CXCR4 inhibitor compositions and methods of making and using

文档序号:589428 发布日期:2021-05-25 浏览:10次 中文

阅读说明:本技术 Cxcr4抑制剂组合物以及制备和使用的方法 (CXCR4 inhibitor compositions and methods of making and using ) 是由 K·M·J·布兰兹 于 2019-08-30 设计创作,主要内容包括:本发明提供了用于治疗、预防或改善与趋化因子受体(例如CXCR4)有关的疾病、病症或病状的组合物和使用方法。(The present invention provides compositions and methods of use for treating, preventing, or ameliorating a disease, disorder, or condition associated with a chemokine receptor (e.g., CXCR 4).)

1. An X4P-001 composition comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and a detectable amount of at least one of the following compounds:

or a pharmaceutically acceptable salt thereof; and wherein the X4P-001 composition does not include a detectable amount of the following compound:

or a pharmaceutically acceptable salt thereof.

2. The X4P-001 composition of claim 1, wherein the X4P-001 composition comprises at least a detectable amount of each of I-2, I-3, I-5, I-6, and I-7; or a pharmaceutically acceptable salt thereof.

3. The X4P-001 composition of claim 1 or 2, wherein the amount of I-2 or a pharmaceutically acceptable salt thereof is less than about 0.3% w/w of the X4P-001 composition.

4. The X4P-001 composition of any one of claims 1-3, wherein the amount of I-3 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

5. The X4P-001 composition of any one of claims 1-4, wherein the amount of I-5 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

6. The X4P-001 composition of any one of claims 1-5, wherein the amount of I-6 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

7. The X4P-001 composition of any one of claims 1-6, wherein the amount of I-7 or a pharmaceutically acceptable salt thereof is less than about 0.25% w/w of the X4P-001 composition.

8. The X4P-001 composition of claim 3, wherein the amount of I-2 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.3% w/w of the X4P-001 composition.

9. The X4P-001 composition of claim 4, wherein the amount of I-3 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

10. The X4P-001 composition of claim 5, wherein the amount of I-5 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

11. The X4P-001 composition of claim 6, wherein the amount of I-6 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

12. The X4P-001 composition of claim 7, wherein the amount of I-7 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.25% w/w of the X4P-001 composition.

13. The X4P-001 composition of claim 1, wherein:

(a) the amount of I-2 or a pharmaceutically acceptable salt thereof is less than about 0.3% w/w of the X4P-001 composition;

(b) the amount of I-3 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition;

(c) the amount of I-5 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition;

(d) wherein the amount of I-6 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition; and is

(e) The amount of I-7 or a pharmaceutically acceptable salt thereof is less than about 0.25% w/w of the X4P-001 composition.

14. The X4P-001 composition of claim 1, wherein:

(a) the amount of I-2 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.3% w/w of the X4P-001 composition;

(b) the amount of I-3 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition;

(c) the amount of I-5 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition;

(d) the amount of I-6 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition; and is

(e) The amount of I-7 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.25% w/w of the X4P-001 composition.

15. The X4P-001 composition of claim 1, wherein:

(a) the amount of I-2 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.2% w/w of the X4P-001 composition;

(b) the amount of I-3 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.2% w/w of the X4P-001 composition;

(c) the amount of I-5 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.2% w/w of the X4P-001 composition;

(d) the amount of I-6 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition; and is

(e) The amount of I-7 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.25% w/w of the X4P-001 composition.

16. The X4P-001 composition of any one of claims 13-15, optionally further comprising the following compound:

or an isomeric form thereof; or a pharmaceutically acceptable salt thereof, in an amount of from about 0.02 to about 0.5% w/w of the X4P-001 composition.

17. A pharmaceutical composition comprising the X4P-001 composition of any one of claims 1-16 and a pharmaceutically acceptable excipient, adjuvant, carrier, or vehicle.

18. The pharmaceutical composition of claim 17, wherein the pharmaceutically acceptable adjuvant comprises at least one diluent, disintegrant, lubricant, and glidant.

19. A unit dosage form comprising a pharmaceutical composition, the pharmaceutical composition comprising:

(a) about 10-20%, by weight of the composition, of the X4P-001 composition of any one of claims 1-16;

(b) from about 70% to about 85%, by weight of the composition, of microcrystalline cellulose;

(c) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(d) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate; and

(e) from about 0.1% to about 1.0% by weight of the composition of colloidal silica.

20. A unit dosage form comprising a pharmaceutical composition, the pharmaceutical composition comprising:

(a) about 10-20%, by weight of the composition, of the X4P-001 composition of any one of claims 1-16;

(b) from about 70% to about 85%, by weight of the composition, of microcrystalline cellulose;

(c) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(d) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate; and

(e) from about 0.1% to about 1.0% by weight of the composition of colloidal silica.

21. A unit dosage form comprising a pharmaceutical composition, the pharmaceutical composition comprising:

(a) from about 35-75%, by weight of the composition, of the X4P-001 composition of any one of claims 1-16;

(b) from about 5% to about 28%, by weight of the composition, of microcrystalline cellulose;

(c) from about 7% to about 30%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 2% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.3% to about 2.5%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.05% to about 1.2%, by weight of the composition, of colloidal silica; and

(g) from about 0.2% to about 1.2%, by weight of the composition, of sodium lauryl sulfate.

22. A method of treating a disease, disorder or condition associated with CXCR4 in a subject in need thereof, comprising administering to the subject an effective amount of the X4P-001 composition of any one of claims 1-16.

23. The method of claim 22, wherein the disease, disorder, or condition is a cancer selected from: renal cell carcinoma, RCC, or renal cancer; hepatocellular carcinoma HCC or hepatoblastoma, or liver cancer; melanoma; breast cancer; colorectal carcinoma, or colorectal carcinoma; colon cancer; rectal cancer; anal cancer; non-small cell lung cancer NSCLC; small cell lung cancer SCLC; epithelial carcinoma of the ovary; ovarian cancer; fallopian tube cancer; papillary serosa adenocarcinoma or uterine papillary serous carcinoma UPSC; prostate cancer; testicular cancer; gallbladder cancer; hepatobiliary cancer; soft tissue sarcoma and bone synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; ewing's sarcoma; undifferentiated thyroid cancer; adrenocortical carcinoma; pancreatic cancer; pancreatic ductal carcinoma; pancreatic cancer; gastrointestinal/gastric GIST cancer; lymphoma; squamous cell carcinoma of the head and neck SCCHN; salivary gland cancer; glioma or brain cancer; neurofibroma-1 associated malignant peripheral nerve sheath tumor MPNST; waldenstrom's macroglobulinemia; or neural tube blastoma.

24. The method of claim 22, wherein the disease, disorder, or condition is a cancer selected from: advanced renal cell carcinoma, clear cell renal carcinoma ccRCC, papillary renal carcinoma, metastatic melanoma, or waldenstrom's macroglobulinemia.

25. The method of claim 22, wherein the disease, disorder, or condition is primary immunodeficiency.

26. The method of claim 22, wherein the disease, disorder or condition is selected from warts, hypogammaglobulinemia, infection, myeloagranulocytosis (WHIM) syndrome, severe congenital neutropenia SCN, GATA2 deficiency (single MAC syndrome), idiopathic CD4+ T lymphopenia ICL, or westkott-aldrich syndrome.

27. The method of claim 22, wherein the disease, disorder, or condition is WHIM syndrome.

28. The method of claim 22, wherein the disease, disorder, or condition is SCN.

Technical Field

The present invention relates to compounds that inhibit the type 4C-X-C receptor (CXCR 4). The invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.

Cross Reference to Related Applications

This application claims the benefit of U.S. provisional patent application No. 62,726,010 filed on 31/8/2018 and the priority of U.S. patent application No. 16/215,963 filed on 11/12/2018, each of which is incorporated herein by reference in its entirety.

Background

The type 4C-X-C chemokine receptor (CXCR4), also known as fusin or clade 184(CD184), is a seven-transmembrane G-protein coupled receptor (GPCR) belonging to the class I GPCR or rhodopsin-like GPCR family. Under normal physiological conditions, CXCR4 plays multiple roles and is expressed primarily in the hematopoietic and immune systems. CXCR4 was initially found to be one of the co-receptors involved in the entry of Human Immunodeficiency Virus (HIV) into cells. Subsequent studies have shown that it is expressed in many tissues, including brain, thymus, lymphoid tissues, spleen, stomach, and small intestine, as well as in specific cell types, such as Hematopoietic Stem Cells (HSCs), mature lymphocytes, and fibroblasts. CXCL12, previously referred to as SDF-1 α, is the only known CXCR4 ligand. CXCR4 mediates stem cell migration during embryonic development and as a response to injury and inflammation. CXCR4 has been demonstrated to play multiple roles in human disease (e.g., cell proliferative disorder, Alzheimer's disease, HIV, rheumatoid arthritis, pulmonary fibrosis, etc.). For example, expression of CXCR4 and CXCL12 has been noted in several tumor types. CXCL12 is expressed by cancer-associated fibroblasts (CAF) and is often present at high levels in the Tumor Microenvironment (TME). In clinical studies of a wide variety of tumor types, including breast, ovarian, renal, lung and melanoma, expression of CXCR4/CXCL12 has been associated with poor prognosis and with increased risk of metastasis to lymph nodes, lung, liver and brain (CXCL12 expression site). CXCR4 is frequently expressed on melanoma cells, particularly on the CD133+ population considered to represent melanoma stem cells; CXCL12 has been shown to be chemotactic for such cells in vitro and in murine models.

In addition, there is evidence that the CXCL12/CXCR4 axis causes loss or loss of tumor response to angiogenesis inhibitors (also referred to as "angiogenesis evasion"). In animal cancer models, interference with CXCR4 function has been shown to alter TME and sensitize tumors to immune attack by multiple mechanisms (e.g., elimination of tumor revascularization and increased ratio of CD8+ T cells to Treg cells). These effects lead to a significant reduction in tumor burden and to an extension of the overall survival of xenograft, syngeneic and transgenic cancer models. See Wanhan nan Tower (Vanharantana) et al (2013) Nature & medicine (Nat Med) 19: 50-56; gal (Gale) and McColl (1999) bioanalysis (BioEssays) 21: 17-28; haifeil (Highfill) et al (2014) science transformation medicine (Sci Transl Med) 6: ra 67; facciabene et al (2011) Nature 475: 226-230.

These data highlight the unmet significant need for CXCR4 inhibitors for the treatment of a variety of diseases and conditions mediated by aberrant or undesired receptor expression (e.g., cell proliferative disorders).

Disclosure of Invention

It has now been found that the presently disclosed X4P-001 compositions and pharmaceutically acceptable compositions thereof are effective as inhibitors of CXC receptor type 4 (CXCR 4). In one aspect, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and at least one compound selected from:

or a pharmaceutically acceptable salt thereof.

The X4P-001 compositions and pharmaceutically acceptable compositions thereof of the invention are useful for treating a variety of diseases, disorders, or conditions associated with CXCR4, such as hyperproliferative conditions, including a variety of cancers. Such diseases, disorders or conditions include those described herein.

Drawings

FIG. 1 shows a detailed summary of the manufacturing process of a capsule (unit dosage form) containing a solid pharmaceutical formulation of X4P-001.

FIG. 2 shows the flow of the original synthetic process (version 2 or Process 2) for the preparation of X4P-001 for clinical trials.

FIG. 3 shows a comparison of Process 3 (modified current synthesis) to Process 2 (original process) for the preparation of X4P-001.

Figure 4 shows a detailed comparison between the version 2 process and the version 3 process in terms of downstream continuous operations with respect to API processing and separation.

Figure 5 shows HPLC and MS data for X4P-001(PTL/ST/0511, batch 3-1 (prepared using process 2), 25 ℃/60% RH, t3 months). HPLC condition 1 (described in detail below); 1mg/mL in methanol, injection volume 100 u L.

Figure 6 shows HPLC and MS data for X4P-001(PTL/ST/0511, batch 3-1 (prepared using process 2), 25 ℃/60% RH, t3 months). HPLC condition 2 (described in detail below); 1mg/mL in methanol, injection volume 100 u L.

Figure 7 shows HPLC and MS data for X4P-001(PTL/ST/0511, batch 3-1 (prepared using process 2), 25 ℃/60% RH, t3 months). HPLC condition 2 (described in detail below); the sample concentration was 10mg/mL in methanol and 100. mu.L was injected.

Figure 8 shows HPLC and MS data for X4P-001 (degraded sample, 80 ℃/80% RH, t ═ 1 day). HPLC condition 2 (described in detail below); the sample concentration was 10mg/mL in methanol, 100. mu.L injection volume.

Figure 9 shows HPLC and MS data for X4P-001 (degraded sample, 80 ℃/80% RH, t ═ 7 days). HPLC condition 2 (described in detail below); the sample concentration was 10mg/mL in methanol, 100. mu.L injection volume.

Figure 10 shows HPLC and MS data for X4P-001(PTL/ST/0511, batch 3-1 (prepared using process 2), 25 ℃/60% RH, t3 months). HPLC condition 3 (described in detail below); the sample concentration was 10mg/mL in methanol, 100. mu.L injection volume.

Detailed Description

1.General description of certain aspects of the invention

In one aspect, the invention provides compounds and compositions thereof useful for treating, preventing, and/or reducing the risk of a disease, disorder, or condition in which CXCR4 is implicated. In some embodiments, such compounds include those of the formulae described herein or a pharmaceutically acceptable salt thereof.

In another aspect, the present invention provides compositions, including formulations and unit dosage forms, comprising X4P-001 (i.e., a compound of formula I, the structure of which is shown below), or a pharmaceutically acceptable salt thereof, wherein such compositions exhibit an improved purity profile. In some embodiments, the disclosed X4P-001 composition has a reduced level of known impurities (e.g., those described herein) and/or a reduced level of unknown impurities as compared to a similar composition prepared in a conventional manner.

In another aspect, the invention provides a composition of X4P-001 comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and at least one compound selected from:

or a pharmaceutically acceptable salt thereof.

2.Definition of

The compounds of the present invention include those compounds described generally above and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For the purposes of the present invention, chemical elements are identified according to CAS version of the periodic Table of the elements (Handbook of Chemistry and Physics, 75 th edition). In addition, the general principles of Organic Chemistry are described in "Organic Chemistry" (Organic Chemistry), "Thomas Sorrell", University Science Books (University Science Books), Shaoshi (Sausaltito): 1999, and "March's Advanced Organic Chemistry" (5 th edition, ed., Smith M.B (Smith, M.B.) and March J. (March, J.), John Wiley parent-Press (John Wiley & Sons), New York: 2001, the entire contents of each of which are incorporated herein by reference.

As used herein, the term "X4P-001 composition" or "disclosed X4P-001 composition" refers to a composition comprising a compound of formula I (i.e., X4P-001) or a pharmaceutically acceptable salt thereof in combination with at least one other compound selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7. For clarity, the "pharmaceutical composition" of the disclosed X4P-001 composition refers to a composition comprising a compound of formula I (i.e., X4P-001) or a pharmaceutically acceptable salt thereof in combination with at least one other compound selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, and a pharmaceutically acceptable excipient, such as an adjuvant, filler, binder, carrier, or vehicle.

For the sake of clarity, and without wishing to be bound by theory, it is believed that compound I-1 exists at least partially in isomeric (e.g., tautomeric) forms; for example, it is believed that Compound I-1 undergoes the following interconversion to become the compound of structure I-1 a:

thus, it is understood that the single isomers of I-1 and I-1a as well as tautomers and other isomeric forms are within the scope of the invention.

Other known or unknown impurities may be present in the disclosed X4P-001 composition. As used herein, the term "impurities" includes one or more degradation products produced during the storage of X4P-001 and/or one or more byproducts formed in the chemical reaction used to make X4P-001. In some embodiments, the impurities result from oxidation, photoinitiated decomposition, reaction with residual solvents (e.g., water or isopropyl acetate), side reactions that occur during the process used to prepare X4P-001, or reaction of X4P-001 with excipients present in the X4P-001 pharmaceutical composition.

The term "inhibitor" as used herein is defined as a compound that binds to and/or inhibits CXCR4 with a measurable affinity. In certain embodiments, the inhibitor has an IC of less than about 100 μ M, less than about 50 μ M, less than about 1 μ M, less than about 500nM, less than about 100nM, less than about 10nM, or less than about 1nM50And/or binding constants.

As used herein, the term "pharmaceutically acceptable salts" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, s.m. berge (s.m. berge) et al, in journal of medical science (j.pharmaceutical Sciences), 1977,66,1-19, describe in detail pharmaceutically acceptable salts, which are incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of the present invention include salts derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, non-toxic acid addition salts include salts of basic groups (e.g., amino groups) with inorganic acids (e.g., hydrochloric, hydrobromic, phosphoric, sulfuric, and perchloric acids) or organic acids (e.g., acetic, oxalic, maleic, tartaric, citric, succinic, or malonic acids), or by using other methods used in the art (e.g., ion exchange). Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, besylate, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodiates, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoates, pectinates, persulfates, laurates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoates, pectinates, persulfates, laurates, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate and the like.

Salts derived from suitable bases include alkali metal salts, alkaline earth metal salts, ammonium salts and N+(C1-4Alkyl radical)4And (3) salt. Representative alkali metal salts or alkaline earth metal salts include sodium salts, lithium salts, potassium salts, calcium salts, magnesium salts, and the like. Other pharmaceutically acceptable salts include, where appropriate, those using, for example, halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, (C)1-6Alkyl) sulfonates and arylsulfonates, and the like.

Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms of the structure; for example, R and S configurations, Z and E double bond isomers, and Z and E conformational isomers for each asymmetric center. Thus, single stereochemical isomers as well as enantiomeric, diastereomeric and geometric (or conformational) isomer mixtures of the compounds of the present invention are within the scope of the invention. Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.

As used herein, "therapeutically effective amount" or "effective amount" means the amount of a substance (e.g., a therapeutic agent, composition, and/or formulation) that elicits a desired biological response. In some embodiments, a therapeutically effective amount of a substance is an amount sufficient to treat, diagnose, prevent, and/or delay the onset of a disease, condition, or disorder when administered as part of a dosing regimen to a subject suffering from or susceptible to the disease, condition, or disorder. As will be appreciated by those skilled in the art, the effective amount of a substance may vary depending on factors such as the desired biological indicator, the substance to be delivered, the target cell or tissue, and the like. For example, an effective amount of a compound in a formulation for treating a disease, condition, or disorder is an amount that alleviates, ameliorates, alleviates, inhibits, prevents, delays onset, reduces the severity of, and/or reduces the incidence of one or more symptoms or features of the disease, condition, or disorder. In some embodiments, a "therapeutically effective amount" is at least the minimum amount of a compound or composition containing a compound sufficient to treat one or more symptoms of a disease or disorder.

As used herein, the terms "treat", "treating" and "treating" refer to partially or completely alleviating, inhibiting, delaying the onset of, preventing, ameliorating and/or alleviating a disease or disorder or one or more symptoms of a disease or disorder as described herein. In some embodiments, the treatment may be administered after one or more symptoms have occurred. In some embodiments, the term "treating" includes preventing or halting the progression of the disease or disorder. In other embodiments, the treatment may be administered in the absence of symptoms. For example, treatment may be administered to susceptible individuals prior to the onset of symptoms (e.g., based on history of symptoms and/or based on genetic or other susceptibility factors). Treatment may also be continued after the symptoms have resolved, e.g., to prevent or delay their recurrence. Thus, in some embodiments, the term "treating" includes preventing the recurrence or recurrence of a disease or disorder.

As used herein, the term "CXCR 4 mediated" when referring to a disorder, disease, and/or condition means any disease, disorder, or condition for which CXCR4 or a mutant thereof is known to play a role. Thus, another embodiment of the invention relates to treating or lessening the severity of one or more diseases for which CXCR4 or a mutant thereof is known to play a role. "CXCR 4 mediated" also includes diseases, disorders, and conditions involving the CXCR4/CXCL12 axis.

As used herein, the term "unit dosage form" refers to a physically discrete unit of a therapeutic formulation suitable for the individual to be treated. It will be appreciated, however, that the total daily amount of the X4P-001 composition of the invention will be determined by the attending physician within the scope of sound medical judgment. The particular effective dosage level for any particular individual or organism will depend upon a variety of factors, including the disorder being treated and the severity of the disorder; the activity of the particular active agent employed; the particular composition employed; age, weight, general health, sex, and diet of the individual; the time of administration and rate of excretion of the particular active agent employed; the duration of treatment; drugs and/or other therapies used in combination or concomitantly with the particular compound employed, and similar factors well known in the medical arts.

3.Description of exemplary Compounds

It has now been found that the compounds of the present invention and compositions thereof are useful for treating, preventing and/or reducing the risk of diseases, disorders or conditions associated with CXCR4 or CXCR4 and its pathogenesis.

In one aspect, the present invention provides a composition comprising X4P-001 having a purity of at least 98.5%, or a pharmaceutically acceptable salt thereof. In some embodiments, X4P-001 in the composition has at least 98.5% purity and contains less than 1.5% w/w impurities. In some embodiments, the composition comprises X4P-001 or a pharmaceutically acceptable salt thereof in at least 98.6%, 98.7%, 98.8%, 98.9%, 99.0%, 99.1%, 99.2%, 99.3%, 99.4%, 99.5%, 99.6%, 99.7%, 99.8%, or 99.9% purity.

In one aspect, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and a detectable amount of at least one of the following compounds:

or a pharmaceutically acceptable salt thereof; and wherein the X4P-001 composition does not include a detectable amount of the following compound:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises each of I-1, I-2, I-3, I-5, I-6, and I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the amount of I-1 or a pharmaceutically acceptable salt thereof is less than about 0.5% w/w of the X4P-001 composition.

In some embodiments, the amount of I-2 or a pharmaceutically acceptable salt thereof is less than about 0.3% w/w of the X4P-001 composition.

In some embodiments, the amount of I-3 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-5 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-6 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-7 or a pharmaceutically acceptable salt thereof is less than about 0.25% w/w of the X4P-001 composition.

In some embodiments, the amount of I-1 or a pharmaceutically acceptable salt thereof is from about 0.02 to about 0.5% w/w of the X4P-001 composition.

In some embodiments, the amount of I-2 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.3% w/w of the X4P-001 composition.

In some embodiments, the amount of I-3 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-5 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-6 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-7 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.25% w/w of the X4P-001 composition.

In one aspect, the invention provides a pharmaceutical composition comprising the disclosed X4P-001 composition and a pharmaceutically acceptable adjuvant, carrier, or vehicle.

In some embodiments, the pharmaceutically acceptable adjuvant comprises at least one diluent, disintegrant, lubricant, and glidant.

In another aspect, the invention provides an X4P-001 composition or a pharmaceutical composition thereof, wherein:

(a) the amount of I-1 or a pharmaceutically acceptable salt thereof is less than about 0.5% w/w of the X4P-001 composition;

(b) the amount of I-2 or a pharmaceutically acceptable salt thereof is less than about 0.3% w/w of the X4P-001 composition;

(c) the amount of I-3 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition;

(d) the amount of I-5 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition;

(e) wherein the amount of I-6 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition; and is

(f) The amount of I-7 or a pharmaceutically acceptable salt thereof is less than about 0.25% w/w of the X4P-001 composition.

