Method for treating malignant rhabdoid tumor of ovary and ovarian small cell carcinoma with hypercalcemia

文档序号:473792 发布日期:2021-12-31 浏览:20次 中文

阅读说明:本技术 治疗卵巢恶性横纹肌样瘤和高钙血症型卵巢小细胞癌的方法 (Method for treating malignant rhabdoid tumor of ovary and ovarian small cell carcinoma with hypercalcemia ) 是由 S·盖特利 于 2020-03-25 设计创作,主要内容包括:本公开提供一种在需要其的受试者中治疗恶性横纹肌样瘤的方法,所述方法包括向所述受试者施用治疗有效量的(S)-N-羟基-2-(2-(4-甲氧基苯基)丁酰胺基)噻唑-5-甲酰胺以及其药学上可接受的盐。在所述方法的某些实施方案中,所述恶性横纹肌样瘤是高钙血症型卵巢小细胞癌(SCCOHT)。(The present disclosure provides a method of treating a malignant rhabdoid tumor in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, and pharmaceutically acceptable salts thereof. In certain embodiments of the methods, the malignant rhabdoid tumor is hypercalcemia-type ovarian Small Cell Cancer (SCCOHT).)

1. A method of treating Malignant Rhabdoid Tumor (MRT), ovarian Malignant Rhabdoid Tumor (MRTO), and/or hypercalcemic ovarian Small Cell Cancer (SCCOHT) in a subject in need thereof, the method comprising:

administering to the subject a therapeutically effective amount of N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof.

2. The method according to claim 1, wherein (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject.

3. The method of claim 2, wherein the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally.

4. The method according to claim 2 or 3, wherein the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered at a dose of from 1 mg/kg/day to 1600 mg/kg/day.

5. The method of any one of the preceding claims, wherein the N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemically protected form thereof, is administered at a dose of about 100,200, 400, 800, or 1600mg per day.

6. The method of any one of the preceding claims, wherein the SCCOHT is SMARCA4 negative.

7. The method of any one of the preceding claims, wherein the subject is SMARCA4 negative.

8. The method of claim 6 or 7, wherein SMARCA4 expression is assessed by a method comprising:

(a) obtaining a biological sample from the subject;

(b) contacting the biological sample or portion thereof with an antibody that specifically binds SMARCA 4; and

(c) the amount of antibody bound to SMARCA4 was detected.

9. The method of any one of claims 6 to 8, wherein SMARCA4 expression and/or function is assessed by a method comprising:

(a) obtaining a biological sample from the subject;

(b) sequencing at least one DNA sequence encoding SMARCA4 protein from the biological sample or portion thereof; and

(c) determining whether at least one DNA sequence encoding SMARCA4 protein comprises a mutation that affects expression and/or function of SMARCA4 protein.

10. The method of any one of the preceding claims, wherein the subject is less than 40 years of age, less than 30 years of age, less than 20 years of age, or between 20 and 30 years of age, inclusive.

11. The method of any one of the preceding claims, wherein the N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemically protected form thereof, prevents and/or inhibits proliferation of SCCOHT cells.

12. A method of treating SCCOHT in a subject in need thereof, wherein the method comprises:

administering to the subject a therapeutically effective amount of N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof, in an oral tablet.

13. The method according to claim 12, wherein (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject.

14. The method according to claim 13, wherein the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered at a dose of 1 mg/kg/day to 1600 mg/kg/day.

15. The method according to any one of claims 12 to 14, wherein the N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate or a chemically protected form thereof, is administered at a dose of about 100,200, 400, 800 or 1600mg per day.

16. A method of treating MRT and/or MRTO in a subject in need thereof, the method comprising

Administering to the subject a therapeutically effective amount of N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof, in an oral tablet.

17. The method according to claim 16, wherein (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to the subject.

18. The method according to claim 17, wherein the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered at a dose of 1 mg/kg/day to 1600 mg/kg/day.

19. The method according to any one of claims 16 to 18, wherein the N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate or a chemically protected form thereof, is administered at a dose of about 100,200, 400, 800 or 1600mg per day.

20. The method of any one of claims 16 to 19, wherein the MRT and/or MRTO is INI1 negative, INI1 deficient, or epithelioid sarcoma.

21. A combination, comprising:

(S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or a pharmaceutically acceptable salt thereof; and

an immune checkpoint molecule selected from the group consisting of an inhibitor of PD-1, an inhibitor of PD-L1, an inhibitor of LAG-3, an inhibitor of TIM-3, an inhibitor of CEACAM and an inhibitor of CTLA-4.

22. The combination according to claim 21, wherein the immune checkpoint molecule is an anti-PD-1 antibody molecule.

23. The combination according to claim 21, wherein the immune checkpoint molecule is an anti-PD-L1 antibody molecule.

24. The combination according to claim 21, wherein the immune checkpoint molecule is an anti-CTLA-4 antibody molecule.

25. The combination according to claim 21, wherein the immune checkpoint molecule is an anti-LAG-3 antibody molecule.

26. The combination according to claim 21, wherein the immune checkpoint molecule is an anti-TIM-3 antibody molecule.

27. The combination of claim 21, wherein the immune checkpoint molecule is an anti-CEACAM antibody molecule.

28. The combination of claim 27, wherein the immune checkpoint molecule is an antibody molecule directed against CEACAM-1, CEACAM-3 or CEACAM-5.

29. The combination according to any one of claims 21 to 28, wherein administration of the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, and the immune checkpoint molecule to a subject in need thereof provides a synergistic effect in the treatment of cancer.

Technical Field

The present disclosure relates to the field of small molecule therapy, cancer, and methods of treating rare cancer types.

Background

Hypercalcemic small cell ovarian cancer (SCCOHT) is a rare, invasive ovarian cancer that has been diagnosed in young women. SCCOHT is often fatal when it spreads beyond the ovary. SCCOHT accounts for no more than 1% of all ovarian cancer diagnoses, with less than 300 cases reported in literature to date. Estel et al, Arch Gynecol Obstet 284: 1277-82(2011) and Young et al, Am J Surg Pathol 18: 1102-16(1994). The mean age at diagnosis is 23 years and, unlike more common types of ovarian cancer patients, most of these women show early stage disease. Harrison et al, Gynecol Oncol 100: 233-8(2006). Although most patients are diagnosed with disease restricted to the ovaries, they relapse and die within 2 years after diagnosis, with a long-term survival rate of only 33% regardless of stage. Seidman, Gynecol Oncol 59: 283-7(1995). While there is no reliable adjuvant therapy to improve outcome, multi-compound chemotherapy is thought to extend survival. Estel et al, Arch Gynecol Obstet 284: 1277-82(2011) and Pautier et al, Ann Oncol 18: 1985-9(2007).

The tissue of origin is still speculative and SCCOHT is still classified as a mixed tumor by the world health organization. Most tumors are unilateral, with sizes over 10cm potentially contributing to prognosis because of the early onset of symptoms leading to stage migration. Estel et al, Arch Gynecol Obstet 284: 1277-82(2011). Histological classification can be challenging, and commonly expressed immunohistochemical markers (e.g., CD10, WT1, and calretin) can be used in conjunction with detectable statin, S100 deletion, and chromogranin expression to exclude histological analogs. Mccluggag, Adv at Pathol 11: 288-96(2004).

Recent studies have shown that the SWI/SNF (baf) chromatin remodeling complex is a major tumor suppressor, as frequent inactivating mutations in at least 7 SWI/SNF subunits have been identified in a variety of cancers. The gene of the SWI/SNF complex was found to be associated with one of the first chromatin remodeling complexes to be identified, many of the subunits of which are conserved from yeast to humans. In mammalian cells, the SWI/SNF complex is composed of 11 to 15 protein subunits including SNF5(SMARCB1) and two mutually exclusive atpases, one of BRG1(SMARCA4) or BRM (SMARCA 2). Genetic alterations in subunits of the SWI/SNF chromatin remodeling complex are key mechanisms in tumorigenesis of several cancers. In the case of rhabdoid tumors, where frequent biallelic deletion of the core SWI/SNF gene SMARCB1 may be the major driver of carcinogenesis. Importantly, up to 20% of patients with rhabdoid tumor carry germline heterozygous mutations in SMARCB1, and inactivated germline mutations of SMARCA4 in patients lacking the SMARCB1 mutation. At the somatic level, however, SMARCA4 is the SWI/SNF subunit most frequently mutated in cancer.

Although the mutation profile of SCCOHT is unclear, the similarity between SCCOHT and rhabdoid tumors (both highly invasive pediatric tumors with primitive histological features, diploid cytogenetics and sometimes familial) suggests that they may have similar molecular genetics. There is a long-felt and unmet need for effective treatments for certain cancers, such as rhabdoid tumors and SCCOHT, which may be carcinogenesis resulting in EZH2 dependency due to genetic alteration or loss of function of subunits of the SWI/SNF chromatin remodeling complex.

Disclosure of Invention

The present disclosure provides effective treatment of INI 1-negative and SMARCA 4-negative tumors (e.g., Malignant Rhabdoid Tumor (MRT) and epithelioid sarcoma). INI1 and SMARCA4 are key proteins of the SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeling complex. In certain embodiments, the MRT may be INI1 negative, INI1 deficient, SMARCA4 negative, SMARCA4 deficient, SMARCA2 negative, SMARCA2 deficient, or comprise a mutation in one or more other components of the SWI/SNF complex.

In some embodiments, the present disclosure provides a method of treating Malignant Rhabdoid Tumor (MRT), ovarian Malignant Rhabdoid Tumor (MRTO), and/or hypercalcemic ovarian Small Cell Carcinoma (SCCOHT) in a subject in need thereof, the method comprising: administering to the subject a therapeutically effective amount of N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof.

In one embodiment, (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered to a subject. In one embodiment, (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered orally.

In some embodiments, (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, is administered at a dose of 1 mg/kg/day to 1600 mg/kg/day.

In other embodiments, N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof, is administered at a dose of about 100,200, 400, 800, or 1600mg per day.

In one embodiment, SCCOHT is SMARCA4 negative. In another embodiment, the subject is negative for SMARCA 4.

