders are also disclosed.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

This application claims priority to U.S. Provisional Application No. 61/805,425, filed Mar. 26, 2013, the entirety of which is incorporated herein by reference.

1. FIELD

Provided herein are solid forms comprising 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione and a coformer. Pharmaceutical compositions comprising such solid forms (e.g., cocrystals) and methods of use for treating, preventing, and managing various disorders are also provided herein.

2. BACKGROUND

2.1 Solid Forms of Pharmaceutical Compounds

The identification and selection of a solid form of a pharmaceutical compound are complex, given that a change in solid form may affect a variety of physical and chemical properties, which may provide benefits or drawbacks in processing, formulation, stability, bioavailability, storage, handling (e.g., shipping), among other important pharmaceutical characteristics. Useful pharmaceutical solids include crystalline solids and amorphous solids, depending on the product and its mode of administration. Amorphous solids are characterized by a lack of long-range structural order, whereas crystalline solids are characterized by structural periodicity. The desired class of pharmaceutical solid depends upon the specific application; amorphous solids are sometimes selected on the basis of, e.g., an enhanced dissolution profile, while crystalline solids may be desirable for properties such as, e.g., physical or chemical stability (see, e.g., S. R. Vippagunta et al., Adv. Drug. Deliv. Rev., (2001) 48:3-26; L. Yu, Adv. Drug. Deliv. Rev., (2001) 48:27-42).

Whether crystalline or amorphous, solid forms of a pharmaceutical compound include single-component and multiple-component solids. Single-component solids consist essentially of the pharmaceutical compound or active ingredient in the absence of other compounds. Variety among single-component crystalline materials may potentially arise from the phenomenon of polymorphism, wherein multiple three-dimensional arrangements exist for a particular pharmaceutical compound (see, e.g., S. R. Byrn et al., Solid State Chemistry of Drugs, (1999) SSCI, West Lafayette). The importance of discovering polymorphs was underscored by the case of Ritonavir™, an HIV protease inhibitor that was formulated as soft gelatin capsules. About two years after the product was launched, the unanticipated precipitation of a new, less soluble polymorph in the formulation necessitated the withdrawal of the product from the market until a more consistent formulation could be developed (see S. R. Chemburkar et al., Org. Process Res. Dev., (2000) 4:413-417).

Additional diversity among the potential solid forms of a pharmaceutical compound may arise from the possibility of multiple-component solids. Crystalline solids comprising two or more ionic species may be termed salts (see, e.g., Handbook of Pharmaceutical Salts: Properties, Selection and Use, P. H. Stahl and C. G. Wermuth, Eds., (2002), Wiley, Weinheim). Additional types of multiple-component solids that may potentially offer other property improvements for a pharmaceutical compound or salt thereof include, e.g., hydrates, solvates, co-crystals and clathrates, among others (see, e.g., S. R. Byrn et al., Solid State Chemistry of Drugs, (1999) SSCI, West Lafayette). Moreover, multiple-component crystal forms may potentially be susceptible to polymorphism, wherein a given multiple-component composition may exist in more than one three-dimensional crystalline arrangement.

Cocrystals are crystalline molecular complexes of two or more non-volatile compounds bound together in a crystal lattice by non-ionic interactions. Pharmaceutical cocrystals are cocrystals of a therapeutic compound, e.g., an active pharmaceutical ingredient (API), and one or more non-volatile compound(s) (referred to herein as coformer). A coformer in a pharmaceutical cocrystal is typically a non-toxic pharmaceutically acceptable molecule, such as, for example, food additives, preservatives, pharmaceutical excipients, or other APIs. In recent years, pharmaceutical cocrystals have emerged as a possible alternative approach to enhance physicochemical properties of drug products.

The variety of possible solid forms creates potential diversity in physical and chemical properties for a given pharmaceutical compound. The discovery and selection of solid forms are of great importance in the development of an effective, stable and marketable pharmaceutical product.

2.2 Lenalidomide

Lenalidomide has a chemical name of 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione and is marketed as Revlimid®. It is an immunomodulatory agent with antiangiogenic and antineoplastic properties. Lenalidomide is a 4-amino-glutamyl analogue of thalidomide that lacks neurological side effects such as sedation and neuropathy commonly associated with thalidomide. Lenalidomide is an immunomodulator and has been shown to affect the cellular and humoral limbs of the immune system. It has also been shown to have anti-antigenic properties wherein it has been shown to act across a spectrum of neoplastic conditions.

Lenalidomide is approved by the Food and Drug Administration (FDA) for use in combination with dexamethasone for the treatment of multiple myeloma (MM) patients who have received at least one prior therapy and for patients with transfusion-dependent anemia due to Low- or Intermediate-1-risk myelodysplastic syndromes (MDS) associated with a deletion 5q cytogenetic abnormality with or without additional cytogenetic abnormalities (Revlimid® webpage).

Lenalidomide is an off-white to pale-yellow solid powder. It is soluble in organic solvent/water mixtures, and buffered aqueous solvents. Lenalidomide is more soluble in organic solvents and low pH solutions. Solubility was significantly lower in less acidic buffers, ranging from about 0.4 to 0.5 mg/ml. Lenalidomide has an asymmetric carbon atom and can exist as the optically active forms S(−) and R(+), and is produced as a racemic mixture with a net optical rotation of zero.

The mechanism of action of lenalidomide remains to be fully characterized. Lenalidomide possesses immunomodulatory, antiangiogenic, and antineoplastic properties. Experiments have demonstrated that lenalidomide inhibits the growth of cells derived from patients with multiple myeloma and del (5q) myelodysplastic syndromes in vitro. Lenalidomide causes a delay in tumor growth in some in vivo nonclinical hematopoietic tumor models, including multiple myeloma. Lenalidomide inhibits the secretion of pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), from peripheral blood mononuclear cells. Lenalidomide also inhibited the expression of cyclooxygenase-2 (COX-2) but not COX-1 in vitro.

Citation of any references in this Section is not to be construed as an admission that such references are prior art to the present application.

3. SUMMARY

Provided herein are solid forms (e.g., crystal forms or amorphous forms, or mixtures thereof) comprising lenalidomide, or pharmaceutically acceptable salts, stereoisomers, solvates (including, hydrates), prodrugs, or clathrates thereof, and a coformer. Also provided are methods of preparing, isolating, and characterizing the solid forms.

Also provided herein are pharmaceutical compositions and single unit dosage forms, which comprise one or more solid forms provided herein.

Also provided herein are methods of treating and managing various diseases or disorders. The methods comprise administering to a patient in need of such treatment or management a therapeutically effective amount of a solid form provided herein.

Also provided herein are methods of preventing various diseases and disorders, which comprise administering to a patient in need of such prevention a prophylactically effective amount of a solid form provided herein.

Also provided herein are methods of treating and managing various diseases or disorders with a therapeutically effective amount of a solid form provided herein and a second, third, or fourth agent.

The various diseases and disorders include, but are not limited to: cancer, including hematologic cancer or solid tumor, for example, multiple myeloma; leukemia including but not limited to acute lymphoblastic leukemia, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia; lymphoma including but not limited to Hodgkins and Non-Hodgkins lymphoma (B-cell and T-cell lymphomas, including follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, indolent lymphomas and others), sarcoma, prostate cancer, or small cell lung cancer; glioma, neuroblastoma, scleroderma; amyloidosis; pain; myelofibrosis; myeloproliferative disease, for example, myelofibrosis with myeloid metaplasia (MMM); myelodysplastic syndromes; diffuse systemic sclerosis; macular degeneration; an immunodeficiency disorder; a CNS disorder; a CNS injury; atherosclerosis; hemoglobinopathy; anemia, for example, sickle cell anemia; an inflammatory disease; an autoimmune disease; a viral disease; a genetic disease; an allergic disease; a bacterial disease; an ocular neovascular disease; a choroidal neovascular disease; a retina neovascular disease; and rubeosis.

4. BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 provides a representative XRPD pattern of a solid form comprising lenalidomide and gallic acid.

FIG. 2 provides a representative XRPD pattern of a solid form comprising lenalidomide and vanillin.

FIG. 3 provides a representative XRPD pattern of a solid form comprising lenalidomide and oxalic acid.

FIG. 4 provides a representative XRPD pattern of a solid form comprising lenalidomide and propyl gallate.

FIG. 5 provides a representative XRPD pattern of a solid form comprising lenalidomide and glycolic acid.

FIG. 6 provides a representative XRPD pattern of a solid form comprising lenalidomide and sodium lauryl sulfate.

FIGS. 7A & 7B provide representative XRPD patterns of a solid form comprising lenalidomide and magnesium bromide.

FIG. 8 provides a representative DSC thermogram and a representative TGA thermogram of a solid form comprising lenalidomide and magnesium bromide.

FIG. 9 provides a representative DVS plot of a solid form comprising lenalidomide and magnesium bromide.

FIG. 10 provides a representative Raman spectrum of a solid form comprising lenalidomide and magnesium bromide.

FIG. 11 provides a representative 1H-NMR spectrum of a solid form comprising lenalidomide and magnesium bromide.

FIG. 12 provides a representative crystal habit of a solid form comprising lenalidomide and magnesium bromide.

FIGS. 13A & 13B provide representative XRPD patterns of a solid form comprising lenalidomide and malonic acid.

FIG. 14 provides a representative DSC thermogram and a representative TGA thermogram of a solid form comprising lenalidomide and malonic acid.

FIG. 15 provides a representative DVS plot of a solid form comprising lenalidomide and malonic acid.

FIG. 16 provides a representative Raman spectrum of a solid form comprising lenalidomide and malonic acid.

FIGS. 17A and 17B provides a representative 1H-NMR spectrum of a solid form comprising lenalidomide and malonic acid.

FIG. 18 provides a representative crystal habit of a solid form comprising lenalidomide and malonic acid.

FIGS. 19A & 19B provide representative XRPD patterns of a solid form comprising lenalidomide and L-tartaric acid.

FIG. 20 provides a representative DSC thermogram and a representative TGA thermogram of a solid form comprising lenalidomide and L-tartaric acid.

FIG. 21 provides a representative DVS plot of a solid form comprising lenalidomide and L-tartaric acid.

FIG. 22 provides a representative 1H-NMR spectrum of a solid form comprising lenalidomide and L-tartaric acid.

FIG. 23 provides a representative crystal habit of a solid form comprising lenalidomide and L-tartaric acid.

FIG. 24 provides a representative XRPD pattern of a solid form comprising lenalidomide and hippuric acid.

FIG. 25 provides a representative XRPD pattern of a solid form comprising lenalidomide and zinc chloride.

FIG. 26 provides a representative XRPD pattern of a solid form comprising lenalidomide and benzoic acid.

FIG. 27 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and gallic acid with a pattern from gallic acid.

FIG. 28 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and oxalic acid with a pattern from lenalidomide form D.

FIG. 29 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and propyl gallate with a pattern from lenalidomide form D.

FIG. 30 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and glycolic acid with a pattern from glycolic acid.

FIG. 31 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and sodium lauryl sulfate with a pattern from lenalidomide form A.

FIG. 32 provides a representative overlay plot of the XRPD patterns of a solid form comprising lenalidomide and magnesium bromide with a pattern from magnesium bromide.

FIG. 33 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and hippuric acid with a pattern from lenalidomide form D.

FIG. 34 provides a representative overlay plot of the XRPD pattern of a solid form comprising lenalidomide and benzoic acid with a pattern from benzoic acid.

5. DETAILED DESCRIPTION 5.1 Definitions

As used herein, and in the specification and the accompanying claims, the indefinite articles “a” and “an” and the definite article “the” include plural as well as single referents, unless the context clearly indicates otherwise.

As used herein, and unless otherwise specified, the compound referred to herein by the name lenalidomide, 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione, or Revlimid®, corresponds to chemical structure (I), depicted below. In certain embodiments, the term lenalidomide, 3-(4-amino-1-oxo-1,3-dihydro-isoindol-2-yl)-piperidine-2,6-dione, may be used herein to refer to either a free base form or an ionized form of a compound of formula (I) (e.g., the molecule is protonated at one or more basic centers).

Unless otherwise specified, the terms “solid form,” “solid forms,” and related terms, when used herein to refer a physical form comprising lenalidomide, which is not predominantly in a liquid or a gaseous state. As used herein, the terms “solid form” and “solid forms” encompass semi-solids. Solid forms may be crystalline, amorphous, partially crystalline, partially amorphous, or mixtures of forms. A “single-component” solid form comprising lenalidomide consists essentially of lenalidomide. A “multiple-component” solid form comprising lenalidomide comprises a significant quantity of one or more additional species, such as ions and/or molecules, within the solid form. For example, in particular embodiments, a crystalline multiple-component solid form comprising lenalidomide further comprises one or more species non-covalently bonded at regular positions in the crystal lattice.

Unless otherwise specified, the term “crystalline” and related terms used herein, when used to describe a substance, component, product, or form, mean that the substance, component, product, or form is substantially crystalline, for example, as determined by X-ray diffraction. (see, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa., 173 (1990); The United States Pharmacopeia, 23rd ed., 1843-1844 (1995)).

Unless otherwise specified, the term “crystal form,” “crystal forms,” and related terms herein refer to crystalline modifications comprising a given substance, including single-component crystal forms and multiple-component crystal forms, and including, but not limited to, polymorphs, solvates, hydrates, co-crystals, other molecular complexes, salts, solvates of salts, hydrates of salts, co-crystals of salts, and other molecular complexes of salts, and polymorphs thereof. In some embodiments, a crystal form of a substance may be substantially free of amorphous forms and/or other crystal forms. In other embodiments, a crystal form of a substance may contain less than about 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% of one or more amorphous form(s) and/or other crystal form(s) on a weight basis. Crystal forms of a substance may be obtained by a number of methods. Such methods include, but are not limited to, melt recrystallization, melt cooling, solvent recrystallization, recrystallization in confined spaces such as, e.g., in nanopores or capillaries, recrystallization on surfaces or templates such as, e.g., on polymers, recrystallization in the presence of additives, such as, e.g., co-crystal counter-molecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, grinding, and solvent-drop grinding.

Unless otherwise specified, the terms “polymorph,” “polymorphic form,” “polymorphs,” “polymorphic forms,” and related terms herein refer to two or more crystal forms that consist essentially of the same molecule, molecules or ions. Different polymorphs may have different physical properties, such as, for example, melting temperatures, heats of fusion, solubilities, dissolution rates, and/or vibrational spectra as a result of a different arrangement or conformation of the molecules or ions in the crystal lattice. The differences in physical properties exhibited by polymorphs may affect pharmaceutical parameters, such as storage stability, compressibility and density (important in formulation and product manufacturing), and dissolution rate (an important factor in bioavailability). Differences in stability can result from changes in chemical reactivity (e.g., differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymorph than when comprised of another polymorph) or mechanical changes (e.g., tablets crumble on storage as a kinetically favored polymorph converts to thermodynamically a more stable polymorph) or both (e.g., tablets of one polymorph are more susceptible to breakdown at high humidity). As a result of solubility/dissolution differences, in the extreme case, some polymorphic transitions may result in lack of potency or, at the other extreme, toxicity. In addition, the physical properties of the crystal may be important in processing; for example, one polymorph might be more likely to form solvates or might be difficult to filter and wash free of impurities (e.g., particle shape and size distribution might be different between polymorphs).

Unless otherwise specified, the term “cocrystal” or “co-crystal,” as used herein, refers to a crystalline material comprised of two or more non-volative compounds bond together in a crystal lattice by non-covalent interactions.

Unless otherwise specified, the term “pharmaceutical cocrystal” or “cocrystal” of an active pharmaceutical ingredient (API), as used herein, refers to a crystalline material comprised of an API and one or more non-volative compound(s) (referred herein as a coformer). The API and the coformer interact through non-covalent forces in a crystal lattice.

Unless otherwise specified, the term “amorphous,” “amorphous form,” and related terms used herein mean that the substance, component, or product referred to is not substantially crystalline as determined by X-ray diffraction. In certain embodiments, an amorphous form of a substance may be substantially free of crystal forms. In other embodiments, an amorphous form of a substance may contain less than about 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45% or 50% of one or more crystal forms on a weight basis. In other embodiments, an amorphous form of a substance may comprise additional components or ingredients (for example, an additive, a polymer, or an excipient that may serve to further stabilize the amorphous form). In some embodiments, amorphous form may be a solid solution. Amorphous forms of a substance can be obtained by a number of methods. Such methods include, but are not limited to, heating, melt cooling, rapid melt cooling, solvent evaporation, rapid solvent evaporation, desolvation, sublimation, grinding, ball-milling, cryo-grinding, spray drying, and freeze drying.

As used herein, and unless otherwise specified, the terms “about” and “approximately,” when used in connection with doses, amounts, or weight percents of ingredients of a composition or a dosage form, mean a dose, amount, or weight percent that is recognized by one of ordinary skill in the art to provide a pharmacological effect equivalent to that obtained from the specified dose, amount, or weight percent. In certain embodiments, the terms “about” and “approximately,” when used in this context, contemplate a dose, amount, or weight percent within 30%, within 20%, within 15%, within 10%, or within 5%, of the specified dose, amount, or weight percent.

As used herein, and unless otherwise specified, the terms “about” and “approximately,” when used in connection with a numeric value or range of values which is provided to characterize a particular solid form, e.g., a specific temperature or temperature range, such as, for example, that describes a melting, dehydration, desolvation, or glass transition temperature; a mass change, such as, for example, a mass change as a function of temperature or humidity; a solvent or water content, in terms of, for example, mass or a percentage; or a peak position, such as, for example, in analysis by, for example, IR or Raman spectroscopy or XRPD; indicate that the value or range of values may deviate to an extent deemed reasonable to one of ordinary skill in the art while still describing the solid form.

Techniques for characterizing crystal forms and amorphous forms include, but are not limited to, thermal gravimetric analysis (TGA), differential scanning calorimetry (DSC), X-ray powder diffractometry (XRPD), single-crystal X-ray diffractometry, vibrational spectroscopy, e.g., infrared (IR) and Raman spectroscopy, solid-state and solution nuclear magnetic resonance (NMR) spectroscopy, optical microscopy, hot stage optical microscopy, scanning electron microscopy (SEM), electron crystallography and quantitative analysis, particle size analysis (PSA), surface area analysis, solubility studies, and dissolution studies.

In certain embodiments, the terms “about” and “approximately,” when used in this context, indicate that the numeric value or range of values may vary within 30%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1.5%, 1%, 0.5%, or 0.25% of the recited value or range of values. For example, in some embodiments, the value of an XRPD peak position may vary by up to ±0.2 degrees two theta while still describing the particular XRPD peak.

As used herein, and unless otherwise specified, a crystalline or amorphous form that is “pure,” i.e., substantially free of other crystalline or amorphous forms, contains less than about 10% by weight of one or more other crystalline or amorphous forms, less than about 5% by weight of one or more other crystalline or amorphous forms, less than about 3% by weight of one or more other crystalline or amorphous forms, or less than about 1% by weight of one or more other crystalline or amorphous forms.

As used herein, and unless otherwise specified, a solid form that is “substantially physically pure” is substantially free from other solid forms. In certain embodiments, a crystal form that is substantially physically pure contains less than about 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.05%, or 0.01% of one or more other solid forms on a weight basis. The detection of other solid forms can be accomplished by any method apparent to a person of ordinary skill in the art, including, but not limited to, diffraction analysis, thermal analysis, elemental combustion analysis and/or spectroscopic analysis.

As used herein, and unless otherwise specified, a solid form that is “substantially chemically pure” is substantially free from other chemical compounds (i.e., chemical impurities). In certain embodiments, a solid form that is substantially chemically pure contains less than about 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.05%, or 0.01% of one or more other chemical compounds on a weight basis. The detection of other chemical compounds can be accomplished by any method apparent to a person of ordinary skill in the art, including, but not limited to, methods of chemical analysis, such as, e.g., mass spectrometry analysis, spectroscopic analysis, thermal analysis, elemental combustion analysis and/or chromatographic analysis.

As used herein, and unless otherwise indicated, a chemical compound, solid form, or composition that is “substantially free” of another chemical compound, solid form, or composition means that the compound, solid form, or composition contains, in certain embodiments, less than about 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2% 0.1%, 0.05%, or 0.01% by weight of the other compound, solid form, or composition.

As used herein, and unless otherwise specified, the term “pharmaceutically acceptable salts” refers to salts prepared from pharmaceutically acceptable, relatively non-toxic acids, including inorganic acids and organic acids. In some embodiments, suitable acids include, but are not limited to, acetic, benzenesulfonic, benzoic, camphorsulfonic, carbonic, citric, dihydrogenphosphoric, ethenesulfonic, fumaric, galactunoric, gluconic, glucuronic, glutamic, hydrobromic, hydrochloric, hydriodic, isobutyric, isethionic, lactic, maleic, malic, malonic, mandelic, methanesulfonic, monohydrogencarbonic, monohydrogen-phosphoric, monohydrogensulfuric, mucic, nitric, pamoic, pantothenic, phosphoric, phthalic, propionic, suberic, succinic, sulfuric, tartaric, toluenesulfonic acid (including p-toluenesulfonic, m-toluenesulfonic, and o-toluenesulfonic acids), and the like (see, e.g., S. M. Berge et al., J. Pharm. Sci., 66:1-19 (1977); and Handbook of Pharmaceutical Salts: Properties, Selection and Use, P. H. Stahl and C. G. Wermuth, Eds., (2002), Wiley, Weinheim). In some embodiments, suitable acids are strong acids (e.g., with pKa less than about 1), including, but not limited to, hydrochloric, hydrobromic, sulfuric, nitric, methanesulfonic, benzene sulfonic, toluene sulfonic, naphthalene sulfonic, naphthalene disulfonic, pyridine-sulfonic, or other substituted sulfonic acids. Also included are salts of other relatively non-toxic compounds that possess acidic character, including amino acids, such as aspartic acid and the like, and other compounds, such as aspirin, ibuprofen, saccharin, and the like. Acid addition salts can be obtained by contacting the neutral form of a compound with a sufficient amount of the desired acid, either neat or in a suitable solvent. As solids, salts can exist in crystalline or amorphous forms, or mixtures thereof. Salts can also exist in polymorphic forms.

Unless otherwise specified, the terms “solvate” and “solvated,” as used herein, refer to a solid form of a substance which contains solvent. The terms “hydrate” and “hydrated” refer to a solvate wherein the solvent is water. “Polymorphs of solvates” refer to the existence of more than one solid form for a particular solvate composition. Similarly, “polymorphs of hydrates” refer to the existence of more than one solid form for a particular hydrate composition. The term “desolvated solvate,” as used herein, refers to a solid form of a substance which can be made by removing the solvent from a solvate. The terms “solvate” and “solvated,” as used herein, can also refer to a solvate of a salt, co-crystal, or molecular complex. The terms “hydrate” and “hydrated,” as used herein, can also refer to a hydrate of a salt, co-crystal, or molecular complex.