In one aspect, the invention provides an X4P-001 composition or a pharmaceutical composition thereof, wherein:

(a) the amount of I-1 or a pharmaceutically acceptable salt thereof is from about 0.02 to about 0.5% w/w of the X4P-001 composition;

(b) the amount of I-2 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.3% w/w of the X4P-001 composition;

(c) the amount of I-3 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition;

(d) the amount of I-5 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition;

(e) the amount of I-6 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition; and is

(f) The amount of I-7 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.25% w/w of the X4P-001 composition.

In one aspect, the present invention provides an X4P-001 composition, wherein:

(a) the amount of I-2 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.2% w/w of the X4P-001 composition;

(b) the amount of I-3 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.2% w/w of the X4P-001 composition;

(c) the amount of I-5 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.2% w/w of the X4P-001 composition;

(d) the amount of I-6 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.4% w/w of the X4P-001 composition; and is

(e) The amount of I-7 or a pharmaceutically acceptable salt thereof is from about 0.01 to about 0.25% w/w of the X4P-001 composition.

In some embodiments, the X4P-001 composition optionally further comprises I-1 or a pharmaceutically acceptable salt thereof in an amount of about 0.02 to about 0.5% w/w of the X4P-001 composition.

In another aspect, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 10% to about 20%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 70% to about 85%, by weight of the composition, of microcrystalline cellulose;

(c) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(d) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate; and

(e) from about 0.1% to about 1.0% by weight of the composition of colloidal silica.

In another aspect, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 35% to about 75%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 5% to about 28%, by weight of the composition, of microcrystalline cellulose;

(c) from about 7% to about 30%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 2% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.3% to about 2.5%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.05% to about 1.2%, by weight of the composition, of colloidal silica; and

(g) from about 0.2% to about 1.2%, by weight of the composition, of sodium lauryl sulfate.

In one aspect, the invention provides a method of treating, preventing, or reducing the risk of a disease, disorder, or condition associated with CXCR4 in an individual in need thereof, comprising administering to the individual an effective amount of a disclosed X4P-001 composition.

In some embodiments, the disease, disorder, or condition is a cancer selected from renal cancer, renal tumor, renal cancer, ovarian cancer, or melanoma.

In one aspect, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and at least one compound selected from:

or a pharmaceutically acceptable salt thereof.

In another aspect, the invention provides a composition of X4P-001 comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and at least one compound selected from:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and a compound of the structure:

or a pharmaceutically acceptable salt thereof. In some embodiments, the total weight of I-6 and any additional impurities present comprises no more than about 0.8% w/w in the X4P-001 composition.

In some embodiments, the X4P-001 composition comprises at least a detectable amount of I-6 or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises two, three, or four compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of two, three, or four compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises three compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of three compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises each of I-1, I-2, I-3, I-5, I-6, and I-7; or a pharmaceutically acceptable salt thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of each of I-1, I-2, I-3, I-5, I-6, and I-7; or a pharmaceutically acceptable salt thereof.

In one aspect, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof; and a detectable amount of at least one of the following compounds:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition does not include a detectable amount of the following compounds:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition does not include a detectable amount of the following compounds:

or a pharmaceutically acceptable salt thereof.

In some embodiments, the total amount of I-6 or a pharmaceutically acceptable salt thereof comprises no more than about 0.2% w/w of the X4P-001 composition, relative to the total weight of the X4P-001 composition of a compound of formula I and one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of I-6 or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises no more than 0.15% w/w of I-6 relative to the total weight of the X4P-001 composition of a compound of formula I and one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of I-6 or a pharmaceutically acceptable salt thereof.

In some embodiments, the total amount of I-6 or a pharmaceutically acceptable salt thereof comprises no more than about 0.2% w/w of the X4P-001 composition, relative to the total weight of the X4P-001 composition of a compound of formula I and one or more compounds selected from I-2, I-3, I-5, I-6, or I-7, or pharmaceutically acceptable salts thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of I-6 or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises no more than 0.15% w/w of I-6 relative to the total weight of the X4P-001 composition of a compound of formula I and one or more compounds selected from I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, the X4P-001 composition comprises at least a detectable amount of I-6 or a pharmaceutically acceptable salt thereof.

In some embodiments, one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, together comprise less than about 3.0% w/w of the X4P-001 composition, relative to the total weight of the X4P-001 composition of a compound of formula I and one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, is present in the X4P-001 composition in at least a detectable amount. In some embodiments, the X4P-001 composition comprises the compound of formula I, or a pharmaceutically acceptable salt thereof, and does not comprise the compound of formula I-4, or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In some embodiments, total organic impurities (including one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or pharmaceutically acceptable salts thereof) comprise less than about 4.0% w/w of the X4P-001 composition.

In some embodiments, one or more compounds selected from I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, together comprise less than about 3.0% w/w of the X4P-001 composition, relative to the total weight of the X4P-001 composition of a compound of formula I and one or more compounds selected from I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, one or more compounds selected from I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, is present in the X4P-001 composition in at least a detectable amount. In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and does not comprise any of compounds I-1 or I-4, or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In some embodiments, total organic impurities (including one or more compounds selected from I-2, I-3, I-5, I-6, or I-7, or pharmaceutically acceptable salts thereof) comprise less than about 4.0% w/w of the X4P-001 composition.

In some embodiments, the amount of I-1 or a pharmaceutically acceptable salt thereof is less than about 0.5% w/w of the X4P-001 composition.

In some embodiments, the amount of I-2 or a pharmaceutically acceptable salt thereof is less than about 0.3% w/w of the X4P-001 composition.

In some embodiments, the amount of I-3 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

In some embodiments, the X4P-001 composition does not include a detectable amount of I-4 or a pharmaceutically acceptable salt thereof.

In some embodiments, the amount of I-5 or a pharmaceutically acceptable salt thereof is less than about 0.07% w/w of the X4P-001 composition.

In some embodiments, the amount of I-6 or a pharmaceutically acceptable salt thereof is less than about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-7 or a pharmaceutically acceptable salt thereof is less than about 0.25% w/w of the X4P-001 composition.

In some embodiments, I-1, I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, is present in the X4P-001 composition in at least a detectable amount.

In some embodiments, the amount of I-1 or a pharmaceutically acceptable salt thereof is from about 0.02 to about 0.5% w/w of the X4P-001 composition.

In some embodiments, the amount of I-2 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.3% w/w of the X4P-001 composition.

In some embodiments, the amount of I-3 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-5 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.07% w/w of the X4P-001 composition.

In some embodiments, the amount of I-6 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.4% w/w of the X4P-001 composition.

In some embodiments, the amount of I-7 or a pharmaceutically acceptable salt thereof is about 0.01 to about 0.25% w/w of the X4P-001 composition.

In some embodiments, the X4P-001 composition comprises a compound of formula I or a pharmaceutically acceptable salt thereof in at least 99.3% purity (by HPLC) and comprises less than 0.7% (as measured by HPLC) of the total additional compounds selected from I-1, I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, the one or more additional compounds are present in at least a detectable amount.

In some embodiments, the X4P-001 composition comprises a compound of formula I or a pharmaceutically acceptable salt thereof in at least 99.3% purity (by HPLC) and comprises less than 0.7% (as measured by HPLC) of the total additional compounds selected from I-2, I-3, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, the one or more additional compounds are present in at least a detectable amount. In some embodiments, each of I-2, I-3, I-5, I-6, and I-7, or a pharmaceutically acceptable salt thereof, is present in at least a detectable amount (as measured by HPLC).

In one aspect, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof, and one or more compounds selected from the group consisting of:

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.5% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.3% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.4% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.5% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.4% w/w of the X4P-001 composition; or

Or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.25% w/w of the X4P-001 composition; wherein each% w/w is measured relative to the total weight of the X4P-001 composition of the compound of formula I and one or more compounds selected from I-1, I-2, I-3, I-5, I-6 or I-7. In some embodiments, the X4P-001 composition comprises the compound of formula I, or a pharmaceutically acceptable salt thereof, and does not comprise the compound of formula I-4, or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In one aspect, the present invention provides X4P-001 compositions comprising a compound of formula I:

or a pharmaceutically acceptable salt thereof, and one or more compounds selected from the group consisting of:

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.3% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.4% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.5% w/w of the X4P-001 composition;

or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.4% w/w of the X4P-001 composition; or

Or a pharmaceutically acceptable salt thereof, in an amount not exceeding about 0.25% w/w of the X4P-001 composition; wherein each% w/w is measured relative to the total weight of the X4P-001 composition of the compound of formula I and one or more compounds selected from I-2, I-3, I-5, I-6 or I-7.

In some embodiments, the chiral purity of the X4P-001 composition is at least about 97% enantiomeric excess (% ee).

In some embodiments, the X4P-001 composition comprises 7000, 6000, 5000, 4500, 4000, 3500, 3000, 2500, 2000, 1750, 1700, 1650, 1600, 1550, 1500, 1450, 1400, or 1350ppm or less of toluene.

In some embodiments, toluene is used as the crystallization solvent for the separation of X4P-001. In certain embodiments, the specification for residual toluene in the X4P-001 free base is that the X4P-001 composition comprises no more than 4500 ppm. In other embodiments, the X4P-001 composition comprises no more than 4000ppm, 3500ppm, 3000ppm, 2500ppm, 2000ppm, 1750ppm, 1700ppm, 1650ppm, 1600ppm, 1550ppm, 1500ppm, 1450ppm, 1400ppm, or 1350ppm of toluene. In some embodiments, a Permissible Daily Exposure (PDE) method is used. The term allowable daily exposure (PDE) is defined as the pharmaceutically acceptable uptake of residual solvent in the drug. See, for example, the Department of Health and Human Services (Department of Health and Human Services), Food and Drug Administration (FDA) published "guide to industry: impurity Q3C: residual solvent (guidelines for Industry: Q3 samples: Residual Solvents).

In some embodiments, the% purity of the X4P-001 composition decreases by less than 1% when the X4P-001 composition is stored at 25 ℃/60% relative humidity for three months as measured by HPLC.

In some embodiments, the X4P-001 composition further comprises one or more of the following:

and wherein compounds I-8, I-9, I-10, and/or I-11 are present in an amount of less than about 25 parts per million (ppm) of the X4P-001 composition.

In some embodiments, compounds I-8, I-9, I-10, and/or I-11 are present in an amount less than about 50, 40, 30, 20, 15, 10, 5, 4,3, 2, or 1ppm of the X4P-001 composition. In some embodiments, compounds I-8, I-9, I-10, and/or I-11 are each independently present in an amount between about 1ppm and about 25ppm or between about 100 parts per billion (ppb) and 4 ppm. In some embodiments, the X4P-001 composition comprises the compound of formula I, or a pharmaceutically acceptable salt thereof, and does not comprise the compound of formula I-4, or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In one aspect, the invention provides a pharmaceutical composition comprising the disclosed X4P-001 composition and a pharmaceutically acceptable adjuvant, carrier, or vehicle.

In one aspect, the invention provides a solid unit dosage form for oral formulation comprising the disclosed X4P-001 composition or pharmaceutical composition.

In some embodiments, the present invention provides a combination of the disclosed X4P-001 composition with another therapeutic agent.

In another aspect, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 10% to about 20%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 70% to about 85%, by weight of the composition, of microcrystalline cellulose;

(c) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(d) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate; and

(e) from about 0.1% to about 1.0% by weight of the composition of colloidal silica.

In another aspect, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 30% to about 40%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 20% to about 25%, by weight of the composition, of microcrystalline cellulose;

(c) from about 30% to about 35%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.1% to about 1.0%, by weight of the composition, of colloidal silica; and

(g) from about 0.1% to about 1.0%, by weight of the composition, of sodium lauryl sulfate.

In another aspect, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 35% to about 75%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 5% to about 28%, by weight of the composition, of microcrystalline cellulose;

(c) from about 7% to about 30%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 2% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.3% to about 2.5%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.05% to about 1.2%, by weight of the composition, of colloidal silica; and

(g) from about 0.2% to about 1.2%, by weight of the composition, of sodium lauryl sulfate.

In some embodiments, the unit dosage form is in the form of a capsule.

In some embodiments, the capsule comprises about 25mg, about 100mg, or about 200mg of X4P-001, or a pharmaceutically acceptable salt thereof.

In another aspect, the invention provides a method of treating, preventing, or reducing the risk of a disease, disorder, or condition associated with CXCR4 in a subject in need thereof, comprising administering to the subject the disclosed X4P-001 composition.

In some embodiments, the disease, disorder, or condition is cancer.

In some embodiments, the cancer is selected from renal cancer, renal tumor, renal cancer (including clear cell carcinoma and papillary renal cancer), ovarian cancer, or melanoma.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and one additional compound selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and two other compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and three additional compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and four other compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises the compound of formula I, or a pharmaceutically acceptable salt thereof, and does not comprise the compound of formula I-4, or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In some embodiments, the one or more additional compounds are present in at least a detectable amount.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and each of I-1, I-2, I-3, I-4, I-5, I-6, and I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and one additional compound selected from I-2, I-3, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and two other compounds selected from I-2, I-3, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and three additional compounds selected from I-2, I-3, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and four other compounds selected from I-2, I-3, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I or a pharmaceutically acceptable salt thereof, and does not comprise a compound of formulae I-1 and I-4 or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In some embodiments, the one or more additional compounds are present in at least a detectable amount.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and each of I-2, I-3, I-5, I-6, and I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition includes a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-1, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-3, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-4, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-5, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-6, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and I-7, or a pharmaceutically acceptable salt thereof.

In some embodiments, the X4P-001 composition comprises a compound of formula I or a pharmaceutically acceptable salt thereof; and I-1, I-2, I-3, I-4, I-5, I-6 and I-7; or a pharmaceutically acceptable salt thereof.

In some embodiments, the% w/w amount of a compound in the disclosed X4P-001 composition is measured by comparing the area percent of the compound in an HPLC chromatogram to any other compound present in the X4P-001 composition. For example, if measured in this manner, the presence of 0.2% w/w of compound I-6 in a composition of X4P-001 comprising I-6 and a compound of formula I means that the composition contains 0.2% (peak area%) of I-6 and 99.8% (peak area%) of a compound of formula I according to HPLC. In other embodiments, the% w/w is measured using another means known to one of ordinary skill in the art (such as those described herein).

In one aspect, the present invention provides a method of making the disclosed X4P-001 composition, wherein the composition is prepared substantially as described in the examples and figures herein.

In another aspect, the present invention provides a compound selected from those depicted in table 1 below.

Table 1: representative Compounds of the invention

In some embodiments, the present invention provides a compound depicted in table 1 above, or a pharmaceutically acceptable salt thereof.

In some embodiments, the present invention provides a pharmaceutical composition comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, and at least one compound depicted in table 1 above, or a pharmaceutically acceptable salt thereof. The composition may comprise 1, 2,3, 4,5, 6 or 7 of said compounds. In some embodiments, the composition does not comprise a detectable amount of compound I-4. In some embodiments, the composition does not comprise a detectable amount of compound I-1. In some embodiments, the composition does not contain a detectable amount of compounds I-1 and I-4.

Typically, the wt% of each impurity is determined by HPLC and measured initially or after storage, and optionally continuously during the shelf life of the X4P-001 composition. In some embodiments, the impurity content is measured after storage of the composition under stress conditions (conditions of elevated temperature, humidity, or both, for estimating the effect of long term storage under ambient conditions).

In some embodiments, a compound of formula I, or a pharmaceutically acceptable salt thereof, is present in an X4P-001 composition in an amount of at least about 96, 97, 97.5, 98, 98.5, 98.7, 98.9, 99.0, 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.75, 99.8, 99.85, 99.9, 99.95, 99.97, or 99.999 weight percent, wherein the percentages are based on the total weight of the free base of the compound and the X4P-001 composition. In other embodiments, the X4P-001 composition contains no more than about 2.0 area% HPLC total organic impurities, or in other embodiments, no more than about 5.0, 4.0, 2.0, 1.9, 1.8, 1.7, 1.6, 1.5, 1.25, 1, 0.75, 0.5, 0.25, 0.2, 0.1, 0.01, 0.005, or 0.001 area% HPLC total organic impurities, relative to the total area of the HPLC chromatogram.

In other embodiments, the X4P-001 composition contains no more than about 5.0, 4.0, 2.0, 1.9, 1.8, 1.7, 1.6, 1.5, 1.25, 1, 0.75, 0.5, 0.25, 0.2, 0.1, 0.01, 0.005, or 0.001 area% (as measured by HPLC) of compounds I-1, I-2, I-3, I-4, I-5, I-6, and I-7 relative to the total area of the HPLC chromatogram.

In other embodiments, the X4P-001 composition comprises a compound of formula I, or a pharmaceutically acceptable salt thereof, and one or more additional compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier. In some embodiments, the X4P-001 composition contains a compound of formula I, or a pharmaceutically acceptable salt thereof, in an amount from about 1% to about 99% by weight, wherein the percentages are based on the free base of the compound and the total weight of the X4P-001 composition. In other embodiments, the X4P-001 composition contains no more than about 2.0 area% HPLC total organic impurities, or in other embodiments, no more than about 5.0, 4.0, 2.0, 1.9, 1.8, 1.7, 1.6, 1.5, 1.25, 1.1, 1.05, 1, 0.95, 0.9, 0.8, 0.75, 0.7, 0.6, 0.5, 0.25, 0.2, 0.1, 0.01, 0.005, or 0.001 area% HPLC total organic impurities, relative to the total area of the HPLC chromatogram.

In some embodiments, one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, collectively or individually, comprise about 0.01 to 0.20 area% of the HPLC chromatogram with respect to a compound of formula I, or a pharmaceutically acceptable salt thereof. In some embodiments, one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, collectively or individually, comprise about 0.02 to 0.18, 0.03 to 0.16, 0.05 to 0.15, 0.075 to 0.13, 0.09 to 0.1, 0.1 to 0.2, or 0.15 to 0.2 area% of the HPLC chromatogram relative to a compound of formula I, or a pharmaceutically acceptable salt thereof. In some embodiments, the above area percentages of the HPLC chromatogram are measured relative to the total area of the HPLC chromatogram, and not relative to the peak area of the compound of formula I, or a pharmaceutically acceptable salt thereof.

In some embodiments, one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, collectively or individually, comprise less than about 5.0 weight percent (% w/w) or about 0.01-5.0% w/w relative to a compound of formula I, or a pharmaceutically acceptable salt thereof. In some embodiments, one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, collectively or individually comprise less than about 3.0% w/w of the X4P-001 composition; or about 0.02-4.0, 0.03-3.5, 0.05-3.1, 0.05-2.9, 0.05-2.5, 0.05-2.0, 0.05-1.8, 0.05-1.6, 0.05-1.5, 0.05-1.4, 0.05-1.2, 0.05-1.1, 0.05-1.0, 0.05-0.9, 0.05-0.8, 0.05-0.7, 0.05-0.6, 0.05-0.5, 0.05-0.4, 0.05-0.3, 0.05-0.2, 0.05-0.1, or about 0.1-0.5% w/w of the X4P-001 composition.

In some embodiments, total organic impurities (including one or more compounds selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or pharmaceutically acceptable salts thereof) comprise less than about 0.05% w/w, about 0.1, 0.5, 1.0, 2.0, 3.0, or about 4.0% w/w or less of the X4P-001 composition. In some embodiments, the total organic impurities comprise about 0.02-4.0, 0.03-3.5, 0.05-3.1, 0.05-2.9, 0.05-2.5, 0.05-2.0, 0.05-1.8, 0.05-1.6, 0.05-1.5, 0.05-1.4, 0.05-1.2, 0.05-1.1, 0.05-1.0, 0.05-0.9, 0.05-0.8, 0.05-0.7, 0.05-0.6, 0.05-0.5, 0.05-0.4, 0.05-0.3, 0.05-0.2, 0.05-0.1, or about 0.1-0.5% w/w of the X4P-001 composition.

In some embodiments, the amount of I-1, or a pharmaceutically acceptable salt thereof, is less than about 1.1%, 1%, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P001 composition. In some embodiments, I-1 or a pharmaceutically acceptable salt thereof is undetectable.

In some embodiments, the amount of I-2 or a pharmaceutically acceptable salt thereof is less than about 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P-001 composition.

In some embodiments, the amount of I-3 or a pharmaceutically acceptable salt thereof is less than about 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P-001 composition.

In some embodiments, the amount of I-4 or a pharmaceutically acceptable salt thereof is less than about 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P-001 composition. In some embodiments, I-4 or a pharmaceutically acceptable salt thereof is undetectable.

In some embodiments, the amount of I-5 or a pharmaceutically acceptable salt thereof is less than about 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P-001 composition.

In some embodiments, the amount of I-6 or a pharmaceutically acceptable salt thereof is less than about 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P-001 composition.

In some embodiments, the amount of I-7, or a pharmaceutically acceptable salt thereof, is less than about 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, or 0.01% w/w of the X4P-001 composition.

In some embodiments, the amount of I-1 or a pharmaceutically acceptable salt thereof is from about 0.001 to about 1.1%, from about 0.01 to about 0.8%, from 0.01 to about 0.7%, from about 0.01 to about 0.6%, from 0.01 to about 0.5%, from 0.01 to about 0.4%, from 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of I-2 or a pharmaceutically acceptable salt thereof is from about 0.001 to about 0.3%, from about 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of I-3 or a pharmaceutically acceptable salt thereof is from about 0.001 to about 0.4%, from about 0.01 to about 0.4%, from 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of I-4 or a pharmaceutically acceptable salt thereof is from about 0.001 to about 0.5%, from about 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of I-5 or a pharmaceutically acceptable salt thereof is from about 0.001 to about 0.5%, from about 0.01 to about 0.5%, from 0.01 to about 0.4%, from 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of I-6 or a pharmaceutically acceptable salt thereof is from about 0.001 to about 0.5%, from about 0.01 to about 0.4%, from about 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of I-7, or a pharmaceutically acceptable salt thereof, is from about 0.001 to about 0.5%, from about 0.01 to about 0.4%, from about 0.01 to about 0.3%, from 0.01 to about 0.2%, from 0.01 to about 0.1%, from 0.01 to about 0.09%, from 0.01 to about 0.08%, from 0.01 to about 0.07%, from 0.01 to about 0.06%, from 0.01 to about 0.05%, from 0.01 to about 0.03%, or from 0.01 to about 0.02% w/w of the X4P-001 composition.

In some embodiments, the amount of any additional or unknown impurity in the X4P-001 composition is from about 0.01 to about 0.2% w/w of the composition.

In some embodiments, the amount of parahydroxybenzoic acid present in the X4P-001 composition is from about 0.01 to about 0.5% w/w of the composition. In some embodiments, the composition is substantially free of parahydroxybenzoic acid. In some embodiments, no detectable amount of parahydroxybenzoic acid is present in the composition. In some embodiments, the X4P-001 composition comprises the compound of formula I, or a pharmaceutically acceptable salt thereof, and does not comprise the compound of formula I-4, or a pharmaceutically acceptable salt thereof, in terms of detectable amounts.

In some embodiments, the chiral purity of the X4P-001 composition is at least about 97% enantiomeric excess (% ee). In some embodiments, the chiral purity of the compound of formula I is at least 97% ee. In some embodiments, the chiral purity of the compound of formula I is at least 98% ee. In some embodiments, the chiral purity of the compound of formula I is at least 99% ee. In some embodiments, the chiral purity of the compound of formula I is at least 99.1, 99.2, 99.3, 99.4, 99.5, 99.6, 99.7, 99.8, or 99.9% ee.