In certain embodiments, SMARCA4 expression is assessed by a method comprising: (a) obtaining a biological sample from the subject; (b) contacting the biological sample or portion thereof with an antibody that specifically binds SMARCA 4; and (c) detecting the amount of said antibody bound to SMARCA 4.

In other embodiments, SMARCA4 expression and/or function is assessed by a method comprising: (a) obtaining a biological sample from the subject; (b) sequencing at least one DNA sequence encoding SMARCA4 protein from the biological sample or portion thereof; and (c) determining whether at least one DNA sequence encoding the SMARCA4 protein comprises a mutation that affects expression and/or function of SMARCA4 protein.

In yet other embodiments, the subject is less than 40 years of age, less than 30 years of age, less than 20 years of age, or between 20 and 30 years of age (inclusive). In one embodiment, the N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemically protected form thereof, prevents and/or inhibits proliferation of SCCOHT cells.

In other embodiments, the present disclosure provides methods of treating SCCOHT in a subject in need thereof, the methods comprising administering to the subject a therapeutically effective amount of N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof, in an oral tablet. In one embodiment, the subject is administered the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or a pharmaceutically acceptable salt thereof.

In some embodiments, the present disclosure provides a method of treating MRT and/or MRTO in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or an enantiomer, a pharmaceutically acceptable salt, a solvate, or a chemoprotected form thereof, in an oral tablet.

In one embodiment, the MRT and/or MRTO is INI1 negative, INI1 deficient, or epithelioid sarcoma.

In certain embodiments of the present disclosure, the MRT is ovarian Malignant Rhabdoid Tumor (MRTO), also known as hypercalcemic ovarian Small Cell Carcinoma (SCCOHT). The present disclosure provides a method of treating SCCOHT in a subject in need thereof comprising administering to the subject a therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, a pharmaceutically acceptable salt, ester, derivative, analog, prodrug, or solvate thereof.

In certain embodiments of the disclosure, the MRT is an epithelioid sarcoma. The present disclosure provides a method of treating an epithelioid sarcoma in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, a pharmaceutically acceptable salt, ester, derivative, analog, prodrug, or solvate thereof.

(S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide may be administered orally. In certain embodiments, (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide may be formulated in oral tablets.

The methods of the present disclosure for treating cancer in a subject in need thereof comprise administering to the subject a therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide. Determining an effective amount of the disclosed compounds is within the ability of those skilled in the art, especially in light of the detailed disclosure provided herein. The effective amount of a pharmaceutical composition for affecting a specific purpose and the toxicity, secretion and overall tolerability of the pharmaceutical composition can be determined in cell cultures or experimental animals by pharmacological and toxicological methods now known to the person skilled in the art or by any similar method not yet disclosed. One example is the determination of the IC of a pharmaceutical composition in an in vitro cell line or target molecule50(half maximal inhibitory concentration). Another example is the determination of LD for a pharmaceutical composition in an experimental animal50(lethal dose resulting in the death of 50% of test animals). The exact technique used to determine the effective amount will depend on factors such as the type and physical/chemical nature of the pharmaceutical compositionThe nature, nature of the test and whether the test is performed in vitro or in vivo. Determination of the effective amount of a pharmaceutical composition is well known to those skilled in the art and will be performed using data obtained from any of the tests. Determining an effective amount of a disclosed compound for addition to cancer cells also includes determining a therapeutically effective amount, including a range of effective dosages of the formulation for in vivo (including in humans) use.

The methods of the present disclosure for treating cancer include treating Malignant Rhabdoid Tumor (MRT). In preferred embodiments, the methods of the present disclosure are used to treat a subject having ovarian Malignant Rhabdoid Tumor (MRTO). MRTO may also be referred to as hypercalcemic small cell carcinoma of the ovary (SCCOHT). In certain embodiments, the MRTO or SCCOHT and/or the subject is characterized as SMARCA4 negative, SMARCA4 deficient, SMARCA2 negative, SMARCA2 deficient, or having a mutation or defect in one or more other components of the SWI/SNF complex. In certain embodiments, the MRTO or SCCOHT and/or the subject is characterized as SMARCA4 negative. In certain embodiments, the MRTO or SCCOHT and/or subject is characterized as SMARCA 4-negative or SMARCA 4-deficient; and SMARCA2 negative or SMARCA2 deficient. As used herein, a cell that is SMARCA4 negative and/or deficient in SMARCA4 contains a mutation in the SMARCA4 gene, the corresponding SMARCA4 transcript (or cDNA copy thereof), or the SMARCA4 protein that prevents transcription of the SMARCA4 gene, prevents translation of the SMARCA4 transcript, and/or reduces/inhibits the activity of the SMARCA4 protein. As used herein, a SMARCA4 negative cell contains a mutation in the SMARCA4 gene, the corresponding SMARCA4 transcript (or cDNA copy thereof), or the SMARCA4 protein that prevents transcription of the SMARCA4 gene, prevents translation of the SMARCA4 transcript, and/or reduces/inhibits the activity of the SMARCA4 protein.

The methods of the present disclosure for treating cancer include treating Malignant Rhabdoid Tumor (MRT). In some preferred embodiments, the methods of the present disclosure are used to treat a subject having an epithelioid sarcoma. In certain embodiments, the epithelioid sarcoma is characterized as SMARCA 4-negative, SMARCA 4-deficient, SMARCA 2-negative, SMARCA 2-deficient, or having a mutation or defect in one or more other components of the SWI/SNF complex. In certain embodiments, the epithelioid sarcoma and/or the subject is characterized as SMARCA4 negative. In certain embodiments, the epithelioid sarcoma and/or the subject is characterized as SMARCA 4-negative or SMARCA 4-deficient; and SMARCA2 negative or SMARCA2 deficient.

The methods of the present disclosure may be used to treat subjects that are SMARCA4 negative or have one or more cells that may be SMARCA4 negative. SMARCA4 expression and/or SMARCA4 function may be assessed by fluorescent and non-fluorescent Immunohistochemical (IHC) methods, including methods well known to those of ordinary skill in the art. In a certain embodiment, the method comprises: (a) obtaining a biological sample from a subject; (b) contacting a biological sample or portion thereof with an antibody that specifically binds SMARCA 4; and (c) detecting the amount of antibody bound to SMARCA 4. Alternatively, or in addition, SMARCA4 expression and/or SMARCA4 function may be assessed by a method comprising: (a) obtaining a biological sample from a subject; (b) sequencing at least one DNA sequence encoding SMARCA4 protein from a biological sample or portion thereof; and (c) determining whether at least one DNA sequence encoding SMARCA4 protein comprises a mutation that affects expression and/or function of SMARCA4 protein. SMARCA4 expression or function of SMARCA4 may be assessed by detecting the amount of antibody that binds to SMARCA4 and by sequencing at least one DNA sequence encoding SMARCA4 protein, optionally using the same biological sample from the subject.

The subject of the present disclosure may be a female. The subject of the present disclosure may be less than 40, 30, or 20 years old. In certain embodiments, the subject of the present disclosure may be between 20 and 30 years of age, inclusive.

In certain embodiments, the present disclosure provides a combination comprising: (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or a pharmaceutically acceptable salt thereof; and an immune checkpoint molecule selected from the group consisting of an inhibitor of PD-1, an inhibitor of PD-L1, an inhibitor of LAG-3, an inhibitor of TIM-3, an inhibitor of CEACAM, and an inhibitor of CTLA-4.

In one aspect, the immune checkpoint molecule is an anti-PD-1 antibody molecule. In another aspect, the immune checkpoint molecule is an anti-PD-L1 antibody molecule. In another aspect, the immune checkpoint molecule is an anti-CTLA-4 antibody molecule. In one aspect, the immune checkpoint molecule is an anti-LAG-3 antibody molecule. In another aspect, the immune checkpoint molecule is an anti-TIM-3 antibody molecule. In another aspect, the immune checkpoint molecule is an anti-CEACAM antibody molecule. In another aspect, the immune checkpoint molecule is an antibody molecule directed against CEACAM-1, CEACAM-3 or CEACAM-5.

In one embodiment, administration of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt thereof, and an immune checkpoint molecule to a subject in need thereof provides a synergistic effect in the treatment of cancer.

Drawings

Figure 1 depicts growth inhibition of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide on SMARCA2 and SMARCA4 double deficient cell lines (i.e., a204, G401, G402, H522, and a 427).

FIG. 2 depicts relative SMARCA2 gene expression in (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) -treated BIN-67 cells.

FIG. 3 depicts SMARCA2 protein expression in (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) -treated BIN-67 cells.

Figure 4 depicts in vivo treatment of tumors in the SCCOHT xenograft model (BIN-67) with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) for 60 days.

Figure 5 depicts in vivo treatment of tumors in a malignant rhabdoid tumor xenograft model (G401) with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) for 30 days.

FIG. 6 depicts the potent antiproliferative activity of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) on human SCCOHT series BIN67, COV434 and SCCOHT-1.

FIG. 7 depicts the activity of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) alone and in combination with anti-mPD-1 and anti-mPD-L1 antibodies in an isogenic CT-26 mouse colon cancer model.

FIG. 8 depicts RNA-Seq analysis in (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) -treated BIN67 cells.

FIG. 9 depicts measurement of increased expression of MHC class I/II genes in (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) -treated BIN67 cells.

Detailed Description

As used herein, the terms "(S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide", "(2S) -N- [5- (hydroxycarbamoyl) thiazol-2-yl ] -2- (4-methoxyphenyl) butanamide" and "GB-3103" are synonyms that refer to compounds having the following chemical structure:

as used herein, the term "treating" may include preventing and/or inhibiting the proliferation of cancer cells (including but not limited to MRTO/SCCOHT cells).

Tumors that are INI1 negative and SMARCA4 negative, e.g., Malignant Rhabdoid Tumor (MRT) and epithelioid sarcoma are severe and failing cancers. The major market worldwide has about 1,400 patients with these tumors per year, and these tumors have no established standard of care. INI1 and SMARCA4 are key proteins of the SWI/SNF complex.

