AGENT FOR CONTROLLING CELLS CONSTITUTING CANCER MICROENVIRONMENT OR INFLAMMATORY MICROENVIRONMENT

- The University of Tokyo

An agent according to the present invention comprises as an effective component any of (1) disulfiram, diethyldithiocarbamate, or a metal complex of diethyldithiocarbamate; (2) a pharmaceutically acceptable salt of (1); or (3) a solvate of (1) or (2), and is used for inhibition of interaction between CR2B or CCR5 and FROUNT protein, inhibition of macrophages, control of cells constituting a cancer microenvironment or inflammatory microenvironment, or enhancement of anticancer activity of an anticancer drug. It is also possible to provide a compound with a reduced side effect and an increased pharmacological effect by identifying a disulfiram derivative having a lower aldehyde dehydrogenase-inhibiting activity and a higher FROUNT-inhibiting activity among derivatives prepared by structural modification of disulfiram.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a Divisional application of co-pending application Ser. No. 15/542,028, filed on Aug. 8, 2017, which is the National Phase under 35 U.S.C. § 371 of International Application No. PCT/JP2016/050214, filed on Jan. 6, 2016, which claims the benefit under 35 U.S.C. § 119(a) to Patent Application No. 2015-000978, filed in Japan on Jan. 6, 2015, all of which are hereby expressly incorporated by reference into the present application.

TECHNICAL FIELD

The present invention relates to a novel FROUNT protein inhibitor, and control of cells constituting a cancer microenvironment or inflammatory microenvironment using the inhibitor.

BACKGROUND ART

FROUNT protein is a cytoplasmic protein that binds to the intracellular C-terminal regions of chemokine receptors CCR2 and CCR5, and positively controls migration signals of macrophages and the like (Patent Document 1, Non-patent Documents 1 and 2). This protein is a novel molecule discovered by the group of the present inventors.

Both CCR2 and CCR5 are known to be involved in cancers and inflammatory diseases, and development of inhibitors for CCR2 and CCR5 has been attempted worldwide aiming at discovery of novel therapeutic agents for these diseases. However, none of these attempts has been successful. The targets of the existing approaches have been the binding between a chemokine CCL2 and a receptor CCR2, the binding between chemokines CCL3 to 5 and a receptor CCR5, and the signal transduction system by PI3K and the like functioning downstream of the receptors. Inhibition of binding of FROUNT protein to the chemokine receptors CCR2 and CCR5 has been expected as a novel drug discovery target (Non-patent Documents 3 and 4).

The present inventors have reported an inhibitor comprising as an effective component a compound represented by the following formula or a salt thereof, as an inhibitor that inhibits interaction between CCR2 or CCR5 and FROUNT protein (Patent Document 1).

(wherein x1 and x2 are the same or different halogens, and R is a lower alkyl.)

Disulfiram has an aldehyde dehydrogenase-inhibiting activity, and inhibits ethanol metabolism in the liver to cause accumulation of acetaldehyde, which is responsible for sickness caused by drinking, in the body. Thus, after taking of disulfiram, symptoms of sickness caused by drinking occur even with a small amount of alcohol. By utilization of this action, disulfiram is used as an anti-alcoholism drug for treatment of chronic alcoholism.

Besides the above-described action, disulfiram is reported to have an action to inhibit the proliferation of cancer cells by induction of their apoptosis (for example, Non-patent Documents 5 to 7). Disulfiram is also reported to have an action to kill hepatic cancer stem cells (Patent Document 2). All these reports are reports on actions for killing cancer cells or cancer stem cells per se. Because of such direct actions on cancer cells, clinical trials targeting cancer are being partially carried out (Non-patent Document 8). However, actions of disulfiram on microenvironment-constituting cells such as immune cells, fibroblasts, vascular endothelial cells and the like present in the vicinity of cancer cells have not been known at all. Actions of disulfiram on inflammatory microenvironments in diseases in which inflammation is involved have not also been known at all.

PRIOR ART DOCUMENT(S) Patent Document(s)

Patent Document 1: JP 5424960 B

Patent Document 2: JP 2013-100268 A

Non-Patent Document(s)

Non-patent Document 1: Nat. Immunol. Vol. 6, pp. 827-835 (2005)

Non-patent Document 2: J Immunol. Vol. 183, pp. 6387-6394 (2009)

Non-patent Document 3: Endocrinology, Diabetology & Metabolism, 35(6): 500-507 (2012)

Non-patent Document 4: Gan Kiban Seibutsugaku—Kakushinteki Seeds Ikusei Ni Mukete—(Cancer Basic Biology—Towards Cultivation of Innovative Seeds—), Nanzando Co., Ltd., 2013, p. 130-136

Non-patent Document 5: Cancer Research, Vol. 66, pp. 10425-10433 (2006)

Non-patent Document 6: Clinical Cancer Research, Vol. 15, pp. 6070-6078 (2009)

Non-patent Document 7: Molecular Cancer Therapeutics, Vol. 1, pp. 197-204 (2002)

Non-patent Document 8: The Oncologist, Vol. 20, pp. 366-367 (2015)

SUMMARY OF THE INVENTION Problems to be Solved by the Invention

In treatment of cancers and treatment of diseases in which inflammation is involved, it is expected that not only agents that directly act on target cells that cause the diseased state, but also appropriate control of cells constituting the cancer lesion or cells constituting the inflammatory lesion, may lead to a cure. An object of the present invention is to provide a substance whose effect on cancers and inflammatory diseases is higher than those of known FROUNT inhibitors, and which is therefore more useful as a pharmaceutical, and to provide novel means for providing such a substance.

Means for Solving the Problems

As a result of intensive screening of a library containing about 130,000 kinds of low molecular compounds, the present inventors newly identified substances having an activity that inhibits FROUNT protein, and discovered that the substances inhibit excessive hyperplasia and accumulation of microenvironment-constituting cells present in the vicinity of cancer cells, especially accumulation of macrophages in the lesion, resulting in inhibition of the proliferation and metastasis of cancer cells, and that the substances also have an action to inhibit accumulation of macrophages in inflammatory sites and migration of leukemia cells, so that the substances are also effective for inflammatory diseases and humoral cancers. The present inventors further identified the spatial structure of the binding region complex of FROUNT protein and CCR2, and obtained a spatial structure information that is useful for development of a stronger FROUNT protein inhibitor, thereby completing the present invention.

That is, the present invention provides an agent for inhibiting macrophages comprising as an effective component any of the following (1) to (3):

    • (1) disulfiram, diethyldithiocarbamate, or a metal complex of diethyldithiocarbamate;
    • (2) a pharmaceutically acceptable salt of (1);
    • (3) a solvate of (1) or (2).

The present invention also provides an agent for controlling cells constituting a cancer microenvironment or inflammatory microenvironment, said agent comprising as an effective component any of the above-described (1) to (3). The present invention further provides an agent for inhibiting interaction between CCR2B or CCR5 and

FROUNT protein, said agent comprising as an effective component any of the above-described (1) to (3). The present invention still further provides an agent for enhancing anticancer activity of an anticancer drug, said agent comprising as an effective component any of the above-described (1) to (3). The present invention still further provides a method for inhibiting macrophages, a method for inhibiting cells constituting a cancer microenvironment or inflammatory microenvironment, a method for inhibiting interaction between CCR2B or CCR5 and FROUNT protein, and a method for enhancing anticancer activity of an anticancer drug, said methods comprising administering an effective amount of any of the above-described (1) to (3) to a subject in need thereof.

The present invention still further provides a method for identifying a disulfiram derivative having an improved ability to inhibit interaction between FROUNT protein and CCR2B or CCR5, said method comprising the steps of: incubating a FROUNT protein fragment containing the region of the 564th to 600th amino acids of FROUNT protein, and a CCR2B fragment containing the region of the 312th to 323rd amino acids in the amino acid sequence of CCR2B shown in SEQ ID NO:6 or a CCR5 fragment containing the region of the 304th to 315th amino acids in the amino acid sequence of CCR5 shown in SEQ ID NO:8, together with a disulfiram derivative library; and selecting a disulfiram derivative having a higher activity to inhibit binding of the FROUNT protein fragment to the CCR2B fragment or CCR5 fragment compared to disulfiram. The present invention still further provides a method for producing an agent for inhibiting interaction between FROUNT protein and CCR2B or CCR5, said method comprising the steps of: identifying a disulfiram derivative having an improved ability to inhibit interaction between FROUNT protein and CCR2B or CCR5 by the method of the present invention described above; and producing the identified disulfiram derivative. The present invention still further provides a method for identifying a compound having an improved ability to inhibit interaction between FROUNT protein and CCR2B or CCR5, said method comprising the steps of: incubating a FROUNT protein fragment containing the region of the 564th to 600th amino acids of FROUNT protein, and a CCR2B fragment containing the region of the 312th to 323rd amino acids in the amino acid sequence of CCR2B shown in SEQ ID NO:6 or a CCR5 fragment containing the region of the 304th to 315th amino acids in the amino acid sequence of CCR5 shown in SEQ ID NO:8, together with a library of derivatives of a candidate FROUNT inhibitor compound; and selecting a derivative having a higher activity to inhibit binding of the FROUNT protein fragment to the CCR2B fragment or the CCR5 fragment compared to the original candidate compound. The present invention still further provides a method for identifying a substance that inhibits interaction between FROUNT protein and CCR2B or CCR5, said method comprising: constructing a binding pocket structure of FROUNT protein constituted by amino acid residues including at least one selected from the group consisting of M564, T565, 1568, A569, M575, L578, and L600 in silico using at least part of the atomic coordinates of FROUNT protein; calculating the strength of binding of the binding pocket structure to a compound library; and selecting a compound that forms a stable complex with FROUNT protein. The present invention still further provides a method for designing a substance that inhibits interaction between FROUNT protein and CCR2B or CCR5, said method comprising: allowing a candidate compound to bind in silico to a binding pocket structure in which FROUNT protein binds to CCR2B or CCR5; and calculating the strength of the binding.