As used herein, and unless otherwise specified, the terms “treat,” “treating” and “treatment” refer to the eradication or amelioration of a disease or disorder, or of one or more symptoms associated with the disease or disorder. In certain embodiments, the terms refer to minimizing the spread or worsening of the disease or disorder resulting from the administration of one or more prophylactic or therapeutic agents to a subject with such a disease or disorder. In some embodiments, the terms refer to the administration of a compound provided herein, with or without other additional active agent, after the onset of symptoms of a particular disease.

As used herein, and unless otherwise specified, the terms “prevent,” “preventing” and “prevention” refer to the prevention of the onset, recurrence or spread of a disease or disorder, or of one or more symptoms thereof. In certain embodiments, the terms refer to the treatment with or administration of a compound provided herein, with or without other additional active compound, prior to the onset of symptoms, particularly to patients at risk of a disease or disorder provided herein. The terms encompass the inhibition or reduction of a symptom of a particular disease. Patients with familial history of a disease in particular are candidates for preventive regimens in certain embodiments. In addition, patients who have a history of recurring symptoms are also potential candidates for the prevention. In this regard, the term “prevention” may be interchangeably used with the term “prophylactic treatment.”

As used herein, and unless otherwise specified, the terms “manage,” “managing” and “management” refer to preventing or slowing the progression, spread, or worsening of a disease or disorder, or of one or more symptoms thereof. Often, the beneficial effects that a subject derives from a prophylactic and/or therapeutic agent do not result in a cure of the disease or disorder. In this regard, the term “managing” encompasses treating a patient who had suffered from the particular disease in an attempt to prevent or minimize the recurrence of the disease or one or more symptoms thereof.

As used herein, and unless otherwise specified, a “therapeutically effective amount” of a compound is an amount sufficient to provide a therapeutic benefit in the treatment or management of a disease or disorder, or to delay or minimize one or more symptoms associated with the disease or disorder. A therapeutically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of the disease or disorder. The term “therapeutically effective amount” can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease or disorder, or enhances the therapeutic efficacy of another therapeutic agent.

As used herein, and unless otherwise specified, a “prophylactically effective amount” of a compound is an amount sufficient to prevent a disease or disorder, or one or more symptoms thereof, or prevent the recurrence of the disease or disorder, or one or more symptoms thereof. A prophylactically effective amount of a compound means an amount of therapeutic agent, alone or in combination with other agents, that provides a prophylactic benefit in the prevention of the disease or disorder. The term “prophylactically effective amount” can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of another prophylactic agent.

Unless otherwise specified, the term “composition” as used herein is intended to encompass a product comprising the specified ingredient(s) (and in the specified amount(s), if indicated), as well as any product which results, directly or indirectly, from combination of the specified ingredient(s) in the specified amount(s). By “pharmaceutically acceptable,” it is meant a diluent, excipient, or carrier in a formulation must be compatible with the other ingredient(s) of the formulation and not deleterious to the recipient thereof.

Unless otherwise specified, the term “subject” is defined herein to include animals, such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, and the like. In specific embodiments, the subject is a human.

As used herein, and unless otherwise specified, the terms “symptom of a hemoglobinopathy” and “symptom of anemia” mean any physiological or biological symptom associated with any hemoglobinopathy or anemia, including, but not limited to, dizziness, shortness of breath, loss of consciousness, tiredness, weakness, hemolysis, pains associated with abnormal hemoglobin, reduced erythrocyte counts (i.e., reduced hematocrit), a reduced ability of a given volume of blood to carry oxygen, as compared with a volume of normal blood, deformities of erythrocytes visible under a microscope, etc. The terms also include negative psychological symptoms such as depression, low self-esteem, perception of illness, perception of limited physical capability, etc.

Unless otherwise specified, to the extent that there is a discrepancy between a depicted chemical structure of a compound provided herein and a chemical name of a compound provided herein, the chemical structure shall control.

5.2 Solid Forms Comprising Lenalidomide and a Coformer

In one embodiment, provided herein are solid forms (e.g., crystal forms, amorphous forms, or mixtures thereof) comprising (a) lenalidomide, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. In one embodiment, provided herein are solid forms (e.g., crystal forms, amorphous forms, or mixtures thereof) comprising (a) a free base of lenalidomide, or a solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. Lenalidomide can be synthesized or obtained according to a method known in the literature or based upon the teachings herein, including the methods described in detail in the examples herein.

In one embodiment, lenalidomide can be prepared according to methods described in, for example, U.S. Pat. Nos. 6,281,230 and 5,635,517, the entireties of which are incorporated herein by reference. For example, the compound can be prepared through catalytic hydrogenation of 3-(4-nitro-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione. 3-(4-Nitro-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione can be obtained by allowing 2,6-dioxopiperidin-3-ammonium chloride to react with methyl 2-bromomethyl-3-nitrobenzoate in dimethylformamide in the presence of triethylamine. The methyl 2-bromomethyl-3-nitrobenzoate in turn is obtained from the corresponding methyl ester of nitro-ortho-toluic acid by conventional bromination with N-bromosuccinimide under the influence of light.

Polymorphs of lenalidomide can be obtained by techniques known in the art, including solvent recrystallization, desolvation, vapor diffusion, rapid evaporation, slow evaporation, rapid cooling and slow cooling. Polymorphs can be made by dissolving a weighed quantity of 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione in various solvents at elevated temperatures. The solutions of the compound can then be filtered and allowed to evaporate either in an open vial (for fast hot evaporation) or in a vial covered with aluminum foil containing pinholes (hot slow evaporation). Polymorphs can also be obtained from slurries. Polymorphs can be crystallized from solutions or slurries using several methods. For example, a solution created at an elevated temperature (e.g., 60° C.) can be filtered quickly then allowed to cool to room temperature. Once at room temperature, the sample that does not crystallize can be moved to a refrigerator then filtered. Alternatively, the solutions can be crash cooled by dissolving the solid in a solvent at an increased temperature (e.g., 45-65° C.) followed by cooling in a dry ice/solvent bath.

The coformer can be any pharmaceutically acceptable coformer known in the art. In one embodiment, the coformer is acetylsalicylic acid, D-glucose, nicotinic acid, aconitic acid, L-glutamic acid, oxalic acid, adipic acid, glutaric acid, L-proline, 4-aminosalicylic acid, glycine, propyl gallate, L-ascorbic acid, glycolic acid, L-pyroglutamic acid, benzoic acid, hippuric acid, saccharin, (+)-camphoric acid, 1-hydroxy-2-naphthoic acid, salicylic acid, capric acid, ketoglutaric acid, sebacic acid, cinnamic acid, L-lysine, sodium lauryl sulfate, citric acid, magnesium bromide, sorbic acid, cyclamic acid, maleic acid, succinic acid, ethyl maltol, L-malic acid, L-tartaric acid, ethyl paraben, malonic acid, urea, D-fructose, maltol, vanillic acid, fumaric acid, D,L-mandelic acid, vanillin, gallic acid, methyl paraben, zinc chloride, gentisic acid, or nicotinamide. In one embodiment, the coformer is benzoic acid, gallic acid, glycolic acid, hippuric acid, magnesium bromide, malonic acid, oxalic acid, propyl gallate, sodium lauryl sulfate, L-tartaric acid, vanillic acid, or zinc chloride.

In one embodiment, solid forms provided herein may be a crystal form or an amorphous form or mixtures thereof (e.g., mixtures of crystal forms, or mixtures of crystal and amorphous forms), which comprises (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. In one embodiment, provided herein is a crystal form comprising (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. In one embodiment, provided herein is a cocrystal comprising (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. In one embodiment, provided herein is an amorphous form comprising (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. In one embodiment, provided herein is a mixture comprising (i) a cocrystal comprising (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer; and (ii) a crystal form of lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof. In one embodiment, provided herein is a mixture comprising (i) a cocrystal comprising (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer; and (ii) an amorphous form of lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof.

In one embodiment, provided herein is an unsolvated solid form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is an anhydrous solid form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is an unsolvated crystal form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is an anhydrous crystal form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is an unsolvated amorphous form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is an anhydrous amorphous form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a solvated solid form comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a hydrated solid form comprising (a) lenalidomide and (b) a coformer (e.g., a hydrate having a stoichiometric or non-stoichiometric amount of water). In one embodiment, provided herein is a hydrated form of (a) lenalidomide and (b) a coformer, including, but not limited to, a hemihydrate, a monohydrate, a dihydrate, a trihydrate, and the like. In one embodiment, the hydrated form is substantially crystalline. In one embodiment, the hydrated form is substantially amorphous. In one embodiment, the anhydrous form is substantially crystalline. In one embodiment, the anhydrous form is substantially amorphous. In one embodiment, provided herein is an unsolvated cocrystal comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is an anhydrous cocrystal comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a hydrated cocrystal comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a solvated cocrystal comprising (a) lenalidomide and (b) a coformer.

Solid forms provided herein can be prepared by the methods described herein, or by techniques, including, but not limited to, heating, cooling, freeze drying, spray drying, lyophilization, quench cooling the melt, rapid solvent evaporation, slow solvent evaporation, solvent recrystallization, antisolvent addition, slurry recrystallization, crystallization from the melt, desolvation, recrystallization in confined spaces, such as, e.g., in nanopores or capillaries, recrystallization on surfaces or templates, such as, e.g., on polymers, recrystallization in the presence of additives, such as, e.g., co-crystal counter-molecules, desolvation, dehydration, rapid cooling, slow cooling, exposure to solvent and/or water, drying, including, e.g., vacuum drying, vapor diffusion, sublimation, grinding (including, e.g., cryo-grinding and solvent-drop grinding), microwave-induced precipitation, sonication-induced precipitation, laser-induced precipitation, and precipitation from a supercritical fluid. The particle size of the resulting solid forms, which can vary (e.g., from nanometer dimensions to millimeter dimensions), can be controlled, e.g., by varying crystallization conditions, such as, e.g., the rate of crystallization and/or the crystallization solvent system, or by particle-size reduction techniques, e.g., grinding, milling, micronizing, or sonication.

In another embodiment, provided herein are compositions comprising one or more solid form(s) comprising (a) lenalidomide or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer. Also provided herein are compositions comprising: (i) one or more solid form(s) provided herein (e.g., one or more crystal forms, one or more amorphous forms, and mixtures thereof), and (ii) other active ingredient(s). Also provided herein are methods of using these compositions in the treatment, prevention, or management of conditions and disorders including, but not limited to: cancer, including hematologic cancer or solid tumor, for example, multiple myeloma; leukemia including but not limited to acute lymphoblastic leukemia, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia; lymphoma including but not limited to Hodgkins and Non-Hodgkins lymphoma (B-cell and T-cell lymphomas, including follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, indolent lymphomas and others), sarcoma, prostate cancer, or small cell lung cancer; glioma, neuroblastoma, scleroderma; amyloidosis; pain; myelofibrosis; myeloproliferative disease, for example, myelofibrosis with myeloid metaplasia (MMM); myelodysplastic syndromes; diffuse systemic sclerosis; macular degeneration; an immunodeficiency disorder; a CNS disorder; a CNS injury; atherosclerosis; hemoglobinopathy; anemia, for example, sickle cell anemia; an inflammatory disease; an autoimmune disease; a viral disease; a genetic disease; an allergic disease; a bacterial disease; an ocular neovascular disease; a choroidal neovascular disease; a retina neovascular disease; and rubeosis. In a specific embodiment, provided herein are methods of using these compositions for the treatment in patients with transfusion-dependent anemia due to Low- or Intermediate-1-risk myelodysplastic syndromes (MDS) associated with a deletion 5q cytogenetic abnormality with or without additional cytogenetic abnormalities.

In another embodiment, provided herein are methods of using these compositions in combination with a second, third, or fourth agent in the treatment, prevention, or management of conditions and disorders including, but not limited to: cancer, including hematologic cancer or solid tumor, for example, multiple myeloma; leukemia including but not limited to acute lymphoblastic leukemia, acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia; lymphoma including but not limited to Hodgkins and Non-Hodgkins lymphoma (B-cell and T-cell lymphomas, including follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, indolent lymphomas and others), sarcoma, prostate cancer, or small cell lung cancer; glioma, neuroblastoma, scleroderma; amyloidosis; pain; myelofibrosis; myeloproliferative disease, for example, myelofibrosis with myeloid metaplasia (MMM); myelodysplastic syndromes; diffuse systemic sclerosis; macular degeneration; an immunodeficiency disorder; a CNS disorder; a CNS injury; atherosclerosis; hemoglobinopathy; anemia, for example, sickle cell anemia; an inflammatory disease; an autoimmune disease; a viral disease; a genetic disease; an allergic disease; a bacterial disease; an ocular neovascular disease; a choroidal neovascular disease; a retina neovascular disease; and rubeosis. In a specific embodiment, provided herein are methods of using these compositions in combination with dexamethasone for the treatment of patients with multiple myeloma who have received at least one prior therapy.

While not intending to be bound by any particular theory, certain solid forms provided herein exhibit physical properties, e.g., stability, solubility and/or dissolution rate, appropriate for use in clinical and therapeutic dosage forms. Moreover, while not wishing to be bound by any particular theory, certain solid forms provided herein exhibit physical properties, e.g., crystal morphology, compressibility and/or hardness, suitable for manufacture of a solid dosage form. In some embodiments, such properties can be determined using techniques such as X-ray diffraction, microscopy, IR spectroscopy and thermal analysis, as described herein and known in the art.

Certain embodiments herein provide solid forms comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a solid form comprising (a) lenalidomide and (b) a coformer that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a solid form comprising a cocrystal comprising (a) lenalidomide and (b) a coformer. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising (a) lenalidomide and (b) a coformer and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising (a) lenalidomide and (b) a coformer and (ii) one or more additional crystal forms of lenalidomide.

In some embodiments, the cocrystal comprising (a) lenalidomide and (b) a coformer can be obtained by crystallization from certain solvent systems, for example, solvent systems comprising one or more of the following solvents: acetone, acetonitrile, methanol, and tetrahydrofuran. N,N-dimethylformamide is another example of a solvent. Other examples of solvent systems are provided herein elsewhere. In certain embodiments, a solid form provided herein (e.g., a cocrystal comprising (a) lenalidomide and (b) a coformer) can be obtained by slurry crystallization, evaporation crystallization, cooling crystallization, and precipitation crystallization.

In some embodiments, the cocrystals is a crystalline material comprised of an API and one or more coformers. In certain embodiments the API and the coformer interact through non-covalent forces in a crystal lattice.

In certain embodiments, the non-covalent forces are one or more hydrogen bonds (H-bonds). The coformer may be H-bonded directly to the API or may be H-bonded to an additional molecule which is bound to the API. The additional molecule may be H-bonded to the API or bound ionically or covalently to the API. The additional molecule could also be a different APT. In certain embodiments, the co-crystals may include one or more solvate molecules in the crystalline lattice, i.e., solvates of co-crystals, or a co-crystal further comprising a solvent or compound that is a liquid at room temperature. In certain embodiments, the co-crystals may be a co-crystal between a coformer and a salt of an API. In certain embodiments, the non-covalent forces are pi-stacking, guest-host complexation and/or van der Waals interactions.

In certain embodiments, hydrogen bonding may result in different intermolecular configurations. In certain embodiments the intermolecular configurations are dimmers, linear chains, or cyclic structures that can additionally include extended hydrogen bond networks and isolated triads.

In certain embodiments, the coformer is a solid under ambient temperature conditions when in its pure form. In certain embodiments, the solid form lenalidomide is solubilized with acetone, acetonitrile, methanol, or tetrahydrofuran.

In certain embodiments, the coformer is selected from acetylsalicylic acid, aconitic acid, adipic acid, 4-aminosalicylic acid, L-ascorbic acid, benzoic acid, (+)-camphoric acid, capric acid, cinnamic acid, citric acid, cyclamic acid, ethyl maltol, ethyl paraben, D-fructose, fumaric acid, gallic acid, gentisic acid, D-glucose, L-glutamic acid, glutaric acid, gycine, glycolic acid, hippuric acid, 1-hydroxy-2-naphthoic acid, ketoglutaric acid, L-lysine, magnesium bromide, maleic acid, L-malic acid, malonic acid, maltol, D,L-mandelic acid, methyl paraben, nicotinamide, nicotinic acid, oxalic acid, L-proline, propyl gallate, L-pyroglutamic acid, saccharin, salicylic acid, sebacic acid, sodium lauryl sulfate, sorbic acid, succinic acid, L-tartaric acid, urea, vanillic acid, vanillin, and zinc chloride. In specific embodiments, the coformer is selected from benzoic acid, gallic acid, hippuric acid, magnesium bromide, malonic acid, maltol, oxalic acid, proplyl gallate, sodium lauryl sulfate, L-tartaric acid, vanillic acid, and zinc chloride. In certain embodiments, the coformer is a second API.

In certain embodiments, the co-crystals include an acid addition salt or base addition salt of an API. Acid addition salts include, but are not limited to, inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, and phosphoric acid, and organic acids such as acetic acid, propionic acid, hexanoic acid, heptanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartatic acid, citric acid, benzoic acid, o-(4-hydroxybenzoyl)benzoic acid, cinnamic acid, madelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, p-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, p-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2,2,2]oct-2-ene-1-carboxylic acid, glucoheptonic acid, 4,4′-methylenebis(3-hydroxy-2-ene-1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutaric acid, hydroxynaphthoic acid, salicylic acid, stearic acid, and muconic acid. Base addition salts include, but are not limited to, inorganic bases such as sodium, potassium, lithium, ammonium, calcium and magnesium salts, and organic bases such as primary, secondary and tertiary amines (e.g., isopropylamine, trimethyl amine, diethyl amine, tri(iso-propyl) amine, tri(n-propyl) amine, ethanolamine, 2-dimethylaminoethanol, tromethamine, lysine, arginine, histidine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, N-alkylglucamines, theobromine, purines, piperazine, piperidine, morpholine, and N-ethylpiperidine).

The ratio of API to coformer may be stoichiometric or non-stoichiometric. In one embodiment, the ratio of API to coformer is about 5:1, 4:1, 3:1, 2.5:1, 2:1, 1.5:1, 1:1, 1:1.5, 1:2, 1:2.5, 1:3, 1:4, or 1:5. In one embodiment, the ratio of API to coformer is about 1:1. In one embodiment, the co-crystal comprises more than one coformers. In one embodiment, the co-crystal comprises two coformers.

In certain embodiments, cocrystals can be prepared using solid-state methods such as solid-state grinding and solvent-drop grinding. In certain embodiments, cocrystals can be prepared using high-throughput screening. In certain embodiments cocrystals can be prepared using solution-based crystallization.

In certain embodiments, cocrystals formation can lead to enhancement of physical properties of the resulting solid forms, such as solubility, dissolution rate, bioavailablity, physical stability, chemical stability, flowability, fractability, or compressibility. In certain embodiments, a given API may form different cocrystals with many different counter-molecules, and some of these cocrystals may exhibit enhanced solubility or stability. In certain embodiments pharmaceutical cocrystals increase the bioavailability or stability profile of a compound without the need for chemical (covalent) modification of the API.

In certain embodiments, the compounds provide herein may also contain an unnatural proportion of an atomic isotope at one or more of the atoms that constitute such a compound. For example, the compound may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) sulfur-35 (35S), or carbon-14 (14C). Radiolabeled compounds are useful as therapeutic agents, e.g., cancer therapeutic agents, research reagents, e.g., binding assay reagents, and diagnostic agents, e.g., in vivo imaging agents. All isotopic variations of the compounds provided herein, whether radioactive or not, are intended to be encompassed herein. In certain embodiments, a compound provided herein contains unnatural proportion(s) of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium (2H), tritium (3H), carbon-11 (11C), carbon-12 (12C), carbon-13 (13C), carbon-14 (14C), nitrogen-13 (13N), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-14 (14O), oxygen-15 (15O), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), fluorine-18 (18F), phosphorus-31 (31P), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-35 (35S), sulfur-36 (36S), chlorine-35 (35Cl), chlorine-36 (36Cl), chlorine-37 (37Cl), bromine-79 (79Br), bromine-81 (81Br), iodine-123 (123I), iodine-125 (125I), iodine-127 (127I), iodine-129 (129I), and iodine-131 (131I). In certain embodiments, a compound provided herein contains unnatural proportion(s) of one or more isotopes in a stable form, that is, non-radioactive, including, but not limited to, hydrogen (1H), deuterium (2H), carbon-12 (12C), carbon-13 (13C), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), phosphorus-31 (31P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-36 (36S), chlorine-35 (35Cl), chlorine-37 (37Cl), bromine-79 (79Br), bromine-81 (81Br), and iodine-127 (127I). In certain embodiments, a compound provided herein contains unnatural proportion(s) of one or more isotopes in an unstable form, that is, radioactive, including, but not limited to, tritium (3H), carbon-11 (11C), carbon-14 (14C), nitrogen-13 (13N), oxygen-14 (14O), oxygen-15 (15O), fluorine-18 (18F), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-35 (35S), chlorine-36 (36Cl), iodine-123 (123I), iodine-125 (125I), iodine-129 (129I), and iodine-131 (131I). In certain embodiments, in a compound as provided herein, any hydrogen can be 2H, for example, or any carbon can be 13C, for example, or any nitrogen can be 15N, for example, or any oxygen can be 180, for example, where feasible according to the judgment of one of skill. In certain embodiments, a compound provided herein contains unnatural proportions of deuterium (D). In exemplary embodiments, provided herein are isotopologues of lenalidomide, as disclosed in U.S. Provisional Application No. 61/500,053, filed Jun. 22, 2011, which is incorporated by reference herein in its entirety. In one embodiment, provided herein are solid forms (e.g., crystal forms, amorphous forms, or mixtures thereof) of isotopologues of lenalidomide provided herein.

In certain embodiments, slurry crystallization is effected by adding solvent or solvent mixtures to a solid substrate, and the slurry is stirred, and optionally heated to various temperatures. In certain embodiments, the slurry is heated at about 25° C., about 50° C., about 80° C., or about 100° C. In certain embodiments, upon heating and cooling, the residual solvents of the slurry can be removed by wicking, or other suitable methods, such as filtration, centrifugation, or decantation, and the crystals can be dried in air or under vacuum.

In certain embodiments, stoichiometric wet milling is effected by charging a PEEK grinding cup with a solvent or solvent mixture with a substrate and a steel grinding ball, which can then be sealed and shaken. In certain embodiments, upon heating and cooling, the residual solvents of the slurry can be removed by wicking, or other suitable methods, such as filtration, centrifugation, or decantation, and the crystals can be dried in air or under vacuum.