Exemplary cancers include malignant rhabdoid tumor of the ovary (MRTO), also known as hypercalcemic ovarian Small Cell Carcinoma (SCCOHT).

A preferred method of treating mrto (sccoht) in a subject in need thereof comprises administering to the subject a therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide.

The (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamides of the present disclosure are effective in treating cancer caused by decreased abundance and/or function of components of the SWI/SNF chromatin remodeling complex, including, for example, decreased abundance and/or function of SMARCA 4. Other components of the SWI/SNF complex that may be tumor markers or drivers are ARID1A, ARID2, ARID1B, SMARCB1, SMARCC1, SMARCA2, or SMARCD 1. At a high level, the SWI/SNF chromatin remodeling complex uses ATP as an energy source for opening chromatin to provide a channel for gene transcription.

According to the methods of the present disclosure, "normal" cells may be used as a basis for comparison of one or more characteristics of cancer cells, including expression and/or function of SMARCA 4. As used herein, a "normal cell" is a cell that cannot be classified as part of a "cell proliferative disorder". A lack of normal cells can result in unregulated growth or abnormal growth, or both, of an undesirable condition or disease progression. Preferably, normal cells contain the wild-type sequence of the SMARCA4 gene, express the SMARCA4 transcript without mutations, and express the SMARCA4 protein without mutations, and retain all functions at normal activity levels.

As used herein, "contacting a cell" refers to conditions in which a composition of a compound or other substance is in direct contact with the cell or is sufficiently close to induce a desired biological effect in the cell.

As used herein, "treating" or "treatment" describes the management and care of a subject for the purpose of combating a disease, condition, or disorder, which comprises administering (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph, or solvate thereof, to alleviate the symptoms or complications of cancer or to eliminate cancer.

As used herein, the term "alleviating" is intended to describe the process of reducing the severity of signs or symptoms of cancer. Importantly, signs or symptoms can be alleviated without elimination. In a preferred embodiment, administration of the pharmaceutical composition of the present disclosure results in elimination of signs or symptoms, however elimination is not necessary. An effective dose is expected to reduce the severity of signs or symptoms. For example, if the severity of cancer decreases in at least one of the multiple locations, signs or symptoms of a condition (e.g., cancer) that may occur at the multiple locations are alleviated.

As used herein, the term "severity" is intended to describe the likelihood of a cancer transitioning from a precancerous or benign state to a malignant state. Alternatively, or in addition, severity is intended to describe the Cancer stage, for example according to TNM systems (accepted by the International Union approach Cancer (UICC) and American Joint Committee on Cancer (AJCC)) or other art-recognized methods. The stage of cancer refers to the degree or severity of the cancer, depending on factors such as the location of the primary tumor, the size of the tumor, the number of tumors, and the involvement of lymph nodes (spread of the cancer to lymph nodes). Alternatively, or in addition, severity is intended to describe tumor grading by art-recognized methods (see National Cancer Institute). Tumor grading is a system for classifying cancer cells based on their microscopic abnormal appearance and the rate at which tumors may grow and spread. Many factors are considered in determining tumor grade, including the structure and growth pattern of the cells. The specific factors used to determine tumor grade vary for each type of cancer. Severity also describes histological grading (also called differentiation), which refers to the extent to which tumor cells are similar to normal cells of the same tissue type (see National Cancer Institute). In addition, the severity describes the nuclear grade, which refers to the size and shape of the nucleus among tumor cells and the percentage of dividing tumor cells (see National Cancer Institute).

In another aspect of the disclosure, severity describes the degree to which a tumor secretes growth factors, degrades the extracellular matrix, becomes vascularized, loses adhesion to parallel tissues, or metastasizes. In addition, severity describes the number of locations to which the primary tumor has metastasized. Finally, severity includes the ease of treatment of different types and locations of tumors. For example, inoperable tumors, those cancers that are more accessible to multiple body systems (hematological and immunological tumors), and those cancers that are most resistant to traditional therapies are considered most severe. In these cases, extending the life expectancy and/or relieving pain, reducing the proportion of cancer cells or confining cells in one system, and improving cancer stage/tumor grade/histological grade/nuclear grade in a subject are considered to be alleviating signs and symptoms of cancer.

As used herein, the term "symptom" (symptom) is defined as an indication of disease, illness, injury, or some discomfort in the body. Individuals experiencing symptoms may feel or notice the symptoms, but others may not readily notice. Others are defined as non-healthcare professionals.

As used herein, the term "sign" is also defined as an indication of some discomfort in the body. But signs are defined as things that a doctor, nurse, or other healthcare professional can find.

Cancer is a group of diseases that can cause almost any sign or symptom. The signs and symptoms will depend on the location of the cancer, the size of the cancer, and the extent to which the cancer affects nearby organs or structures. If the cancer spreads (metastasizes), symptoms may appear in different parts of the body.

As the cancer grows, the cancer begins to press on nearby organs, blood vessels, and nerves. This stress produces some signs and symptoms of cancer. Cancer may develop in places where it does not cause any symptoms until the cancer grows to a considerable size. Ovarian cancer is considered to be a silent killer because until tumors become large or metastasize, the cancer does not produce signs or symptoms severe enough to cause medical intervention.

Cancer can also cause symptoms such as fever, fatigue, or weight loss. This may be because cancer cells consume a large portion of the body's energy supply or cancer cells release substances that alter the body's metabolism. Or the cancer may cause the immune system to respond in a manner that produces these symptoms. While the signs and symptoms listed above are the more common signs and symptoms in cancer, there are many other signs and symptoms that are not common and are not listed here. However, the present disclosure contemplates and encompasses all art-recognized signs and symptoms of cancer.

Treatment of cancer may result in a reduction in tumor size. The reduction in tumor size may also be referred to as "tumor regression". Preferably, tumor size is reduced by 5% or more after treatment according to the methods of the present disclosure relative to the size before treatment; more preferably, the tumor size is reduced by 10% or more; more preferably, a reduction of 20% or more; more preferably, a reduction of 30% or more; more preferably, a reduction of 40% or more; even more preferably, a reduction of 50% or more; most preferably, the reduction is more than 75% or more. The size of the tumor can be measured by any reproducible measurement means. The size of the tumor can be measured as the diameter of the tumor.

Treatment of cancer may result in a reduction in tumor volume. Preferably, tumor volume is reduced by 5% or more after treatment according to the methods of the present disclosure relative to the volume before tumor treatment; more preferably, tumor volume is reduced by 10% or more; more preferably, a reduction of 20% or more; more preferably, a reduction of 30% or more; more preferably, a reduction of 40% or more; even more preferably, a reduction of 50% or more; and most preferably, by more than 75% or more. The volume of the tumor can be measured by any reproducible means of measurement.

Treatment of cancer may result in a reduction in the number of tumors. Preferably, the number of tumors after treatment is reduced by 5% or more relative to the number before treatment; more preferably, the number of tumors is reduced by 10% or more; more preferably, a reduction of 20% or more; more preferably, a reduction of 30% or more; more preferably, a reduction of 40% or more; even more preferably, a reduction of 50% or more; and most preferably, by more than 75%. The number of tumors can be measured by any reproducible means of measurement. The number of tumors can be measured by counting tumors that are visible to the naked eye or at a specified magnification. Preferably, the specified magnification is 2x, 3x, 4x, 5x, 10x or 50 x.

Treatment of cancer may result in a reduction in the number of metastatic lesions in other tissues or organs distant from the primary tumor site. Preferably, the number of metastatic lesions is reduced by 5% or more after treatment according to the methods of the present disclosure relative to the number before treatment; more preferably, the number of transferred lesions is reduced by 10% or more; more preferably, a reduction of 20% or more; more preferably, a reduction of 30% or more; more preferably, a reduction of 40% or more; even more preferably, a reduction of 50% or more; and most preferably, by more than 75%. The number of metastatic lesions can be measured by any reproducible means of measurement. The number of metastatic lesions can be measured by counting the number of metastatic lesions visible to the naked eye or at a given magnification. Preferably, the specified magnification is 2x, 3x, 4x, 5x, 10x or 50 x.

An effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph, or solvate thereof, of the present disclosure is not significantly cytotoxic to normal cells. For example, a therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure is not significantly cytotoxic to normal cells if administration of a therapeutically effective amount of the (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide does not cause cell death in more than 10% of normal cells. A therapeutically effective amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure has no significant effect on the activity of normal cells if the compound is administered in a therapeutically effective amount without causing cell death in more than 10% of normal cells.

Contacting a cell with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph, or solvate thereof, can selectively inhibit HDAC activity in cancer cells. Administration of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure, or a pharmaceutically acceptable salt, prodrug, metabolite, polymorph, or solvate thereof, to a subject in need thereof, can selectively inhibit HDAC activity in cancer cells.

Malignant rhabdoid tumor

Malignant Rhabdoid Tumor (MRT) is a rare childhood tumor that occurs in soft tissues, most commonly starting from the kidneys and brain. Some hallmarks of malignant rhabdoid tumors are loss of function of SMARCB1 (also known as INI 1). INI1 is a key component of the SWI/SNF regulatory complex, a chromatin remodelling agent that acts in opposition to EZH 2. INI 1-negative tumors have altered SWI/SNF function. This activity can be targeted by (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide. INI 1-negative tumors are usually aggressive and currently poorly treated. For example, current treatments for MRT (a well-studied INI 1-negative tumor) include surgery, chemotherapy, and radiation therapy, which are associated with limited efficacy and significant treatment-related morbidity. The annual incidence of INI 1-negative tumor and synovial sarcoma patients in major markets including U.S., e.u., and japan is approximately 2,400. Loss of function of SMARCB1/INI1 also occurs in another rare and aggressive childhood tumor, atypical teratoid rhabdoid tumor of the central nervous system (AT/RT).