In some embodiments, the present invention provides isolated forms of any of the compounds described above and herein. The term "sequestered" as used herein means to provide a compound in a form that is separated from other components that may be present in the common environment of the compound. In certain embodiments, the isolated compound is in solid form. In some embodiments, the isolated compound has at least about 50% purity, as determined by a suitable HPLC method. In certain embodiments, the isolated compound has at least about 60%, 70%, 80%, 90%, 95%, 98%, 99%, 99.5%, 99.6%, 99.7%, 99.8%, 99.9%, 99.95%, 99.99%, or 99.999% purity as determined by a suitable HPLC method. Percent purity can be measured as weight percent of the desired compound (w/w%), as area% relative to the total area of the HPLC chromatogram, or by other methods known in the art.

Methods for the preparation and analysis of certain compounds suitable for use in the present invention are disclosed in US7,354,934, WO 00/56729, USSN60/232,891 and USSN 60/234,510 and ann H. (An, H.); king T. (Wang, T.); mohan, V.); griffey r.h. (r.h.); cook P.D (Cook, P.D.) 1998,54, 3999-4012; each of which is incorporated herein by reference in its entirety.

The disclosed compounds can be purified by any means known in the art. Such means include, for example, silica gel column chromatography; medium Pressure Liquid Chromatography (MPLC); high Pressure Liquid Chromatography (HPLC); preparative HPLC (prep-HPLC); flash Chromatography (FC); liquid Chromatography (LC); supercritical Fluid Chromatography (SFC); thin Layer Chromatography (TLC); preparative TLC (prep-TLC); liquid chromatography-mass spectrometry (LC-MS, LCMS or LC/MS); recrystallizing; precipitating; grinding; distilling; derivatization; acid-base extraction, and the like.

For compounds, the terms "purified", "purified form" or "isolated and purified form" refer to the physical state of the compound after isolation from a synthetic process (e.g., from a reaction mixture) or natural source, or a combination thereof. Thus, for a compound, the terms "purified," "purified form," or "isolated and purified form" refer to the physical state of the compound (or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt or solvate of the compound, the stereoisomer, or the tautomer) after having been obtained from a purification process or a process described herein or well known to those skilled in the art (e.g., chromatography, recrystallization, etc.), the compound having sufficient purity to be suitable for in vivo or pharmaceutical use and/or being characterizable by standard analytical techniques described herein or well known to those skilled in the art.

As used herein, the term "detectable amount" means that a component present in a sample (e.g., a sample of the disclosed X4P-001 composition) is present in an amount that is at least detectable by analytical means known in the art. For example, in some embodiments, a "detectable amount" is an amount that is detectable at least by HPLC, LC-MS, mass spectrometry, NMR, or other analytical methods known to those skilled in the art or described herein.

Mixtures of diastereomers may be separated into their individual diastereomers on the basis of their physicochemical differences by methods well known to those skilled in the art, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting mixtures of enantiomers into mixtures of diastereomers with appropriate optically active compounds, for example chiral adjuvants, such as chiral alcohols or Mosher's acid chloride, separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Enantiomers can also be separated using a chiral HPLC column.

4.Use, formulation and administration

Pharmaceutically acceptable compositions

In one aspect, the invention provides an X4P-001 composition comprising a disclosed compound, a pharmaceutically acceptable salt, or a pharmaceutically acceptable derivative; or the disclosed X4P-001 composition; and a pharmaceutically acceptable excipient, carrier, adjuvant or vehicle. The amount of compound in the compositions of the invention is effective to produce a measurable inhibition of CXCR4 or a mutant thereof in a biological sample or patient. In certain embodiments, the amount of compound in the compositions of the invention is effective to produce a measurable inhibition of CXCR4 or a mutant thereof in a biological sample or patient. In certain embodiments, the compositions of the present invention are formulated for administration to a patient in need of such compositions. In some embodiments, the compositions of the present invention are formulated for oral administration to a patient.

As used herein, the term "patient" means an animal, preferably a mammal, and most preferably a human.

The term "pharmaceutically acceptable carrier, adjuvant or vehicle" refers to a non-toxic carrier, adjuvant or vehicle that does not destroy the pharmacological activity of the compounds formulated together. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of the present invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts), colloidal silica, magnesium trisilicate, polyvinylpyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene block polymers, polyethylene glycol and wool fat.

By "pharmaceutically acceptable derivative" is meant any non-toxic salt, ester, salt of an ester, or other derivative of a compound of the present invention which, upon administration to a recipient, is capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue thereof having inhibitory activity.

As used herein, the term "metabolites or residues thereof having inhibitory activity" means metabolites or residues thereof which are also inhibitors of CXCR4 or mutants thereof.

The compositions of the invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implantable reservoir. As used herein, the term "parenteral" includes subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the composition is administered orally, intraperitoneally, or intravenously. Sterile injectable forms of the compositions of the present invention may be aqueous or oleaginous suspensions. These suspensions may be formulated according to the techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be in the form of a sterile injectable solution or suspension in a parenterally-acceptable non-toxic diluent or solvent, for example as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium.

For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. Fatty acids (e.g., oleic acid and its glyceride derivatives) are suitable for the preparation of injectables, as are pharmaceutically-acceptable natural oils (e.g., olive oil or castor oil, especially in their polyoxyethylated versions). These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersants commonly used in the formulation of pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants such as Tweens, Spans, and other emulsifiers or bioavailability enhancers commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for formulation purposes.

The pharmaceutically acceptable compositions of the present invention may be administered orally in any orally acceptable dosage form, including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers that are commonly used include lactose and corn starch. Lubricating agents such as magnesium stearate are also typically added. For oral administration in capsule form, suitable diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.

Alternatively, the pharmaceutically acceptable compositions of the present invention may be administered in the form of suppositories for rectal administration. These suppositories can be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. The materials include cocoa butter, beeswax and polyethylene glycols.

The pharmaceutically acceptable compositions of the present invention may also be administered topically, particularly when the target of treatment includes areas or organs readily accessible by topical administration, including diseases of the eye, skin or lower intestinal tract. Topical formulations suitable for each of these areas or organs are readily prepared.

Topical administration to the lower intestinal tract may be achieved in rectal suppository formulations (see above) or in suitable enema formulations. Topical transdermal patches may also be used.

For topical administration, the provided pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active ingredient suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of the present invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the provided pharmaceutically acceptable compositions can be formulated as suitable lotions or creams containing the active ingredient suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.

For ophthalmic use, the provided pharmaceutically acceptable compositions may be formulated as micronized suspensions in pH adjusted isotonic sterile saline or, preferably, as solutions in pH adjusted isotonic sterile saline, with or without a preservative such as benzalkonium chloride. Alternatively, for ophthalmic use, the pharmaceutically acceptable composition may be formulated in an ointment such as petrolatum.

The pharmaceutically acceptable compositions of the present invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well known in the art of pharmaceutical formulation and may be prepared as solutions in physiological saline using benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.

The pharmaceutically acceptable compositions of the present invention are most preferably formulated for oral administration. Such formulations may be administered in the presence or absence of a food product. In some embodiments, the pharmaceutically acceptable compositions of the present invention are administered in the absence of food. In other embodiments, the pharmaceutically acceptable compositions of the present invention are administered in the presence of food.

The amount of a compound of the invention that can be combined with a carrier material to produce a composition in a single dosage form will vary depending on the host treated, the particular mode of administration. Preferably, the compositions provided should be formulated so that a dose of between 0.01-100 mg/kg body weight/day of inhibitor can be administered to a patient receiving these compositions.

It will also be understood that the specific dose and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination and the judgment of the treating physician and the severity of the particular disease undergoing therapy. The amount of the compound of the present invention in the composition will also depend on the particular compound in the composition.

The compounds and compositions according to the methods of the invention can be administered in any amount and by any route of administration effective to treat or reduce the severity of cancer, an autoimmune disorder, a primary immunodeficiency, a proliferative disorder, an inflammatory disorder, a neurodegenerative or neurological disorder, schizophrenia, a bone-related disorder, a liver disease, or a cardiac disorder. The exact amount required will vary from individual to individual, depending on the species, age, and general condition of the individual, the severity of the infection, the particular agent, its mode of administration, and the like. In some embodiments, the compounds of the present invention are formulated in unit dosage forms that are easy to administer and are uniform in dosage.

As used herein, the term "patient" or "individual" means an animal, preferably a mammal, and most preferably a human.

The pharmaceutically acceptable compositions of the present invention may be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (e.g., by powders, ointments, or drops), buccally, orally, or nasally, etc., depending on the severity of the infection being treated. In certain embodiments, the compounds of the present invention may be administered orally or parenterally, one or more times a day, at dosage levels of from about 0.01 mg to about 50mg, or for example from about 1mg to about 25mg, per kg of body weight of the individual per day, to achieve the desired therapeutic effect.

Oral liquid dosage forms include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, the oral compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.

Injectable preparations, for example sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that can be employed are water, ringer's solution (u.s.p.), and isotonic sodium chloride solution. In addition, sterile fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids, such as oleic acid, are used in the preparation of injectables.

The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporation of sterilizing agents in the form of sterile solid compositions that can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

In order to prolong the effect of the compounds of the invention, it is generally desirable to slow the absorption of the compounds from subcutaneous or intramuscular injection. This can be achieved by using liquid suspensions of crystalline or amorphous materials with poor water solubility. The rate of absorption of the compound then depends on its rate of dissolution, which in turn may depend on crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is achieved by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming a microcapsule matrix of the compound in a biodegradable polymer, such as polylactide-polyglycolide. Depending on the ratio of compound to polymer and the nature of the particular polymer used, the release rate of the compound can be controlled. Examples of other biodegradable polymers include poly (orthoesters) and poly (anhydrides). Depot injectable formulations are also prepared by entrapping the compounds in liposomes or microemulsions that are compatible with body tissues.

Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of the invention with suitable non-irritating excipients or carriers (e.g., cocoa butter, polyethylene glycol); or a suppository wax that is solid at ambient temperature but liquid at body temperature and therefore melts in the rectum or vaginal cavity and releases the active compound.

Oral solid dosage forms include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is admixed with: at least one pharmaceutically acceptable inert excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; b) binders such as carboxymethyl cellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and acacia; c) humectants, such as glycerol; d) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) dissolution retarders, such as paraffin; f) absorption promoters, such as quaternary ammonium compounds; g) wetting agents, such as cetyl alcohol and glycerol monostearate; h) absorbents such as kaolin and bentonite clay; and i) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.

Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose (lactose/milk sugar) and high molecular weight polyethylene glycols and the like. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. It may optionally contain an opacifying agent and may also have a composition such that it releases only or preferentially the active ingredient, optionally in a certain part of the intestinal tract, in a delayed manner. Examples of embedding compositions that may be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose (lactose/milk sugar) and high molecular weight polyethylene glycols and the like.

The active compound may also be in microencapsulated form with one or more excipients as indicated above. Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release control coatings, and other coatings well known in the pharmaceutical formulating art. In these solid dosage forms, the active compound may be mixed with at least one inert diluent (e.g., sucrose, lactose or starch). Such dosage forms may also contain, as is normal practice, additional substances other than inert diluents, such as tableting lubricants and other tableting aids, such as magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. It may optionally contain an opacifying agent and may also have a composition such that it releases only or preferentially the active ingredient, optionally in a certain part of the intestinal tract, in a delayed manner. Examples of embedding compositions that may be used include polymeric substances and waxes.

Dosage forms for topical or transdermal administration of the compounds of the present invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active ingredient is mixed under sterile conditions with a pharmaceutically acceptable carrier and any required preservatives or buffers as may be required. Ophthalmic formulations, ear drops and eye drops are also encompassed within the scope of the invention. In addition, the present invention contemplates the use of transdermal patches, which have the additional advantage of providing controlled delivery of the compound to the body. The dosage form may be manufactured by dissolving or dispensing the compound in a suitable medium. Absorption enhancers may also be used to increase the flux of the compound through the skin. The rate can be controlled by providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.

In certain embodiments, the composition is formulated for oral administration in the form of a tablet or capsule. In some embodiments, the composition comprising X4P-001 is formulated for oral administration in capsule form.

In certain embodiments, the provided methods comprise administering to a patient one or more capsules comprising 10mg to 1200mg of X4P-001 active ingredient and one or more pharmaceutically acceptable excipients. In certain embodiments, the capsule comprises hard gelatin.

In certain embodiments, the present invention provides a pharmaceutical composition comprising an X4P-001 composition, one or more diluents, disintegrants, lubricants, glidants, and wetting agents. In some embodiments, the present invention provides a pharmaceutical composition comprising 10mg to 1200mg of X4P-001 composition, microcrystalline cellulose, dibasic calcium phosphate dihydrate, croscarmellose sodium, sodium stearyl fumarate, colloidal silicon dioxide, and sodium lauryl sulfate. In some embodiments, the present invention provides a unit dosage form, wherein the unit dosage form comprises a pharmaceutical composition comprising 10-200mg X4P-001 composition, microcrystalline cellulose, dibasic calcium phosphate dihydrate, croscarmellose sodium, sodium stearyl fumarate, colloidal silicon dioxide, and sodium lauryl sulfate. In certain embodiments, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising X4P-001 present in an amount of about 10mg, about 20mg, about 25mg, about 50mg, about 75mg, about 100mg, about 150mg, about 200mg, about 250mg, about 300mg, about 400mg, about 450mg, about 500mg, about 600mg, about 700mg, about 750mg, about 800mg, about 900mg, about 1000mg, about 1100mg, or about 1200 mg. In some embodiments, a provided composition (or unit dosage form) is administered to a patient once a day, twice a day, three times a day, or four times a day. In some embodiments, a provided composition (or unit dosage form) is administered to a patient once a day or twice a day.

In some embodiments, the present invention provides a pharmaceutical composition comprising:

(a) from about 30% to about 40%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 20% to about 25%, by weight of the composition, of microcrystalline cellulose;

(c) from about 30% to about 35%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.1% to about 1.0%, by weight of the composition, of colloidal silica; and

(g) from about 0.1% to about 1.0%, by weight of the composition, of sodium lauryl sulfate.

In some embodiments, the present invention provides a pharmaceutical composition comprising:

(a) from about 8% to about 25%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 65% to about 85%, by weight of the composition, of microcrystalline cellulose;

(c) from about 2% to about 10%, by weight of the composition, of croscarmellose sodium;

(d) from about 0.1% to about 3%, by weight of the composition, of sodium stearyl fumarate; and

(e) from about 0.05% to about 0.7%, by weight of the composition, of colloidal silica.

In some embodiments, the present invention provides a pharmaceutical composition comprising:

(a) from about 25% to 45%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 10% to about 35%, by weight of the composition, of microcrystalline cellulose;

(c) from about 15% to about 45%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 2% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.3% to about 2.5%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.05% to about 1.2%, by weight of the composition, of colloidal silica; and

(g) from about 0.2% to about 1.2%, by weight of the composition, of sodium lauryl sulfate.

In some embodiments, the present invention provides a pharmaceutical composition comprising:

(a) from about 35% to about 75%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 5% to about 28%, by weight of the composition, of microcrystalline cellulose;

(c) from about 7% to about 30%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 2% to about 10%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.3% to about 2.5%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.05% to about 1.2%, by weight of the composition, of colloidal silica; and

(g) from about 0.2% to about 1.2%, by weight of the composition, of sodium lauryl sulfate.

In some embodiments, the X4P-001 composition is present in an amount of about 10mg, about 20mg, about 25mg, about 50mg, about 75mg, about 100mg, about 150mg, about 200mg, about 250mg, about 300mg, about 400mg, about 450mg, about 500mg, about 600mg, about 700mg, about 750mg, about 800mg, about 900mg, about 1000mg, about 1100mg, or about 1200 m.

In some embodiments, the composition comprises about 37 wt% of the disclosed X4P-001 composition.

In some embodiments, the present invention provides a unit dosage form comprising the disclosed X4P-001 composition or pharmaceutical composition.

In some embodiments, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 10% to about 30%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 60% to about 80%, by weight of the composition, of microcrystalline cellulose;

(c) from about 5% to about 10%, by weight of the composition, of croscarmellose sodium;

(d) from about 0.5% to about 2%, by weight of the composition, of sodium stearyl fumarate; and

(e) from about 0.1% to about 1.0% by weight of the composition of colloidal silica.

In some embodiments, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) about 14.7%, by weight of the composition, of the disclosed X4P-001 composition;

(b) about 78.1%, by weight of the composition, of microcrystalline cellulose;

(c) about 6.0%, by weight of the composition, croscarmellose sodium;

(d) about 1.0%, by weight of the composition, sodium stearyl fumarate; and

(e) about 0.2% colloidal silica, by weight of the composition.

In some embodiments, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) from about 10% to about 20%, by weight of the composition, of the disclosed X4P-001 composition;

(b) from about 25% to about 40%, by weight of the composition, of microcrystalline cellulose;

(c) from about 35% to about 55%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) from about 4% to about 15%, by weight of the composition, of croscarmellose sodium;

(e) from about 0.3% to about 2%, by weight of the composition, of sodium stearyl fumarate;

(f) from about 0.1% to about 1.5%, by weight of the composition, of colloidal silica; and

(g) from about 0.1% to about 1.5%, by weight of the composition, of sodium lauryl sulfate.

In some embodiments, the present invention provides a unit dosage form comprising a pharmaceutical composition comprising:

(a) about 12.85%, by weight of the composition, of the disclosed X4P-001 composition;

(b) about 31.92%, by weight of the composition, of microcrystalline cellulose;

(c) about 44.4%, by weight of the composition, dibasic calcium phosphate dihydrate;

(d) about 8.33%, by weight of the composition, croscarmellose sodium;

(e) about 1.38%, by weight of the composition, sodium stearyl fumarate;

(f) about 0.42%, by weight of the composition, of colloidal silica; and

(g) about 0.7%, by weight of the composition, of sodium lauryl sulfate.

Other compositions and methods of use of X4P-001 are described in International patent application No. PCT/US2016/066634, which is incorporated herein by reference in its entirety.

Since it may be desirable to administer a combination of active compounds, for example for the purpose of treating a particular disease or condition, it may be convenient within the scope of the present invention to combine two or more pharmaceutical compositions, at least one of which contains a compound according to the invention, in a kit form suitable for co-administration of the compositions. Thus, the kit of the invention comprises two or more separate pharmaceutical compositions, at least one of which contains the X4P-001 composition of the invention, and means for separately retaining the compositions, such as a container, a dispensing bottle or a dispensing foil packet. Examples of such kits are the familiar blister packs used to encapsulate tablets, capsules, and the like.

The kits of the invention are particularly suitable for administering different dosage forms (e.g. oral and parenteral), for administering the respective compositions at different dosing intervals, or for titrating the respective compositions against each other. To aid compliance, the kit typically includes instructions for administration and may be equipped with memory aids.

Use of compounds and pharmaceutically acceptable compositions

It has been found that during the synthesis of X4P-001 (e.g., the compounds shown in table 1 above or stereoisomers or pharmaceutically acceptable salts thereof), certain impurities are produced. The separation and characterization of various impurities can be used for a number of purposes. Generally, pharmaceutical compositions require a high level of purity to meet regulatory standards for drug quality and purity. For example, in the synthesis of X4P-001, impurities, including degradation products or by-products of manufacture, are often formed which may interfere with the therapeutic effect of X4P-001 and/or, if present in sufficiently high amounts, may be toxic. It is therefore desirable to be able to determine the presence and amount of such impurities and monitor the chemical purity, including stereochemical purity, of X4P-001. For this reason, it is important to identify, isolate and chemically characterize impurities that can be used as standards in chromatographic procedures to confirm the purity of X4P-001.

Thus, in one aspect, the invention provides a method of preparing a disclosed compound, or a pharmaceutically acceptable salt thereof, comprising contacting one or more appropriate starting materials under conditions such as those shown in the examples below to prepare the compound, or a pharmaceutically acceptable salt thereof. In some embodiments, the compound or pharmaceutically acceptable salt thereof is used as a reference standard and/or in a method of determining whether an impurity is present in a sample (e.g., a sample of a compound of formula I or pharmaceutically acceptable salt thereof).

The present invention also provides a method for determining impurities, comprising: injecting a reference solution comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, into an HPLC column under a set of conditions to obtain a first HPLC chromatogram, wherein the amount and/or chemical identity of the compound present in the reference solution is known; injecting a sample solution comprising a compound of formula I, or a pharmaceutically acceptable salt thereof, into an HPLC column under the set of conditions to obtain a second HPLC chromatogram; and determining the presence and/or amount of the compound in the sample solution. In some embodiments, the reference solution is injected multiple times. In some embodiments, determining comprises comparing the retention time of a peak in the first HPLC chromatogram to a peak in the second HPLC chromatogram to determine the presence of the compound in the sample solution. In other embodiments, determining comprises quantifying the peak area of the sample solution and the peak area of the reference solution on the HPLC chromatogram and thereby estimating the amount of the compound in the sample solution. In some embodiments, the HPLC column is a reverse phase column and the column is eluted using a mobile phase comprising water, methanol, trifluoroacetic acid or a mixture thereof.

The present invention also provides a method of determining the presence or amount of an impurity in a sample comprising or consisting essentially of a compound of formula I, or a pharmaceutically acceptable salt thereof, the method comprising: injecting a sample solution containing the material plus a reference compound of known chemical structure (e.g., compound I-1, I-2, I-3, I-4, I-5, I-6 or I-7) into an HPLC column in a single or continuous injection; obtaining an HPLC chromatogram; and determining the presence and/or amount of said compound in said material. In some embodiments, the HPLC column is a reverse phase column and the column is eluted using a mobile phase comprising water, methanol, trifluoroacetic acid or a mixture thereof. The method may further comprise recording the chemical identity of the compound and the amount of the compound as an impurity in written form.

In some embodiments, the method further comprises recording in written form the chemical identity of the compound and the amount of the compound in the material as an impurity. In some cases, the amount of compound in the material is determined as follows: (i) identifying a peak on the chromatogram corresponding to a peak on a control chromatogram of a compound known to have the structure of formula I-1, I-2, I-3, I-4, I-5, I-6, or I-7, (ii) identifying a peak on the chromatogram corresponding to the relative retention time of a compound known to have the structure of formula I-1, I-2, I-3, I-4, I-5, I-6, or I-7; and/or (iii) identifying a peak on the chromatogram corresponding to a known amount of a peak known to be a peak of a compound having the structure of formula I-1, I-2, I-3, I-4, I-5, I-6, or I-7. In some embodiments, the HPLC column is a reverse phase column and the column is eluted using a mobile phase comprising water, methanol, trifluoroacetic acid or a mixture thereof.

In some embodiments, the present invention provides a compound selected from I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, sufficiently pure to enable it to be used as a reference or standard in a variety of analytical methods (e.g., HPLC, GC, SFC, LCMS), as described more fully below. In some embodiments, the compound or pharmaceutically acceptable salt thereof can be isolated in at least 0.5% purity, at least 1% purity, at least 5% purity, at least 10% purity, at least 15% purity, at least 25% purity, at least 50% purity, at least 75% purity, at least 95% purity, or at least 97% purity. In some embodiments, the compound or pharmaceutically acceptable salt thereof is isolated and/or packaged in solid form.