Effect of the Invention

By the present invention, a novel agent for inhibiting microenvironment-constituting cells including macrophages was provided. The agent of the present invention does not kill cancer cells per se, but inhibits excessive hyperplasia and accumulation of microenvironment-constituting cells present in the vicinity of cancer cells, especially accumulation of macrophages in the lesion, resulting in inhibition of the proliferation and metastasis of cancer cells. The agent of the present invention has an action to inhibit accumulation of macrophages in inflammatory sites and migration of leukemia cells, so that the agent is also effective for inflammatory diseases and humoral cancers. Based on the microenvironment controlling action, the agent is expected to be widely applied to various types of cancers and other inflammatory diseases. By use of the agent of the present invention in combination with a known anticancer drug, the anticancer effect of the anticancer drug can be increased. Therefore, the agent of the present invention is expected to be widely applied to patients in whom use of an anticancer drug alone was not effective. It is also expected that side effects and the medical cost can be reduced thanks to reduction of the dose or dosage of an anticancer drug. Disulfiram has been conventionally practically used as an anti-alcoholism drug for treatment of patients with chronic alcoholism. By identifying a disulfiram derivative having a lower aldehyde dehydrogenase-inhibiting activity and a higher FROUNT-inhibiting activity among derivatives prepared by structural modification of disulfiram, a compound having a reduced side effect and an increased pharmacological effect can also be provided.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1A shows the result of a binding inhibition assay according to the HTRF method. Disulfiram selectively inhibited the FROUNT-CCR2 interaction and the FROUNT-CCR5 interaction, but did not inhibit the p53-MDM2 interaction. FIG. 1B is a graph showing patterns of inhibition of the CCR2-FROUNT interaction and the p53-MDM2 interaction by disulfiram.

FIG. 2A shows the result of investigation of binding of disulfiram to FROUNT by SPR. FIG. 2B shows a spatial structure model of the complex between disulfiram and the disulfiram-binding region on a FROUNT C-terminal fragment identified by NMR analysis. The FROUNT C-terminal fragment is shown as a ribbon model, and disulfiram is shown as a hard-sphere model diagram.

FIG. 3 shows the result of measurement of the tumor size in mice orally fed with a disulfiram-containing food or a control food. * indicates a significant difference at p<0.05.

FIG. 4 shows photographs of metastatic nodules in lungs on Day 9 post tumor administration, and a graph showing the result of counting of metastasis.

FIGS. 5A and 5B show the result of investigation of the growth-inhibiting actions and the cytotoxic activities of disulfiram and SFU. FIG. 5A, LLC tumor cells; FIG. 5B, B16F10 melanoma cells.

FIG. 6 shows the expression level of FROUNT in each type of cells collected from peritoneal inflammatory sites in wild-type mice and FROUNT-GFP knock-in mice. The expression level in each type of the cells was detected by flow cytometry analysis using antibodies against surface markers of various immune cells. Gray curves correspond to cells derived from the wild-type mice, and black curves correspond to cells derived from the GFP knock-in mice.

FIGS. 7A and 7B show the tumor volume (FIG. 7A) in and the survival rate (FIG. 7B) of FROUNT-deficient mice (FNT-cKO) in which FROUNT was conditionally knocked out and non-deficient mice (Flox/flox) transplanted with melanoma cells.

FIGS. 8A and 8B show the number and the size of metastatic nodules in lungs in a lung metastasis model of FROUNT-deficient mice (FROUNT-cKO) and non-deficient mice (Flox/flox).

FIG. 9 shows the number of macrophages (upper panel) and the number of neutrophils (lower panel) in cell populations collected from peritoneal inflammatory sites in FROUNT-deficient mice (FROUNT-cKO) and non-deficient mice (Flox/flox).

FIG. 10 shows the ratios of macrophages (left) and neutrophils (right) in cell populations collected from peritoneal inflammatory sites in wild-type mice to which disulfiram was administered. Disulfiram was administered to the mice at the doses shown in the figure.

FIG. 11 shows the result of in vitro investigation of the cell migration-inhibiting activity of disulfiram using a human leukocyte cell line THP-1.

FIG. 12 shows the numbers of macrophages and neutrophils in lung metastasis in each group of mice receiving solvent treatment or disulfiram treatment investigated by flow cytometry (left), and accumulation of macrophages in the vicinity of metastatic nodules investigated by immunohistochemical staining (right). The broken lines in the immunostaining images indicate the positions of tumor metastatic lesions.

FIG. 13 shows the effect of combined use of disulfiram (DSF) and an anti-PD-1 antibody investigated in an LLC tumor-bearing model. The left panel shows changes in the tumor volume with time, and the right panel shows the tumor volume on Day 26 post tumor cell transplantation. Cont/PBS indicates a non-administration group; DSF/PBS indicates a DSF administration group; Cont/PD-1Ab indicates an anti-PD-1 antibody administration group; and DSF/PD-1Ab indicates a DSF +anti-PD-1 antibody combination group.

FIG. 14 shows the effect of combined use of disulfiram (DSF) and an anti-PD-1 antibody investigated in a B16 tumor-bearing model. The left panel shows changes in the tumor volume with time, and the right panel shows the tumor volume on Day 21 post tumor cell transplantation. Cont/PBS indicates a non-administration group; DSF/PBS indicates a DSF administration group; Cont/PD-1Ab indicates an anti-PD-1 antibody administration group; and DSF/PD-1Ab indicates a DSF +anti-PD-1 antibody combination group. ns, no significant difference; *, significant difference at p<0.05.

FIG. 15 shows the effects of combined use of disulfiram (DSF) and an anti-CD4 antibody investigated in an LLC tumor-bearing model (left) and in a B16 tumor-bearing model (right). Cont/PBS indicates a non-administration group; DSF/PBS indicates a DSF administration group; Cont/CD4Ab indicates an anti-CD4 antibody administration group; and DSF/CD4Ab indicates a DSF +anti-CD4 antibody combination group.

MODE FOR CARRYING OUT THE INVENTION

In the present invention, any of the followings is used as an effective component.

    • (1) Disulfiram, diethyldithiocarbamate, or a metal complex of diethyldithiocarbamate.
    • (2) A pharmaceutically acceptable salt of (1).
    • (3) A solvate of (1) or (2).

The present inventors revealed that disulfiram has an action to inhibit binding (interaction) of FROUNT protein to CCR2B and CCR5, and that disulfiram can therefore be used as a FROUNT inhibitor. Disulfiram is metabolized in the body to generate diethyldithiocarbamate. It has been shown that compounds from which diethyldithiocarbamate is generated have a FROUNT-inhibiting action and a cell migration-inhibiting activity and the like produced from the action.

FROUNT protein is a protein that was identified by the present inventors. Its sequence information has been deposited in an NCBI database GenBank under Accession Nos. AF498261 and NM_024844. The sequences shown in SEQ ID NOs:1 and 2 in SEQUENCE LISTING are cDNA of the FROUNT gene deposited under AF498261 and the amino acid sequence of the FROUNT protein encoded thereby.

There are two isoforms in CCR2, that is, CCR2A and CCR2B, having different C-terminal regions. The major isoform is CCR2B. It has been shown that FROUNT protein binds to the membrane-proximal region in the intracellular C-terminal region of CCR2B and CCR5 (EKFRRYLSVFFRKHIT (SEQ ID NO:3) in CCR2B, and EKFRNYLLVFFQKHIA (SEQ ID NO:4) in CCR5) (Biochem. J. (2014) 457, 313-322). In the present description, when the term “CCR2” is simply mentioned, it means CCR2B unless the context clearly indicates otherwise. SEQ ID NOs:5 and 6 show CCR2B sequences (GenBank NM_001123396.1), and SEQ ID NOs:7 and 8 show CCR5 sequences (GenBank NM_000579.3).

Disulfiram controls cells constituting the microenvironments of lesions in cancers and inflammatory diseases by inhibition of FROUNT protein. Disulfiram can therefore be used as an agent for controlling cells constituting the microenvironments of lesions in these diseases. In the present invention, the terms “inhibition of FROUNT protein” and “FROUNT inhibition” mean inhibition of interaction between FROUNT and CCR2 or CCR5.

The microenvironment means an environment in a body in which cells derived from two or more kinds of tissues coexist. Examples of the cells constituting the microenvironments in lesions of cancers and inflammatory diseases (which may also be abbreviated as “microenvironment-constituting cells” in the present description) include immune cells (macrophages, dendritic cells, T cells, B cells, and the like), fibroblasts, vascular endothelial cells, pericytes, inflammatory cells (eosinophils, mast cells, neutrophils, basophils, and the like), and somatic stem cells. In the present invention, the term “cancer microenvironment-constituting cells” means microenvironment-constituting cells other than cancer cells.

The control of microenvironment-constituting cells mainly refers to inhibition of these cells. In cancer microenvironments and inflammatory microenvironments, the cells constituting the environments show abnormal infiltration from the outside and abnormal accumulation in these environments, as well as abnormal hyperplasia in the environments. The control of microenvironment-constituting cell means inhibition of such abnormal infiltration and hyperplasia. By controlling cells that constitute the microenvironments in lesions of cancers and inflammatory diseases, these diseases can be treated or prevented, or their progression, exacerbation, metastasis, or recurrence can be prevented.

Control of microenvironment-constituting cells may be, for example, inhibition of macrophages. Examples of the inhibition of macrophages include inhibition of migration or tissue infiltration of macrophages. It is known that excessive accumulation and infiltration of immune cells such as macrophages are involved in development and exacerbation of a number of cancers and inflammatory diseases, and in induction of metastasis and the like in cancers. Thus, by inhibition of macrophage infiltration in the microenvironments in lesions of cancers and inflammatory diseases, treatment, prophylaxis, prevention of progression, prevention of exacerbation, prevention of metastasis, prevention of recurrence, and the like of these diseases become possible. However, the action to control microenvironment-constituting cells by the agent of the present invention is not limited to inhibition of macrophages.

FROUNT protein, which binds to the intracellular C-terminal region of CCR2 and CCR5 to cause the downstream signal transduction, is inhibited by disulfiram.

Thus, the agent of the present invention is effective for cancers and inflammatory diseases in which CCR2 or CCR5, or their ligand (CCL2 or CCL5) is known to be involved, or cancers and inflammatory diseases for which an inhibitor of these molecules (chemokine inhibitor) is known to be effective. The agent of the present invention is also effective for cancers and inflammatory diseases with microenvironments in which FROUNT is expressed, and the higher the expression level of FROUNT, the higher the effect is expected to be.