Malignant rhabdoid tumor of ovary MRTO (hypercalcemia type Small Cell Carcinoma of Ovary (SCCOHT))

MRTO/SCCOHT is an extremely rare invasive cancer affecting children and young women (average age at diagnosis 23 years). Over 65% of patients succumb to MRTO/SCCOHT disease within 2 years of diagnosis. Like MRT, these tumors are characterized by a gene deletion of the SWI/SNF complex subunit SMARCA 4. SMARCA 4-negative ovarian cancer cells were selectively sensitive to EZH2 inhibition with IC50 values similar to those observed in MRT cells. For example, current treatment of SCCOHT includes tumor reduction surgery and platinum-based chemotherapy, and shows a high rate of relapse. The range of differential diagnosis is wide and includes three subtypes of ovarian cancer: granular cell (sex cord stroma) tumors, dysgerminoma, and high grade serous tumors.

Standard hematoxylin and eosin (H & E) staining showed SCCOHT as striated muscle with small, close-packed, monomorphic, highly proliferative and poorly differentiated cell plate-like arrangement, while IHC showed that SCCOHT is characterized by inactivation of SMARCA4 gene resulting in protein deletion and non-mutated silencing of SMARCA2 protein. (see, e.g., Karnezis et al, J.Pathol.2016; 238: 389-. Some aspects of the present disclosure provide: tumor cells and tumors (e.g., SCCOHT tumors) that exhibit loss of SMARCA4 (e.g., due to mutation) and loss of SMARCA2 (e.g., due to protein loss) are sensitive to HDAC inhibition and thus can be effectively treated with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide.

Epithelioid sarcoma

Epithelioid sarcoma is a rare soft tissue sarcoma, accounting for less than 1% of all soft tissue sarcomas. Epithelioid sarcoma was first well characterized in 1970. The most common gene mutation found in epithelioid sarcomas is the INI-1 deletion (approximately 80-90%). Two variants of epithelioid sarcomas have been reported: distal epithelioid sarcoma is associated with a better prognosis, affecting the distal upper and lower extremities (fingers, hands, forearms, or feet), while proximal epithelioid sarcoma is associated with a worse prognosis, affecting the proximal ends (upper arms, thighs)) and trunk. Epithelioid sarcomas can occur in all age groups, but most commonly occur in young people (the median age at diagnosis is 27 years).

Epithelioid sarcoma is associated with a high recurrence rate after initial treatment, with median survival of less than 2 years when diagnosed as metastatic epithelioid sarcoma. Local recurrence and metastasis, usually to the lymph nodes, lungs, bones, and brain, occur in about 30-50% of patients. Treatment of epithelioid sarcoma includes surgical resection as a preferred treatment. The success rate of conventional chemotherapy and radiation therapy, alone or in combination, is relatively low for inoperable tumors or after recurrence. Approximately 50% of oncologists consider epithelioid sarcomas to be insensitive to chemotherapy.

The present disclosure relates to the compound (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, a pharmaceutically acceptable salt, ester, derivative, analog, prodrug, or solvate thereof.

The present disclosure encompasses any physicochemical form that (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide may take. Non-limiting examples of physicochemical forms include hydrated forms, solvated forms, crystalline (known or not yet disclosed), polymorphic crystalline and amorphous forms, and the like. Methods for producing such physicochemical forms are known to those skilled in the art.

The present disclosure also relates to a pharmaceutical composition for treating Histone Deacetylase (HDAC) related diseases. The pharmaceutical composition comprises at least a first active ingredient selected from the group consisting of: (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, and pharmaceutically acceptable salts, esters, derivatives, analogs, prodrugs, or solvates thereof.

In some aspects, (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, a pharmaceutically acceptable salt, ester, derivative, analog, prodrug, or solvate thereof, comprises 80 to 100% by weight of the first active ingredient, or any percentage range, e.g., between 85 to 100%, 85 to 99.99%, 90 to 99.9%, 92.5% to 99.5%, 95 to 99%, or 97.5 to 99%, etc. In other aspects, #1a, a pharmaceutically acceptable salt, ester, derivative, analog, prodrug, or solvate thereof, constitutes at least 80%, at least 85%, at least 90%, at least 92.5%, at least 95%, at least 97.5%, or at least 99% of the first active ingredient by weight.

Pharmaceutically acceptable salts include any salt derived from an organic or inorganic acid. Examples of such salts include, but are not limited to, the following: hydrobromic acid, hydrochloric acid, nitric acid, phosphoric acid and sulfuric acid. Organic acid addition salts include, for example, acetic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, 2- (4-chlorophenoxy) -2-methylpropionic acid, 1, 2-ethanedisulfonic acid, ethanesulfonic acid, ethylenediaminetetraacetic acid (EDTA), fumaric acid, glucoheptonic acid, gluconic acid, glutamic acid, N-glycolylarsinic acid (N-glycolarsanilic acid), 4-hexylresorcinol, hippuric acid, 2- (4-hydroxybenzoyl) benzoic acid, 1-hydroxy-2-naphthoic acid, 3-hydroxy-2-naphthoic acid, 2-hydroxyethanesulfonic acid, lactobionic acid, N-dodecylsulfuric acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, methylsulfuric acid, mucic acid, 2-naphthalenesulfonic acid, pamoic acid, Pantothenic acid, phosphatidic acid ((4-aminophenyl) phosphonic acid), picric acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, terephthalic acid, p-toluenesulfonic acid, 10-undecenoic acid, or any other salt of such an acid now known or not disclosed. Those skilled in the art will appreciate that these pharmaceutically acceptable salts may be used in the formulation of pharmacological compositions. These salts can be prepared by reacting the disclosed compounds with a suitable acid in a manner known to those skilled in the art.

In a preferred embodiment, the pharmaceutically acceptable salt of #1a is selected from: na (Na)+、K+、Mg2+、Ca2+、Zn2+And Al3 +. In a preferred embodiment, the pharmaceutically acceptable salt of #1 is selected from: na (Na)+、K+、Mg2+、Ca2+、Zn2+And Al3+

The physical form of the pharmaceutical composition depends on many factors. For example, the desired method of administration, the physicochemical form employed by the disclosed compounds or pharmaceutically acceptable salts thereof. Non-limiting examples of physical forms include solids, liquids, gases, sols, gels, aerosols, and the like. In some embodiments, the pharmaceutical composition consists of the disclosed compound or a pharmaceutically acceptable salt thereof, without any other additives.

In other embodiments, the pharmaceutical composition comprises a second active ingredient that is different in formula from (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide or N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide. In some aspects, the second active ingredient has a molecular target that is the same as or similar to the target of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide or N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide. In other embodiments, the second active ingredient acts on one or more biochemical pathways upstream of the molecular target of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide or N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide. In still other embodiments, the second active ingredient acts on one or more biochemical pathways downstream of the molecular target of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide or N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide. Pharmaceutical compositions comprising the disclosed compounds may be prepared using methods well known in the pharmaceutical arts.

In some embodiments, the pharmaceutical composition includes a material capable of modifying the physical form of the dosage unit. In one non-limiting example, the composition includes a material that forms a coating that holds the compound. Non-limiting examples of materials include sugar, shellac, gelatin, and other inert coating agents.

The present invention relates to a method of treating a Histone Deacetylase (HDAC) related disease in a subject, comprising administering to the subject a composition selected from (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide or N-hydroxy-2- (2- (4-methoxyphenyl) butanamide) thiazole-5-carboxamide and pharmaceutically acceptable salts, esters, derivatives, analogs, prodrugs or solvates thereof.

Histone Acetyltransferases (HATs) affect gene expression by controlling the curling and decurling of DNA around histones. Histone acetyltransferases accomplish this by acetylating lysine residues in the core histone, resulting in decreased chromatin tightness and increased transcriptional activity. In contrast, Histone Deacetylase (HDAC) removes acetyl groups from lysine residues, resulting in tighter chromatin and transcriptional silencing. Reversible modification of the terminal tail of the core histone constitutes a major epigenetic mechanism for remodeling the higher chromatin structure and controlling gene expression. HDAC inhibitors (HDI) block this effect and can lead to hyperacetylation of histones, thereby affecting gene expression. Thagalingem s., Cheng K H, Lee H J et al, ann.n.y.acad.sci.983: 84-100,2003; marks P A. Richon V M, Rifkind R A, J.Natl.cancer Inst.92(15)1210-16, 2000; dokmanovic M, Clarke C., Marks P A, mol. cancer Res.5(10) 981-.

Histone Deacetylase (HDAC) inhibitors are a class of cytostatics that inhibit the proliferation of tumor cells in culture and in vivo by inducing cell cycle arrest, differentiation and/or apoptosis. Acetylation and deacetylation play important roles in the regulation of chromatin topology and the regulation of gene transcription. Histone deacetylase inhibitors induce the aggregation of over-acetylated nucleosome core histones in many regions of chromatin, but affect the expression of only a small subset of genes, which results in transcriptional activation of some genes but inhibition of equal or greater numbers of other genes. Non-histone proteins (e.g. transcription factors) are also targets for acetylation with different functional roles. Acetylation enhances the activity of some transcription factors (e.g., tumor suppressor p53 and erythroid differentiation factor GATA-1), but may inhibit the transcriptional activity of other transcription factors (including T-cell factor and co-activator ACTR). Recent studies have shown that estrogen receptor α (era) can be over-acetylated in response to histone deacetylase inhibition, thereby inhibiting ligand sensitivity and regulating transcriptional activation by histone deacetylase inhibitors. The conservation of the acetylated ER α motif in other nuclear receptors suggests that acetylation may play an important regulatory role in a variety of nuclear receptor signaling functions. In animal models, many structurally diverse histone deacetylase inhibitors exhibit potent antitumor efficacy in vivo with little toxicity. Several compounds are currently in the early clinical development stage as potential treatments for solid and hematological cancers, as monotherapies and in combination with cytotoxins and differentiating agents.

