Systems and methods for lysosome-induced immunogenic cell death

文档序号:1078003 发布日期:2020-10-16 浏览:18次 中文

阅读说明:本技术 溶酶体诱导的免疫原性细胞死亡的系统和方法 (Systems and methods for lysosome-induced immunogenic cell death ) 是由 埃里克·T·福塞尔 于 2018-11-15 设计创作,主要内容包括:本发明一般涉及使用免疫原性细胞死亡例如溶酶体诱导的免疫原性细胞死亡治疗癌症的系统和方法。本发明包括可以单独使用或一起使用的至少三个方面。第一方面涉及肿瘤的治疗准备,例如,通过停用和减少抗氧化剂,以及提供n-3至n-6和其他不饱和脂肪酸,和/或使用他汀类药物治疗受试者。第二个方面涉及通过包括CRISPR技术的基因操作恢复p53功能。第三个方面涉及抗体-酶复合物的构建,其中抗体识别肿瘤而酶是氧化酶。可以给药受试者该复合物。也可以给药受试者该酶的底物。这些和/或其他方面可以单独或一起使用,并且可以用于治疗和/或治愈癌症。在某些情况下,可以靶向溶酶体以诱导例如癌细胞中的细胞死亡。(The present invention relates generally to systems and methods for treating cancer using immunogenic cell death, such as lysosome-induced immunogenic cell death. The present invention includes at least three aspects that may be used separately or together. The first aspect relates to the therapeutic preparation of a tumor, for example, by disabling and reducing antioxidants, as well as providing n-3 to n-6 and other unsaturated fatty acids, and/or treating a subject with statins. A second aspect relates to the restoration of p53 function through genetic manipulation including CRISPR technology. A third aspect relates to the construction of an antibody-enzyme complex, wherein the antibody recognizes the tumor and the enzyme is an oxidase. The complex can be administered to a subject. The substrate for the enzyme may also be administered to the subject. These and/or other aspects may be used alone or together and may be used to treat and/or cure cancer. In certain cases, lysosomes can be targeted to induce cell death in, for example, cancer cells.)

1. An article of manufacture, comprising:

a complex comprising an antibody that recognizes a tumor, and an enzyme capable of increasing lysosomal membrane permeability.

2. An article of manufacture, comprising:

a complex comprising an antibody that recognizes a tumor, and an enzyme capable of producing reactive oxygen species.

3. The article of any one of claims 1 or 2, wherein the enzyme is an oxidase.

4. The article of claim 3, wherein the oxidase is glucose oxidase.

5. The article of manufacture of claim 3, wherein said oxidase is a xanthine oxidase.

6. The article of any one of claims 1 or 2, wherein the enzyme is a peroxidase.

7. The article of manufacture of any one of claims 1 to 6, wherein the antibody and the enzyme are directly covalently linked.

8. The article of any one of claims 1 to 7, wherein the antibody and the enzyme are linked by a cross-linking agent.

9. A method, comprising:

administering to a subject a composition comprising a complex of an antibody that recognizes a tumor, and an enzyme capable of increasing lysosomal membrane permeability.

10. A method, comprising:

administering to a subject a composition comprising a complex of an antibody that recognizes a tumor, and an enzyme capable of producing reactive oxygen species.

11. A method, comprising:

administering to a subject a composition comprising a complex of an antibody that recognizes a tumor, and an enzyme capable of causing lysosomal leakage.

12. The method of any one of claims 9 to 11, wherein the enzyme is an oxidase.

13. The method of claim 12, wherein the oxidase is glucose oxidase.

14. The method of claim 12, wherein said oxidase is a xanthine oxidase.

15. The method according to any one of claims 9 to 11, wherein the enzyme is a peroxidase.

16. The method of any one of claims 9 to 15, wherein the antibody and the enzyme are directly covalently linked.

17. The method of any one of claims 9 to 16, wherein the antibody and the enzyme are linked by a cross-linking agent.

18. The method of any one of claims 9 to 17, further comprising administering a fatty acid to the subject.

19. The method of claim 18, wherein the fatty acid comprises an unsaturated C3-C6A fatty acid.

20. The method of any one of claims 18 or 19, wherein the composition comprises the fatty acid.

21. The method of any one of claims 18 to 20, wherein administering the complex and administering the fatty acid are performed simultaneously.

22. The method of any one of claims 18 to 21, wherein administering the complex and administering the fatty acid are performed sequentially.

23. The method of any one of claims 8 to 22, further comprising administering to the subject a statin.

24. The method of claim 23, wherein the composition comprises the statin.

25. The method of any one of claims 23 or 24, wherein administering the complex and administering the statin are performed simultaneously.

26. The method of any one of claims 23 or 24, wherein administering the complex and administering the statin are performed sequentially.

Technical Field

The present invention relates generally to systems and methods for treating cancer using immunogenic cell death. For example, it is an object of certain embodiments of the present invention to treat and/or cure cancer by targeting lysosomes, causing their lysosomal membrane to penetrate and leak degrading enzymes, leading to intracellular digestion, leading to apoptosis, and thus to lysosome-induced immunogenic cell death.

Background

Cancer is a group of diseases associated with abnormal cell growth that may invade or spread to other parts of the body. Although cancer can be treated using techniques such as radiation therapy, surgery, chemotherapy, or targeted therapy, new techniques are still needed in view of the relatively high mortality rate of cancer patients.

Disclosure of Invention

The present invention relates generally to systems and methods for treating cancer using immunogenic cell death, such as lysosome-induced immunogenic cell death. In some cases, the objects of the present invention include related products, alternative solutions to specific problems, and/or a number of different uses for one or more systems and/or articles.

In one aspect, the present invention relates generally to articles. In one set of embodiments, the article of manufacture comprises a complex comprising an antibody that recognizes a tumor and an enzyme capable of increasing lysosomal membrane permeability. In another set of embodiments, the article of manufacture comprises a complex comprising an antibody that recognizes a tumor and an enzyme capable of producing reactive oxygen species.

In another aspect, the invention relates generally to a method. In one set of embodiments, the method comprises administering to the subject a composition comprising a complex of an antibody that recognizes the tumor and an enzyme capable of increasing lysosomal membrane permeability. According to another set of embodiments, the method comprises administering to the subject a composition comprising a complex of an antibody that recognizes the tumor and an enzyme capable of producing reactive oxygen species. In yet another set of embodiments, the method comprises administering to the subject a composition comprising a complex consisting of an antibody that recognizes the tumor and an enzyme capable of causing lysosomal leakage.

Disclosed herein are several methods of administering a compound for preventing or treating a particular disorder to a subject. It will be appreciated that in each of the above aspects of the invention, the invention also specifically includes the compound for use in the treatment or prophylaxis of the particular condition, and the use of the compound in the manufacture of a medicament for the treatment or prophylaxis of the particular condition.

In another aspect, the invention includes a method of making one or more embodiments of a complex described herein, e.g., comprising an antibody that recognizes a tumor and an enzyme capable of producing reactive oxygen species. In yet another aspect, the invention includes methods of using one or more embodiments of the complexes described herein, e.g., comprising an antibody that recognizes a tumor and an enzyme capable of producing reactive oxygen species.

Other advantages and novel features of the invention will become apparent from the following detailed description of various non-limiting embodiments of the invention when considered in conjunction with the drawings.

Drawings

Non-limiting embodiments of the present invention will be described by way of example with reference to the accompanying drawings, which are schematic and not to scale. In the drawings, each identical or nearly identical component that is illustrated is typically represented by a like numeral. For purposes of clarity, not every component may be labeled in every drawing, and not every component may be necessary to understand an embodiment of the invention for those of ordinary skill in the art. In the figure:

FIGS. 1A-1B illustrate lysosomal function in normal and malignant cells following uptake of oxidized LDL into the lysosome by endocytosis, according to certain embodiments of the invention;

FIGS. 2A-2B illustrate cellular components degraded within the autolysosome (autolysome) and released into the cytoplasm in another embodiment of the invention, in contrast to autophagy-induced apoptosis (autophagic-induced apoptosis); and

figure 3 shows a predictive assay for determining immunogenic cell death in yet another embodiment of the invention.

Detailed Description

The present invention relates generally to systems and methods for treating cancer using immunogenic cell death, such as lysosome-induced immunogenic cell death. The present invention includes at least three aspects that may be used separately or together. The first aspect relates to the therapeutic preparation of tumors, e.g. by disabling and reducing antioxidants, as well as providing n-3 to n-6 and other unsaturated fatty acids, and/or treating a subject with statins (statins). A second aspect relates to the restoration of p53 function through genetic manipulation including CRISPR technology. A third aspect relates to the construction of an antibody-enzyme complex, wherein the antibody recognizes the tumor and the enzyme is an oxidase. The complex can be administered to a subject. A substrate for the enzyme may also be provided to the subject. These and/or other aspects may be used alone or together and may be used to treat and/or cure cancer. In certain cases, lysosomes can be targeted to induce cell death in, for example, cancer cells.