Examples of the cancers to be targeted include both solid cancers and humoral cancers, and also include both primary cancers and metastatic cancers. Specific example of the cancers include, but are not limited to, lung cancer, melanoma, gastric cancer, colon cancer, breast cancer, liver cancer, pancreatic cancer, uterine cancer, esophageal cancer, prostate cancer, malignant lymphoma, and leukemia. Known examples of cancers in which CCR2 is involved include melanoma, breast cancer, prostate cancer, lung cancer, myeloma, and brain tumor, and known examples of cancers in which CCR5 is involved include breast cancer, prostate cancer, lung cancer, pancreatic cancer, and myeloma (Scholten DJ, et al., Br J Pharmacol, 165: 1617-1643, 2012). Examples of anticancer drugs in the field of chemokines that have advanced to clinical trials include an anticancer drug for metastatic castration-resistant prostate cancer targeting CCL2, an anticancer drug for non-small cell lung cancer targeting CCL5, an anticancer drug for metastatic cancers targeting CCR2, and an anticancer drug for advanced colon cancer targeting CCR5 (Gan Kiban Seibutsugaku—Kakushinteki Seeds Ikusei Ni Mukete—(Cancer Basic Biology—Towards Cultivation of Innovative Seeds—), Nanzando Co., Ltd., 2013, p. 130-136). These cancers are preferred specific examples to be targeted by the present invention.

The inflammatory diseases to be targeted in the present invention are typically chronic inflammatory diseases. Specific examples of the inflammatory diseases include, but are not limited to, rheumatoid arthritis, fibrosis, peritonitis, multiple sclerosis, arteriosclerosis, diabetes, asthma, Alzheimer's disease, psoriasis, atopic diseases, ischemic heart diseases, and cerebrovascular diseases. Examples of inflammatory diseases in which at least one of CCR2, CCR5, CCL2, and CCL5 is known to be involved include arteriosclerosis, multiple sclerosis, rheumatoid arthritis, psoriasis, type 2 diabetes, inflammatory bowel disease, chronic hepatitis, nephritis, graft-versus-host disease, chronic obstructive lung disease, asthma, and acquired immune deficiency syndrome (Scholten DJ et al., Br J Pharmacol, 165: 1617-1643, 2012; Clinical Immunology & Allergology, 59(3): 386-391, 2013). Other examples of inflammatory diseases in which at least one of CCR2, CCR5, CCL2, and CCL5, or FROUNT is involved include various fibrotic diseases such as pulmonary fibrosis and hepatic fibrosis; peritonitis; and allergic airway hyperresponsiveness (Nippon Rinsho, vol. 70, extra edition 8, 365-371, 2012; and the Examples described below). These inflammatory diseases are preferred specific examples to be targeted by the present invention.

The metal complex of diethyldithiocarbamate may be a complex of any metal. Specific examples of the metal complex include, but are not limited to, zinc complex, iron (II) complex, iron (III) complex, copper complex, and platinum complex.

Disulfiram, diethyldithiocarbamate, or the metal complex of diethyldithiocarbamate may also be used in the form of a pharmaceutically acceptable salt. The salt may be an acid addition salt or a base addition salt. Specific examples of the acid addition salt include inorganic acid salts such as hydrochloric acid salt, hydrobromic acid salt, sulfuric acid salt, hydroiodic acid salt, nitric acid salt, and phosphoric acid salt; and organic acid salts such as citric acid salt, oxalic acid salt, acetic acid salt, formic acid salt, propionic acid salt, benzoic acid salt, trifluoroacetic acid salt, maleic acid salt, tartaric acid salt, methanesulfonic acid salt, benzenesulfonic acid salt, and para-toluenesulfonic acid salt. Specific examples of the base addition salt include inorganic base salts such as sodium salt, potassium salt, calcium salt, magnesium salt, and ammonium salt; and organic base salts such as triethylammonium salt, triethanolammonium salt, pyridinium salt, and diisopropylammonium salt.

Disulfiram, diethyldithiocarbamate, or the metal complex of diethyldithiocarbamate, or the pharmaceutically acceptable salt thereof may also be used in the form of a solvate. Specific examples of the solvate include, but are not limited to, hydrates and ethanolates. The solvate may be any solvate as long as it is a solvate with a pharmaceutically acceptable solvent.

Disulfiram per se is a known compound, and is conventionally used as an anti-alcoholism drug for treatment of chronic alcoholism. Disulfiram is a prescription medication listed in the Japanese Pharmacopoeia, and its production method is well known. Diethyldithiocarbamate and its metal complexes, and the pharmaceutically acceptable salts and solvates of disulfiram and the like described above can also be produced by methods known in the field of chemical synthesis.

When the agent of the present invention is used as a pharmaceutical, the administration route may be systemic administration or local administration, and may be oral administration or parenteral administration. Examples of the parenteral administration include intramuscular administration, subcutaneous administration, intravenous administration, intraarterial administration, and transdermal administration. The agent may be locally administered in the vicinity of the lesion, or, when it is used for cancer, it may be administered to a regional lymph node in the vicinity of the tumor site.

When the agent of the present invention is prepared as a pharmaceutical, disulfiram, diethyldithiocarbamate, a metal complex of diethyldithiocarbamate, a salt of any of these compounds, or a solvate of any of these compounds or salts thereof may be mixed as appropriate with an additive(s) such as a pharmaceutically acceptable carrier, diluent, and/or vehicle suitable for each administration route, to prepare a formulation. Examples of the formulation include oral preparations such as tablets, capsules, granules, powders, and syrups; and parenteral preparations such as inhalants, injection solutions, suppositories, and solutions. Formulation methods and additives which can be used are well known in the field of pharmaceutical preparations, and any of the methods and the additives may be employed.

Techniques for preparing sustained release formulations are also well known. The agent of the present invention may be provided as a sustained release formulation aiming at stabilization and retention of the blood level of the effective component. The term “sustained release” used herein has the same meaning as controlled release, and also includes delayed release and the like. The techniques for preparing sustained release formulations can be classified into the single-unit type and the multiple-unit type based on the form of the sustained release formulation, or can be classified into the reservoir type, matrix type and the like based on the release control mechanism. Hybrid types, in which a plurality of mechanisms are combined, are also known. When the agent of the present invention is prepared as a sustained release formulation, any of the techniques for preparation of sustained release formulations may be used. A DDS such as liposomes may be used for the preparation. The sustained release formulation may be prepared into any dosage form including a tablet, granule, capsule or the like. Specific examples of the sustained release formulation of disulfiram include the disulfiram formulation described in WO 2012/076897 A1, in which liposomes are used as a DDS, and the solid dispersion tablet of disulfiram described in International Journal of Pharmaceutics 497 (2016) 3-11, in which a polyvinyl acetate - polyvinyl pyrrolidone mixture or hypromellose is used as a sustained release polymer. However, the sustained release formulation of disulfiram is not limited to these specific examples.

The administration amount of the agent of the present invention may be any amount as long as it is effective for treatment of the cancer or inflammatory disease to be targeted. The effective amount is appropriately selected depending on, for example, the size of the lesion, symptoms, severity, the age and/or the body weight of the patient, and/or the like. Although the administration amount of the agent of the present invention is not limited, it may be about 0.001 mg to about 10 g, for example, about 0.1 mg to about 1000 mg, or about 5 mg to about 500 mg, or about 5 mg to 200 mg, in terms of the amount of the effective component per administration to an adult (60 kg body weight). The agent may be administered once or several times per day. During the treatment period, the agent may be administered once, or daily for a period of several days or longer, or may be administered multiple times every several days, every several weeks, or every several months. For example, multiple times (for example, about 2 to 5 times) of administration per day may be carried out everyday during the treatment period. As described in the following Examples, the FROUNT-inhibiting ability of disulfiram is obtained by administering disulfiram in a form which can be degraded into DDC, and is lost in a form of the downstream metabolites. Thus, frequent administration is more preferred than once daily administration at a high dose. However, depending on the performance of the sustained release formulation, the frequency of administration can be reduced.

The patient to which the agent of the present invention is administered is a mammal. Although the mammal is not limited, it is typically human.

The agent of the present invention may be used in combination with at least one of known anticancer drugs and anti-inflammatory drugs. The term “used in combination” means that the agent of the present invention and the at least one anticancer drug or anti-inflammatory drug are administered to a patient simultaneously, sequentially, or separately. The agent and a drug(s) to be used in combination may be provided as separate formulations, or, in cases where they are simultaneously administered, the effective components of the agent and a drug(s) may be contained in a single formulation.

The term “anticancer” includes inhibition of development (initiation, metastasis, or recurrence) of cancer and inhibition of growth of cancer. Accordingly, “anticancer drug” includes therapeutic agents, prophylactic agents, metastasis-inhibiting agents, and recurrence-inhibiting agents for cancer.

The anticancer drug that may be used in combination with the agent of the present invention may be an antibody or an antigen-binding fragment thereof. The antibody is preferably a monoclonal antibody, and, in cases where the subject to which it is administered is human, a human type chimeric antibody, humanized antibody (an antibody prepared by transplanting the CDR region of a non-human-derived antibody to the corresponding region of a human antibody), or human antibody (the same antibody as an antibody produced in the body of human, which is prepared using a non-human animal or a human cell line) is preferably used.

Preferred examples of the anticancer drug that may be used in combination include anticancer drugs targeting immune checkpoints. Immune checkpoints are the immune escape mechanism to prevent the immune system from attacking its own body. Immune checkpoint receptors are present on T cells, and interact with ligands expressed on antigen-presenting cells. T cells recognize an antigen presented on the MHC molecule and are activated to generate an immune reaction, whereas the activation of T cells is controlled by an interaction between immune checkpoint receptor and ligand that occurs in parallel. Immune checkpoint receptors can be divided into inhibitory receptors and co-stimulatory receptors, and the T cell activation and the immune reaction are controlled by a balance between both receptors.

Cancer cells utilize such an immune checkpoint mechanism. By expressing a ligand for an inhibitory immune checkpoint receptor, cancer cells suppress the immune function to escape from attack of cytotoxic T cells. Therefore, inhibition of inhibitory immune checkpoint molecules can prevent cancer cells from utilizing the immune checkpoint mechanism, thereby facilitating killing of cancer cells by CD8+ T cells.

Various kinds of anticancer drugs targeting immune checkpoints have been developed, and their clinical trials and practical use have progressed worldwide. Agents that inhibit inhibitory immune checkpoints are also called immune checkpoint inhibitors, and their practical use has especially progressed among the anticancer drugs targeting immune checkpoints. Specific examples of such immune checkpoint inhibitors include antagonistic anti-PD-1 antibodies and antagonistic anti-CTLA-4 antibodies, as well as anti-PD-L1 antibodies and anti-PD-L2 antibodies that bind to PD-L1 and PD-L2, which are ligands for the receptor PD-1, to inhibit binding of the ligands to the receptor.

However, the anticancer drugs targeting immune checkpoints are not limited to immune checkpoint inhibitors (antagonists against inhibitory immune checkpoint molecules), and agonists against co-stimulatory immune checkpoint molecules may also be used in combination with the agent of the present invention. Administration of an agonist against a co-stimulatory immune checkpoint receptor can enhance the immune reaction, by which killing of cancer cells by CD8+ T cells can also be facilitated. The agent of the present invention, an immune checkpoint inhibitor, and an agonist against a co-stimulatory immune checkpoint molecule may be also used in combination.