The HDAC enzyme family constitutes a family of 18 genes, which can be divided into four subclasses based on homology to the corresponding yeast orthologs (orthologs): class I to IV. HDACs belonging to I, II and IV comprise 11 members, HDAC isoforms (isoforms) 1 to 11, commonly referred to as classical HDACs, are metal-dependent hydrolases. Class III HDACs comprising 7 members are known as sirtuins (i.e., sirts 1 to 7) are NAD + dependent hydrolases. Class I HDACs are nuclear proteins with ubiquitous tissue expression. Class II and IV HDACs are found both in the nucleus and cytoplasm and exhibit tissue-specific expression. The class II HDAC family is further subdivided into subclasses IIA and IIB. Class IIA contains the isoforms HDAC4, HDAC5, HDAC7 and HDAC9, while class IIB contains the isoforms HDAC6 and HDAC 10. HDAC6 comprises two sirtuin domains in tandem and a C-terminal zinc finger domain. HDAC10 is structurally related to HDAC6, but has an additional catalytic domain. Table 1 shows the cellular location and tissue expression of classical HDACs (adapted from Witt, O. et al, Cancer Lett., 277: 8-21 (2008)).

TABLE 1 classical HDAC, cell location and tissue expression

Classification Isoforms Cell location Tissue expression
Class I HDAC1 Core Ubiquitous
HDAC2 Core Ubiquitous
HDAC3 Core Ubiquitous
HDAC8 Nucleus/cytoplasm Ubiquitous
Class IIA HDAC4 Nucleus/cytoplasm Heart, smooth muscle, brain
HDAC5 Nucleus/cytoplasm Heart, smooth muscle, brain
HDAC7 Nucleus/cytoplasm Heart, placenta, pancreas, smooth muscle,
HDAC9 Nucleus/cytoplasm Smooth muscle and brain
Class IIB HDAC6 Cytoplasm of cells Kidney, liver, heart, pancreas
HDAC10 Cytoplasm of cells Spleen, kidney and liver
Class IV HDAC11 Nucleus/cytoplasm Heart, smooth muscle, kidney, brain

HDACs play an important role in both normal and abnormal cell proliferation and differentiation. HDACs have been associated with several disease states involving proliferation, including but not limited to cell proliferative diseases and conditions, such as various forms of cancer. (reviewed in Witt, O. et al, Cancer Lett., 277: 8-21 (2008); and Portella A. et al, nat. Biotechnol., 28: 1057-. Class I and II HDACs have been identified as attractive targets for anti-cancer therapy. In particular, different class I and class II HDAC proteins are overexpressed in several cancers, including ovarian cancer (HDAC1-3), gastric cancer (HDAC2), and lung cancer (HDAC1 and 3), among others. In addition, a possible correlation between HDAC8 and Acute Myeloid Leukemia (AML) has been proposed. For class II HDAC proteins, aberrant expression of HDAC6 was induced in some breast cancer cells. Based on the clinical effects of HDAC inhibitors, HDAC inhibitors have been identified that inhibit tumor cell proliferation, induce cell differentiation, and up-regulate key genes associated with anti-cancer efficacy. HDACs also relate to various types of cancer (Bali P, et al, "infection of tissue deacylase 6 acylates and disorders the tissue function of heat shock protein 90: A novel tissue activity of tissue deacylase inhibitors," J.biol.Chem., 2005280: 26729-, Protozoal diseases (see, e.g., U.S. patent No. 5,922,837) and viral diseases (Margolis, d.m. et al, curr. opin. hiv AIDS, 6: 25-29 (2011)).

In recent years, efforts have been made to develop HDAC inhibitors as cancer treatments and/or as adjunctive therapies. Mark P A. et al Expert Opinion on Investigational Drugs 14 (12): 1497-1511(2005). The exact mechanism by which a compound may act is not known, but epigenetic pathways have been investigated to help elucidate the exact biological pathway. Claude Monneret, Anticancer Drugs 18 (4): 363-3702007. For example, HDAC inhibitors have been shown to induce p21(WAFI) expression, said p21 being a modulator of p53 tumor suppressor activity. Rochon V m. et al, proc.natl.acad.sci.u.s.a.97 (18): 10014-10019,2000. HDACs are involved in the pathway by which retinoblastoma protein (pRb) inhibits cell proliferation. The pRb protein is part of a complex that directs HDACs to chromatin such that the HDACs deacetylate histones. Brehm a. et al, Nature 391 (6667): 597-601,1998. HDAC1 down-regulates the cardiovascular transcription factor Kruppel-like factor 5 through direct interactions. Matsumura t. et al, j.biol.chem.280 (13): 12123-12129,2005. Estrogen is known as a mitotic factor that is involved in the tumorigenesis and progression of breast cancer by binding to the estrogen receptor α (era). Recent data indicate that chromatin inactivation mediated by HDAC and DNA methylation is an important component of era silencing of its human breast cancer cells. Zhang z, et al, break Cancer res. treat.94 (1): 11-16,2005.

In some aspects, the composition is administered at 10 to 400mg/kg, or at any value between, e.g., 10 to 350mg/kg, 20 to 300mg/kg, 30 to 250mg/kg, 40 to 200mg/kg, 50 to 150mg/kg, 60 to 150mg/kg, or 60 to 100mg/kg, etc.

In other aspects, the composition is administered about every 4, 8, 12, 16, or 24 hours. In yet other aspects, the composition is administered every 1 to 24 hours or any number between, for example, every 2 to 24 hours, 2 to 18 hours, 3 to 16 hours, 4 to 12 hours, 5 to 8 hours, etc.

In some embodiments, the composition further comprises a second active ingredient selected from the group consisting of chemotherapeutic agents, EZH2 inhibitors, receptor tyrosine kinase inhibitors, CDK4/6 inhibitors, agents that enhance antigen presentation ("antigen presenting combinations"), agents that enhance effector cell responses ("effector cell combinations"), agents that reduce tumor immunosuppression ("anti-tumor immunosuppressive combinations"), and combinations thereof.

Non-limiting examples of chemotherapeutic drugs include: cis-diamminedichloroplatinum (II) (cisplatin), doxorubicin, 5-fluorouracil, paclitaxel and topoisomerase inhibitors such as etoposide, teniposide, irinotecan, topotecan and the like. Non-limiting examples of EZH2 inhibitors include tasetastat (EPZ-6438). Non-limiting examples of receptor tyrosine kinase inhibitors include ponatinib. Non-limiting examples of CDK4/6 inhibitors include rebocillin (Ribociclib), Palbociclib (Palbociclib) (PD-0332991), Abemaciclib (LY2835219), and Trilaciclib (G1T 28).

Antigen presenting combination

Non-limiting examples of agents that enhance antigen presentation include: an agent that enhances antigen presentation, an agent that enhances tumor cell lysis, an agent that stimulates phagocytes, an agent that relieves phagocyte inhibition, an agent that activates dendritic cells, an agent that activates macrophages (e.g., macrophage I), an agent that recruits dendritic cells, or an agent or vaccine that recruits macrophages (e.g., macrophage I), and the like. In certain non-limiting aspects, the agent that enhances antigen presentation enhances tumor antigen presentation.

Non-limiting examples of vaccines include: cell-based vaccines (e.g., dendritic cell-based vaccines, such as provenge.rtm) or antigen-based vaccines (e.g., IL-2 in combination with MUC 1), and the like. A non-limiting example of an agent that enhances tumor cell lysis is an oncolytic virus. Non-limiting examples of agents that stimulate phagocytes are type I Interferon (IFN) activators, e.g., TLR agonists or RIG-I like receptor agonists (RLR) and the like. Non-limiting examples of agents that activate and/or recruit dendritic cells or macrophages include: bispecific cell cement or trispecific cell cement, etc.

In some embodiments, the agent that enhances antigen presentation is selected from the group consisting of agonists of stimulators of Interferon Genes (stimulants of Interferon Genes) (STING agonists), agonists of Toll-like receptors (TLRs), TIM-3 modulators, Vascular Endothelial Growth Factor Receptor (VEGFR) inhibitors, c-Met inhibitors, TGF-beta inhibitors, IDO/TDO inhibitors, A2AR antagonists, oncolytic viruses, vaccines, bispecific cellular binding agents, trispecific cellular binding agents, bispecific antibody molecules, trispecific antibody molecules, IDO/TDO inhibitors, and combinations thereof.

Non-limiting examples of TLRs include: agonists for TLR-3, TLR-4, TLR-5, TLR-7, TLR-8 or TLR-9, and the like. A non-limiting example of a TIM-3 modulator is an anti-TIM-3 antibody molecule. A non-limiting example of a TGF- β inhibitor is an anti-TGF- β antibody. A non-limiting example of a vaccine is the scaffold vaccine (scaffold vaccine). In some aspects, the oncolytic virus expresses a cytokine, e.g., GM-CSF or CSF (e.g., CSFl or CSF2), or the like. Non-limiting examples of bispecific or trispecific cell conjugates include: bispecific or trispecific antibody molecules with or without Fc domains directed against CD47 and CD 19.

Effector cell combinations

Non-limiting examples of agents that enhance effector cell responses include: lymphocyte activators, agents that activate and/or release the inhibition of Tumor Infiltrating Lymphocytes (TILs), NK cell modulators, interleukins or interleukin variants, bispecific or trispecific cell binding agents, NK cell therapies, NK cell inducing vaccines and antigen/immunostimulants, immunomodulators, T cell modulators, bispecific T cell binding agents, inhibitors of IAP (inhibitors of Apoptosis proteins) or of the target of rapamycin (mTOR) and the like.

Non-limiting examples of lymphocyte activators include: NK cell activator or T cell activator, etc. Non-limiting examples of Tumor Infiltrating Lymphocytes (TILs) include: NK cells or T cells, etc. Non-limiting examples of NK cell modulators are modulators (e.g., antibody molecules) of NK receptors, e.g., modulators of NKG2A, KIR3DL, NKp46, MICA, CEACAM1, or combinations thereof, and the like. Non-limiting examples of interleukins include: IL-2, IL-15, IL-21, IL-13R, IL-12 cytokines or combinations thereof and the like. Non-limiting examples of bispecific or trispecific cell-binding agents include: bispecific antibody molecules of NKG2A and CD138, or bispecific antibody molecules of CD3 and TCR, etc. Non-limiting examples of immunomodulators include: activators of co-stimulatory molecules, or inhibitors of immune checkpoint molecules, and the like.