As a non-limiting example, certain embodiments of the present invention generally relate to antibodies that recognize tumors, which antibodies may be complexed with an enzyme, for example, by a bond (such as a covalent bond). The tumor may be, for example, a tumor found in a subject having colorectal cancer. In some cases, colorectal cancer may be particularly difficult to treat, for example, some treatment modalities such as checkpoint inhibitors (checkpoint inhibitors) may be generally ineffective. Thus, these cancers may be treated using, for example, the compositions described herein. For example, in one embodiment, the compositions described herein can produce antigens, e.g., by increasing the permeability and/or leakage of lysosomal membrane and/or by producing active oxygen, etc., as described herein. In this way, the immune system (e.g., T cells) of the subject can recognize the tumor by recognizing antigens produced within the tumor of the subject. Thus, as a non-limiting example, in one embodiment, the composition comprises an anti-epCAM antibody capable of targeting a tumor (e.g., colorectal cancer). The composition may also comprise a suitable oxidase capable of producing an antigen, for example as described herein. For example, in one embodiment, the composition comprises xanthine oxidase (xanthine oxidase). In some cases, the anti-epCAM antibody and xanthine oxidase are covalently linked together, for example, by a thiol-thiol bond. As another example, the linkage may be an amine-mercapto bond. As yet another example, the linkage may be a hydroxyl-mercapto bond. Other possible connections include any of the connections described herein. Furthermore, in some cases, additional treatment steps may be used. For example, in one embodiment, a checkpoint inhibitor, such as a PD-1 inhibitor, is used, e.g., in addition to the above-described compositions.

However, it is to be understood that in addition to the above compositions, other compositions may also be used, for example, to treat various cancers described herein. For example, in certain embodiments, the present invention relates generally to systems and methods for targeting lysosomes within, for example, tumors. Without wishing to be bound by any theory, it is believed that lysosomes can be induced to penetrate and leak out degrading enzymes, thereby producing antigens that can be recognized by the immune system. Thus, for example, in certain instances, targeting the lysosome can result in the production of an antigen that can be recognized by the immune system so that the immune system can recognize the lysosome and/or the tumor containing the lysosome. In this way, "cold" tumors that are not readily recognized by the immune system can be turned into "hot" tumors that are more readily recognized by the immune system by producing antigens. Thus, at least in certain embodiments of the invention, a tumor can be treated by generating an antigen by causing a leak in the lysosome.

However, it should be understood that the above discussion is a non-limiting example of one embodiment of the invention that may be used to treat a subject having cancer. For example, some aspects generally relate to the preparation of a tumor therapy. For example, pretreatment of a subject can prepare the subject for successful attack of the lysosomal membrane, thereby facilitating treatment of the tumor. Thus, in some embodiments, a tumor may be prepared by providing fatty acids, statins, etc. to a subject and/or by removing or reducing antioxidants from the subject. Without wishing to be bound by any theory, it is believed that the oxidative process can be used to treat tumors, as described below. Thus, antioxidants may attenuate or inhibit the above-mentioned tumor treatments, and thus may in some cases facilitate treatment by reducing the concentration of antioxidants in the patient's body, for example. However, it is also to be understood that in all embodiments, pretreatment of the subject is not necessary, and in some cases, pretreatment may not be used.

Various methods may be used for the pre-treatment of tumors, including, for example, the inactivation and reduction of antioxidants, the provision of n-3 to n-6 and other unsaturated fatty acids, and/or the treatment of subjects with statins. Without wishing to be bound by any theory, it is believed that pretreatment of the subject provides for successful attack of the lysosomal membrane. Can reduce the antioxidant status of the subject. Feeding unsaturated fatty acids including n-3 and n-6 fatty acids and/or administering a statin to the subject. These treatments can more effectively attack the lysosomal membranes. However, it should be understood that pre-treatment of a tumor, such as described herein, is not required in all embodiments.

In one set of embodiments, for example, the unsaturated fatty acid is administered to the subject, e.g., fed to the subject. For example, one or more fatty acids can be administered to a subject simultaneously and/or sequentially. Typically, the unsaturated fatty acid may comprise one or more double and/or triple bonds in the fatty acid chain. In some cases, the fatty acid may be a relatively short chain fatty acid. Non-limiting examples of such unsaturated fatty acids include C3、C4、C5、C6、C7And C8A fatty acid. These fatty acids may include monounsaturated and/or polyunsaturated fatty acids. Specific examples include, but are not limited to, CH2=CHCOOH,CH3CH=CHCOOH,CH3CH2CH=CHCOOH,CH3CH2CH2CH=CHCOOH,CH2=CHCH2COOH,CH2=CHCH2CH2COOH,CH2=CHCH2CH2CH2COOH,CH2=CHCH2CH=CHCOOH,CH2CHCH ═ CHCOOH, and so on. Furthermore, in some instances, the fatty acids may be derived from natural sources, such as krill oil, fish oil, safflower oil, soybean oil, linseed oil, rapeseed oil, algal oil, and the like.

The fatty acids can be used in various dosages. In some embodiments, the amount of fatty acid agent administered to a subject (e.g., a human) can be at least 0.1 gram, at least 0.2 gram, at least 0.3 gram, at least 0.4 gram, at least 0.5 gram, at least 0.6 gram, at least 0.7 gram, at least 0.8 gram, at least 0.9 gram, at least 1 gram, at least 2 gram, at least 3 gram, at least 4 gram, at least 5 gram, at least 6 gram, at least 7 gram, at least 8 gram, at least 9 gram, at least 10 gram, at least 11 gram, at least 12 gram, at least 13 gram, at least 14 gram, or at least 15 gram. In some embodiments, the fatty acid is administered in a dose of no more than 15 grams, no more than 14 grams, no more than 13 grams, no more than 12 grams, no more than 11 grams, no more than 10 grams, no more than 9 grams, no more than 8 grams, no more than 7 grams, no more than 6 grams, no more than 5 grams, no more than 4 grams, no more than 3 grams, no more than 2 grams, no more than 1 gram, no more than 0.9 grams, no more than 0.8 grams, no more than 0.7 grams, no more than 0.6 grams, no more than 0.5 grams, no more than 0.4 grams, no more than 0.3 grams, no more than 0.2 grams, or no more than 0.1 grams. In some embodiments, any combination of the above doses is also possible, e.g., the dose may be between 1 and 15 grams, between 1 and 10 grams, between 5 and 10 grams, between 0.5 and 1 gram, etc.

In some embodiments, fatty acids can be administered to a subject (e.g., a human) at the following doses: at least 1 mg/kg, at least 2 mg/kg, at least 3 mg/kg, at least 5 mg/kg, at least 10 mg/kg, at least 20 mg/kg, at least 30 mg/kg, at least 50 mg/kg, at least 100 mg/kg, at least 200 mg/kg, at least 300 mg/kg, at least 500 mg/kg, at least 1 g/kg, or at least 2 mg/kg. In certain instances, the fatty acid can be administered in a dose of no more than 2 g/kg, no more than 1 g/kg, no more than 500 mg/kg, no more than 300 mg/kg, no more than 200 mg/kg, no more than 100 mg/kg, no more than 50 mg/kg, no more than 30 mg/kg, no more than 20 mg/kg, no more than 10 mg/kg, no more than 5 mg/kg, no more than 3 mg/kg, no more than 2 mg/kg, or no more than 1 mg/kg. In some embodiments, any combination of the above dosages is also possible, e.g., the dosage may be 1 to 2 g/kg, 500 mg/kg to 1 g/kg, 400 to 800 mg/kg, etc.

In another set of embodiments, a tumor can be prepared by administering to a subject, for example, a statin in addition to or in place of a fatty acid. Statins, also known as HMG-CoA reductase inhibitors, are lipid-lowering drugs. Non-limiting examples of statins include atorvastatin (atorvastatin), fluvastatin (fluvastatin), lovastatin (lovastatin), pitavastatin (pitavastatin), pravastatin (pravastatin), rosuvastatin (rosuvastatin), cerivastatin (cerivastatin), mevastatin (mevastatin), and simvastatin (simvastatin). For example, one or more statins may be administered to a subject simultaneously and/or sequentially.

A wide dosage range of statins may be used. For example, a subject (e.g., a human) may be administered a statin at the following doses: at least 1 mg, at least 2 mg, at least 3 mg, at least 5 mg, at least 10 mg, at least 20 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 60 mg, at least 70 mg, at least 80 mg, at least 90 mg, or at least 100 mg. In certain instances, the dose of statin administered may be no more than 100 mg, no more than 90 mg, no more than 80 mg, no more than 70 mg, no more than 60 mg, no more than 50 mg, no more than 40 mg, no more than 30 mg, no more than 20 mg, no more than 10 mg, no more than 5 mg, no more than 3 mg, no more than 2 mg, or no more than 1 mg. In some embodiments, any combination of the above dosages is also possible, e.g., the dosage may be 10 to 20 mg, 20 to 40 mg, 40 to 80 mg, 5 to 10 mg, and the like.

Furthermore, in some embodiments, the subject (e.g., human) may be administered a statin at the following doses: at least 1 mg/kg, at least 2 mg/kg, at least 3 mg/kg, at least 4 mg/kg, at least 5 mg/kg, at least 6 mg/kg, at least 7 mg/kg, at least 8 mg/kg, at least 9 mg/kg, at least 10 mg/kg, at least 11 mg/kg, at least 12 mg/kg, at least 13 mg/kg, at least 14 mg/kg, or at least 15 mg/kg. In some embodiments, the dose of statin administered may be no more than 15 mg/kg, no more than 14 mg/kg, no more than 13 mg/kg, no more than 12 mg/kg, no more than 11 mg/kg, no more than 10 mg/kg, no more than 9 mg/kg, no more than 8 mg/kg, no more than 7 mg/kg, no more than 6 mg/kg, no more than 5 mg/kg, no more than 4 mg/kg, no more than 3 mg/kg, no more than 2 mg/kg, or no more than 1 mg/kg. In some embodiments, any combination of the above dosages is also possible, e.g., the dosage may be between 1 and 15 mg/kg, 1 and 10 mg/kg, 5 and 10 mg/kg, etc.