In the present invention, the term “immune checkpoint molecule” includes both receptors and ligands that function as an immune checkpoint.

Specific examples of inhibitory immune checkpoint molecules that can be targeted by anticancer drugs targeting immune checkpoints include receptors such as PD-1, CTLA-4, LAG-3, TIM-3, and BTLA; and ligands such as PD-L1 (ligand for PD-1), PD-L2 (ligand for PD-1), CD80 (ligand for CTLA-4), CD86 (ligand for CTLA-4), GALS (ligand for TIM-3), and HVEM (ligand for BTLA). Specific examples of co-stimulatory immune checkpoint molecules that can be targeted include receptors such as CD137, OX40, and GITR; and ligands such as CD137L (ligand for CD137), OX40L (ligand for OX40), and TNF SF18 (ligand for GITR).

In the present invention, the term “antagonist” includes various substances that interfere with receptor activation induced by binding between receptor and ligand. Examples of the antagonist include substances that interfere with the binding between receptor and ligand by binding to the receptor, and substances that interfere with the binding between receptor and ligand by binding to the ligand.

“An antagonist against an inhibitory immune checkpoint molecule” may be an antagonistic antibody that binds to an inhibitory immune checkpoint receptor (such as antagonistic anti-PD-1 antibody, anti-CTLA-4 antibody, anti-LAG-3 antibody, anti-TIM-3 antibody, anti-BTLA antibody, or the like); an antibody that binds to an inhibitory immune checkpoint ligand to inhibit its binding to the receptor (such as anti-PD-L1 antibody, anti-PD-L2 antibody, anti-CD80 antibody, anti-CD86 antibody, anti-GAL9 antibody, anti-HVEM antibody, or the like); a soluble polypeptide which is designed based on an inhibitory immune checkpoint ligand and does not activate the receptor, or a vector capable of expressing the polypeptide, or the like.

“An agonist against a co-stimulatory immune checkpoint molecule” may be an antibody having agonistic activity that binds to a co-stimulatory immune checkpoint receptor (such as agonistic anti-CD137 antibody, anti-OX40 antibody, anti-GITR antibody, or the like); a soluble polypeptide which is designed based on a co-stimulatory immune checkpoint ligand and has an action to activate the receptor, or a vector capable of expressing the polypeptide, or the like.

Methods for producing an antibody, and methods for producing a polypeptide by chemical synthesis or genetic engineering procedure are well-known conventional methods in the art. Those skilled in the art can prepare an antagonist against an inhibitory immune checkpoint molecule or an agonist against a co-stimulatory immune checkpoint molecule as described above by conventional methods. Methods for preparing a chimeric antibody, humanized antibody, or human antibody have also been established as well-known methods in the art. In cases where the anticancer drug targeting an immune checkpoint molecule is an antibody drug, an antigen-binding fragment such as Fab, F(ab′)2, or scFv (single chain fragment of variable region, single-chain antibody) may also be used, and methods for producing antigen-binding fragments are also well known.

Other preferred examples of the anticancer drugs that may be used in combination include anti-CD4 antibodies having cytotoxic activity; and anti-CD4 antibodies and antigen-binding fragments thereof which antibodies and fragments comprise a cytotoxic component bound thereto. The cytotoxic component herein means a substance having an activity to destroy living cells, and includes biological toxic substances, chemical substances, and radioactive substances. It is known that an anti-CD4 antibody having cytotoxic activity and an anti-CD4 antibody or antigen-binding fragment thereof which antibody or fragment comprises a cytotoxic component bound thereto have an antitumor effect on various cancers including blood cancers and solid cancers (for example, WO 2015/125652). The cytotoxic activity may be antibody-dependent cellular cytotoxicity (ADCC activity) or complement-dependent cellular cytotoxicity (CDC activity). It is necessary to use an antibody having high cytotoxic activity by which a sufficiently high ability to kill CD4+ cells can be exerted.

The term “high cytotoxic activity” in the context of the ADCC activity means that an antibody has a higher ADCC activity than the known anti-CD4 antibody 6G5 or CE9.1 that is known to have an ADCC activity, when the ADCC activity against CD4-expressing cells is measured by a known measurement method. In the context of the CDC activity, the term means that an antibody has a stronger CDC activity than the known anti-CD4 antibody OKT4 that is known to have a CDC activity, when the CDC activity against CD4-expressing cells is measured in an experimental system using the same complements by a known measurement method.

Preferably, an anti-CD4 antibody having a high cytotoxic activity has an ADCC activity that is 10 times or more, more preferably 100 times or more higher than the ADCC activity of the known anti-CD4 antibody 6G5 and/or CE9.1, or has a CDC activity that is 10 times or more, more preferably 100 times or more higher than the CDC activity of the known anti-CD4 antibody OKT4. As used herein, the term “10 times or more” means, for example, that the minimum antibody concentration at which a given antibody exhibits a cytotoxic activity against a certain amount of cells is one-tenth or less of that of the above-described known antibody. As for the affinity of the anti-CD4 antibody to CD4, the antibody binding activity KD may be about 1×10−9M or less.

Methods for measurement of the ADCC activity and the CDC activity of antibodies are known and described in e.g. Cancer Immunol. Immunother., 36, 373 (1993), and kits therefor are commercially available. Whether a given antibody has a higher cytotoxic activity than known anti-CD4 antibodies or not may be evaluated using such a commercially available kit. A specific example of measurement of the cytotoxic activity using a commercially available kit is described in the Examples below. The level of the ADCC activity of anti-CD4 antibody can also be evaluated by, as described in the Examples below, mixing human peripheral blood mononuclear cells with the anti-CD4 antibody, allowing the reaction to proceed at 37° C. for several hours, performing flow cytometry analysis to measure the ratio of CD3+ cells to CD8+ cells in the reaction solution, and then comparing the obtained measurement value with a measurement value obtained using an anti-CD4 antibody having no ADCC activity or a known anti-CD4 antibody described above.

An anti-CD4 antibody having a high cytotoxic activity can be prepared, for example, from a monoclonal anti-CD4 antibody prepared by a known method or from an already established known anti-CD4 antibody, by increasing the cytotoxicity of the antibody by a method known in the art. In cases where an anti-CD4 antibody that specifically recognizes CD4 expressed on the cell surface and has a strong cytotoxicity is known, such an antibody may be used as an effective ingredient of the agent of the present invention. For example, WO 2010/074266 discloses an anti-CD4 antibody having a higher ADCC activity than conventional anti-CD4 antibodies.

Methods for increasing the cytotoxic activity of an antibody are also known, and any of these methods may be used. Specific examples of known methods for enhancing the ADCC activity include the POTELLIGENT (registered trademark) technology, in which fucose (core fucose) contained in a sugar chain present in the Fc region of the antibody is removed (Yamane-Ohnuki N, Satoh M, Production of therapeutic antibodies with controlled fucosylation, MAbs 2009; 1: 230-236); a method in which fucose substrate donation is blocked; and a method in which a sugar chain present in the Fc region of the antibody is converted (M. Schuster et al., Improved effector functions of a therapeutic monoclonal Lewis Y-specific antibody by glycoform engineering, Cancer Res 2005; 65: 7934-7941). Examples of known methods for enhancing the CDC activity include the COMPLEGENT (registered trademark) technology, in which a part of the isotype IgG1 is combined with a sequence of the isotype IgG3 to increase the CDC activity (Natsume A, In M, Takamura H, et al. Engineered antibodies of IgG1/IgG3 mixed isotype with enhanced cytotoxic activities, Cancer Res. 2008; 68: 3863-3872). The AccretaMab (registered trademark) technology, in which the POTELLIGENT (registered trademark) technology and the COMPLEGENT (registered trademark) technology are employed in combination to increase both the ADCC activity and the CDC activity, thereby strongly increasing the cytotoxic activity of an antibody (Natsume A, et al., Improving effector functions of antibodies for cancer treatment: Enhancing ADCC and CDC, Drug Des Devel Ther. 2009; 3: 7-16) is also known.

The anticancer drugs that may be used in combination are not limited, and examples of the anticancer drug include: at least one selected from antagonistic antibodies against inhibitory immune checkpoint receptors and antigen-binding fragments thereof, antibodies against inhibitory immune checkpoint ligands and antigen-binding fragments thereof, agonistic antibodies against co-stimulatory immune checkpoint receptors and antigen-binding fragments thereof, anti-CD4 antibodies having cytotoxic activity, and anti-CD4 antibodies and antigen-binding fragments thereof which antibodies and fragments comprise a cytotoxic component bound thereto; at least one selected from antagonistic antibodies against inhibitory immune checkpoint receptors and antigen-binding fragments thereof, antibodies against inhibitory immune checkpoint ligands and antigen-binding fragments thereof, and anti-CD4 antibodies having cytotoxic activity; or at least one selected from antagonistic antibodies against inhibitory immune checkpoint receptors and antigen-binding fragments thereof, and anti-CD4 antibodies having cytotoxic activity. Especially preferred examples of the antagonistic antibodies against inhibitory immune checkpoint receptors include, but are not limited to, antagonistic anti-PD-1 antibodies and antagonistic anti-CTLA-4 antibodies, especially antagonistic anti-PD-1 antibodies.

In cases where the agent of the present invention is used in combination with a known anticancer drug or anti-inflammatory drug, the anticancer drug or anti-inflammatory drug may be used in the same manner as when it is used alone for treatment of cancer or treatment of an inflammatory disease. It is also possible to reduce the dose or dosage, the frequency of administration, the dosing period, etc. of drugs, since an increased effect is obtained thanks to combined use with the agent of the present invention.

Since a higher anticancer effect can be obtained by combined use of an anticancer drug and the agent of the present invention, the agent of the present invention can also be understood as having an effect to enhance the anticancer activity of an anticancer drug. When the agent of the present invention is used as an agent for enhancing anticancer activity of an anticancer drug, the subject to which the agent is administered is a cancer patient, i.e. a patient who is going to receive or receives treatment with the anticancer drug.

By identifying a derivative having a higher pharmacological effect than disulfiram from a library of derivatives having structures prepared by applying various chemical modifications to disulfiram, a compound having a still higher pharmacological effect as a FROUNT inhibitor or an agent for controlling microenvironment-constituting cells can be obtained. Aldehyde dehydrogenase-inhibiting activity of disulfiram is a side effect in application of disulfiram to a patient with a cancer or an inflammatory disease. Thus, reduction of the aldehyde dehydrogenase-inhibiting activity can also be used as an index for selection of an excellent derivative.