In some embodiments, the T cell modulator is a T cell modulator selected from inhibitors of checkpoint inhibitors. Non-limiting examples of T cell modulators selected from inhibitors (e.g., antibodies) of checkpoint inhibitors include: inhibitors of PD-1, inhibitors of PD-L1, inhibitors of TIM-3, inhibitors of LAG-3, inhibitors of VISTA, inhibitors of Diacylglycerol Kinase (DKG) - α, inhibitors of B7-H3, inhibitors of B7-H4, inhibitors of TIGIT, inhibitors of CTLA4, inhibitors of BTLA, inhibitors of CD160, inhibitors of TIM1, inhibitors of IDO, inhibitors of LAIR1, inhibitors of IL-12, or combinations thereof, and the like.

In other embodiments, the T cell modulator is a T cell modulator selected from an agonist or activator of a costimulatory molecule. Non-limiting examples of T cell modulators selected from agonists or activators of costimulatory molecules include: an agonistic antibody, an antigen-binding fragment thereof, or a soluble fusion thereof, such as a GITR, OX40, ICOS, SLAM (e.g., SLAMF7), HVEM, LIGHT, CD2, CD27, CD28, CDS, ICAM-1, LFA-1(CD11a/CD18), ICOS (CD278), 4-1BB (CD137), CD30, CD40, BAFFR, CD7, NKG2C, NKp80, CD160, B7-H3, or CD83 ligand. Non-limiting examples of bispecific T cell binders are bispecific antibody molecules that bind CD3 and tumor antigens (e.g., Epidermal Growth Factor Receptor (EGFR), PSCA, PSMA, EpCAM, or HER2, etc.).

Anti-tumor immunosuppressive combinations

Non-limiting examples of agents that reduce tumor immunosuppression include: agents that modulate the activity and/or levels of tregs, macrophage 2 and/or MDSCs, agents that increase M2 polarization, Treg depletion and/or T cell recruitment.

Non-limiting examples of agents that reduce tumor immunosuppression include: immunomodulators, CSF-1/1R inhibitors, IL-17 inhibitors, IL-1 beta inhibitors, CXCR2 inhibitors, phosphoinositide 3-kinase inhibitors, BAFF-R inhibitors, MALT-1/BTK inhibitors, JAK inhibitors, CRTH2 inhibitors, VEGFR inhibitors, IL-15 or variants thereof, CTLA-4 inhibitors, IDO/TDO inhibitors, A2AR antagonists, TGF-beta inhibitors or PFKFB3 inhibitors, inhibitors of immune checkpoint molecules and the like.

Non-limiting examples of immunomodulators include: activators of co-stimulatory molecules (e.g., GITR agonists) or inhibitors of immune checkpoint molecules (e.g., PD-1, PD-L1, LAG-3, TIM-3, or CTLA-4, etc.), and the like. A non-limiting example of a CSF-1/1R inhibitor is an inhibitor of macrophage colony stimulating factor (M-CSF). Non-limiting examples of inhibitors of phosphoinositide 3-kinases are PI3K, such as PI3K γ or PI3K δ and the like. Non-limiting examples of immune checkpoint molecule inhibitors include: an inhibitor of PD-1, an inhibitor of PD-L1, an inhibitor of LAG-3, an inhibitor of TIM-3, an inhibitor of CEACAM (e.g., CEACAM-1, CEACAM-3 and/or CEACAM-5, etc.), an inhibitor of CTLA-4, or the like.

In some embodiments, the second active ingredient comprises one or more therapeutic agents that enhance antigen presentation, one or more therapeutic agents that enhance effector cell responses, and/or one or more therapeutic agents that reduce tumor immunosuppression.

In certain embodiments, the second active ingredient is selected from the group consisting of STING agonists, TLR agonists (e.g., TLR7 agonists), TIM-3 modulators (e.g., TIM-3 inhibitors), GITR modulators (e.g., GITR agonists), PD-1 inhibitors (e.g., anti-PD-1 antibody molecules), PD-L1 inhibitors, CSF-1/1R inhibitors (e.g., M-CSF inhibitors), IL-17 inhibitors, IL-1 β inhibitors, and combinations thereof.

Pharmaceutical preparation

The present disclosure also provides pharmaceutical compositions comprising (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide as described herein in combination with at least one pharmaceutically acceptable excipient or carrier.

A "pharmaceutical composition" is a formulation containing (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure in a form suitable for administration to a subject. In one embodiment, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form is in any of a variety of forms including, for example, capsules, IV bags, tablets, a single pump on an aerosol inhaler, or a vial. The amount of active ingredient (e.g., a formulation of a disclosed compound or a salt, hydrate, solvate, or isomer thereof) in a unit dose of the composition is an effective amount and varies with the particular treatment involved. Those skilled in the art will appreciate that routine variations in dosage are sometimes required depending on the age and condition of the patient. The dosage will also depend on the route of administration. A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalation, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for topical or transdermal administration of the compounds of the present disclosure include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In one embodiment, the active compound is mixed under sterile conditions with a pharmaceutically acceptable carrier, and any preservatives, buffers, or propellants which may be required.

As used herein, the phrase "pharmaceutically acceptable" refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

"pharmaceutically acceptable excipient" refers to an excipient that can be used in the preparation of pharmaceutical compositions, which are generally safe, non-toxic, and biologically or otherwise non-adverse, and include acceptable excipients for veterinary use as well as human pharmaceutical use. As used in this disclosure, "pharmaceutically acceptable excipient" includes one and more than one such excipient.

The pharmaceutical compositions of the present disclosure are formulated to be compatible with their intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), and transmucosal administration. Solutions or suspensions for parenteral, intradermal, or subcutaneous application may include the following ingredients: sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents, such as, for example, benzyl alcohol or methyl paraben; antioxidants, such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for adjusting tonicity, for example sodium chloride or dextrose. The pH can be adjusted with an acid or base (e.g., hydrochloric acid or sodium hydroxide). The parenteral formulations may be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.

The compounds or pharmaceutical compositions of the present disclosure can be administered to a subject by a number of well-known methods currently used for chemotherapy. For example, to treat cancer, a compound of the present disclosure may be injected directly into a tumor, injected into the bloodstream or body cavity, or administered orally or transdermally with a patch. The dosage selected should be sufficient to constitute an effective treatment and not so high as to cause unacceptable side effects. During and for a reasonable period of time after treatment, the status of the disease condition (e.g., cancer, pre-cancer, etc.) and the health of the patient should preferably be closely monitored.

The term "therapeutically effective amount" as used herein refers to an amount of (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, a composition or pharmaceutical composition thereof that is effective for treating, ameliorating, or preventing the identified disease or condition, or that exhibits a detectable therapeutic or inhibitory effect. The effect may be detected by any assay known in the art. The precise effective amount for a subject will depend on the weight, size (size) and health of the subject; the nature and extent of the pathology; and selecting a therapeutic agent or combination of therapeutic agents for administration. A therapeutically effective amount for a given situation can be determined by routine experimentation within the skill and judgment of the clinician. In a preferred aspect, the disease or condition to be treated is cancer, including but not limited to, Malignant Rhabdoid Tumor (MRT), ovarian MRT (mrto), and hypercalcemic small cell ovarian cancer (SCCOHT).

For (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure, a therapeutically effective amount may be estimated initially in a cell culture assay (e.g., a culture assay of neoplastic cells) or in an animal model (typically rat, mouse, rabbit, dog, or pig). Animal models can also be used to determine appropriate concentration ranges and routes of administration. Such information can then be used to determine useful doses and routes for administration to humans. Therapeutic/prophylactic efficacy and toxicity can be measured by standard pharmaceutical procedures in cell cultures or experimental animals, e.g. ED50(dose therapeutically effective in 50% of the population) and LD50(dose lethal to 50% of the population). The dose ratio of toxic to therapeutic effect is the therapeutic index, which can be expressed as the ratio LD50/ED50. Pharmaceutical compositions exhibiting a large therapeutic index are preferred. The dosage may vary within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration.

The dosage and administration are adjusted to provide a sufficient level of the active agent or to maintain the desired effect. Factors that may be considered include the severity of the disease state, the general health of the subject, the age, weight and sex of the subject, diet, time and frequency of administration, drug combination, response sensitivity and tolerance/response to treatment. Long-acting pharmaceutical compositions may be administered every 3 to 4 days, weekly, or biweekly, depending on the half-life and clearance of a particular formulation.

The pharmaceutical compositions of the present disclosure containing (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide may be manufactured in a generally known manner, for example by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions may be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and/or auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Of course, the appropriate formulation will depend on the route of administration chosen.

Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL.TM(BASF, Parsippany, n.j.) or Phosphate Buffered Saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms (e.g., bacteria and fungi). The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. Proper fluidity can be maintained, for example, by the use of a coating (e.g., lecithin), by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. The action of microorganisms can be prevented by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it may be preferable to include isotonic agents, for example, sugars, polyalcohols (e.g., mannitol, sorbitol, or sodium chloride) in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.

Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those described above. In the case of sterile powders for the preparation of sterile injectable solutions, the methods of preparation are vacuum drying and freeze-drying to yield a powder of the active ingredient plus any other desired ingredient from a previously sterile-filtered solution thereof.

Oral compositions typically include an inert diluent or an edible pharmaceutically acceptable carrier. It may be encapsulated in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compounds can be incorporated with excipients and used in the form of tablets, dragees or capsules. Oral compositions can also be prepared for use as a mouthwash using a liquid carrier, wherein the compound in the liquid carrier is administered orally and swished (swish) and expectorated or swallowed. Pharmaceutically compatible binding agents and/or adjuvant materials may be included as part of the composition. Tablets, pills, capsules, lozenges and the like may contain any of the following ingredients or compounds of similar nature: binders, for example, microcrystalline cellulose, gum tragacanth or gelatin; excipients, for example, starch or lactose; disintegrating agents, for example, alginic acid, primary gel (Primogel), corn starch, and the like; lubricants, for example, magnesium stearate or Sterotes; glidants, e.g., colloidal silicon dioxide; sweetening agents, for example, sucrose or saccharin; or a flavoring agent, for example, peppermint, methyl salicylate, or orange flavoring.

For administration by inhalation, the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser, or a nebulizer, with a suitable propellant, e.g., a gas such as carbon dioxide.

Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be achieved through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as is well known in the art.

The active compound (i.e., (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide) of the present disclosure may be prepared with a pharmaceutically acceptable carrier that protects the compound from rapid clearance from the body, e.g., a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable biocompatible polymers may be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparing these formulations will be apparent to those skilled in the art. These materials are also commercially available from Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells using monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No.4,522,811.

It is particularly advantageous that the oral or parenteral compositions be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suitable as unitary dosages for the subject to be treated; each unit containing a predetermined amount of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specifications for the dosage unit forms of the present disclosure are dependent upon and directly depend upon the unique characteristics of the active compound and the particular therapeutic effect to be achieved.

In therapeutic applications, the dosage of a pharmaceutical composition used according to the present disclosure depends on the medicament, the age, weight and clinical condition of the patient receiving the treatment, as well as the experience and judgment of the clinician or practitioner administering the treatment, and other factors affecting the selected dosage in addition thereto. In general, the dose should be sufficient to cause a slowing of the growth (and preferably regression) of the tumor and also preferably to cause complete regression of the cancer. An effective amount of a pharmaceutical agent is an amount that provides an objective, identifiable improvement as noted by a clinician or other qualified observer. For example, regression of a tumor in a patient can be measured with reference to the diameter of the tumor. A decrease in tumor diameter indicates regression. Regression was also indicated by the tumor no longer relapsing after treatment cessation. The term "dose-effective manner" as used herein refers to the amount of active compound that produces a desired biological effect in a subject or cell.

The pharmaceutical composition may be contained in a container, package or dispenser together with instructions for administration.

The disclosure also includes any physicochemical or stereochemical form that the disclosed compounds may take. Such forms include diastereomers, racemates, separated enantiomers, hydrated forms, solvated forms, any known or undisclosed crystalline or amorphous form, including all polymorphic crystalline forms. Amorphous forms lack a distinguishable crystal lattice and therefore an ordered arrangement of structural units. Many pharmaceutical compounds have amorphous forms. Methods for producing such chemical forms are well known to those skilled in the art.

The compounds of the present disclosure include possible stereoisomers and include not only racemic compounds but also individual enantiomers and/or diastereomers. When it is desired that the compounds are single enantiomers or diastereomers, the compounds may be obtained by stereospecific synthesis or by resolution (resolution) of the final product or any convenient intermediate. The resolution of the final product, intermediate or starting material may be effected by any suitable method known in the art. See, e.g., E.L.Eliel, S.H.Wilen, and L.N.Mander "Stereochemistry of Organic Compounds" (Wiley-lnterscience, 1994).

The racemates, individual enantiomers, or diastereomers of the disclosed compounds can be prepared by directed synthesis or by resolution by any method now known or as yet undisclosed. For example, a compound may be resolved into its enantiomers through the formation of diastereomeric pairs by salt formation using an optically active acid. Enantiomers are rarely crystallized and the free base is regenerated. In another example, enantiomers may be separated by chromatography. Such chromatography may be any suitable method now known or yet to be disclosed suitable for separating enantiomers, such as HPLC on a chiral column.

The compounds of the present disclosure are capable of further forming salts. All of these forms are also included within the scope of the claimed disclosure.

As used herein, "pharmaceutically acceptable salts" refer to derivatives of the compounds of the present disclosure in which the parent compound is modified by making acid or base salts thereof. Examples of pharmaceutically acceptable salts include, but are not limited to, inorganic or organic acid salts of basic residues (e.g., amines), basic or organic salts of acidic residues (e.g., carboxylic acids), and the like. Pharmaceutically acceptable salts include, for example, the conventional non-toxic salts or the quaternary ammonium salts of the parent compound formed from non-toxic inorganic or organic acids. For example, such conventional non-toxic salts include, but are not limited to, those derived from inorganic or organic acids selected from the group consisting of 2-acetoxybenzoic acid, 2-hydroxyethanesulfonic acid, acetic acid, ascorbic acid, benzenesulfonic acid, benzoic acid, bicarbonate, carbonic acid, citric acid, ethylenediaminetetraacetic acid (edetic), ethanedisulfonic acid, 1, 2-ethanesulfonic acid, fumaric acid, glucoheptose, gluconic acid, glutamic acid, glycolic acid, ethyleneglycolarylaniline, hexylresorcinol (hexylresorcinol), carbamic acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, hydroxymaleic acid, hydroxynaphthoic acid, isoethanesulfonic acid, lactic acid, lactobionic acid, dodecylsulfonic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, naphthalenesulfonic acid, nitric acid, oxalic acid, pamoic acid (pamoic acid), pantothenic acid, phenylacetic acid, phosphoric acid, polygalacturonic acid, propionic acid, salicylic acid, stearic acid, and mixtures thereof, Glycolic acid, succinic acid, sulfamic acid, sulfanilic acid, sulfuric acid, tannic acid, tartaric acid, toluenesulfonic acid and common amino acids, such as glycine, alanine, phenylalanine, arginine and the like.

Other examples of pharmaceutically acceptable salts include hexanoic acid, cyclopentanepropionic acid, pyruvic acid, malonic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo- [2.2.2] -octane-2-ene-1-carboxylic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, muconic acid, and the like. The present disclosure also includes salts formed when an acid proton present in the parent compound is replaced with a metal ion (e.g., an alkali metal ion, an alkaline earth metal ion, or an aluminum ion); or a salt coordinated with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, etc.

It will be understood that all references to pharmaceutically acceptable salts include solvent addition forms (solvates) or crystal forms (polymorphs) of the same salt as defined herein.

The (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure may also be prepared as an ester, e.g., a pharmaceutically acceptable ester. For example, a carboxylic acid functional group in a compound can be converted to its corresponding ester, e.g., methyl, ethyl, or other ester. The alcohol group in the compound may also be converted to its corresponding ester, e.g., an acetate, propionate, or other ester.

The (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide of the present disclosure may also be prepared as a prodrug, e.g., a pharmaceutically acceptable prodrug. The terms "pro-drug" and "prodrug" are used interchangeably herein to refer to any compound that releases an active parent drug in vivo. Since prodrugs are known to enhance many desirable qualities of a drug (e.g., solubility, bioavailability, manufacturing, etc.), the compounds of the present disclosure may be delivered in prodrug form. Accordingly, the present disclosure is intended to encompass prodrugs of the presently claimed compounds, methods of delivering the prodrugs, and compositions comprising the prodrugs. "prodrug" is intended to include any covalently bonded carrier that releases the active parent drug of the present disclosure in vivo when such prodrug is administered to a subject. Prodrugs in the present disclosure are prepared by modifying functional groups present in the compound in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compound. Prodrugs include compounds of the present disclosure wherein a hydroxy, amino, mercapto, carboxyl or carbonyl group is bonded to any group that can be cleaved in vivo to form a free hydroxy, free amino, free mercapto, free carboxyl or free carbonyl group, respectively.

Examples of Prodrugs include, but are not limited to, esters of hydroxyl functional groups (e.g., acetate, dialkylaminoacetate, formate, phosphate, sulfate, and benzoate derivatives), carbamates (e.g., N-dimethylaminocarbonyl), esters of carboxyl functional groups (e.g., ethyl ester, morpholinoethanol ester), N-acyl derivatives of amino functional groups (e.g., N-acetyl) N-mannich bases, schiff bases, and enaminones, oximes, acetals, ketals, and enol esters of ketone and aldehyde functional groups, and the like, see Bundegaard, h.

(S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, or a pharmaceutically acceptable salt, ester, or prodrug thereof, is administered orally, nasally, transdermally, pulmonarily, inhalationally, buccally, sublingually, intraperitoneally, subcutaneously, intramuscularly, intravenously, rectally, intrapleurally, intrathecally, and parenterally. In one embodiment, the compound is administered orally. Those skilled in the art will recognize the advantages of certain routes of administration.

The dosage regimen for the compound is selected in accordance with a variety of factors, including the type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; renal and hepatic function of the patient; and the particular compound or salt thereof used. An ordinarily skilled physician or veterinarian can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.

The dosage regimen may be daily (e.g., every 24 hours) administration of a compound of the disclosure. The dosage regimen may be administered daily for consecutive days, e.g., for at least two consecutive days, for at least three days, for at least four days, for at least five days, for at least six days, or for at least seven days. Administration may be more than once per day, for example two, three or four times per day (every 24 hours). The dosing regimen may be daily administration followed by at least one day, at least two days, at least three days, at least four days, at least five days, or at least six days of non-administration.

Techniques for formulating and administering the disclosed compounds of the present disclosure can be found in Remington: the Science and Practice of Pharmacy, 19 th supplement, Mack Publishing Co., Easton, Pa. (1995). In one embodiment, the compounds described herein and pharmaceutically acceptable salts thereof are used in pharmaceutical formulations in combination with a pharmaceutically acceptable carrier or diluent. Suitable pharmaceutically acceptable carriers include inert solid fillers or diluents and sterile aqueous or organic solutions. The compound will be present in such pharmaceutical compositions in an amount sufficient to provide the desired dosage within the ranges described herein.

The methods of the present disclosure for treating cancer include treating Malignant Rhabdoid Tumor (MRT). In preferred embodiments, the methods of the present disclosure are used to treat a subject having ovarian Malignant Rhabdoid Tumor (MRTO). MRTO may also be referred to as hypercalcemic small cell carcinoma of the ovary (SCCOHT). In certain embodiments, the MRTO or SCCOHT and/or the subject is characterized as SMARCA4 negative. As used herein, a SMARCA4 negative cell contains a mutation in the SMARCA4 gene, the corresponding SMARCA4 transcript (or cDNA copy thereof), or the SMARCA4 protein that prevents transcription of the SMARCA4 gene, prevents translation of the SMARCA4 transcript, and/or reduces/inhibits the activity of the SMARCA4 protein. The SMARCA4 negative status of the cells sensitizes the cells to EZH2 driven tumorigenesis.