In certain embodiments, evaporation crystallization is effected by adding a solvent or solvent mixture to a solid substrate, and allowing the solvent or solvent mixture to evaporate under ambient conditions. In certain embodiments, the residual solvent can be removed by wicking, or other suitable methods, such as filtration, centrifugation, or decantation, and the crystals can be dried in air or under vacuum.

In certain embodiments, precipitation crystallization is effected by adding a solvent or solvent mixture to a solid substrate, and subsequently adding an anti-solvent. In certain embodiments, the resultant mixture stands for a period of time, e.g., overnight, and under certain conditions, for example at room temperature. In certain embodiments, the residual solvent can be removed by wicking, or other suitable methods, such as filtration, centrifugation, or decantation, and the crystals can be dried in air or under vacuum.

In certain embodiments, cooling crystallization is effected by adding a solvent or solvent mixture to a solid substrate at elevated temperature, and allowing the resultant mixture to stand for a period of time at a reduced temperature. In certain embodiments, the elevated temperature is, for example, about 30° C., about 40° C., about 50° C., about 60° C., about 70° C., or about 80° C. In certain embodiments, the reduced temperature is, for example, about 15° C., about 10° C., about 5° C., about 0° C., about −5° C., about −10° C., about −15° C., or about −20° C. The residual solvent can be removed by wicking, or other suitable methods, such as filtration, centrifugation, or decantation, and the crystals can be dried in air or under vacuum.

5.2.1 Cocrystal Compositions Comprising Lenalidomide and Gallic Acid

Certain embodiments herein provide solid forms comprising lenalidomide and gallic acid. In one embodiment, provided herein is a solid form comprising lenalidomide and gallic acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and gallic acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and gallic acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and gallic acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and gallic acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and gallic acid.

In some embodiments, the cocrystal comprising lenalidomide and gallic acid is obtained by mixing lenalidomide and gallic acid in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and gallic acid in a solvent system saturated with gallic acid. In some embodiments, the cocrystal is obtained by mixing lenalidomide and gallic acid in a solvent system saturated with gallic acid, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and gallic acid in a solvent system saturated with gallic acid, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and gallic acid. In some embodiments the solvent system is a mixed solvent of methanol and water. In some embodiments the solvent system is a mixed solvent of methanol and water with a volume ratio of methanol to water of about 1:1.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and gallic acid with a molar ratio of lenalidomide to gallic acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to gallic acid is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and gallic acid is provided in FIG. 1. In some embodiments, provided herein is a solid form comprising lenalidomide and gallic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 5.36, 6.44, 10.03, 13.11, 13.92, 14.31, 15.92, 16.40, 16.56, 17.09, 19.03, 19.37, 19.69, 20.46, 20.98, 21.32, 22.11, 23.49, 23.74, 24.23, 24.40, 25.22, 25.28, 25.38, 25.86, 26.38, 26.47, 27.53, 27.81, 28.52, 28.89, 29.62, 30.00, 30.81, 31.46, 32.04, 32.21, 34.07, 35.30, 35.96, 36.33, 36.61, 37.18, 38.24, 38.76, and 39.35 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and gallic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 13.11, 14.31, 16.40, 16.56, 20.46, 21.32, 23.49, 23.74, 24.23, 24.40, 25.22, 25.28, 25.38, 25.86, 26.47, 27.53, 27.81, 28.52, 28.89, 31.46, and 32.04 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and gallic acid having an XRPD pattern comprising peaks at approximately 13.11, 25.22 (or 25.28), and 27.53 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 16.40 and 21.32 degrees 2θ. In one embodiment, the solid form further comprises peaks at approximately 23.49 and 25.86 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 13.11, 14.31, 16.40, 16.56, 20.46, 21.32, 23.49, 23.74, 24.23, 24.40, 25.22, 25.28, 25.38, 25.86, 26.47, 27.53, 27.81, 28.52, 28.89, 31.46, and 32.04 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and gallic acid having an XRPD pattern comprising peaks at approximately 13.11 and 21.32 degrees 2θ. In one embodiment, the solid form further comprises a peak at approximately 25.86 degree 2θ. In one embodiment, the solid form further comprises a peak at approximately 23.49 degree 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and gallic acid having an XRPD pattern comprising peaks at approximately 23.74, 28.89, and 25.86 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 24.23, 14.31, and 32.04 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 16.40, 23.49 and 21.32 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 13.11, 25.22, 27.53, and 25.28 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and gallic acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 1.

5.2.2 Cocrystal Comprising Lenalidomide and Vanillic Acid

Certain embodiments herein provide solid forms comprising lenalidomide and vanillin. In one embodiment, provided herein is a solid form comprising lenalidomide and vanillin that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and vanillin. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and vanillin. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and vanillin and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and vanillin and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and vanillin.

In some embodiments, the cocrystal comprising lenalidomide and vanillin is obtained by removing solvent from a solution containing lenalidomide and vanillin. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and vanillin on a rotary evaporator at about 65° C. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and vanillin, and subsequently storing the residue at about 75% relative humidity for 1 day. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and vanillin, and subsequently storing the residue at about 60° C. overnight. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing approximately equal molar amount of lenalidomide and vanillin.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and vanillin with a molar ratio of lenalidomide to vanillin of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to vanillin is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and vanillin is provided in FIG. 2. In some embodiments, provided herein is a solid form comprising lenalidomide and vanillin characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, or more peaks) selected from peaks located at the following or approximately the following positions: 5.02, 10.30, 12.06, 15.77, 20.70, and 31.26 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 4 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by 6 of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and vanillin having an XRPD pattern comprising peaks at approximately 12.06, 15.77, and 20.70 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 10.30 and 31.26 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 5.02, 10.30, 12.06, 15.77, 20.70, and 31.26 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and vanillin having an XRPD pattern comprising peaks at approximately 12.06 and 31.26 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 15.77 and 5.02 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 15.77, 5.02 and 20.70 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and vanillin, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 2.

5.2.3 Cocrystal Comprising Lenalidomide and Oxalic Acid

Certain embodiments herein provide solid forms comprising lenalidomide and oxalic acid. In one embodiment, provided herein is a solid form comprising lenalidomide and oxalic acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and oxalic acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and oxalic acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and oxalic acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and oxalic acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and oxalic acid.

In some embodiments, the cocrystal comprising lenalidomide and oxalic acid is obtained by grinding lenalidomide and oxalic acid together in the presence of a minor quantity of a solvent system. In some embodiments, the cocrystal comprising lenalidomide and oxalic acid is obtained by grinding approximately equal molar amount of lenalidomide and oxalic acid together in the presence of a minor quantity of a solvent system. In some embodiments, the solvent system is a mixed solvent of methanol and water. In some embodiments, the solvent system is a mixed solvent of methanol and water with a volume ratio of methanol to water of about 3:1.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and oxalic acid with a molar ratio of lenalidomide to oxalic acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to oxalic acid is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and oxalic acid is provided in FIG. 3. In some embodiments, provided herein is a solid form comprising lenalidomide and oxalic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 5.57, 6.59, 7.80, 10.86, 11.35, 12.06, 13.06, 13.21, 13.76, 14.12, 14.38, 14.78, 15.25, 15.74, 16.19, 16.51, 16.75, 17.19, 17.73, 18.10, 19.08, 19.44, 20.28, 20.69, 20.91, 21.16, 21.58, 22.13, 22.58, 23.88, 24.39, 26.09, 26.39, 26.82, 27.29, 27.60, 27.74, 28.98, 29.27, 29.96, 31.48, 32.08, 32.44, 33.15, 34.50, 34.77, 35.58, 36.03, 37.09, 37.41, 37.93, and 39.68 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and oxalic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 5.57, 10.86, 12.06, 13.06, 13.76, 15.74, 17.73, 19.08, 19.44, 20.28, 21.16, 21.58, 22.13, 22.58, 23.88, 24.39, 26.09, 26.39, 26.82, 27.29, 27.60, 27.74, 29.27, 31.48, 32.44, 33.15, 34.50, and 37.09 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and oxalic acid having an XRPD pattern comprising peaks at approximately 19.44, 22.13, and 26.82 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 13.76, 21.58, and 22.58 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 5.57, 10.86, 12.06, 13.06, 13.76, 15.74, 17.73, 19.08, 19.44, 20.28, 21.16, 21.58, 22.13, 22.58, 23.88, 24.39, 26.09, 26.39, 26.82, 27.29, 27.60, 27.74, 29.27, 31.48, 32.44, 33.15, 34.50, and 37.09 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and oxalic acid having an XRPD pattern comprising peaks at approximately 13.76, 19.44, and 26.09 degrees 2θ. In one embodiment, the solid form further comprises peaks at approximately 17.73, 21.58, and 24.39 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and oxalic acid having an XRPD pattern comprising peaks at approximately 5.57, 33.15, 34.50, 15.74, 20.28 and 10.86 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 13.06, 23.88 and 37.09 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 27.74, 32.44, 21.16, 31.48, 27.60 and 12.06 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 26.39, 24.39, 29.27, 26.09, 17.73, 19.08 and 27.29 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 21.58, 13.76 and 22.58 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 19.44, 26.82 and 22.13 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and oxalic acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 3.

5.2.4 Cocrystal Comprising Lenalidomide and Propyl Gallate

Certain embodiments herein provide solid forms comprising lenalidomide and propyl gallate. In one embodiment, provided herein is a solid form comprising lenalidomide and propyl gallate that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and propyl gallate. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and propyl gallate. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and propyl gallate and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and propyl gallate and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and propyl gallate.

In some embodiments, the cocrystal comprising lenalidomide and propyl gallate is obtained by mixing lenalidomide and propyl gallate in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and propyl gallate in a solvent system saturated with propyl gallate. In some embodiments, the cocrystal is obtained by mixing lenalidomide and propyl gallate in a solvent system saturated with propyl gallate, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and propyl gallate in a solvent system saturated with propyl gallate, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and propyl gallate in a solvent system saturated with propyl gallate. In some embodiments, the solvent system is a mixed solvent of water and methanol. In some embodiments, the solvent system is a mixed solvent of water and methanol with a volume ratio of water to methanol of about 1:1.

In some embodiments, the cocrystal comprising lenalidomide and propyl gallate is obtained by grinding lenalidomide and propyl gallate together in the presence of a minor quantity of a solvent system. In some embodiments, the cocrystal comprising lenalidomide and propyl gallate is obtained by grinding approximately equal molar amount of lenalidomide and propyl gallate together in the presence of a minor quantity of a solvent system. In some embodiments, the solvent system is a mixed solvent of methanol and water. In some embodiments, the solvent system is a mixed solvent of methanol and water with a volume ratio of methanol to water of about 3:1.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and propyl gallate with a molar ratio of lenalidomide to propyl gallate of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to propyl gallate is approximately 1:1.

A representative XRPD patterns of a solid form comprising lenalidomide and propyl gallate is provided in FIG. 4. In some embodiments, provided herein is a solid form comprising lenalidomide and propyl gallate characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 3.72, 7.73, 9.35, 9.89, 11.09, 11.79, 11.96, 12.05, 13.04, 13.17, 13.84, 14.12, 14.42, 14.84, 15.24, 15.70, 16.48, 17.21, 17.66, 18.06, 18.33, 18.47, 18.71, 19.14, 19.40, 19.58, 20.72, 20.92, 21.17, 21.52, 22.09, 22.43, 22.60, 23.89, 24.15, 24.46, 24.83, 26.23, 26.60, 27.05, 27.55, 27.75, 28.24, 29.22, 29.98, 30.22, 31.16, 31.66, 31.80, 32.09, 33.20, 33.74, 34.55, 34.88, 35.39, 37.18, 37.71, and 38.46 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and propyl gallate characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 3.72, 9.89, 11.79, 11.96, 12.05, 13.04, 15.70, 19.14, 19.58, 20.72, 21.17, 21.52, 22.09, 22.60, 23.89, 24.83, 26.23, 26.60, 27.05, 27.75, 29.22, 31.16, and 37.71 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and propyl gallate having an XRPD pattern comprising peaks at approximately 3.72, 22.09, and 26.60 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 11.96, 20.72, and 23.89 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 3.72, 9.89, 11.79, 11.96, 12.05, 13.04, 15.70, 19.14, 19.58, 20.72, 21.17, 21.52, 22.09, 22.60, 23.89, 24.83, 26.23, 26.60, 27.05, 27.75, 29.22, 31.16, and 37.71 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and propyl gallate having an XRPD pattern comprising peaks at approximately 3.72, 23.89, and 26.60 degrees 2θ. In one embodiment, the solid form further comprises peaks at approximately 11.96 and 20.72 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and propyl gallate having an XRPD pattern comprising peaks at approximately 22.60, 27.05, 11.79, 19.14, 24.83, 15.70 and 13.04 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 12.05, 27.75, 29.22, 26.23 and 11.96 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 20.72, 23.89, 22.09, 26.60 and 3.72 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and propyl gallate, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 4.

5.2.5 Cocrystal Comprising Lenalidomide and Glycolic Acid

Certain embodiments herein provide solid forms comprising lenalidomide and glycolic acid. In one embodiment, provided herein is a solid form comprising lenalidomide and glycolic acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and glycolic acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and glycolic acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and glycolic acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and glycolic acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and glycolic acid.

In some embodiments, the cocrystal comprising lenalidomide and glycolic acid is obtained by mixing lenalidomide and glycolic acid in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and glycolic acid in a solvent system saturated with glycolic acid. In some embodiments, the cocrystal is obtained by mixing lenalidomide and glycolic acid in a solvent system saturated with glycolic acid, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and glycolic acid in a solvent system saturated with glycolic acid, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and glycolic acid in a solvent system saturated with glycolic acid. In some embodiments, the solvent system is acetonitrile.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and glycolic acid with a molar ratio of lenalidomide to glycolic acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to glycolic acid is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and glycolic acid is provided in FIG. 5. In some embodiments, provided herein is a solid form comprising lenalidomide and glycolic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 7.91, 9.85, 9.88, 11.32, 11.99, 13.74, 14.36, 14.81, 15.13, 15.83, 16.26, 17.64, 17.78, 18.36, 18.43, 18.73, 19.75, 20.04, 20.16, 20.58, 20.90, 21.70, 21.88, 22.89, 23.80, 24.13, 24.85, 25.10, 25.39, 26.01, 26.14, 26.46, 27.14, 27.35, 28.32, 29.48, 29.80, 30.33, 30.54, 31.09, 31.35, 31.96, 32.59, 32.73, 33.09, 33.55, 33.91, 34.22, 34.92, 35.33, 35.83, 36.27, 38.17, 38.50, 38.99, and 39.71 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and glycolic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 9.85, 14.36, 16.26, 17.64, 17.78, 18.43, 18.73, 19.75, 20.58, 21.70, 22.89, 23.80, 24.13, 25.10, 25.39, 26.01, 26.14, and 29.48 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and glycolic acid having an XRPD pattern comprising peaks at approximately 9.85, 22.89, and 25.39 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 20.58, 24.13, and 29.48 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 9.85, 14.36, 16.26, 17.64, 17.78, 18.43, 18.73, 19.75, 20.58, 21.70, 22.89, 23.80, 24.13, 25.10, 25.39, 26.01, 26.14, and 29.48 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and glycolic acid having an XRPD pattern comprising peaks at approximately 9.85, 22.89, and 29.48 degrees 2θ. In one embodiment, the solid form further comprises peaks at approximately 17.78 and 18.43 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and glycolic acid having an XRPD pattern comprising peaks at approximately 21.70, 26.14, 25.10, 23.80, 19.75 and 14.36 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 17.78, 18.73, 18.43, 16.26 and 26.01 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 17.64, 29.48, 24.13, 20.58, 22.89, 25.39 and 9.85 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and glycolic acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 5.

5.2.6 Cocrystal Comprising Lenalidomide and Sodium Lauryl Sulfate

Certain embodiments herein provide solid forms comprising lenalidomide and sodium lauryl sulfate. In one embodiment, provided herein is a solid form comprising lenalidomide and sodium lauryl sulfate that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and sodium lauryl sulfate. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and sodium lauryl sulfate. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and sodium lauryl sulfate and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and sodium lauryl sulfate and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and sodium lauryl sulfate.

In some embodiments, the cocrystal comprising lenalidomide and sodium lauryl sulfate is obtained by mixing lenalidomide and sodium lauryl sulfate in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and sodium lauryl sulfate in a solvent system saturated with sodium lauryl sulfate. In some embodiments, the cocrystal is obtained by mixing lenalidomide and sodium lauryl sulfate in a solvent system saturated with sodium lauryl sulfate, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and sodium lauryl sulfate in a solvent system saturated with sodium lauryl sulfate, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and sodium lauryl sulfate in a solvent system saturated with sodium lauryl sulfate. In some embodiments, the solvent system is methanol.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and sodium lauryl sulfate with a molar ratio of lenalidomide to sodium lauryl sulfate of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to sodium lauryl sulfate is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and sodium lauryl sulfate is provided in FIG. 6. In some embodiments, provided herein is a solid form comprising lenalidomide and sodium lauryl sulfate characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 2.19, 2.69, 4.38, 5.34, 6.53, 8.00, 10.63, 11.36, 13.30, 14.44, 14.86, 15.85, 16.28, 17.66, 18.06, 20.17, 20.62, 21.03, 21.42, 22.80, 23.85, 24.16, 24.88, 25.29, 26.01, 27.98, 28.36, 29.10, 31.43, 31.99, 32.71, 33.16, 33.59, 35.08, and 36.27 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and sodium lauryl sulfate characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 2.19, 2.69, 4.38, 5.34, 6.53, 8.00, 10.63, 14.44, 14.86, 15.85, 16.28, 17.66, 20.17, 20.62, 21.03, 21.42, 23.85, 24.16, 26.01, and 32.71 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and sodium lauryl sulfate having an XRPD pattern comprising peaks at approximately 2.19, 2.69, and 5.34 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 8.00, 17.66, and 20.62 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 2.19, 2.69, 4.38, 5.34, 6.53, 8.00, 10.63, 14.44, 14.86, 15.85, 16.28, 17.66, 20.17, 20.62, 21.03, 21.42, 23.85, 24.16, 26.01, and 32.71 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and sodium lauryl sulfate having an XRPD pattern comprising peaks at approximately 8.00 and 5.34 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 5.34 and 2.19 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 2.19 and 2.69 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and sodium lauryl sulfate, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 6.

5.2.7 Cocrystal Comprising Lenalidomide and Magnesium Bromide

Certain embodiments herein provide solid forms comprising lenalidomide and magnesium bromide. In one embodiment, provided herein is a solid form comprising lenalidomide and magnesium bromide that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and magnesium bromide. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and magnesium bromide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and magnesium bromide and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and magnesium bromide and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and magnesium bromide.

In some embodiments, the cocrystal comprising lenalidomide and magnesium bromide is obtained by mixing lenalidomide and magnesium bromide in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and magnesium bromide in a solvent system saturated with magnesium bromide. In some embodiments, the cocrystal is obtained by mixing lenalidomide and magnesium bromide in a solvent system saturated with magnesium bromide, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and magnesium bromide in a solvent system saturated with magnesium bromide, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and magnesium bromide in a solvent system saturated with magnesium bromide. In some embodiments, the solvent system is acetone.

In some embodiments, the cocrystal comprising lenalidomide and magnesium bromide is obtained by removing solvent from a solution containing lenalidomide and magnesium bromide. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and magnesium bromide on a rotary evaporator at about 65° C. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and magnesium bromide, and subsequently storing the residue at about 75% relative humidity for 1 day. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and magnesium bromide, and subsequently storing the residue at about 60° C. overnight. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing approximately equal molar amount of lenalidomide and magnesium bromide. In some embodiments, the solvent system is methanol.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and magnesium bromide with a molar ratio of lenalidomide to magnesium bromide of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to magnesium bromide is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and magnesium bromide is provided in FIG. 7A. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 8.43, 8.94, 12.02, 12.78, 13.88, 14.29, 15.06, 15.75, 15.92, 16.22, 16.59, 16.98, 18.06, 18.63, 19.29, 19.68, 20.39, 21.01, 21.82, 21.98, 22.72, 23.21, 23.95, 24.27, 25.44, 25.71, 25.92, 26.07, 26.58, 26.84, 27.59, 27.96, 28.26, 28.52, 28.76, 29.18, 29.59, 29.92, 31.51, 32.07, 32.85, 33.00, 34.43, 36.45, 37.03, 37.90, and 38.72 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 12.02, 14.29, 15.06, 15.75, 15.92, 16.22, 16.59, 16.98, 18.63, 19.29, 21.01, 21.82, 21.98, 23.21, 23.95, 24.27, 25.44, 25.71, 26.07, 26.84, 28.52, 28.76, 29.59, 29.92, 31.51, 32.07, 32.85, 33.00, 37.90, and 38.72 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

A representative XRPD pattern of a solid form comprising lenalidomide and magnesium bromide is provided in FIG. 7B. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 12.02, 14.29, 15.06, 15.75, 15.92, 16.22, 16.59, 16.98, 18.63, 19.29, 21.82, 21.98, 23.21, 23.95, 24.27, 25.44, 25.71, 26.07, 26.84, 28.52, 28.76, 29.59, 29.92, 31.51, 32.07, 37.90, and 38.72 degrees 2θ. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide having an XRPD pattern comprising peaks at approximately 15.75, 21.98, and 28.76 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 18.63, 24.27, and 25.71 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 12.02, 14.29, 15.06, 15.75, 15.92, 16.22, 16.59, 16.98, 18.63, 19.29, 21.82, 21.98, 23.21, 23.95, 24.27, 25.44, 25.71, 26.07, 26.84, 28.52, 28.76, 29.59, 29.92, 31.51, 32.07, 37.90, and 38.72 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide having an XRPD pattern comprising peaks at approximately 25.44, 16.98, 26.07, 38.72, 14.29 and 12.02 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 29.59, 37.90, 15.92, 15.06, 16.59, 25.71 and 26.84 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 24.27, 31.51, 23.95, 16.22 and 33.00 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 23.21, 21.82 and 29.92 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 32.07, 19.29, 15.75 and 18.63 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 28.52 and 21.98 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 32.85 and 21.01 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 7A or 7B.

Representative thermal characteristics (DSC and TGA) of a solid form comprising lenalidomide and magnesium bromide are provided in FIG. 8. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide that exhibits a thermal event, as characterized by DSC, with a peak temperature of about 123° C., or with a temperature of about 180 to about 250° C. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide that exhibits thermal events, as characterized by DSC, with a peak temperature of about 123° C., and with a temperature of about 180 to about 250° C. Without being limited by any particular theory, the event with a temperature of about 180 to about 250° C. corresponds to melting and/or decomposition. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, wherein the solid form is characterized by a DSC thermogram which matches the DSC thermogram presented in FIG. 8.