“Derivatives” are compounds obtained by introducing various chemical modifications into their original compound, and have structures similar to those of the original compound. In the process of optimization of lead compound in drug discovery, derivatives of the lead compound are sometimes called peripheral compounds. A library of such derivatives can be prepared by, for example, application of a method of combinatorial chemistry. Diethyldithiocarbamate is a compound having a structure generated from cleavage of the S—S bond of disulfiram, and the term “disulfiram derivative” in the present invention includes diethyldithiocarbamate derivatives. The disulfiram derivative may be, for example, a compound having the structure shown in the following Formula 1 or Formula 2, although the disulfiram derivative is not limited thereto.

(wherein in Formula 1,

R1 and R2 are each independently methyl that is optionally substituted by a sulfur atom(s), oxygen atom(s), and/or halogen atom(s); and

R3 to R6 are each independently a hydrogen atom, C1-C5 linear alkyl, or C3-C5 branched alkyl, for example, C1-C3 linear alkyl or C3 branched alkyl. The alkyl is optionally substituted by a halogen atom(s), and one or more carbon atoms constituting the alkyl is optionally replaced by a nitrogen atom(s), sulfur atom(s), and/or oxygen atom(s).)

(wherein in Formula 2, R1, R3, and R4 have the same meanings as in the Formula 1, and X is a hydrogen atom, halogen atom, alkali metal atom, or methyl that is optionally substituted by halogen atom.)

The method for identifying a disulfiram derivative having an improved ability to inhibit interaction between FROUNT protein and CCR2B or CCR5 may include, for example, the following steps.

(1) A FROUNT protein fragment, and a CCR2B fragment or a CCR5 fragment are incubated with a library of disulfiram derivatives.

As described later, the binding pocket structure through which FROUNT protein binds to CCR2B or CCR5 is constituted by a plurality of amino acid residues including at least one of the following seven residues: M564, T565, 1568, A569, M575, L578, and L600. Accordingly, a region containing at least these seven residues, that is, a region containing the 564th to 600th amino acids, is used as the FROUNT protein fragment. It has been shown, by an HTRF experiment which was carried out in the past, that the fragment consisting of the region of the 549th to 656th amino acids and the fragment consisting of the region of the 532nd to 650th amino acids have an ability to bind to the C-terminal membrane-proximal region of CCR2 (data not shown). Thus, examples of the FROUNT protein fragment include a fragment containing the region of the 549th to the 650th amino acids, and a fragment containing the region of the 549th to 656th amino acids or the 532nd to 650th amino acids. Or, the FROUNT protein fragment may be a fragment containing the region of the 500th to the 656th amino acids.

The fragment consisting of the region of the 310th to the 325th amino acids of CCR2B (EKFRRYLSVFFRKHIT) is an especially preferred example of the CCR2B fragment used herein. However, even a fragment lacking a small number of terminal amino acid residues can have an ability to bind to FROUNT. Thus, a fragment containing the region of the 312th to 323rd amino acids, for example, a fragment containing the region of the 310th to 325th amino acids, of the CCR2B amino acid sequence shown in SEQ ID NO:6 can be used as the CCR2B fragment.

An especially preferred example of the CCR5 fragment is a fragment consisting of the region of the 302nd to 317th amino acids (EKFRNYLLVFFQKHIA). However, similarly to the above, even a fragment lacking a small number of terminal amino acid residues can have an ability to bind to FROUNT. Thus, a fragment containing the region of the 304th to 315th amino acids, for example, a fragment containing the region of the 302th to 317th amino acids, of the CCR5 amino acid sequence shown in SEQ ID NO:8 can be used as the CCR5 fragment.

The protein fragments described above can be prepared by a well-known chemical synthesis method or genetic engineering method.

(2) A disulfiram derivative having a higher activity to inhibit the binding between the FROUNT protein fragment and the CCR2B fragment or CCR5 fragment compared to disulfiram is selected. This step can be carried out using a well-known binding assay. The selected disulfiram derivative is useful as a disulfiram derivative having an improved ability to inhibit interaction between FROUNT protein and CCR2B or CCR5, and, in turn, as a disulfiram derivative having an improved ability to control microenvironment-constituting cells in lesions of cancers and inflammatory diseases.
(3) The disulfiram derivative may be further subjected to a step of measuring the aldehyde dehydrogenase-inhibiting activity to select a derivative having a lower level of the activity than that of the original disulfiram. This step is usually carried out after the step (2).

Candidate FROUNT inhibitor substances found by screening or the like other than disulfiram may also be subjected to synthesis/development of derivatives and a binding inhibition assay in the same manner as described above to identify a derivative compound having an improved FROUNT-inhibiting ability.

The derivative compound such as a disulfiram derivative identified as described above can be produced by a method well known in the field of chemical synthesis. The derivative has a better performance than disulfiram as an inhibitor of interaction between CCR2B or CCR5 and FROUNT protein, as an agent for controlling microenvironment-constituting cells such as an agent for controlling macrophage infiltration, and, in particular, as a pharmaceutical.

The present inventors have first clarified the spatial structure of the region in which FROUNT protein binds to CCR2B at the atomic coordinate level. By an NMR analysis of the state of binding between a fragment of the binding region of FROUNT protein and a fragment of the binding region of CCR2B, the following seven residues have been identified as candidates of residues constituting the binding pocket structure:

methionine at position 564 (M564), threonine at position 565 (T565), isoleucine at position 568 (1568), alanine at position 569 (A569), methionine at position 575 (M575), leucine at position 578 (L578), and leucine at position 600 (L600). These seven residues are the top seven residues showing large chemical shift changes in the NMR signal due to formation of the complex, and the binding pocket through which FROUNT binds to CCR2B is constituted by a plurality of residues including at least one of these seven residues. The binding regions through which CCR2B and CCR5 bind to FROUNT, respectively, have almost the same spatial structure, and CCR5 also binds to the binding pocket structure identified herein. Since a variety of methods for drug design based on the spatial structure (SBDD: structure based drug design) and for in silico screening are known, more potent FROUNT inhibitors can be developed by utilizing the spatial structure information of FROUNT protein. Table 1 shows the atomic coordinate data of the seven residues described above, in the protein data bank format.