The methods of the present disclosure may be used to treat subjects that are SMARCA4 negative or subjects having one or more cells that may be SMARCA4 negative. SMARCA4 expression and/or SMARCA4 function may be assessed by fluorescent and non-fluorescent Immunohistochemical (IHC) methods, including methods well known to those of ordinary skill in the art. In a certain embodiment, the method comprises: (a) obtaining a biological sample from a subject; (b) contacting a biological sample or portion thereof with an antibody that specifically binds SMARCA 4; and (c) detecting the amount of antibody bound to SMARCA 4. Alternatively, or in addition, SMARCA4 expression and/or SMARCA4 function may be assessed by a method comprising: (a) obtaining a biological sample from a subject; (b) sequencing at least one DNA sequence encoding SMARCA4 protein from a biological sample or portion thereof; and (c) determining whether at least one DNA sequence encoding SMARCA4 protein comprises a mutation that affects expression and/or function of SMARCA4 protein. SMARCA4 expression or function of SMARCA4 may be assessed by detecting the amount of antibody that binds to SMARCA4 and by sequencing at least one DNA sequence encoding SMARCA4 protein, optionally using the same biological sample from the subject.

All percentages and ratios used herein are by weight unless otherwise specified.

Other features and advantages of the disclosure are apparent in the various examples. The examples provided illustrate different components and methods useful in practicing the present disclosure. The examples do not limit the claimed disclosure. Based on the present disclosure, one of ordinary skill in the art may identify and employ other components and methods useful in practicing the present disclosure.

Examples

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

Example 1:

cell viability assay

CellTiter-Cell viability assay to measure cell viability. CellTiter-Luminescent Cell Viability Assay is a homogeneous method that determines the number of viable cells in culture based on the quantification of the presence of ATP, which is indicative of the presence of metabolically active cells. After treatment, CellTiter-Added to the treatment wells and incubated at 37 ℃. The luminescence values were measured using a Molecular Devices Spectramax microplate reader.

Single reagent study

Cells were grown to 70% confluence, trypsinized, counted, and plated at 2.5 × 103-5x103The final concentration of cells/well was seeded in 96-well flat-bottom plates (day 0). Cells were incubated in growth medium for 24 hours. Treatment with test or standard reagents started on day 1 and continued for 72 hours. At 72 th levelAt the time point, the treatment containing the medium was removed. By CellTiter-Cell viability assay to quantify viable cell number. The results of these studies were used to calculate the IC of each compound50Values (concentration of drug that inhibited cell growth by 50% of control).

Data acquisition

For single agent and combination studies, data for each experiment was collected and expressed as% cell growth using the following calculation:

% cell growth ═ fTesting/fCarrier)x100

Wherein f isTestingFor the fluorescence of the sample tested, fCarrierFluorescence of a vehicle that dissolves the drug. Dose response plots and ICs were generated using Prism 6 software (GraphPad) using the following equations50The value:

y ═ top-bottom)/(1 +10((logIC50-X) -slope))

Where X is the logarithm of the concentration and Y is the response. Y extends in an S-shape from the Bottom (Bottom) to the Top (Top).

Results

SCCOHT is characterized by the absence of SMARCA2 and SMARCA 4. Three SCCOHT cell lines tested in a cell proliferation assay (i.e., BIN67, COV434, and SCCOHT-1) were sensitive to (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, IC50Values were 51-293nM (see Table 2).

TABLE 2

Cell lines IC50(μM)
BIN67 0.051
COV434 0.035
SCCOHT-1 0.293

In proliferation assays, it was found that the cell lines that were both deficient in SMARCA2 and SMARCA4 (i.e., A204, G401, G402, H522, and A427) were also sensitive to (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide, IC50Values were 50-200nM (see FIG. 1).

Example 2:

in vitro treatment of BIN-67 cells with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) showed concentration and time dependent induction of SMARCA2 gene re-expression (see fig. 2).

In vitro treatment of BIN-67 cells with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) also showed concentration and time-dependent induction of SMARCA2 protein expression (see fig. 3).

Example 3:

in vivo treatment of tumors in the SCCOHT xenograft model (BIN-67) was evaluated. In vivo xenograft tumors from the SCCOHT cell line BIN-67 were administered with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) for 60 days. The treated tumors showed a statistically significant volume reduction compared to the vehicle control tumors (see FIG. 4).

Example 4:

in vivo treatment of tumors in a malignant rhabdoid tumor xenograft model (G401) was evaluated. In vivo xenograft tumors from MRT line G401 were administered with (S) -N-hydroxy-2- (2- (4-methoxyphenyl) butanamido) thiazole-5-carboxamide (i.e., GB-3103) for 30 days. The treated tumors showed a statistically significant volume reduction compared to the vehicle control tumors (see FIG. 5).

Example 5:

double deletions of SMARCA4/SMARCA2 ATPase in the SWItch/Sucrose Non-Fermentable (SWI/SNF) complex have been reported in hypercalcemic ovarian Small Cell Carcinoma (SCCOHT) and other tumors. The deletion of SMARCA4 was the result of an inactivating mutation, while the deletion of SMARCA2 was the result of the absence of mRNA expression. Repair of SMARCA4 or SMARCA2 may inhibit the growth of these cancers. The inventors have evaluated the activity of a novel, structurally rigid and potent class I/IIb HDAC inhibitor GB-3103 on human SCCOHT and other cell lines lacking the SWI/SNF complex. GB-3103 showed potent antiproliferative activity against human SCCOHT lines BIN67(51nM), COV434(35nM) and SCCOHT-1(293nM) (see FIG. 6), as well as against SWI/SNF-deficient striated muscle-like and lung tumor lines A204(95nM), A427(174nM), G401(138nM), G402(71nM), H522(102nM) (see FIG. 1), with low nM IC (IC) and50the value is obtained.

Treatment of human BIN67 SCCOHT cell line with GB-3103 for 72 hours showed effective concentration and time-dependent induction of SMARCA2 expression at the mRNA and protein levels (see figures 2 and 3). Treatment of mice bearing G401 human malignant rhabdoid tumor xenografts with QDs with GB-3103 at 5mg/kg resulted in 70% Tumor Growth Inhibition (TGI) (P <0.05) (see fig. 5) compared to vehicle controls. Mice bearing BIN67 human tumor xenografts were treated QD with 5mg/kg and 10mg/kg GB-3103, resulting in average tumor regressions of 26% and 33%, respectively, two weeks after initiation of treatment (see FIG. 4).

Example 6:

RNA-Seq analysis of BIN67 cells treated with GB-3103 showed that GB-3103 affected DNA replication and mRNA stability and induced MHC class II protein expression (see FIGS. 8 and 9).

Example 7:

in view of the importance of MHC class II expression and response to checkpoint inhibitor therapy, we tested GB-3103 alone and GB-3103 in combination with anti-mPD-1 and anti-mPD-L1 antibodies for activity in the syngeneic CT-26 mouse colon cancer model. GB-3103 induced 93% of TGI as a single agent. However, tumor growth recovered at day 15 and continued to increase until day 26 (see fig. 7). Surprisingly, GB-3103, when combined with anti-mPD-1 or anti-mPD-L1 (i.e. antibodies against PD-1 or PD-L1) results in regression of established CT-26 tumors, demonstrating the effective immune modulating activity of GB-3103 and the synergistic activity achieved in combination with immune checkpoint inhibitors.

Example 8:

the activity of GB-3103 was determined using HDAC isoforms 1-11, as shown in Table 3. GB-3103, a recent generation of epigenetic immunomodulator, is a potent HDAC isoform-limiting inhibitor that exhibits potent sub-nanomolar inhibition of HDAC3 and irreversible sub-nanomolar inhibitor of HDAC 6. In contrast to pan HDAC inhibition, isoform-restricted HDAC inhibition abrogates immune privilege.

HDAC3 has become a key target for enhancing immune function:

reduction of Treg suppressive function

Increasing natural killer cell ligand expression on tumors

Enhancement of macrophage host defense activity

Upregulation of PD-L1 expression and increased sensitivity to anti-PD 1 treatment

HDAC6 inhibition has potent immunomodulatory benefits including:

reduction of the expression of the anti-inflammatory cytokine IL-10

Enhancement of MHC class I/II gene expression and increase of expression of known tumor antigens

Reduction of immunosuppression and enhancement of immune function of T cells in melanoma patients

GB-3103 exerts potent effects on DNA repair and induces the expression of class I/II MHC proteins and a variety of tumor antigens. GB-3103 shows that an effective single agent regresses BIN67 SCCOHT tumors and inhibits the growth of G-401 tumors, both BIN67 SCCOHT tumors and G-401 tumors contain a double deletion of SMARCA4/SMARCA2 ATPase. GB-3103 shows potent activity in immunocompromised mice alone and in combination with anti-PD-1/anti-PD-L1 checkpoint modulators, and generates a tumor memory response to prevent tumor regrowth. GB-3103 is advancing IND support research into the clinical development of patients with genetically defined cancer, including those carrying a double deletion of SMARCA4/SMARCA 2.

GB-3103 is a novel epigenetic immunomodulator with potent anti-cancer activity against SWI/SNF deficient cancers. Clinical development of GB-3103 in these currently untreatable gene-defined rare cancers offers unique clinical and regulatory opportunities for breakthrough therapy certification, approval of which may be based on smaller one-armed clinical studies.

All publications and patent documents cited herein are incorporated by reference as if each such publication or document were specifically and individually indicated to be incorporated by reference. Citation of publications and patent documents is not intended as an admission that any of them is pertinent prior art, nor does it constitute any admission as to the contents or date thereof. While the invention has been described in terms of what is presently considered to be the written description, those skilled in the art will recognize that the invention can be practiced in various embodiments, and that the foregoing description and the following examples are intended for purposes of illustration and not limitation of the appended claims. The cell line or gene names, abbreviations and names used conform to the nomenclature of the American Type Culture Collection (ATCC) or the National Center for Biotechnology Information (NCBI) unless otherwise indicated or clearly evident from the context.

The present invention may be embodied in other specific forms without departing from its spirit or essential characteristics. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting of the invention described herein. The scope of the invention is, therefore, indicated by the appended claims rather than by the foregoing description, and all changes which come within the meaning and range of equivalency of the claims are intended to be embraced therein.

36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:用于治疗痛风或高尿酸血症的化合物

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!