In another aspect, the invention provides methods for determining the presence and/or amount of I-1, I-2, I-3, I-4, I-5, I-6 or I-7, or a pharmaceutically acceptable salt thereof. For example, a compound or a pharmaceutically acceptable salt thereof may be formed as an impurity during the synthesis of X4P-001. As used herein, the term "impurities" may refer to degradation products generated during storage of X4P-001 and/or byproducts formed in the chemical reaction that produces X4P-001. In one embodiment, the method includes: injecting a reference solution comprising I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, into an HPLC column under a set of conditions to obtain a first HPLC chromatogram, wherein the amount and/or chemical identity of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, present in the reference solution is known; injecting a sample solution comprising X4P-001 into the HPLC column under the same set of conditions to obtain a second HPLC chromatogram; and comparing the first HPLC chromatogram to the second HPLC chromatogram to determine the presence and/or amount of an impurity (I-1, I-2, I-3, I-4, I-5, I-6, or I-7; or a pharmaceutically acceptable salt thereof). A reference solution may be formed by dissolving a sample (e.g., a solid sample) of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in a first solvent, and a sample solution may be formed by dissolving the solid sample in a second solvent. In some embodiments, the reference solution may contain other compounds, where the amounts and/or characteristics of the other compounds are also known. In one embodiment, a sample (e.g., sample solution) can comprise X4P-001. It is to be understood that the invention may encompass other samples suspected of containing a compound selected from I-1, I-2, I-3, I-4, I-5, I-6 or I-7, or a pharmaceutically acceptable salt thereof.

In one embodiment, the presence or absence of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in the sample solution can be determined by comparing the retention time of the peaks in the first HPLC chromatogram to the retention time of the peaks in the second HPLC chromatogram. For example, a standard solution comprising I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, can produce a chromatogram having a peak corresponding to I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, with a particular retention time. The sample solution can then be injected into an HPLC column under the same conditions as the standard solution, and the resulting chromatogram can be studied to determine whether there is a peak having a retention time identical to that of the peak corresponding to I-1, I-2, I-3, I-4, I-5, I-6 or I-7 or a pharmaceutically acceptable salt thereof in the HPLC chromatogram of the standard solution. The presence of such peaks can indicate the presence of I-1, I-2, I-3, I-4, I-5, I-6 or I-7, or a pharmaceutically acceptable salt thereof, in the sample. In another embodiment, the amount of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in the sample solution can be determined by comparing the peak area in the first HPLC chromatogram to the peak area in the second HPLC chromatogram and thereby calculating the amount of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in the sample solution.

In some embodiments, the invention provides a method of determining an impurity in a material consisting essentially of X4P-001, wherein a sample solution containing the material plus a reference compound having a known chemical structure of I-1, I-2, I-3, I-4, I-5, I-6, or I-7 as described herein, or a pharmaceutically acceptable salt thereof, is injected into an HPLC column and an HPLC chromatogram is obtained to determine the presence and/or amount of the compound in the material.

The method of the invention may further comprise recording in written form the chemical identity of the compound and the amount of the compound in the material as an impurity.

In other embodiments, the invention provides methods of determining an impurity in a material consisting essentially of X4P-001, wherein a solution of the material dissolved therein is injected into an HPLC column and an HPLC chromatogram is obtained to determine the amount of a compound known to have the structure of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in the material as described herein. The chemical identity of the compound and the amount of the compound as an impurity in the material can then be recorded. The amount of compound in the material can be determined as follows: (i) identifying peaks on the chromatogram that correspond to peaks on a control chromatogram, (ii) identifying peaks on the chromatogram that correspond to relative retention times for compounds known to have the structure of formula I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof; and/or (iii) identifying a peak on the chromatogram corresponding to a known amount of a peak known to have the structure of formula I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof.

Some embodiments of the invention may be useful for determining the amount and/or presence of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in a sample comprising X4P-001. The sample may be a sample of a newly manufactured material, or the sample may be a sample stored over a given period of time. In one embodiment, a sample of X4P-001 can be stored and periodically analyzed using the methods described herein to determine the presence and/or amount of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in a sample that may have been formed by, for example, X4P-001 degradation. In some cases, the sample may be placed under stress conditions, i.e., conditions that are intended to promote degradation of X4P-001, such as elevated temperature and/or elevated humidity, wherein the sample is periodically analyzed using the methods described herein to determine the presence and/or amount of I-1, I-2, I-3, I-4, I-5, I-6, or I-7, or a pharmaceutically acceptable salt thereof, in the sample.

The compounds and compositions described herein are generally suitable for inhibiting CXCR4 or mutants thereof. Certain compounds and compositions described herein find use in treating, preventing, and/or reducing the risk of a disease, disorder, or condition associated with CXCR 4.

In one aspect, the invention provides a method of inhibiting the activity of CXCR4 or a mutant thereof in a patient comprising the step of administering to the patient the disclosed X4P-001 composition. In other embodiments, the present invention provides a method of treating a disorder mediated by CXCR4 or a mutant thereof in a patient in need thereof, the method comprising the step of administering to the patient a disclosed X4P-001 composition according to the present invention or a pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.

Certain methods of treating a disease or disorder with X4P-001 are described in PCT application PCT/US2018/038776 filed on 21/6/2018, the entire contents of which are incorporated herein by reference. The disclosed compounds and compositions are useful in such methods of treating diseases or disorders.

In certain embodiments, the composition comprising X4P-001 or a pharmaceutically acceptable salt thereof is administered orally in an amount from about 200mg to about 1200mg per day. In certain embodiments, the dosage composition may be provided in divided doses, separated by about 12 hours, twice daily. In other embodiments, the dosage composition may be provided once daily. The terminal half-life of X4P-001 is typically determined to be between about 12 and about 24 hours, or about 14.5 hours. The oral dosage may be from about 100mg to about 1200mg once or twice daily. In certain embodiments, the dose of X4P-0001, or a pharmaceutically acceptable salt thereof, suitable for use in the present invention is from about 200mg to about 800mg per day. In other embodiments, the dosage of X4P-001, or a pharmaceutically acceptable salt thereof, suitable for use in the invention may range from about 200mg to about 600mg, from about 400mg to about 800mg, from about 600mg to about 1000mg, or from about 800mg to about 1200mg per day.

In one aspect, the invention provides a method of treating cancer (e.g., the cancer described herein) by administering to a patient in need thereof an effective amount of a disclosed X4P-001 composition. In some embodiments, the methods comprise co-administering, simultaneously or sequentially, an effective amount of one or more other therapeutic agents, such as those described herein. In some embodiments, the method comprises co-administering one additional therapeutic agent. In some embodiments, the method comprises co-administering two additional therapeutic agents. In some embodiments, the combination of the disclosed compound or composition and one or more other therapeutic agents act synergistically to prevent or reduce immune evasion and/or angiogenic evasion of cancer. In some embodiments, the patient has previously been administered another anti-cancer agent, such as adjuvant therapy or immunotherapy. In some embodiments, the cancer is refractory.

In some embodiments, the disease, disorder or condition associated with CXCR4 is selected from the group consisting of a cell proliferative disorder, alzheimer's disease, HIV, rheumatoid arthritis or pulmonary fibrosis. In some embodiments, the disease, disorder, or condition is a hyperproliferative disorder, such as cancer. In some embodiments, the cancer is breast cancer, ovarian cancer, renal cancer, lung cancer, or melanoma. In some embodiments, the cancer is selected from Renal Cell Carcinoma (RCC), refractory RCC, or clear cell RCC (ccrcc).

In some embodiments, the invention provides methods for treating a patient having a cancer that is exhibited by a solid tumor. In some embodiments, the patient has renal cancer, renal tumor, renal cancer (including clear cell carcinoma and papillary renal cancer), ovarian cancer, or melanoma.

The compounds provided are CXCR4 inhibitors and are therefore useful for treating one or more conditions associated with CXCR4 activity. Thus, in certain embodiments, the present invention provides a method of treating a CXCR4 mediated disorder comprising the step of administering to a patient in need thereof a disclosed X4P-001 composition or a pharmaceutically acceptable composition thereof.

In one aspect, the invention provides a method of treating cancer in a patient in need thereof, wherein the method comprises administering to the patient the disclosed X4P-001 composition in combination with one or more other therapeutic agents (e.g., one or more immunostimulatory therapeutic compounds).

In some embodiments, the one or more immunostimulatory therapeutic compounds are agonists or activators selected from erlotinib (elotuzumab), mifamurtide (mifamurtide), toll-like (toll-like) receptors, or ROR γ t activators.

In some embodiments, the method further comprises administering to the patient a third therapeutic agent, e.g., an immune checkpoint inhibitor. In some embodiments, the method comprises administering to a patient in need thereof three therapeutic agents selected from the group consisting of the disclosed X4P-001 compositions, immunostimulatory therapeutic compounds, and immune checkpoint inhibitors.

In some embodiments, the immune checkpoint inhibitor is selected from nivolumab (nivolumab), periclizumab (pembrolizumab), ipilimumab (ipilimumab), avilimumab (avelumab), dolvacizumab (durvalumab), atelizumab (atezolizumab), or pidilizumab (pidilizumab).

In another aspect, the invention provides a method of treating cancer in a patient in need thereof, wherein the method comprises administering to the patient the disclosed X4P-001 composition in combination with one or more additional therapeutic agents selected from an indoleamine (2,3) -dioxygenase (IDO) inhibitor, a Poly ADP Ribose Polymerase (PARP) inhibitor, a Histone Deacetylase (HDAC) inhibitor, a CDK4/CDK6 inhibitor, or a phosphatidylinositol 3 kinase (PI3K) inhibitor.

In some embodiments, the IDO inhibitor is selected from the group consisting of Epapastat (epacadostat), indoimod (indoximod), cappatinib (capcanitib), GDC-0919, PF-06840003, BMS: F001287, Phy906/KD108, or an enzyme that cleaves kynurenine.

In some embodiments, the PARP inhibitor is selected from olaparib, such as carbopari (rucaparib) or nilapanib (niraparib).

In some embodiments, the HDAC inhibitor is selected from vorinostat (vorinostat), romidepsin (romidepsin), panobinostat (panobinostat), belinostat (belinostat), entinostat (entinostat), or chidamide (chidamide).

In some embodiments, the CDK 4/6 inhibitor is selected from pabociclib (palbociclib), rebocillin (ribociclib), abelmoscib (abemaciclib), or traciclib (trilicib).

In some embodiments, the method further comprises administering to the patient a third therapeutic agent, e.g., an immune checkpoint inhibitor. In some embodiments, the method comprises administering to a patient in need thereof three therapeutic agents selected from the group consisting of: the disclosed X4P-001 compositions; a second therapeutic agent selected from an indoleamine (2,3) -dioxygenase (IDO) inhibitor, a Poly ADP Ribose Polymerase (PARP) inhibitor, a Histone Deacetylase (HDAC) inhibitor, a CDK4/CDK6 inhibitor, or a phosphatidylinositol 3 kinase (PI3K) inhibitor; and a third therapeutic agent selected from an immune checkpoint inhibitor. In some embodiments, the immune checkpoint inhibitor is selected from nivolumab (nivolumab), periclizumab (pembrolizumab), ipilimumab (ipilimumab), avilimumab (avelumab), dolvacizumab (durvalumab), atelizumab (atezolizumab), or pidilizumab (pidilizumab).

In some embodiments, the PI3K inhibitor is selected from idenexib (idelalisib), epenib (alpelisib), tassel ib (taselisib), pickoxib (piculisib), cobicib (copanlisib), duvexib (duvelisib), PQR309, or TGR 1202.

In another aspect, the invention provides a method of treating cancer in a patient in need thereof, wherein the method comprises administering to the patient the disclosed X4P-001 composition in combination with one or more additional therapeutic agents selected from platinum-based therapeutic agents, taxanes, nucleoside inhibitors, or therapeutic agents that interfere with normal DNA synthesis, protein synthesis, cell replication, or otherwise inhibit rapid cell proliferation.

In some embodiments, the platinum-based therapeutic agent is selected from cisplatin (cissplatin), carboplatin (carboplatin), oxaliplatin (oxaliplatin), nedaplatin (nedaplatin), picoplatin (picoplatin), or satraplatin (satraplatin).

In some embodiments, the taxane is selected from paclitaxel (paclitaxel), docetaxel (docetaxel), albumin-bound paclitaxel, cabazitaxel, or SID 530.

In some embodiments, the therapeutic agent that interferes with normal DNA synthesis, protein synthesis, cell replication, or otherwise interferes with rapidly proliferating cell replication is selected from the group consisting of trabectedin, mechlorethamine, vincristine, temozolomide, cytarabine, lomustine, azacitidine, harringtonine, omacetuxine meperianine, Erwinia chrysoidea (Erwinia chrysophamide) asparaginase, eribulin mesylate, capecitabine, bendamustine, ipulone, ixabepilone, nelarabine, clorafacine, clomazone, trospidine, or trifluraline.

In some embodiments, the method further comprises administering to the patient a third therapeutic agent, e.g., an immune checkpoint inhibitor. In some embodiments, the method comprises administering to a patient in need thereof three therapeutic agents selected from the group consisting of: the disclosed X4P-001 compositions; a second therapeutic agent selected from a platinum-based therapeutic agent, a taxane, a nucleoside inhibitor, or a therapeutic agent that interferes with normal DNA synthesis, protein synthesis, cell replication, or otherwise inhibits rapid cell proliferation; and a third therapeutic agent selected from an immune checkpoint inhibitor.

In some embodiments, the immune checkpoint inhibitor is selected from nivolumab (nivolumab), periclizumab (pembrolizumab), ipilimumab (ipilimumab), avilimumab (avelumab), dolvacizumab (durvalumab), atelizumab (atezolizumab), or pidilizumab (pidilizumab).

In some embodiments, any of the above methods further comprises the step of obtaining a biological sample from the patient and measuring the amount of the disease-associated biomarker.

In some embodiments, the biological sample is a blood sample.

In some embodiments, the disease-associated biomarker is selected from circulating CD8+ T cells, or CD8+ T cells to Treg cell ratio.

In some embodiments, the cancer is selected from: hepatocellular carcinoma, ovarian cancer, ovarian epithelial cancer, or fallopian tube cancer; papillary serous cystadenocarcinoma or Uterine Papillary Serous Carcinoma (UPSC); prostate cancer; testicular cancer; gallbladder cancer; hepatobiliary cancer; soft tissue and bone synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; ewing sarcoma (Ewing sarcoma); undifferentiated thyroid cancer; adrenocortical adenoma; pancreatic cancer; ductal pancreatic cancer or pancreatic cancer; gastrointestinal/Gastric (GIST) cancer; lymphoma; squamous Cell Carcinoma of Head and Neck (SCCHN); salivary gland cancer; glioma or brain cancer; neurofibroma-1 associated Malignant Peripheral Nerve Sheath Tumor (MPNST); waldenstrom's macroglobulinemia (Waldenstrom); or neural tube blastoma.

In some embodiments, the cancer is selected from: hepatocellular carcinoma (HCC), hepatoblastoma, colon carcinoma, rectal carcinoma, ovarian epithelial carcinoma, fallopian tube carcinoma, papillary serous cystadenocarcinoma, papillary serous carcinoma of the Uterus (UPSC), hepatobiliary carcinoma, soft tissue and synovial sarcoma, rhabdomyosarcoma, osteosarcoma, undifferentiated thyroid carcinoma, adrenocortical adenoma, pancreatic carcinoma, pancreatic ductal carcinoma, pancreatic carcinoma, glioma, neurofibroma-1 associated Malignant Peripheral Nerve Sheath Tumor (MPNST), waldenstrom's macroglobulinemia, or medulloblastoma.

In some embodiments, the present invention provides a method of treating a cancer exhibiting a solid tumor, such as a sarcoma, carcinoma, or lymphoma, comprising the step of administering to a patient in need thereof a disclosed X4P-001 composition. Solid tumors often contain abnormal tissue masses that typically do not include cysts or fluid regions. In some embodiments, the cancer is selected from renal cell carcinoma or renal cancer; hepatocellular carcinoma (HCC) or hepatoblastoma, or liver cancer; melanoma; breast cancer; colorectal carcinoma, or colorectal carcinoma; colon cancer; rectal cancer; anal cancer; lung cancer, such as non-small cell lung cancer (NSCLC) or Small Cell Lung Cancer (SCLC); ovarian cancer, ovarian epithelial cancer, ovarian carcinoma or fallopian tube cancer; papillary serosa adenocarcinoma or Uterine Papillary Serous Carcinoma (UPSC); prostate cancer; testicular cancer; gallbladder cancer; hepatobiliary cancer; soft tissue and bone synovial sarcoma; rhabdomyosarcoma; osteosarcoma; chondrosarcoma; ewing's sarcoma; undifferentiated thyroid cancer; adrenocortical carcinoma; pancreatic cancer; ductal pancreatic cancer or pancreatic cancer; gastrointestinal/Gastric (GIST) cancer; lymphoma; squamous Cell Carcinoma of Head and Neck (SCCHN); salivary gland cancer; glioma or brain cancer; neurofibroma-1 associated Malignant Peripheral Nerve Sheath Tumor (MPNST); waldenstrom's macroglobulinemia; or neural tube blastoma.

In some embodiments, the cancer is selected from renal cell carcinoma, hepatocellular carcinoma (HCC), hepatoblastoma, colorectal carcinoma, colon carcinoma, rectal carcinoma, anal carcinoma, ovarian carcinoma, fallopian tube carcinoma, papillary serosa adenocarcinoma, papillary serous carcinoma of the Uterus (UPSC), hepatobiliary carcinoma, soft tissue and synovium sarcoma, rhabdomyosarcoma, osteosarcoma, chondrosarcoma, undifferentiated thyroid carcinoma, adrenocortical carcinoma, pancreatic ductal carcinoma, pancreatic carcinoma, glioma, brain carcinoma, neurofibroma-1-associated Malignant Peripheral Nerve Sheath Tumor (MPNST), waldenstrom's macroglobulinemia, or medulloblastoma.

In some embodiments, the cancer is selected from hepatocellular carcinoma (HCC), hepatoblastoma, colon cancer, rectal cancer, ovarian epithelial cancer, ovarian cancer, fallopian tube cancer, papillary serosa adenocarcinoma, papillary serosa carcinoma of the Uterus (UPSC), hepatobiliary carcinoma, soft tissue and synovial sarcoma of the bone, rhabdomyosarcoma, osteosarcoma, undifferentiated thyroid cancer, adrenocortical carcinoma, pancreatic cancer, pancreatic ductal carcinoma, pancreatic cancer, glioma, neurofibroma-1 associated Malignant Peripheral Nerve Sheath Tumor (MPNST), waldenstrom's macroglobulinemia, or neuroblastoma.

In some embodiments, the cancer is hepatocellular carcinoma (HCC). In some embodiments, the cancer is hepatoblastoma. In some embodiments, the cancer is colon cancer. In some embodiments, the cancer is colorectal cancer. In some embodiments, the cancer is ovarian cancer or an ovarian carcinoma. In some embodiments, the cancer is ovarian epithelial cancer. In some embodiments, the cancer is fallopian tube cancer. In some embodiments, the cancer is papillary serosal cystadenocarcinoma. In some embodiments, the cancer is Uterine Papillary Serous Carcinoma (UPSC). In some embodiments, the cancer is hepatobiliary cancer. In some embodiments, the cancer is soft tissue and bone synovial sarcoma. In some embodiments, the cancer is rhabdomyosarcoma. In some embodiments, the cancer is osteosarcoma. In some embodiments, the cancer is undifferentiated thyroid cancer. In some embodiments, the cancer is adrenocortical carcinoma. In some embodiments, the cancer is pancreatic cancer or pancreatic ductal cancer. In some embodiments, the cancer is pancreatic cancer. In some embodiments, the cancer is glioma. In some embodiments, the cancer is Malignant Peripheral Nerve Sheath Tumor (MPNST). In some embodiments, the cancer is neurofibroma-1 associated MPNST. In some embodiments, the cancer is waldenstrom's macroglobulinemia. In some embodiments, the cancer is a medulloblastoma.

In some embodiments, the present invention provides a method of treating a cancer selected from leukemia or hematologic cancer, comprising administering to a patient in need thereof an effective amount of a disclosed X4P-001 composition, optionally in combination with another therapeutic agent (such as those described herein). In some embodiments, the cancer is selected from Acute Myeloid Leukemia (AML), Chronic Myeloid Leukemia (CML), Acute Lymphocytic Leukemia (ALL), Chronic Lymphocytic Leukemia (CLL), or virus-induced leukemia.

In some embodiments, the patient has a resectable solid tumor, meaning that the patient's tumor is considered surgically removable. In other embodiments, the patient has a solid tumor that is unresectable, meaning that the patient's tumor has been deemed to be less susceptible to surgical removal, either in whole or in part.

In some embodiments, the cancer is an advanced cancer, e.g., advanced renal cancer or advanced renal cell carcinoma.

In some embodiments, the present invention provides a method of treating refractory cancer in a patient in need thereof, comprising administering to a patient in need thereof an effective amount of a disclosed X4P-001 composition or a pharmaceutical composition thereof, optionally in combination with another therapeutic agent (such as those described herein).

In certain embodiments, the patient has been previously administered a protein kinase inhibitor. In some embodiments, the patient has been previously administered a VEGF-R antagonist. In certain embodiments, the patient has been previously administered an immune checkpoint inhibitor. In some embodiments, the patient has previously been administered an immune checkpoint inhibitor selected from the group consisting of: nivolumab (a), (b) and (c)Baishimei noble (Bristol-Myers Squ)ibb)), palivizumab (Merck (Merck)), or ipiru (Ipiromab)Baishimei noble).

In some embodiments, the disclosed X4P-001 composition is administered to a patient in a fasting state.

Cell proliferative disorders

The invention features methods and compositions for the diagnosis and prognosis of cell proliferative disorders (e.g., cancer) and the treatment of these disorders by targeting CXCR 4. Cell proliferative disorders described herein include, for example, cancer, obesity, and proliferation-dependent diseases. Such disorders can be diagnosed using methods known in the art.

Cancer treatment

In some embodiments, the cancer includes, but is not limited to, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocytic leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphomas (e.g., Hodgkin's disease or non-Hodgkin's disease), waldenstrom's macroglobulinemia, multiple myeloma, heavy chain diseases, and solid tumors, e.g., sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphoendotheliosarcoma, multiple myeloma, and solid tumors such as sarcoma, Synovioma, mesothelioma, ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon cancer, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, bronchial cancer, renal cell carcinoma, hepatoma, bile duct cancer, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, uterine cancer, testicular cancer, lung cancer, small cell lung cancer, bladder cancer, epithelial cancer, glioma, astrocytoma, undifferentiated glioblastoma (GBM, also known as glioblastoma), neuroblastoma, craniopharyngioblastoma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, meningioma, melanoma, neuroblastoma, cervical carcinoma, bladder carcinoma, neuroblastoma and retinoblastoma).

In some embodiments, the cancer is glioma, astrocytoma, undifferentiated glioblastoma (GBM, also known as glioblastoma), neural tube blastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, schwannoma, neurofibrosarcoma, meningioma, melanoma, neuroblastoma, or retinoblastoma.