In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, which exhibits, as characterized by TGA, about 16% weight loss upon heating from about 70 to about 200° C. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, wherein the solid form is characterized by a TGA thermogram which matches the TGA thermogram presented in FIG. 8.

A representative DVS isotherm plot of a solid form comprising lenalidomide and magnesium bromide is provided in FIG. 9. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, wherein the solid form is characterized by a DVS isotherm plot which matches the DVS isotherm plot presented in FIG. 9. In some embodiments, a mass gain of about 43% occurs when the relative humidity (RH) is increased from about 5% to about 95%. In some embodiments, a mass loss of about 29% occurs when the relative humidity (RH) is decreased from about 95% to about 5%. In one embodiment, the solid form remains unchanged, as characterized by XRPD (data not shown), after it undergoes adsorption/desorption cycles.

A representative Raman spectrum of a solid form comprising lenalidomide and magnesium bromide is provided in FIG. 10. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, wherein the solid form is characterized by a Raman spectrum which matches the Raman spectrum presented in FIG. 10.

A representative 1H-NMR spectrum of a solid form comprising lenalidomide and magnesium bromide is provided in FIG. 11. In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, wherein the solid form is characterized by a 1H-NMR spectrum which matches the 1H-NMR spectrum presented in FIG. 11.

In some embodiments, provided herein is a solid form comprising lenalidomide and magnesium bromide, which has a chunk crystal habit. A representative crystal habit is presented in FIG. 12.

5.2.8 Cocrystal Comprising Lenalidomide and Malonic Acid

Certain embodiments herein provide solid forms comprising lenalidomide and malonic acid. In one embodiment, provided herein is a solid form comprising lenalidomide and malonic acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and malonic acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and malonic acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and malonic acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and malonic acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and malonic acid.

In some embodiments, the cocrystal comprising lenalidomide and malonic acid is obtained by mixing lenalidomide and malonic acid in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and malonic acid in a solvent system saturated with malonic acid. In some embodiments, the cocrystal is obtained by mixing lenalidomide and malonic acid in a solvent system saturated with malonic acid, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and malonic acid in a solvent system saturated with malonic acid, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and malonic acid in a solvent system. In some embodiments, the solvent is acetonitrile.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and malonic acid with a molar ratio of lenalidomide to malonic acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to malonic acid is approximately 1:1. In some embodiments, the molar ratio of lenalidomide to malonic acid is approximately 2:1.

A representative XRPD pattern of a solid form comprising lenalidomide and malonic acid is provided in FIG. 13A. A representative XRPD pattern of a solid form comprising lenalidomide and malonic acid is also provided in FIG. 13B. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or more peaks) selected from peaks located at the following or approximately the following positions: 5.87, 9.28, 9.68, 11.71, 12.03, 13.23, 13.80, 14.15, 15.81, 16.27, 16.60, 17.24, 17.42, 17.58, 17.87, 18.10, 18.31, 18.83, 19.49, 20.71, 21.41, 21.69, 22.14, 22.58, 22.97, 23.10, 23.52, 23.77, 24.49, 24.92, 25.83, 26.69, 27.44, 27.77, 28.26, 28.52, 29.27, 29.49, 30.10, 30.42, 30.85, 31.51, 31.66, 31.89, 32.19, 32.57, 32.87, 33.46, 33.80, 34.28, 34.66, 35.22, 35.74, 36.76, 37.04, 37.37, 37.71, 38.14, 39.23, and 39.37 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or more peaks) selected from peaks located at the following or approximately the following positions: 5.87, 15.81, 18.83, 20.71, 21.69, 22.58, 22.97, 23.10, 23.52, 26.69, 27.44, 28.52, 30.10, 30.85, and 31.89 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid having an XRPD pattern comprising peaks at approximately 18.83, 20.71, and 22.97 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 5.87 and 15.81 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 23.10, 26.69, and 27.44 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 5.87, 15.81, 18.83, 20.71, 21.69, 22.58, 22.97, 23.10, 23.52, 26.69, 27.44, 28.52, 30.10, 30.85, and 31.89 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid having an XRPD pattern comprising peaks at approximately 30.10, 21.69 and 23.52 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 31.89, 30.85, 23.10, 22.58 and 28.52 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 26.69, 5.87, 15.81 and 27.44 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 18.83, 20.71 and 22.97 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 13A or 13B.

Representative thermal characteristics (DSC and TGA) of a solid form comprising lenalidomide and malonic acid are provided in FIG. 14. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid that exhibits a thermal event, as characterized by DSC, with a peak temperature of about 102° C., with a peak temperature of about 111° C., or with a peak temperature of about 159° C. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid that exhibits thermal events, as characterized by DSC, with a peak temperature of about 102° C., with a peak temperature of about 111° C., and with a peak temperature of about 159° C. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, wherein the solid form is characterized by a DSC thermogram which matches the DSC thermogram presented in FIG. 14.

In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, which exhibits, as characterized by TGA, about 43% weight loss upon heating from about 100 to about 180° C. In one embodiment, without being limited by any particular theory, the weight loss corresponds to decomposition. In one embodiment, without being limited by any particular theory, the weight loss corresponds to loss of water. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, wherein the solid form is characterized by a TGA thermogram which matches the TGA thermogram presented in FIG. 14.

A representative DVS isotherm plot of a solid form comprising lenalidomide and malonic acid is provided in FIG. 15. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, wherein the solid form is characterized by a DVS isotherm plot which matches the DVS isotherm plot presented in FIG. 15. In some embodiments, a mass gain of about 42% occurs when the relative humidity (RH) is increased from about 5% to about 95%. In some embodiments, a mass loss of about 25% occurs when the relative humidity (RH) is decreased from about 95% to about 5%. In one embodiment, the solid form remains unchanged, as characterized by XRPD (data not shown), after it undergoes adsorption/desorption cycles.

A representative Raman spectrum of a solid form comprising lenalidomide and malonic acid is provided in FIG. 16. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, wherein the solid form is characterized by a Raman spectrum which matches the Raman spectrum presented in FIG. 16.

A representative 1H-NMR spectrum of a solid form comprising lenalidomide and malonic acid is provided in FIG. 17A. A representative 1H-NMR spectrum of a solid form comprising lenalidomide and malonic acid is also provided in FIG. 17B. In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, wherein the solid form is characterized by a 1H-NMR spectrum which matches the 1H-NMR spectrum presented in FIG. 17A or 17B.

In some embodiments, provided herein is a solid form comprising lenalidomide and malonic acid, which has a chunk crystal habit. A representative crystal habit is presented in FIG. 18.

5.2.9 Cocrystal Comprising Lenalidomide and L-Tartaric Acid

Certain embodiments herein provide solid forms comprising lenalidomide and L-tartaric acid. In one embodiment, provided herein is a solid form comprising lenalidomide and L-tartaric acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and L-tartaric acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and L-tartaric acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and L-tartaric acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and L-tartaric acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and L-tartaric acid.

In some embodiments, the cocrystal comprising lenalidomide and L-tartaric acid is obtained by mixing lenalidomide and L-tartaric acid in a solvent system. In some embodiments, the cocrystal is obtained by mixing lenalidomide and L-tartaric acid in a solvent system saturated with L-tartaric acid. In some embodiments, the cocrystal is obtained by mixing lenalidomide and L-tartaric acid in a solvent system saturated with L-tartaric acid, and subsequently stirring the mixture at room temperature for about 24 hours. In some embodiments, the cocrystal is obtained by mixing lenalidomide and L-tartaric acid in a solvent system saturated with L-tartaric acid, subsequently stirring the mixture at room temperature for about 24 hours, and isolating the solid by centrifugation. In some embodiments, the cocrystal is obtained by mixing approximately equal molar amount of lenalidomide and L-tartaric acid in a solvent system. In some embodiments, the solvent is acetonitrile.

In some embodiments, the cocrystal comprising lenalidomide and L-tartaric acid is obtained by grinding lenalidomide and L-tartaric acid together in the presence of a minor quantity of a solvent system. In some embodiments, the cocrystal comprising lenalidomide and L-tartaric acid is obtained by grinding approximately equal molar amount of lenalidomide and L-tartaric acid together in the presence of a minor quantity of a solvent system. In some embodiments, the solvent system is a mixed solvent of methanol and water. In some embodiments, the solvent system is a mixed solvent of methanol and water with a volume ratio of methanol to water of about 3:1.

In some embodiments, the cocrystal comprising lenalidomide and L-tartaric acid is obtained by removing solvent from a solution containing lenalidomide and L-tartaric acid. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and L-tartaric acid on a rotary evaporator at about 65° C. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and L-tartaric acid, and subsequently storing the residue at about 75% relative humidity for 1 day. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and L-tartaric acid, and subsequently storing the residue at about 60° C. overnight. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing approximately equal molar amount of lenalidomide and L-tartaric acid. In some embodiments, the solvent system is methanol.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and L-tartaric acid with a molar ratio of lenalidomide to L-tartaric acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to L-tartaric acid is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and L-tartaric acid is provided in FIG. 19A. A representative XRPD pattern of a solid form comprising lenalidomide and L-tartaric acid is also provided in FIG. 19B. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 5.66, 9.32, 11.04, 11.32, 12.08, 12.52, 12.82, 13.19, 13.91, 14.09, 14.64, 15.28, 15.78, 16.53, 17.03, 17.49, 18.09, 18.75, 19.18, 19.84, 20.32, 21.36, 22.16, 22.65, 23.86, 24.43, 24.77, 25.21, 25.78, 26.67, 27.32, 27.68, 28.47, 28.79, 29.27, 31.17, 31.95, 34.59, 34.90, 36.94, 37.40, and 37.95 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 5.66, 11.04, 12.08, 13.19, 14.09, 14.64, 15.78, 16.53, 17.03, 17.49, 18.09, 18.75, 19.84, 20.32, 21.36, 22.16, 22.65, 23.86, 24.77, 25.78, 26.67, 27.32, 27.68, 28.47, 29.27, 31.17, 31.95, 34.59, 36.94, and 37.40 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid having an XRPD pattern comprising peaks at approximately 5.66, 18.09, and 24.77 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 19.84, 21.36, and 23.86 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 13.19, 16.53, and 25.78 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 5.66, 11.04, 12.08, 13.19, 14.09, 14.64, 15.78, 16.53, 17.03, 17.49, 18.09, 18.75, 19.84, 20.32, 21.36, 22.16, 22.65, 23.86, 24.77, 25.78, 26.67, 27.32, 27.68, 28.47, 29.27, 31.17, 31.95, 34.59, 36.94, and 37.40 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid having an XRPD pattern comprising peaks at approximately 36.94, 37.40, 25.78, 15.78, 17.03, 14.09 and 34.59 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 27.32, 31.17, 11.04, 17.49, 14.64 and 29.27 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 21.36, 28.47, 31.95, 16.53 and 22.16 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 12.08, 13.19, 23.86 and 20.32 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 27.68, 19.84, 18.75 and 24.77 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 26.67, 18.09, 22.65 and 5.66 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 19A or 19B.

Representative thermal characteristics (DSC and TGA) of a solid form comprising lenalidomide and L-tartaric acid are provided in FIG. 20. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid that exhibits a thermal event, as characterized by DSC, with a peak temperature of about 148° C., or with a temperature of about 180 to about 250° C. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid that exhibits thermal events, as characterized by DSC, with a peak temperature of about 148° C., and with a temperature of about 180 to about 250° C. Without being limited by any particular theory, the event with a temperature of about 180 to about 250° C. corresponds to melting and/or decomposition. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, wherein the solid form is characterized by a DSC thermogram which matches the DSC thermogram presented in FIG. 20.

In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, which exhibits, as characterized by TGA, about 12% weight loss upon heating from about 30 to about 200° C. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, wherein the solid form is characterized by a TGA thermogram which matches the TGA thermogram presented in FIG. 20.

A representative DVS isotherm plot of a solid form comprising lenalidomide and L-tartaric acid is provided in FIG. 21. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, wherein the solid form is characterized by a DVS isotherm plot which matches the DVS isotherm plot presented in FIG. 21. In some embodiments, a mass gain of about 22% occurs when the relative humidity (RH) is increased from about 5% to about 95%. In some embodiments, a mass loss of about 18% occurs when the relative humidity (RH) is decreased from about 95% to about 5%. In one embodiment, the solid form remains unchanged, as characterized by XRPD (data not shown), after it undergoes adsorption/desorption cycles.

A representative 1H-NMR spectrum of a solid form comprising lenalidomide and L-tartaric acid is provided in FIG. 22. In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, wherein the solid form is characterized by a′14-NMR spectrum which matches the 1H-NMR spectrum presented in FIG. 22.

In some embodiments, provided herein is a solid form comprising lenalidomide and L-tartaric acid, which has a chunk crystal habit. A representative crystal habit is presented in FIG. 23.

5.2.10 Cocrystal Comprising Lenalidomide and Hippuric Acid

Certain embodiments herein provide solid forms comprising lenalidomide and hippuric acid. In one embodiment, provided herein is a solid form comprising lenalidomide and hippuric acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and hippuric acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and hippuric acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and hippuric acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and hippuric acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and hippuric acid.

In some embodiments, the cocrystal comprising lenalidomide and hippuric acid is obtained by removing solvent from a solution containing lenalidomide and hippuric acid. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and hippuric acid on a rotary evaporator at about 65° C. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and hippuric acid, and subsequently storing the residue at about 75% relative humidity for 1 day. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and hippuric acid, and subsequently storing the residue at about 60° C. overnight. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing approximately equal molar amount of lenalidomide and hippuric acid. In some embodiments the solvent is methanol.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and hippuric acid with a molar ratio of lenalidomide to hippuric acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to hippuric acid is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and hippuric acid is provided in FIG. 24. In some embodiments, provided herein is a solid form comprising lenalidomide and hippuric acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more peaks) selected from peaks located at the following or approximately the following positions: 11.71, 11.82, 12.05, 12.82, 13.12, 14.09, 15.21, 15.73, 16.55, 17.14, 19.32, 21.42, 22.01, 22.10, 22.51, 23.04, 23.91, 26.27, 26.68, 27.56, 28.89, 33.22, and 35.27 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and hippuric acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more peaks) selected from peaks located at the following or approximately the following positions: 12.05, 12.82, 13.12, 15.73, 19.32, 21.42, 22.10, 23.04, 23.91, 26.68, 27.56, 28.89, and 35.27 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and hippuric acid having an XRPD pattern comprising peaks at approximately 13.12, 21.42, and 28.89 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 22.10, 27.56, and 35.27 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 12.05, 12.82, 13.12, 15.73, 19.32, 21.42, 22.10, 23.04, 23.91, 26.68, 27.56, 28.89, and 35.27 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and hippuric acid having an XRPD pattern comprising peaks at approximately 21.42, 28.89, and 35.27 degrees 2θ. In one embodiment, the solid form further comprises a peak at approximately 12.05 degree 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and hippuric acid having an XRPD pattern comprising peaks at approximately 23.04, 19.32, 15.73 and 12.05 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 23.91, 12.82, 26.68 and 22.10 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 35.27 and 27.56 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 21.42, 13.12 and 28.89 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and hippuric acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 24.

5.2.11 Cocrystal Comprising Lenalidomide and Zinc Chloride

Certain embodiments herein provide solid forms comprising lenalidomide and zinc chloride. In one embodiment, provided herein is a solid form comprising lenalidomide and zinc chloride that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and zinc chloride. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and zinc chloride. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and zinc chloride and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and zinc chloride and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and zinc chloride.

In some embodiments, the cocrystal comprising lenalidomide and zinc chloride is obtained by grinding lenalidomide and zinc chloride together in the presence of a minor quantity of a solvent system. In some embodiments, the cocrystal comprising lenalidomide and zinc chloride is obtained by grinding approximately equal molar amount of lenalidomide and zinc chloride acid together in the presence of a minor quantity of a solvent system. In some embodiments, the solvent system is a mixed solvent of methanol and water. In some embodiments, the solvent system is a mixed solvent of methanol and water with a volume ratio of methanol to water of about 3:1.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and zinc chloride with a molar ratio of lenalidomide to zinc chloride of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to zinc chloride is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and zinc chloride is provided in FIG. 25. In some embodiments, provided herein is a solid form comprising lenalidomide and zinc chloride characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 7.50, 8.98, 14.01, 14.65, 15.02, 15.31, 15.80, 17.27, 18.00, 18.30, 18.68, 19.05, 19.30, 19.54, 20.60, 20.82, 21.07, 22.33, 24.46, 25.40, 26.48, 28.11, 28.86, 29.21, 29.78, 30.30, 30.51, 31.07, 31.97, 33.38, 34.03, 36.19, 37.14, 38.10, and 39.52 degrees 2θ, plus or minus 0.10. In some embodiments, provided herein is a solid form comprising lenalidomide and zinc chloride characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, or more peaks) selected from peaks located at the following or approximately the following positions: 14.01, 14.65, 15.02, 15.31, 15.80, 17.27, 18.00, 18.30, 18.68, 19.05, 19.30, 19.54, 20.60, 20.82, 21.07, 22.33, 24.46, 26.48, 28.11, 28.86, 29.21, 29.78, 30.30, 30.51, 31.97, 34.03, 38.10, and 39.52 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by at least 10 of the peaks. In some embodiments, the solid form is characterized by at least 13 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and zinc chloride having an XRPD pattern comprising peaks at approximately 17.27, 18.00, and 24.46 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 15.80, 20.82, and 22.33 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 15.02, 19.54, and 29.78 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 14.01, 14.65, 15.02, 15.31, 15.80, 17.27, 18.00, 18.30, 18.68, 19.05, 19.30, 19.54, 20.60, 20.82, 21.07, 22.33, 24.46, 26.48, 28.11, 28.86, 29.21, 29.78, 30.30, 30.51, 31.97, 34.03, 38.10, and 39.52 degrees 2θ.

In some embodiments, provided herein is a solid form comprising lenalidomide and zinc chloride having an XRPD pattern comprising peaks at approximately 8.98, 31.07 and 33.38 degrees 2θ. In certain embodiments, the solid form comprises peaks at approximately 7.50, 38.10, 31.97 and 20.60 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 15.31, 30.30, 28.11 and 19.05 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 39.52, 21.07, 14.65 and 19.30 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 34.03, 14.01, 28.86 and 18.30 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 30.51, 26.48, 29.21 and 18.68 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 19.54, 29.78, 15.02 and 15.80 degrees 2θ. In one embodiment, the solid form comprises peaks at approximately 20.82, 22.33, 18.00, 17.27 and 24.46 degrees 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and zinc chloride, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 25.

5.2.12 Cocrystal Comprising Lenalidomide and Benzoic Acid

Certain embodiments herein provide solid forms comprising lenalidomide and benzoic acid. In one embodiment, provided herein is a solid form comprising lenalidomide and benzoic acid that is substantially crystalline. In one embodiment, provided herein is a cocrystal comprising lenalidomide and benzoic acid. In one embodiment, provided herein is a solid form comprising a cocrystal comprising lenalidomide and benzoic acid. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and benzoic acid and (ii) an amorphous form of lenalidomide. In one embodiment, provided herein is a solid form comprising (i) a cocrystal comprising lenalidomide and benzoic acid and (ii) one or more additional crystal forms of lenalidomide. Provided herein are various embodiments, preparations, or modifications of a cocrystal comprising lenalidomide and benzoic acid.

In some embodiments, the cocrystal comprising lenalidomide and benzoic acid is obtained by removing solvent from a solution containing lenalidomide and benzoic acid. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and benzoic acid on a rotary evaporator at about 65° C. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and benzoic acid, and subsequently storing the residue at about 75% relative humidity for 1 day. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing lenalidomide and benzoic acid, and subsequently storing the residue at about 60° C. overnight. In some embodiments, the cocrystal is obtained by removing solvent from a solution containing approximately equal molar amount of lenalidomide and benzoic acid. In some embodiments the solvent is methanol.

In some embodiments, provided herein is a cocrystal comprising lenalidomide and benzoic acid with a molar ratio of lenalidomide to benzoic acid of approximately 2:1 to 1:2. In some embodiments, the molar ratio of lenalidomide to benzoic acid is approximately 1:1.

A representative XRPD pattern of a solid form comprising lenalidomide and benzoic acid is provided in FIG. 26. In some embodiments, provided herein is a solid form comprising lenalidomide and benzoic acid characterized by one or more XRPD peaks (e.g., 1, 2, 3, 4, 5, 6, 7, 8, or more peaks) selected from peaks located at the following or approximately the following positions: 5.14, 15.07, 16.18, 17.12, 23.83, 24.42, 27.66, 29.21, and 32.80 degrees 2θ, plus or minus 0.10. In some embodiments, the solid form is characterized by at least 3 of the peaks. In some embodiments, the solid form is characterized by at least 5 of the peaks. In some embodiments, the solid form is characterized by at least 7 of the peaks. In some embodiments, the solid form is characterized by all of the peaks.

In some embodiments, provided herein is a solid form comprising lenalidomide and benzoic acid having an XRPD pattern comprising peaks at approximately 16.18 and 32.80 degrees 2θ. In certain embodiments, the solid form further comprises peaks at approximately 15.07 and 24.42 degrees 2θ. In certain embodiments, the solid form further comprises a peak at approximately 5.14 degree 2θ. In one embodiment, the solid form comprises peaks at approximately 5.14, 15.07, 16.18, 17.12, 23.83, 24.42, 27.66, 29.21, and 32.80 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and benzoic acid having an XRPD pattern comprising peaks at approximately 15.07, 23.83, and 29.21 degrees 2θ. In one embodiment, the solid form further comprises peaks at approximately 24.42 and 32.80 degrees 2θ.

In one embodiment, provided herein is a solid form comprising lenalidomide and benzoic acid having an XRPD pattern comprising peaks at approximately 15.07 and 29.21 degrees 2θ. In one embodiment, the solid form further comprises a peak at approximately 32.80 degree 2θ.

In certain embodiments, the XRPD peaks above (degrees 2θ peaks) are obtained when analyzed using copper Kα radiation. In some embodiments, provided herein is a solid form comprising lenalidomide and benzoic acid, wherein the solid form is characterized by an XRPD diffraction pattern which matches the XRPD pattern presented in FIG. 26.

5.3 Second Agents

A solid form of the invention can be used with or combined with other pharmacologically active compounds (“second active agents”) in methods and compositions of the invention. It is believed that certain combinations work synergistically in the treatment of particular types of cancers, and certain diseases and conditions associated with, or characterized by, undesired angiogenesis. Immunomodulatory compounds of the invention can also work to alleviate adverse effects associated with certain second active agents, and some second active agents can be used to alleviate adverse effects associated with immunomodulatory compounds of the invention.