According to certain embodiments, if the subject is taking an antioxidant, the antioxidant may be removed or reduced from the subject. For example, antioxidants may be completely withheld as part of the preparation for tumor therapy, or the antioxidant dose may be reduced (e.g., in amount and/or frequency). Non-limiting examples of antioxidants include beta carotene, vitamin a, and vitamin E. Other examples include vitamin C, glutathione (glutathione), lipoic acid (lipoic acid), uric acid, and panthenol (ubiquinol).

It is to be understood that in certain embodiments, any one or more of the above-described medicaments may be used independently, separately or together. For example, a statin, a fatty acid, or both may be administered to a subject. If both are present, they may be administered separately and/or together. If administered separately, the agents may be administered simultaneously and/or sequentially in any suitable order. Any suitable method and regimen of administration may be used, including those discussed herein. In addition, the statin or fatty acid or both may be present in a suitable pharmaceutically acceptable carrier. Furthermore, in some embodiments, antioxidants may be removed or reduced from the subject, whether or not one or both of a statin and a fatty acid are administered.

In some aspects, the systems and methods can be combined with checkpoint inhibitors (e.g., PD-1 inhibitors and/or PD-L1 inhibitors) to treat tumors. For example, in some cases, systems and methods such as those described herein can result in leakage of lysosomes and/or production of antigens that can be recognized by the immune system. Without wishing to be bound by any theory, it is believed that the production of antigens, such as those caused by lysosomal leakage, can help the immune system recognize tumors, thereby attacking the tumor more effectively. In certain instances, it is believed that this response may be enhanced in the presence of checkpoint inhibitors (such as PD-1 inhibitors), thereby allowing the immune system to more effectively attack the tumor. Non-limiting examples of PD-1 or PD-L1 inhibitors include nivolumab (nivolumab), pembrolizumab (pembrolizumab), alezumab (atezolizumab), avizumab (avelumab), Dewar lumab (durvalumab), pidilizumab (pidilizumab), cimiciprizumab (cemipimab), and the like. Other examples of checkpoint inhibitors that may be used include, but are not limited to, ipilimumab (ipilimumab), ofatumumab (ofatumumab), and rituximab (rituximab). Various checkpoint inhibitors are commercially available. However, it is also understood that in some embodiments, checkpoint inhibitors are not used.

The inhibitor (if present) can be provided in any suitable amount or concentration, and can be administered simultaneously and/or sequentially with other compositions (e.g., compositions described herein). Examples of dosages include, but are not limited to, at least 1 mg/kg, at least 2 mg/kg, at least 3 mg/kg, at least 4 mg/kg, at least 5 mg/kg, at least 6 mg/kg, at least 7 mg/kg, at least 8 mg/kg, at least 9 mg/kg, at least 10 mg/kg, and the like. In some embodiments, the dose may be no more than 10 mg/kg, no more than 9 mg/kg, no more than 8 mg/kg, no more than 7 mg/kg, no more than 6 mg/kg, no more than 5 mg/kg, no more than 4 mg/kg, no more than 3 mg/kg, no more than 2 mg/kg, or no more than 1 mg/kg. In some embodiments, any combination of the above dosages is also possible, e.g., the dosage may be between 1 and 15 mg/kg, 1 and 10 mg/kg, 5 and 10 mg/kg, etc.

For example, other cancer immunotherapies may be used in addition to or in place of inhibitors (e.g., checkpoint inhibitors). Non-limiting examples include non-specific immunotherapy, adaptive immunotherapy, monoclonal antibodies, CAR-T cell therapy, cancer vaccines, and the like. In some cases, cancer immunotherapy treatment may use and/or activate T cells, NK cells, and/or macrophages. The above-described therapies may be used, for example, in conjunction with the systems and methods described herein. One of ordinary skill in the art is familiar with systems and methods for producing monoclonal antibodies, identifying T cells, and the like.

For example, in CAR-T therapy, receptors are prepared that consist of antigen binding and T cell activation functions. The general premise of CAR-T cells is that T cells are artificially generated that target cancer cell markers. T cells are removed from the subject, genetically engineered to target cancer cells, and then placed back into the subject. CAR-T cells establish a link between the extracellular ligand recognition domain and an intracellular signaling molecule, which in turn activates the T cell. The extracellular ligand recognition domain may be a single chain variable fragment (scFv). CAR-T cells can be programmed to target antigens present on the surface of a tumor. When contacted with an antigen on a tumor, CAR-T cells are activated by binding to the tumor antigen. CAR-T cells destroy cancer cells through a variety of mechanisms, such as broadly stimulating cell proliferation, increasing the toxicity (i.e., cytotoxicity) of cells to other living cells, and increasing production of cell secreted factors that cause influences on other cells in the organism in the immune system. Such factors include cytokines (cytokines), interleukins (interleukins), interferons (interferons), growth factors, etc.

Another aspect of the invention relates to restoring p53 function by genetic manipulation (including CRISPR-associated techniques), which may be performed independently or in conjunction with tumor therapy preparation, and where performed in conjunction, as described above, the genetic manipulation and tumor therapy preparation may be performed simultaneously and/or sequentially in any suitable order. Without wishing to be bound by any theory, it is believed that in many cancers, p53 is either non-functional or functionally reduced. This may be important in the mechanism of attack of lysosomal membranes. Restoration of its function by genetic means (e.g. CRISPR) can improve the therapeutic ability to attack lysosomal membranes. However, as noted above, this is not required in all embodiments.

Another aspect of the invention relates to complexes such as antibody-enzyme complexes. Such a composite may be used in combination with one or more of the above aspects, or independently of these aspects. The antibody may be an antibody that recognizes a tumor. The enzyme may be an oxidase, or other enzyme capable of producing reactive oxygen species. The complex can be administered to a subject, for example, by infusion or other administration techniques (as described herein). In some cases, a substrate for the subject enzyme may also be provided. Without wishing to be bound by any theory, it is believed that the antibody helps to localize the complex within or near the tumor. Once localized, a substrate for the enzyme can be administered (e.g., infused) to the subject, which can trigger a series of events that lead to attack against the lysosomal membrane, release of lysosomal contents into the cell, and cell death through apoptosis. Thus, certain embodiments of the invention relate to lysosome-induced immunogenic cell death.

Furthermore, it should also be understood that other therapeutic moieties (thereuticmoieties) may be used in addition to enzymes. For example, a complex of an antibody and a drug, or a complex of another targeting moiety (moiety) and a drug may be used. In one set of embodiments, the complex may be an antibody-drug conjugate (or ADC).

Antibodies, including those capable of targeting tumors, are known to those of ordinary skill in the art. For example, in one set of embodiments, the antibody can be an antibody directed against a tumor antigen. Examples of tumor specific antigens include ras and the products of p53, CTAG1B or MAGEA 1. Other examples include tissue differentiation antigens, mutein antigens, oncogenic virus antigens, cancer-testis antigens, and blood vessel or matrix specific antigens. Other examples include carcinoembryonic antigen (oncofetal antigen), such as alpha-fetoprotein (AFP) and carcinoembryonic antigen (CEA). Other further examples include antibodies against CA-125, MUC-1, epithelial tumor antigen (epitalialtumor antigen), tyrosinase (tyrosinase), melanoma-associated antigen (MAGE), epithelial cell adhesion molecule (epCAM), and the like.

Antibodies are typically proteins or glycoproteins of one or more polypeptides substantially encoded by immunoglobulin genes or immunoglobulin gene fragments. Well-known immunoglobulin genes include the kappa, lambda, alpha, gamma, and mu constant region genes, as well as various immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, or, which in turn define the immunoglobulin class, i.e., IgG, IgM, IgA, IgD, and IgE, respectively. Typical immunoglobulin (antibody) building blocks are known to comprise tetramers. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25kD) and one "heavy" chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to the light chain and heavy chain, respectively, described above. Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests the antibody below the disulfide bonds in the hinge region (i.e. towards the Fc domain) to produce f (ab)' 2, a dimer of Fab, which is itself a light chain linked by disulfide bonds to VH-CH 1. F (ab) ' 2 can be reduced under mild conditions to disrupt the disulfide bonds in the hinge region, thereby converting the (Fab ') 2 dimer into a Fab ' monomer. The Fab' monomer is essentially a Fab with a partial hinge region. Although various antibody fragments are defined in terms of digestion of intact antibodies, these fragments can also be synthesized de novo, e.g., chemically by recombinant DNA methods, by "phage display" methods, and the like. Examples of antibodies include single chain antibodies, such as single chain fv (scfv) antibodies in which a variable heavy chain and a variable light chain are linked together (either directly or through a peptide linker) to form a continuous polypeptide. In one embodiment, the antibody is a monoclonal antibody.

The antibody and enzyme may be linked or bound together (e.g., covalently). They may be bound directly to each other or via one or more cross-linking agents. Non-limiting examples include glutaraldehyde (glutaraldehyde), NHS-ester (N-hydroxysuccinimide) (NHS-esters (N-hydroxysuccinimide)) (e.g., dithiobis (succinimidyl propionate) (dithiobis (succinimidyl propionate)), dithiobis (sulfosuccinimidyl propionate) (dithiobis (sulfosuccinimidyl propionate)), etc.), PEG (polyethylene glycol) groups, imidates (imides) (e.g., dimethyladipimidate (dimethyladipimidate), dimethylsubenimidate (dimethylpimelate), dimethylpimelimidate (dimethylpimelimidate), etc.), maleimides (maleimides), pyridyl (pyridines), carbodiimide (carbodiimides), isocyanate (isocyanate), etc. The antibody and enzyme may be conjugated by any suitable system, such as amine-amine, thiol-thiol, amine-thiol, carboxy-amine, thiol-carbohydrate, hydroxy-thiol, and the like. One of ordinary skill in the art is familiar with methods for cross-linking or binding proteins to each other.