In some embodiments, the cancer is an acoustic neuroma, an astrocytoma (e.g., grade I-hairy cell astrocytoma, grade II-low malignancy astrocytoma, grade III-undifferentiated astrocytoma, or grade IV-Glioblastoma (GBM)), chordoma, CNS lymphoma, craniopharyngioma, brain stem glioma, ependymoma, mixed glioma, optic nerve glioma, subintimal tumor, neural tube blastoma, meningioma, metastatic brain tumor, oligodendroglioma, pituitary tumor, Primitive Neuroectodermal (PNET) tumor, or schwannoma. In some embodiments, the cancer is a type more commonly found in children than in adults, such as brain stem glioma, craniopharyngioma, ependymoma, juvenile hairy cell astrocytoma (JPA), medulloblastoma, optic nerve glioma, pineal body tumor, primitive neuroectodermal tumor (PNET), or rhabdoid tumor. In some embodiments, the patient is an adult. In some embodiments, the patient is a pediatric or pediatric patient.

In another embodiment, the cancer includes (but is not limited to): mesothelioma, hepatobiliary (liver and bile duct) cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular melanoma, ovarian cancer, colon cancer, rectal cancer, cancer of the anal region, stomach cancer, gastrointestinal (stomach, colorectal and duodenum) cancer, uterine cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, hodgkin's disease, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, cancer of the prostate, cancer of the testis, chronic or acute leukemia, chronic myelogenous leukemia, lymphobulbar lymphoma, cancer of the bladder, cancer of the kidney or ureter, renal cell carcinoma, carcinoma of the renal pelvis, non-hodgkin's lymphoma, spinal axis tumor, brain stem glioma, pituitary adenoma, cancer of the adrenal cortex, cancer of the gallbladder, multiple myeloma, cancer of, Fibrosarcoma, neuroblastoma, retinoblastoma, or a combination of one or more of the foregoing cancers.

Other features of the invention are methods and compositions for the diagnosis, prognosis and treatment of virus-associated cancers, including Human Immunodeficiency Virus (HIV) -associated solid tumors, Human Papilloma Virus (HPV) -16 positive incurable solid tumors and adult T-cell leukemia caused by human T-cell leukemia type I (HTLV-I), which is a highly invasive form of CD4+ T-cell leukemia with HTLV-I clone integrated into the leukemia cells (see https:// clinical trials. gov/ct2/show/study/NCT 02631746); and virus-associated tumors in gastric cancer, nasopharyngeal cancer, cervical cancer, vaginal cancer, vulvar cancer, head and neck squamous cell carcinoma and Merkel cell carcinoma (Merkel cell carcinoma). (see https:// clinical trials. gov/ct2/show/study/NCT 02488759; also see https:// clinical trials. gov/ct2/show/study/NCT 0240886; https:// clinical trials. gov/ct2/show/NCT02426892)

In some embodiments, the present invention provides a method of treating a tumor in a patient in need thereof, comprising administering to the patient the disclosed X4P-001 composition. In some embodiments, the tumor comprises any one of the cancers described herein. In some embodiments, the tumor comprises melanoma cancer. In some embodiments, the tumor comprises breast cancer. In some embodiments, the tumor comprises lung cancer. In some embodiments, the tumor comprises Small Cell Lung Cancer (SCLC). In some embodiments, the tumor comprises non-small cell lung cancer (NSCLC).

In some embodiments, the tumor is treated by arresting further growth of the tumor. In some embodiments, the tumor is treated by reducing the size (e.g., volume or mass) of the tumor by at least 5%, 10%, 25%, 50%, 75%, 90%, or 99% relative to the size of the tumor before treatment. In some embodiments, the tumor is treated by reducing the number of tumors in the patient by at least 5%, 10%, 25%, 50%, 75%, 90%, or 99% relative to the number of tumors prior to treatment.

Primary immunodeficiency

In some embodiments, the present invention provides a method of treating one or more disorders, diseases, and/or conditions, wherein the disorder, disease, or condition includes (but is not limited to) a primary immunodeficiency disease or disorder, comprising administering to a patient in need thereof an effective amount of a disclosed X4P-001 composition. The primary immunodeficiency that can be treated by the methods of the invention include: warts, hypogammaglobulinemia, infection, myeloagranulocytosis (WHIM) syndrome; severe Congenital Neutropenia (SCN), particularly those caused by G6PC3 deficiency (McDermott et al (2010) Blood 116: 2793-2802); GATA2 deficiency (single MAC Syndrome) (machijweski-Duval et al (2015) journal of leukocyte biology (j.leukoc.biol.). 5MA0815-288R (e-press prior to printing), idiopathic CD4+ T lymphopenia (ICL), and Wiskott-Aldrich Syndrome (Wiskott-Aldrich Syndrome).

In other embodiments, the invention relates to a method of inhibiting CXCR4 activity in a biological sample comprising the step of contacting the biological sample with the disclosed X4P-001 composition.

According to another embodiment, the invention relates to a method of inhibiting the activity of CXCR4 or a mutant thereof in a biological sample comprising the step of contacting the biological sample with the disclosed X4P-001 composition. In other embodiments, the invention relates to a method of inhibiting the activity of CXCR4 or a mutant thereof in a biological sample comprising the step of contacting the biological sample with the disclosed X4P-001 composition.

As used herein, the term "biological sample" includes, but is not limited to, cell cultures or extracts thereof; a biopsy material obtained from a mammal or an extract thereof; and blood, saliva, urine, feces, semen, tears, or other bodily fluids or extracts thereof.

Co-administration with other therapeutic agents

In one aspect, the invention provides a method of treating cancer (e.g., the cancer described herein) by administering to a patient in need thereof the disclosed X4P-001 composition. In some embodiments, the methods comprise co-administering, simultaneously or sequentially, an effective amount of one or more other therapeutic agents, such as those described herein. In some embodiments, the method comprises co-administering one additional therapeutic agent. In some embodiments, the method comprises co-administering two additional therapeutic agents. In some embodiments, the disclosed combination of X4P-001 composition and one or more other therapeutic agents act synergistically to prevent or reduce immune evasion and/or angiogenic evasion of cancer. In some embodiments, the patient has previously been administered another anti-cancer agent, such as adjuvant therapy or immunotherapy. In some embodiments, the cancer is refractory.

Certain methods of treating a disease or disorder by co-administering X4P-001 with one or more other agents are described in PCT application No. PCT/US2018/038776, filed on 21.6.2018, the entire contents of which are incorporated herein by reference.

Depending on the particular condition or disease to be treated, other therapeutic agents normally administered for the treatment of the condition may also be present in the compositions of the invention. As used herein, other therapeutic agents normally administered for the treatment of a particular disease or condition are referred to as "appropriate for the disease or condition being treated.

In some embodiments, the additional therapeutic agent is a kinase inhibitor or a VEGF-R antagonist. Approved VEGF and kinase inhibitors suitable for use in the present invention include: bevacizumab (bevacizumab) (Bevacizumab)Gene tag/Roche (Genentech/Roche)), an anti-VEGF monoclonal antibody; ramucirumab (ramucirumab) ((R))Lilay (Eli Lilly)), an anti-VEGFR-2 antibody; and ziv-aflibercept, also known as VEGF trap (r) ((r))Regenerant/cenofil (Regeneron/Sanofi)); VEGFR inhibitors, e.g. regorafenib (R.) (Bayer (Bayer)); vandetanib (vandetanib) ((vandetanib))Astrikon (AstraZeneca)); axitinib (axitinib) ((a))Pfizer); and lenvatinib (Levatinib) (II)Sanitary materials (Eisai)); raf inhibitors, e.g. sorafenib (sorafenib) ((R))Bayer and ornix (Onyx)); dabrafenib (dabrafenib) ((b))Novartis (Novartis)); and vemurafenib (vemurafenib) ((v))Gene tack/roche); MEK inhibitors, e.g. cobimetinib (cobimetinib) (II)Eloxis (exelixis)/gene tag/roche); trametinib (trametinib) ((R))Noval); Bcr-Abl tyrosine kinase inhibitors, e.g. imatinib (imatinib) (I) or (II) and (III)Noval); nilotinib (nilotinib) ((r))Noval); dasatinib (dasatinib) (Dasatinib)Baishimei noble); bosutinib (bosutinib) (B)Pfeiri); and ponatinib (ponatinib) ((b))Aryad Pharmaceuticals); her2 and EGFR inhibitors, e.g. gefitinib (gefitinib) ((R))Aslicon Corp.); erlotinib (erlotinib) ((iii))Gene tag/roche/anstela (Astellas)); lapatinib (lapatinib) (la)Noval); afatinib (afatinib) (afatinib)Boringer Invitrogen (Boehringer Ingelheim)); oxitinib (osimertinib) (targeting activated EGFR,aslicon Corp.); and brigatinib (brigatinib) (ii)Arriyad pharmaceuticals); c-Met and VEGFR2 inhibitors, e.g. cabozantinib (cabozanib) ((R))Alexis (exellenis)); and multi-kinase inhibitors, such as sunitinib (sunitinib) ((b))Pfeiri); pazopanib (pazopanib) ((pazopanib))Noval); ALK inhibitors, e.g. crizotinib (CRIZOTINib)Pfeiri); ceritinib (ceritinib) ((ii))Noval); and Alletinib (alectinib) ((II))Gene tack/roche); bruton's tyrosine kinase inhibitor, such as ibrutinib (ibrutinib) ((R))Farmsley/Ponsen (pharmaceuticals/Janssen)); and Flt3 receptor inhibitors, such as midostaurin (midostaurin) ((Nova).

Other kinase inhibitors and VEGF-R antagonists under development and that may be used in the present inventionAnti-agents include tivozanib (tivozanib) (avio pharmaceuticals); vantalanib (vatalanib) (Bayer/Nowa); lucitant (lucitanib) (Clovis tumor (Clovis Oncology)); dovirtinib (TKI258, nova); chavanib (chiaanib) (Chipscreen Biosciences); CEP-11981 (Cefalron (Cephalon)); raney nib (liniforib) (Abbott Laboratories); neratinib (HKI-272, buma Biotechnology (Puma Biotechnology)); ladostinib (radotinib)IY5511, korean Il-Yang Pharmaceuticals (Il-Yang Pharmaceuticals, s.korea)); luketinib (ruxolitinib) ((R))Incyte Corporation); PTC299(PTC Therapeutics); CP-547,632 (pyroxene); fratinib (foretinib) (elousis (exellenis), glatiramer smith (GlaxoSmithKline)); quinazatinib (quizartinib) (first san kyo (Daiichi)) and motesanib (Amgen/Takeda)).

In some embodiments, the other therapeutic agent is an mTOR inhibitor that inhibits cell proliferation, angiogenesis, and glucose uptake. Approved mTOR inhibitors suitable for use in the present invention include everolimus (everolimus) ((r))Noval); temsirolimus (temsirolimus) ((temsirolimus))Pfeiri); and sirolimus (sirolimus) ((ii))Pfeiri).

In some embodiments, the other therapeutic agent is a Poly ADP Ribose Polymerase (PARP) inhibitor. Is suitable for the invention

Approved PARP inhibitionThe agent comprises olaparib (olaparib)Aslicon Corp.); such as kappa rui (rucaparib)Cloviss tumor corporation); and nilapanib (niraparib) (ii)Tyxano (Tesaro)). Other PARP inhibitors under investigation that may be used in the present invention include tarazoparib (MDV3800/BMN 673/LT00673, madyvist/pyrosory/bemarlin (Medivation/Pfizer/Biomarin); veliparib (ABT-888, albervie), and BGB-290 (BeiGene Inc.).

In some embodiments, the other therapeutic agent is a phosphatidylinositol 3 kinase (PI3K) inhibitor. Approved PI3K inhibitors suitable for use in the present invention include idecoxib (idelalisib) ((ii))Gillided (Gilead)). Other PI3K inhibitors under investigation that may be used in the present invention include especib (alpelisib) (BYL719, noval); tasselisib (GDC-0032, gene tack/roche); picrorxib (picclisib) (GDC-0941, gene tag/roche); cobicisib (copanlisib) (BAY806946, bayer); duvexib (formerly IPI-145, infinite Pharmaceuticals (Infinity)); PQR309(Piqur Therapeutics, switzerland); and TGR1202 (previously RP5230, TG Therapeutics).

In some embodiments, the additional therapeutic agent is a proteasome inhibitor. Approved proteasome inhibitors suitable for use in the present invention include bortezomib (bortezomib) ((b))Martial arts (Takeda)); carfilzomib (carfilzomib) (ii) aInstallation (Amgen)); and epsomidine (a)Wutian).

In some embodiments, the other therapeutic agent is a Histone Deacetylase (HDAC) inhibitor. Approved HDAC inhibitors suitable for use in the present invention include vorinostat (vorinostat) ((r))Merck); romidepsin (romidepsin) ((R))The Seal gene (Celgene)); panobinostat (panobinostat) (panobinostat)Noval); and belinostat (belinostat) (b)Spectral Pharmaceuticals (spectra Pharmaceuticals)). Other HDAC inhibitors under investigation that may be used in the present invention include entinostat (entinostat) (SNDX-275, Syndax Pharmaceuticals) (NCT 00866333); and chidamide (chidamide) (II)HBI-8000, Chinese Kipp Bioscience (Chipscreen Biosciences, China)).

In some embodiments, the other therapeutic agent is a CDK inhibitor, e.g., a CDK 4/6 inhibitor. Approved inhibitors of CDK 4/6 suitable for use in the present invention include Pabociclib (palbociclib)Pfeiri); and ribociclib (ribociclib) ((R))Nova). Can be used in the present inventionOther CDK 4/6 inhibitors of (i) include abbelib (abemacilib) (Ly2835219, lilac (Eli Lilly)); and traasiril (trilacticb) (G1T28, G1 Therapeutics).

In some embodiments, the additional therapeutic agent is an indoleamine (2,3) -dioxygenase (IDO) inhibitor. Among the IDO inhibitors being investigated that may be used in the present invention include epaptastat (INCB024360, Incyte); indolmod (indoximod) (NLG-8189, NewLink Genetics Corporation); capetinib (capmanitib) (INC280, nova); GDC-0919 (Gene Tack/Roche); PF-06840003 (feverfew); BMS, F001287 (Baishimei noble); phy906/KD108 (Hewlett-packard pharmaceutical (phytoeutica); and Kyn Therapeutics (Kyn Therapeutics) enzymes that decompose kynurenine.

In some embodiments, the additional therapeutic agent is a growth factor antagonist, such as an antagonist of Platelet Derived Growth Factor (PDGF) or Epidermal Growth Factor (EGF) or its receptor (EGFR). Approved PDGF antagonists that can be used in the present invention include olaratumabA gift company). Approved EGFR antagonists that may be used in the present invention include cetuximab (cetuximab), (b) and (c) and (dLilai corporation); lyricitumumab (necitumumab) ((b))Lilai corporation); panitumumab (panitumumab)Mounting); and oxitinib (osimertinib) (targeting activated EGFR,aslikang Corp.).

In some embodiments, the additional therapeutic agent is an aromatase inhibitor. Approved for use in the present inventionAromatase inhibitors include exemestane (A)Pfeiri); anastrozole (a)Aslicon Co.) and letrozole (letrozole) ((R)Nova).

In some embodiments, the additional therapeutic agent is a hedgehog pathway antagonist. Approved hedgehog pathway inhibitors useful in the present invention include sonidegib (sonidegib) ((iii))Sun Pharmaceuticals (Sun Pharmaceuticals)); and vismodegib (vismodegib) (vi)Gene tacg (Genentech)), both of which are used to treat basal cell carcinoma.

In some embodiments, the additional therapeutic agent is a folate inhibitor. Approved folate inhibitors suitable for use in the present invention include pemetrexed (pemetrexed) ((R))A gift company).

In some embodiments, the additional therapeutic agent is a CC chemokine receptor 4(CCR4) inhibitor. CCR4 inhibitors suitable for use in the present study include moglicazumab (mogamulizumab) (M) in the present studyJapan fermented kylin (Kyowa Hakko Kirin, Japan)).

In some embodiments, the other therapeutic agent is an Isocitrate Dehydrogenase (IDH) inhibitor. IDH inhibitors under investigation that may be used in the present invention include AG120 (Seal gene; NCT 02677922); AG221 (Sell Gene, NCT 02677922; NCT 02577406); BAY1436032 (bayer, NCT 02746081); IDH305 (Nowa, NCT 02987010).

In some embodiments, the additional therapeutic agent is an arginase inhibitor. Arginase inhibitors under investigation that may be used in the present invention include AEB1102 (pegylated recombinant arginase, agelia Biotherapeutics), which is being studied in phase 1 clinical trials for acute myeloid leukemia and myelodysplastic syndrome (NCT02732184) and solid tumors (NCT 02561234); and CB-1158 (Calithera Biosciences).

In some embodiments, the other therapeutic agent is a glutaminase inhibitor. Glutaminase inhibitors under investigation which may be used in the present invention include CB-839 (Calif. biosciences).

In some embodiments, the additional therapeutic agent is an antibody that binds to a tumor antigen (i.e., a protein expressed on the cell surface of a tumor cell). Approved antibodies that bind to tumor antigens that can be used in the present invention include rituximab (rituximab) (R) rituximabGene tex/baijiandi (BiogenIdec)); ofatumumab (anti-CD 20,glassware SmithKline (GlaxoSmithKline)); obinutuzumab (obinutuzumab) (anti-CD 20,gene tack); ibbelimumab (anti-CD 20 and yttrium-90,spectral pharmaceutical); dachwitumumab (daratumumab) (anti-CD 38,poplarn biotechnology (Janssen Biotech)); dinu ituximab (anti-glycolipid GD2,combined Therapeutics); trastuzumab (trastuzumab) (anti-HER 2,gene tack); ado-trastuzumab emtansine (anti-HER 2), fused with emtansine (emtansine),gene tack); and pertuzumab (pertuzumab) (anti-HER 2,gene tack); and brentuximab vedottin (brentuximab vedotin) (anti-CD 30-drug conjugate,seattle genes (Seattle Genetics)).

In some embodiments, the additional therapeutic agent is a topoisomerase inhibitor. Approved topoisomerase inhibitors suitable for use in the present invention include irinotecan (irinotecan) ((R))Merrima Pharmaceuticals (Merrimack Pharmaceuticals)); topotecan (topotecan) ((B))Glatiramer). Topoisomerase inhibitors under investigation which may be used in the present invention include pyroxantrone (pixantrone), (xxix), (xxCTI biopharmaceuticals (CTI Biopharma)).

In some embodiments, the other therapeutic agent is a nucleoside inhibitor, or other therapeutic agent that interferes with normal DNA synthesis, protein synthesis, cell replication, or otherwise inhibits rapid cell proliferation. Such coresGlycoside inhibitors or other therapeutic agents include trabectedin (guanidine alkylating agent,popsen anticancer drugs (Janssen Oncology), nitrogen mustards (mechlorothiamine) (alkylating agents,aktelion Pharmaceuticals (Aktelion Pharmaceuticals)); vincristine (A)A salsa company;taihua Pharmaceuticals (Teva Pharmaceuticals);talon pharmaceuticals (Talon Therapeutics)); temozolomide (temozolomide) (prodrug of alkylating agent 5- (3-methyltriazol-1-yl) -imidazole-4-carboxamide (MTIC),merck); cytarabine injections (ara-C, antimetabolite cytidine analog, feverfew); lomustine (lomustine) (alkylating agent,baishimei noble treasure;nanogold Biotechnology (NextSource Biotechnology)); azacitidine (a pyrimidine nucleoside analog of cytidine),seier gene); homoharringtonine (cephalotaxine ester) (protein synthesis inhibitor,taihua medicine preparation); erwinia chrysanthemi asparaginase (an enzyme used to deplete asparagine,Lundbeck;EUSA Pharma); eribulin mesylate (microtubule inhibitor, tubulin-based antimitotic drug,sanitary materials (Eisai)); cabazitaxel (cabazitaxel) (microtubule inhibitors, tubulin-based antimitotic drugs,Xenof-Anthrate corporation (Sanofi-Aventis)); caprenine (a thymidylate synthase inhibitor,gene tack); bendamustine (a bifunctional nitrogen mustard derivative, believed to form interchain DNA crosslinks,cyfadron/tahua (Cephalon/Teva)); semi-synthetic analogs of ixabepilone (epothilone B), microtubule inhibitors, tubulin-based antimitotic drugs,baishimei noble); nelarabine (nelarabine), a prodrug of a deoxyguanosine analog, an inhibitor of nucleoside metabolism,noval); clorfabine (clorafabine), a prodrug of ribonucleotide reductase inhibitor, a competitive inhibitor of deoxycytidine,cenofil-amplat corporation); and trifluridine (trifluridine) and tipiracil (tipiracil) (thymidine based nucleoside analogues and thymidine phosphorylase inhibitors,roc anticancer drugs (Taiho Oncology)).

In some embodiments, the other therapeutic agent is a platinum-based therapeutic agent, also referred to as platinum. Platinum causes DNA cross-linking, and thus it primarily inhibits DNA repair and/or DNA synthesis in rapidly regenerating cells (e.g., cancer cells). Approved platinum-based therapeutics that may be used in the present invention include cisplatin (cissplatin) ((R))Baishimei noble); carboplatin (carboplatin)Baishimei noble treasure; furthermore, taihua; pfeiri); oxaliplatin (oxaliplatin) ((oxaliplatin))Cenofil-amplat corporation); and nedaplatin (nedaplatin) (B)Shionogi, Inc.). Other platinum-based therapeutic agents that have undergone clinical testing and may be used in the present invention include picoplatin (picoplatin) (panier Pharmaceuticals (ponirad Pharmaceuticals)); and satraplatin (JM-216, agiline (Agennix)).

In some embodiments, the additional therapeutic agent is a taxane compound that causes microtubules to rupture, which is necessary for cell division. Approved taxane compounds that may be used in the present invention include paclitaxel (paclitaxelBaishimei noble powder), docetaxelXenoffy-Antont corporation;solar pharmaceutical), albumin-bound paclitaxel (Abolix/Sele genes (Abraxis/Celgene)), and cabazitaxel (cabazitaxel) ((Abraxis/Celgene))Cenofil-amplat). Other taxane compounds that have been clinically tested and may be used in the present invention include SID530(SK chemical) (NCT 00931008).

In some embodiments, the other therapeutic agent is an anti-apoptotic protein inhibitor, such as BCL-2. Approved anti-apoptotic agents that may be used in the present invention include Venetosala (venetocalax) (A), (B), (CEbervian/gene tek (AbbVie/Genentech)); and bluntmomab (blinatumomab) ((blnatumomab))Ann in (Amgen)). Other therapeutic agents that have been clinically tested and that can be used in the present invention to target apoptotic proteins include nevira (navitoclax) (ABT-263, Abbott (Abbott)), BCL-2 inhibitors (NCT 02079740).

As used herein, the term "checkpoint inhibitor" relates to an agent suitable for preventing cancer cells from evading the immune system of a patient. One of the major mechanisms of anti-tumor immune disruption is known as "T cell depletion", which results from chronic exposure to antigens that have caused upregulation of inhibitory receptors. These inhibitory receptors act as immune checkpoints to prevent uncontrolled immune responses.

PD-1 and co-inhibitory receptors, such as cytotoxic T lymphocyte antigen 4(CTLA-4, B and T lymphocyte attenuator (BTLA; CD272), T cell immunoglobulin and mucin domain-3 (Tim-3), lymphocyte activation gene-3 (Lag-3; CD223), etc., are often referred to as checkpoint regulators, which act as molecular "gatekeepers" that determine from extracellular information whether cell cycle progression and other intracellular signaling processes should occur.