TABLE 1 Atomic coordinates of 7 residues defining a binding pocket ATOM 1 N MET A 564 −12.289 7.512 −6.831 1.00 75.12 N ATOM 2 CA MET A 564 −12.530 8.940 −6.665 1.00 14.53 C ATOM 3 CB MET A 564 −13.865 9.335 −7.280 1.00 35.23 C ATOM 4 CG MET A 564 −14.069 10.822 −7.515 1.00 62.34 C ATOM 5 SD MET A 564 −15.851 11.125 −7.713 1.00 64.33 S ATOM 6 CE MET A 564 −16.125 10.429 −9.359 1.00 13.31 C ATOM 7 C MET A 564 −12.553 9.320 −5.187 1.00 65.53 C ATOM 8 O MET A 564 −11.998 10.345 −4.790 1.00 24.41 O ATOM 9 H MET A 564 −12.969 6.961 −7.274 1.00 75.12 H ATOM 10 HA MET A 564 −11.724 9.471 −7.148 1.00 14.53 H ATOM 11 HB2 MET A 564 −13.970 8.818 −8.233 1.00 35.23 H ATOM 12 HB3 MET A 564 −14.657 8.969 −6.612 1.00 35.23 H ATOM 13 HG2 MET A 564 −13.731 11.402 −6.656 1.00 62.34 H ATOM 14 HG3 MET A 564 −13.529 11.188 −8.391 1.00 62.34 H ATOM 15 HE1 MET A 564 −15.641 11.055 −10.107 1.00 13.31 H ATOM 16 HE2 MET A 564 −15.752 9.415 −9.440 1.00 13.31 H ATOM 17 HE3 MET A 564 −17.195 10.405 −9.573 1.00 13.31 H ATOM 18 N THR A 565 −13.198 8.486 −4.377 1.00 4.44 N ATOM 19 CA THR A 565 −13.293 8.735 −2.944 1.00 12.13 C ATOM 20 CB THR A 565 −14.633 8.202 −2.394 1.00 14.53 C ATOM 21 OG1 THR A 565 −14.866 6.864 −2.852 1.00 13.12 O ATOM 22 CG2 THR A 565 −15.796 9.086 −2.827 1.00 53.10 C ATOM 23 C THR A 565 −12.157 8.053 −2.190 1.00 73.43 C ATOM 24 O THR A 565 −11.797 8.463 −1.087 1.00 44.02 O ATOM 25 H THR A 565 −13.620 7.686 −4.753 1.00 4.44 H ATOM 26 HA THR A 565 −13.228 9.802 −2.785 1.00 12.13 H ATOM 27 HB THR A 565 −14.610 8.161 −1.299 1.00 14.53 H ATOM 28 HG21 THR A 565 −15.786 10.036 −2.286 1.00 53.10 H ATOM 29 HG22 THR A 565 −16.747 8.580 −2.648 1.00 53.10 H ATOM 30 HG23 THR A 565 −15.756 9.308 −3.898 1.00 53.10 H ATOM 31 HG1 THR A 565 −15.187 6.921 −3.775 1.00 13.12 H ATOM 32 N ILE A 568 −6.488 6.792 −3.580 1.00 2.13 N ATOM 33 CA ILE A 568 −5.657 6.386 −4.706 1.00 71.23 C ATOM 34 CB ILE A 568 −6.073 4.952 −5.170 1.00 74.31 C ATOM 35 CG2 ILE A 568 −5.149 3.922 −4.498 1.00 22.21 C ATOM 36 CG1 ILE A 568 −7.542 4.597 −4.880 1.00 32.44 C ATOM 37 CD1 ILE A 568 −8.044 3.281 −5.459 1.00 42.43 C ATOM 38 C ILE A 568 −5.717 7.413 −5.832 1.00 22.11 C ATOM 39 O ILE A 568 −4.695 7.761 −6.423 1.00 72.44 O ATOM 40 H ILE A 568 −7.436 6.987 −3.731 1.00 2.13 H ATOM 41 HA ILE A 568 −4.637 6.309 −4.357 1.00 71.23 H ATOM 42 HB ILE A 568 −5.939 4.900 −6.248 1.00 74.31 H ATOM 43 HG12 ILE A 568 −7.671 4.543 −3.795 1.00 32.44 H ATOM 44 HG13 ILE A 568 −8.187 5.396 −5.258 1.00 32.44 H ATOM 45 HG21 ILE A 568 −4.096 4.163 −4.675 1.00 22.21 H ATOM 46 HG22 ILE A 568 −5.309 3.895 −3.415 1.00 22.21 H ATOM 47 HG23 ILE A 568 −5.318 2.916 −4.888 1.00 22.21 H ATOM 48 HD11 ILE A 568 −9.056 3.073 −5.097 1.00 42.43 H ATOM 49 HD12 ILE A 568 −8.102 3.334 −6.547 1.00 42.43 H ATOM 50 HD13 ILE A 568 −7.420 2.430 −5.176 1.00 42.43 H ATOM 51 N ALA A 569 −6.920 7.896 −6.121 1.00 55.14 N ATOM 52 CA ALA A 569 −7.113 8.887 −7.173 1.00 73.42 C ATOM 53 CB ALA A 569 −8.485 8.814 −7.805 1.00 24.43 C ATOM 54 C ALA A 569 −6.657 10.297 −6.653 1.00 32.11 C ATOM 55 O ALA A 569 −7.599 10.823 −5.823 1.00 73.40 O ATOM 56 H ALA A 569 −7.697 7.580 −5.614 1.00 55.14 H ATOM 57 HA ALA A 569 −6.411 8.673 −7.966 1.00 73.42 H ATOM 58 HB1 ALA A 569 −8.533 9.430 −8.705 1.00 24.43 H ATOM 59 HB2 ALA A 569 −8.718 7.788 −8.081 1.00 24.43 H ATOM 60 HB3 ALA A 569 −9.260 9.175 −7.135 1.00 24.43 H ATOM 61 N MET A 575 −11.112 15.090 −11.894 1.00 25.14 N ATOM 62 CA MET A 575 −11.628 15.906 −12.986 1.00 51.31 C ATOM 63 CB MET A 575 −10.501 18.866 −13.456 1.00 0.12 C ATOM 64 CG MET A 575 −10.911 18.314 −13.742 1.00 24.30 C ATOM 65 SD MET A 575 −11.759 18.561 −15.335 1.00 20.34 S ATOM 66 CE MET A 575 −13.473 18.648 −14.761 1.00 11.54 C ATOM 67 C MET A 575 −12.075 15.032 −14.154 1.00 1.33 C ATOM 68 O MET A 575 −13.076 15.319 −14.810 1.00 54.34 O ATOM 69 H MET A 575 −10.232 15.299 −11.516 1.00 25.14 H ATOM 70 HA MET A 575 −12.481 16.455 −12.615 1.00 51.31 H ATOM 71 HB2 MET A 575 −9.742 16.938 −12.665 1.00 0.12 H ATOM 72 HB3 MET A 575 −9.970 16.453 −14.324 1.00 0.12 H ATOM 73 HG2 MET A 575 −11.496 18.734 −12.919 1.00 24.30 H ATOM 74 HG3 MET A 575 −9.996 18.915 −13.802 1.00 24.30 H ATOM 75 HE1 MET A 575 −13.756 17.753 −14.206 1.00 11.54 H ATOM 76 HE2 MET A 575 −13.610 19.529 −14.130 1.00 11.54 H ATOM 77 HE3 MET A 575 −14.136 18.739 −15.625 1.00 11.54 H ATOM 78 N LEU A 578 −15.411 12.762 −12.695 1.00 23.52 N ATOM 79 CA LEU A 578 −16.674 13.490 −12.667 1.00 14.05 C ATOM 80 CB LEU A 578 −16.406 14.884 −12.091 1.00 22.03 C ATOM 81 CG LEU A 578 −16.883 15.021 −10.649 1.00 24.35 C ATOM 82 CD1 LEU A 578 −18.399 15.040 −10.615 1.00 73.11 C ATOM 83 CD2 LEU A 578 −16.278 13.975 −9.733 1.00 23.41 C ATOM 84 C LEU A 578 −17.241 13.653 −14.073 1.00 63.24 C ATOM 85 O LEU A 578 −18.444 13.507 −14.291 1.00 51.34 C ATOM 86 H LEU A 578 −14.633 13.138 −12.234 1.00 23.52 H ATOM 87 HA LEU A 578 −17.373 12.918 −12.074 1.00 14.05 H ATOM 88 HB2 LEU A 578 −15.339 15.138 −12.137 1.00 22.03 H ATOM 89 HB3 LEU A 578 −16.901 15.648 −12.695 1.00 22.03 H ATOM 90 HG LEU A 578 −16.561 15.982 −10.266 1.00 24.35 H ATOM 91 HD11 LEU A 578 −18.829 14.046 −10.754 1.00 73.11 H ATOM 92 HD12 LEU A 578 −18.799 15.682 −11.405 1.00 73.11 H ATOM 93 HD13 LEU A 578 −18.727 15.452 −9.661 1.00 73.11 H ATOM 94 HD21 LEU A 578 −16.750 13.008 −9.897 1.00 23.41 H ATOM 95 HD22 LEU A 578 −16.423 14.239 −8.687 1.00 23.41 H ATOM 96 HD23 LEU A 578 −15.199 13.879 −9.893 1.00 23.41 H ATOM 97 N LEU A 600 −19.544 13.013 −5.696 1.00 1.43 N ATOM 98 CA LEU A 600 −18.998 13.511 −6.954 1.00 22.62 C ATOM 99 CB LEU A 600 −20.004 13.245 −8.081 1.00 34.01 C ATOM 100 CG LEU A 600 −19.798 11.901 −8.793 1.00 34.23 C ATOM 101 CD1 LEU A 600 −19.738 10.710 −7.845 1.00 64.24 C ATOM 102 CD2 LEU A 600 −20.943 11.693 −9.769 1.00 74.25 C ATOM 103 C LEU A 600 −18.631 14.987 −6.843 1.00 23.25 C ATOM 104 O LEU A 600 −17.455 15.348 −6.878 1.00 1.43 O ATOM 105 H LEU A 600 −20.400 12.537 −5.703 1.00 1.43 H ATOM 106 HA LEU A 600 −18.104 12.945 −7.173 1.00 22.52 H ATOM 107 HB2 LEU A 600 −21.030 13.293 −7.713 1.00 34.01 H ATOM 108 HB3 LEU A 600 −19.960 14.035 −8.828 1.00 34.01 H ATOM 109 HG LEU A 600 −18.863 11.943 −9.360 1.00 34.23 H ATOM 110 HD11 LEU A 600 −18.919 10.800 −7.130 1.00 64.24 H ATOM 111 HD12 LEU A 600 −19.562 9.787 −8.403 1.00 64.24 H ATOM 112 HD13 LEU A 600 −20.673 10.596 −7.289 1.00 64.24 H ATOM 113 HD21 LEU A 600 −20.873 10.725 −10.254 1.00 74.25 H ATOM 114 HD22 LEU A 600 −20.958 12.466 −10.541 1.00 74.25 H ATOM 115 HD23 LEU A 600 −21.888 11.707 −9.227 1.00 74.25 H

For example, by constructing a binding pocket structure through which FROUNT protein binds to CCR2B in silico using at least part of the identified atomic coordinates of FROUNT protein, calculating the strength of binding of the binding pocket structure to a compound library, and then selecting a compound that forms a stable complex with FROUNT protein, a novel substance that inhibits interaction between FROUNT protein and CCR2B or CCR5 can be identified. Databases in which the structure information of a number of compounds is registered are known, and such databases can be utilized as a compound library mentioned herein. By performing binding simulation on a library, evaluating the strength of the bindings based on the binding energy (chemical interaction energy), and selecting a compound, a compound expected to be capable of strongly binding to the binding pocket structure through which FROUNT protein binds to CCR2B (that is, capable of inhibiting binding between FROUNT protein and CCR2B or CCR5) can be selected.

By binding a candidate compound in silico to a binding pocket structure through which FROUNT protein binds to CCR2B, which structure is constructed based on the atomic coordinates, and evaluating the strength of the binding by calculation of the binding energy or the like, molecular designing of a candidate compound can be advanced such that a more stable complex can be formed. The structures of disulfiram and other candidate compounds can be modified into more desirable structures.

EXAMPLES

The present invention is described below by way of Examples more concretely. However, the present invention is not limited to the Examples described below. All the animal experiments were carried out in accordance with the guidelines of the Animal Care and Use Committee of the University of Tokyo.

1. Disulfiram Inhibits FROUNT-CCR2 Interaction METHODS High-Throughput Screening Based on HTRF (Homogeneous Time Resolved Fluorescence) Method

For the screening, a compound library containing 131,200 kinds of compounds dissolved in DMSO at a concentration of 10 mM or 2 mM (obtained from Open Innovation Center for Drug Discovery, the University of Tokyo) was used. The HTRF assay was carried out using a 384-well low-dose white microplate (Corning Coaster; catalog number 3676). In each well, 4 μL of a solution of 20 nM GST fusion FROUNT protein (a recombinant polypeptide prepared by fusing GST to aa 500-656 of SEQ ID NO:2), and DMSO or test compounds were mixed with a binding buffer (10 mM HEPES [pH 7.4], 0.2 M potassium fluoride, 10 mM NaCl, 0.1% Tween 20, and 0.5% bovine serum albumin [BSA]), and the plate was incubated at room temperature for 30 minutes. After the incubation, biotinylated CCR2 pro-C peptide (prepared by biotinylation of the 16 residues EKFRRYLSVFFRKHIT in the C-terminal region of CCR2B (SEQ ID NO:3)) at a final concentration of 250 nM, 2.6 ng of a europium cryptate-labelled anti-GST antibody, and 12.5 ng of high-grade XL665-conjugated streptavidin were added to each well. After incubation at room temperature for 20 hours, the HTRF signal was measured at emission wavelengths of 620 nm and 665 nm using a multilabel counter EnVision (PerkinElmer).

Binding Inhibition Assay Based on HTRF Method

By the same HTRF assay as described above, the abilities of disulfiram to inhibit the FROUNT-CCR2 interaction or the FROUNT-CCR5 interaction were investigated. As the CCR5 pro-C peptide, the 16 residues EKFRNYLLVFFQKHIA (SEQ ID NO:4) in the C-terminal region of CCR5 was used. As a control, the p53-MDM2 interaction (which is inhibited by an MDM-2-specific inhibitor Nutlin), which is also a helix peptide-protein interaction like the interaction between FROUNT and CCR2 or CCR5 but is not related thereto, was used.

[Results]

Disulfiram selectively inhibited the interactions between FROUNT-CCR2 and between FROUNT-CCR5, but did not inhibit the interaction between p53-MDM2 (FIG. 1A). The IC50 of disulfiram against FROUNT-CCR2 was calculated as 42 nM, which indicates about 100 times higher inhibitory action than that against p53-MDM2 (FIG. 1B).