As described above, the antibody may be linked to an enzyme. In one set of embodiments, the enzyme may be an oxidase or a peroxidase. Oxidases are generally enzymes that catalyze redox reactions, including oxidation with oxygen (O)2) An enzyme that is an electron acceptor. Oxygen can be reduced to water (H)2O), hydrogen peroxide (H)2O2). Non-limiting examples of oxidases include glucose oxidase (gluceoxidase), monoamine oxidase (monoamine oxidase), cytochrome P450oxidase (cytochrome P450oxidase), NADPH oxidase (NADPH oxidase), xanthine oxidase, L-gulonolactonase, laccase (laccase), lysyl oxidase (lysyl oxidase), and the like. Similarly, peroxidases are enzymes that can act on substrates such as hydrogen peroxide or lipid peroxides. Non-limiting examples of peroxidases include horseradish peroxidase (horseradish peroxidase), cytochrome c peroxidase (cytochrome c peroxidase), ascorbic acid peroxidase (ascorbate peroxidase), chloroperoxidase (chloroperoxidase), glutathione peroxidase (glutathione peroxidase), haloperoxidase (haloperoxidase), lactoperoxidase (lactoperoxidase), manganese peroxidase (manganesepoxidase), myeloperoxidase (myeloperoxidase), thyroid peroxidase (thyroidoperoxidase), vanadium bromoperoxidase (vanadic boroperoxidase), and the like.

In certain cases, the subject may also be administered a substrate for the enzyme. The substrate and enzyme may be administered in any suitable order, for example sequentially and/or simultaneously. Non-limiting examples of substrates include hypoxanthine (hypoxanthine) or xanthine for xanthine oxidase, or glucose for glucose oxidase. As another example, monoamine oxidase may act on serotonin (serotonin), melatonin (melatonin), norepinephrine (norepinephrine), epinephrine (epinephrine), phenethylamine (phenylethylamine), benzylamine (benzylamine), dopamine (dopamine), tyramine (tyramine), tryptamine (tryptamine), and the like for monoamine oxidase; NADPH against NADPH peroxidase; ascorbic acid directed against ascorbic acid peroxidase; and so on.

Without wishing to be bound by any theory, it is believed that oxidases or peroxidases act by oxidizing their substrates to produce reactive oxygen species (e.g., superoxide, peroxide, hydroxyl radicals, etc.). Thus, in some embodiments, a substrate is optionally added to facilitate such a reaction. Reactive oxygen species can interact with the lysosomes of tumor cells, inducing the lysosomal membranes to be more permeable and to leak out of degrading enzymes, which can lead to digestion or other reactions within the cell. For example, reactive oxygen species such as superoxide can cause oxidation of cellular components such as unsaturated lipids, which can be endocytosed and destabilize lysosomal membranes and disperse lysosomal enzymes throughout the cell, which then produce tumor-specific neoantigens that can be recognized by the immune system, and/or which can trigger apoptosis or cell death. Thus, in some embodiments, such reactive oxygen species can be used to cause lysosome-induced immunogenic cell death. Thus, it is believed that tumor cells can be targeted (e.g., using antibodies) to deliver enzymes capable of producing reactive oxygen species, resulting in lysosomal leakage and ultimately tumor cell death. See also example one below.

In some cases, the above enzymes may be facilitated by preparing for tumor therapy, e.g., using fatty acids and/or statins, and/or by removing or reducing antioxidants from the subject. Furthermore, in some cases, p53 function can be restored by genetic manipulation.

Furthermore, it is understood that in some cases, drugs may be used in the complex instead of and/or in addition to active oxygen producing enzymes.

The complex can be administered to a subject, e.g., a human, using any suitable technique, including those described herein. In addition, a variety of dosages may be used. For example, the complex may be administered at the following doses: at least 10 mg, at least 15 mg, at least 20 mg, at least 25 mg, at least 30 mg, at least 40 mg, at least 50 mg, at least 60 mg, at least 70 mg, at least 80 mg, at least 90 mg, at least 100 mg, at least 200 mg, at least 300 mg, at least 500 mg, at least 1000 mg, at least 1500 mg, at least 2000 mg, at least 2500 mg, at least 3000 mg, at least 5000 mg, and the like. In some embodiments, the complex may be administered at the following doses: no more than 5000 mg, no more than 3000 mg, no more than 2500 mg, no more than 2000 mg, no more than 1500 mg, no more than 1000 mg, no more than 500 mg, no more than 300 mg, no more than 200 mg, no more than 100 mg, no more than 90 mg, no more than 80 mg, no more than 70 mg, no more than 60 mg, no more than 50 mg, no more than 40 mg, no more than 30 mg, no more than 25 mg, no more than 20 mg, no more than 15 mg, no more than 10 mg, etc. In some embodiments, any combination of the above dosages is also possible, e.g., the dosage may be 10 to 20 mg, 50 to 100 mg, 100 to 200 mg, and the like.

Furthermore, in some embodiments, the complex may be administered to a subject (e.g., a human) at the following doses: at least 1 mg/kg, at least 2 mg/kg, at least 3 mg/kg, at least 4 mg/kg, at least 5 mg/kg, at least 6 mg/kg, at least 7 mg/kg, at least 8 mg/kg, at least 9 mg/kg, at least 10 mg/kg, at least 11 mg/kg, at least 12 mg/kg, at least 13 mg/kg, at least 14 mg/kg, or at least 15 mg/kg. In some embodiments, the statin may be administered at the following doses: no more than 15 mg/kg, no more than 14 mg/kg, no more than 13 mg/kg, no more than 12 mg/kg, no more than 11 mg/kg, no more than 10 mg/kg, no more than 9 mg/kg, no more than 8 mg/kg, no more than 7 mg/kg, no more than 6 mg/kg, no more than 5 mg/kg, no more than 4 mg/kg, no more than 3 mg/kg, no more than 2 mg/kg, or no more than 1 mg/kg. In some embodiments, any combination of the above dosages is also possible, e.g., the dosage may be between 1 and 15 mg/kg, 1 and 10 mg/kg, 5 and 10 mg/kg, etc.

In another aspect, the compositions described herein can be administered to a subject alone and/or in combination with cofactors, other therapeutic agents, and the like. For example, an antibody-enzyme complex (e.g., comprising an oxidase or other enzyme capable of producing reactive oxygen species) can be administered alone, or in combination with a substrate (e.g., a substrate as described herein) such as hypoxanthine, xanthine, glucose, and the like, and/or in combination with a fatty acid, a statin, and the like. When administered, the compositions can be administered as a pharmaceutically acceptable formulation in a therapeutically effective, pharmaceutically acceptable amount, such as those pharmaceutically acceptable carriers (as described below). The term "effective amount" of a composition (e.g., a complex as described herein) refers to an amount necessary or sufficient to achieve a desired biological effect. For example, an effective amount of a complex to treat a tumor can be an amount sufficient to reduce the size of the tumor. In combination with the teachings provided herein, by selecting among various active compositions and trade-off factors such as potency, relative bioavailability, patient weight, severity of adverse side effects and mode of administration, an effective prophylactic or therapeutic regimen can be planned that does not cause significant toxicity, but is entirely effective for treating a particular subject. The effective amount for any particular application may vary depending on factors such as the disease or disorder being treated, the particular composition being administered, the individual size of the subject, or the severity of the disease or disorder. One of ordinary skill in the art can empirically determine the effective amount of the composition without undue experimentation.

The terms "treat," "treated," "treating," and the like, as used herein, refer to administering to a subject a composition that increases the subject's resistance to the development or further development of a tumor, and administering the composition after the subject has developed a tumor to eliminate or at least control the development of the tumor, and/or to slow the development or lessen the severity of the symptoms caused by the tumor. When administered to a subject, an effective amount may depend on the particular tumor being treated and the desired result. A therapeutically effective dose can be determined by one of ordinary skill in the art, for example, using factors such as those described further below and using no more than routine experimentation.

When used in therapy, an effective amount of the composition can be administered to a subject by any means of delivering the composition to a tumor (e.g., mucosal delivery, systemic delivery, etc.). Administration of the pharmaceutical compositions of the present invention may be accomplished by any means known to those skilled in the art. Examples of routes of administration include, but are not limited to, oral, parenteral, intramuscular, intranasal, sublingual, intratracheal, inhalation, ocular, vaginal, intravenous, transdermal, and rectal. In some cases, intramuscular administration is used. In one set of embodiments, intravenous administration is used.

In administering the compositions to a subject, the dosage, dosing regimen, route of administration, and the like can be selected to affect the known activity of such compositions. The dosage may be estimated based on the results of the experimental model, optionally in combination with the experimental results of the compositions described herein. Depending on the mode of administration, the dosage may be adjusted appropriately to achieve the desired local or systemic drug level. The dosage may be administered once or more times a day. In certain instances, multiple daily administrations are also contemplated to achieve suitable systemic levels of the composition in a subject or within a tumor.

The dosage of the composition administered to the subject can be such that a therapeutically effective amount of the composition reaches a tumor in the subject. In certain instances, the maximum dose may be administered while avoiding or minimizing any potentially harmful side effects produced in the subject. The actual dose of the composition administered may depend on a variety of factors, such as the final concentration desired at the tumor site, the method of administration to the subject, the efficacy of the composition, the longevity of the composition in the subject, the time of administration, the effect of concurrent therapy (e.g., in cocktail therapy), and the like. The dose delivered may also depend on the relevant condition of the subject, and may in some cases vary from subject to subject. For example, the age, sex, weight, size, environment, physical condition, or current health of the subject can also affect the desired dosage and/or concentration of the composition at the active site. Dose variation may occur between individuals or even within the same individual on different days. In some cases, the maximum dose, i.e., the highest safe dose according to sound medical judgment, is used. In some cases, the dosage form has substantially no deleterious effect on the subject.