In one aspect, the checkpoint inhibitor is a biologic therapeutic or a small molecule. In another aspect, the checkpoint inhibitor is a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein, or a combination thereof. In another aspect, the checkpoint inhibitor inhibits a checkpoint protein selected from the group consisting of: CTLA-4, PDLl, PDL2, PDl, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or combinations thereof. In another aspect, the checkpoint inhibitor interacts with a ligand of a checkpoint protein selected from the group consisting of: CTLA-4, PDLl, PDL2, PDl, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or combinations thereof. In one aspect, the checkpoint inhibitor is an immunostimulant, a T cell growth factor, an interleukin, an antibody, a vaccine, or a combination thereof. In another aspect, the interleukin is IL-7 or IL-15. In a particular aspect, the interleukin is glycosylated IL-7. In another aspect, the vaccine is a Dendritic Cell (DC) vaccine.

Checkpoint inhibitors include any agent that blocks or inhibits the inhibitory pathway of the immune system in a statistically significant manner. Such inhibitors may include small molecule inhibitors or may include antibodies or antigen-binding fragments thereof that bind to and block or inhibit an immune checkpoint receptor or antibodies that bind to and block or inhibit an immune checkpoint receptor ligand. Illustrative checkpoint molecules that can be targeted for blocking or inhibition include, but are not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, GAL9, LAG3, TIM3, VISTA, KIR, 2B4 (belonging to the family of CD2 molecules and across all NK, γ δ and memory CD 8)+Expressed on (. alpha.beta.) T cells), CD160 (also known as BY55), CGEN-15049, CHK 1 and CHK2 agonistsEnzymes, A2aR and various B-7 family ligands. B7 family ligands include, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6, and B7-H7. Checkpoint inhibitors include antibodies or antigen-binding fragments thereof, other binding proteins, biotherapeutics, or small molecules that bind to and block or inhibit the activity of one or more of: CTLA-4, PDL1, PDL2, PD1, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, and CGEN-15049. Illustrative immune checkpoint inhibitors include Tremelimumab (Tremelimumab) (CTLA-4 blocking antibody), anti-OX 40, PD-Ll monoclonal antibody (anti-B7-Hl; MEDI4736), MK-3475(PD-1 blocking agent), nivolumab (R-X-Y-H-BMS-936558; anti-PDl antibody), CT-011 (anti-PDl antibody), BY55 monoclonal antibody, AMP224 (anti-PDLl antibody), BMS-936559 (anti-PDLl antibody), MPLDL3280A (anti-PDLl antibody), MSB0010718C (anti-PDLl antibody), and ipilimumab (anti-CTLA-4 checkpoint inhibitor). Checkpoint protein ligands include, but are not limited to: PD-Ll, PD-L2, B7-H3, B7-H4, CD28, CD86 and TIM-3.

In certain embodiments, the immune checkpoint inhibitor is selected from the group consisting of a PD-1 antagonist, a PD-L1 antagonist, and a CTLA-4 antagonist. In some embodiments, a CXCR4 antagonist (e.g., X4P-001 or a pharmaceutically acceptable salt thereof) is administered in combination with: nivolumab (anti-PD-1 antibody,baishimei noble); palivizumab (anti-PD-1 antibody,merck); ipilimumab (anti-CTLA-4 antibody,baishimei noble); dolvacizumab (durvalumab) (anti-PD-L1 antibody,a SiLikang Corp.); or atelizumab (atezolizumab) (anti-PD-L1 antibody,gene tack).

In some embodiments, other immune checkpoint inhibitors suitable for use in the present invention include REGN2810 (regenerant), an anti-PD-1 antibody that has been tested in patients with basal cell carcinoma (NCT03132636), NSCLC (NCT03088540), cutaneous squamous cell carcinoma (NCT02760498), lymphoma (NCT02651662), and melanoma (NCT 03002376); pidilizumab (pidilizumab), also known as CT-011, an antibody that binds to PD-1 in clinical trials against diffuse large B-cell lymphoma and multiple myeloma; abameluumab (A)Pfizer/Merck KGaA), also known as MSB0010718C, a fully human IgG1 anti-PD-L1 antibody in clinical trials for non-small cell lung cancer, Merkel cell carcinoma, mesothelioma, solid tumors, kidney cancer, ovarian cancer, bladder cancer, head and neck cancer, and gastric cancer; and PDR001 (nova), an inhibitory antibody that binds to PD-1, in clinical trials against non-small cell lung cancer, melanoma, triple negative breast cancer, and advanced or metastatic solid tumors. Tremelimumab (CP-675,206; Aslicon, Inc.) is a fully human monoclonal antibody against CTLA-4 that has been studied in clinical trials for a variety of indications, including: mesothelioma, colorectal, renal, breast, lung and non-small cell lung cancers, pancreatic ductal adenocarcinoma, pancreatic cancer, germ cell cancer, head and neck squamous cell carcinoma, hepatocellular, prostate, endometrial, metastatic cancer in the liver, liver cancer, large B-cell lymphoma, ovarian cancer, cervical cancer, metastatic undifferentiated thyroid cancer, urothelial cancer, fallopian tube cancer, multiple myeloma, bladder cancer, soft tissue sarcoma, and melanoma. AGEN-1884 (Agenus) is an anti-CTLA 4 anti-antibody being studied in a phase 1 clinical trial against advanced solid tumorsBody (NCT 02694822).

Nivolumab (a), (b) and (c)BMS-93568/MDX 1106; behcet masforth) is a fully human IgG4 monoclonal antibody that acts as an immunomodulator by binding to the proposed cell death 1(PD-1) receptor and selectively blocking interaction with its ligands PD-L1 and PD-L2. The structure and other properties of nivolumab are described in http:// www.drugbank.ca/drugs/DB09035(2016, 3, 14 days visit), the disclosure of which is hereby incorporated herein. Nivolumab has been approved for the treatment of advanced renal cell carcinoma patients who have received prior anti-angiogenic therapy; as a single agent for certain types of unresectable or metastatic melanoma; for use in the treatment of unresectable or metastatic melanoma, or in combination with ipilimumab for use in the treatment of unresectable or metastatic melanoma; and for the treatment of metastatic non-small cell lung cancer and exacerbations on or following platinum-based chemotherapy. Additionally, nivolumab has been tested or mentioned as a possible therapy for other oncological indications, including solid tumors; cutaneous melanoma; glioblastoma; a glioma; gliosarcoma; astrocytoma; brain cancer; leukemia; acute myeloid leukemia; chronic myelogenous leukemia; chronic lymphocytic leukemia; advanced liver cancer or hepatocellular carcinoma; uveal melanoma; prostate cancer; pancreatic tumors and pancreatic cancers; bladder cancer; colorectal cancer; myelodysplastic syndrome; hodgkin lymphoma; non-hodgkin lymphoma; multiple myeloma; cervical cancer; endometrial cancer; uterine cancer; ovarian cancer and ovarian carcinoma; peritoneal cancer; squamous cell carcinoma of the head and neck; gastric cancer; esophageal cancer; kaposi sarcoma (Kaposi sarcoma); breast tumors, breast cancer and breast cancer; osteosarcoma; soft tissue sarcoma; meningioma; and mesothelioma.

In phase 3 trials, over 800 patients with advanced clear cell renal cell carcinoma who had received prior treatment with one or two anti-angiogenic treatment regimens were randomly assigned to receive either 3mg nivolumab per kg body weight (intravenously, every two weeks) or 10mg tablets of everolimus (orally daily). Nivolumab treated patients exhibited longer median overall survival, decreased mortality risk ratio, and objective response rate (25%) was higher for nivolumab treated patients than for everolimus (5%) (P <0.001), with lower incidence of treatment-related grade 3 or 4 adverse events (mozar et al (2015), "New England Journal of Medicine" (373: 1803-) (1813)). Accordingly, in some embodiments, the present invention provides a method of treating advanced clear cell renal cell carcinoma comprising administering an effective amount of a CXCR4 antagonist (e.g., X4P-001 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof) in combination with nivolumab or everolimus to a patient in need thereof, optionally wherein the patient has received prior treatment with an anti-angiogenic treatment regimen.

In general, the amount of nivolumab or other immune checkpoint inhibitor suitable for use in the present invention will depend on the size, weight, age and condition of the patient being treated, the severity of the disorder or condition, and the judgment of the prescribing physician. For example, in the current prescription label for unresectable or metastatic renal cell carcinoma, the recommended course of nivolumab administration is 3mg/kg, i.v. infusion for 60 minutes, once every two weeks, until the disease worsens or unacceptable toxicity occurs. The clinician, at the discretion, may increase the prescribed dose of nivolumab, e.g., increase the dose and/or frequency, depending on the individual's tolerance. The clinician, at the discretion, in combination with the warning provided by the prescription information, may discontinue administration of nivolumab or reduce the dosage in the event of a significant side effect. In some embodiments, in the methods of the invention, nivolumab is administered according to the label guidelines described above.

In some embodiments, the invention provides a method of treating a patient by administering a CXCR4 antagonist (e.g., X4P-001 or a pharmaceutically acceptable salt thereof) in combination with an immunostimulatory therapeutic agent. Approved immunostimulatory therapeutics that may be used in the present invention include elotuzumab (elotuzumab) (anti-SLAMF 7 antibody,baishimei noble). Immunostimulatory compounds under investigation that may be used in the present invention include mifamurtide (mifamurtide) (iii)Wutian anticancer medicine (Takeda Oncology)).

Another immunostimulatory therapeutic agent that may be used in the present invention is recombinant human interleukin 15 (rhIL-15). rhIL-15 has been tested clinically as a therapy for melanoma and renal cell carcinoma (NCT01021059 and NCT01369888) and leukemia (NCT 02689453). Another immunostimulatory therapeutic agent that may be used in the present invention is recombinant human interleukin 12 (rhIL-12). Another suitable IL-15-based immunotherapeutic is heterodimeric IL-15(hetIL-15, Nowa/Admu (Admune)), a fusion complex composed of endogenous IL-15 complexed with the soluble IL-15 binding protein IL-15 receptor alpha chain in a synthetic form (IL15: sIL-15RA), which has been tested in phase 1 clinical trials for melanoma, renal cell carcinoma, non-small cell lung cancer, and squamous cell carcinoma of the head and neck (NCT 02452268). Recombinant human interleukin 12(rhIL-12) has been tested clinically for a variety of tumor indications, for example as a therapy for lymphoma (NM-IL-12, new biomedical limited (Inc.)) (NCT02544724 and NCT 02542124).

Another example for immune stimulation is the use of oncolytic viruses. In some embodiments, the present invention provides a method of treating a patient by administering the disclosed X4P-001 composition in combination with an immunostimulatory therapy (e.g., an oncolytic virus). Approved immunostimulatory oncolytic viruses that may be used in the present invention include talomogene laherparevec (attenuated live herpes simplex virus,installation).

As used herein, the term "aromatase inhibitor" relates to a compound that inhibits estrogen production (e.g., the substrates androstenedione and testosterone are converted to estrone and estradiol, respectively). The term includes, but is not limited to, steroids, especially aTamestane (atamestane), exemestane (exemestane) and formestane (formestane); and especially non-steroids, especially aminoglutethimide (aminoglutethimide), roglucimide (rogothimide), pirglutethimide (pyridoglucetimide), trilostane (trilostane), testolactone (testolactone), ketoconazole (ketoconazole), vorozole (vorozole), fadrozole (fadrozole), anastrozole (anastrozole) and letrozole (letrozole). Exemestane is available under the trade name AromasinTMAnd (5) selling. Formestane is under the trade name LentaronTMAnd (5) selling. Fadrozole is given the trade name AfemaTMAnd (5) selling. Anastrozole is given the trade name ArimidexTMAnd (5) selling. Letrozole is given the trade name FemaraTMOr FemarTMAnd (5) selling. Aminoglutethimide under the trade name OrimetenTMAnd (5) selling. The combinations of the invention comprising chemotherapeutic agents as aromatase inhibitors are particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.

As used herein, the term "antiestrogen" refers to a compound that antagonizes the effects of estrogen at the estrogen receptor level. The term includes, but is not limited to, tamoxifen (tamoxifen), fulvestrant (fulvestrant), raloxifene (raloxifene), and raloxifene hydrochloride. Tamoxifen is available under the trade name NolvadexTMAnd (5) selling. Ranoxiphenol hydrochloride is under the trade name EvistaTMAnd (5) selling. Fulvestrant may be under the brand name FaslodexTMAnd (4) application. The combinations of the invention comprising chemotherapeutic agents as antiestrogens are particularly useful in the treatment of estrogen receptor positive tumors, such as breast tumors.

As used herein, the term "antiandrogen" refers to any substance capable of inhibiting the biological effects of androgens and includes, but is not limited to, bicalutamide (Casodex)TM). As used herein, the term "gonadoliberin agonist" includes, but is not limited to abarelix (abarelix), goserelin (goserelin), and goserelin acetate. Goserelin may be under the trade name ZoladexTMAnd (4) application.

The term "topoisomerase I inhibitor" as used herein includes, but is not limited to, topotecan (topotecan), gemmacecan (gimatecan), irinotecan (irinotecan), heliotecan (heliotecan)Dendrobine (camptothecan) and its analogs, 9-nitrocamptothecin, and macromolecular camptothecin conjugate PNU-166148. Irinotecan can be, for example, in its form of sale (e.g. under the trademark Camptosar)TM) And (4) application. Topotecan is under the trademark HycamptinTMAnd (5) selling.

As used herein, the term "topoisomerase II inhibitor" includes, but is not limited to, anthracyclines, such as, for example, doxorubicin (doxorubicin) (including liposomal formulations, such as, for example, Caelyx)TM) Daunorubicin (daunorubicin), epirubicin (epirubicin), idarubicin (idarubicin) and nemorubicin (nemorubicin), anthraquinone mitoxantrone (mitoxantrone) and losoxantrone (losoxantrone), and etoposide (etoposide) and teniposide (teniposide). Etoposide is sold under the trademark EtopophosTMAnd (5) selling. Teniposide is sold under the trademark VM 26-Bristol. Raspberry under the trademark AcribilastinTMOr AdriamycinTMAnd (5) selling. Epirubicin is available under the trademark FarmorubicinTMAnd (5) selling. Idarubicin under the trade mark ZavedosTMAnd (5) selling. Mitoxantrone is sold under the trademark Novantron.

The term "microtubule active agent" refers to microtubule stabilizing, microtubule destabilizing compounds and tubulin polymerization inhibitors, including (but not limited to) taxanes such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine (vinorelbine); discodermolide (discodermolide); colchicine (cochine) and epothilone (epothilone) and derivatives thereof. Paclitaxel is available under the trademark TaxolTMAnd (5) selling. Docetaxel under the trademark TaxotereTMAnd (5) selling. Vinblastine sulphate is sold under the trade mark Vinblastin R.PTMAnd (5) selling. Vincristine sulphate under the trade mark FarmistinTMAnd (5) selling.

As used herein, the term "alkylating agent" includes, but is not limited to, cyclophosphamide (cyclophosphamide), ifosfamide (ifosfamide), melphalan (melphalan), or nitrosourea (nitrourea) (BCNU or gliader (Gliadel)). Cyclophosphamide under the trade mark CyclostinTMAnd (5) selling. Ifosfamide under the trademark HoloxanTMAnd (5) selling.

The term "histone deacetylase inhibitor" or "HDAC inhibitor" refers to a compound that inhibits histone deacetylase and has antiproliferative activity. This includes, but is not limited to suberoylanilide hydroxamic acid (SAHA).

The term "antineoplastic antimetabolite" includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds such as 5-azacytidine (5-azacytidine) and decitabine (decitabine), methotrexate (methotrexate) and edatrexate (edatrexate), and folate antagonists such as pemetrexed (pemetrexed). Capecitabine is under the trademark XelodaTMAnd (5) selling. Gemcitabine under the trademark GemzarTMAnd (5) selling.

As used herein, the term "platinum compound" includes, but is not limited to, carboplatin, cisplatin (cis-platinum), cisplatin (cissplatinum), and oxaliplatin (oxaliplatin). Carboplatin can be, for example, in its marketed form (e.g. under the trademark Carboplat)TM) And (4) application. Oxaliplatin can be, for example, in its marketed form (e.g. under the trademark Eloxatin)TM) And (4) application.

As used herein, the term "compound that targets/reduces protein or lipid kinase activity or protein or lipid phosphatase activity", or other anti-angiogenic compound "includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds that target, reduce or inhibit platelet-derived growth factor receptor (PDGFR) activity, such as compounds that target, reduce or inhibit PDGFR activity, particularly compounds that inhibit PDGF receptors, such as N-phenyl-2-pyrimidine-amine derivatives, such as imatinib (imatinib), SU101, SU6668 and GFB-111; b) compounds that target, decrease or inhibit Fibroblast Growth Factor Receptor (FGFR) activity; c) a compound that targets, reduces or inhibits the activity of insulin-like growth factor receptor I (IGF-IR), for example a compound that targets, reduces or inhibits the activity of IGF-IR, in particular a compound that inhibits the kinase activity of IGF-I receptor, or an antibody that targets the extracellular domain of IGF-I receptor or its growth factor; d) targeting, reducing or inhibiting the Trk receptor tyrosine kinase familyAn active compound, or a pterosin B4 inhibitor; e) a compound that targets, reduces or inhibits the activity of the AxI receptor tyrosine kinase family; f) compounds that target, decrease or inhibit Ret receptor tyrosine kinase activity; g) compounds that target, decrease or inhibit the activity of Kit/SCFR receptor tyrosine kinases, such as imatinib; h) compounds that target, decrease or inhibit the activity of the C-Kit receptor tyrosine kinase (part of the PDGFR family), such as compounds that target, decrease or inhibit the activity of the C-Kit receptor tyrosine kinase family, in particular compounds that inhibit the C-Kit receptor, such as imatinib; i) compounds which target, reduce or inhibit the activity of c-Abl family members, their gene fusion products (e.g. BCR-Abl kinase) and mutants, for example compounds which target, reduce or inhibit the activity of c-Abl family members and their gene fusion products, such as N-phenyl-2-pyrimidine-amine derivatives, for example imatinib or nilotinib (nilotinib) (AMN107), PD180970, AG957, NSC 680410, PD173955 from ParkeDavis, or dasatinib (BMS-354825); j) compounds that target, reduce or inhibit the activity of protein kinase c (pkc) and Raf family members of serine/threonine kinases, MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, TYK2, BTK and TEC family members, and/or cyclin dependent kinase family (CDK) members, including staurosporine derivatives, such as midostaurin; examples of other compounds include UCN-01, safrog (safingol), BAY 43-9006, bryodin 1, Perifosine (Perifosine), imofosine (llmofosine), RO 318220 and RO 320432, GO 6976, isis 3521, LY333531/LY379196, isoquinoline compounds, FTI, PD184352 or QAN697(P13K inhibitors) or AT7519(CDK inhibitors); k) compounds which target, reduce or inhibit the activity of protein tyrosine kinase inhibitors, for example compounds which target, reduce or inhibit the activity of protein tyrosine kinase inhibitors, including imatinib mesylate (Gleevec)TM) Or tofukins (tyrphostins), for example tofukins A23/RG-50810, AG 99, tofukins AG 213, tofukins AG 1748, tofukins AG 490, tofukins B44, tofukins B44(+) enantiomer, tofukins AG 555, AG 494, tofukins AG556, AG957 and adapfukins(adaphortin) (4- { [ (2, 5-dihydroxyphenyl) methyl]Amino } -benzoic acid adamantane esters; NSC 680410, adafostin); l) epidermal growth factor family (EGFR) targeting, reducing or inhibiting receptor tyrosine kinases1ErbB2, ErbB3, ErbB4 as homodimers or heterodimers) and mutants thereof, e.g. compounds that target, reduce or inhibit the activity of the epidermal growth factor receptor family, in particular compounds, proteins or antibodies that inhibit EGF receptor tyrosine kinase family members (e.g. EGF receptor, ErbB2, ErbB3 and ErbB4) or bind to EGF or EGF related ligands, CP 358774, ZD 1839, ZM 105180, trastuzumab (Herceptin)TM) Cetuximab (Erbitux)TM) Iressa, Tarceva, OSI-774, Cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or E7.6.3, and 7H-pyrrolo- [2,3-d ]]A pyrimidine derivative; m) a compound that targets, reduces or inhibits the activity of the c-Met receptor, e.g., a compound that targets, reduces or inhibits the activity of c-Met, particularly a compound that inhibits the kinase activity of the c-Met receptor, or an antibody that targets the extracellular domain of c-Met or binds to HGF; n) compounds that target, decrease or inhibit the kinase activity of one or more JAK family members (JAK1/JAK2/JAK3/TYK2 and/or pan JAK) including, but not limited to, PRT-062070, SB-1578, Baritinib (baricitinib), Paritinib (pacitinib), Momelotinib (momelotinib), VX-509, AZD-1480, TG-101348, tofacitinib and lucolinib; o) compounds that target, decrease or inhibit the kinase activity of PI3 kinase (PI3K), including, but not limited to, ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib (buparlisib), pikerxib (pictelisib), PF-4691502, BYL-719, dactyloxib (dactylisib), XL-147, XL-765, and idecoxib (idelalisib); and q) compounds that target, reduce or inhibit the signaling effects of the hedgehog (Hh) or smoothing receptor (SMO) pathway, including, but not limited to, cyclopamine (cyclopamine), vismodegib (vismodegib), itraconazole (itraconazole), imodege (rimodegib), and IPI-926 (saridegib).

As used herein, the term "PI 3K inhibitor" includes, but is not limited to, compounds having inhibitory activity against one or more enzymes of the phosphatidylinositol-3-kinase family, including, but not limited to, PI3K α, PI3K γ, PI3K δ, PI3K β, PI3K-C2 α, PI3K-C2 β, PI3K-C2 γ, Vps34, p110- α, p110- β, p110- γ, p110- δ, p85- α, p85- β, p55- γ, p150, p101, and p 87. Examples of PI3K inhibitors suitable for use in the present invention include, but are not limited to, ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib (buparlisib), pikerxib (pictelisib), PF-4691502, BYL-719, dactulisib (dacylisib), XL-147, XL-765, and idelalisib (idelalisib).

As used herein, the term "Bcl-2 inhibitor" includes, but is not limited to, compounds having inhibitory activity against B-cell lymphoma-2 protein (Bcl-2), including, but not limited to, ABT-199, ABT-731, ABT-737, apogossypol (apocosypol), pan Bcl-2 inhibitors of Ascenta, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (infinite Pharmaceuticals)/Novartis Pharmaceuticals (Novartis Pharmaceuticals)), Narconsis (Genasense) (G3139), 89HA 14-1 (and analogs thereof; see WO2008118802), Navicola (navitoclaxa) (and analogs thereof; see US7390799), NH-1 (Shenayng Pharmaceutical University) (WO 200639), and Glioclad 2006328) (see Glioclad # 4109, Glioclad-2), and analogs thereof (Glioclad-2) (see WO 41063639, Glioclad-2, and analogs thereof), TW series compounds (university of Michigan (univ. of Michigan)) and venetola (venetocalax). In some embodiments, the Bcl-2 inhibitor is a small molecule therapeutic. In some embodiments, the Bcl-2 inhibitor is a peptidomimetic.

As used herein, the term "BTK inhibitor" includes, but is not limited to, compounds having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including, but not limited to, AVL-292 and ibrutinib.

As used herein, the term "SYK inhibitor" includes, but is not limited to, compounds having inhibitory activity against spleen tyrosine kinase (SYK), including, but not limited to, PRT-062070, R-343, R-333, Iselier (Excellair), PRT-062607, and fostertinib (fosomatinib).