One or more second active ingredients or agents can be used in the methods and compositions of the invention together with a solid form of the invention. Second active agents can be large molecules (e.g., proteins, or monoclonal antibodies (e.g., Rituximab or Elotuzmab)) or small molecules (e.g., synthetic inorganic, organometallic, or organic molecules).

One or more second active ingredients or agents can be used in the methods and compositions of the invention together with a solid form of the invention. Second active agents can be large molecules (e.g., proteins) or small molecules (e.g., synthetic inorganic, organometallic, or organic molecules).

Examples of large molecule active agents include, but are not limited to, hematopoietic growth factors, cytokines, and monoclonal and polyclonal antibodies. Typical large molecule active agents are biological molecules, such as naturally occurring or artificially made proteins. Proteins that are particularly useful in this invention include proteins that stimulate the survival and/or proliferation of hematopoietic precursor cells and immunologically active poietic cells in vitro or in vivo. Others stimulate the division and differentiation of committed erythroid progenitors in cells in vitro or in vivo. Particular proteins include, but are not limited to: interleukins, such as IL-2 (including recombinant IL-II (“rIL2”) and canarypox IL-2), IL-10, IL-12, and IL-18; interferons, such as interferon alfa-2a, interferon alfa-2b, interferon alfa-n1, interferon alfa-n3, interferon beta-I a, and interferon gamma-I b; GM-CF and GM-CSF; and EPO.

Particular proteins that can be used in the methods and compositions of the invention include, but are not limited to: filgrastim, which is sold in the United States under the trade name Neupogen® (Amgen, Thousand Oaks, Calif.); sargramostim, which is sold in the United States under the trade name Leukine® (Immunex, Seattle, Wash.); and recombinant EPO, which is sold in the United States under the trade name Epogen® (Amgen, Thousand Oaks, Calif.).

Recombinant and mutated forms of GM-CSF can be prepared as described in U.S. Pat. Nos. 5,391,485; 5,393,870; and 5,229,496; all of which are incorporated herein by reference. Recombinant and mutated forms of G-CSF can be prepared as described in U.S. Pat. Nos. 4,810,643; 4,999,291; 5,528,823; and 5,580,755; all of which are incorporated herein by reference.

Large molecule active agents may be administered in the form of anti-cancer vaccines. For example, vaccines that secrete, or cause the secretion of, cytokines such as IL-2, G-CSF, and GM-CSF can be used in the methods, pharmaceutical compositions, and kits of the invention. See, e.g., Emens, L. A., et al., Curr. Opinion Mol. Ther. 3(1):77-84 (2001).

In one embodiment of the invention, the large molecule active agent reduces, eliminates, or prevents an adverse effect associated with the administration of a solid form of the invention. Depending on the particular immunomodulatory compound of the invention and the disease or disorder begin treated, adverse effects can include, but are not limited to, drowsiness and somnolence, dizziness and orthostatic hypotension, neutropenia, infections that result from neutropenia, increased HIV-viral load, bradycardia, Stevens-Johnson Syndrome and toxic epidermal necrolysis, and seizures (e.g., grand mal convulsions). A specific adverse effect is neutropenia.

Second active agents that are small molecules can also be used to alleviate adverse effects associated with the administration of a solid form of the invention. However, like some large molecules, many are believed to be capable of providing a synergistic effect when administered with (e.g., before, after or simultaneously) a solid form of the invention. Examples of small molecule second active agents include, but are not limited to, anti-cancer agents, antibiotics, immunosuppressive agents, and steroids.

Examples of anti-cancer agents include, but are not limited to: acivicin; aclarubicin; acodazole hydrochloride; acronine; adozelesin; aldesleukin; altretamine; ambomycin; ametantrone acetate; amsacrine; anastrozole; anthramycin; asparaginase; asperlin; azacitidine; azetepa; azotomycin; batimastat; benzodepa; bicalutamide; bisantrene hydrochloride; bisnafide dimesylate; bizelesin; bleomycin sulfate; brequinar sodium; bropirimine; busulfan; cactinomycin; calusterone; caracemide; carbetimer; carboplatin; carmustine; carubicin hydrochloride; carzelesin; cedefingol; celecoxib (COX-2 inhibitor); chlorambucil; cirolemycin; cisplatin; cladribine; crisnatol mesylate; cyclophosphamide; cytarabine; dacarbazine; dactinomycin; daunorubicin hydrochloride; decitabine; dexormaplatin; dezaguanine; dezaguanine mesylate; diaziquone; docetaxel; doxorubicin; doxorubicin hydrochloride; droloxifene; droloxifene citrate; dromostanolone propionate; duazomycin; edatrexate; eflomithine hydrochloride; elsamitrucin; enloplatin; enpromate; epipropidine; epirubicin hydrochloride; erbulozole; esorubicin hydrochloride; estramustine; estramustine phosphate sodium; etanidazole; etoposide; etoposide phosphate; etoprine; fadrozole hydrochloride; fazarabine; fenretinide; floxuridine; fludarabine phosphate; fluorouracil; flurocitabine; fosquidone; fostriecin sodium; gemcitabine; gemcitabine hydrochloride; hydroxyurea; idarubicin hydrochloride; ifosfamide; ilmofosine; iproplatin; irinotecan; irinotecan hydrochloride; lanreotide acetate; letrozole; leuprolide acetate; liarozole hydrochloride; lometrexol sodium; lomustine; losoxantrone hydrochloride; masoprocol; maytansine; mechlorethamine hydrochloride; megestrol acetate; melengestrol acetate; melphalan; menogaril; mercaptopurine; methotrexate; methotrexate sodium; metoprine; meturedepa; mitindomide; mitocarcin; mitocromin; mitogillin; mitomalcin; mitomycin; mitosper; mitotane; mitoxantrone hydrochloride; mycophenolic acid; nocodazole; nogalamycin; ormaplatin; oxisuran; taxane, paclitaxel; pegaspargase; peliomycin; pentamustine; peplomycin sulfate; perfosfamide; pipobroman; piposulfan; piroxantrone hydrochloride; plicamycin; plomestane; porfimer sodium; porfiromycin; prednimustine; procarbazine hydrochloride; puromycin; puromycin hydrochloride; pyrazofurin; riboprine; safingol; safingol hydrochloride; semustine; simtrazene; sparfosate sodium; sparsomycin; spirogermanium hydrochloride; spiromustine; spiroplatin; streptonigrin; streptozocin; sulofenur; talisomycin; tecogalan sodium; taxotere; tegafur; teloxantrone hydrochloride; temoporfin; teniposide; teroxirone; testolactone; thiamiprine; thioguanine; thiotepa; tiazofurin; tirapazamine; toremifene citrate; trestolone acetate; triciribine phosphate; trimetrexate; trimetrexate glucuronate; triptorelin; tubulozole hydrochloride; uracil mustard; uredepa; vapreotide; verteporfin; vinblastine sulfate; vincristine sulfate; vindesine; vindesine sulfate; vinepidine sulfate; vinglycinate sulfate; vinleurosine sulfate; vinorelbine tartrate; vinrosidine sulfate; vinzolidine sulfate; vorozole; zeniplatin; zinostatin; zorubicin hydrochloride, and proteasome inhibitors such as bortezomib, carfilzomib, ixazomib, decitibine, and romedepsin. In one embodiment, the anti-cancer agent is taxane, paclitaxel, bortezomib, carfilzomib, ixazomib, decitibine, or romedepsin.

Other anti-cancer drugs include, but are not limited to: 20-epi-1,25 dihydroxyvitamin D3; 5-ethynyluracil; abiraterone; aclarubicin; acylfulvene; adecypenol; adozelesin; aldesleukin; ALL-TK antagonists; altretamine; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; amsacrine; anagrelide; anastrozole; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti-dorsalizing morphogenetic protein-1; antiandrogen, prostatic carcinoma; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP-DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastatin 1; axinastatin 2; axinastatin 3; azasetron; azatoxin; azatyrosine; baccatin III derivatives; balanol; batimastat; BCR/ABL antagonists; benzochlorins; benzoylstaurosporine; beta lactam derivatives; beta-alethine; betaclamycin B; betulinic acid; bFGF inhibitor; bicalutamide; bisantrene; bisaziridinylspermine; bisnafide; bistratene A; bizelesin; breflate; bropirimine; budotitane; buthionine sulfoximine; calcipotriol; calphostin C; camptothecin derivatives; capecitabine; carboxamide-amino-triazole; carboxyamidotriazole; CaRest M3; CARN 700; cartilage derived inhibitor; carzelesin; casein kinase inhibitors (ICOS); castanospermine; cecropin B; cetrorelix; chlorins; chloroquinoxaline sulfonamide; cicaprost; cis-porphyrin; cladribine; clomifene analogues; clotrimazole; collismycin A; collismycin B; combretastatin A4; combretastatin analogue; conagenin; crambescidin 816; crisnatol; cryptophycin 8; cryptophycin A derivatives; curacin A; cyclopentanthraquinones; cycloplatam; cypemycin; cytarabine ocfosfate; cytolytic factor; cytostatin; dacliximab; decitabine; dehydrodidemnin B; deslorelin; dexamethasone; dexifosfamide; dexrazoxane; dexverapamil; diaziquone; didemnin B; didox; diethylnorspermine; dihydro-5-azacytidine; dihydrotaxol, 9-; dioxamycin; diphenyl spiromustine; docetaxel; docosanol; dolasetron; doxifluridine; doxorubicin; droloxifene; dronabinol; duocarmycin SA; ebselen; ecomustine; edelfosine; edrecolomab; eflornithine; elemene; emitefur; epirubicin; epristeride; estramustine analogue; estrogen agonists; estrogen antagonists; etanidazole; etoposide phosphate; exemestane; fadrozole; fazarabine; fenretinide; filgrastim; finasteride; flavopiridol; flezelastine; fluasterone; fludarabine; fluorodaunorunicin hydrochloride; forfenimex; formestane; fostriecin; fotemustine; gadolinium texaphyrin; gallium nitrate; galocitabine; ganirelix; gelatinase inhibitors; gemcitabine; glutathione inhibitors; hepsulfam; heregulin; hexamethylene bisacetamide; hypericin; ibandronic acid; idarubicin; idoxifene; idramantone; ilmofosine; ilomastat; imatinib (e.g., Gleevec®); imiquimod; immunostimulant peptides; insulin-like growth factor-1 receptor inhibitor; interferon agonists; interferons; interleukins; iobenguane; iododoxorubicin; ipomeanol, 4-; iroplact; irsogladine; isobengazole; isohomohalicondrin B; itasetron; jasplakinolide; kahalalide F; lamellarin-N triacetate; lanreotide; leinamycin; lenograstim; lentinan sulfate; leptolstatin; letrozole; leukemia inhibiting factor; leukocyte alpha interferon; leuprolide+estrogen+progesterone; leuprorelin; levamisole; liarozole; linear polyamine analogue; lipophilic disaccharide peptide; lipophilic platinum compounds; lissoclinamide 7; lobaplatin; lombricine; lometrexol; lonidamine; losoxantrone; loxoribine; lurtotecan; lutetium texaphyrin; lysofylline; lytic peptides; maitansine; mannostatin A; marimastat; masoprocol; maspin; matrilysin inhibitors; matrix metalloproteinase inhibitors; menogaril; merbarone; meterelin; methioninase; metoclopramide; MIF inhibitor; mifepristone; miltefosine; mirimostim; mitoguazone; mitolactol; mitomycin analogues; mitonafide; mitotoxin fibroblast growth factor-saporin; mitoxantrone; mofarotene; molgramostim; Erbitux, human chorionic gonadotrophin; monophosphoryl lipid A+myobacterium cell wall sk; mopidamol; mustard anticancer agent; mycaperoxide B; mycobacterial cell wall extract; myriaporone; N-acetyldinaline; N-substituted benzamides; nafarelin; nagrestip; naloxone+pentazocine; napavin; naphterpin; nartograstim; nedaplatin; nemorubicin; neridronic acid; nilutamide; nisamycin; nitric oxide modulators; nitroxide antioxidant; nitrullyn; oblimersen (Genasense®); O.sup.6-benzylguanine; octrcotide; okicenone; oligonucleotides; onapristone; ondansetron; ondansetron; oracin; oral cytokine inducer; ormaplatin; osaterone; oxaliplatin; oxaunomycin; paclitaxel; paclitaxel analogues; paclitaxel derivatives; palauamine; palmitoylrhizoxin; pamidronic acid; panaxytriol; panomifene; parabactin; pazelliptine; pegaspargase; peldesine; pentosan polysulfate sodium; pentostatin; pentrozole; perflubron; perfosfamide; perillyl alcohol; phenazinomycin; phenylacetate; phosphatase inhibitors; picibanil; pilocarpine hydrochloride; pirarubicin; piritrexim; placetin A; placetin B; plasminogen activator inhibitor; platinum complex; platinum compounds; platinum-triamine complex; porfimer sodium; porfiromycin; prednisone; propyl bis-acridone; prostaglandin J2; proteasome inhibitors; protein A-based immune modulator; protein kinase C inhibitor; protein kinase C inhibitors, microalgal; protein tyrosine phosphatase inhibitors; purine nucleoside phosphorylase inhibitors; purpurins; pyrazoloacridine; pyridoxylated hemoglobin polyoxyethylene conjugate; raf antagonists; raltitrexed; ramosetron; ras farnesyl protein transferase inhibitors; ras inhibitors; ras-GAP inhibitor; retelliptine demethylated; rhenium Re 186 etidronate; rhizoxin; ribozymes; RII retinamide; rohitukine; romurtide; roquinimex; rubiginone B1; ruboxyl; safingol; saintopin; SarCNU; sarcophytol A; sargramostim; Sdi 1 mimetics; semustine; senescence derived inhibitor 1; sense oligonucleotides; signal transduction inhibitors; sizofiran; sobuzoxane; sodium borocaptate; sodium phenylacetate; solverol; somatomedin binding protein; sonermin; sparfosic acid; spicamycin D; spiromustine; splenopentin; spongistatin 1; squalamine; stipiamide; stromelysin inhibitors; sulfinosine; superactive vasoactive intestinal peptide antagonist; suradista; suramin; swainsonine; tallimustine; tamoxifen methiodide; tauromustine; tazarotene; tecogalan sodium; tegafur; tellurapyrylium; telomerase inhibitors; temoporfin; teniposide; tetrachlorodecaoxide; tetrazomine; thaliblastine; thiocoraline; thrombopoietin; thrombopoietin mimetic; thymalfasin; thymopoietin receptor agonist; thymotrinan; thyroid stimulating hormone; tin ethyl etiopurpurin; tirapazamine; titanocene bichloride; topsentin; toremifene; translation inhibitors; tretinoin; triacetyluridine; triciribine; trimetrexate; triptorelin; tropisetron; turosteride; tyrosine kinase inhibitors; tyrphostins; UBC inhibitors; ubenimex; urogenital sinus-derived growth inhibitory factor; urokinase receptor antagonists; vapreotide; variolin B; velaresol; veramine; verdins; verteporfin; vinorelbine; vinxaltine; vitaxin; vorozole; zanoterone; zeniplatin; zilascorb; and zinostatin stimalamer.

Specific second active agents include, but are not limited to, rituximab, oblimersen (Genasense®), remicade, docetaxel, celecoxib, melphalan, dexamethasone (Decadron®), steroids, gemcitabine, cisplatinum, temozolomide, etoposide, cyclophosphamide, temodar, carboplatin, procarbazine, gliadel, tamoxifen, topotecan, methotrexate, Arisa®, taxol, taxotere, fluorouracil, leucovorin, irinotecan, xeloda, CPT-11, interferon alpha, pegylated interferon alpha (e.g., PEG INTRON-A), capecitabine, cisplatin, thiotepa, fludarabine, carboplatin, liposomal daunorubicin, cytarabine, doxetaxol, pacilitaxel, vinblastine, IL-2, GM-CSF, dacarbazine, vinorelbine, zoledronic acid, palmitronate, biaxin, busulphan, prednisone, bisphosphonate, arsenic trioxide, vincristine, doxorubicin (Doxil®), paclitaxel, ganciclovir, adriamycin, estramustine sodium phosphate (Emcyt®), sulindac, and etoposide.

5.4 Methods of Treatments and Prevention

Provided herein are methods of treating, preventing, and/or managing various diseases or disorders using a solid form provided herein. In certain embodiments, provided are methods of treating, managing, and preventing various diseases and disorders, which comprise administering to a patient in need of such treatment, prevention or management a therapeutically or prophylactically effective amount of a solid form provided herein. Examples of diseases and disorders are described herein.

Examples of diseases or disorders include, but are not limited to: cancer including hematologic cancer or solid tumor, for example, multiple myeloma, leukemia, lymphoma, sarcoma, prostate cancer, or lung cancer (e.g., small cell lung cancer); scleroderma; amyloidosis; pain, for example, complex regional pain syndrome (CRPS); myelofibrosis; myeloproliferative disease, for example, MMM; myelodysplastic syndromes (MDS); diffuse systemic sclerosis; macular degeneration; a skin disease; a pulmonary disorder; an asbestos-related disorder; a parasitic disease; an immunodeficiency disorder; a CNS disorder; a CNS injury; atherosclerosis; hemoglobinopathy; anemia, for example, sickle cell anemia; an inflammatory disease; an autoimmune disease; a viral disease; a genetic disease; an allergic disease; a bacterial disease; an ocular neovascular disease; a choroidal neovascular disease; a retina neovascular disease; rubeosis; a sleep disorder; disorders associated with angiogenesis; and TNFα related disorders.

Examples of cancer and precancerous conditions include, but are not limited to, those described in U.S. Pat. Nos. 8,207,200, 6,281,230 and 5,635,517 and in various U.S. patent including publication nos. 2004/0220144A1, published Nov. 4, 2004 (Treatment of Myelodysplastic Syndrome); 2004/0029832A1, published Feb. 12, 2004 (Treatment of Various Types of Cancer); and 2004/0087546, published May 6, 2004 (Treatment of Myeloproliferative Diseases). Examples also include those described in WO 2004/103274, published Dec. 2, 2004. All of these references are incorporated herein in their entireties by reference.

Other examples of diseases or disorders include, but are not limited to, those described in U.S. Pat. Nos. 5,712,291, 7,393,863, and 7,863,297; and U.S. Patent Application Publication Nos. 2005/0143420, 2006/0166932, 2006/0188475, 2007/0048327, 2007/0066512, 2007/0155791, 2008/0051431, 2008/0317708, 2009/0087407, 2009/0088410, 2009/0148853, 2009/0232776, 2009/0232796, 2009/0317385, 2010/0098657, 2010/0099711, and 2011/0184025; all of which are incorporated herein by reference in their entireties.

In one embodiment the disease is multiple myeloma. In certain embodiments the solid form is used to treat multiple myeloma in patients who have received at least one prior therapy. In certain embodiments the solid form is administered in combination with dexamethasone for the treatment of multiple myeloma in patients who have received at least one prior therapy.

In one embodiment the disease is transfusion-dependent anemia due to Low- or Intermediate-1-risk myelodyspastic syndromes associated with a deletion 5q cytogenetic abnormality with or without additional cytogenetic abnormalities.

In one embodiment is provided a method of treating, preventing and/or managing a disease provided herein, comprising administering to a patient in need of such treatment, prevention and/or management a therapeutically or prophylactically effective amount of a solid form comprising lenalidomide and a coformer as described herein and a therapeutically or prophylactically effective amount of a second active agent.

Certain examples of cancer include, but are not limited to, cancers of the skin, such as melanoma; lymph node; breast; cervix; uterus; gastrointestinal tract; lung; ovary; prostate; colon; rectum; mouth; brain; head and neck; throat; testes; kidney; pancreas; bone; spleen; liver; bladder; larynx; nasal passages; and AIDS-related cancers. The solid forms are also useful for treating cancers of the blood and bone marrow, such as multiple myeloma and acute and chronic leukemias, for example, lymphoblastic, myelogenous, lymphocytic, and myelocytic leukemias. The solid forms provided herein can be used for treating, preventing, or managing either primary or metastatic tumors.

Other cancers include, but are not limited to, advanced malignancy, amyloidosis, neuroblastoma, meningioma, hemangiopericytoma, multiple brain metastases, glioblastoma multiforms, glioblastoma, brain stem glioma, poor prognosis malignant brain tumor, malignant glioma, recurrent malignant glioma, anaplastic astrocytoma, anaplastic oligodendroglioma, neuroendocrine tumor, rectal adenocarcinoma, Dukes C & D colorectal cancer, unresectable colorectal carcinoma, metastatic hepatocellular carcinoma, Kaposi's sarcoma, karotype acute myeloblastic leukemia, chronic lymphocytic leukemia (CLL), Hodgkin's lymphoma, non-Hodgkin's lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma, low grade follicular lymphoma, metastatic melanoma, localized melanoma (including, but not limited to, ocular melanoma), malignant mesothelioma, malignant pleural effusion mesothelioma syndrome, peritoneal carcinoma, papillary serous carcinoma, gynecologic sarcoma, soft tissue sarcoma, cutaneous vasculitis, Langerhans cell histiocytosis, leiomyosarcoma, fibrodysplasia ossificans progressive, hormone refractory prostate cancer, resected high-risk soft tissue sarcoma, unresectable hepatocellular carcinoma, Waldenstrom's macroglobulinemia, smoldering myeloma, indolent myeloma, fallopian tube cancer, androgen independent prostate cancer, androgen dependent stage IV non-metastatic prostate cancer, hormone-insensitive prostate cancer, chemotherapy-insensitive prostate cancer, papillary thyroid carcinoma, follicular thyroid carcinoma, medullary thyroid carcinoma, and leiomyoma. In a specific embodiment, the cancer is metastatic. In another embodiment, the cancer is refractory or resistance to chemotherapy or radiation.

In one embodiment is provided a method of treating, preventing, or managing myeloproliferative disease (MPD), comprising administering to a patient in need of such treatment, prevention, or management a therapeutically or prophylactically effective amount of a solid form comprising lenalidomide and a coformer as described herein. The embodiment encompasses the treatment, prevention or management of specific sub-types of MPD such as, but not limited to, polycythemia rubra vera (PRV), primary thrombocythemia (PT), myelofibrosis with myeloid metaplasia (MMM) and agnogenic myeloid metaplasia (AMM). In one embodiment, MPD includes: polycythemia rubra vera (PRV), primary thrombocythemia (PT), and agnogenic myeloid metaplasia (AMM). In a specific embodiment, MPD excludes leukemia. In one embodiment, particular types of MPD are MMM, PRV, PT, and AMM.