The dose of the compounds described herein that is delivered to a subject may range from about 0.1 micrograms to 10 milligrams per administration; depending on the application, it may be administered daily, weekly, or monthly, and any other amount of time in between. In some cases, the dosage range for each administration is about 10 micrograms to 5 milligrams, e.g., about 100 micrograms to 1 milligram, administered 2 to 4 times every several days or weeks. In some embodiments, the dosage range is from 1 microgram to 10 milligrams per administration, most typically from 10 micrograms to 1 milligram per day or week. Other suitable dosages have been described in detail above.

The composition can be administered in multiple doses over an extended period of time. For any of the compounds described herein, a therapeutically effective amount can initially be determined from an animal model. The dosage administered may be adjusted according to the relative bioavailability and potency of the compound administered. It is well within the ability of the ordinarily skilled artisan to adjust dosages based on the above methods and other methods known in the art to achieve maximal efficacy.

The treatment regimens disclosed herein can be administered to any subject, e.g., a human or non-human animal, e.g., a dog, cat, horse, rabbit, cow, pig, sheep, goat, rat (e.g., Rattus norvegicus), mouse (e.g., mus musculus), guinea pig, non-human primate (e.g., monkey, chimpanzee, baboon, ape, gorilla, etc.), and the like.

In certain embodiments, the composition can be administered to a subject with a family history of cancer, or a subject with a genetic predisposition to cancer. In other embodiments, the composition may be administered to a subject who has reached a particular age, or who is more susceptible to a tumor. In other embodiments, the composition is administered to a subject exhibiting symptoms of cancer (e.g., early or late stage). In other embodiments, the composition can be administered to a subject as a prophylactic measure. In some embodiments, the composition can be administered to a subject based on demographic or epidemiological studies, or to a subject in a particular area or profession. Non-limiting examples of cancers that can be treated include brain, pancreatic, hematological, melanoma, multiple melanoma, lung, breast, prostate, ovarian, colon, colorectal, glioblastoma, pancreatic, and other cancers.

Administration of the compositions of the invention may be by any medically acceptable method of bringing the composition to its target (e.g., a tumor). Of course, the particular mode selected may depend on factors such as those previously described, e.g., the particular composition, the severity of the condition in the subject, the dosage required for therapeutic effect, and the like. As used herein, a "medically acceptable" treatment mode refers to a mode that is capable of producing effective levels of the composition in a subject without causing clinically unacceptable side effects.

The composition may be administered to the subject using any medically acceptable method. Depending on the condition to be treated, administration may be local (i.e., to a particular region, physiological system, tissue, organ, or cell type) or systemic. For example, the composition may be administered orally, vaginally, rectally, buccally, pulmonarily, topically, intranasally, transdermally, parenterally, by injection or implantation, by surgical administration, or by any other method of administration that may enter a tumor. In some cases, for example, if two or more compositions are administered, more than one method of administration may be used.

Examples of parenteral treatment modalities that may be used include intravenous, intradermal, subcutaneous, intracavity, intramuscular, intraperitoneal, epidural, or intrathecal. Examples of implantable therapies include any implantable or injectable drug delivery system. Oral administration may be used in some embodiments for convenience of the subject and the dosage regimen. Compositions suitable for oral administration may be presented as discrete units, for example, hard or soft capsules, pills, cachets, tablets, lozenges, or lozenges. Other suitable oral compositions include solutions or suspensions in aqueous or non-aqueous liquids, such as syrups, elixirs or emulsions. In another set of embodiments, the composition can be used to fortify a food or beverage.

When systemic administration is desired, the composition may be formulated for parenteral administration by injection, for example by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, for example, in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.

Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. In addition, suspensions of the active compounds may in some cases be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include, but are not limited to, fatty oils (e.g., sesame oil), or synthetic fatty acid esters (e.g., ethyl oleate or triglycerides), or liposomes (lipomes). Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethylcellulose (sodium carboxymethyl cellulose), sorbitol (sorbitol) or dextran (dextran). Optionally, the suspension may also contain suitable stabilizers or agents to enhance the solubility of the compounds to allow for the preparation of highly concentrated solutions.

In addition to the formulations described above, in some embodiments, the compositions may be formulated as depot formulations. In some cases, such depot formulations may be formulated with suitable polymeric or hydrophobic materials (e.g., emulsions in acceptable oils) or ion exchange resins, or as sparingly soluble derivatives, such as a sparingly soluble salt.

Suitable liquid pharmaceutical dosage forms include, for example, aqueous or saline solution for inhalation, microencapsulated forms, encapsulated in cochleates, coated on microscopic gold particles, contained in liposomes, aerosolized forms, aerosol forms, pellet forms for implantation into the skin, or forms that dry onto sharp objects that are scratched into the skin. Pharmaceutical compositions may also include granules, powders, tablets, coated tablets, (micro) capsules, suppositories, syrups, emulsions, suspensions, creams, drops or sustained-release preparations of the active compounds, in which preparations excipients and additives and/or auxiliaries, such as disintegrants, binders, coatings, swelling agents, lubricants, flavourings, sweeteners or solubilizers, are generally used in the manner described above.

In certain embodiments of the invention, the mode of administration of the compositions as described herein may be designed to result in continuous exposure of the composition over a specified period of time, for example hours, days, weeks, months or years. This can be achieved, for example, by repeated administration of the composition by one of the methods described herein, or by a sustained or controlled release delivery system in which the composition is delivered over an extended period of time without repeated administration. The compositions using such delivery systems may be administered, for example, by oral dosage forms, bolus injection, transdermal patch, subcutaneous implants, or other methods (such as those described herein). In some cases, it may be desirable to maintain the concentration of the composition substantially constant.

Other delivery systems suitable for use in certain embodiments include time release, delayed release, sustained release or controlled release delivery systems. The above system may in many cases avoid repeated administrations and provide greater convenience to the subject. Many types of delivery systems are available and are well known to those of ordinary skill in the art. They include, for example, polymer-based systems such as polylactic and/or polyglycolic acids (polyactics), polyanhydrides (polyanhydrides), polycaprolactones (polycaprolactones), and/or combinations thereof; lipid-based non-polymer systems including sterols such as cholesterol, cholesterol esters, and fatty acids or neutral fats such as mono-, di-, and triglycerides; a hydrogel release system; a liposome-based system; a phospholipid-based system; silicone rubber (silastic) systems; a peptide-based system; a wax coating; compressed tablets using conventional binders and excipients; or a partially fused implant. Specific examples include, but are not limited to, erosion systems, wherein the composition is contained in a form within a matrix, or diffusion systems, wherein the active ingredient controls the release rate. The formulation may be in the form of, for example, microspheres, hydrogels, polymer reservoirs, cholesterol matrices, or polymer systems. In some embodiments, the system can allow for sustained or controlled release of the composition, for example, by controlling the diffusion or erosion/degradation rate of the formulation. Further, a pump-based hardware delivery system may be used in some embodiments.

Long-term release implants may be particularly suitable for use in some embodiments. As used herein, "extended release" refers to the construction and placement of an implant containing a composition described herein to provide therapeutically effective levels for at least 30 or 45 days, or at least 60 or 90 days, or in some cases even longer. Long-term release implants are well known to those of ordinary skill in the art and include some of the release systems described herein.