The S—S bond of disulfiram (DSF) is decomposed by glutathione reductase in the body to generate diethyldithiocarbamate (DDC), and DDC is further metabolized to generate Me-DTC sulfoxide and Me-DTC sulfone. The result of investigation of the abilities of these metabolites to inhibit FROUNT is shown in Table 2. We investigated the FROUNT-inhibiting ability using sodium diethyldithiocarbamate as DDC to find that IC50 was 137 nM. Me-DTCs had an aldehyde dehydrogenase (ALDH)-inhibiting activity, but did not inhibit FROUNT. Thus, it was confirmed that the active substances responsible for inhibition of FROUNT and inhibition of ALDH are different from each other.

TABLE 2 Compound Structure ALDH inhibition FROUNT inhibition p53 inhibition DSF 7400 nM* 42 nM >1000 nM DDC n.d. 137 nM >1000 nM Me-DTC sulfoxide 930 nM* >1000 nM >1000 nM Me-DTC sulfone 530 nM* >1000 nM >1000 nM *Excerpt from Alcohol Clin Exp Res 1996. 20, 595-

We also investigated the FROUNT-inhibiting abilities of the zinc complex and the iron (III) complex of DDC to find that the IC50 values were 114.6 nM and 9.2 nM, respectively. In compounds that were modified such that the S—S bond of disulfiram was not decomposed (by modification of S—S into S—CH2—CH2—S or S—CH2—S), the ability to inhibit the interaction between FROUNT-CCR2 was lost.

REFERENCE

Toda, E. et al. Identification of a binding element for the cytoplasmic regulator FROUNT in the membrane-proximal C-terminal region of chemokine receptors CCR2 and CCR5. Biochem J 457, 313-322, doi:10.1042/BJ20130827 (2014).

2. Disulfiram Directly Binds to FROUNT METHODS

Surface Plasmon Resonance Method

Interactions between FROUNT and compounds in the library were analyzed by surface plasmon resonance (SPR) using Biacore T100 (GE Healthcare). Full-length FROUNT protein (SEQ ID NO:2) (Esaki, K. et al. Protein Expr Purif 77, 86-91, (2011)) was immobilized on a CM5 sensor chip. Solutions of disulfiram prepared by serial dilution in HBS-EP buffer containing 2% DMSO (GE Healthcare) were applied to the sensor chip at a flow rate of 30 μL/min. The resonance unit (RU) was measured during the process from the binding to the washing to analyze the binding dynamics. Biacore T100 evaluation software was used to carry out solvent correction for DMSO.

NMR Analysis of Spatial Structure of FROUNT Binding Region Complex

A fragment consisting of the region of the 500th to 656th amino acids of FROUNT protein and a CCR2 pro-C region fragment of 16 residues (SEQ ID NO:3) were prepared, and the binding state between the two fragments and the binding state between FROUNT protein and disulfiram were investigated by NMR analysis.

RESULTS

The SPR experiment revealed that disulfiram did not bind to CCR2 but directly bound to FROUNT protein (FIG. 2A). In the NMR analysis, the signal derived from FROUNT was affected by addition of disulfiram, suggesting that FROUNT and disulfiram directly interact with each other (FIG. 2B). It was also suggested that dimethyldithiocarbamate, a metabolite of disulfiram, binds to the same binding site on FROUNT as disulfiram (data not shown).

The analysis by chemical shift perturbation using NMR revealed that the residues M564, T565, 1568, A569, M575, L578, and L600 showed large chemical shifts in the NMR signal due to formation of the FROUNT-CCR2 complex. The binding pocket through which CCR2 binds to FROUNT protein is thought to be constituted by a plurality of residues including at least one of these seven residues. The atomic coordinates of these seven residues are as shown in the Table 1 described above.

3. Disulfiram Inhibits Tumor Hyperplasia METHODS Tumor Model

For evaluation of the subcutaneous tumor growth, LLC (Lewis lung cancer) cells (5×105) were suspended in 50 μL of PBS, and subcutaneously administered to the right abdomen of each mouse. The tumor size was measured using a caliper twice a week.

The tumor volume was calculated according to the following equation.


Tumor volume=(tumor shorter diameter)×tumor longer diameter/2

In order to investigate the antitumor effect in the subcutaneous tumor growth model, disulfiram was added in an amount of 0.8 mg/g to CE-2 powder feed (CLEA Japan, Inc.) supplemented with 5% sucrose (Wako), and the mice were fed therewith every day during the period from Day 4 to Day 11 post tumor inoculation. The control group was fed with a feed supplemented with the same amount of sucrose.

RESULTS

In the mouse group to which disulfiram was orally given, the tumor volume was significantly smaller than that in the control group (FIG. 3). It was revealed that, by inhibiting FROUNT with disulfiram, the proliferation of lung cancer cells transplanted into the body can be inhibited.

4. Disulfiram Inhibits Formation of Tumor Metastatic Lesions METHODS

To provide a lung metastasis model, 1×106 B16F10 melanoma cells were suspended in 200 μL of PBS, and administered from the tail vein of each of wild-type C57BL/6 mice. Administration of the inhibitor was carried out one day before, 30 minutes before, and one day after the tumor administration. To provide a control, DMSO and a pyrimidine fluoride anticancer drug 5-FU were administered. The mice were euthanized on Day 9 post tumor administration, and PBS was perfused from the left ventricle, followed by isolation of lungs. Visible lung metastatic lesions in the left lobe were counted.

RESULTS

FIG. 4 shows photographs of metastatic nodules on Day 9 post tumor administration, and a graph showing the result of counting of metastatic lesions. Disulfiram significantly reduced formation of lung metastatic lesions caused by the transplanted B16F 10 cells.

5. Disulfiram Inhibits Tumor Not Depending on Direct Cytocidal Action METHODS Cytotoxicity and Proliferation Assays

The cytotoxicity of inhibitors was tested by using LDH Cytotoxicity Detection Kit (TaKaRa) according to the manufacturer's instructions. Briefly, tumor cells were cultured in the presence or absence of an agent (disulfiram or 5FU) for 72 hours, and the culture supernatant was then collected and tested for the concentration of lactate dehydrogenase released from damaged cells. For measurement of cell proliferation, cells were incubated in a plate well together with an inhibitor for 48 hours. During the last 30 minutes, WST-1 (Dojindo Laboratories) was added to the culture liquid, and the absorbance of each well was measured at 450 nm versus 650 nm reference using EnVision (PerkinElmer) to detect the formazan level.

RESULTS

Recently, it has been reported that disulfiram has an activity to directly kill certain types of tumor cells (Chen, D., et al., Cancer Res 66, 10425-10433, (2006); and Chiba, T. et al., PLoS One 9, e84807, (2014)). To investigate whether the antitumor activity of disulfiram is mediated by direct killing of tumor cells, we compared disulfiram with a cytotoxic anticancer drug 5FU for their anticancer actions and cytotoxic activities. As a result, disulfiram did not show cytotoxic activity at a concentration at which 5FU completely killed LLC tumor cells (FIG. 5A). 5FU also showed cytotoxic activity and growth-inhibiting action on B16F10 melanoma cells, but disulfiram did not show any action on the tumor cells at the same concentration (FIG. 5B). As shown in FIG. 4, 5FU did not reduce metastasis of B16F10 tumor despite its tumor-killing activity, whereas, disulfiram effectively reduced metastasis of the tumor at the same dose. These results indicate that the antitumor effect of disulfiram on B16F10 melanoma cells and LLC cells is not due to the cytotoxic activity, but due to its action on the tumor microenvironment in the host.

6. FROUNT is Highly Expressed in Macrophages METHODS Preparation of FROUNT-gfp-Knock-In Mice

By a conventional method, FROUNT-gfp knock-in mice were prepared by incorporating a GFP gene downstream of the FROUNT promoter on the mouse genome.

Flow Cytometry

Mice were intraperitonealy injected with 2 mL of 4% thioglycolate to induce peritonitis, and infiltrating cells in the peritoneum were collected from these mice. The cells were washed with PBS supplemented with 2% fetal bovine serum, resuspended, and then filtered through a 70-μm strainer. The Fc receptor was blocked by incubation with an anti-mouse CD16/32 antibody (BD biosciences), and thereafter the cells were stained with a fluorescently labeled antibody. Anti-mouse CD11b-Pacific Blue, anti-mouse Ly6C-APC-Cy7, anti-mouse Ly6G-Alexa Fluor 700, anti-mouse CD4-FITC, and anti-mouse B220-PE-Cy7 were purchased from Biolegend. Anti-mouse CD8-Pacific Blue was purchased from BD biosciences.

RESULTS

Flow cytometry analysis of cells derived from the FROUNT-GFP knock-in mice revealed that FROUNT was highly expressed especially in macrophases among the immune cells recruited into the inflammatory sites in the peritonitis model (FIG. 6). It was shown that monocytes/macrophages highly expressing FROUNT were enriched in the cell population mobilized by CCL2, and that FROUNT was highly expressed in cells expressing its receptor CCR2 (data not shown).

In view of the fact that FROUNT is highly expressed in monocytes/macrophages, and the fact that FROUNT has an ability to bind to CCR2 and CCR5 expressed on monocytes/macrophages (Toda, E. et al. J Immunol 183, 6387-6394, (2009)), we subsequently investigated whether deficiency of FROUNT influences infiltration of macrophages into inflammatory sites and tumor sites.

7. Hyperplasia and Metastasis of Cancer Are Reduced in FROUNT-Deficient Mice

To investigate the role of FROUNT in a tumor microenvironment, mice in which FROUNT was knocked out were prepared using the cre/loxP system. Since complete deficiency of FROUNT causes embryonic lethality, conditional knockout was carried out using a system in which induction of recombination reaction was mediated by tamoxifen.

A targeting vector in which the genomic region containing exons 15 to 19 of the FROUNT gene was sandwiched between LoxP sequences was introduced into mice, and heterozygous FNTflox mice were crossed to create homozygous FNTflox/flox mice. Subsequently, B6.Cg-Tg(CAG-cre/Esr1*)5Amca mice (Jaxon Laboratory), to which Cre-ER, a fusion protein of Cre recombinase and a mutant estrogen receptor, was introduced, were crossed with the FNTflox/flox mice to obtain tamoxifen-inducible FROUNT conditional knockout mice FNT-cKO. By treating the FNT-cKO mice with tamoxifen, deletion of FROUNT was induced in both the genomic DNA and mRNA. It was confirmed that the expression of FROUNT mRNA was suppressed to half or less in the FNT-cKO mice treated with tamoxifen.