In one embodiment, a solid form comprising lenalidomide and a coformer is administered to patients who are refractory to conventional treatments for myeloproliferative diseases as well as treatments using thalidomide. As used herein, the term “refractory” means the patient's response to a MPD treatment is not satisfactory by clinical standards, e.g., showing no or little improvement of symptoms or laboratory findings.

In one embodiment is provided a method of reversing, reducing, or avoiding an adverse effect associated with the administration of an active agent used to treat MPD in a patient suffering from MPD, comprising administering to a patient in need thereof a therapeutically or prophylactically effective amount of a solid form comprising lenalidomide and a coformer as described herein. Examples of active agents include, but are not limited to, the second active agents described herein.

Examples of adverse effects associated with active agents used to treat MPD include, but are not limited to: conversion to acute leukemia; severe myelosuppression; gastrointestinal toxicity such as, but not limited to, early and late-forming diarrhea and flatulence; gastrointestinal bleeding; nausea; vomiting; anorexia; leukopenia; anemia; neutropenia; asthenia; abdominal cramping; fever; pain; loss of body weight; dehydration; alopecia; dyspnea; insomnia; dizziness; mucositis; xerostomia; mucocutaneous lesions; and kidney failure.

In one embodiment, provided herein is a method of treating, preventing, or managing MPD, comprising administering to a patient (e.g., a human) a solid form comprising lenalidomide and a coformer as described herein, before, during, or after transplantation therapy.

In one embodiment, provided herein are pharmaceutical compositions, single unit dosage forms, and kits, comprising a solid form comprising lenalidomide and a coformer as described herein, a second active ingredient, and/or blood or cells for transplantation therapy. For example, a kit may comprise a solid form comprising lenalidomide and a coformer as described herein, stem cells for transplantation, an immunosuppressive agent, and an antibiotic or other drug.

The cytokines used in the methods provided herein may be naturally-occurring cytokines, or may be an artificial derivative or analog of the cytokines. For example, analogs or derivatives of erythropoietin that may be used in combination with a solid form or compound provided herein include, but are not limited to, Arancsp™ and Darbopoietin™.

Cytokines used may be purified from natural sources or recombinantly produced. Examples of recombinant cytokines that may be used in the methods provided herein include filgrastim, or recombinant granulocyte-colony stimulating factor (G-CSF), which is sold in the United States under the trade name Neupogen® (Amgen, Thousand Oaks, Calif.); sargramostim, or recombinant GM-CSF, which is sold in the United States under the trade name Leukine® (Immunex, Seattle, Wash.); recombinant Epo, which is sold in the United States under the trade name Epogen® (Amgen, Thousand Oaks, Calif.); and methionyl stem cell factor (SCF), which is sold in the United States under the trade name Ancestim™. Recombinant and mutated forms of GM-CSF can be prepared as described in U.S. Pat. Nos. 5,391,485; 5,393,870; and 5,229,496; all of which are incorporated herein by reference. Recombinant and mutated forms of G-CSF can be prepared as described in U.S. Pat. Nos. 4,810,643; 4,999,291; 5,528,823; and 5,580,755; all of which are incorporated herein by reference.

Other cytokines may be used which encourage the survival and/or proliferation of hematopoietic precursor cells and immunologically active poietic cells in vitro or in vivo, or which stimulate the division and differentiation of committed erythroid progenitors in cells in vitro or in vivo. Such cytokines include, but are not limited to: interleukins, such as IL-2 (including recombinant IL-II (“rIL2”) and canarypox IL-2), IL-10, IL-12, and IL-18; interferons, such as interferon alfa-2a, interferon alfa-2b, interferon alfa-n1, interferon alfa-n3, interferon beta-I a, and interferon gamma-I b; and G-CSF.

When administered to a person having a hemoglobinopathy, a solid form comprising lenalidomide and a coformer as described herein, particularly in the presence of Epo, particularly in the presence of the combination of TNFα, SCF, Flt-3L and GM-CSF, or more particularly in the presence of Epo and SCF, induces the production of erythrocytes, and the production of fetal hemoglobin as well as the production of AHSP. As noted above, cytokines used may include purified or recombinant forms, or analogs or derivatives of specific cytokines.

A solid form comprising lenalidomide and a coformer as described herein may also be administered in conjunction with one or more second compounds known to have, or suspected of having, a beneficial effect on a hemoglobinopathy. In this context, “beneficial effect” means any reduction of any symptom of a hemoglobinopathy or anemia.

For example, with specific reference to the hemoglobinopathy sickle cell anemia, the second compound can be a compound, other than a lenalidomide or a derivative thereof, that is known or suspected to induce the production of fetal hemoglobin. Such compounds include hydroxyurea, and butyrates or butyrate derivatives. The second compound may also be a compound that relaxes blood vessels, such as nitrous oxide, e.g., exogenously-applied or administered nitrous oxide. The second compound may also be a compound that binds directly to hemoglobin S, preventing it from assuming the sickle-inducing conformation. For example, the plant extract known as HEMOXIN™ (NIPRISAN™; see U.S. Pat. No. 5,800,819), which is an extract of a mixture of about 12 to about 17 parts by weight of Piper guineense seeds, from about 15 to about 19 parts by weight of Pterocarpus osun stem, from about 12 to about 18 parts by weight of Eugenia caryophyllata fruit, and from about 25 to about 32 parts by weight of Sorghum bicolor leaves, and optionally 15-22 parts by weight potash, wherein the mixture is extracted with cold water, has antisickling activity. The second compound may also be a Gardos channel antagonist. Examples of Gardos channel antagonists include clotrimazole and triaryl methane derivatives. The second compound may also be one that reduces red blood cell adhesion, thereby reducing the amount of clotting pervasive in sickle cell anemia.

Other hemoglobinopathies may be treated with a second compound known or suspected to be efficacious for the specific condition. For example, β thalassemia may additionally be treated with the second compound Deferoxamine, an iron chelator that helps prevent the buildup of iron in the blood, or folate (vitamin B9). Thalassemia or sickle cell anemia may also be treated with protein C as the second compound (U.S. Pat. No. 6,372,213). There is some evidence that herbal remedies can ameliorate symptoms of hemoglobinopathies, e.g., thalassemia; such remedies, and any of the specific active compounds contained therein, may also be used as a second compound in the method provided herein. See, e.g., Wu Zhikui et al. “The Effect of Bushen Shengxue Fang on β-thalassemia at the Gene Level,” Journal of Traditional Chinese Medicine 18(4): 300-303 (1998); U.S. Pat. No. 6,538,023 “Therapeutic Uses of Green Tea Polyphenols for Sickle Cell Disease”. Treatment of autoimmune hemolytic anemia can include corticosteroids as the second compound.

Second compounds that are proteins may also be derivatives or analogs of other proteins. Such derivatives may include, but are not limited to, proteins that lack carbohydrate moieties normally present in their naturally occurring forms (e.g., nonglycosylated forms), pegylated derivatives, and fusion proteins, such as proteins formed by fusing IgG1 or IgG3 to the protein or active portion of the protein of interest. See, e.g., Penichet, M. L. and Morrison, S. L., J. Immunol. Methods 248:91-101 (2001).

Cytokines and/or other compounds potentially useful in the treatment of anemia or a hemoglobinopathy may be administered at the same time as lenalidomide or a derivative thereof. In this regard, the cytokines or other compounds may be administered as formulations separate from a solid form comprising lenalidomide and a coformer, or, where possible, may be compounded with a solid form comprising lenalidomide and a coformer for administration as a single pharmaceutical composition. Alternatively, the cytokines, the other compounds, or both, may be administered separately from a solid form comprising lenalidomide and a coformer used in the methods provided herein, and may follow the same or different dosing schedules. In one embodiment, a solid form comprising lenalidomide and a coformer, cytokines, and/or any other compound useful to treat anemia or a hemoglobinopathy, are administered at the same time, but in separate pharmaceutical formulations for flexibility in administration.

In addition to the treatment combinations outlined herein, the treated individual may be given transfusions. Such transfusions may be of blood, for example matched blood, or of a blood substitute such as Hemospan™ or Hemospan™ PS (Sangart).

In any of the treatment combinations described herein, the treated individual is eukaryotic. In one embodiment, the treated individual is a mammal, for example a human.

The methods described herein may be used to treat any anemia, including anemia resulting from a hemoglobinopathy. Hemoglobinopathies and anemias treatable by the methods provided herein may be genetic in origin, such as sickle-cell anemia or thalassemias. The hemoglobinopathy may be due to a disease, such as cancer, including, but not limited to, cancers of the hematopoietic or lymphatic systems. Other conditions treatable using the methods provided herein include hypersplenism, splenectomy, bowel resection, and bone marrow infiltration. The methods described herein may also be used to treat anemia resulting from the deliberate or accidental introduction of a poison, toxin, or drug. For example, anemias resulting from cancer chemotherapies may be treated using the methods and solid forms provided herein. As such, the methods described herein may be employed when anemia or a hemoglobinopathy is the primary condition to be treated, or is a secondary condition caused by an underlying disease or treatment regimen.

In one embodiment, the diseases or disorders are various forms of leukemias such as chronic lymphocytic leukemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, and acute myeloblastic leukemia, including leukemias that are relapsed, refractory, or resistant, as disclosed in U.S. publication no. 2006/0030594, published Feb. 9, 2006, which is incorporated in its entirety by reference.

The term “leukemia” refers malignant neoplasms of the blood-forming tissues. The leukemia includes, but is not limited to, chronic lymphocytic leukemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, and acute myeloblastic leukemia. The leukemia can be relapsed, refractory or resistant to conventional therapy. The term “relapsed” refers to a situation where patients who have had a remission of leukemia after therapy have a return of leukemia cells in the marrow and a decrease in normal blood cells. The term “refractory or resistant” refers to a circumstance where patients, even after intensive treatment, have residual leukemia cells in their marrow.

In another embodiment, the diseases or disorders are various types of lymphomas, including Non-Hodgkin's lymphoma (NHL). The term “lymphoma” refers a heterogenous group of neoplasms arising in the reticuloendothelial and lymphatic systems. “NHL” refers to malignant monoclonal proliferation of lymphoid cells in sites of the immune system, including lymph nodes, bone marrow, spleen, liver, and gastrointestinal tract. Examples of NHL include, but are not limited to, mantle cell lymphoma (MCL), lymphocytic lymphoma of intermediate differentiation, intermediate lymphocytic lymphoma (ILL), diffuse poorly differentiated lymphocytic lymphoma (PDL), centrocytic lymphoma, diffuse small-cleaved cell lymphoma (DSCCL), follicular lymphoma, and any type of the mantle cell lymphomas that can be seen under the microscope (nodular, diffuse, blastic and mentle zone lymphoma).

Examples of diseases and disorders associated with, or characterized by, undesired angiogenesis include, but are not limited to, inflammatory diseases, autoimmune diseases, viral diseases, genetic diseases, allergic diseases, bacterial diseases, ocular neovascular diseases, choroidal neovascular diseases, retina neovascular diseases, and rubeosis (neovascularization of the angle). Specific examples of the diseases and disorders associated with, or characterized by, undesired angiogenesis include, but are not limited to, arthritis, endometriosis, Crohn's disease, heart failure, advanced heart failure, renal impairment, endotoxemia, toxic shock syndrome, osteoarthritis, retrovirus replication, wasting, meningitis, silica-induced fibrosis, asbestos-induced fibrosis, veterinary disorder, malignancy-associated hypercalcemia, stroke, circulatory shock, periodontitis, gingivitis, macrocytic anemia, refractory anemia, and 5q-deletion syndrome.

Other disease or disorders treated, prevented, or managed include, but not limited to, viral, genetic, allergic, and autoimmune diseases. Specific examples include, but are not limited to, HIV, hepatitis, adult respiratory distress syndrome, bone resorption diseases, chronic pulmonary inflammatory diseases, dermatitis, cystic fibrosis, septic shock, sepsis, endotoxic shock, hemodynamic shock, sepsis syndrome, post ischemic reperfusion injury, meningitis, psoriasis, fibrotic disease, cachexia, graft versus host disease, graft rejection, auto-immune disease, rheumatoid spondylitis, Crohn's disease, ulcerative colitis, inflammatory-bowel disease, multiple sclerosis, systemic lupus erythrematosus, ENL in leprosy, radiation damage, cancer, asthma, or hyperoxic alveolar injury.

In certain embodiments, a solid form provided herein, or a composition comprising a solid form provided herein, is administered orally, parenterally, topically, or mucosally. Examples of such dosage forms can be found in section 5.8, infra.

In certain embodiments, a solid form provided herein, or a composition comprising a solid form provided herein, is administered at a dosing frequency of once, twice, thrice, or four times daily. In certain embodiments, solid form provided herein, or a composition comprising a solid form provided herein, comprises lenalidomide in an amount of from about 0.1 to about 100 mg, from about 0.5 to about 50 mg, from, about 0.5 to about 25 mg, from about 1 mg to about 10 mg, from about 0.5 to about 5 mg, or from about 1 mg to about 5 mg. In certain embodiments, provided herein is a single unit dosage form suitable for oral administration to a human comprising: an amount equal to or greater than about 1, 2, 3, 4, or 5 mg of a solid form comprising lenalidomide and a coformer provided herein; and a pharmaceutically acceptable excipient. In one embodiment, the amount of the active ingredient is about 0.5 mg. In another embodiment, the amount of the active ingredient is about 1 mg. In another embodiment, the amount of the active ingredient is about 2 mg. In another embodiment, the amount of the active ingredient is about 4 mg.

In one embodiment, the second active agent is administered intravenously or subcutaneously and once or twice daily, once every other day, once every week, once every two weeks, or once every three weeks, in an amount of from about 1 to about 1000 mg, from about 5 to about 500 mg, from about 10 to about 350 mg, or from about 50 to about 200 mg. In one embodiment, the second active agent is administered orally and once or twice daily, once every other day, once every week, once every two weeks, or once every three weeks, in an amount of from about 1 to about 1000 mg, from about 5 to about 500 mg, from about 10 to about 350 mg, from about 10 to about 200 mg, from about 10 to about 100 mg, or from about 20 to about 50 mg. In specific embodiments, the second active agent is administered once every week in an amount of about 40 mg. The specific amount of the second active agent will depend on the specific agent used, the type of disease being treated or managed, the severity and stage of disease, and the amount(s) of compounds provided herein and any optional additional active agents concurrently administered to the patient.

As discussed elsewhere herein, also encompassed is a method of reducing, treating and/or preventing adverse or undesired effects associated with conventional therapy including, but not limited to, surgery, chemotherapy, radiation therapy, hormonal therapy, biological therapy and immunotherapy. Compounds provided herein and other active ingredients can be administered to a patient prior to, during, or after the occurrence of the adverse effect associated with conventional therapy.

5.5 Combination Therapy with a Second Active Agent

Specific methods of the invention comprise administering a solid form of the invention in combination with one or more second active agents, and/or in combination with radiation therapy, blood transfusions, or surgery. Examples of second active agents are also disclosed herein (see, e.g., section 5.3).

Administration of a solid form of this invention and the second active agents to a patient can occur simultaneously or sequentially by the same or different routes of administration. The suitability of a particular route of administration employed for a particular active agent will depend on the active agent itself (e.g., whether it can be administered orally without decomposing prior to entering the blood stream) and the disease being treated. A preferred route of administration for a solid form of the invention is orally. Preferred routes of administration for the second active agents or ingredients of the invention are known to those of ordinary skill in the art. See, e.g., Physicians' Desk Reference, 1755-1760 (56.sup.th ed., 2002).

In one embodiment of the invention, the second active agent is administered intravenously or subcutaneously and once or twice daily in an amount of from about 1 to about 1,000 mg, from about 5 to about 500 mg, from about 10 to about 375 mg, or from about 50 to about 200 mg. The specific amount of the second active agent will depend on the specific agent used, the type of disease being treated or managed, the severity and stage of disease, and the amount(s) of immunomodulatory compounds of the invention and any optional additional active agents concurrently administered to the patient. In a particular embodiment, the second active agent is rituximab, oblimersen (Genasense®), GM-CSF, G-CSF, EPO, taxotere, irinotecan, dacarbazine, transretinoic acid, topotecan, pentoxifylline, ciprofloxacin, dexamethasone, vincristine, doxorubicin, COX-2 inhibitor, IL2, IL8, IL18, IFN, Ara-C, vinorelbine, or a combination thereof.

In a specific embodiment, a solid form of the invention is administered in combination with rituximab to patients with leukemias. In a specific embodiment, the solid form is administered in an amount of from about 5 to about 25 mg per day to patients with chronic lymphocytic leukemia in combination with rituximab in an amount of 375 mg/m.sup.2.

In another embodiment, a solid form of the invention is administered in combination with fludarabine, carboplatin, and/or topotecan to patients with refractory or relapsed or high-risk acute myelogenous leukemia.

In another embodiment, a solid form of the invention is administered in combination with liposomal daunorubicin, topotecan and/or cytarabine to patients with unfavorable karotype acute myeloblastic leukemia.

In another embodiment, a solid form of the invention is administered alone or in combination with a second active ingredient such as vinblastine or fludarabine to patients with various types of lymphoma, including, but not limited to, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cutaneous T-Cell lymphoma, cutaneous B-Cell lymphoma, diffuse large B-Cell lymphoma or relapsed or refractory low grade follicular lymphoma.

In another embodiment, GM-CSF, G-CSF or EPO is administered subcutaneously during about five days in a four or six week cycle in an amount of from about 1 to about 750 mg/m.sup.2/day, preferably in an amount of from about 25 to about 500 mg/m.sup.2/day, more preferably in an amount of from about 50 to about 250 mg/m.sup.2/day, and most preferably in an amount of from about 50 to about 200 mg/m.sup.2/day. In a certain embodiment, GM-CSF may be administered in an amount of from about 60 to about 500 mcg/m.sup.2 intravenously over 2 hours, or from about 5 to about 12 mcg/m.sup.2/day subcutaneously. In a specific embodiment, G-CSF may be administered subcutaneously in an amount of about 1 mcg/kg/day initially and can be adjusted depending on rise of total granulocyte counts. The maintenance dose of G-CSF may be administered in an amount of about 300 (in smaller patients) or 480 mcg subcutaneously. In a certain embodiment, EPO may be administered subcutaneously in an amount of 10,000 Unit 3 times per week.

This invention also encompasses a method of increasing the dosage of an anti-cancer drug or agent that can be safely and effectively administered to a patient, which comprises administering to a patient (e.g., a human) a solid form of the invention, or a pharmaceutically acceptable derivative, salt, solvate, clathrate, hydrate, or prodrug thereof. Patients that can benefit by this method are those likely to suffer from an adverse effect associated with anti-cancer drugs for treating a specific cancer of the blood, skin, subcutaneous tissue, lymph nodes, brain, lung, liver, bone, intestine, colon, heart, pancreas, adrenal, kidney, prostate, breast, colorectal, or combinations thereof. The administration of a solid form of the invention alleviates or reduces adverse effects which are of such severity that it would otherwise limit the amount of anti-cancer drug.

In one embodiment, a solid form of the invention can be administered orally and daily in an amount of from about 0.10 to about 150 mg, and preferably from about 1 to about 50 mg, more preferably from about 5 to about 25 mg prior to, during, or after the occurrence of the adverse effect associated with the administration of an anti-cancer drug to a patient. In a particular embodiment, a solid form of the invention in combination with specific agents such as heparin, aspirin, coumadin, or G-CSF to avoid adverse effects that are associated with anti-cancer drugs such as but not limited to neutropenia or thrombocytopenia.

In another embodiment, this invention encompasses a method of treating, preventing and/or managing cancer, which comprises administering a solid form of the invention in conjunction with (e.g., before, during, or after) conventional therapy including, but not limited to, surgery, immunotherapy, biological therapy, radiation therapy, or other non-drug based therapy presently used to treat, prevent or manage cancer. The combined use of the solid forms of the invention and conventional therapy may provide a unique treatment regimen that is unexpectedly effective in certain patients. Without being limited by theory, it is believed that solid forms of the invention may provide additive or synergistic effects when given concurrently with conventional therapy.

As discussed elsewhere herein, the invention encompasses a method of reducing, treating and/or preventing adverse or undesired effects associated with conventional therapy including, but not limited to, surgery, chemotherapy, radiation therapy, hormonal therapy, biological therapy and immunotherapy. A solid form of the invention and other active ingredient can be administered to a patient prior to, during, or after the occurrence of the adverse effect associated with conventional therapy.

In one embodiment, a solid form of the invention can be administered in an amount of from about 0.10 to about 150 mg, and preferably from about 1 to about 50 mg, more preferably from about 5 to about 25 mg orally and daily alone, or in combination with a second active agent disclosed herein (see, e.g., section 5.2), prior to, during, or after the use of conventional therapy

5.6 Use with Transplantation Therapy

Compounds of the invention can be used to reduce the risk of Graft Versus Host Disease (GVHD). Therefore, the invention encompasses a method of treating, preventing and/or managing cancer, which comprises administering the solid form of the invention in conjunction with transplantation therapy.

As those of ordinary skill in the art are aware, the treatment of cancer is often based on the stages and mechanism of the disease. For example, as inevitable leukemic transformation develops in certain stages of cancer, transplantation of peripheral blood stem cells, hematopoietic stem cell preparation or bone marrow may be necessary. The combined use of the solid form of the invention and transplantation therapy provides a unique and unexpected synergism. In particular, an solid form of the invention exhibits immunomodulatory activity that may provide additive or synergistic effects when given concurrently with transplantation therapy in patients with cancer.

An solid of the invention can work in combination with transplantation therapy reducing complications associated with the invasive procedure of transplantation and risk of GVHD. This invention encompasses a method of treating, preventing and/or managing cancer which comprises administering to a patient (e.g., a human) a solid form the invention before, during, or after the transplantation of umbilical cord blood, placental blood, peripheral blood stem cell, hematopoietic stem cell preparation or bone marrow. Examples of stem cells suitable for use in the methods of the invention are disclosed in U.S. patent publication nos. 2002/0123141, 2003/0235909 and 2003/0032179, by R. Hariri et al., the entireties of which are incorporated herein by reference.

In one embodiment of this method, a solid form of the invention is administered to patients with leukemias before, during, or after the transplantation of autologous peripheral blood progenitor cell.

In another embodiment, a solid form of the invention is administered to patients with relapsed leukemia after the stem cell transplantation.

5.7 Cycling Therapy

In certain embodiments, the prophylactic or therapeutic agents provided herein are cyclically administered to a patient. Cycling therapy involves the administration of an active agent for a period of time, followed by a rest (i.e., discontinuation of the administration) for a period of time, and repeating this sequential administration. Cycling therapy can reduce the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improve the efficacy of the treatment.