The compositions described herein may be administered alone or in combination with other therapeutic agents and/or compositions. For example, non-limiting examples of anti-cancer agents and drugs that can be used in combination with the various compositions described herein include, but are not limited to, any one or more of the following: 20-epi-1,25dihydroxyvitamin D3(20-epi-1,25dihydroxyvitamin D3), 4-epoetin (4-ipomoenol), 5-ethynyluracil (5-ethyluracil), 9-dihydrotaxol (9-dihydrotaxol), abiraterone (abietane), acivicin (acivicin), aclarubicin (aclarubicin), aclodanzozole hydrochloride (acibenzolar hydrochloride), aclonine (acronine), acylfulvene (acylfulvene), adenosine (adecinonol), adolesin (adolesisin), aldesleukin (aldesleukin), total tk antagonist (acetate-tkatoninone), altretamine (acetatemine), amstine (amstine), amytaminomycin (amidomycin), amitriptolide (amidodox), amitriptolide (acetominophen), amitriptolide (amidomycin), amitriptolide (amidodoxylamine), amitriptolide (amidodoxylamine (amidomycin), amitriptolide (amidodoxylamine (amitriptolide), amitriptolide (amidomycin), amitriptolide (amitriptolide), amitriptolide (amidomycin), amitriptolide (amitriptolide), amitriptolide (amidomycin), amitriptolide) and (amidodoxylamine (amicin) in (amidodoxylamine (amisole) in), amdoxylamine (amisole, amdoxylamine (amitriptolide) in (amikainate, amdoxylamine (, Amsacrine (amsacrine), anagrelide (anagrelide), anastrozole (anastrozole), andrographolide (andrographolide), angiogenesis inhibitor, antagonist D (antaconist D), antagonist G, anagrelide (antarilix), anthranomycin (antranamycin), anti-dorsal morphogenetic protein-1 (anti-dorsalsolic protein-1), antiestrogens, antineoplasts (antineoplastins), antisense (antinense) oligonucleotides, aphidicolin glycinate (aphrodistin glycinate), apoptosis gene modulators (aphrodisiamodulators), apoptosis regulatory factors (aphrodisiagenes) depuratives (aprtophases), apurinic acid (apreinascitinase), ARA-CDP-DL-PTBA, arginine depuratinase (arginase), asparginine (asparginine), asparginine (aspartamine), aspartamine (amoxastastine), aspartamine (amoxatrine), aspartamine (1), aspartamine (amoxastastine), arginine depuratin (apraxistatin), amastatin (apraximin), amastatin (amastatin), amastatin (apraximin), amastatin (amastatin), aspartam-DL-L-arginine-L-arginine-L-, Azastatin 3, azacitidine (azacitidine), azasetron (azasetron), azatropin (azatoxin), azatyrine (azatyrosine), azatepa (azetepa), azomycin (azotomycin), baccatin III derivatives (baccatin III derivatives), balanitol (balanol), batimastat (batistat), benzodihydrophenamine (benzochlorin), benzozotepa (benzodepa), benzoylstaurosporine (benzoylstaurosporine), betalactam derivatives (betalactamivatives), betalaicin (betaalethidine), betabetabetalaimycin B (betalaimycin B), betulinic acid (GF), betalaidine inhibitors, betalaidine (benzathine), bizidine hydrochloride (bizidine A), bizidine (bile hydrochloride), bizidine hydrochloride (bizidine A), bizidine hydrochloride (bizidine hydrochloride), bizie (bizie, bizidine hydrochloride (bizidine hydrochloride), bizie (bizidine hydrochloride), bizidine hydrochloride (bizie (bizidine hydrochloride), bizidine hydrochloride (bizie (bizidine hydrochloride, Bleomycin sulphate (bleomycin sulfate), BRC/ABL antagonists, breeford (breffate), brequinar sodium (brequinar sodium), briprimine (bropirimine), butobactam (budotitane), busulfan (busulfan), buthionine sulfimide (buthionine sulfimide), actinomycin (cactinomycin), calcipotriol (calcipotriol), carbostatin C (calciphostin C), carprotosterone (calcisterone), camptothecin derivatives (calcithecin derivitives), canaryil-2 (canarypox IL-2), capecitabine (capitabine), carbinamide (carboximide), carbomer (carbomycin), carboplatin formamide (carboximidamide hydrochloride), carboximidamide (carboximide), carboximidamide (carboximidamycin hydrochloride), carboximide (carboximidamycin (carboximide), carboximide (carboximide), carboximidamycin (carboximide), carboximide (carboximide), carboximidamycin) inhibitors (carboximide), carboximidamycin (carboximide), carboximide (carboximide, Castanospermine (castanospermine), cecropin B (cecropin B), cedarville (cedefingul), cetrorelix (cetrorelix), chlorambucil (chlramucil), diphenoxylate (chlororins), chloroquinoxaline sulfonamide (chloroquinoxaline sulfonimide), cicaprost (cicaprost), cerrolomycin (cirrolomycin), cisplatin (cissplatin), cis-porphyrin (cis-porphyrin), cladribine (cladribine), clomiphene analogs (clomiphene analogs), clotrimazole (clotrimazozole), cole-cloristine A (colchicamycin A), griseofibrinomycin B, combretastatin A4 (crotacta 4), combretastatin analogs, jejunanin (clonidine), bectacrolidine (clacingula), beclomethazine A (clarkestaphylin), non-clavulanate derivatives (noncryptophan A), tacrolipramiperin (816), tacrolipid benzoate (noncryptophan), tacrolidine A (noncrypsin-clavine derivatives (8), tacrolidine A (nonchromtrine, clathraustrine A), clorictam-clavine A, clathromycin derivatives (clathromycin A), clotrimorphin (clathromycin A), clorictam, clathromycin A), clotrimorphin (clorictam-e, clathromycin A), clotrimorphin (clorictam-clathromycin (clorplate, clathromycin derivative (clorictam-e, clathromycin (clorplate, clathromycin A), clorpamide (e, serpentin (cyclophilin), cytarabine (cytarabine), cytarabine phospholipide (cytarabine ocfosfate), cytolytic factor (cytolytic factor), hexestrol phosphate (cytostatin), dacarbazine (dacarbazine), daclizumab (dacylimab), dactinomycin (dactinomycin), daunorubicin hydrochloride (daunorubicin), decitabine (decitabine), dehydromembranocetin B (dihydrodidemnin B), dessertraline (deslorelin), dexifosfamide (dexfosfamide), ormaplatin (dexametplatin), dexrazoxane, dexamethazine (dexamesine), dexamethazine (dexamethyridazine), dexrazine (dexamesine), dexamethazine (doxazone), dexamethazine (doxycycline), dexamectin (doxycycline), dexamesine (5-dihydroazepine), dexametine (dihydrodexazadine), dexamesine (doxycycline), dexamesine (doxycycline) (5) and dexametine (doxycycline), dexametine (doxycycline), dexamesine (doxycycline), dexametine (doxycycline), dexame, Docosanol (docosanol), dolasetron (dolasetron), doxifluridine (doxifluridine), doxorubicin (doxorubicin), doxorubicin hydrochloride (doxorubicin), droloxifene (droloxifene), droloxifene citrate (droloxifene citrate), drotalandrone propionate (dromostanolone propionate), tranabinol (dronabinol), dactinomycin (duazomycin), dactinomycin SA (duocarmycin SA), ebselen (seleben), etokamomustine (ecomoustine), edatrexate (edatrexate), edelfosine (edelfosine), ecolomicromab (ederomomab), efornithine (eflornosine hydrochloride), epirubicin (epirubicin), epirubicin (e (epirubicin), rubicin (e (epirubicin), rubicin), and epirubicin (e (epirubicin), e (e, e, The pharmaceutical composition is prepared from bixin hydrochloride (esorubicin hydrochloride), estramustine (estramustine), estramustine analogue, estramustine sodium phosphate (estramustine phosphate sodium), estrogen agonist, estrogen antagonist, etanidazole (etanidazole), etoposide (etoposide), etoposide phosphate (etoposide phosphate), etoposide (etoposide), exemestane (exemestane), fadrozole (fadrozole), fadrozole hydrochloride (fadrozole hydrochloride), fazarabine (fazabine), fenretinide (fenretinide), filgrastim (filgrastim), finasteride (finasteride), flazidine (fluvirguldol), fluxuridine (fluzelastine), fluxuridine (fluxuridine), fluxuridine hydrochloride (fluxuridine), fluxuridine (fluxuridine), fluxuridine (fluvastatin (flunomide), flunomide (flunarine), flunaringine (flunaringene), flunaringen (flunaringene), flunaringene (flunaringene), flunaringen (flunaringene), flunaringene (flunaringenin), flunaringen, Fostaricin (fostericin), fostretin sodium (fostericin sodium), fotemustine (fotemustine), gadolinium deuteroporphyrin (gadolinium texaphyrin), gallium nitrate (gallimum nitrate), galocitabine (galocitabine), ganirelix (ganirelix), gelatinase inhibitors (gelatinase inhibitors), gemcitabine (gemcitabine), gemcitabine hydrochloride (gemcitibine hydrochloride), glutathione inhibitors, Helsuvin (hepulam), albumin (heregulin), hexamethylenediethylamide (hexamethylene bisacetrimide), hydroxyurea (hydroxyurea), hypericin (hydroperidinic), ibandronic acid (ibandronic acid), idarubicin (ariubicin), idarubicin hydrochloride (hydrastrine), idarubicin (imidazole), imidazole (imidazole-like receptor), imidazole (imidazole), imidazole (imidazole-imidazole), imidazole (imidazole-like receptor (imidazole), imidazole-s (imidazole-2, imidazole-imidazole, imidazole-2, imidazole-like, imidazole-imidazole, imidazole-2-imidazole, imidazole-imidazole, imidazole-2-imidazole, imidazole, Interferon agonists, interferon alpha-2A, interferon alpha-2B, interferon alpha-N1, interferon alpha-N3, interferon beta-1A, interferon gamma-IB, interferon, interleukins, iobenguanide (iobenguane), iododoxorubicin (iododoxorubicin), iproplatin (iproplatin), irinotecan (irinotecan), irinotecan hydrochloride (irinotecan hydrochloride), iprolacin (irinoplast), issorafenib (irsogladine), isogazole (isobazazole), isophoronectin B (isohalophenontin B), itasetron (itasetron), Jerussetate (jasplakinolide acetate), carhalalide F (kalalalide F), lamellarin-N triacetate (melastatin), lentinacanthrin-N-triacetin (luteolin), lentisetrin (flavostatin), lentinacin (flavostatin), lentinacanthin (flavostatin), lentinacin (flavoglastatin), epirubicin (flavoglastrin sulfate), lentinacin (ivastatin (irigenin), epirubicin (irigenin), and their salts, Letrozole (letrozole), leukemia inhibitory factor, leukocyte interferon-alpha, leuprolide acetate (leuprolide acetate), leuprolide (leuprolide)/estrogen/progesterone, leuprolide (leuprolide), levamisole (levamisole), liazole (liarozole), liarozole hydrochloride (liarozole hydrochloride), linear polyamine analogues (linear polyamine analogues), lipophilic dipalmitopeptide (lipohilic disaccharide peptide), lipophilic platinum compounds (lipophilitic platinum compounds), lisoclinamide 7 (lipocalinamide 7), lobaplatin (lobaplatin), earthworm phospholipid (lomicrin), lometroxol (lomexolone), lomethaxol sodium (lobemixolone sodium), lostemustine (losteminum), loxacin (loxacin hydrochloride), loxacin (loxacin), loxacin (loxacin), lox, Maytansine (maitansine), mannostatin A (mannostatin A), marimastat (marimastat), mazoprol (masoprocol), maspin (maspin), matrilysin inhibitor (matrilysin inhibins), matrix metalloproteinase inhibitor (matrix metalloprotease inhibins), maytansine (maytansine), mechlorethamine hydrochloride (mechlorethamine hydrochloride), megestrol acetate (mecystrone acetate), melengestrol acetate (medestrenol acetate), melphalan flange (melphalan), methonuril (menogaril), mellumbone (merloprone), mercaptopurine (mercaptoprine), meterelin (meterelin), methioninase (methioninase), methotrexate (methohexidine), methothrene (methothrene sodium), methothreonine (methothreonine), methoprene (methoprene), methoprim (methoprim), methoprimidine (methoprene), methoprene (methoprim), methoprene (methoprene), methoprene (methoprim), methoprene (methoprim), methoprim (methoprim), methoprene (methoprim), methoprim (methoprim), or (methoprim), or, Mitomycin diamides (mitomycin), mitoxantrone (mitocarcin), mitorubicin (mitomycin), mitogellin (mitogellin), mitoguazone (mitoguazone), dibromodulcitol (mitolactol), mitomycin (mitomycin), mitomycin analogs, mitonaphthylamine (mitonafide), mitosper (mitosper), mitotane (mitotane), mitotoxin fibroblast growth factor-saporin (mitoxin fibroblast growth factor-saponin), mitoxantrone (mitoxantrone), mitoxantrone hydrochloride (mitoxantrone hydrochloride), moraxetin (mofanotene), moraxetin (molum), monoclonal antibody, human membrane hormone (mitosporine), mitoxantrone (mitoxantrone hydrochloride), moraxepinol (mitoxantrone), murabuterol (mitomycin), muramidase (mitomycin), muramyl (mitomycin antagonist, muramyl inhibitor, muramidamycin), muramyl (mitomycin antagonist, mitomycin-based on a gene, mitomycin antagonist, mitomycin-like, mitomycin-inhibitory genes, mitomycin-, Mecanoproxil B (mycaperoxideB), Mycobacteria cell wall extract (mycobactral cell wall extract), mycophenolic acid (mycophenolicacid), Mirisperidone (myciapionone), N-acetyldinaline (N-acetyldinaline), nafarelin (nafarelin), narretest (naggrethip), naloxone/tebuconazole (naloxone/pentazocine), Napavine (napavirin), Naphtheirather (napterin), nartostatin (nartograssin), nedaplatin (nedaplatin), nemorubicin (nemorubicin), neridronic acid (neridronic acid), neutral endopeptidase (nersultap), nilutamide (nilutamide), nisicin (nisicin), nisin (nitrosamine), nitromycin (nitromycin), guanine nitrogen oxide (nitromycin), guanine (guanidium-6), nicotinamide-carbonitrile (nicotinamide-carbonitrile), nicotinamide-imidazole (nicotinamide-carbonitrile (nicotinamide, nicotinamide (nicotinamide-nicotinoidin), nicotinamide (nicotinamide, nitromycin), nicotinamide (nicotinamide, nitromycin-2-nicotinoidin (nicotinamide, naricin), naringin (nicotinamide, naringin, Oxcarbazone (okicenone), oligonucleotide, onapristone (onapristone), ondansetron (ondansetron), olacin (oracin), oral cytokine inducer, ormaplatin (ormaplatin), oxaliplatin (osarone), oxaliplatin (oxaliplatin), enomycin (oxyanomycin), oxisulam (oxisuran), paclitaxel (paclitaxel), paclitaxel analogue, paclitaxel derivative, palatamine (palauaminine), palmitylrhizoxin (palmitylrhizoxin), pamidronic acid (pamidronic acid), panaxytriol (panaxytriol), paminomifene (pancreaticin), palatinomycin (palatinomycin), pamezetidine (palatinomycin), dermatacin (dermatacin), pentostatin (pentostatin), or pentostatin), dextran (sulfastatin), dextran (sulfadoxylamine), or a salt of the compound, or a compound, such as an, Perillyl alcohol (perillyl alcohol), phenazine (phenozinomycin), phenylacetate (phenylacetate), phosphatase inhibitor (phosphatases inhibitors), piceatannol (picibanil), pilocarpine hydrochloride (picocarpine hydrochloride), pipobroman (piphroman), piposulfan (piposulfan), pirarubicin (pirarubicin), pirtrexin (piriteoxime), piroanthraquinone hydrochloride (piroxanthone hydrochloride), practine A (plactina A), practistine B, plasminogen activator inhibitor (plagiogenin activiinhibitor), platinum complex (platinan complex), platinum-triamine complex (planum-propamin), plicamycin (plagiomycine), phenomycin (practicillin J), phenoxazine hydrochloride (phenoxazine hydrochloride), propidium-propidium chloride (practirone hydrochloride), prostanoid (practinoid J), prostanoid (practirone hydrochloride (practine hydrochloride), prostanoid J (practirone hydrochloride), prostanoid (practine hydrochloride), prostanoid J (practine hydrochloride), prostanoid (practirone hydrochloride), prostanoid J), prostanoid (practine hydrochloride), prostanoid J), and (practine hydrochloride), prostanoid I (practine hydrochloride), and (practine hydrochloride), wherein, Proteasome inhibitors (proteasome inhibitors), protein A-based immunomodulators, protein kinase C inhibitors, protein tyrosine phosphatase inhibitors (protein tyrosine phosphatase inhibitors), purine nucleoside phosphorylase inhibitors (purine nucleoside phosphorylase inhibitors), puromycin (puromycin), puromycin hydrochloride (puromycin hydrochloride), purpurins (purpirarins), pyrazolofuranin (pyrazofurin), pyrazoloacridines (pyrazoloacridines), pyridoxalated hemoglobin polyethylene oxide conjugates (pyridoxalated hemoglobin polyoxyethylene conjugates), RAF antagonists, raltitrexed (raltitrexed), ramosetron (ramosetron), farnesetron (ramosetron), farnesyl protein transferase inhibitors (ralysin inhibitors), aminothioredoxin inhibitors (ralysin inhibitors), rhenium reductase inhibitors (riboside I), riboside inhibitors (riboside I), protein kinase inhibitors (riboside I), protein tyrosine kinase inhibitors (riboside I), and pharmaceutically acceptable salts thereof, RNA interference, roglymide (rogletimine), rohitudine (rohitukine), romotide (romurtide), roquinacre (roquinemex), lubiginone B1(rubiginone B1), lupoxl (ruboxyl), safrog (safrogol), safrog hydrochloride (safrogol hydrochloride), santoprene (saintopin), sarcosine nitrosourea (sarcnu), sacatoo A (sarcophytol A), sargramostim (sargramostim), SDI 1mimetics (SDI 1 mimicrics), semustine (semustine), senilisation inhibitor 1, sense (sense) oligonucleotides, signal inhibitors, signal transduction modulators, octreozine (semperrazoxane), single-stranded antigen binding proteins, sizopyrane (buspirosone), sodium boroxine (butroxobin), sodium phosphate (sodium benzoate), sodium phosphocalcipotamone (sodium phosphate), sodium phosphoacetate (sodium benzoate), sodium benzoate (sodium benzoate) phosphate (sodium benzoate) and sodium benzoate (sodium benzoate) phosphate (sodium benzoate) as a salt, Sparamycin (spiramycin), spiramycin D (spicamycin D), germanium spiramine hydrochloride (spirogyramide), spiromustine (spiromustine), spiroplatinum (spiroplatin), slalanin (spirotropin), spongistatin 1(spongistatin 1), squalamine (squalamine), stem cell inhibitors, stem cell division inhibitors, stilbenamide (stipamide), streptavidin (streptonigrin), streptozocin (streptozocin), stromelysin inhibitors (stromysinhibitors), soflavin (sulfinosine), chlorotolurea (sulofenur), superactive vasoactive intestinal peptide antagonists, suradista (suradistidine), suramin (suramin), swainsonine (swainsonine), synthetic polysaccharides (sultamycin), thiotamine (thiamethoxam), thiostatin (furastatin), thiostatin (sodium iodostatin), thiostatin (thiostatin), thiostatin (thiostatin), thiostatin (e), thiostatin (, Telomerase (temomerase) inhibitors, tetroxantrone hydrochloride (teloporphine), temoporfin (temoporfin), temozolomide (temozolomide), teniposide (teniposide), tiruloximone (teroxirone), testolactone (tetoralactone), tetrachlorodecaoxide (tetrachloroxanide), tizoxamine (tetrazoimine), thietalistine (thielavine), thalidomide (thalidomide), thiamimipramine (thiamimiprine), thiocoraline (thiocoraline), thioguanine (thioguanine), thiotepa (thiotepa), thrombopoietin (thymopoietin), thrombopoietin (prothromotene mimetic), fasin (thymopoietin), thyrotoxin (thyrotoxin receptor agonist (thyrotoxin), thyrotropin (thyrotropin hydrochloride), thyrotropine (thyrotropine hydrochloride), thyrotropine (thyrotropine hydrochloride (thyrotropine), thyrotropine (thyrotropine hydrochloride (thyrotropine, thyrotropine hydrochloride), thyrotropine (thyrotropine hydrochloride (thyrotropine, thyrotropine hydrochloride), thyrotropine (thyrotropine, thyrotropine hydrochloride), thyrotropine hydrochloride, thyrotropine hydrochloride (thyrotropine, thyrotropi, Toremifene (toremifene), toremifene citrate (toremifene citrate), totipotent stem cell factor, translation inhibitor, tritulone acetate (tresolone acetate), tretinoin (tretinoin), triacetyluridine (triacetyluridine), triciribine (triciribine), triciribine phosphate (triciribineephosphate), trimetrexate (trimetrexate), trimetrexate glucuronate (trimetrexate), triptorelin (triptorelin), tropisetron (tropisetron), tolbutazine hydrochloride (tubolohydrochloride), tourette (turulone), tyrosine kinase (tyrosinase) inhibitor, tivotene (tyosines), UBC inhibitor, steverameltrexate (uramustine), urokinase (growth factor B), tretinomycin (retinokinase), tretinomycin (tretinomycin), tretinomycin (tretinomycin) inhibitor, tretinomycin (tretinomycin), tretinomycin (tretinomycin) inhibitor, tretinomycin (e-derived receptor (tretinomycin), tretinomycin (inhibitor, tretinomycin (e (tretinomycin) inhibitor, tretinomycin), tretinomycin (e (tretino, Walnuts (verdins), verteporfin (verteporfin), vinblastine sulfate (vinblastine sulfate), vincristine sulfate (vincristine sulfate), vindesine (vindesine), vindesine sulfate (vindesine sulfate), vinepidine sulfate (vinopidine sulfate), vinglycinate sulfate (vinblastine sulfate), vinrosine sulfate (vinleurosine sulfate), vinorelbine (vinorelbine), vinorelbine tartrate (vinorelbine tartrate), vinrosidine sulfate (vinrosidine sulfate), vinblastine (vinxanthene), vinzolidine sulfate (vinzolidine sulfate), vitamin (vitaxin), vorozole (vorozole), zanoterone (zanoterone), zeniplatin (zeniplatin), benzalkonium C (zilascorb), zinostatin (zinostatin), zinostatin stimalamer, and zorubicin hydrochloride (zorubicin hydrochloride), and salts, homologs, analogs, polymorphs, derivatives, enantiomers, and/or functionally equivalent compositions thereof.