From 6 days or 14 days before the experiment, 8- to 16-week-old FNTflox/flox mice and FNT-cKO mice were fed with CE-2 powder feed (CLEA Japan, Inc.) supplemented with tamoxifen citrate (Wako Pure Chemical Industries, Ltd.) in an amount of 0.4 mg/1 g CE-2, to induce expression of Cre. Southern blotting was carried out to confirm the recombination.

(1) Reduction of Cancer Hyperplasia in FNT-cKO Mice

To the right abdomen of each of FNTflux/flux mice and FNT-cKO mice, 5×105 B16 melanoma cells were transplanted. Thereafter, the tumor size was measured using a caliper twice a week, and the tumor volume was calculated. As a result, significant tumor growth inhibition and improved survival rate were observed in FNT-cKO mice compared to non-knockout (FNTflox/flox) mice (FIGS. 7A and 7B).

(2) Reduction of Cancer Metastasis in FNT-cKO Mice

To each of FNTflox/flox mice and FNT-cKO mice, 1×106 B16 melanoma cells were intravenously administered, and the numbers and the sizes of metastatic nodules in lungs were visually observed on Day 8 post administration. As a result, it was revealed that both the numbers and the sizes of metastatic nodules were significantly decreased in FNT-cKO mice (FIGS. 8A and 8B).

8. Macrophage Infiltration into Inflammatory Sites Is Inhibited in FROUNT-Deficient Mice

METHODS In Vivo Chemotaxis Assay

Peritonitis was induced by intraperitoneal administration of 2 mL of 4% thioglycolate medium (Difco) to each of FNTflox/flox mice and FNT-cKO mice. Infiltrating cells in the peritoneum were collected by injecting 5 mL of ice-cold PBS into the abdominal cavity and giving a gentle massage. The collected cells was washed with PBS containing 0.1% FBS, and then subjected to flow cytometry analysis to investigate the cell number and the cell populations.

RESULTS

The results are shown in FIG. 9. The number of macrophages was decreased in FROUNT-deficient mice, confirming that infiltration of macrophages into peritoneal inflammatory sites was reduced in these mice (FIG. 9, upper panel). No difference was found in the number of neutrophils between the FROUNT-deficient mice and the non-deficient mice (FIG. 9, lower panel).

9. Disulfiram Inhibits Infiltration of Macrophages into Inflammatory Sites

METHODS In Vivo Chemotaxis Assay

The same treatment as in the above-described section 8 was carried out on wild-type mice to induce peritonitis, and infiltrating cells in the peritoneum were collected. After washing the collected cells, flow cytometry analysis was carried out to investigate the cell number and the cell populations. For evaluation of disulfiram, disulfiram was dissolved in DMSO, and then diluted in 2% Tween 80-containing physiological saline to each concentration shown in the figure. Each resulting disulfiram solution was administered to mice 1 day before and 30 minutes before the administration of thioglycolate.

RESULTS

Similarly as in the case of FROUNT-deficient mice, infiltration of macrophages into peritoneal inflammatory sites was reduced while infiltration of neutrophils was not influenced in the wild-type mice which received the FROUNT inhibitor disulfiram (FIG. 10).

10. Disulfiram Inhibits Cell Migration METHODS In Vitro Chemotaxis Assay

A human leukocyte cell line THP-1 was counted and resuspended in Boyden buffer (RPMI medium supplemented with 0.1% BSA), and the cells were preincubated with disulfiram before the chemotaxis assay. The cell migration activity was measured using a 96-well ChemoTX Chemotaxis Chamber (Neuro probe) with a polycarbonate filter with a pore size of 5 Chemokine was applied to the lower chamber of the plate, and the cells were applied to the upper chamber. After incubation at 37° C. in 5% CO2 for 90 minutes, the filter was removed, and the number of migrated cells in the lower chamber was counted using Cell Counting Kit F (Dojindo Laboratories).

RESULTS

By treatment with disulfiram at a concentration at which it does not exhibit cytotoxicity (data not shown), cell migration mediated by CCL2 was inhibited (FIG. 11).

11. Disulfiram Inhibits Accumulation of Macrophages in Tumor Sites METHODS Flow Cytometry

Lung cells of the lung metastasis model of wild-type mice described in the section 4 above or lung cells of the lung metastasis model of FNT-cKO mice and FNTflox/flox mice described in the section 7(2) above were obtained from the right lower lobe by digestion with collagenase and DNase. The cells were washed with PBS supplemented with 2% fetal bovine serum, resuspended, and filtered through a 70-μm strainer. The Fc receptor was blocked by incubation with an anti-mouse CD16/32 antibody (BD biosciences), and thereafter the cells were stained with a fluorescently labeled antibody. Anti-mouse CD11b-Brilliant Violet 510, anti-mouse Ly6C-APC-Cy7, and anti-mouse Ly6G-Alexa Fluor 700 were purchased from Biolegend. The stained cells were analyzed using the Gallios flow cytometer (Beckman coulter).

Immunohistochemical Staining

Each mouse was perfused with PBS, and the left lung was isolated. Optimal Cutting Temperature Compound (OCT) (Sakura Finetek) was injected from the trachea to embed the lung in OCT, and the lung was then frozen in liquid nitrogen. Fresh frozen sections with a thickness of 8 μm were prepared, and fixed with 4% paraformaldehyde-PBS. After washing with 0.05% Tween 20-PBS, the sections were blocked with Blocking One reagent (Nacalai Tesque), and sequentially stained with an anti-mouse F4/80 antibody (BioLegend) and Alexa Fluor 594 anti-rat IgG (Life technologies). Fluorescence Images were obtained with an SP5 confocal microscope (Leica Microsystems).

RESULTS

In the wild-type mice which received the FROUNT inhibitor disulfiram, accumulation of macrophages in tumor sites was reduced, compared to mice which received a solvent (FIG. 12). Also in the FROUNT-deficient mice, accumulation of macrophages in cancer metastatic nodules was significantly reduced compared to the non-deficient mice (data not shown).

The above-described results confirmed that mice to which the FROUNT inhibitor disulfiram was administered showed the same phenotypes as FROUNT-deficient mice, that is, inhibition of cancer hyperplasia, inhibition of cancer metastasis, and inhibition of macrophage infiltration into inflammatory sites.

12. Effect of Combined Use of Disulfiram and Anti-PD-1 Antibody METHODS

To provide an example of combined use of an anticancer drug and disulfiram (DSF), the effect of combined use of an anti-PD-1 antibody, which is an anticancer drug targeting an immune checkpoint molecule, and DSF was studied using an LLC tumor-bearing model and a B16 tumor-bearing model. The B16 tumor-bearing model was prepared by transplanting 5×105 B16 melanoma cells to the right abdomen of each of wild-type C57BL/6 mice. Preparation of the LLC tumor-bearing model was carried out in the same manner as in the above-described section 3. A feed supplemented with DSF was provided in the same manner as in the above-described section 3.

The day of the transplantation of the tumor cells to the mice was defined as Day 0. From Day 4, the mice were fed with the feed supplemented with DSF or a control feed every day during the experimental period. An anti-PD-1 antibody (J43, manufactured by BioXcell) was intraperitoneally administered once at a dose of 0.2 mg on each of Day 5, Day 8, Day 14, and Day 18 (four times in total). To provide a control for the antibody administration, PBS was intraperitoneally administered. The tumor size was measured using a caliper twice a week, and the tumor volume was calculated.

RESULTS

The result obtained from the LLC tumor-bearing model is shown in FIG. 13. In the LLC tumor-bearing model, administration of either one of an anti-PD-1 antibody and DSF inhibited hyperplasia of cancer cells, and combined administration of them showed an additive effect, confirming that the effect of an anti-PD-1 antibody to inhibit cancer hyperplasia was additively enhanced by the combined use with DSF.

The result obtained from the B16 tumor-bearing model is shown in FIG. 14. In the B16 tumor-bearing model, an inhibitory effect on cancer hyperplasia was not observed at a significant level when an anti-PD-1 antibody or DSF was administered alone, but combined use of them evidently inhibited hyperplasia of cancer cells, confirming that a synergistic effect was obtained.

13. Effect of Combined Use of Disulfiram and Anti-CD4 Antibody METHODS

To provide another example of combined use of an anticancer drug and DSF, the effect of combined use of an anti-CD4 antibody having high cytotoxic activity and DSF was studied using an LLC tumor-bearing model and a B16 tumor-bearing model. The LLC tumor-bearing model and the B16 tumor-bearing model were prepared in the same manner as in the above-described section 12. A feed supplemented with DSF was provided in the same manner as in the above-described section 12. After the transplantation of the tumor cells, the tumor size was measured using a caliper twice a week, and the tumor volume was calculated.

The day of the transplantation of the tumor cells to the mice was defined as Day 0. From Day 4, the mice were fed with the feed supplemented with DSF or a control feed every day during the experimental period. An anti-CD4 antibody (GK1.5, an antibody known to be capable of depletion of CD4+ cells in the mouse body by the CDC activity; manufactured by BioXcell) was intraperitoneally administered once at a dose of 0.2 mg on each of Day 5 and Day 8 (two times in total). To provide a control for the antibody administration, PBS was intraperitoneally administered.

RESULTS

The result of measurement of the tumor volume with time is shown in FIG. 15. In both the LLC tumor-bearing model (left) and the B16 tumor-bearing model (right), the effect of the anti-CD4 antibody to inhibit cancer hyperplasia was confirmed to be significantly enhanced by the combined use with DSF.

Claims

1. A method comprising inhibiting macrophages in a subject in need thereof by administering an effective amount of any of the following (1) to (3) to the subject:

(1) disulfiram, diethyldithiocarbamate, or a metal complex of diethyldithiocarbamate;
(2) a pharmaceutically acceptable salt of (1);
(3) a solvate of (1) or (2),
wherein said subject is a patient with inflammatory disease.
Patent History
Publication number: 20200253896
Type: Application
Filed: Feb 20, 2020
Publication Date: Aug 13, 2020
Applicant: The University of Tokyo (Tokyo)
Inventors: Yuya TERASHIMA (Tokyo), Kouji MATSUSHIMA (Tokyo), Etsuko TODA (Tokyo), Hiroaki TERASAWA (Kumamoto-shi), Sosuke YOSHINAGA (Kumamoto-shi)
Application Number: 16/796,220
Classifications
International Classification: A61K 31/145 (20060101); G16B 15/00 (20060101); A61K 45/06 (20060101); C12Q 1/32 (20060101); G01N 33/68 (20060101);