Further examples of BTK inhibiting compounds and conditions that such compounds can treat in combination with the compounds of the present invention can be found in WO2008039218 and WO2011090760, the entire contents of which are incorporated herein by reference.

Further examples of SYK inhibiting compounds and conditions that such compounds can treat in combination with the compounds of the present invention may be found in WO2003063794, WO2005007623 and WO2006078846, the entire contents of which are incorporated herein by reference.

Further examples of PI3K inhibiting compounds and conditions that such compounds can treat in combination with the compounds of the invention can be found in WO2004019973, WO2004089925, WO2007016176, US8138347, WO2002088112, WO2007084786, WO2007129161, WO2006122806, WO2005113554 and WO2007044729, the entire contents of which are incorporated herein by reference.

Further examples of JAK inhibitory compounds and conditions that such compounds can treat in combination with the compounds of the present invention can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246 and WO2007070514, the entire contents of which are incorporated herein by reference.

Other anti-angiogenic compounds include compounds with another mechanism of activity (e.g., not associated with protein or lipid kinase inhibition), such as thalidomide (Thalomid)TM) And TNP-470.

Examples of proteasome inhibitors suitable for use in combination with the compounds of the present invention include, but are not limited to, bortezomib (bortezomib), disulfiram (disulphiram), epigallocatechin-3-gallate (EGCG), salinosporin A, carfilzomib (carfilzomib), ONX-0912, CEP-18770 and MLN 9708.

Compounds targeting, reducing or inhibiting the activity of a protein or lipid phosphatase are for example phosphatase 1 inhibitors, phosphatase 2A inhibitors or CDC25 inhibitors, such as okadaic acid (okadaic acid) or derivatives thereof.

Compounds that induce a cell differentiation process include, but are not limited to, retinoic acid, alpha-tocopherol, gamma-tocopherol, or delta-tocopherol, or alpha-tocotrienol, gamma-tocotrienol, or delta-tocotrienol.

As used herein, the term cyclooxygenase inhibitor includes, but is not limited to Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acids, and derivatives, such as celecoxib (Celebrex)TM) Etoricoxib, valdecoxib or 5-alkyl-2-arylaminophenylacetic acids, such as 5-methyl-2- (2 '-chloro-6' -fluoroanilino) phenylacetic acid, lumiracoxib.

As used herein, the term "bisphosphonate" includes, but is not limited to, itration (etodonic acid), clodronic acid (clodronic acid), tiludronic acid (tiludronic acid), pamidronic acid (pamidronic acid), alendronic acid (alendronic acid), ibandronic acid (ibandronic acid), risedronic acid (risedronic acid), and zoledronic acid (zoledronic acid). Etidronic acid under the trade mark DidronelTMAnd (5) selling. Chlorophosphonic acids are under the trade mark BonefosTMAnd (5) selling. Telophosphonic acid is available under the trade name SkelidTMAnd (5) selling. Pamidronic acid as commercial ArediaTMAnd (5) selling. Alendronic acid under the trade mark FosamaxTMAnd (5) selling. Ibandronic acid is under the trade mark BondranatTMAnd (5) selling. Risedronic acid under the trademark ActonelTMAnd (5) selling. Zomet phosphonic acid under the trademark ZometTMAnd (5) selling. The term "mTOR inhibitor" refers to a compound that inhibits the mammalian target of rapamycin (mTOR) and has antiproliferative activity, such as sirolimus (sirolimus) ((mTOR))) Everolimus (Certican)TM) CCI-779 and ABT 578.

As used herein, the term "heparinase inhibitor" refers to a compound that targets, reduces or inhibits the degradation of heparin sulfate. The term includes, but is not limited to, PI-88. As used herein, the term "biological response modifier" refers to a lymphokine or interferon.

As used herein, the term "inhibitor of Ras oncogenic isoforms (e.g., H-Ras, K-Ras, or N-Ras)" refers to compounds that target, reduce, or inhibit the oncogenic activity of Ras; example (b)Such as "farnesyl transferase inhibitors", for example L-744832, DK8G557 or R115777 (Zarnestra)TM). As used herein, the term "telomerase inhibitor" refers to a compound that targets, reduces, or inhibits telomerase activity. Compounds which target, decrease or inhibit telomerase activity are in particular compounds which inhibit the telomerase receptor, for example telomerase (telomestatin).

As used herein, the term "methionine aminopeptidase inhibitor" refers to a compound that targets, reduces, or inhibits methionine aminopeptidase activity. Compounds that target, reduce or inhibit methionine aminopeptidase activity include, but are not limited to, benguanamide (bengamide) or derivatives thereof.

As used herein, the term "proteasome inhibitor" refers to a compound that targets, reduces, or inhibits proteasome activity. Compounds that target, reduce or inhibit proteasome activity include, but are not limited to, bortezomib (Velcade)TM) And MLN 341.

As used herein, the term "matrix metalloproteinase inhibitor" or "MMP" inhibitor "includes, but is not limited to, collagen peptide mimetic and non-peptidomimetic inhibitors, tetracycline derivatives, such as the hydroxamate peptide mimetic inhibitor batimastat (batimastat) and its orally bioavailable analogs marimastat (marimastat) (BB-2516), prinomastat (prinomastat) (AG3340), metamastat (metastat) (NSC 683551), BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ 996.

As used herein, the term "compound for the treatment of hematological malignancies" includes, but is not limited to, FMS-like tyrosine kinase inhibitors, which are compounds that target, reduce or inhibit the activity of FMS-like tyrosine kinase receptor (Flt-3R); interferon, 1-beta-D-arabinofuranosyl cytosine (ara-c) and busulfan; and ALK inhibitors, which are compounds that target, reduce, or inhibit undifferentiated lymphoma kinase.

Compounds which target, reduce or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are in particular compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, staurosporine derivatives, SU11248 and MLN 518.

As used herein, the term "HSP 90 inhibitor" includes, but is not limited to, compounds that target, reduce, or inhibit the intrinsic atpase activity of HSP 90; compounds that degrade, target, reduce or inhibit HSP90 client proteins (client proteins) through the ubiquitin proteosome pathway. Compounds that target, decrease or inhibit the intrinsic atpase activity of HSP90, in particular compounds, proteins or antibodies that inhibit the atpase activity of HSP90, such as 17-allylamino, 17-demethoxygeldanamycin (17AAG) (geldanamycin) derivatives); other geldanamycin related compounds; radicicol (radicicol); and HDAC inhibitors.

The term "anti-proliferative antibody" as used herein includes, but is not limited to, trastuzumab (trastuzumab) (Herceptin)TM) trastuzumab-DM 1, erbitux, bevacizumab AvastinTM) Rituximab (rituximab) ((R))) PRO64553 (anti-CD 40) and 2C4 antibodies. By antibody is meant intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity.

For the treatment of Acute Myeloid Leukemia (AML), the compounds of the invention can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, the compounds of the present invention may be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs suitable for treating AML, such as daunorubicin, doxorubicin (Adriamycin), Ara-C, VP-16, teniposide, mitoxantrone, idamycin, carboplatin (carboplatin) and PKC 412.

Other anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog which is a 2' - α -hydroxyribose (arabinoside) derivative of deoxycytidine. Purine analogs of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate (fludarabine phosphate) are also included. Compounds that target, reduce, or inhibit the activity of Histone Deacetylase (HDAC) inhibitors, such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA), inhibit the activity of an enzyme known as histone deacetylase. Specific HDAC inhibitors include compounds disclosed in MS275, SAHA, FK228 (formerly FR901228), trichostatin a (trichostatin a) and US 6,552,065, including, but not limited to, N-hydroxy-3- [4- [ [ [2- (2-methyl-1H-indol-3-yl) -ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof, and N-hydroxy-3- [4- [ (2-hydroxyethyl) {2- (1H-indol-3-yl) ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof, particularly lactate. As used herein, somatostatin receptor antagonists refer to compounds that target, treat or inhibit somatostatin receptors, such as octreotide (octreotide) and SOM 230. Tumor cell destruction means, for example, ionizing radiation. The term "ionizing radiation" referred to above and hereinafter means ionizing radiation that is carried out in the form of electromagnetic rays (e.g., X-rays and gamma rays) or particles (e.g., alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Helman (Hellman), the Principles of Radiation Therapy for Cancer (Principles of Radiation Therapy, Cancer), the Principles and Practice of Oncology (Principles and Practice of Oncology), Devicat (Devita), et al, 4 th edition, Vol.1, p.248-275 (1993).

Also included are EDG binding agents and ribonucleotide reductase inhibitors. As used herein, the term "EDG binding agent" refers to a class of immunosuppressive agents that modulate lymphocyte recirculation, such as FTY 720. The term "ribonucleotide reductase inhibitor" refers to a pyrimidine or purine nucleoside analog, which includes, but is not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine (cladribine), 6-mercaptopurine (especially in combination with ara-C for ALL) and/or pentostatin (pentostatin). Ribonucleotide reductase inhibitors are in particular hydroxyurea or 2-hydroxy-1H-isoindole-1, 3-dione derivatives.

Especially those compounds, proteins or monoclonal antibodies which also include VEGF, such as 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine or a pharmaceutically acceptable salt thereof, 1- (4-chloroanilino) -4- (4)-pyridylmethyl) phthalazine succinate; angiostatinTM;EndostatinTM(ii) a Anthranilic acid amides; ZD 4190; zd6474; SU 5416; SU 6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab, VEGF aptamers, such as mokugong (Macugon); FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2IgGI antibodies, Amphibin (Angiozyme) (RPI 4610) and bevacizumab (Avastin)TM)。

As used herein, photodynamic therapy refers to therapy using certain chemicals called photosensitizing compounds to treat or prevent cancer. Examples of photodynamic therapy include treatment with, for example, VisudyneTMAnd porfimer sodium.

As used herein, an angiogenesis-inhibiting steroid refers to a compound that blocks or inhibits angiogenesis, such as anecortave (anecortave), triamcinolone (triamcinolone), hydrocortisone (hydrocortisone), 11- α -epihydrocortisone (11- α -anhydrocortisol), deoxycorticosterol (cortisolve), 17 α -hydroxyprogesterone (17 α -hydroxyprogesterone), corticosterone (corticosterone), deoxycorticosterone (desoxycorticosterone), testosterone (testosterone), estrone, and dexamethasone (dexomethasone).

Implants containing corticosteroids refer to compounds such as fluocinolone acetonide (fluocinolone) and dexamethasone.

Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; an antisense oligonucleotide or oligonucleotide derivative; shRNA or siRNA; or hybrid compounds or compounds with other or unknown mechanisms of action.

The structures of active compounds identified by code number, common name or trademark can be obtained from The actual version of The standard profile Merck Index (The Merck Index) or from databases, such as International Patents (Patents International), such as IMS World Publications.

The compounds of the invention may also be used in combination with known therapeutic methods, such as administration of hormones or radiation. In certain embodiments, the provided compounds are useful as radiosensitizers, particularly for treating tumors that are less sensitive to radiotherapy.

The compounds of the invention can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapies employing fixed combinations or staggered administration or administration independently of one another of the compounds of the invention and one or more other therapeutic compounds, or administration of a fixed combination in combination with one or more other therapeutic compounds. Additionally or alternatively, the compounds of the present invention may be administered for tumor therapy, in particular in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention or a combination of these. In the case of other treatment strategies, long-term therapy and adjuvant therapy are likewise possible, as described above. Other possible therapies are those that maintain the patient's state after tumor regression, or even chemopreventive therapies (e.g. for patients at risk).

Those other agents may be administered separately from the disclosed X4P-001 composition as part of a multiple dosing regimen. Alternatively, those agents may be part of a single dosage form, mixed together with the disclosed X4P-001 composition into a single composition. If administered as part of a multiple dosing regimen, the two active agents may be provided simultaneously, sequentially or at a time interval from each other (typically within five hours of each other).

As used herein, the terms "combination", "combination" and related terms refer to the simultaneous or sequential administration of a therapeutic agent according to the present invention. For example, the compound of the present invention may be administered simultaneously or sequentially with another therapeutic agent in separate unit dosage forms or mixed into a single unit dosage form. Thus, the present invention provides a single unit dosage form comprising a compound of the invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.

The amounts of the disclosed X4P-001 composition and other therapeutic agent (present in those compositions comprising another therapeutic agent as described above) that can be combined with a carrier material to produce a single dosage form will vary depending on the host treated and the particular mode of administration. The compositions of the invention should preferably be formulated so as to be capable of administering a dose of between 0.01 and 100mg of a compound of the invention per kg of body weight per day.

In those compositions that comprise other therapeutic agents, the other therapeutic agent and the compound of the invention may act synergistically. Thus, the amount of other therapeutic agent in such compositions will be lower than that required in monotherapy using only that therapeutic agent. In such compositions, other therapeutic agents can be administered at doses between 0.01 and 1,000 μ g per kg body weight per day.

The amount of other therapeutic agent present in the compositions of the present invention will be no greater than that normally administered in compositions containing that therapeutic agent as the only active agent. Preferably, the amount of the other therapeutic agent in the disclosed X4P-001 composition will range from about 50% to 100% of the amount typically present in a composition comprising that agent as the only therapeutically active agent.

The compounds of the present invention or pharmaceutical compositions thereof may also be incorporated into compositions for coating implantable medical devices, such as prostheses, prosthetic valves, vascular prostheses, stents, and catheters. Vascular stents, for example, have been used to overcome restenosis (restenosis of the vessel wall after injury). However, patients using stents or other implantable devices are at risk for clot formation or platelet activation. These undesirable effects can be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor. Implantable devices coated with the compounds of the present invention are another embodiment of the present invention.

In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and should not be construed as limiting the invention in any way.

Illustrative description

As depicted in the examples below, in certain exemplary embodiments, the compounds are prepared according to the following general procedure. It is to be understood that while the general methods depict the synthesis of certain compounds of the invention, the following general methods and other methods known to those of ordinary skill in the art may be applicable to all compounds as described herein and to subclasses and classes of each of these compounds.

Methods for preparing certain compounds suitable for use in the present invention are disclosed in croughard (Crawford) et al (2008) organic process research and development (org. process res.dev.) 12:823 830; US7,354,934, WO 00/56729, USSN60/232,891 and USSN 60/234,510 and ann H. (An, H.); king T. (Wang, T.); mohan, V.); griffoy, r.h. (Griffey, r.h.); cook P.D (Cook, P.D.) 1998,54, 3999-4012; each of which is incorporated herein by reference in its entirety. Such disclosed methods can be modified by one of ordinary skill in the art using no more than routine experimentation to provide alternative means of preparation, testing, and analysis of the compounds of the invention.

Example 1: impurities of methyl imine

According to high performance liquid chromatography-mass spectrometry (HPLC-MS) analysis, a peak at a Relative Retention Time (RRT) of 1.13 shows [ M +1]]+And 362m/z (12 mass units higher than X4P-001). If X4P-001 is dissolved in an acidic aqueous medium, the peak disappears after a few hours. The molecular weight of the impurities and their chemical behavior indicate the presence of a methyl imine structure on the primary amine. Without wishing to be bound by any particular theory, it is believed that compound I-1 is formed in the presence of formaldehyde or a formaldehyde source. The imine impurity I-1 and possible routes of formation are shown in scheme 1 below.

Scheme 1

Example 2: n-formyl impurities

According to HPLC-MS analysis, the RRT 1.28 peak shows [ M +1] + at 378M/z (28 mass units higher than X4P-001), indicating an N-formyl derivative of X4P-001. This has been confirmed by independent synthesis. Direct reaction of X4P-001 with ethyl formate yielded the N-formyl compound I-2, which was identical to the batch impurity according to MS and HPLC data. The formation of the formate ester is shown in scheme 2 below.

Scheme 2

Example 3: aldehyde impurities

The RRT 1.14 peak has the characteristic of I-6. Without wishing to be bound by any particular theory, it is believed that I-6 results from the oxidation of the amine on the alkyl chain of X4P-001, as shown in scheme 3 below:

scheme 3

Example 4: acetamide impurities

According to HPLC-MS analysis, the RRT 1.39 peak shows [ M +1] + 392M/z (42 mass units higher than X4P-001), which is characteristic of X4P-001 acetamide. Formed by the reaction of X4P-001 with isopropyl acetate crystallization solvent at elevated temperature as shown in scheme 4. The structure was confirmed by the independent synthesis of acetamide impurity. Reaction of X4P-001 with acetic anhydride provided acetamide I-5, which was identical to the batch impurity according to MS and HPLC data. Isopropyl acetate is used in some processes for making the free base X4P-001 using p-hydroxybenzoate salts of X4P-001.

Scheme 4

Example 5: benzimidazole impurities

According to LC-MS analysis, the RRT 1.67 peak showed [ M +1] + of 481M/z (131 mass units higher than X4P-001), which is characteristic of the M + benzimidazole compound I-3. Which is formed as an impurity during the protection of 2-chloromethylbenzimidazole with tert-butoxycarbonyl (Boc group) as shown in scheme 5 below. During the N-alkylation reaction (step 2 of the X4P-001 synthesis), the impurities undergo alkylation and are therefore present throughout the remainder of the manufacturing process.

Scheme 5

A separate synthesis involved reaction of secondary amine 2918 with boc-2-chloromethylbenzimidazole 2890(I-9), followed by deprotection provided I-3, which was identical to the impurity observed according to MS and HPLC.

Example 6: aminal impurities

Aminal impurities are present when p-hydroxybenzoic acid salt of X4P-001 is produced from the reaction of imine impurities with p-hydroxybenzoic acid. In the method using p-hydroxybenzoate salt of X4P-001 or an intermediate thereof, such impurities are associated with the production of X4P-001.

The structure of the aminal impurity I-4 and its source are shown in scheme 6 below.

Scheme 6

Example 7: basis for setting acceptance criteria for organic impurities

The impurity results for the relevant batches of X4P-001 and its para-hydroxybenzoic acid (PHB) salt are shown in Table 2 below. The X4P-001 batch used in the initial toxicological study was used primarily to set impurity specifications. P-hydroxybenzoate batches of X4P-001 were used for a longer term toxicological study and have also been shown in Table 2. The total impurities in these six batches ranged from < 0.05% to 1.4%, at the 0.1% level, with individual non-listed impurities > 0.05% present in only one batch. Given the limited number of batches produced and the impurity profile of the X4P-001 batch used for toxicological and clinical studies, the total impurity release profile for the drug substance has been set to ≦ 3.0% w/w, with no more than 0.2% w/w (0.5% in terms of stability) of a single unlisted impurity (Table 2). The individual impurities listed and the p-hydroxybenzoic acid content, with the exception of the imine impurity (which has been set at 1.1% according to the level of this impurity administered in the toxicological study), were ≦ 0.5% w/w. The individual batches of product (not shown in table 2), i.e. X4P-001 batch 3-1 (table 4), provided a high purity drug substance in which the total impurity was 1.20% w/w and the unspecified impurities were greater than 0.07% w/w except for the imine impurity (0.62% w/w). The analysis of the clinical X4P-001 bulk drug batch gave 99.3% w/w and chiral purity > 99% ee.

TABLE 2

Abbreviations used:

avalues below LOQ (limit of quantitation)

bThe only relevant specification NA of the free base clinical drug substance manufactured using X4P-001 p-hydroxybenzoate as starting material is not applicable

ND is not detected

NLT ═ not less than

Not exceeding NMT ═

NR ═ no report

Typically, the wt% of each impurity is determined by HPLC and measured initially or after storage, and optionally continuously during the shelf life of the X4P-001 composition. In some embodiments, the impurity content is measured after storage of the composition under stress conditions (conditions of elevated temperature, humidity, or both, for estimating the effect of long term storage under ambient conditions). Thus, in some embodiments, the present invention provides an X4P-001 composition comprising: not more than 1.1% of imine (I-1); up to 0.3% of formyl groups (I-2); no more than 0.4% benzimidazole (I-3); up to 0.5% of aminal (I-4); not more than 0.5% acetamide (I-5); not more than 0.4% of aldehyde (I-6); no more than 0.3% de-BOC NT-316 (I-7); and no more than 0.2% of a single unlisted impurity. In addition, when the free base clinical drug substance was manufactured via X4P-001 p-hydroxybenzoate, no more than 1.0% of PHB was present in the composition. In some embodiments, no more than 3.0% total impurities are present. In some embodiments, the enantiomeric excess (% ee) of the R-enantiomer is not less than 97.0%.

Example 8: identification of impurities produced after storage

Stability test samples of X4P-001 (PTL/ST/0511) stored for three months at 25 ℃/60% relative humidity (25/60) by MET/CR/1448 analysis showed the formation of two unknown impurities.

Unknown 1-RRT 1.14

Unknown 2-RRT 1.24

The RRT 1.14 impurity is also the major degradation product of PTL/DA/0175 under conditions of high temperature and high humidity (80 ℃/80% RH).

SUMMARY

During the stability study, two impurities formed by X4P-001 at t ═ 3 months at 25 ℃/60% RH and in the forced degradation study at high temperature/high humidity were identified by LC-MS.

Experiment of

Parameters of the instrument

The study was performed on a GMP Waters Alliance HPLC system connected to ZQ 2000 single quadrupole MS. The collected data is entered into the Empower 2 software.

The analysis and impurity method for X4P-001(MET/CR/1448) is not suitable for mass spectrometry because it uses TFA as a modulator in the eluent, which can significantly inhibit ionization. Initially, this was used to replace 0.15% v/v formic acid. The flow rate was also varied to allow direct connection to the inlet of the MS (maximum flow rate 0.3mL/min) and the gradient was adjusted accordingly.

HPLC Condition 1

Column: zorbax Bonus-RP,150x4.6mm,3.5 μm

Injection volume: 100 μ L

And (3) detection: UV @220nm (190-

MS ES+100-700Da,ES-100-700Da

Mobile phase A: 0.15% formic acid/water

A mobile phase B: 0.15% formic acid/MeCN

Time (minutes) %A %B
0.0 95 5
6 95 5
51 5 95
60 5 95
60.1 95 5
75 95 5

Flow rate: 0.3mL/min

Column temperature: environment(s)

The operation time is as follows: 75 minutes

MS tuning parameter 1

Ion source option ES+ ES-
Capillary voltage (kV) 3.5 4
Taper hole voltage (V) 25 25
Extractor (V) 3 0
RF lens 0.4 3
Source temperature C 150 150
Desolventizing temperature DEG C 150 150
Desolventizing gas flow (L/hr) 300 300
Taper hole gas flow (L/hr) 50 50
Analyzer options
LM Res 15 15
HM Res 15 15
Energy of ions 1.8 2
Multiplier 521 521

X4P-001 was not trapped under these conditions because it required ion pairing of TFA. The chromatogram and the resulting mass spectrum are shown in fig. 5.

Experimental qualities of both impurities were obtained. All mass spectra shown have been background corrected (regions taken immediately before and after the peak of interest).

Unknown 1MET/CR/1448RRT 1.14[ M + H ] ═ M/z 349.2

Unknown 2MET/CR/1448RRT 1.24[ M + H ] ═ M/z 392.3

The inventors hypothesize that there is a possibility that the impurity of interest co-elutes with other impurities. Samples were therefore run using chromatographic conditions in MET/CR/1448. The concentration and injection volume were increased to counteract TFA-induced ionization inhibition.