In certain embodiments of the invention, the compositions may be combined with a suitable pharmaceutically acceptable carrier, e.g., incorporated into liposomes, incorporated into a polymeric delivery system, or suspended in a liquid, e.g., in dissolved or colloidal form, or combined in other ways as described herein. Generally, pharmaceutically acceptable carriers suitable for use are well known to those of ordinary skill in the art. As used herein, "pharmaceutically acceptable carrier" refers to a non-toxic substance that does not significantly interfere with the effectiveness of the biological activity of the active compound to be administered, but is used as a formulation ingredient, e.g., to stabilize or protect the active compound within the composition prior to use. The carrier may comprise one or more compatible solid or liquid fillers, diluents or encapsulating substances suitable for administration to humans or other vertebrates. The term "carrier" denotes an organic or inorganic ingredient, which may be natural or synthetic, with which one or more active compounds of the invention are associated to facilitate application. The carriers can be blended or otherwise mixed with one or more of the compositions described herein, and/or with each other, in such a manner that the interaction therebetween does not significantly impair the desired pharmaceutical efficacy. Depending on the application, the carrier may be soluble or insoluble. Examples of well-known carriers include, but are not limited to, glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylase, natural and modified cellulose, polyacrylamide, agarose, and magnetite. The nature of the carrier may be soluble or insoluble.