Consequently, in one embodiment, a compound provided herein is administered daily in a single or divided doses in a four to six week cycle with a rest period of about a week or two weeks. Cycling therapy further allows the frequency, number, and length of dosing cycles to be increased. Thus, another embodiment encompasses the administration of a compound provided herein for more cycles than are typical when it is administered alone. In yet another embodiment, a compound provided herein is administered for a greater number of cycles than would typically cause dose-limiting toxicity in a patient to whom a second active ingredient is not also being administered.

In one embodiment, a compound provided herein is administered daily and continuously for three or four weeks at a dose of from about 0.1 mg to about 5 mg per day, followed by a rest of one or two weeks. In other embodiments, the dose can be from about 1 mg to about 5 mg per day (e.g., 1, 2, 3, or 4 mg/day), given on Days 1-21 of each 28-day cycle until disease progression, followed by a rest of 7 days on Days 22-28 of each 28-day cycle, for example, in patients with relapsed and refractory multiple myeloma who are refractory to their last myeloma therapy and have received at least 2 prior therapies that included lenalidomide and bortezomib.

In a preferred embodiment, the solid form is administered to patients with leukemia in an amount of from about 0.10 to about 150 mg per day for 21 days followed by seven days rest in a 28 day cycle. In the most preferred embodiment, the solid form is administered to patients with refractory or relapsed chronic lymphocytic leukemia in an amount of about 25 mg per day for 21 days followed by seven days rest in a 28 day cycle.

In one embodiment, a compound provided herein and a second active ingredient are administered orally, with administration of the compound provided herein occurring 30 to 60 minutes prior to the second active ingredient, during a cycle of four to six weeks. In another embodiment, the combination of a compound provided herein and a second active ingredient is administered by intravenous infusion over about 90 minutes every cycle.

In a specific embodiment, one cycle comprises the administration of from about 5 to about 25 mg/day of the solid form and from about 50 to about 750 mg/m2/day of a second active ingredient daily for three to four weeks and then one or two weeks of rest. In a preferred embodiment, rituximab can be administered in an amount of 375 mg/m2 as an additional active agent to patients with refractory or relapsed chronic lymphocytic leukemia. Typically, the number of cycles during which the combinatorial treatment is administered to a patient will be from about one to about 24 cycles, more typically from about two to about 16 cycles, and even more typically from about four to about three cycles.

5.8 Pharmaceutical Compositions and Dosage Forms

Pharmaceutical compositions can be used in the preparation of single unit dosage forms comprising one or more solid forms provided herein. In one embodiment, provided herein are pharmaceutical compositions and dosage forms comprising one or more solid forms comprising a compound provided herein, or a pharmaceutically acceptable salt, solvate (e.g., hydrate), stereoisomer, co-crystal, clathrate, or prodrug thereof. Pharmaceutical compositions and dosage forms provided herein can further comprise one or more pharmaceutically acceptable excipients or carriers.

In some embodiments, pharmaceutical compositions and dosage forms provided herein can also comprise one or more additional active ingredients. Examples of optional second, or additional, active ingredients are disclosed herein elsewhere.

In one embodiment, single unit dosage forms provided herein are suitable for oral, parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial), topical (e.g., eye drops or other ophthalmic preparations), mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), or transdermal administration to a patient. Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules or hard gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; powders; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions, or water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; eye drops or other ophthalmic preparations suitable for topical administration; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient. In one embodiment, the single dosage forms provided herein are tablets, caplets, or capsules comprising one or more solid forms provided herein. In one embodiment, the single dosage forms provided herein are tablets or capsules comprising one or more solid forms provided herein.

The composition, shape, and type of dosage forms will typically vary depending on their use. For example, a dosage form used in the acute treatment of a disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease. Similarly, a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease. These and other ways in which specific dosage forms are used will vary from one another will be readily apparent to those skilled in the art. See, e.g., Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990).

In one embodiment, pharmaceutical compositions and dosage forms comprise one or more excipients or carriers. Suitable excipients are known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors known in the art including, but not limited to, the way in which the dosage form will be administered to a patient. For example, oral dosage forms such as tablets may contain excipients not suited for use in parenteral dosage forms. The suitability of a particular excipient may also depend on the specific active ingredients in the dosage form. For example, the decomposition of some active ingredients may be accelerated by some excipients such as lactose, or when exposed to water. Active ingredients that comprise primary or secondary amines are particularly susceptible to such accelerated decomposition. Consequently, in one embodiment, provided are pharmaceutical compositions and dosage forms that contain little, if any, lactose or other mono- or di-saccharides. As used herein, the term “lactose-free” means that the amount of lactose present, if any, is insufficient to substantially increase the degradation rate of an active ingredient. Lactose-free compositions provided herein can comprise excipients which are known in the art and are listed in the U.S. Pharmacopeia (USP) 25-NF20 (2002), which is incorporated herein in its entirety.

Also provided are anhydrous pharmaceutical compositions and dosage forms comprising active ingredient(s), since water may facilitate the degradation of some compounds. Anhydrous pharmaceutical compositions and dosage forms can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, in one embodiment, anhydrous compositions are packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.

Also provided are pharmaceutical compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.

Like the amounts and types of excipients, the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients. In one embodiment, dosage forms comprise the active ingredient or solid form comprising lenalidomide and a coformer provided herein in an amount of from about 0.10 to about 50 mg, or from about 0.50 to about 30 mg. In other embodiments, dosage forms comprise a compound provided herein in an amount of about 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 12.5, 15, 20, or 25 mg. In one embodiment, dosage forms comprise a compound provided herein in an amount of about 1, 2.5, 5, 10, 15, 20, or 25 mg.

In other embodiments, dosage forms comprise a second active ingredient in an amount from about 1 mg to about 1000 mg, from about 5 mg to about 500 mg, from about 10 mg to about 350 mg, from about 5 mg to about 250 mg, from about 5 mg to about 100 mg, from about 10 mg to about 100 mg, from about 10 mg to about 50 mg, or from about 50 mg to about 200 mg. In one embodiment, the specific amount of the second active agent will depend on the specific agent used, the diseases or disorders being treated or managed, and the amount(s) of a compound provided herein, and any optional additional active agents concurrently administered to the patient.

In particular embodiments, provided herein is a pharmaceutical composition comprising a solid form comprising lenalidomide and a coformer provided herein and a pharmaceutically acceptable excipient or carrier. In particular embodiments, provided herein is a pharmaceutical composition comprising a cocrystal comprising lenalidomide and a coformer provided herein and a pharmaceutically acceptable excipient or carrier. In particular embodiments, provided herein is a pharmaceutical composition comprising an amorphous lenalidomide provided herein and a pharmaceutically acceptable excipient or carrier. Exemplary embodiments of formulations of lenalidomide are described in, for example, U.S. Pat. Nos. 5,635,517, 6,335,349, 6,316,471, 6,476,052, 7,041,680, and 7,709,502; and U.S. Patent Application Publication No. 2011/0045064; the entireties of which are incorporated herein by reference.

5.8.1 Oral Dosage Forms

Pharmaceutical compositions that are suitable for oral administration can be provided as discrete dosage forms, such as, but not limited to, tablets, fastmelts, chewable tablets, capsules, pills, strips, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, bulk powders, effervescent or non-effervescent powders or granules, oral mists, solutions, emulsions, suspensions, wafers, sprinkles, elixirs, and syrups. In one embodiment, such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990). As used herein, oral administration also includes buccal, lingual, and sublingual administration.

In one embodiment, the oral dosage form provided herein is a tablet. In one embodiment, the oral dosage form provided herein is a capsule. In one embodiment, the oral dosage form provided herein is a caplet. In particular embodiments,

In one embodiment, oral dosage forms provided herein are prepared by combining the active ingredients in an intimate admixture with one or more pharmaceutically acceptable carrier or excipient, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, flavoring agents, emulsifying agents, suspending and dispersing agents, preservatives, solvents, non-aqueous liquids, organic acids, and sources of carbon dioxide, according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.

In one embodiment, oral dosage forms are tablets or capsules, in which case solid excipients are employed. In specific embodiments, capsules comprising one or more solid forms comprising lenalidomide and a coformer provided herein can be used for oral administration. In one embodiment, the total amount of lenalidomide in the capsule is about 0.5, about 1, about 1.5, about 2, about 2.5, about 3, about 3.5, about 4, about 4.5, about 5, about 6, about 7, about 8, about 9, about 10, about 12.5, about 15, about 20, or about 25 mg. In one embodiment, the total amount of lenalidomide in the capsule is about 1, about 2.5, about 5, about 10, about 15, about 20, or about 25 mg. Each capsule can contain lenalidomide as the active ingredient and one or more of the following inactive ingredients: lactose anhydrous, microcrystalline cellulose, croscarmellose sodium, and magnesium stearate. In specific embodiments, the 5 mg and 25 mg capsule shell can contain gelatin, titanium dioxide and black ink. In specific embodiments, the 2.5 mg and 10 mg capsule shell can contain gelatin, FD&C blue #2, yellow iron oxide, titanium dioxide and black ink. In specific embodiments, the 15 mg capsule shell can contain gelatin, FD&C blue #2, titanium dioxide and black ink. In another embodiment, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.

In certain embodiments, the dosage form is a tablet, wherein the tablet is manufactured using standard, art-recognized tablet processing procedures and equipment. In certain embodiments, the method for forming the tablets is direct compression of a powdered, crystalline and/or granular composition comprising a solid form provided herein, alone or in combination with one or more excipients, such as, for example, carriers, additives, polymers, or the like. In certain embodiments, as an alternative to direct compression, the tablets may be prepared using wet granulation or dry granulation processes. In certain embodiments, the tablets are molded rather than compressed, starting with a moist or otherwise tractable material. In certain embodiments, compression and granulation techniques are used.

In certain embodiments, the dosage form is a capsule, wherein the capsules may be manufactured using standard, art-recognized capsule processing procedures and equipments. In certain embodiments, soft gelatin capsules may be prepared in which the capsules contain a mixture comprising a solid form provided herein and vegetable oil or non-aqueous, water miscible materials, such as, for example, polyethylene glycol and the like. In certain embodiments, hard gelatin capsules may be prepared containing granules of solid forms provided herein in combination with a solid pulverulent carrier, such as, for example, lactose, saccharose, sorbitol, mannitol, potato starch, corn starch, amylopectin, cellulose derivatives, or gelatin. In certain embodiments, a hard gelatin capsule shell may be prepared from a capsule composition comprising gelatin and a small amount of plasticizer such as glycerol. In certain embodiments, as an alternative to gelatin, the capsule shell may be made of a carbohydrate material. In certain embodiments, the capsule composition may additionally include polymers, colorings, flavorings and opacifiers as required. In certain embodiments, the capsule comprises HPMC.

Examples of excipients or carriers that can be used in oral dosage forms provided herein include, but are not limited to, diluents (bulking agents), lubricants, disintegrants, fillers, stabilizers, surfactants, preservatives, coloring agents, flavoring agents, binding agents (binders), excipient supports, glidants, permeation enhancement excipients, plasticizers and the like, e.g., as known in the art. It will be understood by those in the art that some substances serve more than one purpose in a pharmaceutical composition. For instance, some substances are binders that help hold a tablet together after compression, yet are also disintegrants that help break the tablet apart once it reaches the target delivery site. Selection of excipients and amounts to use may be readily determined by the formulation scientist based upon experience and consideration of standard procedures and reference works available in the art.

In certain embodiments, dosage forms provided herein comprise one or more binders. Binders may be used, e.g., to impart cohesive qualities to a tablet or a capsule, and thus ensure that the formulation remains intact after compression. Suitable binders include, but are not limited to, starch (including potato starch, corn starch, and pregelatinized starch), gelatin, sugars (including sucrose, glucose, dextrose and lactose), polyethylene glycol, propylene glycol, waxes, and natural and synthetic gums, e.g., acacia sodium alginate, polyvinylpyrrolidone (PVP), cellulosic polymers (including hydroxypropyl cellulose (HPC), hydroxypropylmethylcellulose (HPMC), methyl cellulose, ethyl cellulose, hydroxyethyl cellulose (HEC), carboxymethyl cellulose and the like), veegum, carbomer (e.g., carbopol), sodium, dextrin, guar gum, hydrogenated vegetable oil, magnesium aluminum silicate, maltodextrin, polymethacrylates, povidone (e.g., KOLLIDON, PLASDONE), microcrystalline cellulose, among others. Binding agents also include, e.g., acacia, agar, alginic acid, cabomers, carrageenan, cellulose acetate phthalate, ceratonia, chitosan, confectioner's sugar, copovidone, dextrates, dextrin, dextrose, ethylcellulose, gelatin, glyceryl behenate, guar gum, hydroxyethyl cellulose, hydroxyethylmethyl cellulose, hydroxypropyl cellulose, hydroxypropyl starch, hypromellose, inulin, lactose, magnesium aluminum silicate, maltodextrin, maltose, methylcellulose, poloxamer, polycarbophil, polydextrose, polyethylene oxide, polymethylacrylates, povidone, sodium alginate, sodium carboxymethylcellulose, starch, pregelatinized starch, stearic acid, sucrose, and zein. In one embodiment, the binding agent can be, relative to the weight of the dosage form, in an amount of from about 50% to about 99% w/w. In certain embodiments, a suitable amount of a particular binder is determined by one of ordinary skill in the art.

Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101, AVICEL-PH-103 AVICEL RC-581, AVICEL-PH-105 (FMC Corporation, Marcus Hook, Pa.), and mixtures thereof. In one embodiment, a specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581. Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103™ and Starch 1500 LM.

Examples of fillers suitable for use in the pharmaceutical compositions and dosage forms provided herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The binder or filler in a pharmaceutical composition is, in one embodiment, present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.

In certain embodiments, dosage forms provided herein comprise one or more diluents. Diluents may be used, e.g., to increase bulk so that a practical size tablet or capsule is ultimately provided. Suitable diluents include dicalcium phosphate, calcium sulfate, lactose, cellulose, kaolin, mannitol, sodium chloride, dry starch, microcrystalline cellulose (e.g., AVICEL), microfine cellulose, pregelitinized starch, calcium carbonate, calcium sulfate, sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, tribasic calcium phosphate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, polymethacrylates (e.g., EUDRAGIT), potassium chloride, sodium chloride, sorbitol and talc, among others. Diluents also include, e.g., ammonium alginate, calcium carbonate, calcium phosphate, calcium sulfate, cellulose acetate, compressible sugar, confectioner's sugar, dextrates, dextrin, dextrose, erythritol, ethylcellulose, fructose, fumaric acid, glyceryl palmitostearate, isomalt, kaolin, lacitol, lactose, mannitol, magnesium carbonate, magnesium oxide, maltodextrin, maltose, medium-chain triglycerides, microcrystalline cellulose, microcrystalline silicified cellulose, powered cellulose, polydextrose, polymethylacrylates, simethicone, sodium alginate, sodium chloride, sorbitol, starch, pregelatinized starch, sucrose, sulfobutylether-β-cyclodextrin, talc, tragacanth, trehalose, and xylitol. Diluents may be used in amounts calculated to obtain a desired volume for a tablet or capsule. The amount of a diluent in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.

Disintegrants may be used in the compositions to provide tablets or capsules that disintegrate when exposed to an aqueous environment. Dosage forms that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredient(s) may be used to form solid oral dosage forms. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. In one embodiment, pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, or from about 1 to about 5 weight percent of disintegrant.

Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.

Lubricants that can be used in pharmaceutical compositions and dosage forms include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL200, manufactured by W.R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Plano, Tex.), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.

Suitable glidants include, but are not limited to, colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston, Mass.), and asbestos-free talc. Suitable coloring agents include, but are not limited to, any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Suitable flavoring agents include, but are not limited to, natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Suitable sweetening agents include, but are not limited to, sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include, but are not limited to, gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate. Suitable suspending and dispersing agents include, but are not limited to, sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable preservatives include, but are not limited to, glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Suitable wetting agents include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Suitable solvents include, but are not limited to, glycerin, sorbitol, ethyl alcohol, and syrup. Suitable non-aqueous liquids utilized in emulsions include, but are not limited to, mineral oil and cottonseed oil. Suitable organic acids include, but are not limited to, citric and tartaric acid. Suitable sources of carbon dioxide include, but are not limited to, sodium bicarbonate and sodium carbonate.

In one embodiment, a solid oral dosage form comprises a compound provided herein, and one or more excipients selector from anhydrous lactose, microcrystalline cellulose, polyvinylpyrrolidone, stearic acid, colloidal anhydrous silica, and gelatin. In one embodiment, capsules comprise one or more solid forms comprising lenalidomide and a coformer provided herein, and one or more of the following inactive ingredients: mannitol, pregelatinized starch, sodium stearyl fumarate, gelatin, titanium dioxide, FD&C blue 2, yellow iron oxide, white ink, black ink, FD&C red 3, and a combination thereof.

The pharmaceutical compositions provided herein for oral administration can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.

The tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.

The pharmaceutical compositions provided herein for oral administration can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.

The pharmaceutical compositions provided herein for oral administration can be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil. Emulsions may include a pharmaceutically acceptable non-aqueous liquid or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.

Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly-alkylene glycol, including, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations can further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.

The pharmaceutical compositions provided herein for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems. Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.

The pharmaceutical compositions provided herein for oral administration can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form. Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.

Coloring and flavoring agents can be used in all of the above dosage forms.

The pharmaceutical compositions provided herein for oral administration can be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.

6. EXAMPLES

Certain embodiments provided herein are illustrated by the following non-limiting examples.

6.1 Preparation of Cocrystal Comprising Lenalidomide and a Coformer Example 1. Solubilities of Lenalidomide

Solubility of lenalidomide in various solvents at ambient temperature was determined and is shown in Table 1. Solubility was estimated by treating a weighed sample of lenalidomide with measured aliquots of the test solvent at ambient temperature, with shaking and/or sonication between aliquots. Dissolution was determined by visual inspection. Solubility numbers were calculated by dividing the total amount of solvent used to dissolve the sample by the weight of the sample. The actual solubilities may be greater than the numbers calculated because of the use of solvent aliquots that were too large or because of slow dissolution rates. The solubility number is expressed as “less than” if dissolution did not occur during the experiment. The solubility number is expressed as “greater than” if dissolution occurred on addition of the first solvent aliquot.

TABLE 1 Estimated Solubility of Lenalidomide Solvent Solubility (mg/mL) acetone 2.2 acetonitrile 3.4 methanol 2.6 tetrahydrofuran 3.0

Example 2. Conformers Used in the Cocrystal Screen of Lenalidomide

A set of 50 coformers was selected from an internal database based on toxicity profiles (pharmaceutical acceptability) and anticipated non-covalent and ionic interactions with lenalidomide.

TABLE 2 Coformers Used in the Cocrystal Screen of Lenalidomide acetylsalicylic acid D-glucose nicotinic acid aconitic acid L-glutamic acid oxalic acid adipic acid glutaric acid L-proline 4-aminosalicylic acid glycine propyl gallate L-ascorbic acid glycolic acid L-pyroglutamic acid benzoic acid hippuric acid saccharin (+)-camphoric acid 1-hydroxy-2-naphthoic salicylic acid acid capric acid ketoglutaric acid sebacic acid cinnamic acid L-lysine sodium lauryl sulfate citric acid magnesium bromide sorbic acid cyclamic acid maleic acid succinic acid ethyl maltol L-malic acid L-tartaric acid ethyl paraben malonic acid urea D-fructose maltol vanillic acid fumaric acid D,L-mandelic acid vanillin gallic acid methyl paraben zinc chloride gentisic acid nicotinamide

Example 3. Preparation of Cocrystal Comprising Lenalidomide and a Coformer Using Stoichiometric Slurry Experiments

Stoichiometric slurry experiments were carried out in glass vials. Each of the vials was charged with about 20 mg of lenalidomide, an approximately equimolar amount of coformer, and 500 μL of a saturated solution of the same coformer in the solvent used for that experiment. A magnetic stir bar was placed in each vial and the rack of vials was placed on a stir plate at room temperature for 24 hours. The solids were isolated by centrifugation.

Example 4. Preparation of Cocrystal Comprising Lenalidomide and a Coformer Using Stoichiometric Wet Milling Experiments

For each experiment, A PEEK grinding cup was charged with about 20 mg of lenalidomide, an approximately equimolar amount of coformer, about 10 μL of a mixture of methanol and water (3:1), and one steel grinding ball. The cup was sealed and shaken on a Retsch mill for 20 min. The solid was collected.

Example 5. Preparation of Cocrystal Comprising Lenalidomide and a Coformer Using Stoichiometric Flash Evaporation Experiments

For each experiment, he solvent was removed from solutions containing about 20 mg of lenalidomide and approximately equimolar amount of coformer on a rotary evaporator using a bath set at 65° C. If the residue was solid it was stored at 75% relative humidity for 1 day. If the residue was oil it was stored at 60° C. overnight. The resulting solids were collected.

Example 6. Potential New Solid Phases From Screening Experiments

One hundred and thirty-nine (139) experiments using fifty (50) coformers were carried out. Sixteen (16) samples made using twelve (12) different coformers were found that exhibit XRPD patterns suggestive of cocrystal formation: benzoic acid, gallic acid, glycolic acid, hippuric acid, magnesium bromide (2 samples), malonic acid, oxalic acid, propyl gallate (2 samples), sodium lauryl sulfate, L-tartaric acid (3 samples), vanillic acid, and zinc chloride. Those patterns contain peaks that do not appear to arise from any polymorph of lenalidomide or the relevant coformer.

Example 7. Solid Forms Comprising Lenalidomide and Gallic Acid

A cocrystal comprising lenalidomide and gallic acid was prepared using the stoichiometric slurry method described in Example 3. A 1:1 mixture of methanol:water was used as the solvent. The XRPD pattern of the solid form is provided in FIG. 1. The overlay plot of the XRPD pattern of the solid form with a pattern from gallic acid is provided in FIG. 27.

Example 8. Solid Forms Comprising Lenalidomide and Vanillic Acid

A cocrystal comprising lenalidomide and vanillic acid was prepared using the stoichiometric flash evaporation method described in Example 5. Methanol was used as the solvent. The XRPD pattern of the solid form is provided in FIG. 2.

Example 9. Solid Forms Comprising Lenalidomide and Oxalic Acid

A cocrystal comprising lenalidomide and oxalic acid was prepared using the stoichiometric wet milling method described in Example 4. The XRPD pattern of the solid form is provided in FIG. 3. The overlay plot of the XRPD pattern of the solid form with a pattern from lenalidomide form D is provided in FIG. 28.

Example 10. Solid Forms Comprising Lenalidomide and Propyl Gallate

A cocrystal comprising lenalidomide and propyl gallate was prepared using the stoichiometric slurry method described in Example 3. A 1:1 mixture of methanol:water was used as the solvent. A cocrystal comprising lenalidomide and propyl gallate was also prepared using the stoichiometric wet milling method described in Example 4. The XRPD pattern of the solid form is provided in FIG. 4. The overlay plot of the XRPD patterns of the solid form with a pattern from lenalidomide form D is provided in FIG. 29.