HPLC Condition 2

Column: zorbax SB-C8,150mmx4.6mm,3.5 μm

Protection of the column: zorbax SB-C8,12.5mmx4.6mm,5 μm

Injection volume: a plurality of

And (3) detection: 270nm UV

Mobile phase: mobile phase A: 0.2% TFA/water

A mobile phase B: 0.1% TFA/acetonitrile

Gradient elution

Time (minutes) %A %B Flow (mL/min)
0 92 8 0.8
5 90 10 0.8
15 89 11 0.8
25 80 20 0.8
28 80 20 0.8
37 55 45 0.8
44 20 80 0.8
47 20 80 0.8
48 92 8 1.2
53 92 8 1.2
54 92 8 0.8
55 92 8 0.8

Initial flow rate: 0.8mL/min (referring to the gradient schedule), 4:1 split to the MS inlet.

Column temperature: 25 deg.C

The operation time is as follows: 55 minutes

The MS parameters follow the tuning parameter 1 described above.

Multiple samples of X4P-001 in which the impurity of interest has been detected were prepared at 1 or 10mg/mL in methanol:

PTL/ST/0511, batch 3-1, 25/60t 3 months

PTL/DA/0175 degraded samples: 80 ℃/80% relative humidity (80/80), t ═ 1 and 7 days.

The resulting data are shown in fig. 6-9.

Mass spectral data obtained from HPLC condition 2 have been confirmed:

unknown 1MET/CR/1448RRT 1.14[ M + H ] ═ M/z 349.2

Unknown 2MET/CR/1448RRT 1.24[ M + H ] ═ M/z 392.3

Finally, the chromatographic conditions were modified to allow flow to enter the MS without splitting. The results from HPLC condition 3 are shown in fig. 10.

HPLC Condition 3

Column: zorbax SB-C8,150mm × 4.6mm,3.5 μm

Protection of the column: zorbax SB-C8,12.5mmx4.6mm,5 μm

Injection volume: a plurality of

And (3) detection: 270nm UV

Mobile phase: mobile phase A: 0.2% TFA/water

A mobile phase B: 0.1% TFA/acetonitrile

Gradient elution

Time (minutes) %A %B
0 92 8
13.3 90 10
39.9 89 11
66.50 80 20
74.48 80 20
98.42 55 45
117 20 80
125 20 80
125.10 92 8
140 92 8

Flow rate: 0.3mL/min, directly into the MS inlet

Column temperature: 25 deg.C

The operation time is as follows: 140 minutes

The MS parameters follow the tuning parameter 1 described above.

Discussion of the results

All the spectra obtained confirmed the initial results, despite the different peak responses and resolution of X4P-001.

Unknown 1MET/CR/1448RRT 1.14[ M + H ] ═ M/z 349.2

Unknown 2MET/CR/1448RRT 1.24[ M + H ] ═ M/z 392.3

Unknown 1RRT 1.14 (aldehyde)

The molecular weight of 348 indicates nitrogen loss (nitrogen rule). This is consistent with the oxidation of amines on the alkyl chain to produce aldehydes. The assumed structure is as follows.

Empirical formula C21H24N4O

Monoisotopic mass 348.195

Unknown 2RRT 1.24 (acetamide)

391 molecular weight was consistent with the I-5 acetamide impurity (formed by reaction of X4P-001 with residual isopropyl acetate). The structure of I-5 is shown below.

Empirical formula ═ C23H29N5O

Monoisotopic mass 391.237

Conclusion

Two unknown impurities were identified from the mass spectral data obtained. The RRT 1.14 impurity is the aldehyde oxidation product and the RRT 1.24 impurity is acetamide impurity I-5.

Example 9: improvement to the X4P-001 manufacturing process for the purpose of reducing and controlling impurity levels

Introduction to the design reside in

The final step in the previous preparation of GMP drug substance supporting clinical studies (version 2 of the process; shown in FIG. 2) was to start with the compounds NT-316 and AMD-2890 (I-9). After studying the structure and source of the most significant impurities formed in this preparation, new processes (3 rd edition processes) have been developed that provide improved control over impurity profiles and API crystallization. Furthermore, this effort has significantly enhanced the reproducibility and robustness of the last step in the process.

Description of synthetic routes and Key intermediates

API X4P-001 was assembled in 2 steps starting from two key fragments (NT-316 and AMD-2890) in a pooled manner. The penultimate intermediate AMD-11070 was not isolated, but was immediately converted to the API, which crystallized after the work-up procedure.

Scheme 7 and scheme 8 show the synthesis of API molecule X4P-001.

Scheme 7: early steps in the synthesis of X4P-001.

And (3) a process 8: late steps in the synthesis of X4P-001.

The key fragment AMD-2890 was synthesized from RM-396(I-8) in one step. Although RM-396 is commercially available, its purity varies greatly, depending on its source. According to HPLC analysis, the purity of commercially derived materials can be presented as higher (>97 area%), whereas true wt% purity has been found to be as low as 90%. RM-396 may undergo self-alkylation leading to the formation of a different mixture of oligomeric impurities. Only after conversion of RM-396 to AMD-2890 does a stable compound with a well-defined impurity profile be obtained.

Another key fragment is NT-316, a well characterized stable crystalline compound that incorporates a large portion of the API structure. NT-316 is obtained by convergent synthesis via reaction of NT-319 with a chiral amine salt NT-272. NT-272 is a stable crystalline compound that can be obtained via custom synthesis.

NT-319 is a highly unstable intermediate that cannot be isolated without degradation. It was prepared from commercially available ABA by 3 steps (no intermediate isolated at each step). ABA purity is difficult to control because it is not only an oil, but it lacks UV chromophores, severely limiting its precise analytical characterization method.

Based on the above, it was the combination of the impurity profile of these two fragments NT-316 and AMD-2890(I-9) with the exact conditions of the last two chemical steps (performed under GMP control) that determined the impurity profile and quality of the API.

In summary, the chemical purity and enantiomeric purity of NT-316 and the chemical purity of AMD-2890 can be directly related to the chemical purity and enantiomeric purity of API (X4P-001).

Description of Key Process variations

Fig. 3 shows how the new process (process 3) differs from the previous version (process 2). In version 2 of the process, it has proven necessary to first isolate the API in its 4-hydroxybenzoate form. This salt is then converted to the corresponding free base by a separate step in order to obtain the API.

The chemical steps of the version 3 process are unchanged. The compounds NT-316 and AMD-2890(I-9) were reacted with each other and the resulting product (Tri-Boc) was not isolated but immediately deprotected to give the crude API. However, in the 3 rd edition of the process, the method for separating the API has been greatly changed.

The most significant impurities produced by the version 2 process are imine a (RRT 1.08) and N-formyl B (RRT 1.28), as shown in scheme 9. These impurities have been shown to originate from the use of dichloromethane as solvent during the extraction of the crude API. In addition, the formation of acetamide impurity C (RRT 1.37) may be associated with the use of isopropyl acetate as a solvent in the final crystallization of the API free base.

And (3) a process 9: impurities found in bulk drug substances

Thus, dichloromethane and isopropyl acetate used for the treatment and isolation of X4P-001 in version 2 were replaced with 1-butanol and a toluene/methanol mixture, respectively, in version 3. We have found that these solvents do not react with the API and therefore we believe that this change has caused a significant reduction in the impurities a (imine), B (N-formyl) and C (acetamide) we have observed.

In addition, the problem of API formation as gum and oil during crystallization of the final product was found in the 2 nd version of the process due to the quality of AMD-2890 starting material. This material was obtained from commercial 2-chloromethylbenzimidazole (RM-396), which can have low wt% purity, even when the area% purity appears good (> 98%). The development of an improved AMD-2890 isolation procedure is an integral part of the 3 rd edition process and thus has continuously led to the production of high quality AMD-2890 with not only excellent% purity, but also excellent analytical% w/w purity.

Further optimization of aeration and carbon treatment during API separation has enabled better control of the color of the separated API, and these operations are also incorporated into the 3 rd edition of the process.

Finally, the crystallization of the free base API in toluene is narrowly defined. Metastable zones and optimal inoculation points were determined. We also developed an optimal cooling rate after seedbed formation, and an appropriate washing and drying protocol.

Figure 4 provides a detailed comparison between the version 2 process and the version 3 process in terms of downstream continuous operations with respect to API processing and separation.

The robustness and reproducibility of the version 3 process was demonstrated by performing three identical laboratory experiments on a 100g scale. As the data in table 3 and table 4 show, this process demonstrates success. We then scale up the process to the 10kg scale. A total of 9.75kg of GMP X4P-001 was prepared in a single batch, and the results were almost the same. Details of these experiments are discussed below.

Description of Process version 3 and improvements of Process 1 and Process 2

The following section provides a brief description of the various operations of API synthesis and separation in the final step of the 3 rd edition of the process.

Synthesis of NT-316

The early process steps for the preparation of the key starting material NT-316 from the custom-synthesized chiral amines NT-272 and ABA are unchanged relative to earlier runs.

Synthesis of AMD-2890

As mentioned above, the synthesis of AMD-2890(I-9) has been improved, especially its isolation and crystallization, in order to accommodate the varying purity and color of commercial 2-chloromethylbenzimidazole (RM-396). Such materials are typically dark brown to black. Thus introducing a carbon treatment to better control the color of AMD-2890.

Thus, 2-chloromethylbenzimidazole (RM-396) was reacted with 1.3 equivalents of di-tert-butyl dicarbonate in 8.6 volumes (vol) of DMF (N, N-dimethylformamide) in the presence of 0.1 equivalents (equiv.) of DIPEA (diisopropylethylamine) at 40 ℃. After the reaction is complete, decolorizing carbon is added. After aging for 1.5 hours at 40 ℃, the mixture was filtered and the solid was washed with 1vol DMF. Water (3.5vol) was slowly added to the filtrate, resulting in a 1% AMD-2890 seeded slightly cloudy mixture. After aging, a slurry was produced, which was allowed to cool slowly to 20 ℃. After slowly adding more water (1vol) and further cooling to 0 ℃, the slurry was filtered. The solid was washed first with a 2:1 mixture of DMF and water, then with water (2X 3vol), all at 0 ℃. The filter cake was dried under a stream of nitrogen to give light yellow AMD-2890. The purity of this compound is typically >99 area% (according to HPLC) and >99 wt% (according to NMR).

Synthesis and isolation of X4P-001

Equimolar amounts of NT-316 and AMD-2890 were reacted as described previously in acetonitrile in the presence of Diisopropylethylamine (DIPEA) and tetrabutylammonium iodide (TBAI) at 60-65 ℃. Once complete, the reaction was cooled to ambient temperature and quenched with 0.3vol ammonia, followed by the addition of 1vol water. The resulting mixture was then added to a mixture of 2vol concentrated hydrochloric acid and 3vol water. Aging in the acidic solution at 35-40 deg.C for several hours to deprotect the protected intermediate to obtain the API. Acetonitrile was subsequently removed via vacuum distillation.

At this point, 2vol of 1-butanol was added and the pH was adjusted to 12 with 20% NaOH solution. The resulting biphasic mixture was bubbled with a 10% mixture of oxygen in nitrogen at 20 ℃ for 2 hours. The pH was then adjusted to 3.0-3.5 with 18% aqueous HCl. The two phases produced were separated and the aqueous layer was washed with 1-butanol (3x3 vol). The organic layers were combined and extracted with 3vol water. All aqueous phases were then combined and 0.4% (wt) carbon was added. After 1-2 hours, the mixture was filtered and the solid was washed with 3vol water.

Toluene (7vol) and methanol (1vol) were added to the combined filtrates and the temperature was increased to 45-55 ℃ followed by adjustment of the pH to 9.5-10.0 with 20% sodium hydroxide solution. The phases were separated and the aqueous layer was extracted twice more with 3vol toluene.

All toluene layers were combined and partially concentrated via vacuum distillation at 45-50 ℃. After several additions of fresh toluene and continued vacuum distillation at 45-50 ℃ to remove additional volatile solvents, a solution of API in about 3vol toluene was obtained. This solution was heated to 60 ℃ and purged via in-line filtration.

After cooling to 50 ℃ the solution was carefully inoculated with up to 0.5 wt% of X4P-001. After the seed bed was generated, the API was allowed to crystallize stably over 2-3 hours, after which the slurry was allowed to cool slowly to 0 ℃. The resulting slurry was gradually warmed back to 30-35 ℃ and then cooled back to 0 ℃ to promote crystal growth. Finally, the slurry was filtered and the final solid was washed with toluene and dried in a drying filter at 60 ℃ under vacuum via heating for 16 hours with occasional careful stirring.

This procedure, which has been demonstrated on a 10kg scale, gives X4P-001 with 99.0-99.5 area% purity (> 99.9% enantiomeric purity) and 1337ppm residual toluene.

Comparison of Process 2 with Process 3

Table 3 provides a comparison of key process parameters and results for each batch recently produced using process 2 and process 3. As mentioned above, the 3 rd version of the process was first demonstrated on a laboratory scale and then scaled up in a plant producing X4P-001. Table 4 provides a comparison of the impurity profiles of these batches.

Most importantly, table 4 shows that switching from version 2 to version 3 processes has consistently reduced the total impurities by 0.5 to 1.1 area% and the total residual solvent in the API is significantly reduced, with no detrimental effect on other key process results, as a whole.

Table 3: comparison of key process parameters and results for each batch produced using version 2 and version 3 processes.

ND is not detected; iPAc ═ isopropyl acetate; DCM ═ dichloromethane; IPA ═ isopropyl alcohol; tol ═ toluene.

Table 4: comparison of HPLC impurity profiles for each batch produced using version 2 and version 3 processes.

Batch numbering 3-1 3-2 3-3 3-4 3-5 3-6
Version of the technology 2 2 3 3 3 3
Analysis (wt%, oab) 99.3 97.9 99.9 96.7 97.8 98.4
Purity (%) 98.8 98.3 99.46 99.40 99.28 99.51
Identified impurities (%)
RRT 0.38(de-Boc NT-316)(I-7) ND ND 0.11 0.09 0.13 0.05
RRT 1.08 (imine) (I-1) 0.62 0.95 0.10 0.13 0.04 0.09
RRT 1.21 (aldehyde) (I-6) NT* NT* 0.09 0.14 0.20 0.14
RRT 1.28 (formyl) (I-2) 0.03 ND 0.03 0.03 0.03 0.05
RRT 1.37 (acetamide) (I-5) 0.07 0.42 0.02 0.04 0.06 ND
RRT 1.93 (benzimidazole) (I-3) 0.01 ND 0.06 0.05 0.04 0.04
Total impurities (%) 1.20 1.67 0.54 0.60 0.72 0.49

ND is not detected.

NT was not tested.

For both batches, it was not known whether any aldehyde impurities were present in the batches.

Improvement of residual solvent level in Process 3

During early clinical development of X4P-001 (formerly AMD110170), the drug substance was isolated directly as the free base using ethyl acetate as the crystallization solvent (process 1). However, difficulties were encountered in removing ethyl acetate from the final API, and the development of batches was laborious, including milling and exposure to hot nitrogen, in order to reduce the levels below the International Conference on harmony (ICH) limit for NMT 5000 ppm. Early efforts in process 1 also used isopropyl acetate as the separation solvent in several batches of API during AMD-110170 development.

In a later development, the Paraben (PHB) of AMD-11070 was isolated as an intermediate and subsequently converted to the free base (Process 2). We have used this process to manufacture drug substances for recent clinical trials. In process 2, after the salt is released using isopropyl acetate instead of ethyl acetate, the free base is isolated. Residual solvent in such API showed similarly high levels as seen in the API prepared via process 1. In addition, we have found that isopropyl acetate is involved in the production of acetamide impurities during production. Thus, a change in the final API-isolation solvent is warranted.

Our recent process development has provided a direct free base isolation procedure (PHB salt is no longer an intermediate in the process) which now uses toluene instead of isopropyl acetate as the final crystallization solvent (process 3). We found that toluene did not react with X4P-001 and thus provided a more suitable separation solvent. As noted above, the exclusion of the use of isopropyl acetate also results in a reduction in the level of impurities. A summary of the residual solvent content and its residual crystallization solvent content in each batch X4P-001 is shown in table 5.

Table 5: residual isolated solvent content in X4P-001 during development

API batches were initially crystallized from one batch using isopropyl acetate and reprocessed using toluene at the final crystallization stage, so both isopropyl acetate (iPAc) and toluene were analyzed in this batch.

Batch 3-1.

Historically, the average residual solvent content of the principal crystallization solvent in the X4P-001 batch has been shown to be in the range of 1971ppm to 9298 ppm. We have set specifications for residual toluene in the X4P-001 free base using the PDE procedure.

Assuming a daily dose of 600mg X4P-001 (exceeding an exemplary clinical dose of 400mg per day to a 50% safety margin), the calculated toluene content in the X4P-001API was no more than 4500 ppm. Thus, in certain embodiments of the invention, the residual toluene specification in X4P-001 does not exceed 4500 ppm. Thus, in some embodiments, the present invention provides X4P-001 compositions comprising no more than 4500ppm toluene or 1350ppm toluene.

Conclusion

In summary, the above-described improvements of the 3 rd edition process already possess a more robust and reproducible final process step compared to the 2 nd edition. Most importantly, the purity of the isolated API X4P-001 was significantly improved compared to previous process versions. Importantly, we have found that the choice of solvent for the treatment and isolation of X4P-001 is critical to achieving the improved impurity profile described above. Specifically, we have replaced the dichloromethane and isopropyl acetate used to treat and isolate X4P-001 in the version 2 process with 1-butanol and a toluene/methanol mixture, respectively, in the version 3. We have found that in batches produced using the version 2 process, there are certain impurities present in the final product that are increased in level due to the reaction of API, X4P-001 with dichloromethane and isopropyl acetate. Another key finding is that the version 2 process forms gums and oils of the API during crystallization of the final product due to the different (and often poorer) quality of the AMD-2890 starting material. As mentioned above, we have introduced an improved AMD-2890 isolation procedure that consistently produces high quality AMD-2890 and avoids the formation of gums and oils from the API.

Example 10: mutagenicity assessment

We have performed a mutagenesis risk assessment for the synthetic process that produced X4P-001, all process intermediates, potential and actual impurities. The evaluation includes an examination of all raw materials, including key starting materials, as well as potential and actual process impurities and degradation products. All evaluations were performed according to ICH M7(R1) guidelines.

Scheme 10 and scheme 11 below depict synthetic process 2 and process 3, respectively, for X4P-001. Scheme 12 depicts the synthesis of NT-319 and AMD-2890 process intermediates.

Table 6 below depicts the identified potential mutagenic impurities in the X4P-001 manufacturing process.

A process 10: synthesis of X4P-001, Process 2.

Scheme 11: X4P-001, synthesis process 3.

And (3) a process 12: synthesis of NT-319 and AMD-2890.

Table 6: identified potential mutagenic impurities in the X4P-001 synthesis process.

The expected duration of treatment in the clinical trial for the patient is less than 10 years. In ICH M7(R1), an acceptable intake of drug for exposure periods >1 to 10 years was 10 μ g of mutagenic impurity per day, according to table 2.

An upper limit of 25ppm of potential mutagenic impurity was acceptable according to X4P-001 at a dose of 400mg per day (10 μ g/day/400 mg-25 ppm (μ g/g)).

The potential impurities I-10 and I-11 were never detected in X4P-001. In addition, the theoretical purification coefficients of synthesis process 3, according to published references such as Tesdale et al 2013,17,221, organic Process Res.Dev., show that the process effectively purifies potential impurities I-10 and I-11, with a theoretical purification coefficient of about 9x108(I-10) and about 9x1010(I-11)。

From the calculated decontamination factor, the theoretical residual amounts of I-10 and I-11 in X4P-001 have been calculated to be about <0.0001ppm I-10 and about <0.000001ppm I-11, indicating that both potentially mutagenic impurities are effectively decontaminated by this process and do not require control over X4P-001.

The potential impurities RM-396 and AMD-2890(I-9) were never detected in X4P-001 (limit of quantitation or LOQ: 2.5ppm), indicating that both potential mutagenic impurities were effectively purified by this process.

Example 11: solid formulations of 25mg, 100mg and 200mg

During formulation development, excipients were selected according to screening studies involving short-term compatibility of different excipients. For capsules of 25mg and lower strength, microcrystalline cellulose was chosen as the diluent/filler. For the 100mg higher strength formulation, dibasic calcium phosphate dihydrate diluent/filler was added to the formulation to improve flowability. The ratio of microcrystalline cellulose to dibasic calcium phosphate dihydrate is selected so as to approximate the bulk density of the drug substance, thereby reducing the chance of segregation during mixing. Sodium stearyl fumarate was chosen as lubricant. To facilitate capsule filling on an automated capsule filling machine, the slip agent colloidal silicon dioxide was added to the formulation as a glidant. Croscarmellose sodium is selected as a disintegrant to achieve automated capsule filling. Sodium lauryl sulfate is added to the formulation to reduce sticking of the drug substance during encapsulation and to act as a wetting agent.

Table 7: exemplary compositions of X4P-00125 mg capsules

Components Reference standard Function(s) Quantity (mg/capsule) %w/w
X4P-001 composition Inner part Active ingredient 25.0 14.7
Microcrystalline cellulose NF Diluent 132.7 78.1
Croscarmellose sodium NF Disintegrating agent 10.2 6.0
Stearyl fumarate sodium salt NF Lubricant agent 1.7 1.0
Colloidal silica USP Glidants 0.4 0.2
Sum of 170.0 100.0
No. 1 hard gelatin capsule USP Package with a metal layer NA NA

Table 8: composition of X4P-001100 mg capsule

Table 9: composition of X4P-001200 mg capsule

Excipient compatibility has been demonstrated by ongoing long-term stability studies of drug products that meet stability requirements in 3 batches of 100mg capsules under specified refrigerated storage conditions. X4P-001100 mg capsule lot 15K227, packaged in 30-count amber glass bottles and sealed in aluminum foil bags, gave results of up to 24 months stability under storage conditions of 5 ℃. + -. 3 ℃ and 25 ℃/60% RH. The results of any stability test parameters (assay, impurities, dissolution, moisture and microbiological tests) up to 24 months showed no significant trend under the recommended storage conditions of 5 ℃ ± 3 ℃.

The stability results for up to 9 months for 100mg capsules stored at 5 ℃ ± 3 ℃ and packaged in amber glass bottles (sealed in aluminum foil bags) or Oxy-Guard HDPE bottles show an increase in imine impurities over time, but well within the specification range. No trend from the initial value was observed for any of the other stability test parameters.

The up to 3 month stability results for 100mg capsules stored at 5 ℃ ± 3 ℃ and 25 ℃/60% RH and packaged in Oxy-Guard HDPE bottles show an increase in imine impurities over time, but well within specification. No trend from the initial value was observed for any of the other stability test parameters. These capsules were produced using the X4P-001 drug substance, which was manufactured using a toluene reprocessing procedure.

Continued use of refrigerated cold chain storage is a desirable requirement for X4P-001 drugs. The primary packaging container for X4P-001 drugs used in phase 3 clinical trials was an Oxy-Guard 60cc HDPE bottle with an inductively sealed 33mm lid. A coil of rayon was placed over the capsules in each bottle and one desiccant packet (0.5g Sorb-It or equivalent) was positioned on top of each bottle, between the coil of rayon and the lid (count 30).

A detailed overview of the manufacturing process is provided in fig. 1.

90页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:微生物混合物、从其衍生的分子、及其使用方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!