In some embodiments, the formulations described herein may be administered in a pharmaceutically acceptable solution, which may typically contain salts, buffers, preservatives, compatible carriers, adjuvants, emulsifiers, diluents, excipients, chelating agents, fillers (fillers), drying agents, antioxidants, antimicrobials, preservatives, binders, fillers (bulking agents), silica, solubilizers, stabilizers, and optionally other therapeutic ingredients, in pharmaceutically acceptable concentrations and with which the active compound may be used. For example, if the formulation is a liquid, the carrier may be a solvent, a partial solvent or a non-solvent, and may be aqueous or organic based. Non-limiting examples of suitable formulation ingredients include diluents such as calcium carbonate, sodium carbonate, lactose, kaolin, calcium phosphate or sodium phosphate; granulating and disintegrating agents, such as corn starch or alginic acid; binding agents, such as starch, gelatin or acacia; lubricants, such as magnesium stearate, stearic acid or talc; a time delay material, such as glyceryl monostearate or glyceryl distearate; suspending agents such as sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone; dispersing or wetting agents, such as lecithin or other natural phospholipids; thickeners such as cetyl alcohol or beeswax; buffers such as acetic acid and salts thereof, citric acid and salts thereof, boric acid and salts thereof, or phosphoric acid and salts thereof; or preservatives, such as benzalkonium chloride, chlorobutanol, parabens, or thimerosal. The composition of the present invention may be formulated into preparations in the form of solid, semi-solid, liquid or gas, such as tablets, capsules, elixirs, powders, granules, ointments, solutions, depots, inhalants or injections, etc.

Suitable buffers include, but are not limited to, acetic acid and salts (1-2% w/v); citric acid and salts (1-3% w/v); boric acid and salts (0.5-2.5% w/v); and phosphoric acid and salts (0.8-2% w/v). Suitable preservatives include, but are not limited to, benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).

Formulations include sterile aqueous or nonaqueous solutions, suspensions, and emulsions, which may, in certain embodiments, be isotonic with the blood of the subject. Non-limiting examples of non-aqueous solvents are polypropylene glycol, polyethylene glycol, vegetable oils such as olive oil, sesame oil, coconut oil, peanut oil (arachis oil), peanut oil (peanout oil), mineral oils, injectable organic esters such as ethyl oleate, or fixed oils including synthetic mono-or diglycerides. Aqueous carriers include, but are not limited to, water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles include sodium chloride solution, 1, 3-butanediol, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Intravenous vehicles include liquid and nutritional supplements, electrolyte supplements (e.g., ringer's dextrose based electrolyte supplements), and the like. Preservatives and other additives may also be present in some embodiments, such as, for example, antimicrobials, antioxidants, chelating agents, and inert gases and the like.

In some embodiments, the compositions described herein may be combined or contacted with a suitable carrier, which may constitute one or more accessory ingredients. The final composition can be prepared by any suitable technique, for example, by uniformly and intimately bringing the composition into association with a liquid carrier, a finely divided solid carrier, or both, optionally with one or more formulation ingredients as described hereinbefore, and then, if necessary, shaping the product.

The compositions discussed herein, and optionally other therapeutic agents, may be administered as such (neat) or in the form of pharmaceutically acceptable salts. When used in medicine, the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may also be conveniently used in the preparation of pharmaceutically acceptable salts thereof. The term "pharmaceutically acceptable salt" includes salts of the compositions described herein, which are prepared, for example, in combination with an acid or a base. Pharmaceutically acceptable salts can be prepared as alkali metal salts, such as lithium, sodium or potassium salts; or an alkaline earth metal salt, such as a beryllium, magnesium or calcium salt. Examples of suitable bases that can be used to form the salts include ammonium or mineral bases such as sodium hydroxide, lithium hydroxide, potassium hydroxide, calcium hydroxide, magnesium hydroxide, and the like. Examples of suitable acids that can be used to form the salts include inorganic or mineral acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, hydrofluoric acid, nitric acid, carbonic acid, monohydroxycarbonic acid, phosphoric acid, monohydroxyphosphoric acid, dihydrogenphosphoric acid, sulfuric acid, monohydroxysulfuric acid, phosphorous acid, and the like. Other suitable acids include organic acids such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, methanesulfonic acid, glucuronic acid, galacturonic acid, salicylic acid, formic acid, naphthalene-2-sulfonic acid, and the like. Other suitable acids also include amino acids such as arginine, aspartic acid, glutamic acid, and the like. In addition, the salts may be prepared as alkali metal or alkaline earth metal salts, such as sodium, potassium or calcium salts of carboxylic acid groups.

In another aspect, the invention also provides any of the compositions described above in a kit, optionally including instructions for use of the composition for treating a tumor. In certain instances, the kit can include a description of the use of the compositions described herein. The kit may also include instructions for the use of a combination of two or more compositions. Instructions for administering the composition by any suitable technique as previously described, e.g., orally, intravenously, by a pump, or by an implantable delivery device, or by another known drug delivery route, may also be provided.

The kits described herein may also comprise one or more containers, which may comprise the compositions and other ingredients as previously described. In certain instances, the kit may further comprise instructions for mixing, diluting, and/or administering the compositions of the present invention. Kits may also include other containers containing one or more solvents, surfactants, preservatives, and/or diluents (e.g., physiological saline (0.9% sodium chloride) or 5% glucose), as well as containers for mixing, diluting, or administering components in a sample to a subject in need of such treatment.

The composition of the kit may be provided in any suitable form, such as a liquid solution or a dry powder. When the composition is provided as a dry powder, the composition may be reconstituted by the addition of a suitable solvent, which may also be provided in some cases. In embodiments where a liquid form of the composition is used, the liquid form may be concentrated or ready for use. The solvent will depend on the composition and the mode of use or administration. Suitable solvents for use in pharmaceutical compositions are well known, e.g. as described previously, and are available in the literature. The solvent will depend on the composition and the mode of use or administration.

In a further aspect, the invention includes the promotion of one or more of the above embodiments, e.g., the promotion of tumor treatment or prevention by in vivo or in vitro administration of a composition as described herein to a subject. As used herein, "promotion" includes all business methods, including educational methods, scientific research, academic research, industrial activities (including pharmaceutical industrial activities), and any advertising or other promotional activities (including any form of written, oral, and electronic communication).

The inventors filed on 16.11.2017, U.S. provisional patent application No. 62/587,207 entitled "a method for lysosomal-induced apoptosis in cancer therapy," and the inventors filed on 3.10.2018, U.S. provisional patent application No. 62/740,477 entitled "system and method for inducing apoptosis to treat cancer," are each incorporated herein by reference in their entirety.

The following examples are intended to illustrate certain embodiments of the invention, but are not intended to illustrate the full scope of the invention.

28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:抗CD40抗体及其用途

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!