Example 11. Solid Forms Comprising Lenalidomide and Glycolic Acid

A cocrystal comprising lenalidomide and glycolic acid was prepared using the stoichiometric slurry method described in Example 3. Acetonitrile was used as the solvent. The XRPD pattern of the solid form is provided in FIG. 5. The overlay plot of the XRPD pattern of the solid form with a pattern from lenalidomide form A and glycolic acid is provided in FIG. 30.

Example 12. Solid Forms Comprising Lenalidomide and Sodium Lauryl Sulfate

A cocrystal comprising lenalidomide and sodium lauryl sulfate was prepared using the stoichiometric slurry method described in Example 3. Methanol was used as the solvent. The XRPD pattern of the solid form is provided in FIG. 6. The overlay plot of the XRPD pattern of the solid form with a pattern from lenalidomide form A is provided in FIG. 31.

Example 13. Solid Forms Comprising Lenalidomide and Magnesium Bromide

A cocrystal comprising lenalidomide and magnesium bromide was prepared using the stoichiometric slurry method described in Example 3. Acetone was used as the solvent. A cocrystal comprising lenalidomide and magnesium bromide was also prepared using the stoichiometric flash evaporation method described in Example 5. Methanol was used as the solvent.

In a scale up experiment, a solution of 66.1 mg (0.255 mmol) of lenalidomide and 75.0 mg (0.257 mmol) of magnesium bromide hexahydrate in 75 mL of methanol was concentrated on a rotary evaporator at 60° C. The flask containing the resulting solid was placed in an oven at 60° C. overnight. A few milliliters of diethyl ether were added to the flask, the solid was scraped from the walls with a spatula, and the mixture was transferred to a vial. The ether was evaporated in a stream of dry air to give 105.8 mg (94% yield) of the lenalidomide/magnesium bromide cocrystal.

The cocrystal was characterized by XRPD (FIG. 7A and FIG. 7B), DSC and TGA (FIG. 8), DVS (FIG. 9), Raman (FIG. 10), 1H-NMR (FIG. 11), and optical microscope (FIG. 12). The overlay plot of the XRPD patterns of the solid form with a pattern from magnesium bromide is provided in FIG. 32.

The TGA showed about 16% weight loss from 70 to 200° C. However, there were no clear events observed by DSC. A ragged endothermic event was present at about 123° C. A noisy appearance of the DSC thermogram around 180-250° C. suggested decomposition may be occurring.

The 1H-NMR spectrum was consistent with the structure of lenalidomide (magnesium bromide is inactive in 1H-NMR spectroscopy).

Dynamic vapor sorption (DVS) analysis showed that the cocrystal is hygroscopic, increasing in weight by 43% from 5-95% RH. Most of that gain was lost in the desorption cycle and the crystalline form of the resulting solid was the same as that of the starting material, suggesting that the cocrystal was still present after the DVS experiment.

Example 14. Solid Forms Comprising Lenalidomide and Malonic Acid

A cocrystal comprising lenalidomide and malonic acid was prepared using the stoichiometric slurry method described in Example 3. Acetonitrile was used as the solvent.

In a scale up experiment, to 1.5 mL of acetonitrile was added malonic acid until solid persisted. The slurry was stirred at ambient temperature overnight and centrifuged. The liquid was decanted and treated with 82.6 mg (0.319 mmol) of lenalidomide and 33.2 mg (0.286 mmol) of malonic acid. The slurry was stirred at ambient temperature for 4 days and centrifuged. The liquid was decanted and the remaining solid was dried in an oven at 60° C. oven to give a lenalidomide/malonic acid cocrystal.

The cocrystal was characterized by XRPD (FIG. 13A and FIG. 13B), DSC and TGA (FIG. 14), DVS (FIG. 15), Raman (FIG. 16), 1H-NMR (FIGS. 17A & 17B), and optical microscope (FIG. 18).

DSC analysis revealed relatively sharp endothermic events at about 102 and 111° C., followed by a broad endothermic event at about 159° C. TGA analysis showed a weight loss of 43% between 100 and 180° C. That large weight loss suggested a decomposition reaction(s) was occurring, but may included water loss as well.

The 1H-NMR spectrum in DMSO-d6 showed peaks from both lenalidomide and malonic acid. 1H-NMR spectra were acquired of lenalidomide and the cocrystal in CH3OH-d4. Peaks from the CH2 group of malonic acid were integrated and the integral obtained was one proton, which indicated that the cocrystal stoichiometry is 2:1 lenalidomide:malonic acid.

Dynamic vapor sorption (DVS) analysis showed that the cocrystal is hygroscopic, increasing in weight by 42% from 5-95% RH. Most of that gain was lost in the desorption cycle and the crystalline form of the resulting solid was the same as that of the starting material, suggesting that the cocrystal was still present after the DVS experiment.

Example 15. Solid Forms Comprising Lenalidomide and L-Tartaric Acid

A cocrystal comprising lenalidomide and L-tartaric acid was prepared using the stoichiometric wet milling method described in Example 4.

In a scale up experiment, a mixture of 59.8 mg (0.332 mmol) of lenalidomide, 68.8 mg (0.335 mmol) of L-tartaric acid, 40 μL of a 1:3 (v:v) mixture of water and methanol, and one stainless steel grinding ball was placed in a polyether ether ketone (PEEK) grinding cup. The cup was closed and agitated on a Retsch mill at 100% power for about 20 minutes. The grinding ball was removed and the contents of the cup were recovered to give the lenalidomide/L-tartaric acid cocrystal.

The cocrystal was characterized by XRPD (FIG. 19A and FIG. 19B), DSC and TGA (FIG. 20), DVS (FIG. 21), 1H-NMR (FIG. 22), and optical microscope (FIG. 23).

The TGA showed continuous weight loss from the start of the experiment, totaling about 12% by 200° C. An endothermic event at about 148° C. was observed by DSC. The noisy appearance of the DSC thermogram around 180-250° C. suggested decomposition may be occurring.

The 1H-NMR spectrum showed peaks from both lenalidomide and L-tartaric acid; integrations indicated that the cocrystal contains a 1:1 molar ratio of those components.

Dynamic vapor sorption (DVS) analysis showed that the cocrystal is hygroscopic, increasing in weight by 22% from 5-95% RH. Most of that gain was lost in the desorption cycle and the crystalline form of the resulting solid was the same as that of the starting material, suggesting that the cocrystal was still present after the DVS experiment.

Example 16. Solid Forms Comprising Lenalidomide and Hippuric Acid

A cocrystal comprising lenalidomide and hippuric acid was prepared using the stoichiometric flash evaporation method described in Example 5. Methanol was used as the solvent. The XRPD pattern of the solid form is provided in FIG. 24. The overlay plot of the XRPD pattern of the solid form with a pattern from lenalidomide form D is provided in FIG. 33.

Example 17. Solid Forms Comprising Lenalidomide and Zinc Chloride

A cocrystal comprising lenalidomide and zinc chloride was prepared using the stoichiometric wet milling method described in Example 4. The XRPD pattern of the solid form is provided in FIG. 25.

Example 18. Solid Forms Comprising Lenalidomide and Benzoic Acid

A cocrystal comprising lenalidomide and benzoic acid was prepared using the stoichiometric flash evaporation method described in Example 5. Methanol was used as the solvent. The XRPD pattern of the solid form is provided in FIG. 26. The overlay plot of the XRPD patterns of the solid form with a pattern from benzoic acid is provided in FIG. 34.

6.2 Characterization of Cocrystal Comprising Lenalidomide and a Coformer

X-Ray Powder Diffraction (XRPD)

The Rigaku Smart-Lab X-ray diffraction system was configured for reflection Bragg-Brentano geometry using a line source X-ray beam. The x-ray source is a Cu Long Fine Focus tube that was operated at 40 kV and 44 ma. That source provides an incident beam profile at the sample that changes from a narrow line at high angles to a broad rectangle at low angles. Beam conditioning slits are used on the line X-ray source to ensure that the maximum beam size is less than 10 mm both along the line and normal to the line. The Bragg-Brentano geometry is a para-focusing geometry controlled by passive divergence and receiving slits with the sample itself acting as the focusing component for the optics. The inherent resolution of Bragg-Brentano geometry is governed in part by the diffractometer radius and the width of the receiving slit used. Typically, the Rigaku Smart-Lab is operated to give peak widths of 0.1° 2θ or less. The axial divergence of the X-ray beam is controlled by 5.0-degree Soller slits in both the incident and diffracted beam paths.

Powder samples were prepared in a low background Si holder using light manual pressure to keep the sample surfaces flat and level with the reference surface of the sample holder. The single-crystal, Si, low-background holder has a small circular recess (7 mm diameter and about 1 mm depth) that holds between 5 and 10 mg of powdered material. Each sample was analyzed from 2 to 40°2θ using a continuous scan of 6°2θ per minute with an effective step size of 0.02°2θ.

Differential Scanning Calorimetry (DSC)

DSC analyses were carried out using a TA Instruments Q2000 instrument. The instrument temperature calibration was performed using indium. The DSC cell was kept under a nitrogen purge of about 50 mL per minute during each analysis. The sample was placed in a standard, crimped, aluminum pan and was heated from 20° C. to 350° C. at a rate of 10° C. per minute.

Thermogravimetric (TG) Analysis

The TG analysis was carried out using a TA Instruments Q50 instrument. The instrument balance was calibrated using class M weights and the temperature calibration was performed using alumel. The nitrogen purge was about 40 mL per minute at the balance and about 60 mL per minute at the furnace. Each sample was placed into a pre-tared platinum pan and heated from 20° C. to 350° C. at a rate of 10° C. per minute.

Dynamic Vapor Sorption

DVS analyses were carried out TA Instruments Q5000 Dynamic Vapor Sorption analyzer. The instrument was calibrated with standard weights and a sodium bromide standard for humidity. Samples were analyzed at 25° C. with a maximum equilibration time of 60 minutes in 10% relative humidity (RH) steps from 5 to 95% RH (adsorption cycle) and from 95 to 5% RH (desorption cycle).

Raman Spectroscopy

Fourier transform (FT) Raman spectra were acquired on a Nicolet model 6700 spectrometer interfaced to a Nexus Raman accessory module. This instrument is configured with a Nd:YAG laser operating at 1024 nm, a CaF2 beamsplitter, and a indium gallium arsenide detector. OMNIC 8.1 software was used for control of data acquisition and processing of the spectra. Samples were packed into a 3-inch glass NMR tube for analysis.

Nuclear Magnetic Resonance (NMR) Spectroscopy

The 1H NMR spectra were acquired on a Bruker DRX-500 spectrometer located at the Chemistry Department of Purdue University. Samples were prepared by dissolving material in DMSO-d6. The solutions were filtered and placed into individual 5-mm NMR tubes for subsequent spectral acquisition. The temperature controlled (298K) 1H NMR spectra acquired on the DRX-500 utilized a 5-mm cryoprobe operating at an observing frequency. Processing of NMR data at Triclinic was done using the program NUTS.

Optical Microscopy

Optical microscopy experiments were carried out on a Leica DM 2500 P compound microscope. Images were captured using a QImaging MicroPublisher 3.3 RTV camera. Images were collected at 10× magnification.

6.3 Assays

6.3.1 TNFα Inhibition Assay in PBMC

Peripheral blood mononuclear cells (PBMC) from normal donors are obtained by Ficoll Hypaque (Pharmacia, Piscataway, N.J., USA) density centrifugation. Cells are cultured in RPMI 1640 (Life Technologies, Grand Island, N.Y., USA) supplemented with 10% AB+human serum (Gemini Bio-products, Woodland, Calif., USA), 2 mM L-glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin (Life Technologies).

PBMC (2×105 cells) are plated in 96-well flat-bottom Costar tissue culture plates (Corning, N.Y., USA) in triplicate. Cells are stimulated with LPS (from Salmonella abortus equi, Sigma cat. no. L-1887, St. Louis, Mo., USA) at 1 ng/mL final in the absence or presence of compounds. Compounds provided herein are dissolved in DMSO (Sigma) and further dilutions are done in culture medium immediately before use. The final DMSO concentration in all assays can be about 0.25%. Compounds are added to cells 1 hour before LPS stimulation. Cells are then incubated for 18-20 hours at 37° C. in 5% CO2, and supernatants are then collected, diluted with culture medium and assayed for TNFα levels by ELISA (Endogen, Boston, Mass., USA). IC50s are calculated using non-linear regression, sigmoidal dose-response, constraining the top to 100% and bottom to 0%, allowing variable slope (GraphPad Prism v3.02).

6.3.2 IL-2 and MIP-3a Production by T Cells

PBMC are depleted of adherent monocytes by placing 1×108 PBMC in 10 ml complete medium (RPMI 1640 supplemented with 10% heat-inactivated fetal bovine serum, 2 mM L-glutamine, 100 U/ml penicillin, and 100 μg/ml streptomycin) per 10 cm tissue culture dish, in 37° C., 5% CO2 incubator for 30-60 minutes. The dish is rinsed with medium to remove all non-adherent PBMC. T cells are purified by negative selection using the following antibody (Pharmingen) and Dynabead (Dynal) mixture for every 1×108 non-adherent PBMC: 0.3 ml Sheep anti-mouse IgG beads, 15 μl anti-CD16, 15 μl anti-CD33, 15 μl anti-CD56, 0.23 ml anti-CD19 beads, 0.23 ml anti-HLA class II beads, and 56 μl anti-CD14 beads. The cells and bead/antibody mixture is rotated end-over-end for 30-60 minutes at 4° C. Purified T cells are removed from beads using a Dynal magnet. Typical yield is about 50% T cells, 87-95% CD3+ by flow cytometry.

Tissue culture 96-well flat-bottom plates are coated with anti-CD3 antibody OKT3 at 5 μg/ml in PBS, 100 μl per well, incubated at 37° C. for 3-6 hours, then washed four times with complete medium 100 μl/well just before T cells are added. Compounds are diluted to 20 times of final in a round bottom tissue culture 96-well plate. Final concentrations are about 10 μM to about 0.00064 μM. A 10 mM stock of compounds provided herein is diluted 1:50 in complete for the first 20× dilution of 200 μM in 2% DMSO and serially diluted 1:5 into 2% DMSO. Compound is added at 10 μl per 200 μl culture, to give a final DMSO concentration of 0.1%. Cultures are incubated at 37° C., 5% CO2 for 2-3 days, and supernatants analyzed for IL-2 and MIP-3a by ELISA (R&D Systems). IL-2 and MIP-3a levels are normalized to the amount produced in the presence of an amount of a compound provided herein, and EC50s calculated using non-linear regression, sigmoidal dose-response, constraining the top to 100% and bottom to 0%, allowing variable slope (GraphPad Prism v3.02).

6.3.3 Cell Proliferation Assay

Cell lines Namalwa, MUTZ-5, and UT-7 are obtained from the Deutsche Sammlung von Mikroorganismen and Zellkulturen GmbH (Braunschweig, Germany). The cell line KG-1 is obtained from the American Type Culture Collection (Manassas, Va., USA). Cell proliferation as indicated by 3H-thymidine incorporation is measured in all cell lines as follows.

Cells are plated in 96-well plates at 6000 cells per well in media. The cells are pre-treated with compounds at about 100, 10, 1, 0.1, 0.01, 0.001, 0.0001 and 0 μM in a final concentration of about 0.25% DMSO in triplicate at 37° C. in a humidified incubator at 5% CO2 for 72 hours. One microcurie of 3H-thymidine (Amersham) is then added to each well, and cells are incubated again at 37° C. in a humidified incubator at 5% CO2 for 6 hours. The cells are harvested onto UniFilter GF/C filter plates (Perkin Elmer) using a cell harvester (Tomtec), and the plates are allowed to dry overnight. Microscint 20 (Packard) (25 μl/well) is added, and plates are analyzed in TopCount NXT (Packard). Each well is counted for one minute. Percent inhibition of cell proliferation is calculated by averaging all triplicates and normalizing to the DMSO control (0% inhibition). Each compound is tested in each cell line in three separate experiments. Final IC50s are calculated using non-linear regression, sigmoidal dose-response, constraining the top to 100% and bottom to 0%, allowing variable slope. (GraphPad Prism v3.02).

6.3.4 Immunoprecipitation and Immunoblot

Namalwa cells are treated with DMSO or an amount of a compound provided herein for 1 hour, then stimulated with 10 U/ml of Epo (R&D Systems) for 30 minutes. Cell lysates are prepared and either immunoprecipitated with Epo receptor Ab or separated immediately by SDS-PAGE. Immunoblots are probed with Akt, phospo-Akt (Ser473 or Thr308), phospho-Gab1 (Y627), Gab1, IRS2, actin and IRF-1 Abs and analyzed on a Storm 860 Imager using ImageQuant software (Molecular Dynamics).

6.3.5 Cell Cycle Analysis

Cells are treated with DMSO or an amount of a compound provided herein overnight. Propidium iodide staining for cell cycle is performed using CycleTEST PLUS (Becton Dickinson) according to manufacturer's protocol. Following staining, cells are analyzed by a FACSCalibur flow cytometer using ModFit LT software (Becton Dickinson).

6.3.6 Apoptosis Analysis

Cells are treated with DMSO or an amount of a compound provided herein at various time points, then washed with annexin-V wash buffer (BD Biosciences). Cells are incubated with annexin-V binding protein and propidium iodide (BD Biosciences) for 10 minutes. Samples are analyzed using flow cytometry.

6.3.7 Luciferase Assay

Namalwa cells are transfected with 4 μg of AP1-luciferase (Stratagene) per 1×106 cells and 3 μl Lipofectamine 2000 (Invitrogen) reagent according to manufacturer's instructions. Six hours post-transfection, cells are treated with DMSO or an amount of a compound provided herein. Luciferase activity is assayed using luciferase lysis buffer and substrate (Promega) and measured using a luminometer (Turner Designs).

The embodiments described above are intended to be merely exemplary, and those skilled in the art will recognize, or will be able to ascertain using no more than routine experimentation, numerous equivalents of specific compounds, materials, and procedures. All such equivalents are considered to be within the scope of the disclosure and are encompassed by the appended claims.

All of the patents, patent applications and publications referred to herein are incorporated herein in their entireties. Citation or identification of any reference in this application is not an admission that such reference is available as prior art. The full scope of the disclosure is better understood with reference to the appended claims.

Claims

1. A solid form comprising (a) 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione, or a pharmaceutically acceptable salt, solvate, hydrate, stereoisomer, prodrug, or clathrate thereof; and (b) a coformer.

2. The solid form of claim 1, wherein the coformer is benzoic acid, gallic acid, glycolic acid, hippuric acid, magnesium bromide, malonic acid, maltol, oxalic acid, propyl gallate, sodium lauryl sulfate, L-tartaric acid, vanillic acid, or zinc chloride.

3. The solid form of claim 2, wherein the coformer is benzoic acid.

4. The solid form of claim 2, wherein the coformer is gallic acid.

5. The solid form of claim 2, wherein the coformer is glycolic acid.

6. The solid form of claim 2, wherein the coformer is hippuric acid.

7. The solid form of claim 2, wherein the coformer is magnesium bromide.

8. The solid form of claim 2, wherein the coformer is malonic acid.

9. The solid form of claim 2, wherein the coformer is maltol.

10. The solid form of claim 2, wherein the coformer is oxalic acid.

11. The solid form of claim 2, wherein the coformer is propyl gallate.

12. The solid form of claim 2, wherein the coformer is sodium lauryl sulfate.

13. The solid form of claim 2, wherein the coformer is L-tartaric acid.

14. The solid form of claim 2, wherein the coformer is vanillic acid.

15. The solid form of claim 2, wherein the coformer is zinc chloride.

16. The solid form of any one of claims 1 to 15, wherein the molar ratio of 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione to the coformer is about 1:1.

17. The solid form of any one of claims 1 to 16, which is substantially crystalline.

18. The solid form of any one of claims 1 to 17, which is substantially a cocrystal.

19. The solid form of any one of claims 1 to 18, which is greater than 80% by weight, greater than 90% by weight, greater than 95% by weight, greater than 97% by weight, or greater than 99% by weight a cocrystal.

20. The solid form any one of claims 1 to 19, which is substantially physically pure.

21. The solid form of claim 20, which is substantially free of other solid forms of 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione.

22. The solid form of any one of claims 1 to 21, which is substantially chemically pure.

23. The solid form of any one of claims 1 to 22, which is substantially free of solvent.

24. The solid form of any one of claims 1 to 23, which is substantially free of water.

25. The solid form of any one of claims 1 to 24, further comprising amorphous 3-(4-amino-1-oxo-1,3 dihydro-isoindol-2-yl)-piperidine-2,6-dione.

26. The solid form of claim any one of claims 1 to 25, which is stable.

27. The solid form of claim any one of claims 1 to 26, which is substantially crystalline and thermally stable.

28. A pharmaceutical composition comprising the solid form of any one of claims 1 to 27.

29. The pharmaceutical composition of claim 28, further comprising a pharmaceutically acceptable excipient or carrier.

30. The pharmaceutical composition of claim 28 or 29, which is a single unit dosage form.

31. The pharmaceutical composition of any one of claims 28 to 30, which is a tablet.

32. The pharmaceutical composition of any one of claims 28 to 31, which is a capsule.

33. The pharmaceutical composition of any one of claims 28 to 32, wherein the lenalidomide is in an amount of from about 0.1 to about 5 mg.

34. A method of treating a disease, comprising administering the solid form of any one of claims 1 to 27 or the pharmaceutical composition of any one of claims 28 to 33 to a subject in need thereof.

35. The method of claim 34, wherein the disease is multiple myeloma.

36. The method of claim 34, wherein the disease is transfusion-dependent anemia due to Low- or Intermediate-1-risk myelodysplastic syndromes (MDS) associated with a deletion 5q cytogenetic abnormality with or without additional cytogenetic abnormalities.

37. The method of any one of claims 34 to 36, further comprising administering a second active agent.

38. The method of any one of claim 37, wherein the second active agent is dexamethasone.

39. The method of any one of claims 34 to 38, wherein the subject was previously treated with a prior therapy.

Patent History
Publication number: 20180000808
Type: Application
Filed: Sep 18, 2017
Publication Date: Jan 4, 2018
Inventors: G. Patrick Stahly (West Lafayette, IN), David Jonaitis (Brookston, IN), Ho-Wah Hui (Basking Ridge, NJ), Kevin J. Klopfer (Flemington, NJ)
Application Number: 15/707,876
Classifications
International Classification: A61K 31/454 (20060101); A61K 31/573 (20060101); C07D 209/46 (20060101); A61K 45/06 (20060101); A61K 9/20 (20060101);